Neuraminidase inhibitor, oseltamivir blocks GM1 ganglioside-regulated excitatory opioid receptor-mediated hyperalgesia, enhances opioid analgesia ...

Page created by Charles Ruiz
 
CONTINUE READING
Brain Research 995 (2004) 260 – 266
                                                                                                                       www.elsevier.com/locate/brainres

                                                                  Research report
 Neuraminidase inhibitor, oseltamivir blocks GM1 ganglioside-regulated
   excitatory opioid receptor-mediated hyperalgesia, enhances opioid
               analgesia and attenuates tolerance in mice
                                                  Stanley M. Crain *, Ke-Fei Shen
           Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University, 1300 Morris Park Ave. Bronx, NY 10461, USA
                                                              Accepted 26 September 2003

Abstract

    The endogenous glycolipid GM1 ganglioside plays a critical role in nociceptive neurons in regulating opioid receptor excitatory signaling
demonstrated to mediate ‘‘paradoxical’’ morphine hyperalgesia and to contribute to opioid tolerance/dependence. Neuraminidase (sialidase)
increases levels of GM1, a monosialoganglioside, in these neurons by enzymatic removal of sialic acid from abundant polysialylated
gangliosides. In this study, acute treatment of mice with the neuraminidase inhibitor, oseltamivir enhanced morphine analgesia. Acute
oseltamivir also reversed ‘‘paradoxical’’ hyperalgesia induced by an extremely low dose of morphine, unmasking potent analgesia. In chronic
studies, co-administration of oseltamivir with morphine prevented and reversed the hyperalgesia associated with morphine tolerance. These
results provide the first evidence indicating that treatment with a neuraminidase inhibitor, oseltamivir, blocks morphine’s hyperalgesic effects
by decreasing neuronal levels of GM1. The present study further implicates GM1 in modulating morphine analgesia and tolerance, via its
effects on the underlying excitatory signaling of Gs-coupled opioid receptors. Finally, this work suggests a remarkable, previously
unrecognized effect of oseltamivir—which is widely used clinically as an antiviral agent against influenza—on glycolipid regulation of
opioid excitability functions in nociceptive neurons.
D 2003 Elsevier B.V. All rights reserved.

Theme: Neurotransmitters, modulators, transporter and receptors
Topic: Opioids: anatomy, physiology and behavior

Keywords: Neuraminidase inhibitor; Excitatory Gs-coupled opioid receptors; Opioid analgesia; Opioid hyperalgesia; Opioid tolerance

1. Introduction                                                                enous GM1 to nociceptive dorsal-root ganglion (DRG)
                                                                               neurons in vitro [24] enhances the efficacy of low-dose
   Previous in vitro and in vivo work demonstrates that the                    opioids in prolonging the calcium-dependent component of
monosialoganglioside GM1 [15,16] regulates opioid recep-                       the action potential duration (APD) [3,27]. The APD pro-
tor signal transduction in nociceptive neurons by enhancing                    longation results from excitatory signaling of opioid recep-
excitatory (Gs-mediated) but not inhibitory (Gi/Go-mediat-                     tors mediated by an adenylate cyclase/cyclic AMP/protein
ed) signaling [2,3,5 – 8,22,23,26,27,34,36,37]. Much exper-                    kinase A second-messenger system linked to K+ and Ca2 +
imental evidence suggests that this excitatory mode of opioid                  channels [1,2,5,21,24]. These directly evoked excitatory
receptors contributes to opioid tolerance and physical de-                     opioid receptor-mediated effects on the APD of DRG neuron
pendence [2,5 – 8,23,26] and mediates the ‘‘paradoxical’’                      perikarya may also result in increased Ca2 + influx in
hyperalgesia seen with very low doses of opioid agonists                       presynaptic DRG terminals and thereby account for the
[9]. Acute application of low (nM) concentrations of exog-                     observed low-dose opioid stimulation of high K+-evoked
                                                                               release of substance P (and probably other excitatory neuro-
                                                                               transmitters) in trigeminal neurons [30], as well as in
    * Corresponding author. Tel.: +1-718-430-2481 or +1-814-867-8609;
fax: +1-718-430-3381 or +1-814-867-7094.
                                                                               dissociated rat DRG neurons in culture [31]. By contrast,
    E-mail addresses: smcrain@aecom.yu.edu, stanleycrain@aol.com               activation of Gi/Go-coupled inhibitory opioid receptors by
(S.M. Crain).                                                                  higher concentrations of opioid agonists shortens the APD of

0006-8993/$ - see front matter D 2003 Elsevier B.V. All rights reserved.
doi:10.1016/j.brainres.2003.09.068
S.M. Crain, K.-F. Shen / Brain Research 995 (2004) 260–266                             261

DRG neurons and attenuates transmitter release [17]. Appli-                       the application of GM1 itself, markedly enhances the
cation of exogenous GM1 alone does not alter the APD of                           efficacy of low-dose opioids in prolonging the APD (Shen,
these DRG neurons [3,5,6,27].                                                     Crain and Ledeen, unpublished). Neuraminidase provides,
    In mice, systemic administration of low doses of GM1                          therefore, a significant metabolic means to supplement
rapidly attenuates morphine’s analgesic effects [7]. In con-                      direct synthesis of GM1 by a cyclic AMP-dependent gly-
trast, co-administration of the non-toxic B-subunit of chol-                      cosyltransferase system [2,6,16,20].
era toxin (CTX-B), which selectively binds to GM1 on cell                            The aim of the present work was to pharmacologically
membranes, appears to interfere with GM1’s modulation of                          decrease levels of endogenous GM1 in vivo in order to
excitatory opioid receptor signaling. Even at low doses,
cotreatment with CTX-B markedly enhances morphine’s
analgesic potency and blocks opioid tolerance/dependence
[26].
    These studies demonstrating opposing effects of GM1
and CTX-B on opioid analgesia were based on electrophys-
iologic analyses of nociceptive DRG neurons in vitro
[1,5,21,24]. In contrast to the enhancement of excitatory
opioid signaling after GM1 [27], application of CTX-B
rapidly blocks excitatory, but not inhibitory, signaling of
opioid receptors stimulated by morphine and other bimo-
dally acting opioid agonists on DRG neurons [22]. This
selective blockade of opioid receptor excitatory signaling by
CTX-B enhances opioid analgesia and attenuates the devel-
opment of opioid tolerance and dependence [5,6,22,23,26].
The blockade of morphine’s effects on excitatory opioid
receptors by CTX-B closely resembles the selective antag-
onism of excitatory opioid receptor signaling by ultra-low-
dose naloxone (NLX) or naltrexone (NTX) [4,5,25]. We
hypothesize that CTX-B binds to and blocks an allosteric
GM1-modulatory site on opioid receptors [5,6,22,26],
whereas ultra-low-dose NLX and NTX act as selective
competitive antagonists at excitatory opioid receptors [4 –
6,25].
    Neuraminidase is present in the plasma membrane of
neurons [16] and in vitro studies demonstrate that adminis-
tration of exogenous neuraminidase markedly increases the
concentrations of GM1 in DRG and other nerve cell
membranes by enzymatic removal of neuraminic (sialic)
acid from abundant polysialylated ligands of the gangliote-
traose series in these neurons [10,14,16,32,33,35,37]. Acute
application of neuraminidase to DRG neurons in vitro, like

Fig. 1. Analgesic effects of morphine are enhanced by cotreatment with
oseltamivir. (Hot water-immersion mouse tail-flick assays.) (a) Morphine
elicited a relatively small analgesic effect lasting < 2 h (z). Cotreatment
with morphine (1 mg/kg) plus oseltamivir (1 mg/kg) produced a dramatic
increase in the magnitude and duration of analgesia, lasting for about 5 h
(q). Oseltamivir alone did not induce analgesia (o) nor did the saline
control (.). (b) Pretreatment of mice with oseltamivir (1 mg/kg) for 16 h
prior to simultaneous cotreatment with morphine resulted in a stronger
enhancement in the magnitude and duration of morphine analgesia, lasting
>6 h (o). Morphine alone elicited a relatively small analgesic effect, lasting
< 2 h (.) and oseltamivir alone did not induce analgesia (z). (c) Histogram
bars indicate mean tail-flick latency (s) calculated during 0 – 6-h time-effect
tests as in (a), following cotreatment with morphine plus oseltamivir at 1,
10, 100 or 1000 Ag/kg. Morphine analgesia was significantly enhanced by
cotreatment with oseltamivir at 10 Ag/kg and higher doses. Note: In all
figures, the data are presented as mean F S.E.M. (n = 8). Asterisks indicate
statistically significant differences between time-points on curves of
cotreated vs. morphine alone groups (*p < 0.05, **p < 0.01).
262                                    S.M. Crain, K.-F. Shen / Brain Research 995 (2004) 260–266

further elucidate the role of GM1 in regulating excitatory             latencies >6 s were excluded from these tests and a 10-s
signaling of opioid receptors and associated hyperalgesic              cutoff was used to minimize tissue damage. Five sequential
effects. Oseltamivir was recently designed to act as a                 control tests were made, each with a 10-min interval. The
neuraminidase inhibitor that can attenuate viral removal of            latencies of the last four tests were averaged to provide a
sialic acid from cell surface glycoproteins (12). It is now            predrug value. Time-effect curves were plotted using tail-
widely used as a clinical antiviral agent to inhibit influenza         flick latencies as the ordinate. Experimentally induced
(13). Because oseltamivir was the first well-defined neur-             increases in control tail-flick latency provide a measure of
aminidase inhibitor to become available for systemic ad-               antinociceptive or analgesic effect, whereas decreases in
ministration in vivo, we decided to determine if it might also         tail-flick latency indicate hyperalgesic effects [9,26]. For
inhibit enzymatic conversion of abundant polysialoganglio-             preparation of the histograms in Figs. 1c and 3a, the mean
sides to GM1 ganglioside in neurons in nociceptive systems.            tail-flick latencies during 0 –6-h test periods were used.
The effects of systemic oseltamivir administration in mice                 All drugs were administered s.c. immediately following
were therefore examined on acute morphine analgesia, on                baseline antinociception measurements. Morphine was ad-
the hyperalgesia caused by low-dose morphine and on                    ministered at 0.1 Ag/kg in the hyperalgesia experiment (Fig.
morphine analgesic tolerance.                                          2) and at 1 mg/kg in all other experiments. Oseltamivir was
                                                                       administered at doses ranging from 1 Ag/kg to 1 mg/kg. In
                                                                       chronic experiments, mice were dosed b.i.d. (at 9 a.m. and 5
2. Materials and methods                                               p.m.), with nociception testing carried out during 6 h after
                                                                       a.m. injection on test days.
2.1. Animal test groups
                                                                       2.4. Statistical analyses
   Swiss – Webster (SW) male mice (20 – 25 g, Charles
River, NY) were housed separately in groups of five,                      The magnitude and duration of antinociception as mea-
maintained on a 12-h light/dark cycle, and provided water              sured by latency to tail-flick was tested by repeated
and food ad libitum for 1– 3 days prior to anti-nociception            measures analysis of variance (ANOVA). Results reported
testing. There were eight mice per treatment group. Com-               as differences between treatments over the full 6 h are F
parative tests were generally carried out on the same day.             tests of the main effects from the repeated ANOVA.
All animal test groups were used for only one assay, except            Overall time by treatment interactions that were significant
in chronic treatment tests where analgesia was tested after            at the 0.05 level or less were followed by paired t-tests of
1 –6 days. The protocols of this research project including            each treatment group vs. the comparison group (e.g., saline
the care and humane use of the mice were approved by the               or oseltamivir at each time point) using the Bonferroni
Animal Institute Committee at the Albert Einstein College              adjustment.
of Medicine.

2.2. Materials

    The oseltamivir phosphate (‘‘Tamiflu’’) used in this study
is a product of Hoffmann-La Roche (Nutley, NJ). Morphine
and NLX were purchased from Sigma (St. Louis, MO).

2.3. Antinociception and hyperalgesia assays

    Antinociceptive and hyperalgesic effects of opioids on
these mice were measured using a hot-water-immersion tail-
flick assay similar to methods previously described [7,9].
Each mouse was permitted to enter a tapered plastic cylinder
(with air holes). The cylinder was slightly larger than the
body size, with the tail freely hanging outside the cylinder.
The cylinder provided a secluded environment into which
the animals voluntarily enter. During the tail-flick assay, the
cylinder was handled without direct contact with the animal.           Fig. 2. Oseltamivir blocks low-dose morphine-induced hyperalgesia,
One-third of the tail from the tip was immersed into a water-          unmasking potent opioid analgesia. A dose of 0.1 Ag/kg morphine alone
bath maintained at 52 jC ( F 0.1j) with an electronic                  (z) elicited hyperalgesia indicated by a decrease in tail-flick latency in the
                                                                       following 4 h. Cotreatment with morphine plus oseltamivir (q) blocked the
thermoregulator (Yellow Springs). The latency to a rapid               morphine-induced hyperalgesia, unmasking prominent analgesia lasting
tail-flick was recorded and the mouse was returned to its              >5 h. Administration of oseltamivir alone (o) did not alter baseline tail-
cage during the period between tests. Mice with control                flick latency nor did the saline control (.).
S.M. Crain, K.-F. Shen / Brain Research 995 (2004) 260–266                                                    263

3. Results                                                                          drug injection and rapidly decreased to baseline levels
                                                                                    during the next hour. By contrast, the tail-flick latency of
3.1. Cotreatment of mice with oseltamivir enhances                                  morphine plus oseltamivir-treated mice reached a peak of
morphine’s analgesic effects                                                        7.2 s at 2 h post-drug injection and continued to be
                                                                                    significantly longer than the latency of saline-control mice
   Cotreatment of mice with 1 mg/kg morphine plus 1 mg/                             through 6 h. Oseltamivir alone did not alter baseline tail-
kg oseltamivir markedly increased both the magnitude and                            flick latency. The effects of morphine alone and of morphine
duration of morphine’s antinociception in a hot water (52                           plus oseltamivir differed from the effect of oseltamivir alone
jC) tail-flick assay (Fig. 1a, P < 0.01). The analgesia of                          ( P < 0.01 in both cases).
cotreated mice lasted >2 h longer than the analgesia from                               A stronger enhancement of the magnitude and duration
morphine alone. The mean baseline tail-flick latency in the                         of morphine analgesia by oseltamivir occurred when an
saline control group was about 4 s. Morphine alone elicited                         additional pretreatment of 1 mg/kg oseltamivir was given 16
a peak tail-flick latency of 6.3 s that occurred at 30 min after                    h prior to simultaneous cotreatment with morphine and

Fig. 3. Chronic cotreatment with morphine plus oseltamivir attenuates development of morphine tolerance. (a) After 5 days of treatment, mice that received
oseltamivir (1 mg/kg) plus morphine (1 mg/kg) retained a prominent analgesic response, whereas mice treated with morphine alone became markedly
hyperalgesic to morphine. Treatment with oseltamivir alone did not alter baseline tail-flick latency. (b) On day 6, a single co-administration of oseltamivir with
morphine (.) to the same group of morphine-tolerant mice shown in (a) restored prominent analgesia. Conversely, analgesic tolerance developed within 1 day
after withdrawal of oseltamivir from the group of mice previously cotreated with morphine plus oseltamivir (o; cf. (a)).
264                                     S.M. Crain, K.-F. Shen / Brain Research 995 (2004) 260–266

oseltamivir (Fig. 1b; P < 0.01 for morphine vs. morphine                with oseltamivir alone showed no change in tail-flick
plus oseltamivir). This group of mice showed peak tail-flick            latency from baseline over all test days. Repeated measures
values of 8.6 s within 30 min after administration of                   ANOVA of days 1, 3 and 5 revealed a main effect of
morphine plus oseltamivir and significant analgesia was                 treatment ( P < 0.001) and a treatment by time interaction
maintained for >6 h (Fig. 1b, cf. Fig. 1a). Morphine alone              ( P = 0.002). There were statistically significant differences
elicited a relatively small analgesic effect, reaching a peak           for morphine vs. morphine plus oseltamivir ( P < 0.001), for
tail-flick value of 6.2 s and lasting < 2 h. Nevertheless, in           morphine plus oseltamivir vs. oseltamivir ( P < 0.001), but
order to demonstrate the remarkable efficacy of oseltamivir             not for morphine vs. oseltamivir ( P = 0.365). While the
to rapidly enhance morphine analgesia, no pretreatment                  difference between morphine vs. oseltamivir averaged over
with oseltamivir was given prior to cotreatments in all of              the 3 days was not significant, on day 5, the mean difference
the subsequent experiments in the present study.                        was statistically significant ( P < 0.001).
    Dose– response assays showed that a dose of oseltamivir                 Furthermore, a single cotreatment of oseltamivir (b.i.d.)
as low as 10 Ag/kg significantly enhanced the analgesic                 plus morphine to morphine-tolerant, hyperalgesic mice
potency of 1 mg/kg morphine with only minor additional                  restored prominent analgesia, seen by a peak tail-flick
enhancement during cotreatments with 100 Ag/kg or 1 mg/                 latency of 7.8 s (Fig. 3b). By contrast, tolerance developed
kg oseltamivir (Fig. 1c, P < 0.01 for 10 Ag/kg, 100 Ag/kg               within 1 day after withdrawal of oseltamivir and adminis-
and 1 mg/kg vs. morphine alone).                                        tration of morphine (b.i.d.) alone to mice that had main-
                                                                        tained prominent analgesia during 5 days of cotreatment
3.2. Oseltamivir blocks acute hyperalgesia elicited by low-             with morphine plus oseltamivir (Fig. 3b, P < 0.01 for day 5
dose morphine in mice and unmasks potent opioid analgesia               vs. day 6 values for this group; cf. Fig. 3a).

    Acute administration of an extremely low dose of mor-
phine (0.1 Ag/kg) elicited prominent thermal hyperalgesia in            4. Discussion
contrast to saline-treated mice ( P < 0.01), manifested by a
decrease in tail-flick latency to a latency as low as 2.6 s                 The effects of the neuraminidase inhibitor oseltamivir
at 1 h ( P = 0.001). It remained significantly lower than               combined with morphine in the present work further sup-
the 4-s baseline tail-flick latency in saline-control mice and          ports the role of GM1 in modulating opioid analgesia and
lasted 4 h after drug injection (Fig. 2). This decrease in tail-        tolerance via its effects on the underlying excitatory signal-
flick latency from 4 to 2.6 s evoked by low-dose morphine               ing by opioid receptors in nociceptive neurons. Inhibition of
treatment indicates a substantial hyperalgesic effect (note             neuraminidase, the enzyme responsible for conversion of
that the minimum possible tail-flick latency in this assay is           abundant polysialogangliosides to GM1 [16], had dramatic
limited by the spinal reflex time, ca. 1.5 s). Acute cotreat-           effects on opioid analgesia and hyperalgesia. Co-adminis-
ment of mice with oseltamivir (1 mg/kg) plus morphine                   tration of oseltamivir with morphine markedly enhanced
(0.1 Ag/kg) blocked the hyperalgesia seen with this dose of             morphine-induced analgesia within the first hour of cotreat-
morphine alone and resulted in dramatic analgesia: the                  ment. A stronger enhancement of the magnitude and dura-
latency increased to a peak of 7.2 s at 1 h and was still 5.6           tion of morphine analgesia by oseltamivir occurred when an
s at 6 h ( P < 0.05 compared to saline; P < 0.01 compared to            additional pretreatment of 1 mg/kg oseltamivir was given 16
morphine alone). Administration of oseltamivir alone did                h prior to simultaneous cotreatment with morphine and
not change tail-flick latency more than 0.3 s in either                 oseltamivir (Fig. 1b). This result can be accounted for by
direction compared to baseline and tail-flick latency of this           a further decrease in GM1 levels in nociceptive neurons
group did not differ from that of the saline group.                     after a longer period of neuraminidase inhibition. Oseltami-
                                                                        vir reversed the well-documented, ‘‘paradoxical’’ hyper-
3.3. Chronic cotreatment with morphine plus oseltamivir                 algesia induced by an extremely low, acute dose of
attenuates opioid tolerance                                             morphine [9], unmasking a prominent analgesia from this
                                                                        dose of morphine lasting for >6 h. Chronic co-administra-
    Chronic cotreatment of mice with 1 mg/kg morphine plus              tion of oseltamivir with morphine strongly attenuated mor-
1 mg/kg oseltamivir for 5 days (b.i.d.) attenuated develop-             phine tolerance, preventing the development of hyperalgesia
ment of opioid tolerance, whereas control mice chronically              after initial analgesia and even reversed an established
treated with 1 mg/kg morphine alone became markedly                     hyperalgesia caused by repeated doses of morphine alone.
hyperalgesic after 3 days (Fig. 3a). After 5 days of treat-                 Our previous work has attributed these anti-analgesic
ment, mice receiving oseltamivir plus morphine retained a               tolerance effects of chronic opioid administration, as well as
prominent analgesic response and showed a mean tail-flick               the hyperalgesia induced by acute, very low-dose morphine,
latency of 6.0 vs. 4.2 s in oseltamivir-alone control mice              to the efficacy of GM1-regulated excitatory signaling by Gs-
( P < 0.05). Mice treated with morphine alone became even               coupled opioid receptors in nociceptive neurons [2,5 – 9].
more hyperalgesic to morphine, resulting in a mean tail-flick           Endogenous GM1 levels are significantly elevated by
latency of 2.9 vs. 4.2 s ( P < 0.05) (Fig. 3a). Mice treated            chronic opioid treatment of neuroblastoma  DRG neuron
S.M. Crain, K.-F. Shen / Brain Research 995 (2004) 260–266                                              265

hybrid F-11 cells [34] (see also Ref. [11]), a finding that            Cotreatment with oseltamivir or other similarly acting
adds support to the hypothesis that oseltamivir blocks                 neuraminidase inhibitors may, therefore, enhance the clini-
chronic morphine-induced hyperalgesia and tolerance by                 cal efficacy of morphine, oxycodone and other bimodally
decreasing GM1 levels in nociceptive neurons.                          acting opioid analgesics by blocking excitatory, GM1-reg-
    In vitro application of a specific neuraminidase inhibitor         ulated opioid receptor-mediated hyperalgesic effects, there-
has previously been shown to decrease GM1 levels in the                by markedly increasing analgesic potency and attenuating
plasma membrane of neuroblastoma cells, indicated by a                 opioid tolerance.
decrease in the binding of 125I-labeled CTX-B [38]. A
functional effect of inhibition of plasma membrane neur-
aminidase in vitro is the blockade of GM1-mediated axonal              Acknowledgements
elongation in hippocampal neurons [19]. Interestingly, elec-
trophysiological tests on DRG neurons in culture showed                   This study was supported by an unrestricted research
that application of polysialogangliosides, e.g. GD1a and               grant from Pain Therapeutics, CA. We thank Dr. Lindsay
GD1b, enhance excitatory opioid effects at 34 jC; however,             Burns for helpful editorial contributions in the preparation
this enhancement is delayed by 1 –2 h at 24 jC, suggesting             of this manuscript and Dr. Polly Bijur for valuable
that the efficacy of neuraminidase in converting the poly-             assistance with the statistical analyses.
sialogangliosides to GM1 is compromised at the lower
temperature [6,27]. Application of GM1 was rapidly effec-
tive at both temperatures [6,27].                                      References
    The present findings concur with previous in vivo results
that systemic administration of CTX-B, known to selective-              [1] S.M. Crain, K.-F. Shen, Opioids can evoke direct receptor-mediated
ly bind GM1 on the plasma membrane [10,15,16], also                         excitatory effects on sensory neurons, Trends Pharmacol. Sci. 11
enhances morphine’s analgesic potency and blocks opioid                     (1990) 77 – 81.
                                                                        [2] S.M. Crain, K.-F. Shen, After chronic opioid exposure sensory neu-
tolerance and withdrawal-associated hyperalgesia [26].                      rons become supersensitive to the excitatory effects of opioid agonists
We have proposed that, by binding to GM1, CTX-B pre-                        and antagonists as occurs after acute elevation of GM1 ganglioside,
vents GM1’s interaction with a putative allosteric regulatory               Brain Res. 575 (1992) 13 – 24.
site on excitatory (Gs-coupled) but not inhibitory (Gi/Go-              [3] S.M. Crain, K.-F. Shen, After GM1 ganglioside treatment of sensory
coupled) opioid receptors in nociceptive neurons                            neurons naloxone paradoxically prolongs the action potential but still
                                                                            antagonizes opioid inhibition, J. Pharmacol. Exp. Ther. 260 (1992)
[2,3,5,6,22,23,26,27,36,37]. Both the present results                       182 – 186.
with oseltamivir and the effects of CTX-B cotreatment with              [4] S.M. Crain, K.-F. Shen, Ultra-low concentrations of naloxone selec-
morphine are remarkably similar to the effects of                           tively antagonize excitatory effects of morphine on sensory neurons,
cotreatment with low-dose opioid antagonists plus opioid                    thereby increasing its antinociceptive potency and attenuating toler-
agonists. Ultra-low-dose NTX dramatically enhances opioid                   ance/dependence during chronic cotreatment, Proc. Natl. Acad. Sci.
                                                                            U. S. A. 92 1995, pp. 10540 – 10544.
analgesia in mice by selective and competitive antagonism               [5] S.M. Crain, K.-F. Shen, Modulation of opioid analgesia, tolerance and
of Gs-coupled opioid receptors that play a major role                       dependence by Gs-coupled, GM1 ganglioside-regulated opioid recep-
in the development of opioid tolerance/dependence                           tor functions, Trends Pharmacol. Sci. 19 (1998) 358 – 365.
[4,8,9,18,25,28,29].                                                    [6] S.M. Crain, K.-F. Shen, GM1 ganglioside-induced modulation of
    The specific effect of neuraminidase on the enzymatic                   opioid receptor-mediated functions, Ann. N.Y. Acad. Sci. 845 (1998)
                                                                            106 – 125.
conversion of polysialylated gangliosides to GM1 in neu-                [7] S.M. Crain, K.-F. Shen, Enhanced analgesic potency and reduced
rons is quite distinct from the role of neuraminidase in                    tolerance of morphine in 129/SvEv mice: evidence for a deficiency
promoting influenza virus release from infected cells and                   in GM1 ganglioside-regulated excitatory opioid receptor functions,
facilitating spread of the virus within the respiratory tract.              Brain Res. 856 (2000) 227 – 235.
                                                                        [8] S.M. Crain K.-F. Shen, Antagonists of excitatory opioid receptor
The antiviral effects of oseltamivir and other neuraminidase
                                                                            functions enhance morphine’s analgesic potency and attenuate opioid
inhibitors attenuate viral removal of sialic acid from cell                 tolerance/dependence liability, Pain 84 (2000) 121 – 131.
surface glycoproteins [12,13]. By contrast, we postulate that           [9] S.M. Crain, K.-F. Shen, Acute thermal hyperalgesia elicited by low-
the opioid modulatory effects of these neuraminidase inhib-                 dose morphine in normal mice is blocked by ultra-low naltrexone,
itors attenuate removal of sialic acid from neuronal glyco-                 unmasking potent opioid analgesia, Brain Res. 888 (2001) 75 – 82.
lipids, thereby decreasing GM1 levels in neurons in                    [10] P. Doherty, F.S. Walsh, Ganglioside GM1 antibodies and B-cholera
                                                                            toxin bind specifically to embryonic chick dorsal root ganglion neu-
nociceptive systems. The present study in mice suggests                     rons but do not modulate neurite regeneration, J. Neurochem. 48
that clinical administration of oseltamivir at doses (ca. 1 mg/             (1987) 1237 – 1244.
kg) that result in inhibition of influenza [12] may also have          [11] S.-F. Fan, S.M. Crain, Dual regulation by A, y and k opioid receptor
an additional effect by decreasing GM1 levels in nociceptive                agonists of K+ conductance of DRG neurons and neuroblasto-
neurons. This interesting dual role of oseltamivir and other                ma  DRG neuron hybrid F11 cells, Brain Res. 696 (1995) 97 – 105.
                                                                       [12] L.V. Gubareva, L. Kaiser, F.G. Hayden, Influenza virus neuramini-
neuraminidase inhibitors in enhancing opioid analgesia in                   dase inhibitors, Lancet 355 (2000) 827 – 835.
addition to attenuating influenza has not been previously              [13] C.U. Kim, W. Lew, M.A. Williams, H. Liu, L. Zhang, S. Swamina-
recognized in the clinical, as well as preclinical, literature.             than, N. Bischofberger, M.S. Chen, D.B. Mendel, C.Y. Tai, W.G.
266                                             S.M. Crain, K.-F. Shen / Brain Research 995 (2004) 260–266

       Laver, R.C. Stevens, Influenza neuraminidase inhibitors possessing a            masking potent opioid analgesia and attenuating opioid tolerance/de-
       novel hydrophobic interaction in the enzyme active site: design, syn-           pendence, Brain Res. 919 (2001) 20 – 30.
       thesis, and structural analysis of carbocyclic sialic acid analogues     [27]   K.-F. Shen, S.M. Crain, R.W. Ledeen, Brief treatment of sensory
       with potent anti-influenza activity, J. Am. Chem. Soc. 119 (1997)               ganglion neurons with GM1 ganglioside enhances the efficacy of
       681 – 690.                                                                      opioid excitatory effects on the action potential, Brain Res. 550
[14]   J. Kopitz, C. von Reitzenstein, K. Sinz, M. Cantz, Selective ganglio-           (1991) 130 – 138.
       side desialylation in the plasma membrane of human neuroblastoma         [28]   K.-F. Shen, S.M. Crain, R. Boston, A. de Kater, P. Moate, G. Schoen-
       cells, Glycobiology 6 (1996) 367 – 376.                                         hard, PTI-555 reverses and prevents morphine-induced tolerance and
[15]   R.W. Ledeen, Gangliosides of the neuron, Trends in Neurosci. 8                  naloxone-precipitated withdrawal in mice chronically treated with
       (1985) 169 – 174.                                                               morphine, J. Pain 3 (2002) 50.
[16]   R.W. Ledeen, Biosynthesis, metabolism, and biological effects of         [29]   K.-F. Shen, S.M. Crain, P. Moate, R. Boston, A. de Kater, G. Schoen-
       gangliosides, in: R.V. Margolis, R.K. Margolis (Eds.), Neurobiology             hard, PTI-801, a novel formulation of oxycodone, shows absence of
       of Glycoconjugates, Plenum, New York, 1989, pp. 43 – 83.                        tolerance, physical dependence and naloxone-precipitated withdrawal
[17]   R.A. North, Opioid receptor types and membrane ion channels,                    effects in mice, J. Pain 3 (2002) 49.
       Trends Neurosci. 9 (1986) 114 – 117.                                     [30]   H. Suarez-Roca, W. Maixner, Activation of kappa opioid receptors
[18]   K.J. Powell, N.S. Abul-Husn, A. Jhamandas, M.C. Olmstead, R.J.                  by U50488H and morphine enhances the release of substance P from
       Beninger, K. Jhamandas, Paradoxical effects of the opioid antagonist            rat trigeminal nucleus slices, J. Pharmacol. Exp. Ther. 264 (1993)
       naltrexone on morphine analgesia, tolerance, and reward in rats, J.             648 – 653.
       Pharmacol. Exp. Ther. 300 (2002) 588 – 596.                              [31]   H. Suarez-Roca, W. Maixner, Morphine produces a biphasic modu-
[19]   J.A. Rodriguez, E. Piddini, T. Hasegawa, T. Miyagi, C.G. Dotti,                 lation of substance P release from cultured dorsal root ganglion neu-
       Plasma membrane ganglioside sialidase regulates axonal growth                   rons, Neurosci. Lett. 194 (1995) 41 – 44.
       and regeneration in hippocampal neurons in culture, J. Neurosci. 21      [32]   A. Wieraszko, W. Seifert, The role of monosialoganglioside GM1 in
       (2001) 8387 – 8395.                                                             the synaptic plasticity: in vitro study on rat hippocampal slices, Brain
[20]   M.A. Scheideler, G. Dawson, Direct demonstration of the activation              Res. 345 (1985) 159 – 164.
       of UDP-N-acetylgalactosamine:[GM3]N-acetylgalactosaminyltrans-           [33]   G. Wu, R.W. Ledeen, Stimulation of neurite outgrowth in neuroblas-
       ferase by cyclic AMP, J. Neurochem. 46 (1986) 1639 – 1643.                      toma cells by neuraminidase: putative role of GM1 ganglioside in
[21]   K.-F. Shen, S.M. Crain, Dual opioid modulation of the action poten-             differentiation, J. Neurochem. 56 (1991) 95 – 104.
       tial duration of mouse dorsal root ganglion neurons in culture, Brain    [34]   G. Wu, S.F. Fan, Z.-H. Lu, R.W. Ledeen, S.M. Crain, Chronic opioid
       Res. 491 (1989) 227 – 242.                                                      treatment of neuroblastoma  dorsal root ganglion neuron hybrid F11
[22]   K.-F. Shen, S.M. Crain, Cholera toxin-B subunit blocks opioid ex-               cells results in elevated GM1 ganglioside and cyclic adenosine mono-
       citatory effects on sensory neuron action potentials indicating that            phosphate levels and onset of naloxone-evoked decreases in mem-
       GM1 ganglioside may regulate Gs-linked opioid receptor functions,               brane K+ currents, J. Neurosci. Res. 42 (1995) 493 – 503.
       Brain Res. 531 (1990) 1 – 7.                                             [35]   G. Wu, Z.-H. Lu, R.W. Ledeen, GM1 ganglioside modulates prosta-
[23]   K.-F. Shen, S.M. Crain, Chronic selective activation of excitatory              glandin E1 stimulated adenylyl cyclase in neuro-2A cells, Glycocon-
       opioid receptor functions in sensory neurons results in opioid ‘‘de-            jugate J. 13 (1996) 235 – 239.
       pendence’’ without tolerance, Brain Res. 597 (1992) 74 – 83.             [36]   G. Wu, Z.-H. Lu, R.W. Ledeen, Interaction of y-opioid receptor with
[24]   K.-F. Shen, S.M. Crain, Nerve growth factor rapidly prolongs the                GM1 ganglioside: conversion from inhibitory to excitatory mode,
       action potential of mature sensory neurons in culture and this effect           Molec. Brain Res. 44 (1997) 341 – 346.
       requires activation of Gs-coupled excitatory kappa opioid receptors      [37]   G. Wu, Z.-H. Lu, J.W. Tzongjer, R.D. Howells, K. Christoffers, R.W.
       on these cells, J. Neurosci. 14 (1994) 5570 – 5579.                             Ledeen, The role of GM1 ganglioside in regulating excitatory opioid
[25]   K.-F. Shen, S.M. Crain, Ultra-low doses of naltrexone or etorphine              effects, Ann. N.Y. Acad. Sci. 845 (1998) 126 – 138.
       increase morphine’s antinociceptive potency and attenuate tolerance/     [38]   C. von Reitzenstein, J. Kopitz, V. Schuhmann, M. Cantz, Differential
       dependence in mice, Brain Res. 757 (1997) 176 – 190.                            functional relevance of a plasma membrane ganglioside sialidase in
[26]   K.-F. Shen, S.M. Crain, Cholera toxin-B subunit blocks excitatory               cholinergic and adrenergic neuroblastoma cell lines, Eur. J. Biochem.
       opioid receptor-mediated hyperalgesic effects in mice, thereby un-              268 (2001) 326 – 333.
You can also read