You are on page 1of 10

Chemico-Biological Interactions 311 (2019) 108790

Contents lists available at ScienceDirect

Chemico-Biological Interactions
journal homepage: www.elsevier.com/locate/chembioint

Modulation of COX-2, INF-ɣ, glutamatergic and opioid systems contributes T


to antinociceptive, anti-inflammatory and anti-hyperalgesic effects of bis(3-
amino-2-pyridine) diselenide
Angélica S. Reisa, Ane G. Vogta, Mikaela P. Pinza, Guilherme T. Vossa, Caren A.R. da Fonsecaa,
Jaini J. Paltiana, Thiago J. Peglowb, Rodrigo A. Vaucherc, Joanna V.Z. Echeniqued,
Mauro P. Soaresd, Ricardo F. Schumacherb, Gelson Perinb, Cristiane Luchesea,**,
Ethel A. Wilhelma,*
a
Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio)- Grupo de Pesquisa em Neurobiotecnologia, CCQFA, Universidade Federal de Pelotas (UFPel), Pelotas,
RS, Brazil
b
Laboratório de Síntese Orgânica Limpa (LASOL), CCQFA, Universidade Federal de Pelotas (UFPel), Pelotas, RS, Brazil
c
Laboratório de Pesquisa em Bioquímica e Biologia Molecular de Micro-organismos (LaPeBBioM), Universidade Federal de Pelotas (UFPel), Pelotas, RS, Brazil
d
Laboratório Regional de Diagnóstico, Faculdade de Veterinária, Universidade Federal de Pelotas(UFPel), Pelotas, RS, Brazil

A R T I C LE I N FO A B S T R A C T

Keywords: Preclinical assays play a key role in research in research on the neurobiology of pain and the development of
Nociception novel analgesics. Drugs available for the treatment of inflammatory pain are not fully effective and show adverse
Analgesic effects. Thus, we investigated the antinociceptive, anti-inflammatory and anti-hyperalgesic effects of bis(3-
Anti-edematogenic amino-2-pyridine) diselenide (BAPD), a new analgesic drug prototype. BAPD effects were investigated using
Allodynia
nociception models induced by chemical (glutamate), immunologic (Freund's Complete Adjuvant - CFA) and
Selenium
thermal stimuli in Swiss mice. Mice were orally (p.o.) treated with BAPD (0.1–50 mg/kg) 30 min prior to the
Pyridine
glutamate and hot-plate tests and a time–course (0.5 up to 8 h) of the antinociceptive effect of BAPD (50 mg/kg,
p. o.) was evaluated in a CFA model. In the CFA model, BAPD effects on cyclooxygenase-2 (COX-2), tumor
necrosis factor (TNFα) and interferon-γ (INF-γ) expression, myeloperoxidase (MPO) activity, oxidative (2,2′-
Azino-bis-3-ethylbenzothiazoline 6-sulfonic acid and 2,2-diphe- nyl-1-picrylhydrazyl levels) and histological
parameters were evaluated. The safety of the compound (50 and 300 mg/kg, p. o.) was verified for 72 h. BAPD
reduced the licking time induced by glutamate and caused an increase in latency response to thermal stimulus.
Naloxone reversed the antinociceptive effect of BAPD. Paw edema formation induced by glutamate or CFA
injection was reduced by BAPD. Mechanical hyperalgesia induced by CFA was attenuated by BAPD. BAPD did
not protect against the increase in MPO activity and decrease of the 2,2′-Azino-bis-3-ethylbenzothiazoline 6-
sulfonic acid and 2,2-diphe- nyl-1-picrylhydrazyl levels induced by CFA. BAPD protected against histological
alterations and reduction on the levels of gene expression COX-2 and INF-γ in the paw of mice exposed to CFA.
BAPD was safe at the doses and time evaluated. BAPD exerts acute antinociceptive, anti-inflammatory and anti-
hyperalgesic actions, suggesting that it may represent an alternative in the future development of new ther-
apeutic strategies.

1. Introduction emotional experience associated with actual or potential tissue damage.


In fact, it represents an essential adaptive response to protect the in-
According to epidemiological studies, the worldwide prevalence of tegrity of the organism. However, pain can also be generated by ma-
pain is approximately 30% [1,2]. The International Association for the ladaptive responses of the organism, affecting daily activities and
Study of Pain (IASP) defines pain as an unpleasant sensory and quality of life [3].

*
Corresponding author. Programa de Pós-graduação em Bioquímica e Bioprospecção (PPGBBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos,
Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, CEP 96010-900, RS, Brazil.
**
Corresponding author.
E-mail addresses: cristiane_luchese@yahoo.com.br (C. Luchese), ethelwilhelm@yahoo.com.br (E.A. Wilhelm).

https://doi.org/10.1016/j.cbi.2019.108790
Received 3 May 2019; Received in revised form 23 July 2019; Accepted 7 August 2019
Available online 07 August 2019
0009-2797/ © 2019 Elsevier B.V. All rights reserved.
A.S. Reis, et al. Chemico-Biological Interactions 311 (2019) 108790

Several drugs are available to treat pain and inflammation [4,5]. maleate (MK-801) was purchased from Sigma (St. Louis, MO, USA).
However, major adverse effects limit the use of these drugs [6]. In this Mice received oral treatment (p.o.) by intragastric gavage at a constant
context, the development of new therapeutic agents for the treatment volume of 10 ml/kg body weight.
pain and inflammation is of great interest. In this sense, our research
group has dedicated itself to studying the synthesis and pharmacolo- 2.2. Animals
gical properties of new compounds with anti-inflammatory and an-
algesic activities [7–9]. Our strategy is based on the design of new All experiments were performed in accordance with the guidelines
compounds objectifying structural improvement. of the Committee on Care and Use of Experimental Animal Resources of
Some authors have suggested that the chemical structure of orga- the Federal University of Pelotas, Brazil, and were approved by the
nochalcogens plays an important role in establishing their biological same Committee (CEEA 1289–2016) and in accordance with the U.K.
applications [10–12]. Although research groups have studied the Animals (Scientific Procedures) Act, 1986 and associated guidelines.
pharmacological properties of organochalcogens [13,14], the study of The experiments were carried out using male adult Swiss mice
pyridin derivatives containing selenium has been little explored. (25–30 g). Animals were maintained at 22 ± 2 °C with free access to
Indeed, a growing body of evidence demonstrates that pyridine water and food, under a 12:12 h light/dark cycle (with lights on at 6:00
compounds have attracted much attention in the field of drug devel- a.m.). The mice were acclimatized to the behavior room for at least 1 h
opment [15–18]. It should be considered that several studies have ad- before the test. Every effort was made to minimize the number of ani-
dressed the potential antinociceptive and anti-inflammatory effects of mals used and their discomfort. The size of the group used for each
pyridine derivatives [19–21]. Similarly, organoselenium compounds experiment was based on studies that used protocols similar to those
exhibit several pharmacological properties, such as antioxidant, anti- proposed here. The number of animals and intensities of noxious sti-
allodynic, anti-hyperalgesic, antinociceptive and anti-inflammatory muli used were the minimum needed to demonstrate the consistent
actions [7,22–26]. It is important to highlight the advantages of re- effects of the treatments. The allocation was concealed using a rando-
searching organoselenium compounds. Among others, antioxidant ac- mization procedure (http://www.randomizer.org/). Behavioral eva-
tion is an important activity because oxidative stress is critical to the luations were performed blindly on drug administration. Each experi-
etiology of many diseases. Therefore, the use of compounds with a ment was repeated two to three times (using two or three animals for
potent antioxidant effect, such as organoselenium compounds, has re- each repetition) and experiments were carried out between 08:00 and
ceived attention from researchers, because they are promising multi- 17:00 h.
target therapeutic agents in the animal model [7,10,13,22]. A literature
survey indicates that only few publications have mentioned the in- 2.3. Experimental protocol
corporation of a selenium atom in the pyridine nucleus. Consequently,
synthesis and biological screening of selenopyridine derivatives may be 2.3.1. Glutamate-induced nociception
considered a relevant research area. Among the selenopyridine deri- The glutamate test was chosen for this study because it is largely
vatives, bis(3-amino-2-pyridine) diselenide (BAPD), a compound ob- used to screen new drugs with an antinociceptive effect. The procedure
tained by simple and environmentally benign protocols, has been stu- used was similar to that described previously [28]. Mice were treated
died by our research group and results obtained after preliminary with BAPD (0.1–50 mg/kg, p. o.) or vehicle (canola oil, p. o., 10 ml/kg)
biological assays show that BAPD has an important potential to act 30 min before intraplantar (i.pl.) injection of glutamate (20 μmol/paw,
against oxidative stress and as an inhibitor of acetylcholinesterase ac- 20 μl) into the right hind paw and saline solution (0.9%, w/v; 20 μl/
tivity [27]. paw) into the left paw. Mice were observed individually for 15 min
In this context, considering that (i) the combination of pyridines and following glutamate injection and the amount of time spent licking the
selenium is scarce, and (ii) the designed synthesis of these compounds injected paw was recorded with a chronometer and it was considered as
has a great potential for the creation of new molecules with biological a nociceptive behavior. The BAPD pretreatment time was defined in
applications, we aimed to evaluate the antinociceptive, anti-in- accordance with previous studies that observed the pharmacological
flammatory and anti-hyperalgesic effects of a new pyridine derivative effects of organoselenium compounds 30 min after pretreatment
containing selenium, BAPD (Fig. 1), in mice. The potential acute toxi- [7–9,29–33].
city and mechanisms involved in the pharmacological actions of BAPD After the observation period, the mice were killed, and the edema
were also investigated. was measured. Paws were removed and weighed. The paw edema was
measured by comparing the difference between the weight of the glu-
tamate-injected paw and the weight of the contralateral paw (saline-
2. Materials and methods treated paw).

2.1. Drugs 2.3.2. Evaluation of the involvement of the N-methyl-d-aspartate (NMDA)


receptor in the antinociceptive effect of BAPD
BAPD was prepared and characterized in our laboratory and ana- The possible contribution of the glutamatergic system to the anti-
lysis of the 1H NMR and 13C NMR spectra showed analytical and nociceptive action of BAPD was investigated using a glutamate-induced
spectroscopic data in full agreement with its assigned structure [27]. nociception test [28]. NMDA receptor involvement in the anti-
The chemical purity of BAPD (99.9%) was determined by GC/MS. nociceptive action of BAPD was evaluated using MK-801 (0.02 mg/kg
BAPD was dissolved in canola oil, while all other drugs were dis- intraperitoneal, i. p.), an uncompetitive antagonist of the NMDA re-
solved in a saline solution (0.9%) and were of analytical grade and ceptor. Fifteen min after the MK-801 or vehicle administration (10 ml/
obtained from standard commercial suppliers. (+)-MK-801 hydrogen kg of body weight, i. p.), the animals received BAPD (50 mg/kg, p. o.)
and after 30 min, the glutamate injection. The pretreatment times,
doses and route of administration were selected in accordance with a
previous study [33].

2.3.3. Hot-plate test


The hot-plate test is used as a model of acute non-inflammatory
nociception to investigate the central effect of analgesic drugs. The
Fig. 1. Chemical structure of bis(3-amino-2-pyridine) diselenide (BAPD). procedure was carried out as described by Woolfre and MacDonald

2
A.S. Reis, et al. Chemico-Biological Interactions 311 (2019) 108790

(1944) [34]. It is a behavioral model of nociception where behaviors 2.4. Ex vivo assays, qT-PCR and histological evaluation
such as jumping and hind paw-licking are elicited following a noxious
thermal stimulus. For this, animals were placed in a glass box on a In order to improve the understanding of the mechanisms involved
heated metal plate maintained at 55 ± 1 °C. The latency of nociceptive in the pharmacological properties of BAPD, the CFA model was used
responses such as licking or shaking one of the paws or jumping was according to the protocol described above. Four hours after the BAPD
recorded as the reaction. In order to avoid damage to the paws of an- treatment, the animals were anesthetized and then killed to collect
imals, time standing on the plate was limited to 45 s. The change in samples from both paws that were cut off at the ankle joint and used for
thermal withdrawal latencies was recorded before treatment and at histological evaluation as well as to determine the myeloperoxidase
30 min after treatment with BAPD (0.1–50 mg/kg, p. o.) or vehicle. The (MPO) activity, 2,2′-azino-bis-3-ethylbenzothiazoline 6-sulfonic acid
BAPD pretreatment time was defined in accordance with previous (ABTS) and 2,2-diphenyl-1-picrylhydrazyl (DPPH) radicals levels. For
studies that observed the pharmacological effects of organoselenium the RNA extraction and expression of cyclooxygenase-2 (COX-2), tumor
compounds at 30 min after pretreatment [7–9,29–33]. The delta of la- necrosis factor (TNFα) and interferon-γ (INF-γ), the samples were im-
tency (Δt) was calculated for each animal: Δt (s) = post-treatment la- mediately processed and adequately stored (−80 °C) until the evalua-
tency – pretreatment latency. tion of the expression.

2.3.4. Evaluation of the involvement of the opioid system in the 2.4.1. MPO assay
antinociceptive effect of BAPD MPO activity was assayed according to the method previously de-
Opioids have been reported to cause analgesia in rodents in the hot- scribed [39] with some modifications. The paws were homogenized in
plate test [35,36]. Thus, the possible participation of the opioid system potassium phosphate buffer (20 mmol/l, pH 7.4) containing ethylene-
in the BAPD antinociceptive effect was investigated in the hot-plate test diaminetetraacetic acid (0.1 mmol/l). After the homogenization, sam-
as described above. To this end, the animals were pretreated with na- ples were centrifuged at 900×g at 4 °C for 10 min to yield a low-speed
loxone (5 mg/kg, subcutaneous, s. c.) or saline (0.9% NaCl) 15 min supernatant fraction (S1). Then, S1 fraction was centrifuged again at
before the administration of vehicle (canola oil, p. o., 10 ml/kg), mor- 20,000×g at 4 °C for 15 min to yield a final pellet (P2) that was re-
phine (2.5 mg/kg, s. c.) or BAPD (50 mg/kg, p. o.). The pretreatment suspended in medium containing potassium phosphate buffer
times, doses and route of administration were selected in accordance (50 mmol/l, pH 6.0) and hexadecyltrimethyl ammonium bromide
with a previous study [23]. (0.5%). Samples were finally frozen and thawed three times for the
later enzymatic assay. For the MPO activity measurement, an aliquot of
resuspended P2 (100 μl) was added to a medium containing suspension
2.3.5. Open field test (OFT) medium and N,N,N′,N'-tetramethylbenzidine (1.5 mmol/l). The kinetic
OFT evaluates the general locomotor and exploratory behaviors of analysis of MPO was started after the addition of H2O2 (0.01%), and
mice. The open-field was made of plywood and surrounded by 30 cm - color reaction was measured at 655 nm at 37 °C. Results were expressed
high walls. The floor of the open-field, 45 cm long and 45 cm wide, was as optic density (OD)/mg protein/min.
divided by masking tape markers into 9 squares (3 rows of 3). Thirty
min after the treatments, each animal was placed at the center of the 2.4.2. DPPH and ABTS radicals scavenging activity assay
open field and observed for 4 min to record the locomotor (number of Paw tissue was collected and immediately homogenized with 500 μl
segments crossed with the four paws) and exploratory (number of of potassium chloride (1.15%), and centrifuged (for 10 min, at 200×g,
rearings on the hind limbs) activities [37]. The arena was cleaned with at 4 °C). The ability of the sample to resist oxidative damage was de-
70% ethanol after each session and individual mice were tested only termined as free radical scavenging ability by ABTS and DPPH assays
once. [40,41]. For an ABTS assay, ABTS solution was diluted with phosphate
buffer saline pH 7.4 (PBS) to an absorbance of 730 nm. Then, 1.0 ml of
2.3.6. Mechanical withdrawal threshold and paw edema induced by diluted ABTS solution was mixed with 20 μl of S1. After 6 min, the
complete Freund's adjuvant (CFA) absorbance was measured at 730 nm. For DPPH assay, DPPH solution
Based on the results obtained in the nociception models, to extend was diluted with ethanol to an absorbance of 517 nm. Then, 1.0 ml of
our knowledge of the pharmacological properties of BAPD, it was tested diluted DPPH solution was mixed with 20 μl of S1. After 6 min, the
in an inflammatory pain model. The effect of BAPD was investigated in absorbance was measured at 517 nm. The results were equated against
an immunologic reaction induced by i. pl. injection of CFA. The me- an ascorbic acid standard curve. The results were expressed as
chanical hyperalgesia was measured as described by Bortolanza et al. equivalents of ascorbic acid per gram of tissue weight in both assays.
(2002) [38]. The frequency of withdrawal was determined before
(baseline) CFA injection, in order to obtain data purely derived from the 2.4.3. RNA extraction and expression of COX-2, TNFα and INF-γ by real-
treatments in CFA allodynia. In this experiment, the time-course of the time PCR
antinociceptive effect of BAPD was evaluated at the dose of 50 mg/kg The total RNA of paw tissue was directly isolated after the com-
(p.o.). Mice received the i. pl. injection of CFA (1 mg/ml of heat killed pletion of treatment using TRIZOL® (Invitrogen, Carlsbad, CA) ac-
Mycobacterium tuberculosis in 85% paraffin oil and 15% mannide cording to the manufacturer's instructions. Total RNA was treated with
monoleate, 20 μl) into the right hind paw and saline solution (0.9%, w/ RNase-free DNase (Invitrogen, Carlsbad, CA) and its quality was as-
v; 20 μl/paw) into the left paw. The inflammatory response was verified sessed by running samples on a 1% formaldehyde agarose gel. RNA was
24 h after CFA injection into the i. pl. surface of the right hind paw in quantified spectrophotometrically. The primers used for the real-time
mice. Therefore, mice that presented hyperalgesia received vehicle PCRs were synthesized by Invitrogen (São Paulo, Brazil). The real-time
(canola oil, p. o., 10 ml/kg) or BAPD (50 mg/kg, p. o.) and the with- PCR amplification reaction was carried out using SYBR® Green One-
drawal response frequency in von frey filament (VHF) was recorded Step qRT-PCR with Rox (Invitrogen, Carlsbad, CA), performed ac-
0.5, 2, 4, 6 and 8 h after treatments. Mechanical hyperalgesia in mice cording to the manufacturer's protocol. cDNA was synthesized from
was measured by using a calibrated nylon VHF of 1 g. 0.5 μg of total RNA, using the specific forward and reverse primers
After the test, the animals were killed, and both paws were cut at (20 μM) for each gene. PCR reactions were run in a 7500 Real time Fast
the ankle joint and immediately weighed on an analytical balance. The thermocycler (Applied Biosystems), under the following conditions:
paw edema was measured by comparing the difference between the 50 °C for 15 min, 95 °C for 52 min, followed by 40 cycles at 95 °C for
weight of the CFA-treated paw and the weight of the contralateral paw 15 s and 60 °C for 30 s, after the dissociation curve was performed at
(vehicle - treated paw). 95 °C for 5 min with a final step of 4 °C. The assay was accomplished for

3
A.S. Reis, et al. Chemico-Biological Interactions 311 (2019) 108790

each gene and included cDNA of the samples treated and control incubation was stopped by adding 10% TCA solution with 10 mM
without template. HgCl2. The reaction product porphobilinogen (PBG) was measured at
Results were obtained as CT (threshold cycle) values. The software 555 nm using modified Ehrlich's reagent. The values were expressed as
determines a threshold line at the base of the baseline fluorescent nmol PBG/mg protein/h.
signal, and the data point that meets the threshold is given as CT, which
is inversely proportional to the starting template copy number. The 2.6. Statistical analysis
differences in CT values between the control group, treated group and
endogenous control β-actin gene for each reaction (ΔCT) were analyzed The data and statistical analysis comply with the recommendations
using the 2−ΔΔCT method. The relative expression genes after incuba- on experimental design and analysis in pharmacology [49]. The raw
tion were determined by dividing expression units of the treated group data supporting the conclusions of this manuscript will be made
by the control group. All measurements were performed in duplicate in available by the authors, without undue reservation, to any qualified
two independent experiments. The results were expressed as relative researcher. The normality of data was evaluated by the D'Agostino and
concentration calculated as described by Giongo et al. (2017) [42]. Pearson omnibus normality test. Statistical analysis was performed
using GraphPad Prism 6.0 software (San Diego, CA, USA). Data were
2.4.4. Protein determination analyzed by one-way or two-way analysis of variance (ANOVA) fol-
The protein concentration was measured by the method of Bradford lowed by the Newman–Keuls or Tukey's test when appropriate. Main
(1976) [43], using bovine serum albumin as the standard. effects are presented only when the higher second order interaction was
non-significant. For mechanical withdrawal threshold assays the data
2.4.5. Histological examination were analyzed using a non-paired t-test followed by the Mann-Whitney
Paws were collected and fixed by immersion in 10% buffered for- test. Data were expressed as mean ± standard error of the mean
malin solution for 24 h. Subsequently, the hind paws were decalcified in (S.E.M.). Probability values less than 0.05 (P < 0.05) were considered
8% hydrochloric acid and formic acid solution at a ratio of 1:1 over a statistically significant. Post hoc tests were performed only when the F-
period of approximately 5 days. The samples were crosscut and routi- value achieved the necessary level of statistical significance (P < 0.05)
nely processed, embedded in paraffin, cut into of 3–4 μm sections, and when there was no significant variance in homogeneity [49].
stained with hematoxylin-eosin (HE) and examined under an optical
microscope by two independent pathologists.
3. Results
2.5. Acute toxicity
3.1. Experimental protocol
For evaluation of acute toxicity, mice received a single oral dose of
BAPD (50 and 300 mg/kg) or vehicle. After drug administration, ani- 3.1.1. Glutamate-induced nociception
mals were observed for up to 72 h to determine the lethal potential of The antinociceptive effects exerted by BAPD in the glutamate test
this compound. After this time of exposure, mice were anesthetized for are presented in Table 1. The one-way ANOVA revealed that pretreat-
blood collection by heart puncture in tubes containing heparin. Plasma ment with BAPD reduced the nociception induced by glutamate (F
was obtained by centrifugation at 900×g for 10 min and used for bio- (4,25) = 78.91, P < 0.0001). The BAPD antinociceptive effect on the

chemical assays, which were performed with commercial tests kits. glutamate test was evidenced at the doses of 0.1–50 mg/kg and it was
Plasma aspartate (AST) and alanine aminotransferase (ALT) activities, effective in reducing the licking time induced by glutamate in 100%
used as the biochemical markers for the early acute hepatic damage, (P < 0.0001) at the dose of 50 mg/kg.
were determined by the colorimetric method [44]. Renal function was Additionally, the pretreatment with BAPD reduced the paw edema
analyzed by determining plasma urea [45]. AST and ALT activities were formation induced by glutamate injection in 58% (5 mg/kg;
expressed as units (U)/l and urea levels were expressed as mg/dl. P < 0.0001), 50% (25 mg/kg; P < 0.0001) and 36% (50 mg/kg;
Samples of kidney, liver and brain were also collected to determine P < 0.0001) (F (4,25) = 26.28, P < 0.0001) (Table 1).
non-protein thiols (NPSH) and thiobarbituric acid reactive species
(TBARS) levels and δ-aminolevulinate dehydratase (δ-ALA-D) activity. 3.1.2. Evaluation of the involvement of the NMDA receptor in the
Samples were homogenized in 50 mM Tris/HCl pH 7.5 (1:10 for liver/ antinociceptive effect of BAPD
kidney and 1:5 for brain, w/v) and centrifuged at 900×g for 10 min at Fig. 2A llustrates the effect of BAPD administration on glutamate
4 °C to yield a S1. test after the pretreatment of mice with MK-801. The two-way ANOVA
TBARS levels, a measure of lipid peroxidation, were determined as revealed non significant BAPD x MK-801 (F (1,20) = 0.1458, P > 0.05)
described by Ohkawa et al. (1976) [46]. An aliquot of S1 was incubated interaction, but significant main effect of BAPD (F (1,20) = 74.01,
with 0.8% thiobarbituric acid (TBA), acetic acid buffer pH 3.4 and 8.1% P < 0.0001). However, the pretreatment of animals with MK-801 did
sodium dodecil sulphate at 95 °C for 2 h. The color reaction was mea-
sured at 532 nm. TBARS levels were expressed as nmol mal- Table 1
ondialdehyde (MDA)/mg protein. Effects of BAPD on glutamate and hot plate tests in mice.
NPSH levels were determined as described by Ellman (1968) [47]. Glutamate test Hot-plate test
An aliquot of S1 was mixed (1:1) with 10% trichloroacetic acid (TCA)
and centrifuged at 900×g for 10 min. After the centrifugation, the Groups Licking (s) Paw Edema (mg) Δ Latency (s)
protein pellet was discarded and free –SH groups were determined in
Induced 101.5 ± 9.1 78.7 ± 2.6 −6.0 ± 1.9
the clear supernatant. An aliquot of S1 was added in 1 M potassium BAPD
phosphate buffer, pH 7.4, and 10 mM 5,5-dithiobis (2-nitrobenzoic 0.1 mg/kg 74.0 ± 6.3*** 68.9 ± 5.6 5.6 ± 2.3
acid) (DTNB). The color reaction was measured at 412 nm. NPSH levels 5 mg/kg 1.0 ± 4.1**** 33.5 ± 3.2**** −0.5 ± 0.9
were expressed as μmol NPSH/g tissue. 25 mg/kg 0.0 ± 0.8**** 39.2 ± 4.0**** 17.6 ± 3.6***
50 mg/kg 0.0 ± 0.0**** 49.4 ± 2.3**** 12.7 ± 6.0**
δ-ALA-D activity was assayed by the method of Sassa (1982) [48].
An aliquot of S1 was pre-incubated for 10 min at 37 °C. The enzymatic Each column represents the mean ± S.E.M. of 6 mice in each group. BAPD
reaction was initiated by adding the substrate (δ-ALA) to a final con- means bis(3-amino-2-pyridine)diselenide. Asterisks denote significance levels
centration of 2.2 mM in a medium containing 45 mM phosphate buffer, when compared with the induced group (**) P < 0.01; (***) P < 0.001 and
pH 6.8 and incubated for 3 h (brain) or 1 h (liver/kidney) at 37 °C. The (****) P < 0.0001 (one-way ANOVA followed by the Newman– Keuls' test).

4
A.S. Reis, et al. Chemico-Biological Interactions 311 (2019) 108790

Fig. 2. Investigation of the involvement of NMDA receptor and opioid system in


the antinociceptive action of bis(3-amino-2-pyridine) diselenide (BAPD) in
mice. (A) Effect of MK-801 (0.02 mg/kg, i. p.) on the antinociceptive action of
BAPD (50 mg/kg, p. o.) on the licking behavior induced by glutamate in mice;
(B) effect of naloxone (5 mg/kg, s. c.) on the antinociceptive action of BAPD
(50 mg/kg, p. o.) or morphine (2.5 mg/kg, s. c.) on the hot-plate test. Each
column represents the mean ± S.E.M. of 6 mice in each group. (****)
P < 0.0001 and (**) P < 0.01 denotes significance levels when compared with
the control group, (##) P < 0.01 denotes significance levels when compared
with the MK-801 group, (&&) P < 0.01 and (&&&&) P < 0.0001 denotes sig-
nificance levels when compared with the naloxone group (Two-way ANOVA
followed by the Newman– Keuls' test).

not reverse the antinociceptive effect of BAPD (50 mg/kg) in the glu- Fig. 3. Effect of bis(3-amino-2-pyridine) diselenide (BAPD) (50 mg/kg, p. o.) on
tamate test. mechanical hyperalgesia induced by CFA in the ipsilateral paw (A) and in the
contralateral paw (B) of mice in response to 10 applications of 1.0 g VFH. The
animals received vehicle (●) or BAPD (Δ) 24 h after CFA injection. The baseline
3.1.3. Hot-plate test (□) was recorded before CFA injection. The measure that follows was 24 h after
CFA-injection (0) and (0.5, 1, 2, 4, 6 and 8 h) subsequent to BAPD treatment.
Effects of BAPD administration in a hot-plate test were demon-
(C) Effect of BAPD on edema CFA-induced 4 h subsequent to treatment. Each
strated in Table 1. The one-way ANOVA revealed that the pretreatment
column represents the mean ± S.E.M. of 7 mice in each group. (*) P < 0.05
with BAPD increased response latency to thermal stimulus when com- and (***) P < 0.001 denote significance levels when compared with the control
pared with the control group (F (4,25) = 7.671, P < 0.001). BAPD group and (#) P < 0.05 denotes significance levels when compared with the
showed significant antinociceptive activity at the doses of 25 and baseline group. Statistical analysis was performed by nonpaired t-test followed
50 mg/kg, increasing the latency time by 24% (P < 0.001) and 19% by the Mann Whitney test.
(P < 0.01), respectively.
3.1.5. Open field test (OFT)
The possible effects of treatments on locomotor and exploratory
3.1.4. Evaluation of the involvement of the opioid system in the activities of mice were evaluated in the OFT. The data analysis of OFT
antinociceptive effect of BAPD revealed that no alteration in the number of crossings (F (4,
The two-way ANOVA revealed significant BAPD x naloxone (F
25) = 0.3085, P > 0.05) and rearings (F (4, 25) = 2.369, P > 0.05)
(2,30) = 11.74, P < 0.001) and morphine x naloxone (F (1,20) = 16.83, were observed after the different treatments of mice (supplementary
P < 0.001) interactions. Pretreatment of animals with naloxone re- material).
versed the antinociceptive effect of morphine and BAPD on the hot-
plate test (Fig. 2B).

5
A.S. Reis, et al. Chemico-Biological Interactions 311 (2019) 108790

Table 2 with the induced group (Fig. 4).


Effects of BAPD (50 mg/kg) on oxidative and inflammatory parameters in the
paw of mice exposed to a VHF test induced by CFA in the 4 h after treatment.
3.2.4. Histological examination
MPO (OD/mg DPPH (equivalents of ABTS (equivalents of Briefly, results demonstrated that the mice of the control group had
Groups protein/min) ascorbic acid/g of ascorbic acid/g of preserved tissues and no inflammatory process. Meanwhile, the mice of
tissue) tissue)
the induced group (CFA) showed subcutaneous tissue with marked
Control −0.001 ± 0.0 108.2 ± 5.9 50.2 ± 1.3 expansion, edema, prominent lymphatic vessels dilatation and severe
Induced 0.128 ± 0.0* 89.4 ± 4.2* 34.5 ± 2.9* eosinophilic inflammatory infiltrate. Additionally, the CFA caused ex-
BAPD 0.131 ± 0.0* 89.8 ± 1.9* 40.9 ± 3.4* tensive damage in the articular region of the mice paw, such as thick-
ened synovial membrane, epithelial cell necrosis, hemorrhage, eosino-
Each column represents the mean ± S.E.M. of 6 mice in each group. BAPD
philic inflammatory infiltrate and marked lymphatic vessels dilatation.
means bis(3-amino-2-pyridine) diselenide. BAPD was evaluated at the dose of
50 mg/kg. Asterisks denote significance levels when compared with the control However, mice treated with BAPD demonstrated a partial reversal of
group (*) P < 0.05 (one-way ANOVA followed by the Newman– Keuls' test). the damage caused by the CFA, with reduction of the edema and in-
flammatory process.
3.1.6. Mechanical withdrawal threshold and paw edema induced by CFA As shown in Fig. 5, mice exposed to CFA (Fig. 5B, E, H) presented a
The animals that received BAPD (50 mg/kg, p. o.) demonstrated a metatarsal-phalangeal region that was enlarged twice the control
reduction of mechanical hyperalgesia induced by CFA. This hy- group, (Fig. 5A, D, G). CFA induced the flattening of cutaneous folds
persensitivity reduction started at 0.5 h and remained significant up to due to severe edema and eosinophilic inflammatory infiltrate (Fig. 5B).
6 h after administration of BAPD (50 mg/kg) (Fig. 3A). The maximal The intra-articular space was expanded by severely diffuse eosinophilic
inhibition observed was 91% at 4 h and the sensitivity of animals was inflammatory infiltrate and the articular surfaces were dissociated.
similar to baseline values (Fig. 3A). No significant difference was ob- Cellular debris, edema and hemorrhage were observed in the articular
served in the contralateral paw when compared to the basal (Fig. 3B). space. The synovial membrane was thickened by severe eosinophilic
Additionally, as verified in Fig. 3C, the treatment with BAPD 50 mg/kg inflammatory infiltrate admixed with chondrocyte necrosis, hemor-
reduced the paw edema formation (df = 12; t = 4.109, P < 0.01) in- rhage and lymphatic vessel dilatation (Fig. 5E). The interosseous con-
duced by CFA in 39%. nective and subcutaneous tissues were expanded due to edema, fibrin
and severe eosinophilic inflammatory infiltrate. Additionally, there was
3.2. Ex vivo assays prominent lymphatic vessels dilation and eosinophilic perivasculitis
(Fig. 5B).
3.2.1. MPO assay BAPD pretreatment reduced the severe articulation and metatarsal-
The one-way ANOVA revealed that CFA significantly increased the phalangeal region enlargement, when compared with a CFA group. In
MPO activity (128 times) in mice paws, when compared with the this group, distinct cutaneous folds were present (Fig. 5C). The inter-
control group (F (2,15) = 34.66; P < 0.0001). No protection against the osseous connective and subcutaneous tissue presented less severe his-
increase in the MPO activity was elicited after pretreatment with BAPD tological alterations. Mild lymphatic vessel dilatation, moderate eosi-
(50 mg/kg) in mice (Table 2). nophilic inflammatory infiltrate, edema and in some areas calcification
secondary to necrosis were observed (Fig. 5I). The synovial membrane
3.2.2. DPPH and ABTS radicals scavenging activity assay showed coating epithelial cell loss with less severe congestion and mild
As shown in Table 2, CFA significantly decreased the DPPH (17%) eosinophilic inflammatory infiltrate. There were remaining cellular
(F (2,15) = 5.930, P < 0.05) and ABTS (31%) (F (2,15) = 8.14, debris, hemorrhage and fibrin within the intra-articular space (Fig. 5F).
P < 0.01) levels in mice paws when compared with the control group
(Table 2). The one-way ANOVA demonstrated that BAPD treatment 3.3. Acute toxicity
(50 mg/kg) did not cause changes in the DPPH and ABTS levels in the
paws with the immunologic reaction induced by CFA (Table 2). A single oral administration of BAPD (50 and 300 mg/kg) did not
cause the death of exposed mice. As demonstrated in Table 3, the ac-
3.2.3. COX-2, TNFα and INF-γ of expression level tivities of ALT (F (2, 6) = 0.8086, P > 0.05), AST (F (2, 6) = 2.475,
As shown in Fig. 4, CFA significantly increased the levels of gene P > 0.05) and the levels of urea remained (F (2, 6) = 0.2183,
expression COX-2 (293.2%) (P < 0.0001) and INF-γ (92.1%) P > 0.05) unaltered after BAPD treatment, when compared to those of
(P < 0.001) in mice paws, when compared with the control group. The mice in the control group.
results showed that there was no significant difference in expression of In addition, effects of a single oral administration of BAPD (50 and
TNFα after the treatments. The one-way ANOVA demonstrated that 300 mg/kg) on oxidative stress parameters in the brain, kidney and
BAPD (50 mg/kg, p. o.) reduced the levels of gene expression COX-2 liver of mice are shown in Table 4. As revealed by statistical analysis,
(P < 0.0001) and INF-γ (P < 0.05) in mice paws, when compared TBARS (F (2, 6) = 1.738, P > 0.05 for brain; F (2, 6) = 1.647, P > 0.05

Fig. 4. Effect of bis(3-amino-2-pyridine) dis-


elenide (BAPD) (50 mg/kg, p. o.) on levels of ex-
pression of COX-2, TNF-α and INF-γ related genes
in the paw tissue of mice exposed to CFA. The
relative expression ratios are the average values
from three replicate cultures. COX-2, TNF-α and
INF-γ expression levels were normalized using the
geometric mean of β-actin gene. (*) P < 0.05,
(***) P < 0.001 and (****) P < 0.0001 denote
significance levels when compared with the con-
trol group and (#) P < 0.05 and (####)
P < 0.0001 denote significance levels when
compared with the induced group.

6
A.S. Reis, et al. Chemico-Biological Interactions 311 (2019) 108790

Fig. 5. Photomicrograph of the morphological


alterations of mice paws: (A) Control: Tarsal re-
gion. HE. 40X. (B) CFA (induced): Tarsus region.
HE 40X. (C) BAPD + CFA: Tarsus region. HE 40X.
(D) Control: Articular region. HE. 200X. (E) CFA
(induced): Articular region. Thickened synovial
membrane presents epithelial cell necrosis, he-
morrhage, eosinophilic inflammatory infiltrate
and marked lymphatic vessel dilatation (*). The
appearance of the articular surface is rough due to
the irregularity in the membrane surface. In the
articular space, there was cellular debris and he-
morrhage (arrow). HE. 200X. (F) BAPD + CFA:
Articular region. HE. 200X. (G) Control: Tarsus
region. HE. 200X. (H) CFA (induced): Interosseous
connective tissue. Severe eosinophilic in-
flammatory infiltrate admixed with hemorrhage,
edema and fibrin (*). There is marked lymphatic
vessel dilatation (L). HE. 200X. (I) BAPD + CFA:
Interosseous connective tissue. HE. 200X.

Table 3 thermal nociception models, without causing locomotor disturbances


Effect of a single oral administration of BAPD (50 and 300 mg/kg) on bio- and toxicity. Our data indicate that opioid and glutamatergic systems
chemical parameters in mice. are related to the antinociceptive properties of BAPD in mice. In ad-
Groups AST (U/l) ALT (U/l) Urea (mg/dl) dition, CFA injection induced hyperalgesia, allodynia and histological
alterations in the mice paws, and these alterations were reversed by the
Control 77.2 ± 10.4 104.5 ± 27.2 70.2 ± 9.5 BAPD treatment. Additionally, we showed that BAPD reduced the in-
BAPD
flammatory process induced by CFA by reducing levels of gene ex-
50 mg/kg 66.6 ± 9.4 57.3 ± 3.4 61.9 ± 1.6
300 mg/kg 49.7 ± 5.5 90.5 ± 37.7 68.8 ± 13.4 pression COX-2 and INF-γ.
Glutamate and its receptors play a crucial role in perception and
Data are reported as the mean ± S.E.M. of 3 mice in each group. BAPD means integration of nociceptive signals and in their relay to supraspinal
Bis(3-amino-2-pyridine) diselenide. Statistical analysis was performed using centers. Indeed, glutamate is widely distributed throughout virtually all
one-way ANOVA followed by the Newman– Keuls' test. circuits in the entire nervous system and also co-localized with its re-
ceptors in areas of the brain, spinal cord and periphery that are in-
for kidney; F (2, 6) = 0.4617, P > 0.05 for liver) and NPSH (F (2, volved in pain sensation and transmission [50,51]. In this regard, in-
6) = 0.001852, P > 0.05 for brain; F (2, 6) = 0.2459, P > 0.05 for itially, we sought to determine the antinociceptive activity of BAPD
kidney; F (2, 6) = 1.206, P > 0.05 for liver)levels and δ-ALA-D activity using a model of nociception induced by glutamate. As expected, i. pl.
(F (2, 6) = 0.3795, P > 0.05 for brain; F (2, 6) = 0.4230, P > 0.05 for injection of glutamate in the mouse hind paw induced a nociceptive
kidney; F (2, 6) = 1.177, P > 0.05 for liver) remained unaltered after response and edema. Agonists at these receptors or subcutaneous in-
BAPD (50 and 300 mg/kg) treatment, when compared to the control jection of glutamate into the hind paw have been shown to reduce pain
group (Table 4). thresholds [52–55], and conversely antagonists promote analgesia in
peripheral pain [56]. Our results revealed that BAPD inhibited the
4. Discussion nociceptive response and edema caused by glutamate injection, sug-
gesting potential antinociceptive and anti-inflammatory actions pos-
The findings of the present study indicated, for the first time, that sibly by modulation of the glutamatergic system. Importantly, some
BAPD exerts antinociceptive, anti-inflammatory and anti-hyperalgesic researchers have demonstrated that other organoselenium compounds
actions. BAPD treatment reversed the nociception in chemical and

Table 4
Effect of a single oral administration of BAPD (50 and 300 mg/kg) on oxidative parameters in brain, kidney and liver of mice.
NPSH (μmol/g tissue) TBARS (nmol MDA/mg protein) δ- ALA-D (nmol PBG/mg protein/h)

Brain Kidney Liver Brain Kidney Liver Brain Kidney Liver

Control 1.73 ± 0.06 2.29 ± 0.09 6.55 ± 0.19 0.62 ± 0.04 1.70 ± 0.08 2.16 ± 0.16 1.09 ± 0.02 0.79 ± 0.10 8.83 ± 0.29
BAPD
50 mg/kg 1.73 ± 0.01 2.18 ± 0.27 5.57 ± 0.63 0.61 ± 0.02 2.21 ± 0.31 2.02 ± 0.01 1.12 ± 0.04 0.78 ± 0.02 8.32 ± 0.53
300 mg/kg 1.73 ± 0.03 2.36 ± 0.13 6.26 ± 0.43 0.50 ± 0.07 1.69 ± 0.23 2.02 ± 0.12 1.07 ± 0.03 0.70 ± 0.07 8.09 ± 0.44

Date are reported as the mean ± S.E.M. of 3 mice in each group. BAPD means bis(3-amino-2-pyridine) diselenide. Statistical analysis was performed using one-way
ANOVA followed by the Newman– Keuls' test.

7
A.S. Reis, et al. Chemico-Biological Interactions 311 (2019) 108790

exhibit potential antinociceptive in the glutamate test [30–33]. The results suggest that BAPD anti-inflammatory, antinociceptive and anti-
antinociceptive effect of organoselenium compounds, at doses equal to hyperalgesic actions are not related to MPO or scavenger activities. Our
or higher than 0.1 mg/kg, in the glutamate test has been reported results on MPO are according to the data published. Chagas et al. [26]
[30,31]. Here, BAPD has a similar or higher antinociceptive potency in demonstrated that CFA increased the MPO activity in the paw tissue of
the glutamate test when compared to other organoselenium com- C57BL/6 mice and, the bis(phenylimidazoselenazolyl) diselenide, an
pounds. In line with these results, the involvement of NMDA receptor in organoselenium compound, did not reverse this alteration.
the antinociceptive action of BAPD was investigated using MK-801 (an As a result of CFA-induced tissue damage several chemical media-
uncompetitive antagonist of the NMDA receptor). Data obtained sug- tors, such as interleukin, TNFα, prostaglandin, INF-γ, are released from
gested that the modulation of NMDA receptor did not contribute to the the injured cells and the infiltrating immune cells to create a peripheral
antinociceptive effect of BAPD. However, considering the results in the inflammatory process. In this process, the peripheral terminals of no-
glutamate test, we cannot rule out the involvement of other glutama- ciceptors express receptors for many of these inflammatory mediators.
tergic receptors in this pharmacological action of BAPD. This increase in the peripheral sensitivity of nociceptors in inflamed
Considering that glutamate is released both peripherally and cen- tissue contributes to inflammatory pain hypersensitivity or hyperalgesia
trally in response to nociceptive stimulation [57,58] and the effects of [66–68]. In this case, tissue damage that involves up-regulation of INF-γ
BAPD on the glutamate test, we had objectively evaluated the anti- contributes to inflammation and probably increases the excitability of
nociceptive effect of BAPD on thermal stimulus in the hot-plate test. nociceptive neurons. Earlier studies demonstrated that INF-γ could in-
Acute non-inflammatory nociception is produced in the hot-plate test, duce nitric oxide synthase (iNOS) and activity immune [69,70]. Our
and it is a good model to investigate the central effect of analgesic drugs results revealed that BAPD presented a promising anti-inflammatory
with selectivity for opioid-like drugs [59,60]. Supporting the BAPD effect, since it reduced the paw edema formation induced by CFA and
antinociceptive action, the current findings revealed that BAPD reduced glutamate. This effect can be due to the potential inhibitory effect of
the nociceptive behavior of the thermal stimulus, suggesting a central BAPD on INF-γ, reducing maintenance of the inflammatory process.
action of this compound. In line with these results, it was imperative to Additionally, tissue damage promotes the release of neuro-
understand whether the opioid system is associated with the anti- transmitters from the central terminals of nociceptors and increases the
nociceptive effect of BAPD. Significantly, the pretreatment of animals production of prostaglandin or pro-inflammatory cytokines in the spinal
with naloxone, a nonselective opioid receptor antagonist, reversed the cord. This leads to additional excitation and disinhibition of dorsal horn
antinociceptive effect caused by oral administration of BAPD, demon- neurons and causes abnormal responses to sensory signals from the
strating that the opioid system is implicated in this effect. Indeed, these periphery [71]. In this sense, prostanoids play a pivotal role in in-
results are consistent with previous findings that demonstrated the ef- flammation and pain. The role of COX in inflammatory pain has been
fect of selol, an organoselenium compound, on the opioid system. Bu- extensively studied. Here, it is important to highlight that our results
jalska and collaborators [61] demonstrated that pretreatment with selol suggest that the single oral administration of BAPD markedly reduced
enhanced the analgesic activity of opioids drugs. Evidence for the in- the levels of COX-2 expression in injured tissue suggesting an anti-in-
volvement of μ-opioid and δ-opioid receptors in the antinociceptive flammatory effect of this compound. Indeed, inhibitors of COX, also
effect caused by oral administration of m-trifluoromethyl-diphenyl known as nonsteroidal anti-inflammatory drugs (NSAIDs), exert anti-
diselenide in mice was provided by Brüning and collaborators [25]. The inflammatory action and are mainly used in the treatment of degen-
results indicate that m-trifluoromethyl-diphenyl diselenide elicited an- erative joint disease, rheumatoid diseases, metabolic disorders and
tinociception in different models of pain through mechanisms that seem other diseases that are associated with pain and inflammation [72].
to involve an interaction with the central opioid system, more specifi- This result is in agreement with the study performed by Marcondes Sari
cally μ-opioid and δ-opioid receptors. In addition, m-trifluoromethyl- and collaborators [73] that revealed the effectiveness of the organose-
diphenyl diselenide was effective to abolish the phenotype induced by lenium compound p,p'-methoxyl-diphenyl diselenide in an animal
repeated forced swim stress, which was accompanied by changes in the model of chronic pain by restoring the molecular changes in the COX-2
contents of cortical μ- and κ-opioid receptors [62]. content.
Additionally, a further study was performed to extend our knowl- In addition, the present study revealed that BAPD also was able to
edge about the anti-inflammatory potential of BAPD. A CFA-induced reduce the mechanical allodynia in the inflamed paws and the noci-
inflammatory pain model was used because it effectively mimics some ceptive behavior from thermal stimulus in mice. This can be due to its
characteristics of human arthritis, such as the development of me- inhibitory potential effect in the expression of INF-γ and COX-2, as well
chanical allodynia, edema and induction of inflammation [63]. In the as to modulating the opioid and glutamatergic pathways. In addition,
current study, it was demonstrated that a single oral administration of these results corroborate those found in the histopathological analysis
BAPD produced antinociceptive, anti-hyperalgesic and anti-edemato- that demonstrated a reduction in the inflammatory process since BAPD
genic actions in CFA-induced painful inflammation in mice. Revealing a reduced lymphatic vessel dilatation and eosinophilic inflammatory in-
rapid and long action, BAPD reduced the response frequency of VHF filtrate.
stimulation at 0.5 h and this effect was maintained for up to 6 h with the Here, we also demonstrated that the single oral administration of
maximal inhibition of 91%. The pharmacological actions of other dis- BAPD did not elicit any sign of acute toxicity and did not modify the
elenides in CFA-induced painful inflammation have been demonstrated locomotor and exploratory activities of animals. BAPD, at doses of 50
by other authors [24,26]. and 300 mg/kg, did not cause death or any significant acute tox-
In an attempt to identify the mechanisms involved in these effects of icological effect. Concerning biochemical parameters of hepatic (ALT
BAPD, the MPO activity, DPPH and ABTS radical scavenging activities and AST activities) and renal (urea) function, BAPD did not alter any of
were determined in paws of mice exposed to CFA. Oxidative stress has these parameters in plasma, suggesting that this compound does not
been implicated as a mechanism in the induction and maintenance of have any potentially toxic effect on these organs. δ-ALA-D activity, the
pain and its occurrence has been described in several animal models of parameter of potential organoselenium toxicity [22], was not altered in
pain [60]. Although this hypothesis is strengthened by the fact that brain, kidney and liver by treatment with the single oral administration
some antioxidant substances present antinociceptive effects [7,64], of BAPD (50 and 300 mg/kg). Additionally, BAPD did not modify
here BPAD did not protect against the reduction of DPPH and ABTS TBARS and NPSH levels.
levels induced by CFA. Additionally, concerning the stimulation of MPO
activity found in animals exposed to CFA, our results demonstrated that 5. Conclusion
BAPD did not exert a protective effect. MPO is a heme peroxidase highly
expressed in the neutrophils and activated microglial cells [65]. Our In summary, BAPD induced antinociceptive, anti-hyperalgesic and

8
A.S. Reis, et al. Chemico-Biological Interactions 311 (2019) 108790

anti-inflammatory effects in mice without altering locomotor activity or oxidative damage, Chem. Biol. Interact. 176 (2008) 129–136.
inducing toxicity. These effects of BAPD seem to be due to a modulation [11] B.M. Gai, A.L. Stein, J.A. Roehrs, et al., Synthesis and antidepressant-like activity of
selenophenes obtained via iron(III)-PhSeSePh-mediated cyclization of Z-selenoe-
of opioid and glutamatergic systems as well as to reduction of the levels nynes, Org. Biomol. Chem. 10 (2012) 798–807.
of expression COX-2 and INF-γ. Our results suggest that BAPD may be a [12] A.G. Vogt, G.T. Voss, R.L. de Oliveira, et al., Organoselenium group is critical for
good, more effective and/or potent prototype for the treatment of pain antioxidant activity of 7-chloro-4-phenylselenyl-quinoline, Chem. Biol. Interact.
282 (2018) 7–12.
and inflammation. [13] M. Ibrahim, W. Hassan, D.F. Meinerz, et al., Antioxidant properties of diorganoyl
diselenides and ditellurides: modulation by organic aryl or naphthyl moiety, Mol.
Funding information/acknowledgments Cell. Biochem. 371 (2012) 97–104.
[14] H.L. Seng, E.R. Tiekink, Anti-cancer potential of selenium‐and tellurium‐containing
species: opportunities abound, Appl. Organomet. Chem. 26 (2012) 655–662.
We are grateful to UFPel, CNPq (UNIVERSAL 408874/2016-3, [15] N.R. Challa, B. Mamidisetty, M.R. Ghanta, et al., Synthesis and pharmacological
429859/2018-0) and FAPERGS (PqG 17/2551-0001013-2; PRONEM evaluation of 5-[2′-(1H-tetrazol-5-yl)-biphenyl-4-ylmethyl]-4, 5, 6, 7-tetrahydro-
thieno [3, 2-c] pyridine derivatives as platelet aggregation inhibitors, J Saudi Chem
16/2551-0000240-1 and PRONUPEQ 16/2551- 0000526-5) for fi-
Soc 18 (2014) 513–519.
nancial support. C.L., E.A.W. and G.P are recipients of CNPq fellowship. [16] S. Lee, D.W. Kang, H. Ryu, et al., t-Butyl pyridine and phenyl C-region analogues of
This study was financed in part by the Coordenação de 2-(3-fluoro-4-methylsulfonylaminophenyl) propanamides as potent TRPV1 an-
Aperfeiçoamento de Pessoal de Nível Superior - Brasil (CAPES) - tagonists, Bioorg. Med. Chem. Lett 25 (2017) 2451–2462.
[17] K. Umei, Y. Nishigaya, A. Kondo, et al., Novel pyrazolo [1, 5-a] pyridines as orally
Finance Code 001. active EP 1 receptor antagonists: synthesis, structure-activity relationship studies,
and biological evaluation, Bioorg. Med. Chem. 25 (2017) 2635–2642.
Conflicts of interest [18] J.B. Xi, Y.F. Fang, B. Frett, et al., Structure-based design and synthesis of imidazo
[1,2-a]pyridine derivatives as novel and potent Nek2 inhibitors with in vitro and in
vivo antitumor activities, Eur. J. Med. Chem. 126 (2017) 1083–1106.
The authors declare that they have no conflict of interest. [19] T.C. Mendes, J.M. Raimundo, N.M. Nascimento-Junior, et al., Sedation and anti-
nociception induced by a new pyrazolo [3, 4-b] pyrrolo [3, 4-d] pyridine derivative
(LASSBio-873) is modulated by activation of muscarinic receptors, Pharmacol.
Author contributions Biochem. Behav. 94 (2009) 70–74.
[20] T.H. Ha, H. Ryu, S.E. Kim, et al., TRPV1 antagonist with high analgesic efficacy: 2-
A.R, C.L and E.A.W conceived and designed the study. A.V, M.P, Thio pyridine C-region analogues of 2-(3-fluoro-4-methylsulfonylaminophenyl)
propanamides, Bioorg. Med. Chem. 21 (2013) 6657–6664.
G.V, C·F, J.P, T.P, R.V, J.E, M.S, R.S and G.P carried out the acquisition, [21] M.H. Helal, S.A. El-Awdan, M.A. Salem, et al., Synthesis, biological evaluation and
analysis and interpretation of data. A.R, C.L and E.A.W wrote the molecular modeling of novel series of pyridine derivatives as anticancer, anti-in-
manuscript. E.A.W and C.L supervised the study. flammatory and analgesic agents, Spectrochim. Acta A Mol. Biomol. Spectrosc. 135
(2015) 764–773.
[22] C.W. Nogueira, G. Zeni, J.B. Rocha, Organoselenium and organotellurium com-
Declaration of interests pounds: toxicology and pharmacology, Chem. Rev. 104 (2004) 6255–6285.
[23] E.A. Wilhelm, C.R. Jesse, C.F. Bortolatto, et al., Antinociceptive and antiallodynic
The authors declare the following financial interests/personal re- effects of 3-alkynyl selenophene on different models of nociception in mice,
Pharmacol. Biochem. Behav. 93 (2009) 419–425.
lationships which may be considered as potential competing interests: [24] F. Donato, N.F. Pavin, A.T. Goes, et al., Antinociceptive and anti-hyperalgesic ef-
fects of bis(4-methylbenzoyl) diselenide in mice: evidence for the mechanism of
Appendix A. Supplementary data action, Pharm. Biol. 53 (2015) 395–403.
[25] C.A. Bruning, M. Prigol, J.A. Roehrs, G. Zeni, et al., Evidence for the involvement of
l-opioid and d-opioid receptors in the antinociceptive effect caused by oral ad-
Supplementary data to this article can be found online at https:// ministration of m-trifluoromethyl-diphenyl diselenide in mice, Behav. Pharmacol.
doi.org/10.1016/j.cbi.2019.108790. 21 (2010) 621–626.
[26] P.M. Chagas, B.D.C.W. Fulco, A.P. Pesarico, et al., Effectiveness of bis (phenylimi-
dazoselenazolyl) diselenide on a mouse model of inflammatory nociception,
References Biomed. Pharmacother. 96 (2017) 56–63.
[27] T.J. Peglow, R.F. Schumacher, R. Cargnelutti, et al., Preparation of bis (2-pyridyl)
diselenide derivatives: synthesis of selenazolo [5, 4-b] pyridines and unsymmetrical
[1] R.A. Elzahaf, O.A. Tashani, B.A. Unsworth, et al., The prevalence of chronic pain
diorganyl selenides, and evaluation of antioxidant and anticholinesterasic activities,
with an analysis of countries with a Human Development Index less than 0.9: a
Tetrahedron Lett. 58 (2017) 3734–3738.
systematic review without meta-analysis, Curr. Med. Res. Opin. 28 (2012)
[28] A. Beirith, A.R. Santos, J.B. Calixto, Mechanisms underlying the nociception and
1221–1229.
paw oedema caused by injection of glutamate into the mouse paw, Brain Res. 924
[2] S.R.D.T. Siqueira, T.T. Vilela, A.A. Florindo, Prevalence of headache and orofacial
(2002) 219–228.
pain in adults and elders in a Brazilian community: an epidemiological study,
[29] A.S. Reis, M. Pinz, L.F.B. Duarte, J.A. Roehrs, et al., 4-phenylselenyl-7-chlor-
Gerodontology 32 (2015) 123–131.
oquinoline, a novel multitarget compound with anxiolytic activity: contribution of
[3] I. Brusco, C. Camponogara, F.B. Carvalho, et al., α‐Spinasterol: a COX inhibitor and
the glutamatergic system, J. Psychiatr. Res. 84 (2017) 191–199.
a transient receptor potential vanilloid 1 antagonist presents an antinociceptive
[30] F.S. Sousa, R.G. Anversa, P.T. Birmann, et al., Contribution of dopaminergic and
effect in clinically relevant models of pain in mice, Br. J. Clin. Pharmacol. 174
noradrenergic systems in the antinociceptive effect of α-(phenylalanyl) acet-
(2017) 4247–4262.
ophenone, Pharmacol. Rep. 69 (2017) 871–877.
[4] B.E. Lacy, F. Wang, S. Bhowal, et al., On-demand hyoscine butylbromide for the
[31] R.G. Anversa, F.S.S. Sousa, P.T. Birmann, et al., Antinociceptive and anti‐in-
treatment of self-reported functional cramping abdominal pain, Scand. J.
flammatory effects of 1, 2‐bis‐(4 methoxyphenylselanyl) styrene in mice: involve-
Gastroenterol. 48 (2013) 926–935.
ment of the serotonergic system, J. Pharm. Pharmacol. 70 (2018) 901–909.
[5] E.M. Pogatzki-Zahn, D. Segelcke, S.A. Schug, Postoperative pain—from mechanisms
[32] P.T. Birmann, F.S. Sousa, D.H. de Oliveira, 3-(4-Chlorophenylselanyl)-1-methyl-1H-
to treatment, Pain Rep 2 (2017) e588.
indole, a new selenium compound elicits an antinociceptive and anti-inflammatory
[6] R. Chou, D.B. Gordon, O. de Leon-Casasola, et al., Management of postoperative
effect in mice, Eur. J. Pharmacol. 827 (2018) 71–79.
pain: a clinical practice guideline from the American pain society, the American
[33] M.S. Meymandi, F. Keyhanfar, O. Yazdanpanah, et al., The role of NMDARs ligands
society of regional anesthesia and pain medicine, and the American society of an-
on antinociceptive effects of pregabalin in the tail flick test, Anesthesiol. Pain Med.
esthesiologists committee on regional anesthesia, executive committee, J. Pain 17
5 (2015) e28968.
(2016) 131–157.
[34] H.G. Woolfre, A.D. MacDonald, The evaluation of the analgesic action of pethidine
[7] M. Pinz, A.S. Reis, V. Duarte, et al., 4-Phenylselenyl-7-chloroquinoline, a new
hydrochloride, J. Pharmacol. Exp. Ther. 80 (1944) 300–307.
quinoline derivative containing selenium, has potential antinociceptive and anti-
[35] S.I. Ankier, New hot plate tests to quantify antinociceptive and narcotic antagonists
inflammatory actions, 780 (2016) 122–128.
activities, Eur. J. Pharmacol. 27 (1974) 1–4.
[8] V.D. Silva, A.S. Reis, M. Pinz, et al., Further analysis of acute antinociceptive and
[36] S. Hunskaar, O.G. Berge, K. Hole, A modified hot plate test sensitive to mild an-
anti‐inflammatory actions of 4-phenylselenyl-7-chloroquinoline in mice, Fundam.
algesics, Behav. Brain Res. 21 (1986) 101–108.
Clin. Pharmacol. 31 (2017) 513–525.
[37] R.N. Walsh, R.A. Cummins, The open-field test: a critical review, Psychol. Bull. 83
[9] E.A. Wilhelm, A.T. Ferreira, M. Pinz, et al., Antioxidant effect of quinoline deri-
(1976) 482–504.
vatives containing or not selenium: relationship with antinociceptive action qui-
[38] L.B. Bortolanza, J. Ferreira, S.C. Hess, et al., Anti-allodynic action of the tormentic
nolines are antioxidant and antinociceptive, An. Acad. Bras. Cienc. 89 (2017)
acid, a triterpene isolated from plant, against neuropathic and inflammatory per-
457–467.
sistent pain in mice, Eur. J. Pharmacol. 453 (2002) 203–208.
[10] M. Prigol, E.A. Wilhelm, C.C. Schneider, et al., Protective effect of unsymmetrical
[39] K. Suzuki, H. Ota, S. Sasagawa, et al., Assay method for myeloperoxidase in human
dichalcogenide, a novel antioxidant agent, in vitro and an in vivo model of brain
polymorphonuclear leukocytes, Anal. Biochem. 132 (1983) 345–352.

9
A.S. Reis, et al. Chemico-Biological Interactions 311 (2019) 108790

[40] R. Re, N. Pellegrini, A. Proteggente, et al., Antioxidant activity applying an im- glutamate receptors contribute to nociceptive behaviors in the rat formalin test,
proved ABTS radical cation decolorization assay, Free Radic. Biol. Med. 26 (1999) Neuroreport 8 (1997) 941–946.
1231–1237. [57] K. Omote, T. Kawamata, M. Kawamata, et al., Formalin-induced release of ex-
[41] C.W. Choi, S.C. Kim, S.S. Hwang, et al., Antioxidant activity and free radical citatory amino acids in the skin of the rat hindpaw, Brain Res. 787 (1998) 161–164.
scavenging capacity between Korean medicinal plants and flavonoids by assay- [58] M. Sasaki, C. Tohda, Y. Kuraishi, Region-specific increase in glutamate release from
guided comparison, Plant Sci. 163 (2002) 1161–1168. dorsal horn of rats with adjuvant inflammation, Neuroreport 9 (1998) 3219–3222.
[42] J.L. Giongo, V.R. De Almeida, M.R. Sagrillo, et al., Anti-inflammatory effect of [59] L.G. Fischer, D. Santos, C. Serafin, et al., Further antinociceptive properties of ex-
geranium nanoemulsion macrophages induced with soluble protein of Candida al- tracts and phenolic compounds from Pliniaglomerata (Myrtaceae) leaves, Biol.
bicans, Microb pathogenesis 110 (2017) 694–702. Pharm. Bull. 31 (2008) 235–239.
[43] M.M. Bradford, A rapid and sensitive method for the quantification of microgram [60] F.S. Oliveira, D.P. De Sousa, R.N. De Almeida, Antinociceptive effect of hydro-
quantities of protein utilizing the principle of protein-dye binding, Anal. Biochem. xydihydrocarvone, Biol. Pharm. Bull. 31 (2008) 588–591.
72 (1976) 248–254. [61] M. Bujalska, R. Winecka, S.W. Gumułka, Effect of selol on the opioids activity in
[44] S. Reitman, S. Frankel, A colorimetric method for the determination of serum streptozotocin induced hyperalgesia, Acta Pol Pharm Drug Res 65 (2008) 691–696.
glutamic oxalacetic and glutamic pyruvic transaminases, Am. J. Clin. Pathol. 28 [62] S.G. Rosa, A.P. Pesarico, F. Martini, et al., m-Trifluoromethyl-diphenyl diselenide
(1957) 56–63. regulates prefrontal cortical MOR and KOR protein levels and abolishes the phe-
[45] E.M. Mackay, L.L. Mackay, The concentration of urea in the blood of normal in- notype induced by repeated forced swim stress in mice, Mol. Neurobiol. 55 (2018)
dividuals, J. Clin. Investig. 4 (1927) 295–306. 8991–9000.
[46] H. Ohkawa, N. Ohishi, K. Yagi, Assay for lipid peroxides in animal tissues by [63] B. Joe, R.L. Wilder, Animal models of rheumatoid arthritis, Mol. Med. Today 5
thiobarbituric acid reaction, Anal. Biochem. 95 (1976) 351–358. (1999) 367–369.
[47] J. Sedlak, R.H. Lindsay, Estimation of total, protein-bound, and non protein sul- [64] H.K. Kim, S.K. Park, J.L. Zhou, et al., Reactive oxygen species (ROS) play an im-
phydryl groups in tissue with Ellman's reagent, Anal. Biochem. 25 (1968) 192–205. portant role in a rat model of neuropathic pain, Pain 111 (2004) 116–124.
[48] S. Sassa, Delta-aminolevulinic acid dehydratase assay, Enzyme 28 (1982) 133–145. [65] D.K. Choi, S. Pennathur, C. Perier, et al., Ablation of the inflammatory enzyme
[49] M.J. Curtis, R.A. Bond, D. Spina, et al., Experimental design and analysis and their myeloperoxidase mitigates features of Parkinson's disease in mice, J. Neurosci. 25
reporting: new guidance for publication in BJP, Br. J. Pharmacol. 172 (2015) (2005) 6594–6600.
3461–3471. [66] T. Rosenbaum, S.A. Simon, W.B. Liedtke, S. Heller (Eds.), TRP Ion Channel Function
[50] L.T. Graham Jr., R.P. Shank, R. Werman, et al., Distribution of some synaptic in Sensory Transduction and Cellular Signaling Cascades, 2007.
transmitter suspects in cat spinal cord: glutamic acid, aspartic acid, gamma-ami- [67] A.I. Basbaum, D.M. Bautista, G. Scherrer, et al., Cellular and molecular mechanisms
nobutyric acid, glycine and glutamine, J. Neurochem. 14 (1967) 465–472. of pain, Cell 139 (2009) 267–284.
[51] J.L. Johnson, The excitant amino acids glutamic and aspartic acid as transmitter [68] F.R. Nieto, E.J. Cobos, M.A. Tejada, et al., Tetrodotoxin (TTX) as a therapeutic
candidates in the vertebrate central nervous system, Prog. Neurobiol. 10 (1978) agent for pain, Mar. Drugs 10 (2012) 281–305.
155–202. [69] B. Amoah-Apraku, L.J. Chandler, J.K. Harrison, J.K., et al., NF-kappa B and tran-
[52] S.M. Carlton, G.L. Hargett, R.E. Coggeshall, Localization and activation of gluta- scriptional control of renal epithelial-inducible nitric oxide synthase, Kidney Int. 48
mate receptors in unmyelinated axons of rat glabrous skin, Neurosci. Lett. 197 (1995) 674–682.
(1995) 25–28. [70] T. Salim, C.L. Sershen, E.E. May, Investigating the role of TNF-alpha and IFNgamma
[53] D.L. Jackson, C.B. Graff, J.D. Richardson, et al., Glutamate participates in the activation on the dynamics of iNOS gene expression in LPS stimulated macro-
peripheral modulation of thermal hyperalgesia in rats, Eur. J. Pharmacol. 284 phages, PLoS One 11 (2016) e0153289.
(1995) 321–325. [71] M. Costigan, C.J. Woofl, Pain: molecular mechanisms, J. Pain 1 (2000) 35–44.
[54] S. Zhou, L. Bonasera, S.M. Carlton, Peripheral administration of NMDA, AMPA or [72] M. Regulski, K. Regulska, W. Prukała, et al., COX-2 inhibitors: a novel strategy in
KA results in pain behaviors in rats, Neuroreport 7 (1996) 895–900. the management of breast cancer, Drug Discov. Today 21 (2016) 598–615.
[55] N.B. Lawand, W.D. Willis, K.N. Westlund, Excitatory amino acid receptor involve- [73] M.H.M. Sari, V.A. Zborowski, L.M. Ferreira, et al., Enhanced pharmacological ac-
ment in peripheral nociceptive transmission in rats, Eur. J. Pharmacol. 324 (1997) tions of p, p’-methoxyl-diphenyl diselenide-loaded polymeric nanocapsules in a
169–177. mouse model of neuropathic pain: behavioral and molecular insights, J. Trace Elem.
[56] E.M. Davidson, R.E. Coggeshall, S.M. Carlton, Peripheral NMDA and non-NMDA Med. Biol. 46 (2018) 17–25.

10

You might also like