ArticlePDF Available

A Comparative Study of the Antiemetic Effects of α2-Adrenergic Receptor Agonists Clonidine and Dexmedetomidine against Diverse Emetogens in the Least Shrew (Cryptotis parva) Model of Emesis

MDPI
International Journal of Molecular Sciences (IJMS)
Authors:

Abstract and Figures

In contrast to cats and dogs, here we report that the α2-adrenergic receptor antagonist yohimbine is emetic and corresponding agonists clonidine and dexmedetomidine behave as antiemetics in the least shrew model of vomiting. Yohimbine (0, 0.5, 0.75, 1, 1.5, 2, and 3 mg/kg, i.p.) caused vomiting in shrews in a bell-shaped and dose-dependent manner, with a maximum frequency (0.85 ± 0.22) at 1 mg/kg, which was accompanied by a key central contribution as indicated by increased expression of c-fos, serotonin and substance P release in the shrew brainstem emetic nuclei. Our comparative study in shrews demonstrates that clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p.) and dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg, i.p.) not only suppress yohimbine (1 mg/kg, i.p.)-evoked vomiting in a dose-dependent manner, but also display broad-spectrum antiemetic effects against diverse well-known emetogens, including 2-Methyl-5-HT, GR73632, McN-A-343, quinpirole, FPL64176, SR141716A, thapsigargin, rolipram, and ZD7288. The antiemetic inhibitory ID50 values of dexmedetomidine against the evoked emetogens are much lower than those of clonidine. At its antiemetic doses, clonidine decreased shrews’ locomotor activity parameters (distance moved and rearing), whereas dexmedetomidine did not do so. The results suggest that dexmedetomidine represents a better candidate for antiemetic potential with advantages over clonidine.
This content is subject to copyright.
Citation: Sun, Y.; Darmani, N.A. A
Comparative Study of the Antiemetic
Effects of α2-Adrenergic Receptor
Agonists Clonidine and
Dexmedetomidine against Diverse
Emetogens in the Least Shrew
(Cryptotis parva) Model of Emesis. Int.
J. Mol. Sci. 2024,25, 4603. https://
doi.org/10.3390/ijms25094603
Academic Editor: Alain Couvineau
Received: 11 March 2024
Revised: 9 April 2024
Accepted: 19 April 2024
Published: 23 April 2024
Copyright: © 2024 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
International Journal of
Molecular Sciences
Article
A Comparative Study of the Antiemetic Effects of
α2
-Adrenergic
Receptor Agonists Clonidine and Dexmedetomidine against
Diverse Emetogens in the Least Shrew (Cryptotis parva) Model
of Emesis
Yina Sun and Nissar A. Darmani *
Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University
of Health Sciences, 309 East Second Street, Pomona, CA 91766, USA; yinasun@westernu.edu
*Correspondence: ndarmani@westernu.edu; Tel.: +1-909-469-5654; Fax: +1-909-469-5535
Abstract: In contrast to cats and dogs, here we report that the
α2
-adrenergic receptor antagonist
yohimbine is emetic and corresponding agonists clonidine and dexmedetomidine behave as anti-
emetics in the least shrew model of vomiting. Yohimbine (0, 0.5, 0.75, 1, 1.5, 2, and 3 mg/kg, i.p.)
caused vomiting in shrews in a bell-shaped and dose-dependent manner, with a maximum frequency
(0.85
±
0.22) at 1 mg/kg, which was accompanied by a key central contribution as indicated by
increased expression of c-fos, serotonin and substance P release in the shrew brainstem emetic nuclei.
Our comparative study in shrews demonstrates that clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p.) and
dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg, i.p.) not only suppress yohimbine (1 mg/kg, i.p.)-
evoked vomiting in a dose-dependent manner, but also display broad-spectrum antiemetic effects
against diverse well-known emetogens, including 2-Methyl-5-HT, GR73632, McN-A-343, quinpirole,
FPL64176, SR141716A, thapsigargin, rolipram, and ZD7288. The antiemetic inhibitory ID
50
values
of dexmedetomidine against the evoked emetogens are much lower than those of clonidine. At
its antiemetic doses, clonidine decreased shrews’ locomotor activity parameters (distance moved
and rearing), whereas dexmedetomidine did not do so. The results suggest that dexmedetomidine
represents a better candidate for antiemetic potential with advantages over clonidine.
Keywords:
α2
-adrenergic receptors; yohimbine; clonidine; dexmedetomidine; emesis; least shrew;
dorsal vagal complex
1. Introduction
Vomiting (emesis) is a protective reflex [
1
] which helps to remove ingested toxins from
the gastrointestinal tract (GIT) [
2
]. The emetic loci include the nucleus tractus solitarius
(NTS), the dorsal motor nucleus of the vagus (DMNX), and the area postrema (AP) in the
brainstem, as well as the enteric nervous system (ENS) and enterochromaffin cells (EC cells)
in the GIT [
3
]. The functional pathophysiology of vomiting indicates that emetic processes
are controlled via interactions between the gastrointestinal enteric nervous system, the
vagus, and the central nervous system (CNS) [
3
]. The emetic response involves multiple
neurotransmitters/mediators such as dopamine (DA), serotonin (5-HT), substance P (SP),
acetylcholine, histamine, prostaglandins, leukotrienes, and opiates, several of which have
been recognized as mediators of chemotherapy-evoked vomiting [
3
5
]. The act of emesis
may result from either direct activation of the brainstem dorsal vagal complex (DVC) emetic
nuclei including the AP, NTS, and DMNX via a blood-borne pathway, and/or indirect
stimulation of the DVC via activation of peripheral emetic loci such as the neurons of
the ENS and release of emetic neurotransmitters from the gastrointestinal EC cells which
subsequently activate the gastrointestinal vagal afferents to the brainstem [6,7].
α2
-Adrenergic receptors are expressed both presynaptically (auto-receptors) and post-
synaptically [
8
10
] in terminal fields in the nervous system, as well as on platelets which
Int. J. Mol. Sci. 2024,25, 4603. https://doi.org/10.3390/ijms25094603 https://www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2024,25, 4603 2 of 28
regulate several key physiological processes, including blood pressure, platelet aggre-
gation, insulin secretion, lipolysis, and neurotransmitter release [
11
]. Moreover, acti-
vation or blockade of presynaptic
α2
-adrenergic receptors respectively suppress or en-
hance norepinephrine (NE) release. In contrast, stimulation or antagonism of postsynaptic
α2
-adrenergic receptors exert direct inhibitory or excitatory postsynaptic effects, respec-
tively [
12
,
13
]. Systemic
α2
-adrenergic receptor perturbation can affect NE neurons in the
nucleus of the solitary tract [
14
,
15
], as well as many
α2
-adrenergic receptor-expressing
regions in the nervous system [1618].
α2
-Adrenergic receptor agonists (e.g., clonidine, xylazine) have been shown to evoke
emesis in both cats and dogs [
19
26
]. Moreover, clonidine appears to be the most po-
tent emetic
α2
-adrenergic receptor agonist with an ED
50
of 25
µ
g/kg (i.m.) in dogs and
0.075
µ
g/kg (i.c.v.) in cats [
20
,
21
]. The evoked emesis is thought to be mediated by
α2
-adrenergic receptors, since it was blocked by the prototypical monoterpenoid indole
alkaloid
α2
-adrenergic receptor antagonist, yohimbine [
19
21
]. In addition, several stud-
ies also demonstrate that dexmedetomidine can evoke dose-dependent emesis in cats
and dogs [
26
29
]. However, in contrast to its emetic effects in cats and dogs, clonidine
failed to trigger vomiting even at large doses in ferrets [
30
], pigeons [
31
], or least shrews
(present study). On the contrary, clonidine was found to prevent vomiting induced by
type 4 phosphodiesterase (PDE4) inhibitors in ferrets, and reserpine-evoked emesis in
pigeons, whereas administration of yohimbine alone in both species produced unexpected
vomiting [
30
,
31
]. Moreover, similar emetic responses were observed in ferrets following
administration of either peripherally acting
α2
-adrenergic receptor antagonist MK-467, or
its brain-penetrating analog MK-912, indicating both a peripheral and a central locus of
action [
30
]. In humans, nausea and vomiting are also common side-effects of parenterally
administered yohimbine [
32
]. In the clinical setting, a number of meta-analysis studies
show that the incidence of post-operative nausea and vomiting (PONV) can be significantly
reduced by both clonidine [3336] and dexmedetomidine [36,37] in patients.
Shrews are assigned to the order of Insectivora and are among the earliest animals.
Shrews are considered closer to primates than rodents, lagomorphs, and carnivores in the
phylogenetic system [
38
]. Unlike the large emetic animal models discussed above, the least
shrew (Cryptotis parva) is relatively much smaller (3–6 g in weight), and it is a well-validated
experimental emetic model that has been used to test the emetic and antiemetic potential of
diverse drugs [6,39].
Due to the discussed species differences, the present study sought to investigate the
emetic/antiemetic potential of
α2
-adrenergic receptor ligands in the least shrew animal
model of vomiting. Thus, we initially investigated whether intraperitoneal (i.p.) administra-
tion of varying doses of clonidine, dexmedetomidine, or yohimbine can evoke vomiting in
least shrews. We found that the
α2
-adrenergic receptor agonists clonidine and dexmedeto-
midine do not induce vomiting in least shrews, whereas its corresponding antagonist
yohimbine evoked emesis in a dose-dependent and bell-shaped manner. Secondly, we
examined via immunohistochemistry whether the dorsal vagal emetic loci in the least
shrew brainstem mediate the emetic effect of a maximally effective dose of yohimbine
(1.0 mg/kg, i.p.) through induction of c-fos, 5-HT-, and/or SP-release. Thereafter, we further
investigated whether clonidine and dexmedetomidine display broad-spectrum antiemetic
efficacy against diverse emetogens, such as selective agonists of serotonin 5-HT
3
(2-Methyl-
5-HT)-, substance P (SP) neurokinin NK
1
(GR73632)-, muscarinic M
1(McN-A-343)
, and
dopamine D
2
(quinpirole)-receptors, as well as the cannabinoid CB
1
receptor inverse ago-
nist/antagonist SR141716A, and Ca
2+
channel modulators, including the selective L-type
Ca
2+
channel (LTCC) agonist FPL6417 and the sarco/endoplasmic reticulum Ca
2+
-ATPase
(SERCA) inhibitor thapsigargin, which evoke pronounced vomiting in least shrews [
40
,
41
].
We also investigated the antiemetic potential of clonidine and dexmedetomidine against
the PDE4 inhibitor rolipram- [
30
,
42
] and the hyperpolarization-activated cyclic nucleotide-
gated (HCN) channel blocker ZD7288-induced vomiting [43].
Int. J. Mol. Sci. 2024,25, 4603 3 of 28
2. Results
2.1. Dose-Response Emetic Effect of Yohimbine in Least Shrews
Intraperitoneal injection of yohimbine (3 mg/kg) can evoke vomiting in all tested
ferrets [
30
]. In the present study, we initially assessed the emetic/antiemetic potential of
clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p., n= 6–10 per group), dexmedetomidine (0, 0.01,
0.05, 0.1, 0.5, and 1 mg/kg, i.p., n= 6–10 per group), and yohimbine (0, 0.5, 0.75, 1, 1.5, 2,
and 3 mg/kg, i.p., n= 7–14 per group) in the least shrew. Only yohimbine (0, 0.5, 0.75, 1, 1.5,
2, and 3 mg/kg, i.p.) increased both the frequency of emesis and the percentage of shrews
vomiting in a bell-shaped and dose-dependent manner (Figure 1A,B). A Kruskal–Wallis
(KW) non-parametric ANOVA test showed that, relative to the vehicle-pretreated control
group, yohimbine significantly increased the mean vomiting frequency (KW (6, 65) = 14.16;
p= 0.0279) with a maximum (0.85
±
0.22) occurring at 1 mg/kg (p= 0.0259; Dunn’s test).
At a dose of 3 mg/kg, few vomits were observed (Figure 1A). The chi-square test indicated
that the percentage of animals exhibiting emesis in response to yohimbine also showed a
bell-shaped and dose-dependent increase (
χ2
(6, 65) = 9.276, p= 0.1586), but a significant
difference was only observed at the 1 mg/kg dose, which evoked vomits in 61.54% of tested
shrews (p= 0.0445; chi-square test; Figure 1B).
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 3 of 28
investigated the antiemetic potential of clonidine and dexmedetomidine against the PDE4
inhibitor rolipram- [30,42] and the hyperpolarization-activated cyclic nucleotide-gated
(HCN) channel blocker ZD7288-induced vomiting [43].
2. Results
2.1. Dose-Response Emetic Eect of Yohimbine in Least Shrews
Intraperitoneal injection of yohimbine (3 mg/kg) can evoke vomiting in all tested fer-
rets [30]. In the present study, we initially assessed the emetic/antiemetic potential of
clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p., n = 6–10 per group), dexmedetomidine (0, 0.01,
0.05, 0.1, 0.5, and 1 mg/kg, i.p., n = 6–10 per group), and yohimbine (0, 0.5, 0.75, 1, 1.5, 2,
and 3 mg/kg, i.p., n = 7–14 per group) in the least shrew. Only yohimbine (0, 0.5, 0.75, 1,
1.5, 2, and 3 mg/kg, i.p.) increased both the frequency of emesis and the percentage of
shrews vomiting in a bell-shaped and dose-dependent manner (Figure 1A,B). A Kruskal–
Wallis (KW) non-parametric ANOVA test showed that, relative to the vehicle-pretreated
control group, yohimbine signicantly increased the mean vomiting frequency (KW (6,
65) = 14.16; p = 0.0279) with a maximum (0.85 ± 0.22) occurring at 1 mg/kg (p = 0.0259;
Dunn’s test). At a dose of 3 mg/kg, few vomits were observed (Figure 1A). The chi-square
test indicated that the percentage of animals exhibiting emesis in response to yohimbine
also showed a bell-shaped and dose-dependent increase (χ2 (6, 65) = 9.276, p = 0.1586), but
a signicant dierence was only observed at the 1 mg/kg dose, which evoked vomits in
61.54% of tested shrews (p = 0.0445; chi-square test; Figure 1B).
Figure 1. The pro-emetic effect of the α2-adrenergic receptor antagonist yohimbi ne and the correspond-
ing antiemetic efficacy of the α2-adrenergic receptor agonists clonidine and dexmedetomidine in least
shrews. Different groups of least shrews were given varying doses of yohimbine (0, 0.5, 0.75, 1, 1.5, 2,
3 mg/kg, i.p., n = 7–14 shrews per group (A,B). Emetic parameters were recorded for the next 30 min.
Figure 1. The pro-emetic effect of the
α2
-adrenergic receptor antagonist yohimbine and the corre-
sponding antiemetic efficacy of the
α2
-adrenergic receptor agonists clonidine and dexmedetomidine
in least shrews. Different groups of least shrews were given varying doses of yohimbine (0, 0.5, 0.75,
1, 1.5, 2, 3 mg/kg, i.p., n= 7–14 shrews per group (A,B). Emetic parameters were recorded for the
next 30 min. In drug interaction studies, different groups of least shrews were given an injection
(i.p.) of either the corresponding vehicle, or varying doses of clonidine (0.1, 1, 5, and10 mg/kg, i.p.,
n= 7–13 shrews per group (C,D) or dexmedetomidine (0.01, 0.05, and 0.1 mg/kg, i.p., n= 8–13 shrews
per group (E,F), 30 min prior to an injection of yohimbine (1 mg/kg, i.p.), and were observed for the
next 30 min. The frequency of emesis was analyzed with Kruskal–Wallis non-parametric one-way
ANOVA followed by Dunn’s post hoc test and presented as mean
±
SEM (A,C,E). The percentage
of shrews vomiting was analyzed with chi-square test and presented as mean (B,D,F). * p< 0.05,
** p< 0.01 vs. 0 mg/kg. The number of animals in each group is presented on the top of the
corresponding column.
Int. J. Mol. Sci. 2024,25, 4603 4 of 28
As shown in Figure 1C, the
α2
-adrenergic receptor agonist clonidine dose-depen-
dently attenuated the frequency of yohimbine (1 mg/kg, i.p.)-induced vomiting (KW
(4, 42) = 10.64; p= 0.0309) with an ID
50
value of 0.205 (0.01437–1.882) mg/kg. Clonidine
completely prevented the evoked vomiting at its 10 mg/kg dose (p= 0.0133). Clonidine
also reduced the percentage of shrews vomiting (
χ2
(4, 42) = 10.98, p= 0.0268) with an
ID
50
value of 0.2093 (0.02186–1.626) mg/kg, with significant reduction occurring at its
5 mg/kg (p= 0.0428) and complete protection at 10 mg/kg (p= 0.0048) (Figure 1D).
Likewise, dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg) reduced both the frequency
(KW (3, 34) = 8.073; p= 0.0445) and percentage (
χ2
(3, 34) = 8.78; p= 0.0324) of shrews
vomiting in response to yohimbine (1 mg/kg, i.p.) in a dose-dependent manner with
ID
50
values of 0.02113 (0.002887–0.0981) mg/kg and 0.02253 (0.004506–0.08576) mg/kg,
respectively (Figure 1E,F). Dexmedetomidine at 0.1 mg/kg completely suppressed both the
mean frequency (p= 0.0177) and the percentage (p= 0.0048) of shrews vomiting in response
to yohimbine (1 mg/kg, i.p.; Figure 1E,F).
2.2. Yohimbine Activates the Brainstem Emetic Nuclei
Since the 1 mg/kg (i.p.) dose of yohimbine caused maximal frequency of emesis in
the tested shrews, we conducted immunohistochemistry to determine c-fos responsiveness
following intraperitoneal administration of this dose of yohimbine. Figure 2A,B show that
very few c-fos positive cells were observed in the DVC emetic nuclei in shrew brainstem
sections from vehicle-treated controls, with mean values of 10.33
±
1.65, 56.5
±
4.99, and
9.67
±
1.15 in the AP, NTS, and DMNX, respectively (Figure 2E). Relative to the vehicle-
treated control group, a 1 mg/kg (i.p.) dose of yohimbine caused significant increases
in c-fos expression in the brainstem throughout the three DVC emetic nuclei. Following
vomiting induced by yohimbine, the average numbers of c-fos positive cells were increased
to 50.17
±
3.98 in the AP (p< 0.0001 vs. Vehicle), 166.3
±
15.41 in the NTS (p< 0.0001 vs.
Vehicle), and 36.17 ±4.73 in DMNX (p= 0.0003 vs. Vehicle), respectively (Figure 2C–E).
2.3. Effect of Yohimbine on 5-HT- and SP-Release in Brainstem Emetic Nuclei
Our previous studies have shown that emetogens (e.g., cisplatin, FPL64176, and
thapsigargin)-evoked emesis were accompanied by increases in 5-HT or SP immunoreactiv-
ity in brainstem emetic nuclei [
41
,
44
,
45
]. In the current study, we tested whether yohimbine
administration (1 mg/kg., i.p.) could also increase 5-HT and SP immunoreactivity in brain-
stem DVC containing emetic nuclei AP, NTS, and DMNX. Least shrews were euthanized at
15 min and 30 min post yohimbine treatment and were subjected to immunohistochemistry
to label 5-HT and SP. A representative image (Figure 3A) of the brainstem DVC emetic
nuclei after 5-HT immunolabelling shows that in vehicle-treated control group, the highest
density of 5-HT-positive fibers are in the dorsomedial subdivision of the NTS. A lower
density of the 5-HT immunoreactive profile is noted in the adjacent subnuclei of NTS
and DMNX. The AP had a few 5-HT-containing neurons. Figure 3D,J show that shrews
exhibited significant increases in 5-HT immunoreactivity in the AP (p= 0.0337 vs. vehicle),
NTS (p= 0.0298 vs. vehicle), and DMNX (p= 0.0446 vs. vehicle) at 15 min post yohimbine
treatment. Figure 3G,J indicate that at 30 min post-injection, 5-HT immunoreactivity in the
NTS, DMNX, and AP returned to basal levels.
Int. J. Mol. Sci. 2024,25, 4603 5 of 28
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 5 of 28
Figure 2. Immunohistochemical analysis of c-fos following emesis induced by systemic administra-
tion of the α2-adrenergic receptor antagonist yohimbine. Least shrews were sacriced 90 min post
vehicle treatment, or after the rst vomiting occurred post systemic administration of yohimbine (1
mg/kg, i.p., n = 6 shrews per group). Shrew brainstem sections (20 µm) were stained with rabbit c-
fos primary antibody and Alexa Fluor 594 donkey anti-rabbit secondary antibody. Nuclei were
stained with DAPI in blue. Representative tile-scanned images (20×) show robust c-fos induction in
the area postrema (AP), the nucleus tractus solitarius (NTS), and the dorsal motor nucleus of the
vagus (DMNX) in response to yohimbine (1 mg/kg, i.p.; (C,D)) when compared to the vehicle-
treated group (A,B). Scale bar = 100 µm. Quantied data show the yohimbine-induced c-fos expres-
sion in the AP, NTS, and DMNX in least shrew brainstem (E). Values represent the mean number of
c-fos-positive nuclei of each region (AP/NTS/DMNV) per section and are presented as mean ± SEM
(n = 6 shrews per group). *** p < 0.001, **** p < 0.0001 vs. Vehicle, Unpaired t-test.
Representative images acquired from SP immunolabeling are presented as Figure
3B,E,H. In the brainstem DVC of the vehicle-treated control group (Figure 3B), SP-immu-
noreactive bers were found in highest concentration within the DMNX and to a lesser
extent in the NTS, but rarely in the AP. Figure 3E,K show that yohimbine caused a signif-
icant increase in SP immunoreactivity only in the DMNX region of the shrew DVC (p =
0.0371 vs. vehicle) at 15 min post yohimbine treatment. Figure 3H,K show the intensity of
SP staining in the DMNX returned to basal level at 30 min post yohimbine injection.
Figure 2. Immunohistochemical analysis of c-fos following emesis induced by systemic administration
of the
α2
-adrenergic receptor antagonist yohimbine. Least shrews were sacrificed 90 min post vehicle
treatment, or after the first vomiting occurred post systemic administration of yohimbine (1 mg/kg,
i.p., n= 6 shrews per group). Shrew brainstem sections (20
µ
m) were stained with rabbit c-fos primary
antibody and Alexa Fluor 594 donkey anti-rabbit secondary antibody. Nuclei were stained with DAPI
in blue. Representative tile-scanned images (20
×
) show robust c-fos induction in the area postrema
(AP), the nucleus tractus solitarius (NTS), and the dorsal motor nucleus of the vagus (DMNX) in
response to yohimbine (1 mg/kg, i.p.; (C,D)) when compared to the vehicle-treated group (A,B).
Scale bar = 100
µ
m. Quantified data show the yohimbine-induced c-fos expression in the AP, NTS,
and DMNX in least shrew brainstem (E). Values represent the mean number of c-fos-positive nuclei
of each region (AP/NTS/DMNV) per section and are presented as mean
±
SEM (n= 6 shrews per
group). *** p< 0.001, **** p< 0.0001 vs. Vehicle, Unpaired t-test.
Int. J. Mol. Sci. 2024,25, 4603 6 of 28
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 6 of 28
Following 5-HT and SP labeling, sections were counterstained with DAPI to visualize cel-
lular nuclei (Figure 3C,F,I).
Figure 3. Immunohistochemical analysis of 5-HT and SP involvement following emesis induced by
systemic administration of the α2-adrenergic receptor antagonist yohimbine. Shrews (n = 5–6 per
gro up) we re euthanized at 15 and 30 min post vehic le or yohim bine (1 mg/kg, i.p.) injection. C orona l
brain sections (20 µm) containing the brainstem DVC emetic nuclei [area postrema (AP), nucleus of
the solitary tract (NTS), and dorsal motor nucleus of the vagus (DMNX)] were immunolabeled with
goat anti-5-HT antibody and rat anti-SP antibody overnight followed by Alexa Fluor 488 donkey
anti-goat and cy3-conjugated donkey anti-rat secondary antibody incubation. After counterstaining
with DAPI, images were acquired using a confocal microscope. Representative tile-scanned images
(20×) images (AI) show sections from vehicle, 15, and 30 min post yohimbine treated groups la-
beled with anti-5-HT (green), anti-SP antibodies (red), and merged with DAPI (blue), respectively.
Scale bar = 100 µm. Quantied data show the yohimbine-induced release of 5-HT and SP in the AP,
NTS, and DMNX regions of least shrew brainstem, respectively (J,K). Values represent the mean
gray value of released 5-HT and SP of each region (AP/NTS/DMNV) per section and are presented
as mean ± SEM (n = 5–6 shrews per group). * p < 0.05 vs. Vehicle, one-way ANOVA followed by
Dunne’s post hoc test.
Figure 3. Immunohistochemical analysis of 5-HT and SP involvement following emesis induced by
systemic administration of the
α2
-adrenergic receptor antagonist yohimbine. Shrews (n= 5–6 per
group) were euthanized at 15 and 30 min post vehicle or yohimbine (1 mg/kg, i.p.) injection. Coronal
brain sections (20
µ
m) containing the brainstem DVC emetic nuclei [area postrema (AP), nucleus of
the solitary tract (NTS), and dorsal motor nucleus of the vagus (DMNX)] were immunolabeled with
goat anti-5-HT antibody and rat anti-SP antibody overnight followed by Alexa Fluor 488 donkey
anti-goat and cy3-conjugated donkey anti-rat secondary antibody incubation. After counterstaining
with DAPI, images were acquired using a confocal microscope. Representative tile-scanned images
(20
×
) images (AI) show sections from vehicle, 15, and 30 min post yohimbine treated groups labeled
with anti-5-HT (green), anti-SP antibodies (red), and merged with DAPI (blue), respectively. Scale
bar = 100
µ
m. Quantified data show the yohimbine-induced release of 5-HT and SP in the AP, NTS,
and DMNX regions of least shrew brainstem, respectively (J,K). Values represent the mean gray value
of released 5-HT and SP of each region (AP/NTS/DMNV) per section and are presented as mean
±
SEM (n= 5–6 shrews per group). * p< 0.05 vs. Vehicle, one-way ANOVA followed by Dunnett’s
post hoc test.
Representative images acquired from SP immunolabeling are presented as Figure 3B,E,H.
In the brainstem DVC of the vehicle-treated control group (Figure 3B), SP-immunoreactive
fibers were found in highest concentration within the DMNX and to a lesser extent in the
NTS, but rarely in the AP. Figure 3E,K show that yohimbine caused a significant increase in
SP immunoreactivity only in the DMNX region of the shrew DVC (p= 0.0371 vs. vehicle)
at 15 min post yohimbine treatment. Figure 3H,K show the intensity of SP staining in the
DMNX returned to basal level at 30 min post yohimbine injection. Following 5-HT and SP
labeling, sections were counterstained with DAPI to visualize cellular nuclei (Figure 3C,F,I).
Int. J. Mol. Sci. 2024,25, 4603 7 of 28
2.4. The Broad-Spectrum Antiemetic Potential of the α2-Adrenergic Receptor Agonists Clonidine
and Dexmedetomidine against Vomiting Evoked by Diverse Receptor-Selective Emetogens
Figures 4and 5demonstrate the broad-spectrum antiemetic potential of both clonidine
and dexmedetomidine against vomiting evoked by diverse receptor-selective emetogens, re-
spectively. In fact, pretreatment with clonidine (0, 0.1, 1, 5, and 10 mg/kg, n= 7–10 per group),
reduced both the frequency (KW (4, 40) = 26.40; p< 0.0001) and percentage (
χ2
(4, 40) = 26.52;
p< 0.0001) of shrews vomiting in a dose-dependent manner in response to the administration
of the 5-HT
3
receptor-selective agonist, 2-Methyl-5-HT (5 mg/kg, i.p.), with respective ID
50
values of 0.6431 (0.224–1.494) mg/kg and 1.353 (0.7354–2.368) mg/kg (Figure 4A,B). The mean
frequency of 2-Methyl-5-HT-induced emesis was significantly reduced at its 5 mg/kg dose
(p= 0.0003) and with complete suppression at its 10 mg/kg dose (p= 0.0003). Signifi-
cant decreases in the percentage of shrews vomiting were also noted at its 5 mg/kg dose
(90%; p< 0.0001) and complete protection at 10 mg/kg dose (100%; p< 0.0001). Like-
wise, shrews pretreated with dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg, n= 8–10
per group) also reduced both the frequency (KW (3, 31) = 8.331; p= 0.0397) and percentage (
χ2
(3, 31) = 10.79; p= 0.0129) of shrews vomiting in a dose-dependent manner in response to 2-
Methyl-5-HT (5 mg/kg, i.p.) with ID
50
values of 0.02641 (0.007458–0.07622) mg/kg and 0.05137
(0.02349–0.1112) mg/kg, respectively (Figure 5A,B). Dexmedetomidine at 0.1 mg/kg signifi-
cantly suppressed both the mean frequency (p= 0.0124) and the percentage (75%; p= 0.005) of
shrews vomiting in response to 2-Methyl-5-HT (5 mg/kg, i.p.).
Next, the antiemetic effect of varying doses of clonidine and dexmedetomidine were
tested against vomiting caused by the NK
1
receptor selective agonist GR73632 (5 mg/kg,
i.p.). As shown in Figure 4C, clonidine (0, 0.1, 1, 5, and 10 mg/kg, n= 7–10 per group) also
caused dose-dependent decreases in the mean frequency (KW (4, 39) = 22.46;
p= 0.0002) of vomits induced by GR73632 with significant reductions occurring at its 1 mg/kg
(p= 0.0108)
, 5 mg/kg (p= 0.005), and 10 mg/kg (p= 0.0002) doses, and with an ID
50
value of
0.333 (0.1035–0.9538) mg/kg. The percentage of shrews vomiting was also reduced in a
dose-dependent fashion (
χ2
(4, 39) = 12.51; p= 0.0139) with significant reductions at its 5
(33.33%; p= 0.0466) and 10 mg/kg (71.43%; p= 0.0015) doses, and with an ID
50
value of 6.226
(2.959–13.5) mg/kg (Figure 4D). Figure 5C,D show that dexmedetomidine (0, 0.01, 0.05, and
0.1 mg/kg, n= 7–9 per group) dose-dependently suppressed vomiting caused by GR73632
(5 mg/kg, i.p.). The frequency of GR73632-induced emesis (KW (3, 28) = 8.195; p= 0.0421) was
significantly reduced at 0.1 mg/kg (p= 0.0156) with an ID
50
value of 0.04907 (0.0164–0.1427)
mg/kg (Figure 5C). Likewise, a significant decrease (
χ2
(3, 28) = 9.429; p= 0.0241) in the
percentage of animals vomiting were also noted at its 0.1 mg/kg dose (62.5%; p= 0.0048) with
an ID50 value of 0.08766 (0.04151–0.2017) mg/kg (Figure 5D).
Next, the antiemetic effect of clonidine and dexmedetomidine were assessed against
vomiting caused by the muscarinic M
1
receptor agonist, McN-A-343 (2 mg/kg, i.p.).
Figure 4E,F show that clonidine (0, 0.1, 1, 5, and 10 mg/kg, n= 6–9 per group) dose-
dependently suppressed vomiting (KW (4, 34) = 18.96; p= 0.0008) caused by McN-A-343
(2 mg/kg, i.p.). The frequency of McN-A-343-induced emesis was significantly reduced
at its 5 mg/kg dose (p= 0.0084) and completely reduced at its 10 mg/kg dose (p= 0.001)
with an ID
50
value of 0.6007 (0.1618–1.76) mg/kg (Figure 4E). Likewise, significant de-
creases (
χ2
(4, 34) = 20.92; p= 0.0003) in the percentage of animals vomiting were also
noted at its 5 mg/kg dose (75%; p= 0.0019) with complete protection at the 10 mg/kg
dose (100%; p= 0.0002), having an ID
50
value of 1.588 (0.744–3.202) mg/kg (Figure 4F).
Figure 5E,F show that dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg, n= 7–8 per group)
also dose-dependently suppressed vomiting caused by McN-A-343 (2 mg/kg, i.p.). The
frequency of McN-A-343-induced emesis was significantly reduced (KW (3, 27) = 11.11;
p= 0.0111) at 0.05 mg/kg (p= 0.0166) and 0.1 mg/kg (p= 0.0089) with an ID
50
value
of 0.02732 (0.009468–0.06793) mg/kg (Figure 5E). However, dexmedetomidine failed to
significantly protect shrews from vomiting (
χ2
(3, 27) = 6.285; p= 0.0985) having an ID
50
value of 0.08931 (0.03922–0.2283) mg/kg (Figure 5F).
Int. J. Mol. Sci. 2024,25, 4603 8 of 28
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 8 of 28
Figure 4. The antiemetic eects of the α2-adrenergic receptor agonist clonidine against vomiting
evoked by diverse emetogens. Varying doses of clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p.) were in-
jected to dierent groups of shrews 30 min prior to an injection of a fully eective emetic dose of
selective serotonin 5-HT3 receptor agonist 2-Methyl-5-HT (5 mg/kg, i.p., n = 7–10 shrews per group)
(A,B), the selective neurokinin NK1 receptor agonist GR73632 (5 mg/kg, i.p., n = 7–10 shrews per
group) (C,D), the muscarinic M1 receptor agonist McN-A-343 (2 mg/kg, i.p., n = 6–9 shrews per
group) (E,F), the dopamine D2/3 receptor preferring agonist quinpirole (2 mg/kg, i.p., n = 10 shrews
per group) (G,H), or the cannabinoid CB1 receptor-selective inverse agonist/antagonist SR141716A
(20 mg/kg, i.p., n = 8–10 shrews per group) (I,J). Emetic parameters were recorded for the next 30
min. The frequency of emesis was analyzed with KruskalWallis non-parametric one-way ANOVA
followed by Dunn’s post hoc test and presented as mean ± SEM (A,C,E,G,I). The percentage of
shrews vomiting was analyzed with chi-square test and presented as the mean (B,D,F,H,J). * p <
0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001 vs. 0 mg/kg (controls pretreated with vehicle of clonidine).
The number of animals in each group is presented on the top of the corresponding column.
Thereafter, the antiemetic eect of varying doses of clonidine (0, 0.1, 1, 5, and 10
mg/kg, n = 10 per group) were examined against vomiting caused by the dopamine D2/3
receptor agonist quinpirole (2 mg/kg, i.p.; Figure 4G,H). The mean frequency of
quinpirole-induced emesis (KW (4, 45) = 17.19; p = 0.0018) was signicantly reduced by
clonidine at its 1 mg/kg (p = 0.025), 5 mg/kg (p = 0.0051) and 10 mg/kg (p = 0.0013) doses,
with an ID50 value of 0.7356 (0.1996–2.239) mg/kg (Figure 4G). However, there was not a
signicant decrease (χ2 (4, 45) = 9.201; p = 0.0563) in the percentage of animals vomiting
with an ID50 value of 3.934 (1.30410.17) mg/kg (Figure 4H). Pretreatment with dexme-
detomidine (0, 0.01, 0.05, and 0.1 mg/kg, n = 911 per group) also dose-dependently sup-
pressed the mean frequency of vomiting (KW (3, 36) = 11.31; p = 0.0102) caused by
quinpirole (2 mg/kg, i.p.; Figure 5G). Signicant decreases in the frequency of vomiting
were noted at its 0.05 (p = 0.0188) and 0.1 mg/kg (p = 0.0079) doses with an ID50 value of
0.05819 (0.02685–0.1258) mg/kg (Figure 5G). The percentage of shrews vomiting (χ2 (3, 36)
Figure 4. The antiemetic effects of the
α2
-adrenergic receptor agonist clonidine against vomiting
evoked by diverse emetogens. Varying doses of clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p.) were
injected to different groups of shrews 30 min prior to an injection of a fully effective emetic dose
of selective serotonin 5-HT
3
receptor agonist 2-Methyl-5-HT (5 mg/kg, i.p., n= 7–10 shrews per
group) (A,B), the selective neurokinin NK
1
receptor agonist GR73632 (5 mg/kg, i.p., n= 7–10 shrews
per group) (C,D), the muscarinic M
1
receptor agonist McN-A-343 (2 mg/kg, i.p., n= 6–9 shrews
per group) (E,F), the dopamine D
2/3
receptor preferring agonist quinpirole (2 mg/kg, i.p., n= 10
shrews per group) (G,H), or the cannabinoid CB
1
receptor-selective inverse agonist/antagonist
SR141716A (20 mg/kg, i.p., n= 8–10 shrews per group) (I,J). Emetic parameters were recorded
for the next
30 min
. The frequency of emesis was analyzed with Kruskal–Wallis non-parametric
one-way ANOVA followed by Dunn’s post hoc test and presented as mean
±
SEM (A,C,E,G,I).
The percentage of shrews vomiting was analyzed with chi-square test and presented as the mean
(B,D,F,H,J).
*p< 0.05,
** p< 0.01, *** p< 0.001, **** p< 0.0001 vs. 0 mg/kg (controls pretreated
with vehicle of clonidine). The number of animals in each group is presented on the top of the
corresponding column.
Thereafter, the antiemetic effect of varying doses of clonidine (0, 0.1, 1, 5, and
10 mg/kg,
n= 10 per group) were examined against vomiting caused by the dopamine D
2/3
receptor
agonist quinpirole (2 mg/kg, i.p.; Figure 4G,H). The mean frequency of quinpirole-induced
emesis (KW (4, 45) = 17.19; p= 0.0018) was significantly reduced by clonidine at its 1 mg/kg
(p= 0.025), 5 mg/kg (p= 0.0051) and 10 mg/kg (p= 0.0013) doses, with an ID
50
value of
0.7356 (0.1996–2.239) mg/kg (Figure 4G). However, there was not a significant decrease
(
χ2
(4, 45) = 9.201; p= 0.0563) in the percentage of animals vomiting with an ID
50
value
of 3.934 (1.304–10.17) mg/kg (Figure 4H). Pretreatment with dexmedetomidine (0, 0.01,
0.05, and 0.1 mg/kg, n= 9–11 per group) also dose-dependently suppressed the mean
frequency of vomiting (KW (3, 36) = 11.31; p= 0.0102) caused by quinpirole (2 mg/kg,
i.p.; Figure 5G). Significant decreases in the frequency of vomiting were noted at its 0.05
(p= 0.0188) and 0.1 mg/kg (p= 0.0079) doses with an ID
50
value of 0.05819 (0.02685–0.1258)
Int. J. Mol. Sci. 2024,25, 4603 9 of 28
mg/kg (Figure 5G). The percentage of shrews vomiting (
χ2
(3, 36) = 11.72; p= 0.0084) was
significantly reduced at its 0.1 mg/kg dose (36.36%, p= 0.0341) with an ID
50
value of 0.276
(0.1526–0.6753) mg/kg (Figure 5H).
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 9 of 28
= 11.72; p = 0.0084) was signicantly reduced at its 0.1 mg/kg dose (36.36%, p = 0.0341) with
an ID50 value of 0.276 (0.1526–0.6753) mg/kg (Figure 5H).
Figure 5. The antiemetic eects of the α2-adrenergic receptor agonist dexmedetomidine against
vomiting evoked by diverse receptor-selective emetogens. Varying doses of dexmedetomidine (0,
0.01, 0.05, and 0.1 mg/kg, i.p.) were injected to dierent groups of shrews 30 min prior t o an injection
of a fully eective emetic dose of the selective serotonin 5-HT3 receptor agonist 2-Methyl-5-HT (5
mg/kg, i.p., n = 8–10 shrews per group) (A,B), the selective neurokinin NK1 receptor agonist GR73632
(5 mg/kg, i.p., n = 7–9 shrews per group) (C,D), the muscarinic M1 receptor agonist McN-A-343 (2
mg/kg, i.p., n = 7–8 shrews per group) (E,F), the dopamine D2/3 receptor preferring agonist quinpirole
(2 mg/kg, i.p., n = 9–11 shrews per group) (G,H), or the cannabinoid CB1 receptor-selective inverse
agonist/antagonist SR141716A (20 mg/kg, i.p., n = 8–9 shrews per group) (I,J). Emetic parameters
were recorded for the next 30 min. The frequency of emesis was analyzed with Kruskal–Wallis non-
parametric one-way ANOVA followed by Dunn’s post hoc test and presented as mean ± SEM
(A,C,E,G,I). The percentage of shrews vomiting was analyzed with chi-square test and presented as
the mean (B,D,F,H,J). * p < 0.05, ** p < 0.01 vs. 0 mg/kg (controls pretreated with vehicle of dexme-
detomidine). The number of animals in each group is presented on the top of the corresponding
column.
We also investigated the antiemetic potential of clonidine and dexmedetomidine
against vomiting evoked by the CB1 receptor inverse agonist/antagonist SR141716A (20
mg/kg, i.p.). As shown in Figure 4I, clonidine (0, 0.1, 1, 5, and 10 mg/kg, n = 8–10 per group)
caused a dose-dependent decrease in the frequency of SR141716A-evoked vomits (KW (4,
41) = 23.22; p = 0.0001) with signicant reductions occurring at its 1 mg/kg (p = 0.0244), 5
Figure 5. The antiemetic effects of the
α2
-adrenergic receptor agonist dexmedetomidine against
vomiting evoked by diverse receptor-selective emetogens. Varying doses of dexmedetomidine (0,
0.01, 0.05, and 0.1 mg/kg, i.p.) were injected to different groups of shrews 30 min prior to an injection
of a fully effective emetic dose of the selective serotonin 5-HT
3
receptor agonist 2-Methyl-5-HT
(5 mg/kg, i.p., n= 8–10 shrews per group) (A,B), the selective neurokinin NK
1
receptor agonist
GR73632 (5 mg/kg, i.p., n= 7–9 shrews per group) (C,D), the muscarinic M
1
receptor agonist
McN-A-343 (2 mg/kg, i.p., n= 7–8 shrews per group) (E,F), the dopamine D
2/3
receptor preferring
agonist quinpirole (2 mg/kg, i.p., n= 9–11 shrews per group) (G,H), or the cannabinoid CB
1
receptor-
selective inverse agonist/antagonist SR141716A (20 mg/kg, i.p., n= 8–9 shrews per group) (I,J).
Emetic parameters were recorded for the next 30 min. The frequency of emesis was analyzed with
Kruskal–Wallis non-parametric one-way ANOVA followed by Dunn’s post hoc test and presented
as mean
±
SEM (A,C,E,G,I). The percentage of shrews vomiting was analyzed with chi-square test
and presented as the mean (B,D,F,H,J). * p< 0.05, ** p< 0.01 vs. 0 mg/kg (controls pretreated with
vehicle of dexmedetomidine). The number of animals in each group is presented on the top of the
corresponding column.
We also investigated the antiemetic potential of clonidine and dexmedetomidine
against vomiting evoked by the CB
1
receptor inverse agonist/antagonist SR141716A
(20 mg/kg, i.p.). As shown in Figure 4I, clonidine (0, 0.1, 1, 5, and 10 mg/kg,
n= 8–10
per group) caused a dose-dependent decrease in the frequency of SR141716A-evoked
Int. J. Mol. Sci. 2024,25, 4603 10 of 28
vomits (KW (4, 41) = 23.22; p= 0.0001) with significant reductions occurring at its 1 mg/kg
(p= 0.0244), 5 mg/kg (p= 0.0022), and 10 mg/kg (p< 0.0001) doses, and with an ID
50
value of 0.0875 (0.02819–0.2546) mg/kg. The chi-square test indicates that the number
of shrews vomiting in response to SR141716A was also significantly attenuated by cloni-
dine (
χ2
(4, 41) = 19.1; p= 0.0008) with 40% protection at its 0.1 mg/kg (p= 0.0253) and
1 mg/kg (p= 0.0253) doses, 62.5% protection at 5 mg/kg (p= 0.0033), and complete protec-
tion at 10 mg/kg (p< 0.0001) with an ID
50
value of 1.291 (0.3871–3.31) mg/kg (Figure 4J).
Figure 5I,J show that dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg, n= 8–9 per group) also
dose-dependently suppressed vomiting caused by SR141716A (20 mg/kg, i.p.). The mean
frequency of SR141716A-induced emesis was significantly reduced (KW
(3, 29) = 13.21
;
p= 0.0042) at 0.1 mg/kg (p= 0.004) with an ID
50
value of 0.02453 (0.007966–0.06693) mg/kg
(Figure 5I). The percentage of shrews vomiting (
χ2
(3, 29) = 17.81; p= 0.0005) was signifi-
cantly reduced at its 0.05 mg/kg (62.5%, p= 0.007)- and 0.1 mg/kg (75%, p= 0.0019) doses
with an ID50 value of 0.04303 (0.02993–0.08087) mg/kg (Figure 5J).
2.5. The Antiemetic Potential of the α2-Adrenergic Receptor Agonists Clonidine and
Dexmedetomidine against Vomiting Evoked by Ca2+ Channel Regulators
We then investigated the antiemetic effect of clonidine and dexmedetomidine against
vomiting evoked by Ca
2+
channel regulators, such as the LTCC agonist FPL64176 (
10 mg/kg,
i.p.) and the SERCA inhibitor thapsigargin (0.5 mg/kg, i.p.) (Figures 6and 7).
Pretreatment with clonidine (0, 0.1, 1, 5, and 10 mg/kg, n= 6–9 per group) significantly
attenuated the mean frequency of FPL64176-induced vomiting in a dose-dependent manner
(KW (4, 34) = 19.15; p= 0.0007) with significant reductions occurring at its 5 mg/kg
(p= 0.01) and 10 mg/kg (p= 0.0008; ID
50
value: 0.404 (0.1078–1.332) mg/kg; Figure 6A)
doses. In addition, the percentage of shrews vomiting in response to FPL64176 was
also suppressed by clonidine (
χ2
(4, 34) = 18.34; p= 0.0011) at its 5 mg/kg dose (66.67%,
p= 0.0042) and completely protected at its 10 mg/kg dose (100%, p= 0.0001) with ID
50
value
of 1.661 (0.6772–3.796) mg/kg (Figure 6B). Dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg,
n= 7–11 per group) also dose-dependently suppressed vomiting caused by FPL64176 (KW
(3, 35) = 16.43; p= 0.0009) with significant reductions occurring at its 0.05 (p= 0.0028) and
0.1 mg/kg (p= 0.0009) doses, with an ID
50
value of 0.006993 (0.001346–0.02088) mg/kg
(Figure 7A). Likewise, significant decreases (
χ2
(3, 35) = 11.46; p= 0.0095) in the percentage
of animals vomiting were also noted at its 0.05 (54.55%; p= 0.0057) and 0.1 mg/kg (71.43%;
p= 0.0015) doses with an ID50 value of 0.03667 (0.01645–0.0796) mg/kg (Figure 7B).
Figure 6C,D show that clonidine (0, 0.1, 1, 5, and 10 mg/kg, n= 7–9 per group) dose-
dependently suppressed vomiting caused by thapsigargin (0.5 mg/kg, i.p.). The frequency
of thapsigargin-induced emesis (KW (4, 36) = 25.05; p< 0.0001) was significantly reduced
at 1 mg/kg (p= 0.0457), 5 mg/kg (p= 0.0013), and 10 mg/kg (p= 0.0003) doses, with
an ID
50
value of 0.1488 (0.04487–0.4809) mg/kg (Figure 6C). Significant decreases in the
percentage of animals vomiting (
χ2
(4, 36) = 16.38; p= 0.0026) occurred at its 5 mg/kg
(55.56%; p= 0.0121) and 10 mg/kg (66.67%; p= 0.0041) doses with an ID
50
value of 4.691
(2.37–9.161) mg/kg (Figure 6D). As shown in Figure 7C,D, dexmedetomidine (0, 0.01, 0.05,
and 0.1 mg/kg, n= 8–9 per group) also caused a dose-dependent decrease in the mean
frequency of vomits (KW (3, 30) = 8.296; p= 0.0403) induced by the thapsigargin (0.5 mg/kg,
i.p.) with a significant reduction at its 0.1 mg/kg dose (p= 0.0135; ID
50
value: 0.05317
(0.017–0.2617) mg/kg; Figure 7C), but no significant decrease (
χ2
(3, 30) = 5.542; p= 0.1362)
in the percentage of animals vomiting was observed (ID
50
value: 0.1372 (0.06515–0.363)
mg/kg; Figure 7D).
Int. J. Mol. Sci. 2024,25, 4603 11 of 28
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 10 of 28
mg/kg (p = 0.0022), and 10 mg/kg (p < 0.0001) doses, and with an ID50 value of 0.0875
(0.02819–0.2546) mg/kg. The chi-square test indicates that the number of shrews vomiting
in response to SR141716A was also signicantly aenuated by clonidine (χ2 (4, 41) = 19.1;
p = 0.0008) with 40% protection at its 0.1 mg/kg (p = 0.0253) and 1 mg/kg (p = 0.0253) doses,
62.5% protection at 5 mg/kg (p = 0.0033), and complete protection at 10 mg/kg (p < 0.0001)
with an ID50 value of 1.291 (0.3871–3.31) mg/kg (Figure 4J). Figure 5I,J show that dexme-
detomidine (0, 0.01, 0.05, and 0.1 mg/kg, n = 8–9 per group) also dose-dependently sup-
pressed vomiting caused by SR141716A (20 mg/kg, i.p.). The mean frequency of
SR141716A-induced emesis was signicantly reduced (KW (3, 29) = 13.21; p = 0.0042) at
0.1 mg/kg (p = 0.004) with an ID50 value of 0.02453 (0.007966–0.06693) mg/kg (Figure 5I).
The percentage of shrews vomiting (χ2 (3, 29) = 17.81; p = 0.0005) was signicantly reduced
at its 0.05 mg/kg (62.5%, p = 0.007)- and 0.1 mg/kg (75%, p = 0.0019) doses with an ID50
value of 0.04303 (0.02993–0.08087) mg/kg (Figure 5J).
2.5. The Antiemetic Potential of the α2-Adrenergic Receptor Agonists Clonidine and
Dexmedetomidine against Vomiting Evoked by Ca2+ Channel Regulators
We then investigated the antiemetic eect of clonidine and dexmedetomidine against
vomiting evoked by Ca2+ channel regulators, such as the LTCC agonist FPL64176 (10
mg/kg, i.p.) and the SERCA inhibitor thapsigargin (0.5 mg/kg, i.p.) (Figures 6 and 7).
Figure 6. The antiemetic eects of the α2-adrenergic receptor agonist clonidine against vomiting
caused by Ca2+ channel regulators, the LTCC agonist FPL64176 and the SERCA inhibitor thapsigar-
gin. Varying doses of clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p.) were injected to dierent groups of
shrews 30 min prior to an injection of a fully eective emetic dose of FPL64176 (10 mg/kg, i.p., n =
Figure 6. The antiemetic effects of the
α2
-adrenergic receptor agonist clonidine against vomiting caused
by Ca
2+
channel regulators, the LTCC agonist FPL64176 and the SERCA inhibitor thapsigargin. Varying
doses of clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p.) were injected to different groups of shrews 30 min
prior to an injection of a fully effective emetic dose of FPL64176 (10 mg/kg, i.p., n= 6–9 per group)
(A,B) or thapsigargin (0.5 mg/kg, i.p., n= 7–9 per group) (C,D). Emetic parameters were recorded
for the next 30 min. The frequency of emesis was analyzed with Kruskal–Wallis non-parametric one-
way ANOVA followed by Dunn’s post hoc test and presented as mean
±
SEM (A,C). The percentage
of shrews vomiting was analyzed with chi-square test and presented as the mean (B,D). * p< 0.05,
** p< 0.01, *** p< 0.001 vs. 0 mg/kg (controls pretreated with vehicle of clonidine). The number of
animals in each group is presented on the top of the corresponding column.
Int. J. Mol. Sci. 2024,25, 4603 12 of 28
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 11 of 28
6–9 per group) (A,B) or thapsigargin (0.5 mg/kg, i.p., n = 7–9 per group) (C,D). Emetic parameters
were recorded for the next 30 min. The frequency of emesis was analyzed with Kruskal–Wallis non-
parametric one-way ANOVA followed by Dunn’s post hoc test and presented as mean ± SEM (A,
C). The percentage of shrews vomiting was analyzed with chi-square test and presented as the mean
(B, D). * p < 0.05, ** p < 0.01, *** p < 0.001 vs. 0 mg/kg (controls pretreated with vehicle of clonidine).
The number of animals in each group is presented on the top of the corresponding column.
Figure 7. The antiemetic eects of the α2-adrenergic receptor agonist dexmedetomidine against
vomiting caused by Ca2+ channel regulators, the LTCC agonist FPL64176 and the SERCA inhibitor
thapsigargin. Varying doses of dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg, i.p.) were injected to
dierent groups of shrews 30 min prior to an injection of a fully eective emetic dose of FPL64176
(10 mg/kg, i.p., n = 7–11 per group) (A,B) or thapsigargin (0.5 mg/kg, i.p., n = 8–9 per group) (C,D).
Emetic parameters were recorded for the next 30 min. The frequency of emesis was analyzed with
Kruskal–Wallis non-parametric one-way ANOVA followed by Dunn’s post hoc test and presented
as mean ± SEM (A,C). The percentage of shrews vomiting was analyzed with chi-square test and
presented as the mean (B,D). * p < 0.05, ** p < 0.01, *** p < 0.001 vs. 0 mg/kg (controls pretreated with
vehicle of dexmedetomidine). The number of animals in each group is presented on the top of the
corresponding column.
Pretreatment with clonidine (0, 0.1, 1, 5, and 10 mg/kg, n = 6–9 per group) signicantly
aenuated the mean frequency of FPL64176-induced vomiting in a dose-dependent man-
ner (KW (4, 34) = 19.15; p = 0.0007) with signicant reductions occurring at its 5 mg/kg (p
= 0.01) and 10 mg/kg (p = 0.0008; ID50 value: 0.404 (0.1078–1.332) mg/kg; Figure 6A) doses.
In addition, the percentage of shrews vomiting in response to FPL64176 was also
Figure 7. The antiemetic effects of the
α2
-adrenergic receptor agonist dexmedetomidine against
vomiting caused by Ca
2+
channel regulators, the LTCC agonist FPL64176 and the SERCA inhibitor
thapsigargin. Varying doses of dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg, i.p.) were injected to
different groups of shrews 30 min prior to an injection of a fully effective emetic dose of FPL64176
(10 mg/kg, i.p., n= 7–11 per group) (A,B) or thapsigargin (0.5 mg/kg, i.p., n= 8–9 per group) (C,D).
Emetic parameters were recorded for the next 30 min. The frequency of emesis was analyzed with
Kruskal–Wallis non-parametric one-way ANOVA followed by Dunn’s post hoc test and presented
as mean
±
SEM (A,C). The percentage of shrews vomiting was analyzed with chi-square test and
presented as the mean (B,D). * p< 0.05, ** p< 0.01, *** p< 0.001 vs. 0 mg/kg (controls pretreated with
vehicle of dexmedetomidine). The number of animals in each group is presented on the top of the
corresponding column.
2.6. The Antiemetic Potential of Clonidine and Dexmedetomidine against Vomiting Evoked
by Rolipram, the Inhibitor of PDE4 in Shrews
In the ferret, clonidine was found to prevent emesis induced by PDE4 inhibitors,
such as PMNPQ (i.e., 6-(4-pyridylmethyl)-8-(3-nitrophenyl) quinoline, rolipram, and CT
2450 (i.e., (R)-N-{4-[1-(3-cyclopentyloxy-4-methoxyphenyl)-2-(4- pyridyl)ethyl]phenyl}N9-
ethylurea) [
30
]. In the present study, we explored the antiemetic potential of the
α2
-
adrenergic receptor agonists clonidine and dexmedetomidine against rolipram (1 mg/kg,
i.p.)-induced vomiting in shrews [42].
As shown in Figure 8A, clonidine (0, 0.1, 1, 5, and 10 mg/kg, n= 8–10 per group) caused
dose-dependent decreases in the frequency of rolipram-evoked vomits ((KW (4, 41) = 10.9;
p= 0.0277) with significant reductions occurring at its 5 mg/kg (p= 0.0332) and 10 mg/kg
(p= 0.0182) doses, with an ID
50
value of 1.024 (0.1497–4.052) mg/kg. The percentage of
animals vomiting in response to rolipram (
χ2
(4, 41) = 11.21; p= 0.0243) was also significantly
reduced at its 0.1 mg/kg (40%, p= 0.0253), 1 mg/kg (60%, p= 0.0034), 5 mg/kg (62.5%,
p= 0.0033) and 10 mg/kg (62.5%, p= 0.0033) doses with an ID
50
value of 0.7897
(0.1143–3.64)
Int. J. Mol. Sci. 2024,25, 4603 13 of 28
mg/kg (Figure 8B). Dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg, n= 10 per group)
also dose-dependently suppressed the mean vomiting frequency evoked by rolipram (KW
(3, 36) = 10.7; p= 0.0135) with significant reductions at its 0.05 mg/kg (p= 0.0305) and
0.1 mg/kg (p= 0.0084) doses, and with an ID
50
value of 0.01748 (0.004243–0.05612) mg/kg
(Figure 8C). Significant decreases (
χ2
(3, 36) = 9.167; p= 0.0272) in the percentage of animals
vomiting were also noted at its 0.01 mg/kg (50%; p= 0.0098), 0.05 mg/kg (50%; p= 0.0098)
and 0.1 mg/kg (60%; p= 0.0034) doses with an ID
50
value of 0.03517 (0.011–0.09391) mg/kg
(Figure 8D).
Figure 8. The antiemetic effects of the
α2
-adrenergic receptor agonists clonidine and dexmedetomidine
against vomiting caused by the PDE4 inhibitor rolipram (1 mg/kg, i.p.). Varying doses of clonidine (0,
0.1, 1, 5, and 10 mg/kg, i.p., n= 8–10 per group; (A,B)) or dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg,
i.p., n= 10 per group; (C,D)) were injected to different groups of shrews 30 min prior to an injection
of a fully effective emetic dose of PDE4 inhibitor rolipram (1 mg/kg, i.p.). Emetic parameters were
recorded for the next 30 min. The frequency of emesis was analyzed with Kruskal–Wallis non-parametric
one-way ANOVA followed by Dunn’s post hoc test and presented as mean
±
SEM (A,C). The percentage
of shrews vomiting was analyzed with chi-square test and presented as the mean (B,D). * p< 0.05,
** p< 0.01 vs. 0 mg/kg (controls pretreated with vehicle of clonidine or dexmedetomidine). The number
of animals in each group is presented on the top of the corresponding column.
2.7. The Antiemetic Effect of Clonidine and Dexmedetomidine against Vomiting Evoked by the
HCN Channel Blocker ZD7288
Our previous study has shown that the HCN channel blocker ZD7288 induces vomit-
ing in a dose-dependent manner, with maximal efficacy of 100% at 1 mg/kg (i.p.) [
43
]. In
the current study, the antiemetic potential of clonidine and dexmedetomidine on ZD7288
(1 mg/kg, i.p.)-induced vomiting was also examined (Figure 9). Pretreatment with clonidine
(0, 0.1, 1, 5, and 10 mg/kg, n= 10 per group) significantly attenuated the mean frequency
of ZD7288-induced vomiting in a dose-dependent manner (KW (4, 40) = 29.08; p< 0.0001)
Int. J. Mol. Sci. 2024,25, 4603 14 of 28
with significant reductions occurring at its 5 mg/kg (p= 0.0003) and 10 mg/kg (p< 0.0001;
ID
50
value: 0.4023 (0.1636–0.8948) mg/kg; Figure 9A) doses. Significant decreases in the
percentage of animals vomiting (
χ2
(4, 40) = 17.22; p= 0.0017) also occurred at its 5 mg/kg
(44.44%; p= 0.0233) and 10 mg/kg (55.56%; p= 0.0085) doses with an ID
50
value of 7.819
(4.363–14.69) mg/kg (Figure 9B). Administration of dexmedetomidine (0, 0.01, 0.05, and
0.1 mg/kg, n= 7–10 per group) also suppressed the mean frequency of vomiting evoked by
ZD7288 (KW (3, 30) = 8.11; p= 0.0438) with a significant reduction at its 0.1 mg/kg dose
(p= 0.0155), and with an ID
50
value of 0.08931 (0.03732–0.2473) mg/kg (Figure 9C). How-
ever, dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg) failed to significantly protect shrews
from vomiting (χ2(3, 30) = 3.974; p= 0.2643; Figure 9D).
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 14 of 28
value of 7.819 (4.363–14.69) mg/kg (Figure 9B). Administration of dexmedetomidine (0,
0.01, 0.05, and 0.1 mg/kg, n = 7–10 per group) also suppressed the mean frequency of vom-
iting evoked by ZD7288 (KW (3, 30) = 8.11; p = 0.0438) with a signicant reduction at its
0.1 mg/kg dose (p = 0.0155), and with an ID50 value of 0.08931 (0.03732–0.2473) mg/kg (Fig-
ure 9C). However, dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg) failed to signicantly
protect shrews from vomiting (χ2 (3, 30) = 3.974; p = 0.2643; Figure 9D).
Figure 9. The antiemetic eects of the α2-adrenergic receptor agonists clonidine and dexmedetomi-
dine against vomiting caused by the HCN channel blocker ZD7288 (1 mg/kg, i.p.). Varying doses of
clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p., n = 9 per group; (A,B)) or dexmedetomidine (0, 0.01, 0.05,
and 0.1 mg/kg, i.p., n = 7–10 per group; (C,D)) were injected to dierent groups of shrews 30 min
prior to an injection of a fully eective emetic dose of ZD7288 (1 mg/kg, i.p.). Emetic parameters
were recorded for the next 30 min. The frequency of emesis was analyzed with Kruskal–Wallis non-
parametric one-way AN OVA followed by Dunns post hoc test and presented as mean ± SEM (A,C).
The percentage of shrews vomiting was analyzed with chi-square test and presented as the mean
(B,D). * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001 vs. 0 mg/kg (controls pretreated with vehicle
of clonidine or dexmedetomidine). The number of animals in each group is presented on the top of
the corresponding column.
2.8. Open-Field Locomotor Studies
Figure 10 shows the eects of corresponding vehicle, varying doses of clonidine (0.01,
0.1, 1, 5, and 10 mg/kg, i.p.), and dexmedetomidine (0.01, 0.05, 0.1, and 0.5 mg/kg, i.p.) on
the locomotor activities of shrews using the open-eld locomotor test. A one-way ANOVA
analysis demonstrated that clonidine signicantly decreased (F 5, 39 = 5.255; p = 0.0009) the
Figure 9. The antiemetic effects of the
α2
-adrenergic receptor agonists clonidine and dexmedetomi-
dine against vomiting caused by the HCN channel blocker ZD7288 (1 mg/kg, i.p.). Varying doses of
clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p., n= 9 per group; (A,B)) or dexmedetomidine (0, 0.01, 0.05,
and 0.1 mg/kg, i.p., n= 7–10 per group; (C,D)) were injected to different groups of shrews 30 min
prior to an injection of a fully effective emetic dose of ZD7288 (1 mg/kg, i.p.). Emetic parameters
were recorded for the next 30 min. The frequency of emesis was analyzed with Kruskal–Wallis
non-parametric one-way ANOVA followed by Dunn’s post hoc test and presented as mean
±
SEM
(A,C). The percentage of shrews vomiting was analyzed with chi-square test and presented as the
mean (B,D). * p< 0.05, ** p< 0.01, *** p< 0.001, **** p< 0.0001 vs. 0 mg/kg (controls pretreated with
vehicle of clonidine or dexmedetomidine). The number of animals in each group is presented on the
top of the corresponding column.
2.8. Open-Field Locomotor Studies
Figure 10 shows the effects of corresponding vehicle, varying doses of clonidine (0.01,
0.1, 1, 5, and 10 mg/kg, i.p.), and dexmedetomidine (0.01, 0.05, 0.1, and 0.5 mg/kg, i.p.) on
the locomotor activities of shrews using the open-field locomotor test. A one-way ANOVA
analysis demonstrated that clonidine significantly decreased (F
5, 39
= 5.255; p= 0.0009)
Int. J. Mol. Sci. 2024,25, 4603 15 of 28
the total distance moved by the shrews at its 1 mg/kg (p= 0.0015), 5 mg/kg (p= 0.0225),
and 10 mg/kg (p= 0.0114) doses in the 30 min observation period (Figure 10A). Like-
wise, the frequencies of rearing behavior were also significantly attenuated (F
5, 39
= 5.223;
p= 0.0009) by clonidine at its 1 mg/kg (p= 0.0299), 5 mg/kg (p= 0.0013), and
10 mg/kg
(p= 0.0014; Figure 10B) doses. Relative to the corresponding vehicle-pretreated control
group, dexmedetomidine only significantly decreased the total distance moved
(F
4, 32
= 6.877; p= 0.0004) and the frequency of rearing behavior (F
4, 32
= 4.758;
p= 0.004) at its 0.5 mg/kg dose (p= 0.0002 for distance moved, Figure 10C; p= 0.0009 for
rearing frequency, Figure 10D), which is larger than its antiemetic doses.
Int. J. Mol. Sci. 2024, 25, x FOR PEER REVIEW 15 of 28
total distance moved by the shrews at its 1 mg/kg (p = 0.0015), 5 mg/kg (p = 0.0225), and
10 mg/kg (p = 0.0114) doses in the 30 min observation period (Figure 10A). Likewise, the
frequencies of rearing behavior were also signicantly aenuated (F 5, 39 = 5.223; p = 0.0009)
by clonidine at its 1 mg/kg (p = 0.0299), 5 mg/kg (p = 0.0013), and 10 mg/kg (p = 0.0014;
Figure 10B) doses. Relative to the corresponding vehicle-pretreated control group, dex-
medetomidine only signicantly decreased the total distance moved (F 4, 32 = 6.877; p =
0.0004) and the frequency of rearing behavior (F 4, 32 = 4.758; p = 0.004) at its 0.5 mg/kg dose
(p = 0.0002 for distance moved, Figure 10C; p = 0.0009 for rearing frequency, Figure 10D),
which is larger than its antiemetic doses.
Figure 10. The eect of the α2-adrenergic receptor agonists clonidine and dexmedetomidine on
open-eld locomotor behaviors. Dierent groups of shrews were injected with either vehicle or var-
ying doses of clonidine (0.1, 1, 5, and 10 mg/kg, i.p., n = 6–10; (A,B))/dexmedetomidine (0.01, 0.05,
and 0.1 mg/kg, i.p., n = 5–10; (C,D)) at 0 min. Thirty minutes later, each shrew was individually
placed in a white observation cage (27.5 × 27.5 × 28 cm), and the locomotor activity [distance moved
(A,C) and rearing frequency (B,D)] were recorded for 30 min by a computerized video tracking,
motion analysis, and behavior recognition system (Ethovision, version XT 9). The dose-response
eects of clonidine and dexmedetomidine on dierent groups of least shrews were analyzed using
the Noldus software. Signicant dierence relative to the corresponding vehicle group is indicated
as * p < 0.05, ** p < 0.01, *** p < 0.001. The number of animals in each group is presented on the top
of the corresponding column.
3. Discussion Software
3.1. Signicance of the Present Study
Per the Introduction Section, currently there is signicant confusion in the published
literature on the ability of α2-adrenergic receptor ligands evoking emetic/antiemetic ef-
fects, which are probably due to: (i) species variation in the array of animal models (e.g.,
cats, dogs, ferrets, pigeons, least shrews, humans) employed for emesis studies in dierent
laboratories, (ii) lack of published comprehensive studies investigating the emetic/antie-
metic potential of α2-adrenergic receptor agonists and antagonists in a given laboratory
using one emesis model, and (iii) full pharmacological assessment of α2-adrenergic
Figure 10. The effect of the
α2
-adrenergic receptor agonists clonidine and dexmedetomidine on
open-field locomotor behaviors. Different groups of shrews were injected with either vehicle or
varying doses of clonidine (0.1, 1, 5, and 10 mg/kg, i.p., n= 6–10; (A,B))/dexmedetomidine (0.01,
0.05, and 0.1 mg/kg, i.p., n= 5–10; (C,D)) at 0 min. Thirty minutes later, each shrew was individually
placed in a white observation cage (27.5
×
27.5
×
28 cm), and the locomotor activity [distance moved
(A,C) and rearing frequency (B,D)] were recorded for 30 min by a computerized video tracking,
motion analysis, and behavior recognition system (Ethovision, version XT 9). The dose-response
effects of clonidine and dexmedetomidine on different groups of least shrews were analyzed using
the Noldus software. Significant difference relative to the corresponding vehicle group is indicated as
*p< 0.05, ** p< 0.01, *** p< 0.001. The number of animals in each group is presented on the top of the
corresponding column.
3. Discussion
3.1. Significance of the Present Study
Per the Introduction Section, currently there is significant confusion in the published
literature on the ability of
α2
-adrenergic receptor ligands evoking emetic/antiemetic effects,
which are probably due to: (i) species variation in the array of animal models (e.g., cats,
dogs, ferrets, pigeons, least shrews, humans) employed for emesis studies in different labo-
ratories, (ii) lack of published comprehensive studies investigating the emetic/antiemetic
potential of
α2
-adrenergic receptor agonists and antagonists in a given laboratory using one
emesis model, and (iii) full pharmacological assessment of
α2
-adrenergic receptor ligands
against diverse emetogens. In the present study, we investigated the emetic/antiemetic
Int. J. Mol. Sci. 2024,25, 4603 16 of 28
potential of two
α2
-adrenergic receptor agonists (clonidine and dexmedetomidine), as
well as an
α2
-adrenergic receptor antagonist yohimbine, in the least shrew animal model
of vomiting. Only yohimbine increased both the mean frequency of emesis and the per-
centage of shrews vomiting. The evoked vomiting was bell-shaped and occurred in a
dose-dependent manner with a maximal efficacy of 1 mg/kg. However, only 61.5% of
least shrews vomited at the 1 mg/kg dose, whereas larger doses of yohimbine were less
efficacious. Our immunohistochemical findings demonstrated that yohimbine (1 mg/kg,
i.p.) causes significant increases in both c-fos expression and release of 5-HT and SP in the
shrew brainstem DVC emetic nuclei. Furthermore, both
α2
-adrenergic receptor agonists
not only suppressed yohimbine-induced vomiting in a dose-dependent manner, but also
emesis evoked by: (1) selective agonists of diverse emetic receptors including serotonin
5-HT
3
(2-Methyl-5-HT), SP neurokinin NK
1
(GR73632), muscarinic M
1
(MCN-A-343), and
dopamine D
2/3
(quinpirole); (2) the selective CB
1
receptor inverse agonist/antagonist
(SR141716A); (3) Ca
2+
channel modulators including the LTCC agonist FPL64176 and the
SERCA inhibitor thapsigargin; (4) the PDE4 inhibitor rolipram; and 5) the HCN channel
blocker ZD7288. Clonidine at antiemetic doses (1, 5, and 10 mg/kg, i.p.) decreased the
locomotor activity of shrews in the open-field test, whereas dexmedetomidine did not
influence shrew locomotion at its tested antiemetic doses.
3.2. Emetic Effect of Yohimbine in the Least Shrew
Existing experimental evidence suggests a role for
α2
-adrenergic receptor in emesis.
The
α2
-adrenergic receptor agonist clonidine has been shown to evoke vomiting in a
dose-dependent fashion in both cats and dogs in an
α2
-adrenergic receptor antagonist
(yohimbine)-sensitive manner [
19
21
]. However, clonidine was found to prevent vomiting
caused by either rolipram (an inhibitor of PDE4) in ferrets [
30
], or reserpine (a monoamine
releaser) in pigeons [
31
]. Moreover, yohimbine not only induced dose-dependent emesis in
both ferrets and pigeons, but also potentiated reserpine-induced vomiting and reversed
the inhibitory effect of clonidine against reserpine-evoked vomiting [
30
,
31
]. However, the
discussed emetic studies in pigeons and ferrets were limited in scope since yohimbine’s
full dose-response emetic effects were not evaluated, but 100% of both species vomited
in response to an i.p. injection of its 0.5 or 3 mg/kg doses, respectively [
30
,
31
]. In the
present study, yohimbine evoked vomiting in a dose-dependent, but bell-shaped manner,
with a maximal efficacy of 0.85
±
0.22 vomits at 1 mg/kg dose in 61.54% of tested shrews,
whereas its larger doses were less efficacious. A similar bell-shaped dose-response effect
has also been observed in aggression studies where smaller doses of yohimbine increased,
and larger doses decreased aggressive behavior in rats [
46
]. The bell-shaped dose-response
effects may be due to the non-selective nature of yohimbine since it does not exclusively
bind to
α2
-adrenergic receptors as it also (i) has moderate to weak affinity for dopamine D
2
-,
adrenergic
α1
-, and serotonergic 5-HT
1A
receptors [
47
49
], and (ii) can release monoamines
(NE, DA, and 5-HT) both in the periphery and the CNS [50,51].
3.3. Yohimbine-Induced Expression of c-fos and Release of 5-HT and SP in the DVC Central
Emetic Loci
Induction of c-fos immunoreactivity is an indirect but classical tool to evaluate neu-
ronal activation following peripheral agonist administration [
52
]. Our lab has also utilized
c-fos induction in the least shrew brainstem DVC emetic nuclei to demonstrate central
responsiveness to peripheral administration of diverse emetogens [
41
,
53
]. In the current
study, relative to vehicle injection, vomiting induced by yohimbine (i.p.) was followed by
increased expression of c-fos in all the DVC emetic nuclei (the AP, NTS, and DMNV) in
the shrew brainstem. Likewise, we have previously shown that the nonspecific emetogen
cisplatin and the more selective 5-HT
3
receptor agonist 2-Methyl-5-HT can evoke vomiting
in least shrews and c-fos expression in their corresponding DVC emetic nuclei AP, NTS, and
DMNV [
53
]. Such a pattern of c-fos expression in the DVC emetic nuclei is not surprising
since the AP, NTS, and DMNX are populated by neurons containing a number of emetic
Int. J. Mol. Sci. 2024,25, 4603 17 of 28
neurotransmitters (e.g., 5-HT, DA) and corresponding receptors, and systemic administra-
tion of yohimbine promotes their release both in the periphery and brainstem [
50
,
51
]. In
fact, prior depletion of emetic monoamines with reserpine can completely prevent vomiting
caused by yohimbine or reserpine in pigeons, suggesting both yohimbine and reserpine
may induce emesis by releasing monoamines, albeit via different mechanisms [31].
5-HT is an important gastrointestinal signaling molecule. In the CNS it regulates
appetite and emesis, and in the gastrointestinal tract 5-HT is an important mediator of
sensation (e.g., nausea and emesis) between the intestine and the brainstem [
54
]. It can
evoke vomiting via peripheral stimulation of 5-HT
3
receptors in least shrews, whereas
its brain penetrant analog 2-Methyl-5-HT involves both central and peripheral compo-
nents
[3,55]
. We have previously demonstrated that the LTCC agonist FPL64176-induced
emesis was accompanied by an increase in 5-HT immunoreactivity in the dorsomedial
subdivision of the NTS [
45
]. In the present study, compared to vehicle pretreatment, a 15
min exposure to yohimbine resulted in a moderate enhancement of 5-HT immunoreac-
tivity in the AP, NTS, and DMNX. Indeed, yohimbine has been shown to release emetic
monoamines, including 5-HT, via blockade of presynaptic
α2
-adrenergic receptors [
55
59
].
In the current study, the yohimbine-evoked increases in 5-HT-positive fibers were most
abundant in the least shrew dorsomedial area of the NTS, followed by smaller increases in
the adjacent subnuclei of the NTS and DMNX, but the AP contained fewer 5-HT-containg
neurons. In the current study, yohimbine also evoked an increase in SP immunoreactivity in
the DMNX at 15 min post treatment, which is not surprising, since release of stimuli-evoked
SP in the rat or rabbit spinal dorsal horn can be modulated by yohimbine [
60
,
61
]. The
current finding agrees with our published study which demonstrated that a 15–30 min
thapsigargin exposure (0.5 mg/kg, i.p.), can increase the SP tissue content up to ~3 times
over the basal level in the least shrew DMNX [
41
]. The DMNX is known to send appropriate
output to the gastrointestinal tract to alter gastric motility [
53
,
62
]. Thus, yohimbine-evoked
enhancements in 5-HT and SP immunoreactivity in the DVC suggests that both 5-HT and
SP are likely to be involved in the evoked emesis.
3.4. Effects of α2-Adrenergic Receptor Agonists Clonidine and Dexmedetomidine
on Locomotor Activity
A major concern regarding
α2
-adrenergic receptor agonists is the possibility of se-
dation [
63
]. Although the affinity of dexmedetomidine is eight times greater than cloni-
dine [
64
], similar low doses of both
α2
-adrenergic agonists (0.03–0.1 mg/kg, i.p.) reduce
spontaneous locomotor activity in rodents [
65
68
]. Thus, we examined the motor suppres-
sive effects of both clonidine and dexmedetomidine. Clonidine at 0.01 and 0.1 mg/kg doses
did not significantly affect either spontaneous locomotor activity (i.e., distance moved) or
the frequency of rearing behavior in least shrews, but significantly decreased both behaviors
at its antiemetic doses (1, 5, and 10 mg/kg) in the 30 min observation period. This should
not be surprising since, unlike rodents, least shrews are constantly on the move and rarely
remain motionless. Some clinical findings suggest that although sedation is a well-known
side-effect of clonidine [69], it does not appear to be related to its antiemetic property. For
example, Kumar et al. [
70
] have shown that in elderly patients undergoing intraocular
surgery, clonidine had no effect on emesis despite the fact that it induced more seda-
tion. In contrast to clonidine, the tested antiemetic doses of dexmedetomidine (0.01, 0.05,
and 0.1 mg/kg) in the current study did not significantly decrease either shrew motor
parameters. Overall, our data provide evidence for the safe use of dexmedetomidine as an
antiemetic drug at human doses equivalent to less than 0.1 mg/kg in least shrews.
3.5. Antiemetic Effects of α2-Adrenergic Receptor Agonists Clonidine and Dexmedetomidine
against Yohimbine and Diverse Emetogens
In contrast to cats and dogs (see introduction), both the non-selective (clonidine)
and the more selective
α2
-adrenergic receptor agonist dexmedetomidine failed to trigger
vomiting in shrews even at doses 333–400 times greater than their corresponding ED
50
values reported in dogs (clonidine: 25
µ
g/kg, i.m.; dexmedetomidine: 0.003 mg/kg,
Int. J. Mol. Sci. 2024,25, 4603 18 of 28
s.c.; [
20
,
26
]). Subsequently, we investigated the antiemetic potential of clonidine and
dexmedetomidine, and found that both
α2
-adrenergic receptor agonists dose-dependently
reduce to varying degrees both the frequency and percentage of shrews vomiting caused
by diverse emetogens, including the following.
1.
The
α2
-adrenergic receptor antagonist yohimbine (1 mg/kg, i.p.): Both clonidine and
dexmedetomidine reduced the mean frequency and percentage of shrews vomiting
in response to yohimbine in a dose-dependent manner with respective ID
50
values
ranging between 0.21 mg/kg and 0.021 mg/kg, respectively. Thus, dexmedetomidine
appears to be ten times more potent antiemetic than clonidine against yohimbine-
evoked emesis, and both agents completely protected shrews from vomiting at their
maximal tested dose of 0.1 and 10 mg/kg, respectively. Likewise, dexmedetomi-
dine is reported to be eight times more potent than clonidine in the clinic [
71
]. It
is suggested that yohimbine-induced vomiting could be due to blockade of the in-
hibitory
α2
-adrenergic receptors, which is associated with the enhanced release of
monoamines NE, DA, and 5-HT [
50
]. Clonidine overrides the ability of yohimbine to
release monoamines, and thus prevents vomiting evoked by the monoamine releaser,
reserpine [
31
]. These maximal tested doses of clonidine and dexmedetomidine in
least shrews were subsequently used as their utmost tested doses against vomiting
produced by the following emetogens.
2.
The more selective and centrally/peripherally-acting 5-HT
3
receptor agonist, 2-methyl-
5-HT (5 mg/kg, i.p.): 5-HT
3
receptor selective antagonists, such as tropisetron [
72
] or
palonosetron [
40
], can suppress vomiting caused by 2-Methyl-5-HT. Here, clonidine
and dexmedetomidine suppressed the mean frequency of vomiting and protected
shrews from emesis in a dose-dependent manner. In the latter case, dexmedetomidine
was 25 times more potent than clonidine in suppressing 2-Methyl-5-HT-evoked vom-
iting. However, clonidine completely protected shrews from vomiting at its highest
tested dose (10 mg/kg), whereas the largest dose of dexmedetomidine (0.1 mg/kg)
only prevented 75% of shrews from vomiting. This difference probably reflects the
nonselective nature of clonidine since the most potent and selective antagonist of
5-HT
3
receptors, palonosetron (10 mg/kg, i.p.), could not completely prevent least
shrews from 2-Methyl-5-HT-induced vomiting [
40
]. Ca
2+
mobilization via extracellu-
lar Ca
2+
influx through 5-HT
3
receptors and L-type Ca
2+
channels, and intracellular
Ca
2+
release via ryanodine receptors (RyRs) present on the endoplasmic reticulum
(ER), initiate Ca
2+
-dependent sequential activation of CaMKIIa and signal-regulated
kinase 1/2 (ERK
1/2
), which contribute to 2-Methyl-5-HT-evoked emesis [
73
]. The
most studied signaling pathway of
α2
-adrenergic receptor is through G protein-
coupled receptors, which consists of direct inhibition (“membrane-delimited”) by
G protein
βγ
(G
βγ
) subunit complexes of Ca
2+
entry through voltage-gated Ca
2+
(Cav) channels [
74
]. For
α2
-adrenergic receptor, the L-type Cav channel is the main
effector [
75
]. Typically, L-type calcium currents are maximally inhibited by 50% upon
α2
-adrenergic agonist application [
74
]. We suggest that the antiemetic effects of cloni-
dine and dexmedetomidine on 2-Methyl-5-HT-evoked emesis is probably related to
the inhibition of the L-type calcium currents.
3.
The selective SP neurokinin NK
1
receptor agonist GR73632 (5 mg/kg, i.p.): At this
dose, GR73632 causes robust vomiting in least shrews through the activation of SP
neurokinin NK
1
receptors [
76
], which can be completely blocked by the selective and
potent NK
1
receptor antagonist netupitant (10 mg/kg, i.p.) in least shrews [
77
]. In
the present study, both clonidine and dexmedetomidine prevented GR73632-evoked
emesis in a dose-dependent manner. However, the largest tested dose of clonidine
(10 mg/kg, i.p.) reduced the frequency of GR73632-evoked vomiting by 94% and
the percentage of least shrews vomiting by 71.4%. Also, dexmedetomidine at its
maximum tested dose (0.1 mg/kg) significantly but partially attenuated both the
mean vomit frequency (76.3%) and the percentage of least shrews vomiting (62.5%)
in response to GR73632. Their ID
50
values indicate that dexmedetomidine is more
Int. J. Mol. Sci. 2024,25, 4603 19 of 28
potent than clonidine in preventing GR73632-evoked vomiting. The NK
1
receptor is
G-protein coupled and can increase cytoplasmic Ca
2+
concentration [
78
80
]. In fact,
GR73632 evokes an increase in intracellular Ca
2+
concentration via both Ca
2+
release
from intracellular Ca
2+
stores, and extracellular Ca
2+
influx through the transient
receptor potential channels [
79
]. In addition, we have shown that LTCC blockers
amlodipine and nifedipine also suppress vomiting caused by GR73632 in a dose-
dependent manner [
40
]. Per our discussion above, we suggest that the antiemetic
effects of clonidine and dexmedetomidine on GR73632-induced emesis may involve
suppression of intracellular Ca2+ concentration.
4.
The cholinergic M
1
receptor agonist McN-A-343 (2 mg/kg, i.p.): Clonidine reduced
both the mean frequency and the percentage of least shrews vomiting in response to
McN-A-343 in a dose-dependent fashion, with complete emesis protection occurring at
its 10 mg/kg dose. However, dexmedetomidine significantly but partially reduced the
mean frequency of vomiting (66.7%) at its tested maximal dose of 0.1 mg/kg but failed
to significantly protect shrews from vomiting. McN-A-343 can activate PKC and PKA
phosphorylates to enhance Ca
2+
influx through LTCC channels [
81
,
82
]. Moreover, the
L-type calcium antagonist nifedipine can significantly and completely prevent in a
potent and dose-dependent manner both the percentage of shrews vomiting and the
mean frequency of emesis evoked by McN-A-343 (2 mg/kg, i.p.) in the least shrew [
83
].
The antiemetic effects of clonidine and dexmedetomidine on McN-A-343-induced
emesis may also involve suppression of intracellular Ca2+ mobilization.
5.
The dopamine D
2/3
receptor preferring agonist quinpirole (2 mg/kg, i.p.): Clonidine
only significantly but partially reduced the mean frequency of quinpirole-evoked
vomiting by 80.4% at its 10 mg/kg maximal tested dose but failed to completely
protect shrews from vomiting. Likewise, dexmedetomidine significantly reduced
both the mean frequency (57.8% reduction at 0.1 mg/kg) and the percentage of least
shrews vomiting (36.4% protection at 0.1 mg/kg) in response to quinpirole, but
these reductions were only partial. Although the dopamine D
2
receptor-preferring
antagonist sulpiride can completely prevent apomorphine (2 mg/kg, i.p.)-induced
vomiting in least shrews at 2 mg/kg (s.c.), but it cannot fully protect shrews from
quinpirole (2 mg/kg, i.p.)-evoked emesis even up to 8 mg/kg [
84
]. Thus, it appears
that quinpirole-evoked emesis cannot be easily subdued by sulpiride. Mechanistically,
we have previously demonstrated the involvement of the PI3K/mTOR/Akt signaling
pathway in dopamine D
2
receptor-evoked vomiting [
85
]. Since the
α2
-adrenergic
receptor agonist tizanidine can reduce the expression levels of PI3K/Akt [
86
], it is
possible that clonidine and dexmedetomidine may also affect this signaling system to
suppress the evoked vomiting, but this remains to be investigated.
6.
The cannabinoid CB
1
receptor-selective inverse agonist/antagonist SR141716A
(20 mg/kg, i.p.): SR141716A can induce vomiting in the least shrew at large doses
(20–40 mg/kg, i.p.) which can be fully prevented by CB
1
receptor agonists, includ-
ing
9
-THC [
87
]. Clonidine significantly reduced both the mean frequency and the
percentage of shrews vomiting in response to SR141716A in a dose-dependent manner
with complete emesis protection at it 10 mg/kg dose. Dexmedetomidine reduced the
mean frequency of vomiting by 88% and protected shrews from vomiting by 75% at
its maximal tested dose of 0.1 mg/kg. SR141716A increases the release and turnover
of monoamines DA, NE, and 5-HT [
88
] in several brain regions of rodents and least
shrews, and can enhance capsaicin-evoked release of SP in the mouse spinal cord [
89
].
As discussed earlier, since clonidine can prevent vomiting caused by the monoamine
releaser reserpine [
31
], it probably prevents SR141716A-evoked vomiting via a similar
mechanism.
7.
The selective LTCC agonist FPL64176 (10 m/kg, i.p.): The LTCC regulates extracel-
lular Ca
2+
influx into the cytosol [
90
]. FPL64176 is an extracellular Ca
2+
-mobilizing
agent and evokes vomiting in all tested shrews at 10 mg/kg [
40
,
83
]. Clonidine sig-
nificantly and completely reduced the mean vomiting frequency and the percentage
Int. J. Mol. Sci. 2024,25, 4603 20 of 28
of shrews vomiting in response to FPL64176 challenge in a dose-dependent manner.
Dexmedetomidine significantly and dose-dependently reduced the mean vomit fre-
quency by 92.3% following its maximal tested dose (0.1 mg/kg), and protected 71.4%
of shrews from FPL64176 (10 mg/kg, i.p.)-induced vomiting. However, their ID
50
values indicate that dexmedetomidine is more potent than clonidine in preventing
FPL64176-evoked vomiting. As was discussed earlier, the antiemetic effects of cloni-
dine and dexmedetomidine on FPL64176-evoked emesis may also involve inhibition
of L-type calcium currents.
8.
Specific inhibitor of the SERCA pump thapsigargin (0.5 mg/kg, i.p.): The SERCA
pump is a main mechanism that transports free cytosolic Ca
2+
into endoplasmic retic-
ulum (ER) Ca
2+
stores. Release of Ca
2+
from the ER stores into the cytosol occurs
through the inositol trisphosphate (IP
3
)- and ryanodine (RyR)-receptor ion chan-
nels localized on the ER membrane [
91
,
92
]. Thapsigargin is a specific and potent
inhibitor of SERCA pumps and causes a rapid elevation in cytosolic Ca
2+
concentra-
tions. Thapsigargin can also release Ca
2+
from the ER stores into the cytosol via IP
3
and RyR calcium channels [
93
95
]. Thapsigargin causes vomiting by triggering an
initial elevation in the cytoplasmic Ca
2+
concentration by inhibiting the SERCA as
well as releasing Ca
2+
from the ER into the cytoplasm via both RyR- and IP
3
-receptors
(IP3Rs), which is followed by an extracellular Ca
2+
influx through LTCCs prior to
the intracellular activation of the Ca
2+
-CaMKII-ERK1/2 cascade [
41
]. In the present
study, clonidine significantly reduced the mean frequency of thapsigargin-evoked
vomiting by 93.3% at its 10 mg/kg dose, but only partially protected 66.7% of shrews
at this maximal dose. Moreover, dexmedetomidine only reduced the mean frequency
of the evoked vomiting by 70.8% at its 0.1 mg/kg maximal dose but failed to sig-
nificantly protect shrews from vomiting. Since the LTCC inhibitor nifedipine can
dose-dependently inhibit thapsigargin-induced vomiting [
41
], we speculate that cloni-
dine and dexmedetomidine may partially suppress extracellular Ca
2+
influx through
LTCCs, which could then attenuate the above-discussed signaling cascade, leading to
a limited reduction in thapsigargin-evoked vomiting.
9.
The PDE4 inhibitor rolipram (1 mg/kg, i.p.): PDE4 inhibitors prevent metabolism
of second messengers such as cAMP and increase their tissue levels. They have
procognitive and antidepressant properties [
96
]. The emetic effect of some PDE4
inhibitors is thought to be a consequence of inhibition of PDE4 and the subsequent
increase in cAMP levels in the brainstem DVC [
30
,
42
]. PDE4 inhibitors may mimic
the pharmacological effect of
α2
-adrenergic receptor antagonists, which elevate in-
tracellular levels of cAMP in noradrenergic neurons [
30
]. In contrast,
α2
-adrenergic
receptor activation decreases intracellular levels of cAMP in noradrenergic neurons.
PDE4 inhibitors are thought to modulate the release of emetic mediators including
5-HT, SP, and noradrenaline which are involved in the onset of the emetic reflex [30].
We have previously shown that the PDE4 inhibitor rolipram (1 mg/kg, i.p.) evokes
both vomiting as well as significant increases in shrew brainstem cAMP levels, while
pretreatment with SQ22536, an inhibitor of adenylyl cyclase, prevented the evoked
emesis [
42
]. In the present study, clonidine dose-dependently and significantly, albeit
partially, reduced both the mean vomit frequency (80.3%) and the percentage (62.5%)
of shrews vomiting in response to rolipram at its highest tested dose, 10 mg/kg.
Likewise, dexmedetomidine produced similar dose-dependent but partial reductions
in both the mean vomit frequency (73.7%) and the percentage of shrews vomiting
(60%) at its maximal tested dose, 0.1 mg/kg. The antiemetic effect of clonidine and
dexmedetomidine against rolipram-induced vomiting might be due to suppression
of increased intracellular tissue levels of cAMP evoked by rolipram in the shrew
brainstem. In fact, administration of cAMP analogs such as 8-chloro-cAMP cause
vomiting in cancer patients [97].
Int. J. Mol. Sci. 2024,25, 4603 21 of 28
10.
The HCN blocker ZD7288 (1 mg/kg, i.p.): The HCN channels are a class of voltage-
gated ion-channels permeable to Na
+
and K
+
and constitutively open at voltages near
the resting membrane potential [
98
,
99
]. The hyperpolarization-activated currents (I
h
)
mediated by HCN channels elicit membrane depolarization toward a threshold for
action potential generation, which plays a pivotal role in controlling neuronal excitabil-
ity [
98
,
100
]. The HCN channel blocker ZD7288 can reduce apomorphine-induced
conditioned taste aversion to saccharin preference and depress the excitability of the
AP since it blocks HCN channel activation [
101
]. We have recently demonstrated that
ZD7288 (1 mg/kg, i.p.) evokes both vomiting in a dose-dependent manner as well
as a robust expression of c-fos and ERK
1/2
phosphorylation in the shrew brainstem
DVC, indicating a central contribution to the evoked vomiting [
43
]. In the present
study, clonidine significantly and dose-dependently reduced ZD7288-evoked mean
vomit frequency (92.4% reduction at 10 mg/kg), but only partially protected shrews
from vomiting (55.6% protection at 10 mg/kg). Dexmedetomidine only significantly
reduced the mean frequency (63.1% protection at 0.1 mg/kg) and failed to signifi-
cantly protect shrews from ZD7288-evoked vomiting. Given the well-known negative
coupling of
α2
-adrenergic receptors to adenylate cyclase via a heterotrimeric G pro-
tein [
102
], any reduction in the cAMP level would decrease I
h
. Dexmedetomidine
via
α2
-adrenergic receptors activates G-protein-coupled K
+
channels and inhibits
I
h
, which leads to membrane hyperpolarization [
103
]. In addition, clonidine can
directly inhibit I
f
current [
11
]. Hence, we speculate that administration of clonidine
and dexmedetomidine reverse the inhibitory effect of ZD7288 on the HCN channels
and consequently suppresses ZD7288-induced vomiting in shrews.
As discussed in the introduction section, although both clonidine and dexmedetomi-
dine are used as antiemetics for suppression of PONV, to date no basic or clinical study
has compared their antiemetic potential against diverse emetogens. In the present study
their antiemetic ID
50
values are comparatively determined against various emetogens, and
dexmedetomidine appears to be 3–69 times more potent than clonidine, demonstrating
that dexmedetomidine has more efficacious antiemetic potential at non-sedating doses.
4. Materials and Methods
4.1. Animals
A colony of adult least shrews between 45–60 days old and weighing between 4 and
6 g from the Western University of Health Sciences Animal Facilities were employed in
this study. Shrews were housed in groups of 5–10 on a 14:10 light: dark cycle and were
fed and watered ad libitum. Animal experiments were conducted in accordance with the
principles and procedures of the National Institutes of Health Guide for the Care and Use
of Laboratory Animals. All protocols were approved by the Institutional Animal Care and
Use Committee of Western University of Health Sciences (Protocol number R20IACUC018).
All efforts were made to minimize animal suffering and to reduce the number of animals
used in the experiments.
4.2. Chemicals
The following drugs were used in the present study: clonidine, dexmedetomidine,
yohimbine, GR73632, SR141716A, FPL64176, thapsigargin, and ZD7288 were purchased
from Tocris (Minneapolis, MN, USA); McN-A-343, quinpirole HCl, and rolipram were
purchased from Sigma-Aldrich (St. Louis, MO, USA); 2-methyl-serotonin maleate salt (2-
Methyl-5-HT) was purchased from Santa Cruz Biotechnology (Dallas, TX, USA). SR141716A
was dissolved to twice the stated drug dose in a 1:1:18 solution of ethanol:Emulphor™:0.9%
saline and was then diluted further with an equal volume of saline. FPL64176 was dissolved
in 25% DMSO in water. Thapsigargin was dissolved in 10% DMSO in distilled water.
Other drugs were dissolved in distilled water. All drugs were administered at a volume
of 0.1 mL/10 g of body weight. The doses and routes used for the emetogens were based
upon previous publications from our laboratory [6,43,87].
Int. J. Mol. Sci. 2024,25, 4603 22 of 28
4.3. Behavioral Emesis Studies
On the day of the experiment, both male and female shrews [
43
] were brought from
the animal facility, randomly separated into individual cages, and allowed to adapt to the
experimental condition for at least 2 h. Daily food was withheld 2 h prior to the start of the
experiment, but shrews were given 4 mealworms each 30 min prior to emetogen injection
to help identify wet vomits as described previously [
39
]. Experiments were performed
between 8:00 a.m. and 5:00 p.m.
To identify the emetic/antiemetic potential of either the
α2
-adrenergic receptor ago-
nists clonidine and dexmedetomidine, or the corresponding antagonist yohimbine, different
groups of shrews were injected with varying doses of either the corresponding vehicle,
clonidine (0.1, 1, 5, and 10 mg/kg, i.p., n= 6–10), dexmedetomidine (0.01, 0.05, 0.1, 0.5,
and 1 mg/kg, i.p., n= 6–10), or yohimbine (0.5, 0.75, 1, 1.5, 2, and 3 mg/kg, i.p., n= 7–14).
Immediately following the injection, each shrew was placed in the observation cage and
the frequency of emesis was recorded for the next 30 min. Based on the results obtained,
the corresponding vehicles, clonidine, or dexmedetomidine at all tested doses failed to
evoke vomiting in least shrews. However, the i.p. administration of yohimbine produced
vomiting in shrews in a dose-dependent and bell-shaped response manner, and a 1 mg/kg
dose of yohimbine caused maximal mean frequency of emesis in shrews, and thus was
chosen for subsequent immunofluorescence staining and drug interaction studies.
Since the
α2
-adrenergic receptor antagonist yohimbine caused vomiting, we initially
tested the antiemetic potential of its corresponding agonists clonidine and dexmedetomi-
dine in the following manner: different groups of shrews were pretreated with an injection
of either corresponding vehicles or varying doses of clonidine (0.1, 1, 5, and 10 mg/kg, i.p.)
or dexmedetomidine (0.01, 0.05, and 0.1 mg/kg, i.p.) at 0 min. Following 30 min, the pre-
treated shrews were challenged for vomiting with a maximally effective dose of yohimbine
(1 mg/kg, i.p.). Each shrew was then placed in the observation cage and the frequency of
vomiting was recorded for the next 30 min. Using the same procedure, we subsequently
investigated the antiemetic potential of these
α2
-adrenergic receptor agonists against fully
efficacious emetic dose of the following emetogens in separate experiments [
40
,
41
,
43
]:
(1) the selective serotonin 5-HT
3
receptor agonist 2-Methyl-5-HT (5 mg/kg, i.p.) [
73
];
(2) the selective SP neurokinin NK
1
receptor agonist GR73632 (5 mg/kg, i.p.) [
76
,
77
];
(3) the muscarinic M
1
receptor agonist McN-A-343 (2 mg/kg, i.p.) [
83
]; (4) the dopamine
D
2/3
preferring receptor agonist quinpirole (2 mg/kg, i.p.) [
84
]; (5) the CB
1
receptor in-
verse agonist/antagonist SR141716A (20 mg/kg, i.p.) [
87
]; (6) the LTCC agonist FPL64176
(10 mg/kg, i.p.) [
45
]; (7) the SERCA inhibitor thapsigargin (0.5 mg/kg, i.p.) [
41
];
(8) the
PDE4 inhibitor rolipram (1 mg/kg, i.p.) [
42
]; (9) the HCN channel blocker ZD7288
(1 mg/kg, i.p.) [
43
]. In the emesis studies the observer was unaware of treatment conditions.
Each shrew was used once, then euthanized with isoflurane (3%) in the anesthesia chamber
following the termination of each behavioral experiment.
4.4. Immunohistochemistry and Image Analysis
4.4.1. c-fos Staining and Image Analysis
Immunohistochemistry of the least shrew brainstem (20
µ
m) was conducted as previ-
ously reported [
43
,
77
]. Following vehicle or yohimbine (1 mg/kg, i.p.) injection, vomiting
shrews were subjected to c-fos staining (n= 6 shrews per group). Following 90 min after
the first emesis occurred, shrews were deeply anesthetized with isoflurane (3%), then
transcardially perfused with 0.01 M phosphate buffered saline (PBS) followed by ice cold
4% paraformaldehyde (pH 7.4) in 0.01 M PBS for 10 min. Brainstems were post-fixed in
the same fixative for 2 h, then placed in 0.1 M PB containing 30% sucrose at 4°C until they
sank. The brainstem was cut in 20
µ
m sections using a cryostat (Leica, Bannockburn, IL,
USA), and pre-incubated in the blocking buffer (0.01 M PBS containing 10% normal donkey
serum and 0.3% Triton X-100) for 1 h at room temperature. The slices were then incubated
in a rabbit anti-c-fos primary antibody (1:1000, ab190289, Abcam, Cambridge, UK) in 0.01M
PBS containing 5% normal donkey serum, 0.05% sodium azide, and 0.3% Triton X-100 at
Int. J. Mol. Sci. 2024,25, 4603 23 of 28
4°C overnight. The sections were washed 3 times (10 min each) in PBS and incubated in
an Alexa Fluor 594 donkey anti-rabbit secondary antibody (1:500, A-21207, Invitrogen,
Waltham, USA) in 0.01 M PBS containing 0.3% Triton X-100 for 2 h at room temperature,
then washed 3 times (10 min each) and were mounted and coverslipped with an anti-fade
mounting medium containing DAPI (Vector Laboratories, Newark, USA). Tile-scanning
images of the brainstem sections containing the brainstem emetic nuclei (AP/NTS/DMNV)
were taken by a confocal microscope (Zeiss LMS 880, Oberkochen, Germany) at 1024
×
1024
pixels with Zen software using Plan-Apochromat 20
×
/0.8 M27 objective. Cytoarchitectonic
differences in the AP, NTS, and DMNV of the least shrew brainstem have been described in
our published studies [
41
,
45
,
76
]. For each animal, c-fos positive cells in the AP and both
sides of NTS and DMNX from 3 sections at 90
µ
m intervals were counted manually by
an experimenter blind to the experimental conditions. The average value was used in
statistical analysis.
4.4.2. 5-HT and SP Immunohistochemistry
Shrews (n= 5–6 shrews per group) were treated with either vehicle or yohimbine
(1 mg/kg, i.p.) and rapidly anesthetized with isoflurane and subjected to perfusion at
15 min and 30 min post-treatment to examine 5-HT and SP immunoreactivity. The experi-
mental procedure prior to staining was performed as described above for Section 4.4.1. c-fos
Staining and Image Analysis. Coronal brainstem sections (20
µ
m) were blocked with 0.1 M
PBS containing 10% donkey serum and 0.3% Triton X-100, then incubated overnight at 4
C
with a mix of goat anti-5-HT primary antibody (1:1000, ab66047, Abcam) and rat anti-SP
primary antibody (1:400, MAB356, EMD Millipore, Burlington, VT, USA) in 0.1 M PBS
containing 5% donkey serum and 0.3% Triton X-100. Sections were washed 3 times (10 min
each) in PBS and incubated in a mix of Alexa Fluor 488 donkey anti-goat (1:500, ab150133,
Abcam) and cy3-conjugated donkey anti-rat (1:500, AP189C, EMD Millipore) secondary
antibody in 0.1 M PBS containing 0.3% Triton X-100 for 2 h at room temperature. After
washing with PBS 3 times (10 min each), sections were mounted with anti-fade mounting
medium containing DAPI (Vector Laboratories). Images for the DVC were acquired using
a confocal microscope (Zeiss LMS 880) as described above. Fluorescence intensity (mean
gray value) of 5-HT and SP values were acquired using ImageJ software, as described
previously [45].
4.5. Locomotor Activity Studies
Because
α2
-adrenergic receptor agonists have potent analgesic, sedative/hypnotic
properties in humans and in experimental animals [
104
107
], the locomotor activity param-
eters (i.e., total distance moved and rearing behavior) were measured in shrews following
injection of varying doses of clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p.) or dexmedetomidine
(0, 0.01, 0.05, 0.1, and 0.5 mg/kg, i.p.). The Ethovision (version XT 9) locomotion analysis and
behavior recognition system (Noldus Information Technology, Wageningen, The Netherlands)
was used, as reported previously [108]. The parameters of Ethovision were set to record two
locomotor activities: (1) total distance moved in centimeters (spontaneous locomotor activity),
and (2) rearing frequency, which was recorded when the shrew was standing upright with a
5% reduction in body surface area as seen by the overhead video camera.
On the day of the experiment, both male and female shrews were brought in their home
cages to the experimental room and were allowed to acclimate to a semi-dark environment
for 1 h. Each shrew was further acclimated in an empty white plastic observation cage
(27.5
×
27.5
×
28 cm) for 1 h before testing. Different groups of shrews were injected with
either corresponding vehicle or varying doses of clonidine (0.1, 1, 5, and 10 mg/kg, i.p.,
n= 6–10) or dexmedetomidine (0.01, 0.05, 0.1, and 0.5 mg/kg, i.p., n= 6–10) at 0 min.
Following 30 min, each shrew was individually placed in a white observation cage of the
same dimension, and the two motor parameters were recorded for 30 min by an overhead
camera and data were analyzed using the Noldus software. The chamber was thoroughly
Int. J. Mol. Sci. 2024,25, 4603 24 of 28
cleaned with 70% ethanol and dried to eliminate animal odors between test sessions. Each
shrew was used once and euthanized using isoflurane at the end of the experiment.
4.6. Statistical Analysis
Statistical analyses were done using Graphpad Prism 8 (Graphpad software Inc., San
Diego, CA, USA). The frequencies of vomits were analyzed using the Kruskal–Wallis non-
parametric one-way analysis of variance (ANOVA) followed by Dunn’s post hoc test and
expressed as the mean
±
SEM. The percentage of animals vomiting across treatment groups
at different doses was compared using the chi-square test. The locomotor activities and the
differences of 5-HT/SP mean gray values among groups were compared with an ordinary
ANOVA test followed by Dunnett’s post hoc test. The numbers of c-fos-positive cells
between two groups were tested by unpaired t-test. p< 0.05 was considered statistically
significant. Sample size calculations were determined as described by Chow [109].
5. Conclusions
Our results demonstrate that the
α2
-adrenergic receptor antagonist yohimbine is pro-
emetic in least shrews, and its corresponding agonists clonidine and dexmedetomidine
possess broad-spectrum antiemetic potential. In fact, yohimbine caused vomiting in a
bell-shaped and dose-dependent manner, accompanied by increased c-fos expression and
release of 5-HT and SP in the brainstem DVC emetic nuclei. Clonidine and dexmedeto-
midine not only suppressed yohimbine-evoked emesis in a dose-dependent manner, but
also vomiting caused by other well-investigated emetogens of varied mechanism of actions.
The broad-spectrum antiemetic effects of dexmedetomidine occur at much lower doses
than those of clonidine. Furthermore, the antiemetic doses (1, 5, and 10 mg/kg, i.p.) of
clonidine decreased the least shrew locomotor activity parameters, whereas dexmedetomi-
dine lacked motor suppressive behaviors. The current results suggest both clonidine and
dexmedetomidine possess broad-spectrum antiemetic potential, but dexmedetomidine’s
antiemetic ability occurs at non-sedative doses.
Author Contributions: N.A.D. and Y.S. conceived and designed the project, contributed to tissue
dissection and collection, completed immunohistochemical and behavioral experiments, contributed
toward statistical analyses, and wrote the manuscript. All authors have read and agreed to the
published version of the manuscript.
Funding: This work was funded in part by the NIH-NCI grant (CA207287) and the WesternU
intramural startup fund (1395) to N.A.D.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: The raw data supporting the conclusions of this article will be made
available by the authors, without undue reservation.
Conflicts of Interest: The authors declare no conflicts of interest.
References
1. Carpenter, D.O. Neural mechanisms of emesis. Can. J. Physiol. Pharmacol. 1990,68, 230–236. [CrossRef]
2. Horn, C.C. Why is the neurobiology of nausea and vomiting so important? Appetite 2008,50, 430–434. [CrossRef]
3.
Darmani, N.A.; Ray, A.P. Evidence for a re-evaluation of the neurochemical and anatomical bases of chemotherapy-induced
vomiting. Chem. Rev. 2009,109, 3158–3199. [CrossRef] [PubMed]
4. Hesketh, P.J. Chemotherapy-induced nausea and vomiting. N. Engl. J. Med. 2008,358, 2482–2494. [CrossRef] [PubMed]
5.
Singh, P.; Yoon, S.S.; Kuo, B. Nausea: A review of pathophysiology and therapeutics. Therap. Adv. Gastroenterol. 2016,9, 98–112.
[CrossRef] [PubMed]
6.
Zhong, W.; Shahbaz, O.; Teskey, G.; Beever, A.; Kachour, N.; Venketaraman, V.; Darmani, N.A. Mechanisms of Nausea and
Vomiting: Current Knowledge and Recent Advances in Intracellular Emetic Signaling Systems. Int. J. Mol. Sci. 2021,22, 5797.
[CrossRef]
7.
Babic, T.; Browning, K.N. The role of vagal neurocircuits in the regulation of nausea and vomiting. Eur. J. Pharmacol. 2014,722, 38–47.
[CrossRef]
Int. J. Mol. Sci. 2024,25, 4603 25 of 28
8.
Pertovaara, A. The noradrenergic pain regulation system: A potential target for pain therapy. Eur. J. Pharmacol. 2013,716, 2–7.
[CrossRef] [PubMed]
9.
Phillips, J.K.; McLean, A.J.; Hill, C.E. Receptors involved in nerve-mediated vasoconstriction in small arteries of the rat hepatic
mesentery. Br. J. Pharmacol. 1998,124, 1403–1412. [CrossRef]
10.
Leao, R.M.; Von Gersdorff, H. Noradrenaline increases high-frequency firing at the calyx of Held synapse during development by
inhibiting glutamate release. J. Neurophysiol. 2002,87, 2297–2306. [CrossRef]
11.
Knaus, A.E.; Muthig, V.; Schickinger, S.; Moura, E.; Beetz, N.; Gilsbach, R.; Hein, L. Alpha2-adrenoceptor subtypes--unexpected
functions for receptors and ligands derived from gene-targeted mouse models. Neurochem. Int. 2007,51, 277–281. [CrossRef]
[PubMed]
12.
Davis, R.; Wilde, M.I. Mirtazapine: A Review of its Pharmacology and Therapeutic Potential in the Management of Major
Depression. CNS Drugs 1996,5, 389–402. [CrossRef] [PubMed]
13.
Van Bockstaele, E.J. Morphological substrates underlying opioid, epinephrine and gamma-aminobutyric acid inhibitory actions
in the rat locus coeruleus. Brain Res. Bull. 1998,47, 1–15. [CrossRef] [PubMed]
14.
Kirouac, G.J. Placing the paraventricular nucleus of the thalamus within the brain circuits that control behavior. Neurosci. Biobehav.
Rev. 2015,56, 315–329. [CrossRef] [PubMed]
15.
Van Bockstaele, E.J.; Peoples, J.; Telegan, P. Efferent projections of the nucleus of the solitary tract to peri-locus coeruleus dendrites
in rat brain: Evidence for a monosynaptic pathway. J. Comp. Neurol. 1999,412, 410–428. [CrossRef]
16.
McCune, S.K.; Voigt, M.M.; Hill, J.M. Expression of multiple alpha adrenergic receptor subtype messenger RNAs in the adult rat
brain. Neuroscience 1993,57, 143–151. [CrossRef] [PubMed]
17.
Nicholas, A.P.; Pieribone, V.; Hokfelt, T. Distributions of mRNAs for alpha-2 adrenergic receptor subtypes in rat brain: An in situ
hybridization study. J. Comp. Neurol. 1993,328, 575–594. [CrossRef] [PubMed]
18.
Robertson, S.D.; Plummer, N.W.; de Marchena, J.; Jensen, P. Developmental origins of central norepinephrine neuron diversity.
Nat. Neurosci. 2013,16, 1016–1023. [CrossRef]
19.
Hikasa, Y.; Takase, K.; Ogasawara, S. Evidence for the involvement of alpha 2-adrenoceptors in the emetic action of xylazine in
cats. Am. J. Vet. Res. 1989,50, 1348–1351.
20.
Hikasa, Y.; Ogasawara, S.; Takase, K. Alpha adrenoceptor subtypes involved in the emetic action in dogs. J. Pharmacol. Exp. Ther.
1992,261, 746–754.
21.
Hikasa, Y.; Akiba, T.; Iino, Y.; Matsukura, M.; Takase, K.; Ogasawara, S. Central alpha-adrenoceptor subtypes involved in the
emetic pathway in cats. Eur. J. Pharmacol. 1992,229, 241–251.
22.
Japundzic-Zigon, N.; Samardzic, R.; Beleslin, D.B. Clonidine-induced emesis: A multitransmitter pathway concept. Pharmacol.
Res. 1997,35, 287–297. [CrossRef]
23.
McSweeney, P.M.; Martin, D.D.; Ramsey, D.S.; McKusick, B.C. Clinical efficacy and safety of dexmedetomidine used as a
preanesthetic prior to general anesthesia in cats. J. Am. Vet. Med. Assoc. 2012,240, 404–412. [CrossRef] [PubMed]
24.
Santos, L.C.; Ludders, J.W.; Erb, H.N.; Martin-Flores, M.; Basher, K.L.; Kirch, P. A randomized, blinded, controlled trial of the
antiemetic effect of ondansetron on dexmedetomidine-induced emesis in cats. Vet. Anaesth. Analg. 2011,38, 320–327. [CrossRef]
[PubMed]
25.
Granholm, M.; McKusick, B.C.; Westerholm, F.C.; Aspegren, J.C. Evaluation of the clinical efficacy and safety of dexmedetomidine
or medetomidine in cats and their reversal with atipamezole. Vet. Anaesth. Analg. 2006,33, 214–223. [CrossRef]
26.
Brioschi, F.A.; Gioeni, D.; Jacchetti, A.; Carotenuto, A.M. Effect of metoclopramide on nausea and emesis in dogs premedicated
with morphine and dexmedetomidine. Vet. Anaesth. Analg. 2018,45, 190–194. [CrossRef] [PubMed]
27.
Nystrom, M.R.; Odunayo, A.; Okafor, C.C. Assessment of hydromorphone and dexmedetomidine for emesis induction in cats.
J. Vet. Emerg. Crit. Care 2019,29, 360–365. [CrossRef]
28.
Papastefanou, A.K.; Galatos, A.D.; Pappa, E.; Lymperis, A.G.; Kostoulas, P. The effect of butorphanol on the incidence of
dexmedetomidine-induced emesis in cats. Vet. Anaesth. Analg. 2015,42, 608–613. [CrossRef] [PubMed]
29.
Hassen, K.M.; Posner, L.P.; Campbell, N.B. The effect of aquapuncture at Pericardium 6 (PC-6) on dexmedetomidine-induced
nausea and vomiting in cats. Vet. Anaesth. Analg. 2019,46, 308–314. [CrossRef]
30.
Robichaud, A.; Savoie, C.; Stamatiou, P.B.; Tattersall, F.D.; Chan, C.C. PDE4 inhibitors induce emesis in ferrets via a noradrenergic
pathway. Neuropharmacology 2001,40, 262–269. [CrossRef]
31.
Khandker, S.K.; Mukerjee, D.; Gurtu, S.; Pant, K.K.; Dhawan, K.N.; Sinha, J.N. Modification of reserpine-induced emetic response
in pigeons by alpha 2-adrenoceptors. Pharmacol. Res. 1994,29, 383–387. [CrossRef] [PubMed]
32.
Giampreti, A.; Lonati, D.; Locatelli, C.; Rocchi, L.; Campailla, M.T. Acute neurotoxicity after yohimbine ingestion by a body
builder. Clin. Toxicol. 2009,47, 827–829. [CrossRef] [PubMed]
33.
Sanchez Munoz, M.C.; De Kock, M.; Forget, P. What is the place of clonidine in anesthesia? Systematic review and meta-analyses
of randomized controlled trials. J. Clin. Anesth. 2017,38, 140–153. [CrossRef] [PubMed]
34.
Goyal, S.; Sharma, A.; Goswami, D.; Kothari, N.; Goyal, A.; Vyas, V.; Kirubakaran, R.; Sahu, R.; Singh, S. Clonidine and Morphine
as Adjuvants for Caudal Anaesthesia in Children: A Systematic Review and Meta-Analysis of Randomised Controlled Trials.
Turk. J. Anaesthesiol. Reanim. 2020,48, 265–272. [CrossRef] [PubMed]
35.
Zhang, Y.; Zhang, X.; Wang, Y.; Zhang, J. Effect of Clonidine on Hemodynamic Responses During Laparoscopic Cholecystectomy:
A Systematic Review and Meta-Analysis. Surg. Laparosc. Endosc. Percutan. Tech. 2017,27, 335–340. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024,25, 4603 26 of 28
36.
Yang, Y.; Yu, L.Y.; Zhang, W.S. Clonidine versus other adjuncts added to local anesthetics for pediatric neuraxial blocks:
A systematic review and meta-analysis. J. Pain Res. 2018,11, 1027–1036. [CrossRef] [PubMed]
37.
Zhu, M.; Wang, H.; Zhu, A.; Niu, K.; Wang, G. Meta-analysis of dexmedetomidine on emergence agitation and recovery profiles
in children after sevoflurane anesthesia: Different administration and different dosage. PLoS ONE 2015,10, e0123728. [CrossRef]
[PubMed]
38. Colbert, E.H. Tetrapod Extinctions at the End of the Triassic Period. Proc. Natl. Acad. Sci. USA 1958,44, 973–977. [CrossRef]
39.
Darmani, N.A. Serotonin 5-HT
3
receptor antagonists prevent cisplatin-induced emesis in Cryptotis parva: A new experimental
model of emesis. J. Neural. Transm. 1998,105, 1143–1154. [CrossRef]
40.
Darmani, N.A.; Zhong, W.; Chebolu, S.; Vaezi, M.; Alkam, T. Broad-spectrum antiemetic potential of the L-type calcium channel
antagonist nifedipine and evidence for its additive antiemetic interaction with the 5-HT (3) receptor antagonist palonosetron in
the least shrew (Cryptotis parva). Eur. J. Pharmacol. 2014,722, 2–12. [CrossRef]
41.
Zhong, W.; Chebolu, S.; Darmani, N.A. Thapsigargin-induced activation of Ca (2+)-CaMKII-ERK in brainstem contributes to
substance P release and induction of emesis in the least shrew. Neuropharmacology 2016,103, 195–210. [CrossRef] [PubMed]
42.
Alkam, T.; Chebolu, S.; Darmani, N.A. Cyclophosphamide causes activation of protein kinase A (PKA) in the brainstem of
vomiting least shrews (Cryptotis parva). Eur. J. Pharmacol. 2014,722, 156–164. [CrossRef]
43.
Zhong, W.; Darmani, N.A. The HCN Channel Blocker ZD7288 Induces Emesis in the Least Shrew (Cryptotis parva). Front. Pharmacol.
2021,12, 647021. [CrossRef]
44.
Andrews, P.L.; Naylor, R.J.; Joss, R.A. Neuropharmacology of emesis and its relevance to anti-emetic therapy. Consensus and
controversies. Support. Care Cancer 1998,6, 197–203. [CrossRef]
45.
Zhong, W.; Chebolu, S.; Darmani, N.A. Intracellular emetic signaling evoked by the L-type Ca(2+) channel agonist FPL64176 in
the least shrew (Cryptotis parva). Eur. J. Pharmacol. 2018,834, 157–168. [CrossRef] [PubMed]
46.
Haller, J.; Barna, I.; Kovacs, J.L. Alpha 2-adrenoceptor blockade, pituitary-adrenal hormones, and agonistic interactions in rats.
Psychopharmacology 1994,115, 478–484. [CrossRef] [PubMed]
47.
Cornil, C.A.; Ball, G.F. Interplay among catecholamine systems: Dopamine binds to alpha2-adrenergic receptors in birds and
mammals. J. Comp. Neurol. 2008,511, 610–627. [CrossRef]
48.
Millan, M.J.; Newman-Tancredi, A.; Audinot, V.; Cussac, D.; Lejeune, F.; Nicolas, J.P.; Coge, F.; Galizzi, J.P.; Boutin, J.A.; Rivet,
J.M.; et al. Agonist and antagonist actions of yohimbine as compared to fluparoxan at alpha(2)-adrenergic receptors (AR)s,
serotonin (5-HT)(1A), 5-HT(1B), 5-HT(1D) and dopamine D(2) and D(3) receptors. Significance for the modulation of frontocortical
monoaminergic transmission and depressive states. Synapse 2000,35, 79–95. [PubMed]
49.
Uhlen, S.; Dambrova, M.; Nasman, J.; Schioth, H.B.; Gu, Y.; Wikberg-Matsson, A.; Wikberg, J.E. [3H]RS79948-197 binding to
human, rat, guinea pig and pig alpha2A-, alpha2B- and alpha2C-adrenoceptors. Comparison with MK912, RX821002, rauwolscine
and yohimbine. Eur. J. Pharmacol. 1998,343, 93–101. [CrossRef]
50.
Brannan, T.; Martinez-Tica, J.; Yahr, M.D. Effect of yohimbine on brain monoamines: An
in vivo
study. J. Neural. Transm. Park. Dis.
Dement. Sect. 1991,3, 81–87. [CrossRef]
51.
Dunbar, J.C.; Clough-Helfman, C.; Barraco, R.A.; Anderson, G.F. Effect of insulin and clonidine on the evoked release of
norepinephrine and serotonin from the nucleus tractus solitarius of the diabetic rat. Pharmacology 1995,51, 370–380. [CrossRef]
[PubMed]
52.
Bullitt, E. Expression of c-fos-like protein as a marker for neuronal activity following noxious stimulation in the rat. J. Comp. Neurol.
1990,296, 517–530. [CrossRef] [PubMed]
53.
Ray, A.P.; Chebolu, S.; Darmani, N.A. Receptor-selective agonists induce emesis and Fos expression in the brain and enteric
nervous system of the least shrew (Cryptotis parva). Pharmacol. Biochem. Behav. 2009,94, 211–218. [CrossRef] [PubMed]
54.
Gershon, M.D.; Tack, J. The serotonin signaling system: From basic understanding to drug development for functional GI
disorders. Gastroenterology 2007,132, 397–414. [CrossRef] [PubMed]
55.
Johnston, K.D.; Lu, Z.; Rudd, J.A. Looking beyond 5-HT (3) receptors: A review of the wider role of serotonin in the pharmacology
of nausea and vomiting. Eur. J. Pharmacol. 2014,722, 13–25. [CrossRef] [PubMed]
56. Langer, S.Z. Presynaptic regulation of the release of catecholamines. Pharmacol. Rev. 1980,32, 337–362. [CrossRef] [PubMed]
57. Westfall, T.C. Neuroeffector mechanisms. Annu. Rev. Physiol. 1980,42, 383–397. [CrossRef] [PubMed]
58.
Cheng, C.H.; Costall, B.; Ge, J.; Naylor, R.J. The profiles of interaction of yohimbine with anxiolytic and putative anxiolytic agents
to modify 5-HT release in the frontal cortex of freely-moving rats. Br. J. Pharmacol. 1993,110, 1079–1084. [CrossRef]
59.
Ellison, D.W.; Campbell, I.C. Studies on the role of alpha 2-adrenoceptors in the control of synaptosomal [3H]5-hydroxytryptamine
release: Effects of antidepressant drugs. J. Neurochem. 1986,46, 218–223. [CrossRef]
60.
Takano, M.; Takano, Y.; Yaksh, T.L. Release of calcitonin gene-related peptide (CGRP), substance P (SP), and vasoactive intestinal
polypeptide (VIP) from rat spinal cord: Modulation by alpha 2 agonists. Peptides 1993,14, 371–378. [CrossRef]
61.
Kuraishi, Y.; Hirota, N.; Sato, Y.; Kaneko, S.; Satoh, M.; Takagi, H. Noradrenergic inhibition of the release of substance P from the
primary afferents in the rabbit spinal dorsal horn. Brain Res. 1985,359, 177–182. [CrossRef] [PubMed]
62.
Fukuda, H.; Koga, T. Non-respiratory neurons in the Botzinger complex exhibiting appropriate firing patterns to generate the
emetic act in dogs. Neurosci. Res. 1992,14, 180–194. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024,25, 4603 27 of 28
63.
Cruickshank, M.; Henderson, L.; MacLennan, G.; Fraser, C.; Campbell, M.; Blackwood, B.; Gordon, A.; Brazzelli, M. Alpha-2
agonists for sedation of mechanically ventilated adults in intensive care units: A systematic review. Health Technol. Assess. 2016,
20, v–xx, 1–117. [CrossRef] [PubMed]
64. Gavino, L.; Willaredt, M.; Chiche, J.D.; Ben-Hamouda, N. Alpha-2 adrenoreceptor agonists for the intensive care physician. Rev.
Med. Suisse 2023,19, 872–877. [PubMed]
65.
Durcan, M.J.; Lister, R.G.; Linnoila, M. Interactions of alpha 2-adrenoceptor antagonists with medetomidine and with ethanol in a
holeboard test. Neuropharmacology 1989,28, 275–281. [CrossRef] [PubMed]
66.
Malinge, M.; Colombel, M.C.; Bourin, M. Mechanism of action of clonidine in the forced-swimming test in mice. Encephale 1989,
15, 37–41. [PubMed]
67.
Smith, D.F. Lithium attenuates clonidine-induced hypoactivity: Further studies in inbred mouse strains. Psychopharmacology 1988,
94, 428–430. [CrossRef] [PubMed]
68.
Hunter, J.C.; Fontana, D.J.; Hedley, L.R.; Jasper, J.R.; Lewis, R.; Link, R.E.; Secchi, R.; Sutton, J.; Eglen, R.M. Assessment of the role
of alpha2-adrenoceptor subtypes in the antinociceptive, sedative and hypothermic action of dexmedetomidine in transgenic mice.
Br. J. Pharmacol. 1997,122, 1339–1344. [CrossRef]
69.
Houston, M.C. Clonidine hydrochloride: Review of pharmacologic and clinical aspects. Prog. Cardiovasc. Dis. 1981,23, 337–350.
[CrossRef]
70.
Kumar, A.; Bose, S.; Bhattacharya, A.; Tandon, O.P.; Kundra, P. Oral clonidine premedication for elderly patients undergoing
intraocular surgery. Acta Anaesthesiol. Scand. 1992,36, 159–164. [CrossRef]
71.
Ramadhyani, U.; Park, J.L.; Carollo, D.S.; Waterman, R.S.; Nossaman, B.D. Dexmedetomidine: Clinical application as an adjunct
for intravenous regional anesthesia. Anesthesiol. Clin. 2010,28, 709–722. [CrossRef]
72.
Darmani, N.A.; Chebolu, S.; Amos, B.; Alkam, T. Synergistic antiemetic interactions between serotonergic 5-HT
3
and tachykinin-
ergic NK
1
-receptor antagonists in the least shrew (Cryptotis parva). Pharmacol. Biochem. Behav. 2011,99, 573–579. [CrossRef]
[PubMed]
73.
Zhong, W.; Hutchinson, T.E.; Chebolu, S.; Darmani, N.A. Serotonin 5-HT
3
receptor-mediated vomiting occurs via the activation of
Ca
2+
/CaMKII-dependent ERK
1/2
signaling in the least shrew (Cryptotis parva). PLoS ONE 2014,9, e104718. [CrossRef] [PubMed]
74.
Hernandez-Guijo, J.M.; Carabelli, V.; Gandia, L.; Garcia, A.G.; Carbone, E. Voltage-independent autocrine modulation of L-type
channels mediated by ATP, opioids and catecholamines in rat chromaffin cells. Eur. J. Neurosci. 1999,11, 3574–3584. [CrossRef]
[PubMed]
75.
Kleppisch, T.; Ahnert-Hilger, G.; Gollasch, M.; Spicher, K.; Hescheler, J.; Schultz, G.; Rosenthal, W. Inhibition of voltage-dependent
Ca
2+
channels via alpha 2-adrenergic and opioid receptors in cultured bovine adrenal chromaffin cells. Pflugers Arch. 1992,421,
131–137. [CrossRef] [PubMed]
76.
Darmani, N.A.; Wang, Y.; Abad, J.; Ray, A.P.; Thrush, G.R.; Ramirez, J. Utilization of the least shrew as a rapid and selective
screening model for the antiemetic potential and brain penetration of substance P and NK
1
receptor antagonists. Brain Res. 2008,
1214, 58–72. [CrossRef] [PubMed]
77.
Zhong, W.; Chebolu, S.; Darmani, N.A. Intracellular emetic signaling cascades by which the selective neurokinin type 1 receptor
(NK(1)R) agonist GR73632 evokes vomiting in the least shrew (Cryptotis parva). Neurochem. Int. 2019,122, 106–119. [CrossRef]
[PubMed]
78.
Lin, Y.R.; Kao, P.C.; Chan, M.H. Involvement of Ca
2+
signaling in tachykinin-mediated contractile responses in swine trachea.
J. Biomed. Sci. 2005,12, 547–558. [CrossRef]
79.
Miyano, K.; Morioka, N.; Sugimoto, T.; Shiraishi, S.; Uezono, Y.; Nakata, Y. Activation of the neurokinin-1 receptor in rat spinal
astrocytes induces Ca
2+
release from IP3-sensitive Ca
2+
stores and extracellular Ca
2+
influx through TRPC3. Neurochem. Int. 2010,
57, 923–934. [CrossRef]
80.
Suzuki, Y.; Inoue, T.; Ra, C. L-type Ca
2+
channels: A new player in the regulation of Ca
2+
signaling, cell activation and cell
survival in immune cells. Mol. Immunol. 2010,47, 640–648. [CrossRef]
81.
Sculptoreanu, A.; Yoshimura, N.; de Groat, W.C.; Somogyi, G.T. Protein kinase C is involved in M
1
-muscarinic receptor-mediated
facilitation of L-type Ca
2+
channels in neurons of the major pelvic ganglion of the adult male rat. Neurochem. Res. 2001,26, 933–942.
[CrossRef] [PubMed]
82.
Oliveira, L.; Correia-de-Sa, P. Protein kinase A and Ca(v)1 (L-Type) channels are common targets to facilitatory adenosine A2A
and muscarinic M1receptors on rat motoneurons. Neurosignals 2005,14, 262–272. [CrossRef] [PubMed]
83.
Zhong, W.; Chebolu, S.; Darmani, N.A. Broad-spectrum antiemetic efficacy of the L-type calcium channel blocker amlodipine in
the least shrew (Cryptotis parva). Pharmacol. Biochem. Behav. 2014,120, 124–132. [CrossRef]
84.
Darmani, N.A.; Zhao, W.; Ahmad, B. The role of D
2
and D
3
dopamine receptors in the mediation of emesis in Cryptotis parva (the
least shrew). J. Neural. Transm. 1999,106, 1045–1061. [CrossRef] [PubMed]
85.
Belkacemi, L.; Zhong, W.; Darmani, N.A. Signal transduction pathways involved in dopamine D(2) receptor-evoked emesis in the
least shrew (Cryptotis parva). Auton. Neurosci. 2021,233, 102807. [CrossRef] [PubMed]
86.
Xing, X.W.; Sun, Y.F.; Zhao, J.; Pan, Z.X.; Jiang, W.X. Tizanidine hydrochloride exhibits a cytotoxic effect on osteosarcoma cells
through the PI3K/AKT signaling pathway. J. Int. Med. Res. 2019,47, 3792–3802. [CrossRef]
87.
Darmani, N.A. Delta (9)-tetrahydrocannabinol and synthetic cannabinoids prevent emesis produced by the cannabinoid CB (1)
receptor antagonist/inverse agonist SR 141716A. Neuropsychopharmacology 2001,24, 198–203. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024,25, 4603 28 of 28
88.
Tzavara, E.T.; Davis, R.J.; Perry, K.W.; Li, X.; Salhoff, C.; Bymaster, F.P.; Witkin, J.M.; Nomikos, G.G. The CB
1
receptor antagonist
SR141716A selectively increases monoaminergic neurotransmission in the medial prefrontal cortex: Implications for therapeutic
actions. Br. J. Pharmacol. 2003,138, 544–553. [CrossRef] [PubMed]
89.
Lever, I.J.; Malcangio, M. CB (1) receptor antagonist SR141716A increases capsaicin-evoked release of Substance P from the adult
mouse spinal cord. Br. J. Pharmacol. 2002,135, 21–24. [CrossRef]
90.
Zamponi, G.W.; Striessnig, J.; Koschak, A.; Dolphin, A.C. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium
Channels and Their Future Therapeutic Potential. Pharmacol. Rev. 2015,67, 821–870. [CrossRef]
91.
Garaschuk, O.; Yaari, Y.; Konnerth, A. Release and sequestration of calcium by ryanodine-sensitive stores in rat hippocampal
neurones. J. Physiol. 1997,502 Pt 1, 13–30. [CrossRef]
92.
Gomez-Viquez, N.L.; Guerrero-Serna, G.; Arvizu, F.; Garcia, U.; Guerrero-Hernandez, A. Inhibition of SERCA pumps induces
desynchronized RyR activation in overloaded internal Ca
2+
stores in smooth muscle cells. Am. J. Physiol. Cell Physiol. 2010,298,
C1038–C1046. [CrossRef]
93.
Beltran-Parrazal, L.; Fernandez-Ruiz, J.; Toledo, R.; Manzo, J.; Morgado-Valle, C. Inhibition of endoplasmic reticulum Ca
2+
ATPase in preBotzinger complex of neonatal rat does not affect respiratory rhythm generation. Neuroscience 2012,224, 116–124.
[CrossRef] [PubMed]
94.
Michelangeli, F.; East, J.M. A diversity of SERCA Ca
2+
pump inhibitors. Biochem. Soc. Trans. 2011,39, 789–797. [CrossRef]
[PubMed]
95. Solovyova, N.; Verkhratsky, A. Neuronal endoplasmic reticulum acts as a single functional Ca2+ store shared by ryanodine and
inositol-1,4,5-trisphosphate receptors as revealed by intra-ER [Ca
2+
] recordings in single rat sensory neurones. Pflugers Arch. 2003,
446, 447–454. [CrossRef] [PubMed]
96.
Richter, W.; Menniti, F.S.; Zhang, H.T.; Conti, M. PDE4 as a target for cognition enhancement. Expert. Opin. Ther. Targets 2013,17,
1011–1027. [CrossRef] [PubMed]
97.
Propper, D.J.; Saunders, M.P.; Salisbury, A.J.; Long, L.; O’Byrne, K.J.; Braybrooke, J.P.; Dowsett, M.; Taylor, M.; Talbot, D.C.;
Ganesan, T.S.; et al. Phase I study of the novel cyclic AMP (cAMP) analogue 8-chloro-cAMP in patients with cancer: Toxicity,
hormonal, and immunological effects. Clin. Cancer Res. 1999,5, 1682–1689. [PubMed]
98. Benarroch, E.E. HCN channels: Function and clinical implications. Neurology 2013,80, 304–310. [CrossRef]
99.
McGovern, A.E.; Robusto, J.; Rakoczy, J.; Simmons, D.G.; Phipps, S.; Mazzone, S.B. The effect of hyperpolarization-activated
cyclic nucleotide-gated ion channel inhibitors on the vagal control of guinea pig airway smooth muscle tone. Br. J. Pharmacol.
2014,171, 3633–3650. [CrossRef]
100.
Gill, C.H.; Randall, A.; Bates, S.A.; Hill, K.; Owen, D.; Larkman, P.M.; Cairns, W.; Yusaf, S.P.; Murdock, P.R.; Strijbos, P.J.; et al.
Characterization of the human HCN1 channel and its inhibition by capsazepine. Br. J. Pharmacol. 2004,143, 411–421. [CrossRef]
101.
Shinpo, K.; Hirai, Y.; Maezawa, H.; Totsuka, Y.; Funahashi, M. The role of area postrema neurons expressing H-channels in the
induction mechanism of nausea and vomiting. Physiol. Behav. 2012,107, 98–103. [CrossRef] [PubMed]
102.
Docherty, J.R. Subtypes of functional alpha1- and alpha2-adrenoceptors. Eur. J. Pharmacol. 1998,361, 1–15. [CrossRef] [PubMed]
103.
Yang, Y.C.; Meng, Q.T.; Pan, X.; Xia, Z.Y.; Chen, X.D. Dexmedetomidine produced analgesic effect via inhibition of HCN currents.
Eur. J. Pharmacol. 2014,740, 560–564. [CrossRef] [PubMed]
104.
Vulliemoz, Y.; Shen, H.; Virag, L. Alpha-2 adrenoceptor agonists decrease cyclic guanosine 3
,5
-monophosphate in the mouse
brain. Anesthesiology 1996,85, 544–550. [CrossRef] [PubMed]
105.
Glaess, S.S.; Attridge, R.L.; Christina Gutierrez, G. Clonidine as a strategy for discontinuing dexmedetomidine sedation in
critically ill patients: A narrative review. Am. J. Health Syst. Pharm. 2020,77, 515–522. [CrossRef]
106.
Keating, G.M. Dexmedetomidine: A Review of Its Use for Sedation in the Intensive Care Setting. Drugs 2015,75, 1119–1130.
[CrossRef]
107. Nguyen, V.; Tiemann, D.; Park, E.; Salehi, A. Alpha-2 Agonists. Anesthesiol. Clin. 2017,35, 233–245. [CrossRef]
108.
Belkacemi, L.; Sun, Y.; Darmani, N.A. Evidence for Bell-Shaped Dose-Response Emetic Effects of Temsirolimus and Analogs: The
Broad-Spectrum Antiemetic Efficacy of a Large Dose of Temsirolimus Against Diverse Emetogens in the Least Shrew (Cryptotis
parva). Front. Pharmacol. 2022,13, 848673. [CrossRef]
109.
Chow, S.-C.; Wang, H.; Shao, J. Sample Size Calculations in Clinical Research, 2nd ed.; Chapman and Hall/CRC: New York, NY,
USA, 2007; p. 480. ISBN 9780429144639.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... They trigger various stimuli that are responsible for the emetic process [16,17]. Also, various types of adrenergic (α 2 ), CB 1 , and GABA B receptors are responsible for activating emesis [18][19][20]. ...
Article
Full-text available
Background This study emphasizes evaluating the antiemetic efficacy of the natural food component caffeic acid (CAF) using a copper sulfate pentahydrate (CuSO4.5H2O)‐induced emetic model on chicks, and an in silico approach was also adopted to estimate the possible underlying mechanisms. Methods Two doses (25 and 50 mg/kg b.w.) of CAF and several referral drugs considered positive controls (PCs), including domperidone (6 mg/kg), hyoscine (21 mg/kg), aprepitant (16 mg/kg), diphenhydramine (10 mg/kg), and ondansetron (5 mg/kg), were orally administered to chicks. The vehicle served as the control group. Co‐treatments of CAF with referral drugs were also provided to chicks to evaluate the modulatory action of the test compound. Results According to the results, CAF delayed the emetic onset and decreased the frequency of retches in a dose‐dependent manner compared to the vehicle group. CAF (40 mg/kg) represented a notable delayed latency period (60.17 ± 3.16 s) and a diminished number of retchings (18.33 ± 1.74 times) compared to the control group. Further, in the co‐treatments, CAF increased the latency period and reduced the number of retches, except for domperidone. In the in silico investigation, CAF showed notable binding affinity toward the D2 (−7.3 kcal/mol), 5HT3 (−7.1 kcal/mol), and M5 (−7 kcal/mol) receptors in the same binding site as the referral ligand. Conclusion This research indicated that CAF has mild antiemetic properties by interacting with the D2, 5TH3, and M5 receptors. Therefore, several preclinical and clinical studies are necessary to assess the effectiveness and safety profile of this food ingredient.
... Our study reveals a novel role of DEX in modulating intestinal function, particularly in the context of diabetes. DEX significantly influences intestinal motility and immune responses by interacting with the MMP23B pathway [19,20]. This finding is pivotal as it suggests that DEX counteracts diabetes-related intestinal tissue damage by regulating the activity of M1 macrophages and expression of MMP23B, a key inflammatory factor. ...
Article
BACKGROUND Diabetes is often associated with gastrointestinal dysfunctions, which can lead to hypoglycemia. Dexmedetomidine (DEX) is a commonly used sedative in perioperative diabetic patients and may affect gastrointestinal function. AIM To investigate whether sedative doses of DEX alleviate diabetes-caused intestinal dysfunction. METHODS Sedation/anesthesia scores and vital signs of streptozotocin (STZ)-induced diabetic mice under DEX sedation were observed. Diabetic mice were divided into saline and DEX groups. After injecting sedatives intraperitoneally, tight junctions (TJs) and apoptotic levels were evaluated 24 hours later to assess the intestinal barrier function. The role of DEX was validated using Villin-MMP23B flox/flox mice with intestinal epithelial deletion. In vitro , high glucose and hyperosmolarity were used to culture Caco-2 monolayer cells with STZ inter-vention. Immunofluorescence techniques were used to monitor the barrier and mitochondrial functions. RESULTS MMP23B protein levels in the intestinal tissue of STZ-induced diabetic mice were significantly higher than those in the intestinal tissue of control mice, with the DEX group displaying decreased MMP23B levels. Diabetes-mediated TJ dis-ruption, increased intestinal mucosal permeability, and systemic inflammation in wild-type mice might be reversed by DEX. In Caco-2 cells, MMP23B was associated with increased reactive oxygen species accumulation, mitochondrial membrane potential depolarization, and TJ disruption. CONCLUSION DEX reduces MMP23B, which may potentially contribute to STZ-induced intestinal barrier dysfunction, affecting TJ modification through mitochondrial dysfunction.
Article
Full-text available
Temsirolimus is a prodrug form of sirolimus (rapamycin). With its analogs (everolimus, ridaforolimus, and rapamycin), it forms a group of anticancer agents that block the activity of one of the two mammalian targets of rapamycin (mTOR) complexes, mTORC1. We investigated the emetic potential of varying doses (0, 0.5, 1, 2.5, 5, 10, 20, and 40 mg/kg, i.p.) of temsirolimus in the least shrew. Temsirolimus caused a bell-shaped and dose-dependent increase in both the mean vomit frequency and the number of shrews vomiting with maximal efficacy at 10 mg/kg (p < 0.05 and p < 0.02, respectively). Its larger doses (20 or 40 mg/kg) had no significant emetic effect. We also evaluated the emetic potential of its analogs (5, 10, and 20 mg/kg, i.p.), all of which exhibited a similar emetic profile. Our observational studies indicated that temsirolimus can reduce the shrew motor activity at 40 mg/kg, and subsequently, we examined the motor effects of its lower doses. At 10 and 20 mg/kg, it did not affect the spontaneous locomotor activity (distance moved) but attenuated the mean rearing frequency in a U-shaped manner at 10 mg/kg (p < 0.05). We then determined the broad-spectrum antiemetic potential of a 20 mg/kg (i.p.) dose of temsirolimus against diverse emetogens, including selective and nonselective agonists of 1) dopaminergic D2/3 receptors (apomorphine and quinpirole); 2) serotonergic 5-HT3 receptors [5-HT (serotonin) and 2-methyl-5-HT]; 3) cholinergic M1 receptors (pilocarpine and McN-A-343); 4) substance P neurokinin NK1 receptors (GR73632); 5) the L-type calcium (Ca²⁺) channel (LTCC) (FPL64176); 6) the sarcoplasmic endoplasmic reticulum Ca²⁺ ATPase inhibitor, thapsigargin; 7) the CB1 receptor inverse agonist/antagonist, SR141716A; and 8) the chemotherapeutic cisplatin. Temsirolimus prevented vomiting evoked by the aforementioned emetogens with varying degrees. The mechanisms underlying the pro- and antiemetic effects of temsirolimus evaluated by immunochemistry for c-fos expression demonstrated a c-fos induction in the AP and NTS, but not DMNX with the 10 mg/kg emetic dose of temsirolimus, whereas its larger antiemetic dose (20 mg/kg) had no significant effect. Our study is the first to provide preclinical evidence demonstrating the promising antiemetic potential of high doses of temsirolimus and possibly its analogs in least shrews.
Article
Full-text available
Nausea and vomiting are common gastrointestinal complaints that can be triggered by diverse emetic stimuli through central and/or peripheral nervous systems. Both nausea and vomiting are considered as defense mechanisms when threatening toxins/drugs/bacteria/viruses/fungi enter the body either via the enteral (e.g., the gastrointestinal tract) or parenteral routes, including the blood, skin, and respiratory systems. While vomiting is the act of forceful removal of gastrointestinal contents, nausea is believed to be a subjective sensation that is more difficult to study in nonhuman species. In this review, the authors discuss the anatomical structures, neurotransmitters/mediators, and corresponding receptors, as well as intracellular emetic signaling pathways involved in the processes of nausea and vomiting in diverse animal models as well as humans. While blockade of emetic receptors in the prevention of vomiting is fairly well understood, the potential of new classes of antiemetics altering postreceptor signal transduction mechanisms is currently evolving, which is also reviewed. Finally, future directions within the field will be discussed in terms of important questions that remain to be resolved and advances in technology that may help provide potential answers.
Article
Full-text available
Subtypes (1–4) of the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are widely expressed in the central and peripheral nervous systems, as well as the cells of smooth muscles in many organs. They mainly serve to regulate cellular excitability in these tissues. The HCN channel blocker ZD7288 has been shown to reduce apomorphine-induced conditioned taste aversion on saccharin preference in rats suggesting potential antinausea/antiemetic effects. Currently, in the least shew model of emesis we find that ZD7288 induces vomiting in a dose-dependent manner, with maximal efficacies of 100% at 1 mg/kg (i.p.) and 83.3% at 10 µg (i.c.v.). HCN channel subtype (1–4) expression was assessed using immunohistochemistry in the least shrew brainstem dorsal vagal complex (DVC) containing the emetic nuclei (area postrema (AP), nucleus tractus solitarius and dorsal motor nucleus of the vagus). Highly enriched HCN1 and HCN4 subtypes are present in the AP. A 1 mg/kg (i.p.) dose of ZD7288 strongly evoked c-Fos expression and ERK1/2 phosphorylation in the shrew brainstem DVC, but not in the in the enteric nervous system in the jejunum, suggesting a central contribution to the evoked vomiting. The ZD7288-evoked c-Fos expression exclusively occurred in tryptophan hydroxylase 2-positive serotonin neurons of the dorsal vagal complex, indicating activation of serotonin neurons may contribute to ZD7288-induced vomiting. To reveal its mechanism(s) of emetic action, we evaluated the efficacy of diverse antiemetics against ZD7288-evoked vomiting including the antagonists/inhibitors of: ERK1/2 (U0126), L-type Ca²⁺ channel (nifedipine); store-operated Ca²⁺ entry (MRS 1845); T-type Ca²⁺ channel (Z944), IP3R (2-APB), RyR receptor (dantrolene); the serotoninergic type 3 receptor (palonosetron); neurokinin 1 receptor (netupitant), dopamine type 2 receptor (sulpride), and the transient receptor potential vanilloid 1 receptor agonist, resiniferatoxin. All tested antiemetics except sulpride attenuated ZD7288-evoked vomiting to varying degrees. In sum, ZD7288 has emetic potential mainly via central mechanisms, a process which involves Ca²⁺ signaling and several emetic receptors. HCN channel blockers have been reported to have emetic potential in the clinic since they are currently used/investigated as therapeutic candidates for cancer therapy related- or unrelated-heart failure, pain, and cognitive impairment.
Article
Full-text available
With its five receptor subtypes (D1–5), dopamine is implicated in a myriad of neurological illnesses. Dopamine D2 receptor-based agonist therapy evokes nausea and vomiting. The signaling mechanisms by which dopamine D2 receptors evoke vomiting remains unknown. Phosphatidylinositol 3-kinases (PI3K)- and protein kinase C (PKC)-related signaling cascades stimulate vomiting post-injection of various emetogens in emetically competent animals. This study investigated potential mechanisms involved in dopamine D2 receptor-mediated vomiting using least shrews. We found that vomiting evoked by the selective dopamine D2 receptor agonist quinpirole (2 mg/kg, i.p.) was significantly suppressed by: i) a dopamine D2 preferring antagonist, sulpiride (s.c.); ii) a selective PI3K inhibitor, LY294002 (i.p.); iii) a PKCαβII inhibitor, GF109203X (i.p.); and iv) a selective inhibitor of extracellular signal-regulated protein kinase1/2 (ERK1/2), U0126 (i.p.). Quinpirole-evoked c-fos immunofluorescence in the nucleus tractus solitarius (NTS) was suppressed by pretreatment with sulpiride (8 mg/kg, s.c.). Western blot analysis of shrew brainstem emetic loci protein lysates revealed a significant and time-dependent increase in phosphorylation of Akt (protein kinase B (PKB)) at Ser473 following a 30-min exposure to quinpirole (2 mg/kg, i.p.). Pretreatment with effective antiemetic doses of sulpiride, LY294002, GF109203X, or U0126 significantly reduced quinpirole-stimulated phosphorylation of emesis-associated proteins including p-85PI3K, mTOR (Ser2448/2481), PKCαβII (Thr638/641), ERK1/2 (Thr202/204), and Akt (Ser473). Our results substantiate the implication of PI3K/mTOR/Akt and PI3K/PKCαβII/ERK1/2/Akt signaling pathways in dopamine D2 receptor-mediated vomiting. Potential novel antiemetics targeting emetic proteins associated with these signaling cascades may offer enhanced potency and/or efficacy against emesis.
Article
Full-text available
Objective: The aim of this systematic review and meta-analysis is to compare the outcomes of morphine vs. clonidine use as adjuvants in caudal anaesthesia. We are specifically focused on analgesic and side effect profiles. Methods: We searched databases and trial registration sites and include here randomised controlled trials that compare the analgesic effects of caudal clonidine vs. morphine as adjuvants on postoperative pain. The risk ratio for evaluating pain scores, the need for rescue analgesia and all adverse effects were assessed. The i2 statistic was used to assess heterogeneity. We also assessed risk of bias with Cochrane's Collaboration tool. The quality of evidence was assessed with Grading of Recommendations Assessment, Development and Evaluation (GRADE) system. Results: Four randomised controlled trials (including 166 patients) that evaluated the use of clonidine vs. morphine as adjuvants in caudal block were included in this systematic review and meta-analysis. The pooled estimate for postoperative analgesia revealed no statistically significant differences between the clonidine group compared to morphine group (MD=2.90; 95% CI 4.05 to 9.85; i2 93%). Significantly less postoperative nausea and vomiting were reported among the patients that received clonidine vs. those that were treated with morphine (RR 0.57, 95% CI -0.36 to -0.90, i2 26%). There were no statistically significant differences between the two groups in assessments that included urinary retention, pain scores or need for rescue analgesia at 24 hours. Conclusion: Clonidine is just as effective as morphine when used an adjuvant to local anaesthetic for caudal block, and has a more desirable side effect profile, particularly with respect to postoperative nausea and vomiting.
Article
Full-text available
Objective To compare the efficacy of hydromorphone and dexmedetomidine at inducing emesis in cats. Design Prospective, blinded, randomized crossover study. Setting Veterinary university teaching hospital. Animals 12 healthy purpose‐bred cats. Interventions Cats were randomly assigned to receive hydromorphone (0.1 mg/kg, subcutaneously) or dexmedetomidine (7 μg/kg, IM). Following administration, the incidences of emesis, number of emetic events, signs of nausea (hypersalivation, lip licking), temperature, heart rate, respiratory rate, and sedation score were recorded for 6 hours. Measurements and Main Results Emesis was successful in 9 of 12 (75%) cats when treated with hydromorphone and in 7 of 12 (58%) cats when treated with dexmedetomidine (P = 0.67). Dexmedetomidine was more likely to cause sedation than hydromorphone (P < 0.001). Heart rate in cats was significantly decreased at 1 and 2 hours post‐hydromorphone (P = 0.003, 0.014, respectively) and at 1, 2, 3, 5, 6 hours post‐dexmedetomidine (P = 0.001, 0.003, 0.038, 0.013, 0.001, respectively). Cats were more likely to develop an increase in body temperature with hydromorphone administration although this was not clinically significant. Conclusions Results of the present study indicate that hydromorphone is an effective alternative to dexmedetomidine for the induction of emesis in cats. Hydromorphone appears to cause less sedation and less decrease in heart rate. Further investigation into the most adequate dose of hydromorphone for optimizing emesis is warranted.
Article
Full-text available
Objectives: α2-adrenergic receptors are reportedly involved in cancer cell proliferation, invasion, and apoptosis through regulation of diverse molecules, which implies that it contributes to tumor progression. However, the functional significance of α2-adrenergic receptors in osteosarcoma (OS) is unclear. Tizanidine hydrochloride (THC), an α2-adrenergic receptor agonist, is often used to alleviate symptoms of spasticity. This study investigated the functional implications of THC treatment on human OS cells and the underlying mechanisms of resulting changes. Methods: The proliferation of U2 OS cells was assessed by Cell Counting Kit-8; the migration and invasion capacities of U2 OS cells were then analyzed by transwell assay. Moreover, apoptosis in U2 OS cells was evaluated by flow cytometry and western blot analyses. Additionally, expression levels of key proteins in the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) signaling pathway were measured. Results: THC inhibited the proliferation, migration, and invasion of U2 OS cells, but promoted apoptosis within these cells. Expression levels of p-AKT, p-mTOR, and p-P70S6K were reduced by exposure to THC, suggesting involvement of PI3K/AKT signaling in THC-induced cytotoxicity within OS cells. Conclusions: THC may play a novel role in OS cell cytotoxicity, and these findings suggest a potent therapeutic strategy for OS treatment.
Article
Clonidine and dexmedetomidine are two α2-adrenoreceptors agonists available for the intensivist in the clinical practice. The affinity of dexmedetomidine is eight times greater than clonidine affinity for the α2 receptors. Their main effect is sedation. They act by inhibition of noradrenaline release in the locus coeruleus in the brainstem. α2-agonists are used primarily for sedation, analgesia, and management of delirium. Nowadays, dexmedetomidine application is increasing in critically ill patients showing a good safety. Most frequent side effects include bradycardia and hypotension.
Article
Purpose To review the efficacy and safety of transitioning from dexmedetomidine to clonidine to facilitate weaning of patients from sedation with dexmedetomidine. There is a paucity of data describing dexmedetomidine withdrawal syndrome (DWS) as well as clonidine’s place in therapy for DWS. This review will describe and analyze current literature to provide clinical recommendations. Summary A MEDLINE literature search was performed to identify original research articles describing DWS and/or transitioning from dexmedetomidine to clonidine for the purpose of weaning patients from sedation with dexmedetomidine. Four case reports describing DWS, 3 case reports describing the use of clonidine to treat DWS, and 3 observational studies describing the use of clonidine to facilitate dexmedetomidine weaning were identified. The incidence of and risk factors for DWS are unknown; factors including patient age and dexmedetomidine infusion rate, loading dose, and discontinuation strategy have inconsistent associations with DWS. All cases of DWS have been associated with infusion durations greater than 72 hours. While there are limited data describing clonidine use for the treatment of dexmedetomidine withdrawal, clonidine appears to be beneficial for dexmedetomidine weaning and its use for that purpose has been well described. Clonidine dosages that have been assessed for discontinuing dexmedetomidine vary from 0.1 to 0.3 mg orally or enterally every 6 to 8 hours; one study assessed use of transdermal clonidine (100 µg/24 h patch). Patients with extensive cardiac comorbidities may be more susceptible to adverse effects of clonidine, which may limit the drug’s use for DWS intervention. Conclusion Despite limited supportive data, clonidine provides a promising option for sedation management in adult ICU patients, with successful transitions from dexmedetomidine reported within 24 hours after clonidine initiation.
Article
Objective To determine the effect of aquapuncture at acupuncture point Pericardium 6 (PC-6) on the incidence of dexmedetomidine-induced vomiting and nausea in cats. Study design Randomized, prospective, crossover study. Animals A group of 22 cats, 14 females and eight males, aged 1–12 years and weighing 3.8–5.9 kg. Methods Each cat was administered treatments in random order at ≥1 week intervals. For treatment (DEX–A), cats were administered PC-6 stimulation by aquapuncture (0.25 mL/250 μg vitamin B12 injection subcutaneously at PC-6). After 30 minutes, dexmedetomidine (10 μg kg–1) was administered intramuscularly (IM). For control treatment (DEX), cats were administered only dexmedetomidine (10 μg kg–1) IM. Incidence of vomiting, number of vomiting episodes and time to first vomiting were recorded by an observer unaware of treatment allocation. At 30 minutes after dexmedetomidine administration, atipamezole (0.1 mg kg–1) was injected IM. Behavior was video recorded and later scored by two observers for clinical signs of nausea. A regression model (analysis of covariance) was used to detect the influence of aquapuncture on vomiting and nausea. Significance was set at p < 0.05. Results Of 21 cats, 18 (85%) and 16 cats (76%) vomited in DEX–A and DEX, respectively. There was no significant difference in the incidence of vomiting (p = 0.55), number of vomiting episodes (p = 0.55), mean time to vomit (p = 0.88) or nausea score (p = 0.51) between DEX–A and DEX. Conclusions and clinical relevance PC-6 aquapuncture did not reduce the incidence of dexmedetomidine-induced vomiting or severity of nausea in cats.