ArticlePDF Available

NSAID Antinociception Measured in a Chemical and a Thermal Assay in Mice

Wiley
Pain Research and Management
Authors:
  • Univesidad de Chile

Abstract and Figures

The antinociceptive activity of several nonsteroidal anti-inflammatory drugs (NSAIDs) that were administered either intraperitoneally or intrathecally was assessed in mice by two algesiometric tests. The first was the writhing test, which assessed the abdominal constrictions that were induced by intraperitoneal acetic acid (a chemical test), and the second was the tail flick test, which measured pain responses to heat stimuli. The corresponding effective doses and their relative potencies were compared because these tests use different nociceptive stimuli with different transmission pathways. The intraperitoneal and intrathecal dose-response curves for the antinociception induced by every NSAID that was tested were parallel in the writhing test. In the tail flick test, however, only the intraperitoneal and intrathecal dose-response curves for ketoprofen, piroxicam, naproxen, nimesulide, paracetamol and diclofenac were parallel. The results obtained in this study confirm that NSAIDs possess different abilities to induce inhibition of cyclooxygenase, and they can be indirectly assessed by their different antinociceptive activities, depending on the algesiometric assays that are used. The antinociception of most NSAIDs might involve central mechanisms. The findings demonstrate the increasing importance of the spinal cord in processing and modulating nociceptive input, because intrathecal administration of NSAIDs is always more effective (in terms of potency) than systemic administration; thus, the antinociceptive efficacy of NSAIDs strongly depends on the algesiometric assay that is used and on the type of the nociceptive stimulus to which the test subject is exposed.
Content may be subject to copyright.
ORIGINAL ARTICLE
NSAID antinociception
measured in a chemical and a
thermal assay in mice
HF Miranda PhD, J Lopez MA, F Sierralta DDS, A Correa MA, G Pinardi MD
Pharmacology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
Correspondence: Dr HF Miranda, Faculty of Medicine, Universidad de Chile, Box 70.000, Santiago 7, Chile. Telephone +56-2-678-6052,
fax +56-2-737-2783, e-mail hmiranda@machi.med.uchile.cl
Received for publication August 30, 2000. Accepted August 29, 2001
HF Miranda, J Lopez, F Sierralta, A Correa, G Pinardi.
NSAID antinociception measured in a chemical and a
thermal assay in mice.
Pain Res Manage 2001;6(4):190-196.
The antinociceptive activity of several nonsteroidal anti-inflam-
matory drugs (NSAIDs) that were administered either intraperi-
toneally or intrathecally was assessed in mice by two
algesiometric tests. The first was the writhing test, which
assessed the abdominal constrictions that were induced by
intraperitoneal acetic acid (a chemical test), and the second was
the tail flick test, which measured pain responses to heat stimuli.
The corresponding effective doses and their relative potencies
were compared because these tests use different nociceptive
stimuli with different transmission pathways. The intraperitoneal
and intrathecal dose-response curves for the antinociception
induced by every NSAID that was tested were parallel in the
writhing test. In the tail flick test, however, only the intraperi-
toneal and intrathecal dose-response curves for ketoprofen,
piroxicam, naproxen, nimesulide, paracetamol and diclofenac
were parallel. The results obtained in this study confirm that
NSAIDs possess different abilities to induce inhibition of
cyclooxygenase, and they can be indirectly assessed by their dif-
ferent antinociceptive activities, depending on the algesiometric
assays that are used. The antinociception of most NSAIDs might
involve central mechanisms. The findings demonstrate the
increasing importance of the spinal cord in processing and mod-
ulating nociceptive input, because intrathecal administration of
NSAIDs is always more effective (in terms of potency) than sys-
temic administration; thus, the antinociceptive efficacy of
NSAIDs strongly depends on the algesiometric assay that is used
and on the type of the nociceptive stimulus to which the test sub-
ject is exposed.
Key Words: Antinociception; Nonsteroidal anti-inflammatory
drugs; Tail flick test; Writhing test
L’antinociception amenée par les AINS telle
que mesurée dans des tests chimique et ther-
mique chez la souris
RÉSUMÉ : L’activité antinociceptive de plusieurs anti-inflam-
matoires non stéroïdiens (AINS) qui ont été administrés soit par
voie intrapéritonéale, soit par voie intrathécale a été évaluée chez
des souris au moyen de deux tests algésiométrique. Le premier a
été le test de torsion qui visait à évaluer les contractions abdo-
minales induites par l’acide acétique intrapéritonéal (test chi-
mique) et le second était le test du battement de la queue qui
mesurait les réponses douloureuses aux stimuli thermiques (test
thermique). Les doses efficaces correspondantes et leur puis-
sance relative ont été comparées, parce que ces tests utilisent des
stimuli nociceptifs différents dont les voies de transmission sont
également différentes. Les courbes dose-réponse intra-péri-
tonéales et intrathécales pour l’antinociception induite par
chaque AINS testé ont été parallèles dans les tests de torsion.
Dans les tests de battement de la queue, par contre, seules les
courbes dose-réponse intra-péritonéales et intrathécales du kéto-
tifène, du piroxicam, du naproxène, du nimésulide, du paracéta-
mol et du diclofénac ont été parallèles. Les résultats obtenus lors
de cette étude confirment que les AINS induisent, à divers
degrés, l’inhibition de la cyclo-oxygénase et qu’ils peuvent indi-
rectement être évalués par leurs activités antinociceptives dis-
tinctes selon les tests algésiométriques utilisés. L’antinociception
de la plupart des AINS pourrait mettre en jeu des mécanismes
centraux. Les conclusions confirment l’importance croissante de
la moelle épinière dans le processus et la modulation de l’influx
nociceptif, parce que l’administration intrathécale des AINS est
toujours plus efficace (sur le plan de la puissance) que l’adminis-
tration systémique. Ainsi, l’efficacité antinociceptive des AINS
dépend fortement du test algésiométrique utilisé et du type de
stimulus nociceptif auquel le sujet est exposé.
190
Pain Res Manage Vol 6 No 4 Winter 2001
N
onsteroidal anti-inflammatory drugs (NSAIDs) are a
group of agents that possess both analgesic and anti-
inflammatory properties. They are used in the treatment of a
variety of pain and inflammatory disorders. There is sub-
stantial evidence that NSAID-induced analgesia is due to
the inhibition of prostaglandin synthesis in both the periph-
eral and central nervous systems (1). Prostaglandins are
generated through two isoforms of the enzyme cyclooxyge-
nase (COX): the constitutive isoform (COX-1) and the iso-
form that is induced at sites of inflammation (COX-2). Most
NSAIDs are principally inhibitors of COX-1, while others
(eg, meloxicam, nimesulide, celecoxib and rofecoxib) have
pharmacological profiles that are clearly different and are
characterized by a more selective inhibition of COX-2 (2).
Meloxicam, compared with other NSAIDs, is the most
potent inhibitor of prostaglandin biosynthesis in pleural and
peritoneal exudates, but is only a weak inhibitor in the gas-
tric tract and kidney. Unlike other NSAIDs, meloxicam
preferentially inhibits inducible human COX-2 in COS cells
(3,4). The activity of nimesulide against COX-1 and COX-2,
measured in vitro and in vivo, demonstrated that it is a
selective inhibitor of COX-2 (5). Several additional sub-
stances have been reported to be potentially involved in the
modulation of the antinociceptive activity of NSAIDs.
These include substance P, nitric oxide, adenosine triphos-
phate, glutamate, serotonin, neurokinin, noradrenaline, ion
channels and acetylcholine (6-13).
Previous studies have demonstrated that NSAIDs can
induce antinociception in several algesiometric tests; how-
ever, a detailed comparative study of the dose-response
characteristics of several NSAIDs in two different algesio-
metric tests has not been performed. The writhing test (vis-
ceral pain) and the tail flick test (somatic pain) are animal
models of acute pain in which the nociceptive stimulus and
the transmission pathways are different (14). The purpose of
the present study was to examine the antinociceptive effect
of NSAIDs that were administered intraperitoneally and
intrathecally in these chemical and thermal algesiometric
assays, to determine their corresponding effective doses and
to discuss their relative potencies.
ANIMALS AND METHODS
CF-1 mice that weighed 25±3 g were used throughout the
study. The animals were acclimated to the laboratory envi-
ronment for a minimum of 2 h before being used, and the
ethical standard guidelines were followed (14). In particular,
the durations of the experiments were as short as possible,
the number of animals involved was kept to a minimum, and
the animals were killed by the administration of an anes-
thetic overdose immediately after the recording period.
Each animal was used only once and received only one dose
of the drugs that were tested. All drugs were freshly pre-
pared by dissolving them in normal saline or in a slightly
hyperbaric solution of glucose (6%), and doses were calcu-
lated on the basis of the drug salts. All observations during
the assay were made in a randomized and blind manner.
Antinociceptive activity was evaluated as previously
reported (15). Briefly, intraperitoneal administration was
accomplished 30 min before the algesiometric test by inject-
ing the total dose in a constant volume of 10 mL/kg. The
Hylden and Wilcox (16) technique was used for intrathecal
administration 15 min before the algesiometric test, and the
doses of the drugs (dissolved in a slightly hyperbaric solu-
tion of glucose [6%] to limit diffusion) were injected in a
constant volume of 5 µL/g. The procedure was performed
rapidly, with a high degree of accuracy and reproducibility.
These times (30 min for intraperitoneal administration and
15 min for intrathecal administration) were found in previ-
ous experiments to be near the time of the onset of maxi-
mum analgesic effect.
Algesiometric tests
Writhing test: Mice were injected intraperitoneally with
10 mL/kg of 0.6% acetic acid, and starting 5 min after the
administration of the acetic acid solution, the number of
writhes during a 5 min period was counted. A writhe was
defined as a contraction of the abdominal muscles, accom-
panied by an elongation of the body and an extension of the
hind limbs. Dose-response curves, which were determined
near the time of peak effect, were constructed to assess the
antinociceptive actions of the different NSAIDs that were
administered either intraperitoneally or intrathecally. Eight
animals were injected at each of four doses to determine a
dose-response curve. The dose that produced 50% of
antinociception (AD
50
: 50% reduction of control writhes)
was calculated by a standard linear regression analysis of
the dose-response curve. Antinociceptive activity was
expressed as a percentage of inhibition of the usual number
of writhes that were observed in control animals that
received intraperitoneal saline (24.3±1.0, n=42) or intrathe-
cal glucose (23.9±0.5, n=42).
Tail flick test: A radiant heat tail flick test algesiometer unit
was used to measure response latencies according to the
method that was described by D’Amour and Smith (17).
Animals responded to a focused heat stimulus by flicking or
removing their inflicted tails from the heat source. The
intensity of the light beam was adjusted so that the control
latency (baseline) readings were approximately 2.5 s, and an
8 s cutoff time was used to prevent tissue damage. Anti-
nociception activity was expressed according to the follow-
ing formula:
% antinociception = 100 × [(test latency-control
latency)/(cutoff time-control latency)]
A dose-response curve was determined by using at least
four doses of each NSAID, and the AD
50
was determined by
linear regression analysis.
All of the drugs used in the study were dissolved in saline
solution when they were administered intraperitoneally or,
in the case of intrathecal administration, in a slightly hyper-
baric solution of glucose (6%) to limit diffusion (with the
exception of ibuprofen and nimesulide, which were dis-
solved in 5 mm dimethylsulphoxide; this vehicle was
NSAID antinociception
Pain Res Manage Vol 6 No 4 Winter 2001
191
Miranda et al
Pain Res Manage Vol 6 No 4 Winter 2001
192
Figure 1) Dose-response curves of the antinociceptive activity of naproxen, clonixin, ibuprofen and diclofenac administered intraperitoneally (i.p.) and
intrathecally (i.t.) in the writhing test in mice
Figure 2) Dose-response curves of the antinociceptive activity of meloxicam, piroxicam, paracetamol and nimesulide administered intraperitoneally (i.p.)
and intrathecally (i.t.) in the writhing test in mice
devoid of antinociceptive activity when tested by itself).
The following drugs, provided by local laboratories in
Chile, were used: ketoprofen (Rhône Poulenc Rorer), pirox-
icam (Pfizer), diclofenac (Novartis), naproxen (Saval),
ibuprofen and clonixine (Pharma Investi of Chile), metami-
zol (dipyrone, Sanderson), paracetamol (Pasteur), meloxi-
cam (Recalcine) and nimesulide (Schering Plough).
Results were presented as AD
50
values with 95% confi-
dence limits (95% CL). Parallelism of the lines that were
obtained by linear regression analysis of the dose-response
curves and relative potencies was calculated according to
the method described by Tallarida and Murray (18), and sig-
nificance was accepted at the 0.05 level.
RESULTS
Effects of NSAIDs in the writhing test
The intraperitoneal or intrathecal administration of the
NSAIDs produced dose-dependent antinociceptive activi-
ties in this chemical algesiometric test. The dose-response
curves are shown in Figures 1, 2 and 3. The intraperitoneal
and intrathecal dose-response curves for each NSAID were
statistically parallel. The corresponding AD
50
(with 95%
CLs) and the relative intraperitoneal/intrathecal potencies
are shown in Table 1. These values vary widely, depending
on the route of administration and the algesiometric test that
was performed. In the writhing test, AD
50
values ranged
from 0.85 to 221 mg/kg after intraperitoneal administration,
and ranged from 0.05 to 9.3 mg/kg after intrathecal admin-
istration.
The order of potency, from greatest to least, of the
antinociceptive activities that were displayed by NSAIDs
(given by intraperitoneal injection) was ibuprofen, meloxi-
cam, diclofenac, nimesulide, piroxicam, clonixine, metami-
zol, ketoprofen, naproxen and paracetamol. The order of
potency, from greatest to least, given by intrathecal injection
was ibuprofen, nimesulide, meloxicam, diclofenac, piroxi-
cam, metamizol, ketoprofen, clonixine, naproxen and para-
cetamol.
Effects of NSAIDs in the tail flick test
In the thermal algesiometric tail flick test, the NSAIDs that
were administered either intraperitoneally or intrathecally
produced dose-dependent antinociceptive activities. The
corresponding AD
50
values and their 95% CLs are shown in
Table 2. Dose-response curves for intraperitoneal and
intrathecal ketoprofen, piroxicam, naproxen, nimesulide,
paracetamol and diclofenac were statistically parallel and
are shown in Figures 4 and 5. The relative potencies (calcu-
NSAID antinociception
Pain Res Manage Vol 6 No 4 Winter 2001
193
Figure 3) Dose-response curves of the antinociceptive activity of metamizol and ketoprofen administered intraperitoneally (i.p.) and intrathecally (i.t.) in
the writhing test in mice
TABLE 1
Dose that produced 50% antinociception (AD
50
) values
with 95% confidence limits (CL) and relative potency of
nonsteroidal anti-inflammatory drugs (NSAIDs)
administered intraperitoneally (IP) and intrathecally (IT) in
the writhing test
AD
50
mg/kg Relative
(95% CL ) Potency
NSAID IP IT IT/IP
Naproxen 46.5 6.5 5.04
(20.7-104.1) (2.1-21.1)
Clonixine 11.3 1.1 9.46
(6.1-21.2) (0.38-3.1)
Ibuprofen 0.85 0.05 16.14
(0.12-6.1 ) (0.02-0.13)
Diclofenac 7.2 0.44 18.08
(2.6-19.9) (0.08-2.6)
Meloxicam 6.5 0.30 20.80
(3.5-11.8) (0.07-1.3)
Piroxicam 9.0 0.51 20.85
(3.3-24.1) (0.38-0.69)
Paracetamol 221.0 9.3 21.33
(140-346) (1.9-10.8)
Nimesulide 7.6 0.27 24.25
(3.7-15.4) (0.21-0.39)
Metamizol 28.5 0.79 24.92
(19.8-41.1) (0.4-1.5)
Ketoprofen 30.3 0.86 30.33
(11.3-81.6) (0.28-2.6)
The drugs are listed according to the increment in the relative potency
lated by the intrathecal/intraperitoneal ratio) of the NSAIDs
in which dose-response curves were parallel are summa-
rized in Table 2.
The order of potency, from greatest to least, of the
antinociceptive activities displayed by NSAIDs that were
given by intraperitoneal injection was nimesulide, ketopro-
fen, ibuprofen, meloxicam, clonixine, piroxicam, naproxen,
diclofenac, metamizol and paracetamol. Given by intrathe-
cal injection, the order of potency, from greatest to least,
was meloxicam, ketoprofen, diclofenac, nimesulide, clonix-
ine, naproxen, piroxicam, paracetamol, metamizol and
ibuprofen.
DISCUSSION
The results that were obtained in this study confirm that
NSAIDs possess different capacities to induce the inhibition
of COX, as reflected in their different antinociceptive activ-
ities in two nociceptive tests. In the tail flick test, intraperi-
toneal administration resulted in an AD
50
range of 28.6 to
572.2 mg/kg and intrathecal administration resulted in an
AD
50
range of 1.18 to 221.9 mg/kg. The dose-response
curves for intraperitoneal and intrathecal ibuprofen, clonix-
ine, meloxicam and metamizol, however, significantly devi-
ated from parallelism, which suggested that the
antinociceptive activities of these NSAIDs were produced
by the activation of different peripheral and spinal pathways
(19). In the chemical nociceptive test, ibuprofen was the
most potent drug administered either intraperitoneally or
intrathecally, while it was clearly less potent in the tail flick
test. Ibuprofen has been characterized as a purely competi-
tive inhibitor of COX-1 and COX-2, producing a readily
reversible effect (20). On the other hand, naproxen, which
was a low potency analgesic in the writhing test, is a more
selective COX-2 inhibitor. Naproxen shows a time-depend-
ent effect (involving a second step), which occurs within
seconds to minutes and may reflect the induction of a pro-
tein conformational change with the formation of a complex
in which the inhibitor is bound more tightly to the enzyme
(20). Because the tail flick test did not involve an inflamma-
tory component, COX-2 induction should not have been
present, and time-dependent inhibitors should conceivably
have exhibited lower antinociceptive activities.
In the rat spinal cord/tail in vitro preparation, very low
doses of paracetamol (10 to 50 µM) induced inhibition of
COX and blocked bradykinin-activation of nociceptors
(21). Even if it is not possible to establish a direct pharma-
cological correlation with this report (because of the nature
of the assays), paracetamol-induced antinociception with
algesiometric tests in the present study was evident at doses
that were four to 76 times higher than in other studies (6),
which strongly suggests the involvement of central neural
mechanisms in addition to COX inhibition.
In a study of NSAID suppression of evoked nociceptive
activity in individual thalamic neurons, Jurna (22) obtained
low AD
50
values for diclofenac and ibuprofen; these results
support the idea that central effects of NSAIDs contribute to
their analgesic efficacy because they do not completely
inhibit prostaglandin biosynthesis. The findings on the
effects of diclofenac and ibuprofen in the present acetic acid
writhing test are comparable with these previous results and
with those reported by Bjorkman (6), who demonstrated that
diclofenac induced antinociception in visceral pain models
(writhing test) but not in somatosensory models (tail flick
test).
Reports that ketoprofen is more potent than diclofenac as
an analgesic agent in vivo (23) do not agree with the results
of the writhing test in the present study. This finding could
be explained by differences in experimental models – the
route of administration (oral and intravenous compared with
intraperitoneal and intrathecal) and the active enantiomers
that were used (dexketoprofen compared with racemic keto-
profen).
The two models of animal pain that were used in this
study have been used extensively over time. They represent
different models of acute pain because the writhing test is a
chemical assay that may produce some inflammation and is
used as a model of visceral pain, while the tail flick test is a
somatic pain model, without inflammation. NSAIDs are less
effective (24,25) in the tail flick test than in the writhing
test. Our results show that, irrespective of the mechanisms
related to the quality and intensity of the pain stimuli, the
NSAIDs always elicited antinociception in both tests.
Miranda et al
Pain Res Manage Vol 6 No 4 Winter 2001
194
TABLE 2
Dose that produced 50% antinociception (AD
50
) values
with 95% confidence limits (CL) and relative potency of
nonsteroidal anti-inflammatory drugs (NSAIDs)
administered intraperitoneally (IP) and intrathecally (IT) in
the tail flick test
AD
50
mg/kg Relative
(95% CL ) Potency
NSAID IP IT IT/IP
Ketoprofen 43.7 4.3 8.16
(12.6-151.6) (1.6-11.6)
Piroxicam 160.2 25.5 13.50
(64.4-398.2) (1.3-509.4)
Naproxen 218.1 10.0 17.06
(58.9-807.7) (2.7-37.8)
Nimesulide 28.6 5.0 18.91
(9.4-86.8) (3.0-8.3)
Paracetamol 572.2 28.4 32.64
(172-1903) (13-61.8)
Diclofenac 424.1 4.5 33.61
(60.5-2972) (1.4-14.3)
Ibuprofen 57.3 221.9 N/A
(21.9-149.6) (197.5-296.3)
Clonixine 118.0 8.9 N/A
(62.8-221.6) (1.2-64.5)
Meloxicam 100.4 1.2 N/A
(42.3-238.5) (0.6-2.4)
Metamizol 467.4 179.8 N/A
(423.1-516.3) (35.9-900.4)
The drugs are listed according to the increment in the relative potency.
NA Not applicable
NSAID antinociception
Pain Res Manage Vol 6 No 4 Winter 2001
195
The results of this study indicate that antinociception
after the administration of most NSAIDs might involve cen-
tral neural mechanisms, which add to the peripheral
antinociception that is induced by COX inhibition. These
findings demonstrate the increasing importance of the spinal
cord in processing and modulating nociceptive input,
because intrathecal injections of NSAIDs are always more
effective in terms of potency than systemic administration.
The antinociceptive efficacy of NSAIDs, therefore, strongly
depends on the algesiometric assay that is used and on the
type of the nociceptive stimulus to which the test subject is
exposed.
ACKNOWLEDGEMENTS: This work was supported by grant
number 1990842 from FONDECYT, Chile.
REFERENCES
1. Walker JS. NSAID: an update on their analgesic effects. Clin Exp
Pharmacol Physiol 1995;22:855-60.
2. Hamilton LC, Warner TD. Interactions between inducible isoforms of
nitric oxide synthase and cyclo-oxygenase in vivo: investigations
using the selective inhibitors, 1400 W and celecoxib. Br J Pharmacol
1998;125:335-40.
3. Engelhardt G. Pharmacology of meloxicam, a new non-steroidal
anti-inflammatory drug with an improved safety profile through
preferential inhibition of COX-2. Br J Rheumatol
1996;35(Suppl 1):4-12.
Figure 4) Dose-response curves of the antinociceptive activity of ketoprofen, piroxicam, naproxen and nimesulide administered intraperitoneally (i.p.) and
intrathecally (i.t.) in the tail flick test in mice
Figure 5) Dose-response curves of the antinociceptive activity of paracetamol and diclofenac administered intraperitoneally (i.p.) and intrathecally (i.t.)
in the tail flick test in mice
Miranda et al
Pain Res Manage Vol 6 No 4 Winter 2001
196
4. Wolfe MM. Future trends in the development of safer nonsteroidal
anti-inflammatory drugs. Am J Med 1998;105:S44-52.
5. Cullen L, Kelly L, Connor SO, Fitzgerald DJ. Selective
cyclooxygenase-2 inhibition by nimesulide in man.
J Pharmacol Exp Ther 1998;287:578-82.
6. Bjorkman R. Central antinociceptive effects of non-steroidal anti-
inflammatory drugs and paracetamol. Experimental studies in the rat.
Acta Anaesthesiol Scand 1995;103:1-44.
7. Bovil JG. Mechanisms of actions of opioids and non-steroidal anti-
inflammatory drugs. Eur J Anesthesiol 1997;15:9-15.
8. Furst S. Transmitter involved in antinociception in the spinal cord.
Brain Res Bull 1999;48:129-41.
9. Papworth J, Colville-Nash P, Alam C, Seed M, Willoughby D.
The depletion of substance P by diclofenac in the mouse.
Eur J Pharmacol 1997;325:R1-2.
10. Bjorkman R, Hallman KM, Hedner J, Hedner T, Henning M.
Acetaminophen blocks spinal hyperalgesia induced by NMDA and
susbtance P. Pain 1994;57:259-64.
11. Granados-Soto V, Flores-Murrieta FJ, Castaneda-Hernández G,
López-Muñoz FJ. Evidence for the involvement of nitric oxide
in the antinociceptive effect of ketorolac. Eur J Pharmacol
1995;277:281-4.
12. Menzel JE, Kolarz G. Modulation of nitric oxide synthase activity by
ibuprofen. Inflammation 1997;21:451-61.
13. Tjolsen A, Lund A, Hole K. Antinociceptive effect of paracetamol in
rats is partly dependent on spinal serotonergic systems.
Eur J Pharmacol 1991;193:193-201.
14. Walker K, Fox AJ, Urban LA. Animal models for pain research. Mol
Med Today 1999;5:319-21.
15. Miranda HF, Sierralta F, Pinardi G. Previous administration of
indomethacin or naloxone did not influence ketorolac antinociception
in mice. Anesth Analg 1993;77:750-3.
16. Hylden JL, Wilcox GL. Intrathecal morphine in mice: a new
technique. Eur J Pharmacol 1980;67:313-6.
17. D’Amour FE, Smith DL. A method for determining loss of pain
sensation. J Pharmacol Exp Ther 1941;72:74-9.
18. Tallarida RJ, Murray RB. Manual of Pharmacologic Calculations with
Computer Programs. New York: Springer-Verlag, 1986.
19. Goldstein A, Aronov L, Kalman SM. Drug Action: The Basis of
Pharmacology, 2nd edn. New York: John Wiley & Sons, 1974.
20. Marnett LA, Kalgutkar AS. Cyclooxygenase 2 inhibitors:
discovery, selectivity and the future. Trends Pharmacol Sci
1999;20:465-9.
21. Dray A, Patel IA, Perkins MN, Rueff A. Bradykinin-induced
activation of nociceptors: receptor and mechanistic studies on the
neonatal rat spinal cord-tail preparations in vitro. Br J Pharmacol
1992;107:1129-34.
22. Jurna I. Central analgesic effects of non-steroidal anti-rheumatic
agents. Z Rheumatol 1991;50:7-13.
23. Cabre F, Fernández MF, Calvo L, Ferrer X, García ML,
Mauleón D. Analgesic, antiinflammatory and antipyretic
effects of S(+)-ketoprofen in vivo. J Clin Pharmacol
1998;38 (Suppl 12):S3-10.
24. Walker K, Fox AJ, Urban LA. Animal models for pain research.
Mol Med Today 1999;5:319-21.
25. Yaksh TL. Spinal systems and pain processing: development of novel
analgesic drugs with mechanistically defined models.
Trends Pharmacol Sci 1999;20:329-37.
Submit your manuscripts at
http://www.hindawi.com
Stem Cells
International
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
M ED IATO RS
INFLAMMATION
of
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Behavioural
Neurology
Endocrinology
International Journal of
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Disease Markers
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
BioMed
Research International
Oncology
Journal of
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Oxidative Medicine and
Cellular Longevity
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
PPAR Research
The Scientic
World Journal
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Immunology Research
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Journal of
Obesity
Journal of
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Computational and
Mathematical Methods
in Medicine
Ophthalmology
Journal of
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Diabetes Research
Journal of
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Research and Treatment
AIDS
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Gastroenterology
Research and Practice
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Parkinson’s
Disease
Evidence-Based
Complementary and
Alternative Medicine
Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com
... 3,7 The tail flick test is a thermal latency assay that has been extensively used to evaluate the anti-nociceptive effect of various drugs given systemically in both rats and mice. 8,9 The aim of present study was to compare the antinociceptive effects of intraperitoneal (IP) single dose of morphine, tramadol and meloxicam when used alone or in combination, using tail flick test in the male Wistar rats. ...
... Meloxicam, as a preferential inhibitor of inducible COX-2, may be less effective in the tail flick test as a somatic pain model without inflammation. 9 In conclusion, the results of present study indicate that TR and MO provide acceptable anti-nociceptive effects in rats. Combination of TR and MO showed strong antinociceptive effects. ...
Article
Full-text available
The purpose of the present study was to evaluate anti-nociceptive effects of morphine, tramadol, meloxicam and their combinations in rats. Seventy male Wistar rats were divided into seven equal groups and randomly assigned to receive intraperitoneal saline (S) (control group, 1.0 mL kg(-1)), morphine (MO) (4.0 mg kg(-1)), tramadol (TR) (12.5 mg kg(-1)), meloxicam (ML) (1.0 mg kg(-1)), tramadol- morphine (TR-MO), meloxicam-morphine (ML-MO) and meloxicam-tramadol (ML-TR) at the same doses. Anti-nociception was evaluated using tail flick latency (TFL) test at 45, 60, 75, 90 and 120 min after drug injection. The TFL was significantly higher in TR and MO groups compared to S group for 90 and 120 min, respectively. No significant change in TFL from baseline values was observed at all time points in ML group. Among rats that received combination of analgesics, those that received TR-MO had significantly greater TFL. There was no significant difference in TFL between ML-TR and ML-MO groups. In conclusion, TR, MO and their combination all provided acceptable anti-nociceptive effects in rats. Meloxicam at the given dosage (1.0 mg kg(-1)) did not demonstrate any anti-nociceptive effect when evaluated by TFL test.
... The pain pathway, characterized by its multisynaptic nature, offers opportunities for pain modulation at various action sites 25 . While NSAIDs are recognized for their impact on nociceptors, they also exhibit central mechanisms of pain relief 26,27 . ULEAF demonstrated significant analgesic activity in both tests. ...
Article
Full-text available
Background: The primary aim of this study is to identify the most active fraction from Urena lobata leaves and subsequently evaluate the analgesic and antioxidant accomplishments of the identified potent extract. The focus is on determining the fraction with the highest efficacy for potential therapeutic applications. Methods: U. lobata leaves were consecutively extracted with solvents like n-hexane, chloroform, ethyl acetate, and methanol. The concentrated fractions underwent initial phytochemical screening. The fraction exhibiting the highest activity, as determined by the carrageenan model, was further evaluated for its analgesic potential through the hot plate and acetic acid-induced writhing methods. In-vitro antioxidant activity was evaluated through DPPH and FRAP assays. Results: The preliminary phytochemical analysis of U. lobata leaf fractions exposed the presence of steroids, triterpenes, alkaloids, carbohydrates, tannins, and flavonoids. In the carrageenan-induced hind paw edema model, the ethyl acetate fraction of U. lobata leaf (ULEAF) demonstrated the highest anti-inflammatory effects, comparable to the standard drug Diclofenac sodium. Evaluation of analgesic potential using the hot-plate method and acetic acid-induced writhing test showed dose-dependent efficacy, with 500 mg/kg ULEAF exhibiting effects similar to ibuprofen. In vitro, antioxidant assays indicated potent radical scavenging and reducing power in the ethyl acetate fraction. Conclusion: In conclusion, U. lobata leaf fractions exhibited potent anti-inflammatory properties, with the ethyl acetate fraction demonstrating notable analgesic and antioxidant activity. These findings support the plant’s therapeutic potential for further exploration in pharmaceutical development.
... 2 Although analgesic properties of TA can be assessed in several screening pain tests for NSAIDs, including a mouse writhing test, formalin test or the rat Randal-Selitto test, in this research we focused on the NTG model. [40][41][42] Firstly because analgesic properties of TA in other animal pain and inflammation models were described previously, and secondly, our study was the firstin-animal experiment that assessed novel TA forms and it was aimed to be focused on their potential use in migraine. 43,44 Therefore we selected a pain model specific for migraine. ...
Article
Full-text available
Purpose: The poorly soluble nonsteroidal anti-inflammatory drug (NSAID), tolfenamic acid (TA), was studied to maximize its solubility, permeability through biological membranes, and pharmacological activity. Methods: A mixture with magnesium stearate (MS) - microenvironment pH-modifier was prepared, as well as systems additionally containing incorporating substances methyl-β-cyclodextrin (M-β-CD) and 2-hydroxypropyl-β-cyclodextrin (HP-β-CD). The identification of TA-MS-CD systems was confirmed using experimental methods: X-ray powder diffraction (XRPD) and Fourier transform infrared spectroscopy (FT-IR) with the theoretical support. Apparent solubility study was performed using the paddle apparatus, while in vitro gastrointestinal tract (GIT) and blood-brain barrier (BBB) permeability were conducted by using PAMPA (Parallel Artificial Membrane Permeability Assay). The in vivo part of the study used the mouse nitroglycerin (NTG)-induced migraine pain model. Results: From practically insoluble substance, TA in TA-MS-M-β-CD system dissolved up to 80.13% ± 2.77%, and in TA-MS-HP-β-CD up to 92.39% ± 3.25% in 180 minutes. An increase in TA permeability was also obtained in the TA-MS-M-β-CD and TA-MS-HP-β-CD systems through GIT membranes (Papp values 2.057 x 10-5 cm s-1 and 2.091 x 10-5 cm s-1, respectively) and through BBB (Papp values 3.658 x 10-5 cm s-1 and 3.629 x 10-5 cm s-1, respectively). The enlargement of the solubility and permeability impacted analgesia. The dose 25 mg/kg of both TA-MS-HP-β-CD and TA-MS-M-β-CD was almost equally effective and only slightly less effective than the dose 50 mg/kg of pure TA. Both TA-MS-HP-β-CD and TA-MS-M-β-CD used at 50 mg/kg more effectively attenuated tactile allodynia in NTG-treated mice than the same dose of pure TA. None of TA forms influenced heat hyperalgesia. Conclusion: Increasing solubility of TA caused an increase of its analgesic effect in an animal model of migraine pain.
... This corroborates the fact that analgesic action of NSAIDs also involve central mechanisms. 21 Despite the gaps in knowledge of the mechanism of pain and pain relief that exist at present, our study points out that zaltoprofen can be utilized as an effective alternative to standard analgesic in treatment of painful conditions. In a randomized, comparative, multicentric, double blind, double-dummy, Phase III clinical study conducted in patients with primary knee osteoarthritis, comparing zaltoprofen with diclofenac, it was concluded that the efficacy and safety of zaltoprofen is clinically non-inferior to that of diclofenac. ...
Article
Full-text available
Purpose: Pain is an unpleasant sensation, but a protective mechanism of our body. It is the most common medical complaint requiring a visit to a physician. The new non-steroidal anti-inflammatory drug (NSAID) - zaltoprofen, is a preferential COX-2 inhibitor. It also inhibits bradykinin-induced nociceptive responses by blocking the B2 receptor-mediated pathway in the primary sensory neurons. The present study was conducted to evaluate and compare the anti-nociceptive activity of zaltoprofen with a conventional NSAID - piroxicam, in a mouse model of acute pain using hot plate and tail flick tests. Materials and methods: Twenty-four adult Swiss albino mice (20-25 g) of either sex were used in this study. Oral zaltoprofen and piroxicam were used as test and standard drugs respectively. Anti-nociceptive activity was evaluated and compared using hot plate and tail flick tests. Results: In comparison to the control group (vehicle), zaltoprofen showed a significant increase in reaction time at various time periods in the hot plate and tail flick tests. In the hot plate method, zaltoprofen groups (15 and 20 mg/kg) showed a significant elevation in pain threshold in comparison to control group (vehicle) (p<0.001). In the tail flick model also, zaltoprofen groups (15 and 20 mg/kg) showed a significant increase in the reaction time in comparison to control group (vehicle). In both the analgesiometer assays, zaltoprofen was found to be non-inferior compared to a standard drug - piroxicam (positive control). Conclusion: Our study concludes that zaltoprofen is an effective analgesic agent in various pain models. Our results support that zaltoprofen has therapeutic potential for treating pain disorders and is non-inferior to a standard drug - piroxicam.
... En adéquation directe avec ce qui se passe en clinique, il a été mis en évidence l'existence d'une interaction synergique entre les AINS et les opioïdes (Kolesnikov et al, 2003), (Zelcer et al, 2005). En effet, l'injection intrathécale (injection dans l'espace sous-arachnoïdien, permettant un effet central au niveau de la moelle épinière et du liquide céphalo-rachidien) d'AINS est toujours plus efficace que l'injection systémique (effet périphérique) (Miranda et al, 2001). De plus, l'inhibition de la synthèse de sérotonine induit une diminution de l'activité antalgique des AINS dans différents types de douleurs (Miranda et al, 2003). ...
Article
Chronic, inflammatory, neuropathic, or incisional pain is affecting about 20 % of the adult population and up to 50 % of the elderly population. It thus represent a real public health issue. Despite the existence of large families of analgesics, treatments are often ineffective. This is due in large part to a lack of knowledge of the patho-physiological mechanisms of pain. During my PhD, I have been interested in the roles and regulation of molecular sensors of the pain recently highlighted: ion channels (ASICs "Acid Sensing Ion Channels "). ASICs constitute a family of excitatory cationic channels. The ASIC3 channels, in particular, are present in sensory neurons that innervate the skin, muscles, organs and joints. They are activated by low extracellular acidification occurring in many patho-physiological mechanisms such as inflammation, ischemia, tumor growth, or the subsequent tissue damage, for example, surgery. In a first study, we showed that ASIC3 channels play a crucial role in the development of post -operative pain, including postural pain, close to clinical cases. From a plantar incision model in rats, we demonstrated an over-expression of ASIC3 channels in sensory neurons innervating the operated hindpaw. Pharmacological inhibition (with toxin) and invalidation (siRNA) of ASIC3 in vivo reduce pain behavior. Our second study focused on the human ASIC3 channel, not yet extensively studied. I demonstrated that this channel has a unique and inducible property which gives it a sensitivity not only to acidification, but also to the extracellular alkalinization. This alkaline sensitivity is an intrinsic characteristic of the channel. It involves two specific arginine residues in the human channel that are present in its extracellular loop.Thus the human ASIC3 channel adapts its activity at different pH environments, and could participate in the fine regulation of membrane potential and neuronal sensitization. More recently, I have studied the regulation of ASIC3 channel by inflammatory lipids and there effects on pain. Interestingly, I showed that lysophosphatidylcholine (LPC), a lipid produced from the degradation of the membrane during inflammation, is a new activator of ASIC3 channel under normal pH conditions. Moreover, in synergy with moderate acidosis (pH 7.0), the LPC and its non-metabolizable analogue produce spontaneous pain in rats. This pain is reduced in the presence of the ASIC3 inhibitory toxin.
... Peripheral analgesics such as aspirin and paracetamol are exerting their antinociceptive action mainly via an inhibition of cyclooxygenases (COX), either at the peripheral site of inflammation, which is the case for aspirin (Smith et al. 2000), or in the central nervous system which is the case for paracetamol (Bujalska 2003). In addition to the COX inhibitory effect, several studies have suggested a role for aspirin and paracetamol in other, noninflammatory pain conditions, such as thermal, visceral and mechanical pain in rats and mice (Björkman 1995;Miranda et al. 2001). The mechanisms underlying this non-inflammatory pain modulation are not fully understood, but it has been suggested that central antinociceptive receptor mechanisms such as the adrenergic , serotonergic (Courade et al. 2001), opioid (Pini et al. 1997) and cholinergic Pinardi et al. 2003) systems are involved. ...
Chapter
The pharmacological premise upon which the prostaglandin G/H synthase II (PGHS-II) or cyclo-oxygenase-2 (COX-2) inhibitors was developed was essentially that founded in the identification of two different genes coding for the cyclo-oxygenases-1 and -2 [1–3]. COX-1 is considered to be a constitutive enzyme that is responsible for “housekeeping” or physiological functions (e.g., some gastrointestinal (GI) mucosal, renal and haemostatic functions and protective effects) while COX-2, which is induced by inflammatory stimuli, is considered to be important in inflammation, pain and fever [1–5]. Comprehensive reviews on the regulation and roles of these two isoenzymes in inflammation are found in [1–9] and Figure 1 summarizes their essential features.
Article
Full-text available
The pharmacological and toxicological properties of nimesulide have been previously reviewed (see [1-8]). The major part of these reviews has been concerned with the preclinical actions of the drug. A key issue concerning the interpretation of the in vitro effects of nimesulide has been the relationship of these to the plasma or synovial fluid concentrations of the drug, which are found during therapy. The review by Bennett and Villa [4] is noteworthy for having discriminated the in vitro effects, which are known to occur at therapeutic drug concentrations with those which are above this range. Thus, generally speaking although nimesulide has preferential COX-2 selectivity it is also an inhibitor of histamine release and actions, leukotriene B4 and C4, and platelet activating factor (PAF) production, the adherence and activation of neutrophils, collagenase and other metalloproteinases, glucocorticoid receptor phosphorylation, interleukin-6 production, calcium channel activation and is an antioxidant within the range of drug concentrations encountered therapeutically [4]. Thus, nimesulide can be regarded as having multifactorial actions in relation to its anti-inflammatory activity.
Article
This study was designed to evaluate the extent of the antinociceptive interaction between codeine and diclofenac at the local, spinal and systemic level. The effects of individual and fixed-ratio combinations of locally, spinally or orally given codeine and diclofenac were assayed using the formalin test in rats. Isobolographic analysis was employed to characterize the synergism produced by the combinations. Codeine, diclofenac and fixed-ratio codeine–diclofenac combinations produced a dose-dependent antinociceptive effect when administered locally, spinally or systemically. ED30 values were estimated for the individual drugs and isobolograms were constructed. Theoretical ED30 values for the combination estimated from the isobolograms were 422.2±50.5 μg/paw, 138.5±9.2 μg/rat, and 9.3±1.1 mg/kg for the local, spinal and oral routes, respectively. These values were significantly higher than the actually observed ED30 values which were 211.1±13.6 μg/paw, 45.9±3.9 μg/rat, and 2.5±0.2 mg/kg, indicating a synergistic interaction. Systemic administration resulted in the highest increase in potency, being about fourfold, while spinal and local administration increased potency in two- and threefold, respectively. The fact that the highest synergism was observed after systemic administration suggests that the interaction is occurring at several anatomical sites. The results support the clinical use of this combination in pain management.
Article
NSAIDs, including acetylsalicylic acid, are frequently classified as peripherally-acting analgesics. This is based on the fact that these substances, among other effects, inhibit the biosynthesis of prostaglandines. A few solid arguments, however, stand against an exclusively peripheral mode of the analgesic action of NSAIDs in the sense of an inhibition of prostaglandin synthesis. It should be noted here that the analgesically-effective doses do not suffice to block prostaglandin synthesis. Furthermore, the inhibiting effects of paracetamol and metamizol are far weaker or are not at all present, although both substances are reliably-effective analgesics. The NSAIDs indometacin, ibuprofen, and diclofenac are capable of suppressing the sensory response of the nociceptive system via a central effect. In experimental studies with rats under urethane anesthesia, the nociceptive activity of individual neurons of the thalamus (the dorsomedial part of the ventral nucleus) was measured. The activity was triggered via an electric stimulation of afferent C-fibres in the ipsilateral or contralateral sural nerve. The NSAIDs named suppressed the evoked nociceptive activity in a dose-dependent manner. At the highest doses, the suppression resulted in a difference of approx. 60% of the control activity. The ED50 values were 5 mg/kg for indometacin, 10.9 mg/kg for diclofenac, and 15.6 mg/kg for ibuprofen. These results support the theory that the central effects of NSAIDs contribute to their analgesic efficacy. The possible mechanisms of these effects will be discussed. A practical significance of the central analgesic effects of NSAIDs could be that their therapeutic applicability is not limited only to the treatment of pain, which results from an activation of nociceptors.
Article
The possible involvement of bulbo-spinal monoaminergic pathways in the antinociceptive effect of paracetamol was investigated in rats. Serotonergic pathways were lesioned with intrathecal 5,6-dihydroxytryptamine (5,6-DHT), and noradrenergic pathways with 6-hydroxydopamine (6-OHDA). Intact and lesioned rats were tested in the formalin test after i.p. paracetamol (400 mg/kg) or vehicle. Behaviour was scored for 1 h after the dorsal injection of 100 μ1 of 5% formalin into one hind paw. Behavioural variables were evaluated with a multivariate statistical procedure, as well as an analysis of variance. Paracetamol itself reduced pain-related behaviour and increased normal motor activity. This antinociceptive effect was reduced in rats lesioned with 5,6-DHT. In lesioned rats paracetamol caused a change in nociceptive behaviour from active, focused behaviour towards passive, protective and non-focused behaviour in the early phase of the formalin test. No significant effect of lesioning with 6-OHDA upon the paracetamol effect was found. These results show that activation of spinal serotonergic systems is involved in the antinociceptive effect of paracetamol. The relative importance of this mechanism in the central effect of paracetamol and the mechanisms that cause the activation remain to be determined.
Article
The effects of bradykinin on nociceptors have been characterized on a preparation of the neonatal rat spinal cord with functionally connected tail maintained in vitro . Administration of bradykinin to the tail activated capsaicin‐sensitive peripheral fibres and evoked a concentration‐dependent (EC 50 = 130 n m ) depolarization recorded from a spinal ventral root (L 3 ‐L 5 ). The response to bradykinin was unaffected by the peptidase inhibitors, bestatin (0.4 m m ), thiorphan (1 μ m ), phosphoramidon (1 μ m ) and MERGETPA (10 μ m ) or by the presence of calcium blocking agents, cadmium (200 μ m ) and nifedipine (10 μ m ). Inhibition of cyclo‐oxygenase with indomethacin (1–5 μ m ), aspirin (1–10 μ m ) and paracetamol (10–50 μ m ) consistently attenuated responses to bradykinin. The effect of bradykinin was mimicked by the phorbol ester PDBu, an activator of protein kinase C. The response to bradykinin was attenuated following desensitization to PDBu but desensitization to bradykinin did not induce a cross‐desensitization to PDBu. The protein kinase C inhibitor staurosporine (10–500 n m ) consistently attenuated the effects of PDBu and bradykinin. Bradykinin responses were reversibly enhanced by dibutyryl cyclic AMP (100 μ m ). However dibutyryl cyclic GMP (0.5 m m ) and nitroprusside (10 μ m ) produced prolonged block of responsiveness to bradykinin. Prolonged superfusion with pertussis toxin did not affect responses to bradykinin. The B 1 ‐receptor agonist des Arg ⁹ ‐bradykinin (10–100 μ m ) was ineffective alone or after prolonged exposure of the tail to lipopolysaccharide (100 ng ml ⁻¹ ) or epidermal growth factor (100 ng ml ⁻¹ ) to induce B 1 receptors. The B 1 ‐receptor antagonist, des Arg ⁹ Leu ⁸ ‐bradykinin (10 μ m ) did not attenuate the response to bradykinin. A number of bradykinin B 2 antagonists selectively and reversibly attenuated the response to bradykinin. The rank order potency was Hoe 140 > LysLys [Hyp ³ ,Thi 5,8 , d ‐Phe ⁷ ]‐bradykinin > d ‐Arg[Hyp ³ , Thi 5,8 , d ‐Phe ⁷ ]‐bradykinin = d ‐Arg[Hyp ² ,Thi 5,8 , d ‐Phe ⁷ ]‐bradykinin. These data show that bradykinin produces concentration‐dependent activation of peripheral nociceptors in the neonatal rat tail. The responses were unaffected by calcium channel block and were partially dependent on the production of prostanoids. Bradykinin‐evoked responses were consistent with the activation of protein kinase C‐dependent mechanisms. Cyclic GMP‐dependent mechanisms may be involved in bradykinin‐receptor desensitization whereas cyclic‐AMP dependent mechanisms increase fibre excitability and facilitate bradykinin‐induced responses. The effects of bradykinin were mediated by a B 2 receptor.
Article
NSAIDs, including acetylsalicylic acid, are frequently classified as peripherally-acting analgesics. This is based on the fact that these substances, among other effects, inhibit the biosynthesis of prostaglandines. A few solid arguments, however, stand against an exclusively peripheral mode of the analgesic action of NSAIDs in the sense of an inhibition of prostaglandin synthesis. It should be noted here that the analgesically-effective doses do not suffice to block prostaglandin synthesis. Furthermore, the inhibiting effects of paracetamol and metamizol are far weaker or are not at all present, although both substances are reliably-effective analgesics. The NSAIDs indometacin, ibuprofen, and diclofenac are capable of suppressing the sensory response of the nociceptive system via a central effect. In experimental studies with rats under urethane anesthesia, the nociceptive activity of individual neurons of the thalamus (the dorsomedial part of the ventral nucleus) was measured. The activity was triggered via an electric stimulation of afferent C-fibres in the ipsilateral or contralateral sural nerve. The NSAIDs named suppressed the evoked nociceptive activity in a dose-dependent manner. At the highest doses, the suppression resulted in a difference of approx. 60% of the control activity. The ED50 values were 5 mg/kg for indometacin, 10.9 mg/kg for diclofenac, and 15.6 mg/kg for ibuprofen. These results support the theory that the central effects of NSAIDs contribute to their analgesic efficacy. The possible mechanisms of these effects will be discussed. A practical significance of the central analgesic effects of NSAIDs could be that their therapeutic applicability is not limited only to the treatment of pain, which results from an activation of nociceptors.
Article
The involvement of nitric oxide in the antinociception produced by ketorolac was assessed using the pain-induced functional impairment model in the rat: 800 micrograms of NG-nitro-L-arginine methyl ester, an inhibitor of nitric oxide synthesis, or saline was injected intra-articularly in a hind limb joint previously injured with uric acid. Animals then received ketorolac, dipyrone or no drug. Ketorolac and dipyrone produced a significant antinociceptive effect which was reduced by pretreatment with NG-nitro-L-arginine methyl ester, but not with saline. It is concluded that the antinociceptive effect of both drugs involves the local participation of nitric oxide.