Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

| |

Received: 5 May 2021    Revised: 25 June 2021    Accepted: 30 June 2021

DOI: 10.1111/jfbc.13863

ORIGINAL ARTICLE

Menthol, a bioactive constituent of Mentha, attenuates motion


sickness in mice model: Involvement of dopaminergic system

Uma Maheswari Deshetty | Anand Tamatam  | Mahantesh Mallikarjun Patil

Nutrition, Biochemistry & Toxicology


Division, Defence Food Research Abstract
Laboratory, Mysore, India Motion sickness (MS) occurs due to contradicting vestibular and visual inputs to the
Correspondence brain causing nausea and vomiting. Antidopaminergic drugs being effective in re-
Anand Tamatam, Nutrition, Biochemistry & ducing MS create a path for effective therapy against MS by regulating dopamine
Toxicology Division, Defence Food Research
Laboratory, Siddarthanagar, Mysore 570011, levels. We aimed to evaluate the role of the striatum and brainstem dopamine and
India. dopamine D2 receptor (DRD2) in MS and the efficacy of menthol (MNT) to modulate
Email: anand@dfrl.drdo.in
dopamine and DRD2 in vitro and in vivo for possible amelioration of MS. Evaluation
of efficacy of MNT to inhibit dopamine release from PC12 cells and anti-­MS efficacy
in BALB/c mice model was performed. Dopamine, DRD2 expression in PC12 cells,
mice striatum, and brainstem were detected using HPLC-­ECD, RT-­PCR, and Western
blot analysis, respectively. DRD2 expression increased in calcium ionophore-­treated
PC12 cells compared with control cells. Pretreatment with 50 μg/ml menthol de-
creased dopamine and DRD2 expression. Similarly, dopamine and DRD2 levels in
mice striatum and brainstem of MS group (rotation induced) increased significantly
compared with control group NC (no rotation). Pretreatment with menthol at 50 mg/
kg concentration (rotation induced) showed decreased dopamine and DRD2 expres-
sion, thus indicating ameliorative effect on MS. Hence, we suggest that increased
striatum and brainstem dopamine and DRD2 levels might lead to MS symptoms, and
menthol could be used as a potent herbal alternative medicine for MS.
Practical applications
1. Antidopaminergic drugs being effective in reducing motion sickness (MS) creates
a path for effective therapy against MS by regulating dopamine levels.
2. Increased striatum and brainstem dopamine and Dopamine D2 receptor (DRD2)
levels might lead to the MS symptoms induced by rotation stimulation in mice
model.
3. Menthol showed a prophylactic effect on rotation-­induced MS by reducing stri-
atal and brainstem dopamine levels, DRD2 mRNA, and protein expression.
4. Menthol could be used as an herbal alternative to antidopaminergics to minimize
the associated adverse effects.

KEYWORDS

brainstem, dopamine, dopamine D2 receptor, menthol, motion sickness, striatum

J Food Biochem. 2021;45:e13863. wileyonlinelibrary.com/journal/jfbc |


© 2021 Wiley Periodicals LLC.     1 of 11
https://doi.org/10.1111/jfbc.13863
|
2 of 11       DESHETTY et al.

1 |  I NTRO D U C TI O N Menthol [5-­m ethyl-­2-­(1-­m ethyl ethyl) cyclohexanol; 2-­


isopropyl-­5-­m ethylcyclohexanol or p-­m ethan-­3-­ol, MNT] is a nat-
Motion sickness (MS) is described by a set of autonomic symptoms urally occurring cyclic monoterpene alcohol majorly found in the
which is caused by incongruent sensory inputs under conditions Mentha species. Presently, MNT is widely used in the cosmetic
of motion (Koch et al., 2018). Neural mismatch theory explains industry, pharmaceuticals, oral hygiene products, confectionery,
that the intersensory contradiction between visual and vestib- pesticides, and as a flavoring agent (Oz et al., 2017). Regarding
ular systems initiates the release of acetylcholine and histamine its medicinal uses, MNT-­
containing medications are currently
which activates the vestibular nuclei in the brainstem leading to available for conditions such as gastrointestinal disorders, respi-
nausea and vomiting (Reason, 1978; Takeda et al., 2001). This the- ratory diseases, musculoskeletal pain, and common cold (Patel
ory was elucidated based on evidence that antihistaminics and et al., 2007). MNT is known to exert potent antiemetic properties
anticholinergics are the widely used medications for MS (Koch by acting on the 5HT3 receptor by binding to a modulatory site
et al., 2018). (Heimes et al., 2011). MNT is also known to reduce the dopamine
It is reported that dopamine is the key biomarker involved in neuron firing frequency in the midbrain (Henderson et al., 2016).
the etiology of nausea and vomiting and subsequently dopamine Hence, the present study was aimed to evaluate the role of the
receptor D2 (DRD2) antagonists such as metoclopramide, dom- striatum and brainstem dopamine and DRD2 levels in MS and the
peridone, and sulpiride are being used as antiemetics for chemo- efficacy of MNT to modulate dopamine and DRD2 levels in vivo
therapy and apomorphine-­i nduced emesis (Carpenter et al., 1984; for the possible amelioration of MS.
Leslie et al., 1990; Sakata et al., 1986). Nausea and vomiting are
the prominent symptoms of MS which paved the way to the hy-
pothesis that antidopaminergics might be effective against MS. 2 | M ATE R I A L S A N D M E TH O DS
However, certain reports stated against the involvement of do-
pamine and DRD2 in the motion-­induced emesis and also the in- 2.1 | Chemicals and reagents
effectiveness of metoclopramide against MS (Kohl, 1987; Takeda
et al., 1993). Contrary to this, it was reported that dopamine re- Menthol, dopamine, calcium ionphore (A23187), metoclopramide, horse
ceptor antagonists are effective in reducing MS in animals but not serum, fetal bovine serum (FBS), RPMI-­1640, penicillin-­streptomycin
in humans (Lucot, 1998). solution, 3-­(4,5 dimethylthiazol-­2-­yl)-­2,5diphenyltetrazolium bromide
However, it was established that DRD2 antagonists metoclo- (MTT), EDTA, citric acid, triethylamine, DL-­10-­camphorsulfonic acid,
pramide and sulpiride reduced MS symptoms in human patients saccharin, RIPA lysis buffer, protease cocktail inhibitor, p-­coumaric
and squirrel monkeys, respectively (Miller & Brizzee, 1987; Rubio acid, and luminol were purchased from Sigma Aldrich (St. Louis, MO,
et al., 2011). Moreover, metoclopramide exhibits antiemetic action USA). Methanol, Dimethylsulfoxide (DMSO) and disodium hydrogen
by exhibiting DRD2 antagonism at chemoreceptor trigger zone orthophosphate were purchased from SRL (Mumbai, MH, India).
(CTZ) in central nervous system (CNS) and is also known to show ELISA kits of adrenocorticotropic hormone (ACTH), cortisol, and cor-
antiemetic effect by 5-­hydroxytryptamine receptor 3 (5HT3) an- ticosterone were procured from Enzo Life Sciences Inc. (New York,
tagonism. Since metoclopramide binds more avidly to the DRD2, NY, USA) and Cayman Chemical (Ann Arbor, MI, USA), respectively.
a very high dosage of metoclopramide is required to show an anti- Arginine vasopressin (AVP) ELISA kit was purchased from Elabscience,
emetic effect through 5HT3 antagonism (Holland & Pollock, 2010). Hubei, China. Primary antibody (rabbit antidopamine receptor D2,
Concurrent with this, Takeda et al. (1989) also postulated that the PAA673Mu01) and secondary antibody (HRP-­labeled Goat antirab-
two major pathways of dopaminergic nervous system, mesolimbic bit IgG) were purchased from Cloud-­Clone Corp. (Houston, TX, USA).
and nigrostriatal pathways, might be involved in MS. Therefore, it SV total RNA isolation system and High-­capacity cDNA reverse tran-
might be postulated that the antiemetic effect of dopamine receptor scription kits were procured from Promega (Madison, WI, USA) and
antagonists might be by affecting the DRD2 through the nigrostria- Applied Biosystems (Waltham, MA, USA), respectively.
tal or mesolimbic pathway. Hence, the role of dopamine, DRD2 and
DRD2 antagonists in motion-­induced emesis is not clearly defined
and needs to be elucidated. 2.2 | In vitro analysis
Additionally, it is also known that though potential antiemetics
DRD2 antagonists, such as metoclopramide and sulpiride, are consid- 2.2.1 | Cell maintenance and pretreatment
ered to be effective against MS, these are accompanied by adverse
side effects, such as sedation, restlessness, orthostatic hypotension, Rat pheochromocytoma (PC12) cells were procured from National
headache, confusion, focal dystonia, and tardive dyskinesia, which Centre for Cell Sciences, Pune, India. PC12 cells were grown in an
limits their long-­term use among travelers (Baldessarini, 1990; Flake RPMI-­1640 growth medium supplemented with 10% horse serum,
et al., 2004). Hence, herbal extracts and plant-­derived bioactive 5% FBS, penicillin (100 U/ml), and streptomycin (100 µg/ml). Cells
compounds have gained attention as an alternative medicine for possessing 95% cell viability were utilized for experiments. Cells
managing MS.
DESHETTY et al. |
      3 of 11

were pretreated with different concentrations of MNT for 1 hr in mice (20–­25  g) were obtained from Central Animal House Facility,
serum-­free media. DFRL, Mysore. Mice were housed in polycarbonate cages at 25 ± 2℃,
12 hr light/dark cycle, and fed with water and pellets ad libitum.

2.2.2 | Cell viability assay


2.3.2 | Induction of MS
Cell viability of PC12 cells was evaluated by MTT assay. Cells were
seeded in 24-­well plate at a density of 1 × 106 cells/well and grown for Grouping
24 hr. Treatment of cells with MNT (20–­140 µg/ml concentration range) Thirty-­six BALB/c mice (female) were randomly divided into six
was carried out and incubated for 24  hr. After treatment, MTT was groups (n  = 6) such as normal control (NC, devoid rotation), MNT
added and assay performed according to the method described in our at 50 mg/kg body weight control (MNT50, devoid rotation), motion
previous study (Deshetty et al., 2020). The concentration of MNT pro- sickness (MS, induced rotation), metoclopramide (standard drug) at
viding 50% inhibition (IC50) was evaluated from the graph of concentra- 20 mg/kg body weight + rotation (MCD + MS), MNT at 25 mg/kg
tion of MNT versus inhibition percentage in a dose-­dependent manner. body weight + rotation (MNT25 + MS), and MNT at 50 mg/kg body
weight + rotation (MNT50 + MS).

2.2.3 | Lactate dehydrogenase (LDH) leakage assay Rotation device


MS was induced in mice by clockwise rotation at an angular velocity
PC12 cells were seeded, grown in 24-­well plate, and were treated of 50 rpm for 30 min using a custom-­designed centrifuge machine
with 20 µg/ml–­140  µg/ml concentration range of MNT. After 24 hr, with the specifications as described in our previous study (Deshetty
LDH activity was estimated and expressed as U/L using the method et al., 2020).
demonstrated in our prior study (Deshetty et al., 2020).
Experimental procedure
Conditioned taste aversion (CTA) method involving the usage of sac-
2.2.4 | Measurement of dopamine release charin solution (SS, 0.15%) was used as an index of MS. The method
of two bottle choice test (TBCT), which includes a choice between
PC12 cells (1 × 106 cells/well) were grown in 6-­well plate for 24 hr. The saccharin solution (SS) and drinking water (DW) being provided
growth medium was replenished with serum-­free media after 24 hr. to mice (Ossenkopp & Frisken, 1982). The experimental period of
Treatment of cells with different concentrations of MNT (12.5 µg/ml 21 days was divided into two phases (conditioning phase and test
[12.5 MNT], 25 µg/ml [25 MNT], and 50 µg/ml [50MNT]) was carried phase) as described in our prior study (Deshetty et al., 2020).
out, incubated for 1 hr followed by replacing the culture medium
with stimulation buffer containing 5 µM of A23187. Cells were in- Conditioning phase. Mice were conditioned to the water deprivation
cubated for 2 hr at 37℃ with 5% CO2 (Maheswari et al., 2020). Cell schedule of 4 hr until the 7th day in a manner similar to our earlier
supernatant was collected and centrifuged for 5 min at 8,000 rpm. study, with the exception of rotation provided to groups MS,
0.4 N perchloric acid was added to the cell supernatant and filtered MCD + MS, MNT25 + MS, and MNT50 + MS (Deshetty et al., 2020).
through 0.22 µm filter (Bieger et al., 2002).
Dopamine released in the cell supernatant was estimated using Test phase. In this phase, mice were provided with 4 hr water
HPLC (Electrochemical detector) as described in our previous study deprivation schedule, which included oral intubation with MCD or
(Maheswari et al., 2020). The concentration of dopamine in cell su- MNT at 3 hr of the schedule. After 4 hr, mice were provided access
pernatant samples was evaluated by comparing the peak area of the to SS and DW for 1 hr. This was immediately followed by rotation
samples with the peak area of standard dopamine. The concentra- (30 min, 50 rpm) to mice from MS, MCD + MS, MNT25 + MS, and
tion of dopamine was expressed as ng/ml of sample. MNT50 + MS groups. Again, mice were given access to DW/SS and
consumption was measured for the next 2 hr (Deshetty et al., 2020).

2.3 | In vivo study


2.3.3 | Evaluation of behavioral parameters
2.3.1 | Animals
Determination of consumption of saccharin solution/drinking water
Animal study was conducted as per the guidelines of Committee The volume of SS/DW consumed by mice belonging to different groups
for the Purpose of the Control and Supervision of Experiments on before and after rotation was calculated using the following formula:
Animals (CPCSEA). This experiment was approved by the DFRL IAEC
(Institutional Animal Ethical Committee), approval number (IAEC-­ Volume SS∕DW intake before MS induction
% SS∕DWintake = × 100
2016/BN/1) and comply with ARRIVE guidelines. Female BALB/c Volume SS∕DW intake after MS induction
|
4 of 11       DESHETTY et al.

Behavioral scoring test pellet was lysed using the RNA lysis buffer. Mice brainstem and
The behavioral scoring test was performed after 30 min of rotation striatum samples were lysed using RNA lysis buffer. Total RNA
as per the method described by Yu et al. (2007) with minor modi- was extracted and converted to cDNA as described in our pre-
fications. The sum of all scores assigned to each symptom was ex- vious study using the specific kits. Housekeeping gene used
pressed as the MS index (Deshetty et al., 2020). for comparison was β-­a ctin. Primers targeting the DRD2 gene
in both rat PC12 cells and mice striatum and brainstem sam-
Open field test ples were KiCqStart primers purchased from Sigma Aldrich (St.
Open field test (OFT) known as an animal behavioral test was per- Louis, MO, USA). The primer sequences used for RT-­P CR were
formed to detect the spontaneous locomotor activity of mice after rat DRD2: sense primer 5′-­C AACAATACAGACCAGAATGAG-­3′,
30 min of rotation (Brown et al., 1999). The specifications of the ap- antisense primer 5′-­G GAGGACGATGTAGATTTTG-­3′; mouse
paratus used and the protocol followed are as described in our previ- DRD2: sense primer 5′-­T TGTTCTTGGTGTGTTCATC-­3′, anti-
ous study (Deshetty et al., 2020). sense primer 5′-­TATAGATGATGGGGTTCACG-­3′; rat β-­a ctin:
Mice were sacrificed instantly after rotation on the 21st day of sense primer 5′-­A AGACCTCTATGCCAACAC-­3′, antisense
the experimental protocol. Brain and blood samples were collected primer 5′-­TGATCTTCATGGTGCTAGG-­3′; and mouse β-­a ctin:
for analysis. Mice were anesthetized and blood was collected retro-­ sense primer 5′-­G TCAAGATCATTGCTCCTC-­3′, antisense primer
orbitally followed by cervical dislocation to confirm euthanasia. Brain 5′-­T TGTCAAGAAAGGGTGTAAC-­3′. The PCR reaction mix and
was harvested out and further microdissected to obtain specific stri- reaction cycles were set up as mentioned in our previous study
atum and brainstem regions. However, the test was performed on using DRD2 primers. Changes in gene expression were expressed
ΔΔCt
every alternate day in the test phase for six times which also involved as mean fold change by using the 2-­ method (Deshetty
three sessions of OFT that was conducted immediately after the et al., 2020).
rotation to avoid adaptation to MS. Therefore, a sufficient gap was
provided between the experimental trials to prevent the adaptation
process. Data were represented by calculating an average of all the 2.3.7 | Western blot analysis of Dopamine D2
values from the experimental trials performed in the test phase. receptor (DRD2) protein

PC12 cells (1 × 106) were seeded 25 cm2 flasks and treated with MNT
2.3.4 | Estimation of dopamine levels as described earlier. The cell pellet, brainstem, and striatum samples
were homogenized in protease cocktail inhibitor-­containing RIPA
The brainstem and striatum samples were homogenized in 0.9% per- lysis buffer followed by centrifugation (10,000× g, 4℃) for 20 min.
chloric acid, centrifuged for 15 min at 10,000 rpm. Supernatants were The protein in cells and tissue homogenates was determined (Lowry
separated, filtered with 0.22-­μm filters, and the filtrate was analyzed et al., 1951). Protein sample (50 µg), primary antibodies (GAPDH and
for dopamine levels using HPLC analysis (Alburges et al., 1993). The DRD2), and secondary antibody (HRP-­labeled Goat antirabbit IgG)
concentration of dopamine in the samples was calculated by com- were used to perform the western blotting analysis, and band in-
paring the peak area of the samples with the peak area of standard tensity was measured as mentioned in our previous study (Deshetty
dopamine and expressed as ng/g tissue. et al., 2020).

2.3.5 | Estimation of stress hormones 2.4 | Statistical analysis

The levels of stress hormones, such as ACTH (Enzo Life Sciences Inc., The in vitro data were represented as mean ±  SEM of re-
New York, NY, USA), cortisol and corticosterone (Cayman Chemical, sults calculated from three independent experiments con-
MI, USA) in plasma samples, were estimated using a 96-­well plate ducted in triplicates, respectively. The in vivo data were
ELISA kit. Plasma AVP was detected using 96-­well plate ELISA kit represented as the mean ±  SD of results attained from three
(Elabscience, Hubei, China). The assays were performed as per the experimental trials (n  =  6), respectively. GraphPad Prism ver-
instructions of the manufacturer and the concentration of stress sion 6 software (LaJolla, CA, USA) was used for the statistical
hormones was calculated and represented as pg/ml plasma samples. analysis. The MS group was compared with NC group (control)
and MCD + MS, MNT25 + MS, MNT50 + MS groups were
compared with the MS group. Data were analyzed by one-­
2.3.6 | Total RNA Isolation, cDNA synthesis, and RT-­ way ANOVA and comparisons among different groups were
PCR analysis made using Tukey's multiple comparisons test, where *p  < .05,
**p  < .01, ***p  < .001 compared with NC group and #p  < .05,
PC12 cells (1 × 10 6) were seeded and grown in 25 cm2 flasks. ##
p  < .01, ###
p  < .001 compared with MS group was considered
Cells were treated with MNT as described earlier and the cell as significant.
DESHETTY et al. |
      5 of 11

3 | R E S U LT S 1.103  ± 0.069 fold (25 MNT), and 0.71 ± 0.041 fold (50 MNT) in
comparison with A23187-­treated PC12 cells (Figure  2a). Increased
3.1 | In vitro analysis DRD2 protein expression was detected in A23187-­treated PC12
cells (215.34% ± 9.33%) compared with control PC12 cells (100%).
3.1.1 | Effect of MNT on PC12 cell viability PC12 cells pretreated with MNT showed significantly reduced the
DRD2 protein expression in 12.5 MNT (182.62% ± 6.17%), 25 MNT
The IC50 value of MNT in PC12 cells was 100 µg/ml (Figure 1a). (169.26% ± 7.23%), 50 MNT (124.87% ± 8.11%) compared with PC12
Based on the IC50 value, the concentration of MNT was further fixed cells treated with A23187 (Figure 2b,c).
for dopamine release assay.

3.2 | In vivo analysis


3.1.2 | Effect of MNT on LDH leakage
3.2.1 | Effect of MNT on drinking water (DW)/
Control PC12 cells released 30.85 ± 4.11 U/L LDH and MNT treated saccharin solution (SS) consumption
PC12 cells released between 56.99 ± 8.11 and 289.46 ± 11.18 U/L.
The LDH release increased with increase in the MNT concentration The SS consumption of mice from MS group (16.54% ± 0.95%) de-
(Figure 1b). This in turn indicates the cytotoxicity of MNT at differ- creased compared with the control groups NC (26.24% ± 0.58%)
ent concentrations and IC50 value. and MNT50 (25.69% ± 0.47%). Pretreatment with MCD + MS
(25.11% ± 0.68%), MNT25 + MS (20.32% ± 0.93%), and MNT50 + MS
(24.92%  ± 0.65%) increased the SS in comparison with MS group
3.1.3 | Effect of MNT on dopamine release from (Figure 3a).
PC12 cells

Control PC12 cells released 47.28 ± 4.99 ng/ml, whereas the cal- 3.2.2 | Effect of MNT on behavioral scores
cium ionophore (A23187)-­treated cells released 377.43 ± 10.16 ng/
ml. Cells pretreated with 50 MNT exhibited inhibition in dopamine The behavioral score exhibited by mice from the MS group
release to maximum extent (57.12 ± 9.12 ng/ml) compared with (16.60 ± 1.12) was higher than the control groups NC (2.2 ± 0.68)
12.5 MNT (165.78 ± 11.11 ng/ml) and 25 MNT (64.50 ± 8.12 ng/ and MNT50 (2.20 ± 0.62). The behavioral score of mice from the pre-
ml) (Figure 1c). treated groups MCD + MS (4.8 ± 0.66), MNT25 + MS (8.34 ± 0.72),
and MNT50 + MS (5.20 ± 0.56) reduced in comparison with MS
group (Figure 3b).
3.1.4 | Effect of MNT on DRD2 mRNA and protein
expression in PC12 cells
3.2.3 | Effect of MNT on open field test scores
PC12 cells treated with A23187 exhibited upregulated DRD2
mRNA expression by 2.074  ± 0.049 folds in comparison with the Mice belonging to the MS group exhibited significantly increased
PC12 control cells. PC12 cells pretreated with MNT showed re- freezing duration and decreased rearing, number of line cross-
duced DRD2 mRNA expression by 1.30  ± 0.061 fold (12.5 MNT), ings, and total distance traveled compared with the control groups

F I G U R E 1   Dose-­dependent effect of menthol pretreatment on (a) PC12 cell viability, (b) extracellular LDH leakage from PC12 cells (U/L),
(c) concentration of dopamine release from PC12 cells (ng/ml). Values are expressed as mean ± SEM of three independent experiments.
***p < .001 compared with PC12 control cells; ##p < .01 and ###p < .001 compared with A23187 treated cells
|
6 of 11       DESHETTY et al.

F I G U R E 2   Effect of menthol on (a) DRD2 mRNA expression in PC12 cells, (b,c) DRD2 protein expression in PC12 cells. Values are
expressed as mean ± SEM for three trials. **p < .01 compared with control PC12 cells; #p < .05, ##p < .01, and ###p < .001 compared with
A23187-­treated PC12 cells

F I G U R E 3   (a) Volume (%) of saccharin solution (SS) and drinking water (DW) consumed per mice after rotation of 30 min; (b) behavioral
score exhibited by mice after rotation. Values are expressed as mean ± SD for six mice per group. *p < .05 and **p < .01 compared with NC
group; #p < .05, ##p < .01, and ###p < .001 compared with MS group

(NC and MNT50). Mice from the pretreatment groups MCD + MS, 3.2.5 | Effect of MNT on stress hormone levels
MNT25 + MS, MNT50 + MS showed decreased freezing ­duration and
significantly increased rearing, number of line crossings, and total AVP, ACTH, cortisol, and corticosterone levels in plasma samples
distance traveled. The behavioral pattern was similar to the control were increased in mice from the MS group compared with NC and
groups, indicating the amelioration of MS (Figure 4). MNT50 (control groups). Pretreatment with MCD and MNT reduced
the levels in the plasma samples and were comparable with control
groups (Table 1).
3.2.4 | Effect of MNT on dopamine levels

Dopamine levels were increased in the striatum sample of MS group 3.2.6 | Effect of MNT on DRD2 mRNA expression
(158.62 ± 8.68) compared with the control groups NC (72.15 ± 7.99)
and MNT50 (70.55 ± 8.56). Pretreatment with MCD + MS DRD2 mRNA expression in striatum samples of MS group was up-
(73.68  ± 7.92), MNT25 + MS (111.73 ± 7.02), and MNT50 + MS regulated by 2.040 ± 0.058 folds compared with control group.
(76.11 ± 7.54) exhibited decreased dopamine levels compared with Expression was downregulated in MCD + MS (0.51 ± 0.091),
the MS group. Brainstem dopamine levels in NC, MNT50, and MS MNT25 + MS (1.11 ± 0.08), MNT50 + MS (0.75 ± 0.032) compared
groups were 50.22 ± 8.89, 48.22 ± 9.18, and 139.69 ± 9.68, respec- with the MS group. The brainstem DRD2 mRNA expression got
tively. Pretreatment with MCD + MS (59.11 ± 7.45), MNT25 + MS upregulated in MS group by 3.11 ± 0.092 folds compared with the
(102.46 ± 6.99), and MNT50 + MS (73.11 ± 7.54) reduced the dopa- control group. Whereas, in pretreatment groups, expression was de-
mine levels (Figure 5a). creased by MCD + MS (0.73 ± 0.083), MNT25 + MS (1.54 ± 0.095),
DESHETTY et al. |
      7 of 11

F I G U R E 4   Open field test scores exhibited by mice from different groups after 30 min rotation. (a) Number of line crossings, (b) rearing,
(c) total distance traveled (cm), and (d) freezing duration (s). Values are expressed as mean ± SD for six mice per group. *p < .05, **p < .01, and
***p < .001 compared with NC group; #p < .05, ##p < .01, and ###p < .001 compared with MS group

F I G U R E 5   Effect of MNT on (a) dopamine levels (ng/g tissue) and (b) DRD2 mRNA expression in striatum and brainstem samples of mice.
Values are expressed as mean ± SD for six mice per group. **p < .01 and ***p < .001 compared with NC group; ##p < .01 and ###p < .001
compared with MS group

MNT50  + MS (0.83 ± 0.05) folds compared with the MS group pretreated groups exhibited decreased DRD2 expression, MCD + MS
(Figure 5b). (130.46% ± 8.32%), MNT25 + MS (161.55% ± 7.11%), MNT50 + MS
(135.34%  ± 7.56%), compared with the MS group. The brainstem
DRD2 protein expression was increased to 196.28% ± 8.75% in the
3.2.7 | Effect of MNT on DRD2 protein expression MS group compared with the control group (100%). The pretreated
groups exhibited decreased DRD2 protein expression, MCD + MS
Increased striatum DRD2 protein expression (185.12% ± 6.45%) was (135.62% ± 9.44%), MNT25 + MS (165.12% ± 9.46%), MNT50 + MS
observed in the MS group compared with the control group (100%). The (143.06% ± 8.09%), compared with the MS group (Figure 6).
8 of 11       | DESHETTY et al.

TA B L E 1   Stress hormone levels in


Corticosterone
plasma samples of mice (pg/ml sample)
Groups AVP (pg/ml) ACTH (pg/ml) Cortisol (pg/ml) (pg/ml)

NC 23.29 ± 3.86 101.61 ± 6.42 168.63 ± 9.51 96.10 ± 8.63


MNT50 22.88 ± 3.72 102.67 ± 4.66 172.43 ± 5.44 100.02 ± 7.51
MS 81.63 ± 6.43* 198.86 ± 8.11* 384.94 ± 5.98** 208.44 ± 6.56*
MCD + MS 35.67 ± 5.11###  121.22 ± 4.98##  210.34 ± 5.66 ##  120.38 ± 5.11## 
# ## #
MNT25 + MS 52.58 ± 4.89   153.23 ± 7.11   248.34 ± 6.12   143.67 ± 5.12# 
MNT50 + MS 46.34 ± 5.11##  136.37 ± 4.18##  218.27 ± 3.84 ##  128.41 ± 4.48## 

Note:: Values are expressed as mean ± SD for six mice per group.
Abbreviations: ACTH, adrenocorticotropic hormone; AVP, arginine vasopressin.
*p < .05,; **p < .01 compared with NC group;
#
p < .05,
##
p < .01,
###
p < .001 compared with MS group.

F I G U R E 6   Effect of MNT on DRD2 protein expression in striatum and brainstem samples of mice. Values are expressed as mean ± SD for
six mice per group. **p < .01 compared with NC group; #p < .05 compared with MS group

4 |  D I S CU S S I O N role of dopamine, DRD2, DRD2 antagonists in MS is obscure and


needs to be explored. Few reports suggested the noninvolvement
Antihistaminics and anticholinergics are the widely used class of of dopamine and DRD2 in motion-­induced nausea and vomiting due
drugs for MS as histamine and acetylcholine elevations are consid- to the ineffectiveness of DRD2 antagonists such as metoclopramide
ered to be the primary reason for MS as described by sensory conflict (Kohl, 1987; Takeda et al., 1993). However, contrary to these antido-
theory (Koch et al., 2018). Dopamine has been considered to play a paminergics such as DRD2 antagonists (metoclopramide, sulpiride)
role in etiology of nausea and vomiting induced by chemotherapy, are reported to reduce MS symptoms in animals and humans (Miller
postoperative, drug (apomorphine) as DRD2 antagonists are effec- & Brizzee, 1987; Rubio et al., 2011). Concomitant with this, Takeda
tive by acting on DRD2 at chemoreceptor trigger zone (CTZ) in area et al. (1989) hypothesized that the mesolimbic or nigrostriatal dopa-
prostema (Holland & Pollock, 2010; Smith et al., 2012). However, the minergic pathway might be involved in MS. In view of this, dopamine
DESHETTY et al. |
      9 of 11

might be considered as an important biomarker and its role in MS and defecation in MS group which resembles earlier published
needs to be elucidated by exploring the role of dopamine, DRD2, reports (Qi et al., 2019; Wang et al., 2017; Zheng et al., 2014;
and DRD2 antagonist via the nigrostriatal or mesolimbic pathway. Zhou et al., 2017). Pretreatment with MCD and MNT exhibited
However, it is known that though DRD2 antagonists are potential a reduction in the score of the symptoms (Figure 3b). Rotation
antiemetic and considered effective against MS, it causes side ef- significantly decreased the rearing, total distance traveled, num-
fects, such as sedation, restlessness, orthostatic hypotension, ber of line crossings and increased the freezing duration in the
headache, confusion, tardive dyskinesia, and focal dystonia, which MS group compared with the control groups (NC and MNT50),
limits their long-­term use among travelers (Baldessarini, 1990; Flake which is in accord with a prior report (Wang et al., 2012). Whereas
et al., 2004). Hence, natural bioactive molecules from plant origin mice belonging to treatment groups (MCD + MS, MNT25 + MS,
are gaining importance to be used as alternative medicine as they MNT50  + MS) exhibited restored spontaneous locomotor activ-
are free from adverse side effects (Hwang et al., 2019). MNT, a cy- ity, determining the protective effect of MCD and MNT at 10 and
clic monoterpene found majorly in Mentha species is known to have 50 mg/kg b.wt. concentration, respectively, on rotation induced
a potent antiemetic property (Heimes et al., 2011; Thawkar, 2016). MS symptoms (Figure 4).
Hence, the present study was performed to evaluate the role of the Concurrent with this, it was found that dopamine and DRD2
striatum and brainstem dopamine and DRD2 in MS and the efficacy mRNA and protein expression in striatum and brainstem regions
of MNT to modulate dopamine and DRD2 levels in vitro and in vivo of the mice from MS group (rotation induced) upregulated signifi-
for possible amelioration of MS. cantly compared with the NC group (control group and no rotation)
PC12 cells can synthesize, store, metabolize, and release dopa- (Figures 5 and 6). This outcome may be correlated to the fact that
mine in a method similar to that observed in vivo (Greene & Rein, histamine release during MS leads to vasopressin release which in
1977). A23187 increases the calcium ions permeability into the cells turn induces stress (Horii et al., 2001; Kjaer et al., 1998; Sharman &
which are essential for the release of dopamine from cells (Garcia Low, 2008). It was found that levels of hormones AVP, ACTH, corti-
et al., 1975). Hence, this could be used as an efficient in vitro model sol, and corticosterone increased in plasma samples of the MS group
for evaluating the effect of MNT by checking the extent of inhibi- compared with the control groups NC and MNT50 (Table 1). This may
tion on dopamine release and DRD2 expression from PC12 cells be correlated to the fact that AVP is an etiological hormone and con-
(Maheswari et al., 2020). It was found that dopamine levels in- tributes to MS development (Xu et al., 2015). AVP, in turn, activates
creased and DRD2 expression was upregulated in cells treated with the hypothalamic-­pituitary-­adrenal (HPA) axis causing the release of
A23187 compared with the control cells. The pretreatment with stress hormones (ACTH, cortisol, and corticosterone) and glucocor-
50MNT (50 μg/ml) decreased both dopamine and DRD2 expres- ticoids which eventually induces stress and promotes the release of
sion to a maximum extent compared with cells treated with A23187 catecholamines such as dopamine (Antoni,  1993; Chrousos,  2009;
(Figures 1 and 2). Therefore, it was established in vitro that MNT Smith & Vale, 2006). It is also reported earlier that induction of stress
could decrease dopamine levels and reduce DRD2 (mRNA and pro- causes the release of dopamine in the striatum, nucleus accumbens,
tein) expression. and medial frontal cortex (Abercrombie et  al.,  1989). Hence, the
Concomitantly, BALB/c mice model was used to evaluate the stress induced due to rotation might be the reason for the release
effect of MCD and MNT on striatal and brainstem dopamine and of dopamine from the striatum which activated the DRD2 receptors
DRD2 levels in MS. CTA and behavioral parameters such as be- in the striatum. It is also known that dopaminergic innervations from
havioral scoring and OFT were used as MS index. In the present striatum extend to the substantia nigra compacta which is present
study, when provided a choice between DW and SS, mice from all in the midbrain (a part of the brainstem) (Pliszka, 2016). These in-
the groups preferred SS rather than DW before rotation. It was ob- nervations might have activated the dopaminergic neurons in the
served that after 30 min of rotation, mice from MS group (rotation brainstem causing the dopamine release and activation of DRD2
induced) and preferred DW avoided SS compared with NC group receptors in the brainstem which in turn activates the vomiting cen-
(control group, no rotation), thus demonstrating aversive behav- ter in the brainstem through DRD2 and causing symptoms of MS.
ior toward SS. This was in agreement with prior published results However, mice from treatment groups MCD + MS, MNT25 + MS,
(Chen et al., 2018; Ossenkopp & Frisken, 1982). Whereas pretreated MNT50  + MS (rotation induced) exhibited decreased stress hor-
groups MCD + MS, MNT25 + MS, MNT50 + MS preferred SS rather mones, dopamine levels, DRD2 mRNA/protein expression compared
than DW. Hence, indicating a protective effect of MCD and MNT at with the MS group (Table 1, Figures 5 and 6).
10 and 50 mg/kg bodyweight concentration, respectively, on rota- Moreover, it has been reported that elevated striatal dopamine
tion induced MS symptoms in mice (Figure 3a). levels further facilitate conditioned avoidance behavior (Cooper
Simultaneously in behavioral scoring tests, specific symptoms et al., 1974). In correlation, we found that mice from the MS group
such as piloerection, urination, tremor, and defecation were ob- (rotation induced) showed conditioned aversive behavior toward SS
served and indexed with a scoring system (Yu et al., 2007). It was (Figure 3a), which could be due to the increased dopamine levels in
found that mice belonging to the MS group exhibited a higher the striatum of these mice (Figure 5a). Further, stress-­induced during
score compared with the control groups NC and MNT50. This rotation stimulation might also be a possible reason for decreased
indicates that rotation increased piloerection, urination, tremor, locomotor activity in the mice belonging to the MS group in the OFT
|
10 of 11       DESHETTY et al.

(Figure 4), as stress is thought to impair vestibular compensation by Biomedical Chromatography, 7, 306–­ 310. https://doi.org/10.1002/
bmc.11300​70605
causing behavioral changes (Yamamoto, 2000). But pretreatment of
Antoni, F. A. (1993). Vasopressinergic control of pituitary adrenocorti-
mice with MCD (10 mg/kg b.wt) and MNT (50 mg/kg b.wt) showed cotropin secretion comes of age. Frontiers in Neuroendocrinology, 14,
the restorative effect on behavioral patterns indicating possible pro- 76–­122. https://doi.org/10.1006/frne.1993.1004
tective effect against MS. Baldessarini, R. J. (1990). Drugs and the treatment of psychiatric disor-
Hence, the present study suggests that increased striatum and ders. Goodman and Gilman's the Pharmacological Basis of Therapeutics,
8, 383–­435.
brainstem dopamine and DRD2 levels might lead to the symptoms of
Bieger, S., Morinville, A., & Maysinger, D. (2002). Bisperoxovanadium
MS induced by rotation stimulation in the BALB/c mice model. We complex promotes dopamine exocytosis in PC12 cells.
also established that MNT showed a protective effect on rotation-­ Neurochemistry International, 40, 307–­314. https://doi.org/10.1016/
induced MS by decreasing the striatal and brainstem dopamine and S0197​- ­0186(01)00093​-­6
Brown, R. E., Corey, S. C., & Moore, A. K. (1999). Differences in measures
DRD2 mRNA, protein levels in a manner almost similar to metoclopr-
of exploration and fear in MHC-­congenic C57BL/6J and B6-­H-­2K
amide. Thus, MNT with no known adverse effects or toxicity at this mice. Behavior Genetics, 29, 263–­271.
dosage could be used as an herbal antidote and alternative medicine Carpenter, D. O., Briggs, D. B., & Strominger, N. (1984). Peptide-­induced
to antidopaminergic medications to minimize the related adverse ef- emesis in dogs. Behavioural Brain Research, 11, 277–­281. https://doi.
org/10.1016/0166-­4328(84)90220 ​-­1
fects and discomfort of travelers. However, the role of dopamine
Chen, M. M., Xu, L. H., Chang, L., Yin, P., & Jiang, Z. L. (2018). Reduction
and DRD2 could also be analyzed real-­time by microdialysis as a part of motion sickness through targeting histamine N-­methyltransferase
of future studies. Also, the effect of MNT on other pathways such in the dorsal vagal complex of the brain. Journal of Pharmacology and
as serotonergic and GABAergic in MS could be explored. However, Experimental Therapeutics, 364, 367–­376.
Chrousos, G. P. (2009). Stress and disorders of the stress system.
further studies in human volunteers are required to determine the
Nature Reviews Endocrinology, 5, 374–­381. https://doi.org/10.1038/
efficacy of MNT to reduce MS. nrendo.2009.106
Cooper, B. R., Howard, J. L., Grant, L. D., Smith, R. D., & Breese, G. R.
AC K N OW L E D G M E N T S (1974). Alteration of avoidance and ingestive behavior after destruc-
tion of central catecholamine pathways with 6-­hydroxydopamine.
The authors extend their deep sense of gratitude to Director,
Pharmacology Biochemistry and Behavior, 2, 639–­ 6 49. https://doi.
Defence Research and Development Organisation-­Defence Food org/10.1016/0091-­3 057(74)90033​-­1
Research Laboratory (DRDO-­DFRL) for his constant encouragement Deshetty, U. M., Tamatam, A., Bhattacharjee, M., Perumal, E., Natarajan,
and support throughout the study. G., & Khanum, F. (2020). Ameliorative effect of hesperidin against
motion sickness by modulating histamine and histamine H1 recep-
tor expression. Neurochemical Research, 45, 371–­ 384. https://doi.
C O N FL I C T O F I N T E R E S T
org/10.1007/s1106​4-­019-­02923​-­0
The authors declare that they have no known competing financial Flake, Z. A., Scalley, R. D., & Bailey, A. G. (2004). Practical selection of
interests or personal relationships that could have appeared to influ- antiemetics. American Academy of Family Physicians, 69, 1169–­1174.
ence the work reported in this paper. Garcia, A. G., Kirpekar, S. M., & Prat, J. C. (1975). A calcium ionophore
stimulating the secretion of catecholamines from the cat adrenal.
Journal of Physiology, 244, 253–­262. https://doi.org/10.1113/jphys​
AU T H O R C O N T R I B U T I O N S iol.1975.sp010795
Uma Maheswari Deshetty: Conceptualization; Data curation; Formal Greene, L. A., & Rein, G. (1977). Release, storage and uptake of catechol-
analysis; Investigation; Methodology; Writing-­original draft. Anand amines by a clonal cell line of nerve growth factor (NGF) responsive
pheochromocytoma cells. Brain Research, 129, 247–­263. https://doi.
Tamatam: Project administration; Supervision; Writing-­review & ed-
org/10.1016/0006-­8993(77)90005​-­1
iting. M M Patil: Software; Validation. Heimes, K., Hauk, F., & Verspohl, E. J. (2011). Mode of action of pepper-
mint oil and (-­)-­menthol with respect to 5-­HT3 receptor subtypes:
DATA AVA I L A B I L I T Y S TAT E M E N T Binding studies, cation uptake by receptor channels and contraction
of isolated rat ileum. Phytotherapy Research, 25, 702–­708.
The data that support the findings of this study are available from
Henderson, B. J., Wall, T. R., Henley, B. M., Kim, C. H., Nichols, W. A.,
the corresponding author upon reasonable request. Moaddel, R., Xiao, C., & Lester, H. A. (2016). Menthol alone upregu-
lates midbrain nAChRs, alters nAChR subtype stoichiometry, alters
ORCID dopamine neuron firing frequency, and prevents nicotine reward.
Journal of Neuroscience, 36, 2957–­ 2974. https://doi.org/10.1523/
Anand Tamatam  https://orcid.org/0000-0003-2387-730X
JNEUR​OSCI.4194-­15.2016
Holland, J. F., & Pollock, R. E. (2010). Holland-­Frei cancer medicine.
REFERENCES PMPH-­USA, 8, 213–­218.
Abercrombie, E. D., Keefe, K. A., DiFrischia, D. S., & Zigmond, M. J. Horii, A., Koike, K., Uno, A., Uno, Y., & Kubo, T. (2001). Vestibular modu-
(1989). Differential effect of stress on in vivo dopamine release in lation of plasma vasopressin levels in rats. Brain Research, 914, 179–­
striatum, nucleus accumbens, and medial frontal cortex. Journal 184. https://doi.org/10.1016/S0006​-­8993(01)02799​-­8
of Neurochemistry, 52, 1655–­ 1658. https://doi.org/10.1111/ Hwang, E., Kuhn, S. M., & Lange, B. (2019). Motion Sickness. In Travel
j.1471-­4159.1989.tb092​24.x Medicine, 2, 423–­428.
Alburges, M. E., Narang, N., & Wamsley, J. K. (1993). A sensitive and rapid Kjaer, A., Larsen, P. J., Knigge, U., Jorgensen, H., & Warberg, J. (1998).
HPLC-­ECD method for the simultaneous analysis of norepinephrine, Neuronal histamine and expression of corticotropin-­releasing hor-
dopamine, serotonin and their primary metabolites in brain tissue. mone, vasopressin and oxytocin in the hypothalamus: Relative
DESHETTY et al. |
      11 of 11

importance of H1 and H2 receptors. European Journal of Endocrinology, Sharman, A., & Low, J. (2008). Vasopressin and its role in critical care.
139, 238–­243. https://doi.org/10.1530/eje.0.1390238 Continuing Education Anaesthesia Critical Care Pain, 8, 134–­137.
Koch, A., Cascorbi, I., Westhofen, M., Dafotakis, M., Klapa, S., https://doi.org/10.1093/bjace​accp/mkn021
& Kuhtz-­B uschbeck, J. P. (2018). The Neurophysiology and Smith, H. S., Cox, L. R., & Smith, B. R. (2012). Dopamine receptor antago-
Treatment of motion sickness. Deutsches Arzteblatt International, nists. Annals of Palliative Medicine, 1, 137–­142.
115, 687–­696. Smith, S. M., & Vale, W. W. (2006). The role of the hypothalamic-­pituitary-­
Kohl, R. L. (1987). Failure of metoclopramide to control emesis or nau- adrenal axis in neuroendocrine responses to stress. Dialogues in
sea due to stressful angular or linear acceleration. Aviation Space and Clinical Neuroscience, 8, 383–­395.
Environmental Medicine, 58, 125–­131. Takeda, N., Morita, M., Hasegawa, S., Horii, A., Kubo, T., & Matsunaga, T.
Leslie, R. A., Shah, Y., Thejomayen, M., Murphy, K. M., & Robertson, H. (1993). Neuropharmacology of motion sickness and emesis: A review.
A. (1990). The neuropharmacology of emesis: The role of receptors Acta Otolaryngologica, 113, 10–­ 15. https://doi.org/10.3109/00016​
in neuromodulation of nausea and vomiting. Canadian Journal of 48930​9126205
Physiology and Pharmacology, 68, 279–­288. https://doi.org/10.1139/ Takeda, N., Morita, M., Hasegawa, S., Kubo, T., & Matsunaga,
y90-­0 42 T. (1989). Neurochemical mechanisms of motion sickness.
Lowry, O. H., Rosebrough, N. J., Farr, A. L., & Randall, R. J. (1951). American Journal of Otolaryngology, 10, 351–­ 359. https://doi.
Protein measurement with the Folin phenol reagent. Journal of org/10.1016/0196- ­0709(89)90112​-­9
Biological Chemistry, 193, 265–­ 275. https://doi.org/10.1016/ Takeda, N., Morita, M., Horii, A., Nishiike, S., Kitahara, T., & Uno, A.
S0021  -­9258(19)52451​-­6 (2001). Neural mechanisms of Motion sickness. The Journal of Medical
Lucot, J. B. (1998). Pharmacology of motion sickness. Journal of Vestibular Investigation, 48, 44–­59.
Research, 8, 61–­66. https://doi.org/10.3233/VES-­1998-­8109 Thawkar, B. S. (2016). Phytochemical and pharmacological review of
Maheswari, D. U., Anand, T., Padma, A., Ilaiyaraja, N., & Khanum, F. Mentha arvensis. International Journal of Green Pharmacy, 10, 71–­76.
(2020). Evaluation of the effect of herbal extracts and their bioactive Wang, J. Q., Li, H. X., Chen, X. M., Mo, F. F., Qi, R. R., Guo, J. S., & Cai, Y.
compounds against motion sickness by regulating neurotransmitter L. (2012). Temporal change in NMDA receptor signaling and GABA A
levels in vitro and in vivo. South African Journal of Botany, 130, 130–­ receptor expression in rat caudal vestibular nucleus during motion
140. https://doi.org/10.1016/j.sajb.2019.12.012 sickness habituation. Brain Research, 1461, 30–­4 0.
Miller, J. D., & Brizzee, K. R. (1987). The anti-­emetic properties of 1-­sulpiride Wang, J., Liu, J., Pan, L., Qi, R., Liu, P., Zhou, W., & Cai, Y. (2017). Storage
in a ground-­based model of space motion sickness. Life Sciences, 41, of passive motion pattern in hippocampal CA1 region depends on
1815–­1822. https://doi.org/10.1016/0024-­3205(87)90700​- ­4 CaMKII/CREB signaling pathway in a motion sickness rodent model.
Ossenkopp, K. P., & Frisken, N. L. (1982). Defecation as an index of mo- Scientific Reports, 7, 1–­12. https://doi.org/10.1038/srep4​3385
tion sickness in the rat. Physiological Psychology, 10, 355–­360. https:// Xu, L. H., Tang, G. R., Yang, J. J., Liu, H. X., Li, J. C., & Jiang, Z. L. (2015). AVP
doi.org/10.3758/BF033​32964 modulation of the vestibular nucleus via V1b receptors potentially
Oz, M., El Nebrisi, E. G., Yang, K. H. S., Howarth, F. C., & Al Kury, contributes to the development of motion sickness in rat. Molecular
L. T. (2017). Cellular and molecular targets of menthol actions. Brain, 8, 86–­98. https://doi.org/10.1186/s1304​1-­015-­0175-­1
Frontiers in Pharmacology, 8, 472–­ 485. https://doi.org/10.3389/ Yamamoto, T. (2000). The effect of stress application on vestibular
fphar.2017.00472 compensation. Acta Otolaryngologica, 120, 504–­ 507. https://doi.
Patel, T., Ishiuji, Y., & Yosipovitch, G. (2007). Menthol: A refreshing look at org/10.1080/00016​48007​50046009
this ancient compound. Journal of American Academy of Dermatology, Yu, X. H., Cai, G. J., Liu, A. J., Chu, Z. X., & Su, D. F. (2007). A novel an-
57, 873–­878. https://doi.org/10.1016/j.jaad.2007.04.008 imal model for motion sickness and its first application in rodents.
Pliszka, S. R. (2016). Neuroscience of mental health clinician. Guilford Physiology and Behavior, 92, 702–­ 707. https://doi.org/10.1016/j.
Publications, 2, 316–­324. physb​eh.2007.05.067
Qi, R., Su, Y., Pan, L., Mao, Y., Liang, L., Dai, Z., Wang, J., & Cai, Y. (2019). Zheng, Y., Wang, X. L., Mo, F. F., & Li, M. (2014). Dexamethasone allevi-
Anti-­cholinergics mecamylamine and scopolamine alleviate motion ates MS in rats in part by enhancing the endocannabinoid system.
sickness-­induced gastrointestinal symptoms through both peripheral European Journal of Pharmacology, 727, 99–­105.
and central actions. Neuropharmacology, 146, 252–­263. https://doi. Zhou, W., Wang, J., Pan, L., Qi, R., Liu, P., Liu, J., & Cai, Y. (2017). Sex and
org/10.1016/j.neuro​pharm.2018.12.006 age differences in motion sickness in rats: The correlation with blood
Reason, J. T. (1978). Motion sickness adaptation: A neural mismatch hormone responses and neuronal activation in the vestibular and au-
model. Journal of the Royal Society of Medicine, 71, 819–­829. https:// tonomic nuclei. Frontiers in Aging Neuroscience, 9, 29–­43. https://doi.
doi.org/10.1177/01410​76878​07101109 org/10.3389/fnagi.2017.00029
Rubio, S., Weichenthal, L., & Andrews, J. (2011). Motion sickness:
Comparison of metoclopramide and diphenhydramine to pla-
cebo. Prehospital and Disaster Medicine, 26, 305–­ 3 09. https://doi. How to cite this article: Deshetty, U. M., Tamatam, A., &
org/10.1017/S1049​023X1​1006431 Patil, M. M. (2021). Menthol, a bioactive constituent of
Sakata, Y., Suzuki, H., Satoh, T., Kawatsu, S., Tanabe, K., Okamoto, K., Mentha, attenuates motion sickness in mice model:
Takagi, S., Komatsu, Y., Kawakami, K., & Yoshida, Y. (1986). Clinical
Involvement of dopaminergic system. Journal of Food
evaluation of sulpiride against nausea and vomiting during cancer
chemotherapy compared with domperidone-­envelope method. Gan Biochemistry, 45, e13863. https://doi.org/10.1111/jfbc.13863
to kagaku ryoho. Cancer Chemotherapy, 13, 2415–­2418.

You might also like