1 s2.0 S0753332222006424 Main

You might also like

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

Biomedicine & Pharmacotherapy 152 (2022) 113253

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Antinociceptive effect of Mansoa alliacea polar extracts involves opioid


receptors and nitric oxide in experimental nociception in mice
María Guadalupe Valle-Dorado a, Alberto Hernández-León b, Andrés Nani-Vázquez b, Guadalupe
Esther Ángeles-López a, María Eva González-Trujano b, *, Rosa Ventura-Martínez a, *
a
Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad No. 3000, Col. Ciudad Universitaria, Alcaldía
Coyoacán, 04510 Ciudad de México, Mexico
b
Laboratorio de Neurofarmacología de Productos Naturales, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría “Ramón de la Fuente
Muñiz”, Av. México-Xochimilco 101, Col. San Lorenzo Huipulco, 14370 Ciudad de México, Mexico

A R T I C L E I N F O A B S T R A C T

Key words: To evaluate the antinociceptive effect and the possible mechanism of action of two polar extracts of Mansoa
Mansoa alliacea alliacea, a medicinal plant used in Perú, Brazil, and Mexico to treat rheumatic pain, we used the formalin and hot-
Polar extracts plate tests in mice. We found that ethanolic (MA-EtOH) and aqueous (MA-AQ) extracts of M. alliacea induced
Antinociceptive effect
antinociceptive effects in both nociceptive tests. The antinociceptive efficacy of the highest dosage (300 mg/kg)
Nitric oxide pathway
Opioid receptors
of both extracts were also compared by using intraperitoneal and oral administration in the formalin test. Results
Mechanism of action showed that intraperitoneal injection of the two extracts produced better antinociceptive effects than that ob­
tained by their oral administration. The mechanism of action involved in their antinociceptive activity was
determined in the formalin test. Results showed that the presence of A784168 (TRPV1 antagonist) did not alter
the antinociceptive effect induced by any of the M. alliacea extracts, whereas naltrexone (opioid antagonist)
partially prevented the antinociceptive effect only of MA-EtOH in both phases of the formalin test. Furthermore,
the effects of the extracts were diminished by L-NAME (inhibitor of nitric oxide synthase), but not by ODQ
(inhibitor of the soluble guanylyl cyclase) or glibenclamide (blocker of K+ ATP channels) in the neurogenic phase.
However, the effect of MA-AQ was diminished by all the inhibitors in the inflammatory phase. These results
support the use of M. alliacea as a potential natural product with efficacy for pain relief depending on the form of
preparation and the route of administration by involving opioid receptors and the production of nitric oxide.

1. Introduction inflammatory process as well as in the generation of pain [3,4]. How­


ever, despite their clinical efficacy, most NSAIDs cause adverse effects as
Inflammatory pain is a public health problem that affects almost 30% gastric, renal, and/or cardiovascular damage; hence, limiting their
of the world population [1]. It is known that non-steroidal anti-in­ long-term use [5,6]. In addition to NSAIDs, among the therapeutic al­
flammatory drugs (NSAIDs) are the most prescribed medications for the ternatives for the relief of inflammatory pain, the use of medicinal plants
relief of inflammatory pain [2]. Their analgesic effects are due to the is also considered [7,8].
inhibition of cyclooxygenase (COX), an enzyme involved in the pro­ Mansoa alliacea (Lam.) AH Gentry (M. alliacea) is a plant native to the
duction of prostaglandins, a mediator that participates in the tropical forests of South America such as Perú, Brazil, and Mexico that

Abbreviations: M. alliacea, Mansoa alliacea; MA-EtOH, ethanolic extract of M. alliacea; MA-AQ, aqueous extract of M. alliacea; DIC, diclofenac; COX, cyclo­
oxygenase; CFA, Complete Freund’s adjuvant; NSAIDs, non-steroidal anti-inflammatory drugs; GLIB, glibenclamide; L-NAME, N(G)-L-nitro-arginine methyl ester;
NOS, nitric oxide synthase; ODQ, 1-H-(1,2,4)-oxadiazolo-(4,2-a)-quinoxalin-1-one; NTX, naltrexone; TRPV1, transient receptor potential cation channel subfamily V
member 1; SS, saline solution; i.p., intraperitoneal administration; p.o., oral administration; VEH, vehicle; S.E.M., standard error of the mean; AUC, area under the
curve; ANOVA, analysis of variance; au, area units; TCs, temporal courses; OECD, Organization for Economic Cooperation and Development guideline; LD50, lethal
dose 50.
* Corresponding authors.
E-mail addresses: lvalle-59@hotmail.com (M.G. Valle-Dorado), albertoh-leon@hotmail.com (A. Hernández-León), nani@imp.edu.mx (A. Nani-Vázquez),
geangeles@yahoo.com.mx (G.E. Ángeles-López), evag@imp.edu.mx (M.E. González-Trujano), rventuram7@hotmail.com (R. Ventura-Martínez).

https://doi.org/10.1016/j.biopha.2022.113253
Received 3 May 2022; Received in revised form 1 June 2022; Accepted 2 June 2022
Available online 10 June 2022
0753-3322/© 2022 The Author(s). Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
M.G. Valle-Dorado et al. Biomedicine & Pharmacotherapy 152 (2022) 113253

belongs to the Bignoniaceae family and is popularly known as "ajillo", Institute’s Bioethics Committee (CONBIOETICA-09-CEI-01–20170316).
“ajo sacha” or false garlic [9]. In the Southern Mexico, M. alliacea grows This protocol was approved by the Ethics and Research Committees as
wild in the states of Chiapas, Tabasco, and Veracruz [10]. The name of well as by the Internal Committee for the Care and Use of Laboratory
false garlic is due to the smell like that of bulb garlic (Allium sativum) that Animals of the Facultad de Medicina at UNAM (FM/DI/054/2018 and
is produced when its leaves are crushed [11]. M. alliacea is a perennial 009-CIC-2018) and by the INP (No. NC123280.0 and NC17073.0).
climbing shrub, with leaves, semi-woody branches, and violet flowers
[12]. In traditional medicine, the infusion of the dried aerial parts of this 2.3. Compounds
plant has been used to treat fever, headache, or rheumatic pain [12,13].
The alcoholic maceration of the bark of the roots and the patch of the Diclofenac (DIC) and morphine, used as positive controls, was pur­
leaves are considered anti-rheumatic and antiarthritic [14]. It has also chased from Novartis Farmacéutica and Pisa, respectively; A7841683
been used as insecticidal, antioxidant or as a condiment [14]. Despite its (6-dihydro-3’-(trifluoromethyl)-N-[4-[(trifluoromethyl) sulfonyl]
use for pain relief, in the biomedical literature there is only one report phenyl]-[1(2 H), 2’-bipyridine]-4-carboxamide), an antagonist of
describing the antiallodynic, antihyperalgesic, and antiinflammatory TRPV1 receptor, was purchased from TOCRIS Bioscience (Bristol); L-
effects of an hydroethanolic extract of M. alliacea using a model of NAME (N(G)-L-nitro-arginine methyl ester), a non-selective inhibitor of
chronic inflammation induced by the intraplantar administration of nitric oxide synthase; ODQ (1-H-(1,2,4)-oxadiazolo-(4,2-a)-quinoxalin-
Complete Freund’s Adjuvant (CFA) in mice [15]. To date, there are no 1-one), a selective inhibitor of soluble guanylate cyclase; naltrexone
studies on the antinociceptive effect of other extracts of this species. (NTX), an antagonist of opioid receptor, and glibenclamide (GLIB), a
Therefore, the objective of this study was determined the anti­ blocker of ATP-sensitive potassium channels (K+ ATP) were purchased
nociceptive activity of the aqueous and ethanolic extracts of M. alliacea from Sigma (Sigma Chemical Co., MO, USA). All compounds were
leaves in the formalin and hot-plate tests, two acute nociceptive pain administered intraperitoneally (i.p.) in a volume of 10 mL/kg using
models. The antinociceptive efficacy of both extracts was compared saline solution (SS, 0.9% NaCl) for their dissolution, except A784168,
using enteral and parenteral administration, as well as with diclofenac, which was dissolved with 0.1% DMSO/saline. A 1% formalin solution (J.
an analgesic of clinical use, in the formalin test. The possible mechanism T. Baker) was used as the allogeneic agent.
of action involved in the antinociceptive activity of this medicinal plant
was also analyzed. 2.4. Nociception tests

2. Material and methods 2.4.1. Formalin test


In this test, nociception was induced by the intradermal adminis­
2.1. Vegetal material and extracts preparation tration of 20 µl of 1% formalin into the dorsal surface of the right paw in
mice. Immediately after formalin administration, each mouse was
M. alliacea was collected in March 2019 in Gutiérrez Zamora in the placed in an acrylic cylinder surrounded by mirrors to quantify the
state of Veracruz in eastern Mexico. It was identified by the taxonomist number of flinches in the administered limb. The number of flinches was
Martha Juana Martínez Gordillo and deposited in the Herbarium from recorded during one minute at 5-minute intervals for a total time of 30
Facultad de Ciencias at UNAM with voucher number 178051. min. The formalin test consists of two phases: an early phase known as
Ethanolic extract of M. alliacea (MA-EtOH) was prepared with 400 g neurogenic (0–10 min), and a late phase known as inflammatory (10–30
of the dried and crushed aerial parts of the plant. The material was min) after administration of the allogeneic agent [17,18]. Animals were
macerated with ethanol (3.14 L) for seven days at room temperature habituated to the acrylic cylinder for at least 30 min before formalin
with occasional agitation. We used gravity filtration to separate the administration.
extract from the plant residues, the preparation was gravity filtered. The
filtrate was concentrated under vacuum, obtaining 18.6 g of a dark 2.4.2. Hot-plate test
green solid that represents a yield of 4.6% of the crude ethanolic extract. In this test, animals were placed, one by one, in an open cylinder with
Aqueous extract of M. alliacea (MA-AQ) was obtained by infusion a floor of a metallic plate that was previously heated to 55 ± 1 ◦ C. The
with 150 g of the dried and crushed aerial parts in 600 mL of boiling heated plate produces two kinds of behavioral responses: paw licking
water for 20 min. The solid residues of the extract were separated by and jumping, both are considered nociceptive responses. The latency to
gravity filtration and the filtrate was lyophilized for 12 h (Heto model, the first nociceptive response was recorded and the animal was imme­
FD3 Lab). A sample of 5.3 g of the final crude aqueous extract was ob­ diately removed from the hot plate. The first trial served as the control
tained as a light brown powder, which represented a yield of 3.5%. The reaction time, animals with base line latencies of more than 20 s were
dry extracts were stored at 4 ◦ C until further use. excluded from the study [19].
For their administration in mice, MA-EtOH extract was suspended in
0.5% tween 80 in saline solution (SS, 0.9% NaCl), while MA-AQ extract 2.5. Experimental design
was suspended only in SS. The extracts were given intraperitoneally (i.
p.) or by gavage (p.o) in a volume of 10 mL/kg. Control groups received To determine the antinociceptive effect of the extracts of M. alliacea
vehicle by the same route of administration. in the formalin test, eight groups of mice were used (n = 6 for group).
Three groups of animals were administered with MA-EtOH (30, 100, and
2.2. Animals 300 mg/kg), three received MA-AQ (30, 100, and 300 mg/kg), one
group received diclofenac (DIC, 10 mg/kg as a positive control), and
One hundred seventy-seven male mice (Swiss Webster, Taconic; another was the saline group (VEH). All treatments were administered
25–30 g) obtained from the Bioterium of the Instituto Nacional de Psi­ intraperitoneally (i.p.) 30 min before the administration of formalin. A
quiatría (INP) “Ramón de la Fuente Muñiz” and from the Central Bio­ decrease in the number of formalin-induced flinches in the presence of
terium of the Facultad de Medicina at Universidad Nacional Autónoma treatments was considered as an antinociceptive effect.
de México (UNAM) were used in this study. The animals were housed in To determine the antinociceptive effects of the extracts of M. alliacea
a temperature-controlled room (22 ± 1 ◦ C), with access to food and in the hot plate test, eight groups of mice were used (n = 6 for group).
water ad libitum and an automatic 12-h light/dark cycle. The experi­ Three groups of animals were administered with MA-EtOH (100, 300,
mental protocols were carried out under international guidelines for the and 600 mg/kg), three received MA-AQ (100, 300 and 600 mg/kg), one
use of laboratory animals, in addition to the Official Mexican Standard group received morphine (10 mg/kg as a positive control), and another
for the care and handling of animals [16] and the guidelines of the was the saline group (VEH). All treatments were administered

2
M.G. Valle-Dorado et al. Biomedicine & Pharmacotherapy 152 (2022) 113253

intraperitoneally (i.p.) 30 min before the hot-plate test. A decrease in the of the acute toxicity study of both extracts were analyzed with two-way
latency time to licking a paw or jumping was considered as an anti­ ANOVA test. Differences between means with a P-value < 0.05 were
nociceptive effect. Measurements were performed 30, 60, 90, and 120 considered significant at the 95% confidence level. Statistical analysis
min after treatment, with a cut-off time of 20 s to prevent development was performed using GraphPad Prism version 9.1.1 (GraphPad Software
of paw lesions. INC, La Jolla, CA, USA).
To know if the antinociceptive effect of the extracts of M. alliacea was
affected by the route of administration, we evaluated this effect 3. Results
exploring the highest dosage of MA-EtOH and MA-AQ by intragastric (p.
o.) and intraperitoneal (i.p.) administration used in the formalin test. 3.1. Antinociceptive effect of the ethanolic (MA-EtOH) and aqueous
For this, two groups of mice with n = 6 for each group were used. (MA-AQ) extracts of M. alliacea in the formalin test
To determine the possible mechanism of action involved in the
antinociceptive effect of MA-ETOH or MA-AQ, we used the formalin test Formalin injection produced a rapid flinches response in the
with dosage of 300 mg/kg, i.p., of each extract. For this, ten groups of administered limb (28.7 ± 1.3) in the VEH group that decreased at 10
mice with n = 6 for each group were used. Different groups of animals min (8.4 ± 0.8) (neurogenic phase) and increased again until reaching
received intraperitoneally the following treatment: A784168 (antago­ the maximum number of flinches at 25 min in the inflammatory phase
nist of TRPV1 receptor, 1 mg/kg), NTX (antagonist of opioid receptor, 5 (17.9 ± 1.1), which was maintained until 30 min (16.6 ± 0.9) (Fig. 1a,
mg/kg), L-NAME (inhibitor of nitric oxide synthetase, 20 mg/kg), GLIB b). The AUC of the nociceptive effect induced by formalin in the animals
(blocker of K+ATP channel, 10 mg/kg) or ODQ (inhibitor of soluble gua­ with vehicle (VEH) was 160 ± 14.8 au in the neurogenic phase (Fig. 1c,
nylate cyclase, 5 mg/kg), 15 min before the extracts. The anti­ d) and 280.4 ± 19.0 au in the inflammatory phase (Fig. 1e, f).
nociceptive effect of each M. alliacea extract was compared in the The administration of both MA-ETOH and MA-AQ extracts as well as
presence or in the absence of antagonists, blockers, or inhibitors. Doses DIC, used as positive control, decreased the number of flinches induced
of all drugs were chosen from previous studies in our laboratory [20,21]. by formalin from the first 5 min of evaluation (Fig. 1a, b). This dimi­
nution of the number of flinches was considered as an antinociceptive
2.6. Acute toxicity study effect. To know if this diminution produced by each treatment was
significant, the AUC of each phase in the formalin test was compared.
The oral acute toxicity of the M. alliacea extracts was carried out DIC significantly diminished the AUC of the nociceptive effect
according to procedure of Organization for Economic Cooperation and induced by formalin in comparison with VEH in the neurogenic phase
Development (OECD) guideline No. 425 [22]. Briefly, three groups of (0–10 min) (92.9 ± 4.3 vs 160 ± 14.8 au, respectively; with P-value =
mice were used to determine the acute toxicity of extracts of M. alliacea. 0.0014, t = 4.351, df = 10; using an unpaired t test of two-tailed)
For this, two groups of mice (n = 3 per group) were treated with a single (Fig. 1c, d) and in the inflammatory phase (10–30 min) (194.2 ± 21.7 vs
dose of 2000 mg/kg of each extract (MA-AQ or MA-EtOH) and the other 280.4 ± 19.0 au, respectively; with P-value = 0.0135, t = 2.99, df = 10;
was administered with vehicle (VEH). The extracts or VEH were using an unpaired t test of two-tailed) (Fig. 1e, f).
administered orally using gastric feeding tube. All animals were Regarding the effect of M. alliacea extracts, the analysis of the AUC
observed individually during 4 h every 30 min, and daily thereafter, for (0–10 min) of the neurogenic phase showed that the MA-EtOH extract
a total of 14 days for any clinical signs of toxicity or mortality. The body significantly diminished the nociceptive behaviors induced by formalin
weight of all animals was recorded before the administration of treat­ with all doses administered by i.p. (30, 100, 300 mg/kg) and p.o.
ment (day 0) and every 24 h for 14 days. (300 mg/kg) in comparison with that in VEH (99.6 ± 7.8, 81.3 ± 7.6,
After the acute administration of each treatment, locomotor activity 61.3 ± 5.7 and, 120.0 ± 3.8 vs 160 ± 14.8 au, respectively; with P-
(rotarod test) and exploratory behavioral was recorded every 30 min for value <0.0001, F(4,24) = 18.01; using one-way ANOVA followed by
2 h. For rotarod test, animals were placed in the rotarod apparatus (4 Dunnett’s post-hoc test). Furthermore, the diminution of the nociception
bars of 2.5 cm diameter, 25 cm above the floor) which was turned to 10 induced by formalin of this extract at 300 mg/kg by p.o. was signifi­
rpm for a period of 2 min. The duration of permanence in the bar in cantly lower than that obtained with the same dosage by i.p. (120 ± 3.8
seconds (s), and the number of falls of animals was recorded. To assess vs 61.3 ± 5.7 au, respectively; with P-value < 0.0001, t = 8.16, df = 9;
the exploratory activity of animal, they were placed in an acrylic cage using an unpaired t test of two-tailed) (Fig. 1c).
with 9 squares of 5 cm and the number of explored squares was On the other hand, the analysis of the AUC (0–10 min) of the MA-AQ
recorded. extract in the same phase also showed a significant reduction in the
nociceptive behaviors induced by formalin with all doses administered
2.7. Statistical analysis by i.p. (30, 100, 300 mg/kg) and p.o. (300 mg/kg)) in comparison with
that in VEH (106.7 ± 11.5, 112.5 ± 14.2, 88.3 ± 9.7 and, 78.0 ± 3.5 vs
Data are expressed as the mean ± standard error of the mean (S.E. 160 ± 14.8 au, respectively; with P-value = 0.0007, F(4,24) = 7.04;
M.) of six mice per group. Considering that 28 groups of animals (n = 6 using one-way ANOVA followed by Dunnett’s post-hoc test). However,
animals per group) and 3 groups for the acute toxicity (n = 3 animals per in this case, there was no difference between the effect of the MA-AQ
group) were used, the total number of animals used in this study was one extract (300 mg/kg) administered p.o. compared to i.p. (78.0 ± 3.5 vs
hundred seventy-seven. 88.3 ± 9.7 au, respectively; with P-value = 0.3782, t = 0.927, df = 9;
Temporal courses (TCs) of the number of flinches in the formalin test using an unpaired t test of two-tailed) (Fig. 1d).
and the latency to licking a paw or jumping in the hot-plate test in the In relation to the analysis of AUC (10–30 min) of the inflammatory
presence of different treatments were constructed. The areas under the phase, all doses of the MA-EtOH extract (30, 100, 300 mg/kg, i.p. and,
curve (AUC) of the neurogenic phase (from 0 to 10 min) and of the in­ 300 mg/kg, p.o.) significantly reduced the nociception induced by
flammatory phase (from 10 to 30 min) in the formalin test were calcu­ formalin in comparison with that by VEH (172.9 ± 18.1, 105.8 ± 15.5,
lated from the TCs with the trapezoid addition method [23] and the 63.8 ± 9.2 and, 169.0 ± 23.8 vs 280.4 ± 19.0 au, respectively; with P-
corresponding bar graphs were constructed for statistical analysis. Sta­ value < 0.0001, F(4,24) = 23.2; using one-way ANOVA followed by
tistical differences of treatments were obtained through a one-way Dunnett’s post-hoc test). Like in the neurogenic phase, the diminution of
analysis of variance (ANOVA) for independent samples followed by the nociception induced by formalin of 300 mg/kg of this extract by p.o.
Dunnett’s post-hoc test, which was used only if F achieved P < 0.05 and was significantly lower than that obtained with the same dosage used by
there was no significant variance in homogeneity. Differences between i.p. way in the inflammatory phase (169.0 ± 23.8 vs 63.8 ± 9.2 au,
two groups were determined using an unpaired t test of two tailed. Data respectively; with P-value = 0.0017, t = 4.42, df = 9; using an unpaired

3
M.G. Valle-Dorado et al. Biomedicine & Pharmacotherapy 152 (2022) 113253

Fig. 1. Temporal courses (TCs) of the number of flinches induced by formalin in mice in the absence (VEH) and in the presence of DIC and different doses of the
ethanolic or aqueous extracts of M. alliacea (MA-EtOH or MA-AQ) (a, b). AUC of the neurogenic phase calculated from TCs of each treatment (0–10 min) (c, d) and,
AUC of the inflammatory phase calculated from TCs of each treatment (10–30 min) (e, f). Each point or bar represents the mean ± S.E.M. of a n = 6 animals per
treatment. *P < 0.05, **P < 0.01 and, ***P < 0.001 using one-way ANOVA followed by Dunnett’s test compared with VEH. ##P < 0.01 and ###P < 0.001 using
an unpaired t test for each comparison.

4
M.G. Valle-Dorado et al. Biomedicine & Pharmacotherapy 152 (2022) 113253

t test of two-tailed) (Fig. 1e). two-way ANOVA followed by Dunnett’s multiple comparison test)
In the same phase in the formalin test, the analysis of AUC (10–30 min) (Fig. 2b).
of MA-AQ extract showed that only two doses (100 and 300 mg/kg, i.p.)
significantly reduced the nociception in comparison with that in VEH
(87.5 ± 25.1 and 67.9 ± 17.1 vs 280.4 ± 19.0 au, respectively; with P- 3.3. Participation of TRPV1 channels and opioids receptors in the
value = 0.0002, F(4,24) = 8.83; using one-way ANOVA followed by antinociceptive effects of M. alliacea extracts
Dunnett’s post-hoc test). Finally, unlike in the neurogenic phase, the
effect of MA-AQ extract (300 mg/kg) by p.o. in the nociception In the temporal courses obtained after the formalin injection,
formalin-induced was significantly lower than that obtained with the administration of A784168 (a TRPV1 receptor antagonist) did not alter
same dosage by i.p. in the inflammatory phase (185.5 ± 16.0 vs 67.9 the antinociceptive effects induced by MA-EtOH neither MA-AQ ex­
± 17.1 au, respectively; with P-value = 0.0008, t = 4.93, df = 9; using tracts; while naltrexone (NTX, an opioid receptor antagonist) prevented
an unpaired t test of two-tailed) (Fig. 1 f). the antinociceptive effect induced by MA-EtOH but did not alter the
effect of MA-AQ (Fig. 3a, b). To know if some of the pre-treatment with
A78168 or NTX significantly altered the diminution in the number of
3.2. Antinociceptive effect of the ethanolic (MA-EtOH) and aqueous flinches produced by MA-EtOH or MA-AQ extracts at 300 mg/kg, i.p.,
(MA-AQ) extracts of M. alliacea in the hot-plate test their AUCs in each phase of the formalin test were compared.
Results showed that both MA-EtOH and MA-AQ extracts significantly
On the other hand, in the hot-plate test, the administration of reduced the AUC (0–10 min) in the neurogenic phase of the number of
300 mg/kg of MA-EtOH extract significantly increased the latency at the flinches produced by formalin in comparison with that in VEH (61.3
first nociceptive response (paw licking) at 60, 90, and 120 min of ± 5.8 vs 160.0 ± 14.8 au with P-value < 0.0001, t = 6.22, df = 10; and,
evaluation (13.2 ± 1.6, 14.0 ± 0.6 and 13.6 ± 1.4 s, respectively); and 88.3 ± 9.7 vs 160.0 ± 14.8 au with P-value = 0.0023, t = 4.05, df = 10,
with 600 mg/kg at 90 and 120 min (11.0 ± 2 and 10.7 ± 1.2 s, respectively; in both comparisons an unpaired t test of two-tailed was
respectively); as well as morphine did at 30, 60, 90 and 120 min (14.8 used) (Fig. 3c, d and 4c, d). Like in the neurogenic phase, both MA-EtOH
± 1.7, 19.5 ± 0.4, 18.9 ± 0.6 and 19.1 ± 0.8 s, respectively) in com­ and MA-AQ extracts also significantly reduced the AUC (10–30 min) in the
parison with that in VEH at the respective time of evaluation: 30, 60, 90 inflammatory phase in the formalin test (63.8 ± 9.2 vs 280.4 ± 19.0 au
and 120 min (5.8 ± 0.7, 7.7 ± 0.6, 7.4 ± 0.5 and 7.2 ± 0.8 s, respec­ with P-value < 0.0001, t = 10.3, df = 10; and, 67.9 ± 17.1 vs 280.4
tively) (with P-value < 0.0001, F(3,75) = 16.1; using two-way ANOVA ± 19.0 au with P-value < 0.0001, t = 8.31, df = 10, respectively; in both
followed by Dunnett’s multiple comparison test) (Fig. 2a). comparisons an unpaired t test of two-tailed was used) (Figs. 3e, f and 4
Regarding the effect of MA-AQ extract in this test, 100 mg/kg e, f).
significantly increased the latency at the first paw licking at 90 min Otherwise, in the neurogenic phase in the formalin test the admin­
(12.0 ± 1.0 s), 300 mg/kg at 60 min (13.8 ± 1.6 s) and, 600 mg/kg at istration of A784168 did not alter the AUC (0–10 min) produced by MA-
60, 90 and 120 min (14.7 ± 2.4, 14.8 ± 2.0 and 12.2 ± 1.2 s, respec­ EtOH extract as NTX did in comparison with the extract alone (84.6
tively) in comparison with that in VEH at the respective time of evalu­ ± 15.5 and 122.9 ± 13.0 vs 61.3 ± 5.8 au, respectively; P-value =
ation: 30, 60, 90 and 120 min (5.8 ± 0.7, 7.7 ± 0.6, 7.4 ± 0.5 and 7.2 0.008, F(2,15) = 6.6; using one-way ANOVA followed by Dunnett’s
± 0.8 s, respectively) (with P-value < 0.0001, F(3,100) = 9.94; using post-hoc test) (Fig. 3c); while neither A784168 nor NTX altered the AUC

Fig. 2. Latency (s) in the hot-plate test in mice


with VEH or different doses of the ethanolic
extract (MA-EtOH) (a) or of the aqueous extract
(MA-AQ) of M. alliacea (b) evaluated in each
group every 30 min during 2 h. VEH group is
the negative control and morphine group is the
positive control. Each treatment was adminis­
tered 30 min before the test. The cut-off time of
this test in each time of evaluation was 20 s.
Each bar represents the mean ± S.E.M. of a
n = 6 animals per treatment. *P < 0.05,
**P < 0.01 and, ***P < 0.001 using two-way
ANOVA followed by Dunnett’s multiple com­
parison test compared with VEH at the corre­
sponding time.

5
M.G. Valle-Dorado et al. Biomedicine & Pharmacotherapy 152 (2022) 113253

Fig. 3. Temporal courses (TCs) of the effect of MA-EtOH or MA-AQ on 1% formalin-induced nociception in mice in the presence of NTX (opioids receptor antagonist)
and A784168 (TRPV1 receptor antagonist) (a, b). AUC of the neurogenic phase of the formalin test calculated from TCs of each treatment (0–10 min) (c, d) and, AUC
of the inflammatory phase of the formalin test calculated from TCs of each treatment (10–30 min) (e, f). Each point or bar represents the mean ± S.E.M. of a n = 6
animals per treatment. ###P < 0.001 using an unpaired t test of two-tailed in each comparison; **P < 0.01 using one-way ANOVA followed by Dunnett’s post-hoc
test compared with the extract alone.

6
M.G. Valle-Dorado et al. Biomedicine & Pharmacotherapy 152 (2022) 113253

produced by MA-AQ extract alone (85.8 ± 7.7 and 112.5 ± 17


(0–10 min) did not alter the AUC (10–30 min) of the effect produced by MA-EtOH
vs 88.3 ± 9.7 au, respectively; P-value = 0.261, F(2,15) = 1.47; using extract as NTX did in comparison with the extract alone (56.7 ± 12.9
one-way ANOVA test) (Fig. 3d). and 125.4 ± 17.0 vs 63.8 ± 9.2 au, respectively; P-value = 0.0003, F
While, in the inflammatory phase, the administration of A784168 (2,15) = 14.36; using one-way ANOVA followed by Dunnett’s post-hoc

Fig. 4. Temporal courses (TCs) of the effect of MA-EtOH or MA-AQ on 1% formalin-induced nociception in mice in the presence of L-NAME (SON inhibitor), ODQ
(sGC inhibitor) or GLIB (blocker of K+
ATP channels) (a, b). AUC of the neurogenic phase of the formalin test calculated from TCs of each treatment (0–10 min) (c, d),
and AUC of the inflammatory phase of the formalin test calculated from TCs of each treatment (10–30 min) (e, f). Each point or bar represents the mean ± S.E.M. of a
n = 6 animals. ###P < 0.001 using an unpaired t test of two-tailed in each comparison; **P < 0.01 and ***P < 0.001 using one-way ANOVA followed by Dunnett’s
post-hoc test compared with the extract alone.

7
M.G. Valle-Dorado et al. Biomedicine & Pharmacotherapy 152 (2022) 113253

test) (Fig. 3e), while neither A784168 nor NTX alter the AUC (10–30 min)
± 6.3, 82.5 ± 13.4 and 120.8 ± 12.8 vs 61.3 ± 5.8 au, respectively; P-
produced by MA-AQ extract alone (81.7 ± 16.6 and 80.4 ± 19.5 vs 67.9
value = 0.004, F(3,20) = 6.1; using one-way ANOVA followed by
± 17.1 au, respectively; P-value = 0.835, F(2,15) = 0.183; using one-
Dunnett’s post-hoc test) (Fig. 4c). The same situation occurred with the
way ANOVA test) (Fig. 3 f).
MA-AQ extract: neither of ODQ nor GLIB alter the AUC (0–10 min) of the
effect produced by the extract as L-NAME did in comparison with the
3.4. Participation of the NO-cGMP-K+ATP channels pathway in the extract alone (105.8 ± 13.7, 73.8 ± 7.3 and 160.0 ± 12.0 vs 88.3 ± 9.7
antinociceptive effects of M. alliacea extracts au, respectively; P-value = 0.0001, F(3,20) = 11.9; using one-way
ANOVA followed by Dunnett’s post-hoc test) (Fig. 4d).
To know the possible participation of the NO-cGMP-K+ In regard to the analysis of AUC (10–30 min) in the inflammatory phase,
ATP pathway in
the antinociceptive effect of MA-EtOH and MA-AQ extracts in the results showed that neither the administration of L-NAME, ODQ nor
formalin test, we used several inhibitors of this pathway. Results showed GLIB alter the AUC (10–30 min) of the effect produced by MA-EtOH extract
that the administration of L-NAME (an inhibitor of NO synthase) alone (67.9 ± 19.8, 135.4 ± 29.4 and 150.0 ± 33.6 vs 63.8 ± 9.2 au,
partially inhibited the effect of MA-EtOH extract (300 mg/kg, i.p.) on respectively; P-value = 0.073, F(3,20) = 3.25; using one way ANOVA
the nociceptive responses induced by formalin, whereas neither ODQ (a test) (Fig. 4e); while, on the contrary, the L-NAME, ODQ and GLIB
soluble guanylate cyclase inhibitor) nor GLIB (a K+ significantly alter the AUC (10–30 min) of the effect produced by MA-EtOH
ATP channel blocker)
altered it (Fig. 4a). On the other hand, administration of L-NAME, ODQ extract alone (177.5 ± 8.9, 200.8 ± 34.4 and 180.4 ± 28.3 vs 67.9
and GLIB seemed to alter the effect of MA-AQ extract (300 mg/kg, i.p.) ± 17.1 au, respectively; P-value = 0.004, F(3,20) = 6.11; using one-way
on the nociceptive responses induced by formalin (Fig. 4b). To know if ANOVA followed by Dunnett’s post-hoc test) (Fig. 4 f).
the pretreatment with L-NAME, ODQ or GLIB significantly altered the
diminution in the number of flinches produced by MA-EtOH or MA-AQ 3.5. Acute toxicity of M. alliacea extracts
extracts in each phase of the formalin test, their AUCs were compared.
In this sense, neither the administration of ODQ nor GLIB alter the The oral dose of 2000 mg/kg of MA-AQ or MA-EtOH extracts did not
AUC (0–10 min) of the effect produced by MA-EtOH extract in the neuro­ cause death in mice during the short and long-term observation period,
genic phase as L-NAME did in comparison with the extract alone (77.5 and no signs of toxicity were observed in the animals throughout the 14

Fig. 5. Acute toxicity tests. Exploratory activity of animals administered with an oral dose of 2000 mg/kg of MA-EtOH or MA-AQ on the acrylic cage with 9 squares
of 5 cm each one evaluated in each group every 30 min during 2 h (a). Body weight of animals administered with an oral dose of 2000 mg/kg of MA-EtOH, MA-AQ or
vehicle (VEH). Each point or bar represents the mean ± S.E.M. of a n = 3 animals.

8
M.G. Valle-Dorado et al. Biomedicine & Pharmacotherapy 152 (2022) 113253

days study period. The lethal doses 50 (LD50) of both extracts were morphine activates the descending inhibitory pathway of the CNS and
estimated to be greater than 2000 mg/kg. Regarding the exploratory inhibits the nociceptive afferent neurons of the peripheral nervous sys­
behavioral, the mice pretreated with MA-AQ or MA-EtOH (2000 mg/kg) tem, which leads to a reduction of the nociceptive transmission [28].
did no show changes in the number of explored squares at any time In this work, we also compared the antinociceptive effects of
evaluated compared with those of the VEH (P-value = 0.671, F(8, 30) M. alliacea prepared in two polar extracts (MA-EtOH and MA-AQ) using
= 0.721; using two-way ANOVA test) (Fig. 5a). Also, in relation to the enteral and parenteral routes of administration. Results showed that the
locomotor activity, the mice pretreated with MA-AQ or MA-EtOH antinociceptive efficacy of both extracts was better by intraperitoneal
(2000 mg/kg) did no show motor performance alterations in the than intragastric administration in the formalin test using the same
rotarod test compared with those of VEH since all animals were held in dosage. As in other extracts of medicinal plants, antinociceptive effects
the rotarod for 2 min at each evaluation time (data not shown). There of M. alliacea extracts might depend on the bioactive constituents pre­
were also no differences in the body weight of the animals with any of sent in preparations of this species. Phytochemical analysis of an
the treatments during the 14 days of registration (P-value = 0.9989, F hydroethanol extract of M. alliacea by UHPLC-MS/MS revelated the
(28,90) = 0.347; using two-way ANOVA test) (Fig. 5b). presence of higher concentrations of phenolic compounds such as p-
coumaric acid, luteolin, ferulic acid, chlorogenic acid, and apigenin
4. Discussion [15], a triterpene (betulinic acid) identified in an ethanol extract
through analysis by HPLC-MS [29] and two pyranonaphthoquinonas
In the present study, the antinociceptive effects of polar extracts (mansonin A and mansonin B) determined by spectroscopic analysis
(ethanol and aqueous) from M. alliacea leaves were demonstrated in the [30]. In contrast, sulfur compounds have been identified in the essential
formalin and in the hot-plate tests in mice: two models that represent an oil from M. alliacea fresh leaves, whose characteristic smell to garlic
acute nociceptive pain. M. alliacea has been used in the traditional depends on these kinds of constituents [31] as well as estigmasterol and
medicine [9] for relieving rheumatic-type pain [11,24,25], headache, γ-sitosterol identified in the no-polar extract of this species [32]. These
muscle fatigue [11], to treat colds, pneumonia [26], fever, and as insect kinds of metabolites might be obtained in the aqueous extract that was
repellent; as well as a condiment in food preparation due to its smell and prepared by decoction involving boiling water.
taste of garlic [9,11]. The aerial parts of the plant are used as infusion or One of the bioactive metabolites identified in the polar extracts is
decoction, as well as a preparation using the roots or leaves macerated in apigenin, whose antinociceptive effect has been reported in the hot-
ethanol [14]. plate, formalin, and writhing tests in mice [33,34]. Also, the chloro­
Results of this study show that both ethanol and aqueous extracts of genic [35,36] and ferulic acids [37] have produced antinociceptive ef­
M. alliacea (MA-EtOH and MA-AQ) produced antinociceptive effects in fects in a neuropathic pain model induced by ligation of the sciatic
both phases of the formalin test. The antinociceptive effects of both nerve. Whereas luteolin has shown anti-inflammatory effect in the CFA
extracts were better than those shown by diclofenac, a NSAID. In gen­ model in rats [38] and p-coumarinic acid induces analgesic and
eral, these results showed that the antinociceptive effects of both ex­ anti-inflammatory effects in the gouty arthritis model induced with acid
tracts were better in the inflammatory phase than in the neurogenic crystals monosodium uric acid in rats [39].
phase of this test. It is well known that formalin test is an assay used as a To know the possible mechanisms involved in the antinociceptive
model for searching compounds with antinociceptive and anti- effects of MA-EtOH and MA-AQ extracts, the participation of TRPV1 and
inflammatory effects. In this test, nociception is induced by the intra­ opioid receptors, as well as the NO-cGMP-K+ ATP pathway were investi­
dermal administration of 1% formalin into the dorsal surface of right gated in this work. It is known that opioid receptors are one of the most
paw in mouse. The number of formalin-induced flinches is quantified, important signaling pathways in the descending modulation of noci­
and an increase is related to the degree of nociception [27]. This test is ception [21]. It has also been reported that some natural products such
biphasic where different mechanisms and/or nociceptive mediators as flavonoids and terpenoids participated through this signaling
participate and can be distinguished pharmacologically. The first phase pathway [21,40]. In this study, the administration of naltrexone, a
is characterized by central neurogenic pain caused by direct chemical non-selective antagonist of opioid receptors, produced a partial inhibi­
stimulation of C-fiber nociceptors and occurs within the first (0–10 min) tion of the antinociceptive effect of MA-ETOH extract but not of the
minutes of formalin administration. The nociceptive behaviors in this MA-AQ extract in both phases in the formalin test. These results agreed
phase can be attenuated with opioid drugs because of their central ef­ with a previous report where the antinociceptive effect of the hydro­
fect. The second phase is characterized by an inflammatory process in alcoholic extract was mediated by the participation of opioids, mainly δ
which several mediators are involved such as prostaglandins, neuro­ receptors [15]. It is important to mention that in binding assays, the
peptides, and nitric oxide. This phase occurs from 10–30 min after the apigenin has shown affinity for δ and μ opioid receptors [41]. In addition
administration of formalin and the nociceptive behaviors in this phase to this, an activation of opioid receptors was involved in the anti­
can be attenuated by NSAIDs for their peripheral effects [17]. The fact nociceptive effects showed by luteolin [42], ferulic acid [37], and
that both extracts of M. alliacea in this study induced antinociceptive chlorogenic acid [43].
effects in the two phases of the formalin test suggests that it acts by On the other hand, the TRPV1 receptor is one of the most intensively
central and peripheral mechanisms by blocking inhibitory receptors studied channel for being one of the main receptors involved in the
and/or by inhibiting the formation and/or release of inflammatory sensitization and the maintenance of nociception in the inflammatory
mediators. To demonstrate that in the antinociceptive effects of pain [44]. For this, its participation in the antinociceptive effect of MA
M. alliacea extracts, central mechanisms are involved, the hot-plate test extracts in this study was assessed using a A784168 as specific antago­
was used in this study. The hot-plate test is also a quick and relatively nist of this receptor [45]. Results showed that antinociceptive effects of
inexpensive way to assess acute, thermal pain, and it has been consid­ both extracts of M. alliacea did not change in the presence of this
ered a sensitive test to repeated measurements. Also, it is well known antagonist, suggesting that the participation of TRPV1 receptors is not
that the behavioral responses induced in this thermal nociceptive test involved in the MA antinociceptive properties.
(paw licking and jumping) are considered supraspinally integrated re­ Evidence in literature indicates that participation of the NO–cGMP
sponses [19]. Results of this study confirmed that in the antinociceptive pathway is involved in the antinociceptive activity of several drugs [46,
effect of both M. alliacea extracts central mechanisms are involved, such 47]. Nitric oxide (NO) is a small gaseous molecule with a short half-life
as their effects in the hot-plate test resembling the effect of morphine, that is synthesized by nitric oxide synthetase (NOS) [48]. NO can in­
whose antinociceptive effect occur centrally. Morphine is considered the crease the concentration of cGMP by activation of soluble guanyl cyclase
classic opioid analgesic that produces its analgesic effects by binding to (sGC) in different types of cells. The increase of cGMP activates the
opioid receptors within the central nervous system (CNS). In this way, ATP-sensitive potassium channels (K+ ATP). Finally, the opening of these

9
M.G. Valle-Dorado et al. Biomedicine & Pharmacotherapy 152 (2022) 113253

channels induces the membrane hyperpolarization reducing the action Declaration of interest
potential transmission abilities of neurons producing analgesia [49,50].
In this study, the inhibition of the NOS using L-NAME significantly The authors declare that there is no conflict of interest.
decreased the antinociceptive effect of both MA-EtOH and MA-AQ ex­
tracts in the neurogenic phase of the formalin test, while the adminis­ Acknowledgments
tration of ODQ (an inhibitor of sGC) or glibenclamide (a blocker of K+ ATP
channels) did not alter the antinociceptive effect of these extracts in this The authors would like to thank Mrs. Josefina Bolado, Head of the
phase. As previously mentioned, it is known that in the neurogenic Scientific Paper Translation Department, from División de Investigación
phase of the formalin test, mainly drugs like opioids induce anti­ at Facultad de Medicina, at UNAM for the diligent proofreading of the
nociceptive effects. In this sense, the participation of NO-cGMP pathway English-language version of this manuscript. We also thanks to Andrea
in the modulation of antinociceptive activity of morphine has been re­ Bañuelos Sánchez and Miguel A. Zumaya for their technical assistance.
ported [51]. These results suggest that the antinociceptive effect of ex­ PhD. Maria G. Valle-Dorado thanks the fellowship from Dirección
tracts of M. alliacea involve central mechanisms such as the participation General de Asuntos del Personal Académico at Universidad Nacional
of ON as an analgesic mediator, but it is produced by an independent Autónoma de México (DGAPA-UNAM, 080/2021).
mechanism that the classic cGMP-ATP-sensitive K+ channel pathway as
it has been observed with some natural products such as pure com­
Funding sources
pounds or complete extract, for example ursolic acid [20] or Clinacan­
thus nutans methanol extract [52].
Funding: This work was supported by the Programa de Apoyo a
On the other hand, the NO-cGMP-K+ ATP pathway did not participate in
Proyectos de Investigación e Innovación Tecnológica, UNAM, Mexico
the antinociceptive effect of MA-EtOH extract in the inflammatory phase
[UNAM-PAPIIT, grant number IN201820]; Consejo Nacional de Ciencia
of the formalin test since L-NAME, ODQ or GLIB did not alter its anti­
y Tecnología, Mexico [CONACYT, grant numbers 226454 and 256448];
nociceptive effect. Whereas this NO-cGMP-K+ ATP pathway participated in
and Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz”,
the antinociceptive effect obtained with the MA-AQ extract in the in­
Mexico [grant numbers NC123280.0 and NC17073.0]. PhD. Maria G.
flammatory phase of the formalin test. These results reinforced the po­
Valle-Dorado has received the fellowship from Dirección General de
tential use of M. alliacea in inflammatory pain and they agree with the
Asuntos del Personal Académico at UNAM, Mexico (DGAPA-UNAM,
already reported antinociceptive effect of the hydroalcoholic extract of
080/2021).
this species collected in the Amazonian region using the intraplantar
administration of the complete Freund’s adjuvant in mice, a model
representing a condition of chronic inflammatory pain like those pre­ Data statement
sented by patients with arthritis [15]. Previous studies in cell culture
have shown that apigenin can activate the NO pathway [53,54] and The datasets generated and/or analyzed during the current study are
some other studies indicate that ferulic acid [55] and p-coumarin available from the corresponding author on reasonable request.
decrease NO levels to produce antinociceptive effects [39].
Finally, we also evaluated the possible adverse effects of M. alliacea References
extracts and the results of the oral administration of 2000 mg/kg of MA-
[1] A.J. Barragán-Berlanga, S. Mejía-Arango, L.M. Gutiérrez-Robledo, Dolor en adultos
AQ and MA-EtOH extracts suggesting that both extracts may be safe at mayores de 50 años: prevalencia y factores asociados [Pain in the elderly:
these doses and the oral LD50 considered greater than 2000 mg/kg in prevalence and associated factors], Salud Publica Mex. 49 (2007) S488–S494,
mice, according to OECD guideline No. 425 [22]. These results are ac­ https://doi.org/10.1590/s0036-36342007001000008.
[2] V.K. Bournia, G. Kitas, A.D. Protogerou, P.P. Sfikakis, Impact of non-steroidal anti-
cording with a study where it was demonstrated that the M. alliacea
inflammatory drugs on cardiovascular risk: Is it the same in osteoarthritis and
hydroethanolic extract did not produce constipation, alterations in the rheumatoid arthritis? Mod. Rheumatol. 27 (2017) 559–569, https://doi.org/
motor coordination, hypothermia, gastric lesions neither hepatic, nor 10.1080/14397595.2016.1232332.
[3] J.N. Cashman, The mechanisms of action of NSAIDs in analgesia, Drugs 52 (1996)
renal failure, considering that this species is a safe medicinal plant [15].
13–23, https://doi.org/10.2165/00003495-199600525-00004.
[4] N. Leyva-López, E.P. Gutierrez-Grijalva, D.L. Ambriz-Perez, J.B. Heredia,
5. Conclusions Flavonoids as cytokine modulators: a possible therapy for inflammation-related
diseases, Int. J. Mol. Sci. 17 (2016) 921, https://doi.org/10.3390/ijms17060921.
[5] M.M. Wolfe, D.R. Lichtenstein, G. Singh, Gastrointestinal toxicity of nonsteroidal
In conclusion, our study corroborates that M. alliacea prepared in anti-inflammatory drugs, N. Engl. J. Med. 340 (1999) 1888–1899, https://doi.org/
both ethanolic and aqueous extracts can produce antinociceptive effects 10.1056/NEJM199906173402407.
with similar efficacy in acute nociceptive pain, thus contributing to the [6] C.K. O’Neil, J.T. Hanlon, Z.A. Marcum, Adverse effects of analgesics commonly
used by older adults with osteoarthritis: focus on non-opioid and opioid analgesics,
scientific evidence that supports the ethnopharmacological knowledge Am. J. Geriatr. Pharmacother. 10 (2012) 331–342, https://doi.org/10.1016/j.
of this species for pain relief. It seems that concentrations and kinds of amjopharm.2012.09.004.
constituents in the polar nature of the extracts can influence the anti­ [7] J.B. Calixto, Twenty-five years of research on medicinal plants in Latin America: A
personal view, J. Ethnopharmacol. 100 (2005) 131–134, https://doi.org/10.1016/
nociceptive mechanism of action involved. Since for the ethanolic j.jep.2005.06.004.
extract it had influence on the opioid receptors while for the aqueous [8] B.B. Mishra, V.K. Tiwari, Natural products: An evolving role in future drug
extract, the participation of all the NO-cGMP/K+ ATP pathway was discovery, Eur. J. Med. Chem. 46 (2011) 4769–4807, https://doi.org/10.1016/j.
ejmech.2011.07.057.
observed.
[9] L.G. Lohmann, M. Ch. Taylor, A new generic classification of Bignonieae
(Bignoniaceae), Ann. Mo. Bot. Gard. 99 (2014) 348 489, https://doi.org/10.3417/
CRediT authorship contribution statement 2003187.
[10] CONABIO. Comisión Nacional para el conocimiento y uso de la biodiversidad.
〈https://enciclovida.mx/especies/202399〉.
Rosa Ventura-Martínez, María E. González-Trujano: Resources, [11] M.G. Bichara Zoghbi, J. Oliveira, G.M. Pinheiro Guilhon, The genus Mansoa
Writing − review & editing, Visualization; Funding acquisition. María (Bignoniaceae): A source of organosulfur compounds, Rev. Bras. Farmacogn. 19
E. González-Trujano, María G. Valle-Dorado: Conceptualization, (2009) 795–804, https://doi.org/10.1590/S0102-695X2009000500025.
[12] A. Tasambay-Salazar, L. Scalvenzi, A.S. Piedra-Lascano, M. Radice,
Writing − original draft. María G. Valle-Dorado, Alberto Hernández- Ethnopharmacology, biological activity and chemical characterization of Mansoa
León, Guadalupe E. Ángeles-López: Methodology, Formal analysis, alliacea, A Rev. A Promis. Plant Amazon. Reg., MOL2NET´17 3 (2017) 15, https://
Investigation. Andrés Nani-Vázquez: Methodology, Collected the me­ doi.org/10.3390/mol2net-03-04617.
[13] E. Pagani, J. Santos, E. Rodrigues, Culture-Bound Syndromes of a Brazilian Amazon
dicinal plant and prepared the extracts. Riverine population: Tentative correspondence between traditional and
conventional medicine terms and possible ethnopharmacological implications,

10
M.G. Valle-Dorado et al. Biomedicine & Pharmacotherapy 152 (2022) 113253

J. Ethnopharmacol. 203 (2017) 80–89, https://doi.org/10.1016/j. involvement of descending monoaminergic system and opioid receptors,
jep.2017.03.024. Oncotarget 7 (2016) 20455–20468, https://doi.org/10.18632/oncotarget.7973.
[14] I. Patel, S. Sipai, D. Rathod, G. Shrimali, A. Patel, E. Rami, Phytochemical studies [38] R. Jeyadevi, T. Sivasudha, A. Rameshkumar, L. Dinesh Kumar, Anti-arthritic
on Mansoa alliacea (Lam.), Int. J. Adv. Pharm. Res. 4 (2013) 1823–1828. activity of the Indian leafy vegetable Cardiospermum halicacabum in Wistar rats and
[15] F.R. Hamann, I. Brusco, G. de Campos Severo, L.M. de Carvalho, H. Faccin, UPLC–QTOF–MS/MS identification of the putative active phenolic components,
L. Gobo, S.M. Oliveira, M.A. Rubin, Mansoa alliacea extract presents Inflamm. Res. 62 (2013) 115–126, https://doi.org/10.1007/s00011-012-0558-z.
antinociceptive effect in a chronic inflammatory pain model in mice through [39] S.J. Pragasan, M. Rasool, Dietary component p-coumaric acid suppresses
opioid mechanisms, Neurochem. Int. 122 (2019) 157–169, https://doi.org/ monosodium urate crystal-induced inflammation in rats, Inflamm. Res. 62 (2013)
10.1016/j.neuint.2018.11.017. 489–498, https://doi.org/10.1007/s00011-013-0602-7.
[16] NOM-062-ZOO,1999, Official Mexican Norm for Animal Care and Management. [40] F. Moreno-Perez, A. Hernandez-Leon, M.G. Valle-Dorado, A. Cano-Martínez,
Secretaría de salud. Especificaciones técnicas para la producción, cuidado y uso de F. Narváes-González, E. Aguirre-Hernández, H. Salgado-Ceballos, M.E. González-
los animales de laboratorio. 〈https://www.gob.mx/senasica/documentos/nom- Trujano, Neo-clerodane diterpenic influence in the antinociceptive and anti-
062-zoo-1999〉. inflammatory properties of Salvia circinnata Cav, J. Ethnopharmacol. 268 (2021),
[17] A. Tjølen, O.G. Berge, S. Hunskaar, J.H. Rosland, K. Hole, The formalin test: an 113550, https://doi.org/10.1016/j.jep.2020.113550.
evaluation of the method, Pain 51 (1992) 5–17, https://doi.org/10.1016/0304- [41] D.E. Webster, Y. He, S.N. Chen, G.F. Pauli, N.R. Farnsworth, Z.J. Wang, Opioidergic
3959(92)90003-T. mechanisms underlying the actions of Vitex agnus-castus L, Biochem. Pharmacol.
[18] S. Hunskaar, O.G. Berge, K. Hole, Dissociation between antinociceptive and anti- 81 (2011) 170–177, https://doi.org/10.1016/j.bcp.2010.09.013.
inflammatory effects of acetylsalicylic acid and indomethacin in the formalin test, [42] K. Hara, Y. Haranishi, T. Terada, Y. Takahashi, M. Nakamura, T. Sata, Effects of
Pain 25 (1986) 125–132, https://doi.org/10.1016/0304-3959(86)90014-X. intrathecal and intracerebroventricular administration of luteolin in a rat
[19] N.B. Eddy, D. Leimbach, Synthetic analgesics. II. Dithienylbutenyl- and neuropathic pain model, Pharmacol. Biochem. Behav. 125 (2014) 78–84, https://
dithienylbutylamines, J. Pharm. Exp. Ther. 107 (1953) 385–393. doi.org/10.1016/j.pbb.2014.08.011.
[20] J. Verano, M.E. González-Trujano, M. Déciga-Campos, R. Ventura-Martínez, [43] O. Guadarrama-Enríquez, M.E. González-Trujano, R. Ventura-Martínez,
F. Pellicer, Ursolic acid from Agastache mexicana aerial parts produces R. Rodríguez, G.E. Ángeles-López, R. Reyes-Chilpa, D.A. Moreno, Broccoli sprouts
antinociceptive activity involving TRPV1 receptors, cGMP and a serotonergic produce abdominal antinociception but not spasmolytic effects like its bioactive
synergism, Pharmacol. Biochem. Behav. 110 (2013) 255–264, https://doi.org/ metabolite sulforaphane, Biomed. Pharmacother. 107 (2018) 1770–1778, https://
10.1016/j.pbb.2013.07.020. doi.org/10.1016/j.biopha.2018.09.010.
[21] A. Hernandez-Leon, A. Fernández-Guasti, M.E. González-Trujano, Rutin [44] M.K. Chung, S.J. Jung, S.B. Oh, Role of TRP channels in pain sensation, Adv. Exp.
antinociception involves opioidergic mechanism and descending modulation of Med. Biol. 704 (2011) 615–636, https://doi.org/10.1007/978-94-007-0265-3_33.
ventrolateral periaqueductal grey matter in rats, Eur. J. Pain. 20 (2016) 274–283, [45] M. Cui, P. Honore, C. Zhong, D. Gauvin, J. Mikusa, G. Hernandez, P. Chandran,
https://doi.org/10.1002/ejp.720. A. Gomtsyan, B. Brown, E.K. Bayburt, K. Marsh, B. Bianchi, H. McDonald,
[22] OECD. Test No. 425: Acute Oral Toxicity: Up-and-Down Procedure. OECD W. Niforatos, T.R. Neelands, R.B. Moreland, M.W. Decker, C.H. Lee, J.P. Sullivan,
Guidelines for the Testing of Chemicals, Section 4, 2008. OECD Publishing, Paris. C.R. Faltynek, TRPV1 receptors in the CNS play a key role in broad-spectrum
〈https://doi.org/10.1787/9789264071049-en〉. analgesia of TRPV1 antagonists, J. Neurosci. 26 (2006) 9385–9393, https://doi.
[23] M. Rowland, T.N. Tozer, Clinical Pharmacokinetics: Concepts and Applications, org/10.1523/JNEUROSCI.1246-06.2006.
second ed., Lea & Febiger, Philadelphia, 1989. [46] A. Hernández-Pacheco, C.I. Araiza-Saldaña, V. Granados-Soto, T. Mixcoatl-Zecuatl,
[24] H. Itokawa, K. Matsumoto, H. Morita, K. Takeya, Cytotoxic naphtoquinones from Possible participation of the nitric oxide-cyclic GMP-protein kinase G-K+ channels
Mansoa alliacea, Phytochem 31 (1992) 1061–1062, https://doi.org/10.1016/0031- pathway in the peripheral antinociception of melatonin, Eur. J. Pharmacol. 596
9422(92)80077-R. (2008) 70–76, https://doi.org/10.1016/j.ejphar.2008.07.068.
[25] J.A. Hasrat, J.P. De Backer, G. Vauquelin, A.J. Vlietinck, Medicinal plants in [47] Y. Cury, G. Picolo, V. Pacciari, S. Henrique, Pain and analgesia: The dual effect of
Suriname: screening of plant extracts for receptor binding activity, Phytomedicine nitric oxide in the nociceptive system, Nitric Oxide 25 (2011) 243–254, https://
4 (1997) 59–65, https://doi.org/10.1016/S0944-7113(97)80029-3. doi.org/10.1016/j.niox.2011.06.004.
[26] C. Desmachelier, M. Repetto, J. Coussio, S. Llesuy, G. Ciccia, Total reactive [48] N. Gamper, L. Ooi, Redox and nitric oxide-mediated regulation of sensory neuron
antioxidant potential (TRAP) and total antioxidant reactivity (TAR) of medicinal ion channel function, Antioxid. Redox Signal. 22 (2015) 486–504, https://doi.org/
plants used in Southwest Amazonia (Bolivia and Peru), Int. J. Pharm. 35 (1997) 10.1089/ars.2014.5884.
1–9, https://doi.org/10.1076/phbi.35.4.288.13303. [49] A.R.A. Rodrigues, I.D.G. Duarte, The peripheral antinociceptive effect induced by
[27] T.J. Coderre, F.V. Abbott, J. Sawynok, Formalin Test, in: G.F. Gebhart, R.F. morphine is associated with ATP-sensitive K(+) channels, Br. J. Pharm. 129 (2000)
Schmidt (Eds), Encyclopedia of Pain, Springer, Berlin, Heidelberg, 2013, pp. 110–114, https://doi.org/10.1038/sj.bjp.0703038.
1303–1308. 〈https://doi.org/10.1007/978–3-642–28753-4〉. [50] F. Gomes, F.Q. Cunha, T.M. Cunha, Peripheral nitric oxide signaling directly blocks
[28] P.B. Murphy, S. Bechmann, M.J. Barrett, Morphine. [Updated 2021 May 30]. In: inflammatory pain, Biochem. Pharm. 176 (2020), 113862, https://doi.org/
StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 Jan-. 10.1016/j.bcp.2020.113862.
Available from: 〈https://www.ncbi.nlm.nih.gov/books/NBK526115/〉. [51] V. Granados-Soto, M.D.O. Rufino, L.D. Gomes Lopes, S.H. Ferreira, Evidence for the
[29] L.A. Gobo, C. Viana, A. Lameira, L.M. Carvalho, A liquid chromatography- involvement of the nitric oxide-cGMP pathway in the antinociception of morphine
atmospheric pressure photoionization tandem mass spectrometric (LC-APPI-MS/ in the formalin test, Eur. J. Pharmacol. 340 (1997) 177–180, https://doi.org/
MS) method for the determination of triterpenoids in medicinal plant extracts, 10.1016/s0014-2999(97)01399-x.
J. Mass. Spectrom. 51 (2016) 558–565, https://doi.org/10.1002/jms.3783. [52] M.H. Abdul Rahim, Z.A. Zakaria, M.H. Mohd Sani, M.H. Omar, Y. Yakob, M.
[30] J. Luo, Z.L. Wu, X.J. Huang, X.Q. Zhang, C.L. Fan, W.C. Ye, Y. Wang, Two new S. Cheema, S.M. Ching, Z. Ahmad, A. Abdul Kadir, Methanolic Extract of
pyranonaphthoquinones from Mansoa alliacea, Zhongguo Zhong Yao Za Zhi 46 Clinacanthus nutans exerts antinociceptive activity via the opioid/nitric oxide-
(2021) 3364–3367, https://doi.org/10.19540/j.cnki.cjcmm.20210317.201. mediated, but cGMP-Independent, pathways, Evid. Based Complement. Altern.
[31] M. Olivera-Condori, J. Flores-Arizaca, T. Vásquez-Zavaleta, E. Ocsa-Borda, Med (2016), 1494981, https://doi.org/10.1155/2016/1494981.
Propiedades Fisicoquímicas y Bioactivas in Vitro del Aceite Esencial de Mansoa [53] J.H. Lee, H.Y. Zhou, S.Y. Cho, Y.S. Kim, Y.S. Lee, C.S. Jeong, Anti-inflammatory
alliacea (LAM.) A. Gentry, Ceprosimad 2 (2013) 96–102. 〈https://journal.cepro mechanisms of apigenin: inhibition of cyclooxygenase-2 expression, adhesion of
simad.com/index.php/ceprosimad/article/view/12〉. monocytes to human umbilical vein endothelial cells, and expression of cellular
[32] J.E. Grovas Llamocca, E.A. Condor Cuyubamba, V.M. Reyna Pinedo, I.E. Collantes adhesion molecules, Arch. Pharm. Res. 30 (2007) 1318–1327, https://doi.org/
Díaz, Esteroles presentes en el extracto apolar de las raíces de ajo sacha Mansoa 10.1007/BF02980273.
alliacea, Rev. Soc. Quím. Perú 84 (2018) 513–521. 〈http://www.scielo.org.pe/sciel [54] A. Erdogan, A.K. Most, B. Wienecke, A. Fehsecke, C. Leckband, R. Voss, M.T. Grebe,
o.php?script=sci_arttext&pid=S1810-634X2018000400011&lng=es&nrm=iso〉. H. Tillmanns, C.A. Schaefer, C.R. Kuhlmann, Apigenin-induced nitric oxide
[33] M.M. Pinheiro, F. Boylan, P.D. Fernandes, Antinociceptive effect of the Orbignya production involves calcium-activated potassium channels and is responsible for
speciosa Mart. (Babassu) leaves: evidence for the involvement of apigenin, Life Sci. antiangiogenic effects, J. Thromb. Haemost. 5 (2007) 1774–1781, https://doi.org/
91 (2012) 293–300, https://doi.org/10.1016/j.lfs.2012.06.013. 10.1111/j.1538-7836.2007.02615.x.
[34] G.A. El-Shoubaky, M.M. Abdel-Daim, M.H. Mansour, E.A. Salem, Isolation and [55] M. Aswar, V. Patil, Ferulic acid ameliorates chronic constriction injury induced
identification of a flavone apigenin from marine red alga Acanthophora spicifera painful neuropathy in rats, Inflammopharmacology 24 (2016) 181–188, https://
with antinociceptive and anti-inflammatory activities, J. Exp. Neurosci. 10 (2016) doi.org/10.1007/s10787-016-0272-5.
21–29, https://doi.org/10.4137/JEN.S25096.
[35] K. Hara, Y. Haranishi, K. Kataoka, Y. Takahashi, T. Terada, M. Nakamura, T. Sata,
Rosa Ventura-Martínez, Departamento de Farmacología, Facultad de Medicina, Uni­
Chlorogenic acid administered intrathecally alleviates mechanical and cold
versidad Nacional Autónoma de México, Av. Universidad No. 3000, Col. Ciudad Uni­
hyperalgesia in a rat neuropathic pain model, Eur. J. Pharm. 723 (2014) 459–464,
versitaria, alcaldía Coyoacán, 04510, Ciudad de México, México. Tel.: + 52 5 55 6232162;
https://doi.org/10.1016/j.ejphar.2013.10.046.
fax: + 52 5 55 6232162. Email: rventuram7 @hotmail.com, rventuram@comunidad.
[36] D. Bagdas, Z. Gul, J.A. Meade, B. Cam, N. Cinkilic, M.S. Gurun, Pharmacologic
unam.mx
overview of chlorogenic acid and its metabolites in chronic pain and inflammation,
Curr. Neuropharmacol. 18 (2020) 216–228, https://doi.org/10.2174/
1570159X17666191021111809. María Eva González-Trujano, Dirección de Investigaciones en Neurociencias, Instituto
[37] Y. Xu, D. Lin, X. Yu, X. Xie, L. Wang, L. Lian, N. Fei, J. Chen, N. Zhu, G. Wang, Nacional de Psiquiatría “Ramón de la Fuente Muñiz”, Av. México-Xochimilco 101, Col. San
X. Huang, J. Pan, The antinociceptive effects of ferulic acid on neuropathic pain: Lorenzo Huipulco, 14370, Ciudad de México, México. Email: evag@imp.edu.mx

11

You might also like