ArticlePDF Available

Abstract and Figures

This study investigates the efficacy of diphenyl diselenide [(PhSe)2] in attenuating methylmercury- (MeHg-)induced toxicity in rats. Adult rats were treated with MeHg [5 mg/kg/day, intragastrically (i.g.)] and/ or (PhSe)2 [1 mg/kg/day, intraperitoneally (i.p.)] for 21 days. Body weight gain and motor deficits were evaluated prior to treatment, on treatment days 11 and 21. In addition, hepatic and cerebral mitochondrial function (reactive oxygen species (ROS) formation, total and nonprotein thiol levels, membrane potential (ΔΨm), metabolic function, and swelling), hepatic, cerebral, and muscular mercury levels, and hepatic, cerebral, and renal thioredoxin reductase (TrxR) activity were evaluated. MeHg caused hepatic and cerebral mitochondrial dysfunction and inhibited TrxR activity in liver (38,9%), brain (64,3%), and kidney (73,8%). Cotreatment with (PhSe)2 protected hepatic and cerebral mitochondrial thiols from depletion by MeHg but failed to completely reverse MeHg's effect on hepatic and cerebral mitochondrial dysfunction or hepatic, cerebral, and renal inhibition of TrxR activity. Additionally, the cotreatment with (PhSe)2 increased Hg accumulation in the liver (50,5%) and brain (49,4%) and increased the MeHg-induced motor deficits and body-weight loss. In conclusion, these results indicate that (PhSe)2 can increase Hg body burden as well as the neurotoxic effects induced by MeHg exposure in rats.
This content is subject to copyright. Terms and conditions apply.
Hindawi Publishing Corporation
BioMed Research International
Volume , Article ID ,  pages
http://dx.doi.org/.//
Research Article
Effects of Diphenyl Diselenide on Methylmercury
Toxicity in Rats
Cristiane L. Dalla Corte,1Caroline Wagner,2Jéssie H. Sudati,2
Bruna Comparsi,3Gerlania O. Leite,4Alcindo Busanello,1Félix A. A. Soares,1
Michael Aschner,5,6 and João B. T. Rocha1
1Biochemistry and Molecular Biology Department, Graduation Program in Biological Sciences: Toxicological Biochemistry,
Natural and Exact Sciences Center, Federal University of Santa Maria, 97105-900 Santa Maria, RS, Brazil
2Federal University of Pampa—Cac¸apava do Sul Campus, Avenida Pedro Anunciac¸˜
ao, Vila Batista,
96570-000 Cac¸apava do Sul, RS, Brazil
3Higher Education Cenecista Institute of Santo ˆ
Angelo—IESA, Rua Dr. Jo˜
ao Augusto Rodrigues 471,
98801-015 Santo ˆ
Angelo, RS, Brazil
4Regional University of Cariri, Pharmacology and Molecular Chemistry Laboratory, Rua Cel. Antˆ
onio Lu´
ıs 1161,
63100-000 Crato, CE, Brazil
5Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
6Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
Correspondence should be addressed to Cristiane L. Dalla Corte; crisbioq@yahoo.com.br
and Jo˜
ao B. T. Rocha; jbtrocha@yahoo.com.br
Received  September ; Revised  November ; Accepted  November 
Academic Editor: Fernando Barbosa Jr.
Copyright ©  Cristiane L. Dalla Corte et al. is is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly
cited.
is study investigates the ecacy of diphenyl diselenide [(PhSe)2] in attenuating methylmercury- (MeHg-)induced toxicity in
rats. Adult rats were treated with MeHg [ mg/kg/day, intragastrically (i.g.)] and/ or (PhSe)2[ mg/kg/day, intraperitoneally (i.p.)]
for  days. Body weight gain and motor decits were evaluated prior to treatment, on treatment days  and . In addition,
hepatic and cerebral mitochondrial function (reactive oxygen species (ROS) formation, total and nonprotein thiol levels, membrane
potential (ΔΨm), metabolic function, and swelling), hepatic, cerebral, and muscular mercury levels, and hepatic, cerebral, and
renal thioredoxin reductase (TrxR) activity were evaluated. MeHg caused hepatic and cerebral mitochondrial dysfunction and
inhibited TrxR activity in liver (,%), brain (,%), and kidney (,%). Cotreatment with (PhSe)2protected hepatic and cerebral
mitochondrial thiols from depletion by MeHg but failed to completely reverse MeHg’s eect on hepatic and cerebral mitochondrial
dysfunction or hepatic, cerebral, and renal inhibition of TrxR activity. Additionally, the cotreatment with (PhSe)2increased Hg
accumulation in the liver (,%) and brain (,%) and increased the MeHg-induced motor decits and body-weight loss. In
conclusion, these results indicate that (PhSe)2can increase Hg body burden as well as the neurotoxic eects induced by MeHg
exposure in rats.
1. Introduction
MeHg is one of the most poisonous environmental contam-
inants, causing toxic eects in humans and experimental
animals [,]. Environmental MeHg is largely derived from
inorganic mercury biomethylation carried out primarily by
aquatic microorganisms []withsubsequentaccumulationin
the aquatic food chain and human consumption []. MeHg
causes acute and chronic damage to multiple organs, most
profoundly to the central nervous system (CNS), in partic-
ular when exposures occur during the neurodevelopmental
period [,,].
e events that mediate MeHg toxicity are largely depen-
dent upon its electrophilic properties, which allow for its
BioMed Research International
interaction with so nucleophilic groups (mainly thiols
and selenols) from either low- or high-molecular-weight
biomolecules []. e interaction of MeHg with so nucle-
ophilic groups from biomolecules is responsible, at least in
part, for decreased antioxidant capacity and increased ROS
generation [,]. Notably, MeHg can disrupt the activity of
thiol- and selenol-containing proteins, such as glutathione
peroxidase (GPx), thioredoxin (Trx), and TrxR [,].
ese proteins are important components of the cellular
antioxidant system, and their inhibition contributes to the
disruption of the normal redox balance of cells [].
In addition, MeHg can disrupt mitochondrial function
by targeting specic thiol-containing proteins, including
respiratory chain complexes [,]. e inhibition of these
complexes or enzymes can contribute to mitochondrial depo-
larization and swelling upon MeHg exposure. Mitochondrial
targeting by MeHg has also been associated with increased
mitochondrial ROS generation, which can further exacerbate
thetoxicityofMeHgbyattackingadditionalnucleophiliccen-
ters in mitochondria and in other subcellular compartments
[,,], leading to a vicious cycle of cell demise.
Several studies demonstrated that organic and inorganic
selenium (Se) compounds inuence the deposition and
toxicity of MeHg [,,]. Se is an essential trace element
for a wide range of living organisms, including humans [].
Se is necessary for the expression of approximately  Se-
dependent proteins, including GPx, TrxR, and several other
enzymes and proteins, which can modulate the cellular redox
and antioxidant status [].
In addition to inorganic and naturally occurring organo-
selenium compounds, synthetic organoselenium compounds
can also exhibit protective eects against MeHg. For example,
ebselen and (PhSe)2have been shown to exert benecial
eects against in vitro and in vivo MeHg-induced toxicity [
]. (PhSe)2(which is the simplest of the diaryl diselenides
[]) protected against an array of toxic eects of MeHg and
lowered the Hg burden in the brain, liver, and kidneys of adult
mice []. e molecular mechanism(s) which underlie(s) the
protective eects of (PhSe)2in mice likely reect the direct
interaction of MeHg with “selenol intermediate” of (PhSe)2
aer its reaction with thiols, or indirectly, by modulating
oxidative stress levels [,]. In short, the protective eects
of (PhSe)2against MeHg-induced toxicity are likely related
to its antioxidant properties and its ability to form stable
complexes with MeHg, which can increase Hg excretion and
decrease the MeHg body burden.
Of particular pharmacological signicance, the toxicity
and pharmacokinetics of MeHg [] are dierent in mice
and rat which can be explained by the higher binding anity
of rat hemoglobin, which contains more cysteinyl residues
than mice protein, for MeHg when compared to the mice
hemoglobin []. (PhSe)2toxicity and pharmacokinetics
dierences between mice and rat also exist and could be
explained by a faster metabolization of (PhSe)2in mice [
].
erefore, the aim of the present study was to investigate
the potential protective eects of (PhSe)2against MeHg-
induced toxicity and mitochondrial dysfunction in rats. To
accomplish this goal, the eects of (PhSe)2on Hg deposition
in liver and brain and on behavioral and biochemical param-
eters were studied in rats.
2. Materials and Methods
2.1. Chemicals. Chemicals, including ethylene glycol-bis(𝛽-
aminoethylether)-N,N,N󸀠,N 󸀠-tetraacetic acid (EGTA), -
(-hydroxyethyl)--piperazineethanesulfonic acid (HEPES),
, dinitrophenol (, DNP), -(,-dimethylthiazol--yl)-
,-diphenyltetrazolium bromide (MTT), glutamic acid,
safranin O, 󸀠,󸀠-dichlorouorescin diacetate (H2-DCFDA),
and methylmercury chloride were obtained from Sigma
Aldrich (St. Louis, MO, USA). (PhSe)2was synthesized
according to the method by Paulmier []. All other chemi-
cals were of analytical reagent grade and purchased from local
commercial suppliers.
2.2. Animals. Male Wistar rats, weighing –g and with
age from  to . months, from our own breeding colony
were kept in cages (four animals in each). Rats were placed
in a room with controlled temperature (22 ± 3C) on a  h
light/dark cycle (lights on at : a.m.) and had continuous
access to food and water. All experiments were conducted
in accordance with the Committee on Care and Use of
Experimental Animal Resources of the Federal University of
Santa Maria, Brazil.
2.3. Treatment. Sixteen rats were equally divided into four
experimental groups as follows: () control ( mL/Kg of
water i.g. and  mL/Kg of soybean oil i.p.); () (PhSe)2
( mL/Kg of water i.g. and  mg/Kg of (PhSe)2i.p.); () MeHg
( mg/Kg of MeHg i.g. and  mL/Kg of soybean oil i.p.); and
() (PhSe)2+ MeHg (mg/Kg of MeHg i.g. and mg/Kg
of (PhSe)2i.p.). Based on previous studies, exposures were
performed daily over a -day period [,,]. Twenty-four
hours aer the last exposure, the animals were sacriced and
the livers, brains, kidneys, and skeletal muscle were quickly
removed, placed on ice and homogenized.
2.4. Determination of Hg Levels. Tissue levels of total Hg
were measured in liver, brain, and skeletal muscle collected
at the time of euthanasia []. Approximately . g (wet
weight) of the tissues was weighed and digested with  mL
of HNO3acid (%). Digested samples were diluted to  mL
with ultrapure water before analysis using a Multitype ICP
Emission Spectrometer (ICPE-, Shimadzu). Calibration
standard curve was prepared freshly using mercury stock
standard solution.
2.5. Motor Coordination Tests
2.5.1. Open Field Test. General locomotor activity was eval-
uated as previously described []. e number of line
crossings (number of segments crossed with the four paws)
and rearings was measured over  min and taken as an
indicator of locomotor activity. e test was carried out at
 time points:  hours prior to treatment (basal), and on
treatment days  and .
BioMed Research International
2.5.2. Rotarod Test. Motor coordination was tested on the
rotarod apparatus as described previously [,]. e
latency to fall and the number of falls from the apparatus were
recorded for  s. e tests were conducted  times:  hours
prior to treatment (basal), and on treatment days  and .
2.6. TrxR
2.6.1. TrxR Purication. TrxR was partially puried by a
modication of the method by Holmgren and Bjornstedt
[]. Tissues were homogenized in buered saline ( mM
NaCl, . mM KCl, . mM Na2HPO4,and.mMKH
2PO4,
pH .). Livers, brains, and kidneys (. g) were homogenized
in,,andvolumesofbueredsaline,respectively.
Homogenates were centrifuged at , g for  min. e
protein concentration in the supernatant was measured and
adjusted to  mg/mL. e supernatant was dialyzed against
buered saline for  h to remove endogenous glutathione
(GSH) and Trx. e dialysate was heated at Cformin,
cooled, and centrifuged at , g for  min to remove
denatured protein.
2.6.2. TrxR Activity. TrxR activity was measured by the
method of Holmgren and Bjornstedt []. e reaction
mixture consisted of the following: . mM NADPH,
 mM EDTA,  mM potassium phosphate buer (pH .),
 mg/mL ,󸀠dithiobis--nitrobenzoic acid (DTNB), and
. mg/mL of BSA. e partially puried TrxR was added
(to nal concentration of – 𝜇gofprotein)tothecuvette
containing the reaction mixture, and the absorbance was
followed at  nm for a maximum of  min.
2.7. Isolation of Rat Brain and Liver Mitochondria. Rat brain
and liver mitochondria were isolated as previously described
by Brustovetsky and Dubinsky [], with some modications.
Brain and liver were rapidly weighed and homogenized in  : 
(w/v) ice-cold “isolation buer I” containing  mM man-
nitol,  mM sucrose,  mM K+-EGTA, .% bovine serum
albumin (BSA), and  mM K+-HEPES,pH..etissuewas
then manually homogenized with a potter glass. e result-
ing suspension was centrifuged for  min at , g. Aer
centrifugation the supernatant was recentrifuged for  min
at , g. e pellet was resuspended in “isolation buer
II” containing  mM mannitol,  mM sucrose,  mM K+-
EGTA, and  mM K+-HEPES pH . and recentrifuged
at , g for  min. e supernatant was discarded and
the nal pellet gently washed and resuspended in “isolation
buer II” without EGTA.
2.8. Mitochondrial Nonprotein and Total iol Content. Mito-
chondrial nonprotein and total thiol content were measured
according to the method of Ellman []. To determine total
thiol groups, mitochondria (.mg protein) were added to
the reaction medium containing  mM Tris-HCl pH ., %
SDS, and  mM DTNB. Nonprotein thiol content was mea-
sured by adding  𝜇L%TCAto𝜇Lofthemitochondria
(. mg protein). Aer centrifugation (, ×gat
Cfor
 min), the protein pellet was discarded and an aliquot of
the clear supernatant, neutralized with .M NaOH, was
added to the medium containing  mM Tris-HCl pH .
and  mM DTNB. e samples absorbance was measured
spectrophotometrically at a wavelength of  nm.
2.9. Measurements of Mitochondrial ΔΨm. Mitochondrial
ΔΨm was estimated by uorescence changes in safranin
O ( mM) recorded by RF- Shimadzu spectrouorom-
eter (Kyoto, Japan) operating at excitation and emission
wavelengths of  and  nm, with slit widths of . nm
[]. Data on ΔΨm in the gures is presented in Arbitrary
Fluorescence Units (AFU).
2.10. Estimation of ROS Production. e mitochondrial
generation of ROS was determined spectrouorimetrically,
using the membrane permeable uorescent dye H2-DCFDA
recorded by RF- Shimadzu spectrouorometer (Kyoto,
Japan) operating at excitation and emission wavelengths of
 and  nm, with slit widths of  nm []. Data of
ROS production in the gures is presented as Arbitrary
Fluorescence Units (AFU).
2.11. Assessment of Mitochondrial Metabolic Function. e
mitochondrial metabolic function was assessed by the con-
version of MTT to a dark violet formazan product by mito-
chondrial dehydrogenases []. e rate of MTT reduction
was measured spectrophotometrically at a wavelength of
 nm. Results were expressed as the percentage of MTT
reductionrelativetocontrolvalues.
2.12. Assessment of Mitochondrial Swelling. Measurement of
mitochondrial swelling was performed in a RF- Shi-
madzu spectrouorometer at nm (slit . nm for exci-
tation and emission) []. Data for mitochondrial swelling
are expressed as Arbitrary Absorbance Units (AAU). e
dierence (ΔA) between the initial absorbance reading and
thenalabsorbancereadingwasusedforstatisticalanalysis.
2.13. Protein Measurement. Protein was assayed by the
method of Bradford []withbovineserumalbuminas
standard.
2.14. Statistical Analysis. Normality assumption was tested
with Kolmogorov-Smirnov test and the distribution of the
majority of results is not normal. Data were analyzed sta-
tistically by Mann-Whitney or Kruskal-Wallis, followed by
Dunn’s post-hoc tests when appropriate. e results were
considered statistically signicant at 𝑃 < 0.05.Allstatistical
analyses were conducted using GraphPad Prism  (Version
., GraphPad Soware, Inc., USA).
3. Results
3.1. Eects of (PhSe)2and MeHg on Body Weight. Treat me n t
with MeHg led to body-weight loss from the second week
until the end of the treatment compared to controls (𝑃<
0.05,Figure ). Rats cotreated with (PhSe)2and MeHg also
BioMed Research International
0 7 14 21
160
210
260
310
360
Control MeHg
Days
Weight (g)
(PhSe)2(PhSe)2+ MeHg
F : Eect of MeHg and/or (PhSe)2on the body weight gain in
adult rats. Data are expressed as mean ±S.D., 𝑛=4.()represents
𝑃 < 0.05 as compared to controls by Mann-Whitney test.
showed a decrease in the body weight when compared to
the control group (𝑃 < 0.05,Figure ). Rats treated with
(PhSe)2lost weight aer the rst week of treatment (𝑃<
0.05) but showed a trend towards a recovery and were
statistically indistinguishable from the controls at the end of
the treatment (Figure ).
3.2. Eects of (PhSe)2andMeHgonHgDeposition. Tre at m e n t
with MeHg increased the levels of Hg in liver, brain, and
skeletal muscle compared with controls (𝑃 < 0.05,Figure ).
e cotreatment with (PhSe)2caused a greater increase
in brain Hg deposition when compared to MeHg alone
treatment, both in brain (Figure (b))andliver(Figure (a)),
and showed a trend towards increased deposition in skeletal
muscle (Figure (c)).
3.3. Eects of (PhSe)2and MeHg on Motor Coordination
and Spontaneous Locomotor Activity. e eects of MeHg
and/or (PhSe)2on locomotion and motor coordination were
assessed by the open-eld and rotarod tests, respectively.
Aer  days, rats treated with MeHg showed increased
number of falls on the rotarod and decreased latency to the
rst fall when compared to controls (𝑃 < 0.05,Figures(a)
and (b)). Rats treated with (PhSe)2did not show statistically
signicant dierences on the rotarod test when compared
to controls; however, rats cotreated with (PhSe)2and MeHg
showed increased loss of motor coordination as evidenced by
increased number of falls and reduced latency to the rst fall
(𝑃 < 0.05,Figures(a) and (b)). e rotarod test could not
be performed at the end of the treatment in rats receiving
MeHgsincetheywereunabletoremainintheapparatusdue
to severe motor impairment caused by MeHg.
Rats treated with MeHg showed a decrease in the number
of crossings and rearings in the open-eld at the end of the
treatment compared to the control rats (𝑃 < 0.05,Figures
(c) and (d)). Rats cotreated with MeHg and (PhSe)2also
showed a signicant decrease in the number of crossings
aer  days of treatment and a decrease in the number of
rearings at the end of the treatment (𝑃 < 0.05,Figure (d)).
Tre at m e n t w it h ( PhS e ) 2didnotaecttherats’performancein
the open-eld. e decrease in the number of crossings and
rearings observed in all groups on treatment days  and 
was expected given that the animals habituate to the open-
eld arena [].
3.4. Eects of (PhSe)2and MeHg on
Mitochondrial Dysfunction
3.4.1. Mitochondrial Metabolic Function. e hepatic mito-
chondrialmetabolicintegrity(MTTreduction)wasnotaec-
ted by MeHg and/or (PhSe)2(Figure (a)). Treatment with
MeHg or cotreatment with MeHg and (PhSe)2decreased the
capacity of brain mitochondrial dehydrogenases to reduce
MTT compared to controls (𝑃 < 0.05,Figure (b)). Treat-
ment with (PhSe)2alonedidnotaectthecerebralmito-
chondrial metabolic function.
3.4.2. Mitochondrial Total and Nonprotein iols. MeHg
treatment decreased the total mitochondrial thiol levels in
brainandliverwhencomparedtocontrols(𝑃 < 0.05,
Figure ). Treatment with (PhSe)2alone did not alter the
mitochondrial total thiol levels in liver and brain (Figure ).
e cotreatment with (PhSe)2blunted the MeHg-induced
mitochondrial total thiol level depletion in rats’ liver and
brain (𝑃 < 0.05,Figure ). Rats treated with MeHg showed
decreased mitochondrial nonprotein thiol levels in the liver
compared to controls, and coadministration of (PhSe)2
blunted the MeHg-induced decrease in hepatic nonprotein
thiol content (𝑃 < 0.05,Figure (a)). Brain mitochondrial
nonprotein thiol levels were not aected by any of the
treatments (Figure (b)).
3.4.3. Mitochondrial Swelling. Treatment with MeHg sig-
nicantly increased hepatic mitochondrial swelling when
compared to controls (𝑃 < 0.05,Figure (a)). Cotreatment
with (PhSe)2partially prevented the MeHg-induced mito-
chondrial swelling in liver (Figure (a)). Similarly, treatment
with MeHg showed a trend towards increased mitochondrial
swelling in brain (Figure (b)). e cotreatment with MeHg
and (PhSe)2signicantly increased cerebral mitochondrial
swelling when compared to controls (𝑃 < 0.05,Figure (b)).
Tre at m e n t w it h ( PhS e ) 2alone did not alter the mitochondrial
swelling in brain or liver compared to the controls (Figures
(a) and (b)).
3.4.4. Mitochondrial ROS Production. Mitochondrial ROS
production (DCFH oxidation) was signicantly increased in
livers of rats treated with MeHg or cotreated with MeHg and
(PhSe)2(𝑃 < 0.05,Figure (a)). Rats treated with (PhSe)2
showed hepatic mitochondrial ROS levels indistinguishable
from controls. ROS production in cerebral mitochondria was
not aected by any of the treatments (Figure (b)).
3.4.5. Mitochondrial ΔΨm. Polarization (ΔΨm) of mitochon-
driafromliverofratscotreatedwithMeHgand(PhSe)
2
BioMed Research International
0
50
100
150
200
250
(𝜇g Hg/g tissue)
Control (PhSe)2MeHg (PhSe)2+ MeH
g
#
(a)
0
10
20
30
40
50
Control (PhSe)2MeHg (PhSe)2+ MeH
g
#
(𝜇g Hg/g tissue)
(b)
0
20
40
60
80
Control (PhSe)2MeHg (PhSe)2+ MeHg
(𝜇g Hg/g tissue)
(c)
F : Hg content in liver (a), brain (b), and muscle (c) of rats exposed to MeHg and/or (PhSe)2. Data are expressed as mean ±S.D., 𝑛=4.
()represents𝑃 < 0.05 as compared to controls by Mann-Whitney test. () represents 𝑃 < 0.05 as compared to MeHg by Mann-Whitney
test.
showed only a trend towards decreased (Figures (a) and
(c)). Treatment with (PhSe)2and MeHg alone did not cause
mitochondrial depolarization in liver of rats (Figures (a) and
(c)). Treatment with (PhSe)2and/or MeHg had no eect on
mitochondrial ΔΨm in brain of rats (Figures (b) and (d)).
3.5. Eects of (PhSe)2andMeHgonTrxRActivity.MeHg
is known to inhibit TrxR activity both in vitro and in vivo
[,,]. (PhSe)2treatment signicantly increased renal TrxR
activities when compared to controls (𝑃 < 0.05,Figures(a)
and (b)). Hepatic and cerebral TrxR activity showed a trend
towards increased in rats treated with (PhSe)2(Figure (c)).
MeHg treatment also led to signicant inhibition of TrxR
in liver, kidney, and brain compared to controls (𝑃 < 0.05,
Figure ). Cotreatment with (PhSe)2failed to signicantly
attenuate the MeHg-induced inhibition of TrxR activity in the
liver, kidney, or brain (Figure ).
4. Discussion
e present study investigated the ecacy of (PhSe)2,an
organoselenium compound, in attenuating MeHg-induced
toxicity in rats. Our results established that MeHg decreased
body weight (Figure ) and induced motor decits (Figure )
as well hepatic and cerebral mitochondrial dysfunction (Fig-
ures (b),,(a),and(a))andinhibitedTrxRactivityin
liver, brain, and kidney (Figure ) in the rat. e cotreatment
with (PhSe)2and MeHg increased Hg accumulation in the
liver and brain (Figure ). Furthermore, the cotreatment with
(PhSe)2protected hepatic and cerebral mitochondrial thiols
from depletion by MeHg (Figure ) but did not prevent
hepatic and cerebral mitochondrial dysfunction (Figures
(b),(b),and(a)) nor did it reverse the MeHg-induced
motor decits (Figure ), body-weight loss (Figure ), and the
MeHg-induced inhibition of TrxR activity in liver, brain, and
kidney (Figure ).
Cotreatment with (PhSe)2and MeHg increased Hg depo-
sition in the brain and liver of exposed rats (Figure ).
ese results dier from those of de Freitas et al. []where
(PhSe)2led to a signicant reduction in Hg concentrations
in brain, liver, and kidney of MeHg-exposed mice. e
discrepancies between the  studies may be attributed to
metabolic dierences between the species and the route of
administration. e toxicity and pharmacokinetics of MeHg
[] are dierent in mice and rat which can be explained
BioMed Research International
0
2
4
6
8
10
Basal
Day 11
Number of falls
Control (PhSe)2MeHg (PhSe)2+ MeH
g
(a)
Basal
Day 11
0
25
50
75
100
125
150
Latency to rst fall (s)
Control (PhSe)2MeHg (PhSe)2+ MeHg
(b)
0
20
40
60
80
100
Basal
Day 11
Day 21
Control (PhSe)2MeHg (PhSe)2+ MeH
g
Ambulation (number of crossings/5 min)
(c)
Basal
Day 11
Day 21
0
10
20
30
40
50
Control (PhSe)2MeHg (PhSe)2+ MeHg
Rearing (number of times/5 min)
#
(d)
F : Rotarod and open eld tests in rats exposed to MeHg and/or (PhSe)2. e number of falls (a) and latency for the rst fall (b)
ambulation (crossing) (a) and rearing (b) were recorded. Data are expressed as mean ±S.D., 𝑛=4.()represents𝑃 < 0.05 as compared to
controls by Kruskal-Wallis test followed by multiple comparison test. () represents 𝑃 < 0.05 as compared to (PhSe)2by Kruskal-Wallis test
followed by multiple comparison test.
by the higher binding anity of rat hemoglobin, containing
more cysteinyl residues, for MeHg when compared to the
mice hemoglobin []. (PhSe)2toxicity and pharmacokinet-
ics dierences between mice and rat also exist and could
be explained by a faster metabolization of (PhSe)2in mice
[]. Notably, herein rats were administered (PhSe)2i.p.,
whilst in the study by de Freitas et al. [](PhSe)
2was
subcutaneously (s.c.) administered to the mice. Another
dierence between the two works is in relation to the dose
of MeHg: in our study we used a dose . times higher than
in the study of de Freitas et al. (mg/Kg). However, the
duration of the treatment was shorter in our study,  versus
 days. On the other hand, the dose of (PhSe)2was similar
betweenthetwostudies.ehigherdoseofMeHgusedinour
study may have contributed to the discrepancies since it could
generate a more severe toxicity which could not be prevented
by (PhSe)2. However, we realize that the dierences in the
pharmacokinetics between rats and mice for the (PhSe)2is
the major factor involved in the discrepancies found here
[].
In the study by de Freitas et al. []theproposed
mechanism for the reduction Hg’s organ burden by (PhSe)2
was the formation of a selenol/selenolate (PhSeH/PhSe)
intermediate, which could interact with MeHg, generating
the readily excretable PhSeHgMe complex. One possible
explanation for the increase in hepatic and cerebral Hg depo-
sition (Figures (a) and (b), resp.) by the cotreatment with
(PhSe)2observed herein may be the conversion of (PhSe)2
to inorganic selenium, which is subsequently metabolized
to selenhidric acid (HSe). HSecouldbindtoMeHgto
form a less soluble complex [], which can be degraded to
HgSe [,]. In addition, Palmer and Parkin []showed
BioMed Research International
Control (PhSe)2MeHg (PhSe)2+ MeHg
0
50
100
150
MTT reduction (% of control)
(a)
Control (PhSe)2MeHg (PhSe)2+ MeHg
0
50
100
150
MTT reduction (% of control)
(b)
F : MTT reduction in liver (a) and brain (b) mitochondria of rats exposed to MeHg and/or (PhSe)2. Data are expressed as mean ±
S.D., 𝑛=4.()represents𝑃 < 0.05 as compared to controls by Mann-Whitney test.
Control (PhSe)2MeHg (PhSe)2+ MeHg
0
2
4
6
8
10
12
Total thiols (nmol/mg protein)
(a)
Control (PhSe)2MeHg (PhSe)2+ MeHg
0
2
4
6
8
10
12
Total thiols (nmol/mg protein)
(b)
Control (PhSe)2MeHg (PhSe)2+ MeHg
0.0
0.2
0.4
0.6
0.8
Nonprotein thiols (nmol/mg protein)
(c)
Control (PhSe)2MeHg (PhSe)2+ MeH
g
0.0
0.2
0.4
0.6
Nonprotein thiols (nmol/mg protein)
(d)
F : Total and nonprotein thiol content in liver (a), (c) and brain (b), (d) mitochondria of rats exposed to MeHg and/or (PhSe)2.Data
are expressed as mean ±S.D., 𝑛=4.()represents𝑃 < 0.05 as compared to controls by Mann-Whitney test.
that organoselenium can also form a complex with mercury.
us the increase in hepatic and cerebral Hg deposition
by the cotreatment with (PhSe)2possibly involves Hg:Se
interactions and the formation of a less excretable compound
that accumulates in these organs []. ese results are in
agreement with other studies that showed elevated deposition
of Hg in key brain regions upon oral Se administration [,
].Ithasbeenspeculatedthattheformationofinsoluble
HgSe salt could reduce the toxicity of MeHg. However, exper-
imental evidence supporting this assumption has yet to be
BioMed Research International
Control (PhSe)2MeHg (PhSe)2+ MeHg
0
10
20
30
40
50
Swelling (ΔA)
(a)
Control (PhSe)2MeHg (PhSe)2+ MeH
0
10
20
30
40
Swelling (ΔA)
(b)
F : Mitochondrial swelling in liver (a) and brain (b) of rats exposed to MeHg and/or (PhSe)2. Data are expressed as mean ±S.D., 𝑛=4.
()represents𝑃 < 0.05 as compared to controls by Mann-Whitney test.
Control (PhSe)2MeHg (PhSe)2+ MeHg
∗∗
0
50
100
150
H2-DCFH oxidation (AFU)
(a)
Control (PhSe)2MeHg (PhSe)2+ MeH
g
0
50
100
150
200
250
H2-DCFH oxidation (AFU)
(b)
F : ROS production (H2-DCFH oxidation) in liver (a) and brain (b) mitochondria of rats exposed to MeHg and/or (PhSe)2.Dataare
expressed as mean ±S.D., 𝑛=4.()represents𝑃 < 0.05 as compared to controls by Mann-Whitney test.
generated. Although the cotreatment with (PhSe)2increased
Hg levels in brain and liver, these were accompanied by
a partial protection against MeHg-induced mitochondrial
dysfunction. We suggest that the formation of an insoluble
and inert complex between Hg and Se could decrease the
availability of MeHg that could react with important cellular
components decreasing its toxicity.
Decreased weight gain and weight loss are prominent
and readily observed features of severe MeHg toxicity. In
this study, rats treated with MeHg showed body-weight
loss (Figure ). Notably, the most severe eect on weight
loss occurred in rats cotreated with (PhSe)2and MeHg
(Figure ). In addition, rats treated with MeHg showed
decreased locomotor activity (Figure ). Cotreatment with
(PhSe)2andMeHgincreasedtheseverityofmotordysfunc-
tion (rotarod test) (Figure ), likely as a result of increased
Hg deposition in the brain (Figure (b)). Motor decits
are the most apparent neurological eects following MeHg
exposure []. In vivo studies in rodents point to impairment
in intracellular calcium homeostasis, alteration in glutamate
homeostasis, and oxidative stress as critical mediators of
MeHg-induced neurotoxicity []. e overactivation of
N-methyl-D-aspartate- (NMDA-) type glutamate receptors
increases Ca2+ inux into neurons, thereby leading to cell
death []. Alternatively, Ca2+ taken up by mitochondria may
stimulate the generation of ROS [].
Several studies corroborate MeHg’s ability to induce
mitochondrial dysfunction and ROS generation [,,].
e high anity binding of MeHg to thiol groups inactivates
enzymes, including respiratory chain complexes [,,],
decreasing mitochondrial dehydrogenases activity. Inhibi-
tion of these complexes may contribute to mitochondrial
swelling and ROS production aer MeHg exposure (Figures
and ). However, in brain, the MeHg-induced decrease in
mitochondrial dehydrogenases activity (MTT reduction) was
not accompanied by an increase in ROS production. ese
BioMed Research International
0 50 100 150 200 250 300
0
100
200
300 Mitochondria 2,4-DNP
Time (s)
Membrane potential (AFU)
Control
(PhSe)2
MeHg
(PhSe)2+ MeHg
(a)
0 50 100 150 200 250 300
0
100
200
300
Mitochondria 2,4-DNP
Time (s)
Membrane potential (AFU)
Control
(PhSe)2
MeHg
(PhSe)2+ MeHg
(b)
0
50
100
150
200
250
Control (PhSe)2MeHg (PhSe)2+ MeHg
Membrane potential (ΔΨm)
ΔΨm1
ΔΨm2
(c)
0
50
100
150
200
250
Control (PhSe)2MeHg (PhSe)2+ MeH
g
Membrane potential (ΔΨm)
ΔΨm1
ΔΨm2
(d)
F : Mitochondrial depolarization in liver (a), (c) and brain (b), (d) of rats exposed to MeHg and/or (PhSe)2. Figures (a) and (b) show
mitochondrial membrane potential (AFU). Figures (c) and (d) show mitochondrial ΔΨm. ΔΨm = delta of uorescence before (time ) and
aer addition of mitochondria (time  seconds) and ΔΨm = delta of uorescence before (time  seconds) and aer addition of , DNP
(time  seconds). Data are expressed as mean ±S.D., 𝑛=4.
results are corroborated by the fact that MeHg aected total
thiols but not nonprotein thiol levels in brain mitochondria.
MeHg caused a decrease in the total mitochondrial thiol
levels in brain, which is related mainly with protein thiols,
andisinagreementwiththeinhibitionofmitochondrial
dehydrogenases activity in this tissue. On the other hand,
MeHg did not aect nonprotein thiol levels (mainly GSH)
in brain mitochondria, which can explain the normal ROS
production, since GSH is the main antioxidant in brain.
e cotreatment with (PhSe)2prevented the MeHg-
induced mitochondrial total and nonprotein thiol groups
depletion in the brain and liver (Figure ). e ecacy of
(PhSe)2in preventing thiol depletion may reside in its ability
to form a complex with MeHg, thus eectively reducing
MeHg binding to protein and free thiols. Treatment with
(PhSe)2also partially protected the liver from mitochondrial
MeHg-induced swelling (Figure (a)). However, the cotreat-
ment with (PhSe)2failed to reverse the MeHg-induced mito-
chondrial swelling (Figure (b))anddecreasedmitochon-
drial metabolic function (Figure (b))inthebrainaswellas
increased mitochondrial ROS production (Figure (a))inthe
liver. ese results indicate that mechanisms other than the
interaction with important free and protein thiols are likely
involved in the MeHg-induced mitochondrial dysfunction.
us, the preferential anity of MeHg for specic, and as
of yet unidentied, mitochondrial protein targets may have
acriticalroleinMeHgstoxicity.
Previous studies have demonstrated that MeHg can
directly inhibit TrxR activity both in vitro and in vivo [,
,]. Mammalian TrxR is a selenoenzyme containing a
 BioMed Research International
Control (PhSe)2MeHg (PhSe)2+ MeH
g
0
3
6
9
12
(nmol of TNB/min/mg protein)
(a)
Control (PhSe)2MeHg (PhSe)2+ MeH
g
#
0
2
4
6
8
10
(nmol of TNB/min/mg protein)
(b)
Control (PhSe)2MeHg (PhSe)2+ MeHg
0
1
2
3
4
(nmol of TNB/min/mg protein)
(c)
F : Tr xR activity in liver (a), kidney (b), and brain (c) of rats exposed to MeHg and/or (PhSe)2. Data are expressed as mean ±S.D., 𝑛=4.
()represents𝑃 < 0.05 as compared to controls by Mann-Whitney test. () represents 𝑃 < 0.05 as compared to controls by Mann-Whitney
test.
unique, catalytically active selenolthiol/selenenylsulde in
the conserved C-terminal sequence (-Gly-Cys-Sec-Gly) [].
ree mammalian TrxR selenoenzymes have been identi-
ed, the cytosolic enzyme TrxR, the mitochondrial enzyme
TrxR, and a testis-specic enzyme thioredoxin-glutathione
reductase (TGR/TrxR) []. Here, we show that MeHg
treatment inhibited rat TrxR activity in brain, liver, and
kidney (Figure ). MeHg forms covalent bonds between its
Hg moiety and the Se of the selenocysteine of the enzyme,
thus directly inhibiting the activity of TrxR []. Since TrxR is
critical for cellular antioxidant defense system the inhibition
ofthisenzymelikelyhasacentralroleinmediatingthe
toxicity of MeHg.
Recently, diphenyl diselenide was demonstrated to be
a substrate for cerebral and hepatic rat TrxR, which could
account, at least in part, for the antioxidant properties of
(PhSe)2[]. Herein, rats treated solely with (PhSe) showed
an increase in the activity of renal TrxR (Figures (a) and
(b), resp.). e formation of selenhidric acid from (PhSe)2
couldalsoexplaintheincreaseinTrxRactivity,sincethis
inorganic form of Se can be converted to selenocysteine
and incorporated to selenoenzymes, such as TrxR [,
]. Accordingly, Zhang et al. [] have demonstrated that
organoselenium compounds (including diselenide) increase
theexpressionofTrxRinwhitebloodcellslinesinculture.
e cotreatment with (PhSe)2and MeHg was ineective in
attenuating the inhibition of MeHg-induced TrxR in liver,
kidney, and brain (Figure ). Similarly, studies in vitro and
in vivo have previously corroborated that selenite was able
to recover the activity of HgCl2-induced TrxR inhibition but
not in response to MeHg. e eect of Se (as selenide) was
attributed to the displacement of Hg from the active site,
giving rise to mercury selenide and regenerating the TrxR
selenol [,].
5. Conclusions
In conclusion, the results of this study established that
(PhSe)2can increase Hg body burden (likely associated with
release of inorganic Se from (PhSe)2) and MeHg neurotoxic-
ityinratsdespitethefactthat(PhSe)
2blunted the deleterious
eects of MeHg on thiol levels. e results presented herein
also reinforce the central role of mitochondrial dysfunction in
mediating the aberrant eects of MeHg in vivo,aswellasthe
role of TrxR as a molecular target for MeHg in the rat. Further
research into MeHg-(PhSe)2interactions will be helpful in
characterizing the consequences concomitant exposures to
theseandrelatedcompounds.
BioMed Research International 
Conflict of Interests
e authors declare that there is no conict of interests regar-
ding the publication of this paper.
Acknowledgments
is work was supported by Fundac¸˜
ao de Amparo a Pesquisa
do Estado do Rio Grande do Sul (FAPERGS), CAPES
(Coordenac¸˜
ao de Aperfeic¸oamento de Pessoal de N´
ıvel
Superior), CNPq (Conselho Nacional de Desenvolvimento
Cient´
ıco e Tecnol ´
ogico), FINEP (Rede Instituto Brasileiro
de Neurociˆ
encia (IBN-Net) no. ..-), FAPERGS-
PRONEX-CNPQ, and INCT-EN (Instituto Nacional de
Ciˆ
encia e Tecnologia em Excitotoxicidade e Neuroprotec¸˜
ao).
Cristiane L. Dalla Corte is recipient of CAPES fellowship.
F´
elix A. A. Soares and Jo˜
ao B. T. Rocha are recipients of
CNPq fellowships. Michael Aschner was partially supported
by grants from the US PHS, R ES and R ES.
References
[] C. M. L. Carvalho, J. Lu, X. Zhang, E. S. J. Arn´
er, and A. Holm-
gren, “Eects of selenite and chelating agents on mammalian
thioredoxin reductase inhibited by mercury: implications for
treatment of mercury poisoning,e FASEB Journal,vol.,
no. , pp. –, .
[] T.W.Clarkson,L.Magos,andG.J.Myers,“etoxicologyof
mercury—current exposures and clinical manifestations,e
New England Journal of Medicine,vol.,no.,pp.,
.
[] G. C. Compeau and R. Bartha, “Sulfate-reducing bacteria:
principal methylators of mercury in anoxic estuarine sediment,
Applied and Environmental Microbiology,vol.,no.,pp.
, .
[] J.G.D
´
orea, “Persistent, bioaccumulative and toxic substances
in sh: human health considerations,Science of the Total
Environment,vol.,no.,pp.,.
[] P. Grandjean and K. T. Herz, “Methylmercury and brain devel-
opment: imprecision and underestimation of developmental
neurotoxicity in humans,Mount Sinai Journal of Medicine,vol.
, no. , pp. –, .
[]C.Johansson,A.F.Castoldi,N.Onishchenko,L.Manzo,M.
Vahter, and S. Ceccatelli, “Neurobehavioural and molecular
changes induced by methylmercury exposure during develop-
ment,Neurotoxicity Research, vol. , no. -, pp. –, .
[] M.Farina,M.Aschner,andJ.B.T.Rocha,“Oxidativestressin
MeHg-induced neurotoxicity,” Toxicology and Applied Pharma-
cology,vol.,no.,pp.,.
[] J. L. Franco, H. C. Braga, J. Stringari et al., “Mercurial-induced
hydrogen peroxide generation in mouse brain mitochondria:
protective eects of quercetin,Chemical Research in Toxicology,
vol. , no. , pp. –, .
[] V. Branco, J. Can´
ario, A. Holmgren, and C. Carvalho, “Inhibi-
tionofthethioredoxinsysteminthebrainandliverofzebra-
seabreams exposed to waterborne methylmercury,Toxicolog y
and Applied Pharmacology,vol.,no.,pp.,.
[] J. L. Franco, T. Posser, P. R. Dunkley et al., “Methylmercury
neurotoxicity is associated with inhibition of the antioxidant
enzyme glutathione peroxidase,Free Radical Biology and
Medicine,vol.,no.,pp.,.
[] C. Wagner, J. H. Sudati, C. W. Nogueira, and J. B. T. Rocha, “In
vivo and in vitro inhibition of mice thioredoxin reductase by
methylmercury,BioMetals,vol.,no.,pp.,.
[] A. Dreiem and R. F. Seegal, “Methylmercury-induced changes
in mitochondrial function in striatal synaptosomesare calcium-
dependent and ROS-independent,NeuroToxicology,vol.,
no. , pp. –, .
[] V. Glaser, E. M. Nazari, Y. M. R. M¨
uller et al., “Eects of
inorganic selenium administration in methylmercury-induced
neurotoxicity in mouse cerebral cortex,International Journal of
Developmental Neuroscience,vol.,no.,pp.,.
[] D. H. Roos, R. L. Puntel, M. Farina et al., “Modulation of
methylmercury uptake by methionine: prevention of mitochon-
drial dysfunction in rat liver slices by a mimicry mechanism,
Toxicology and Applied Pharmacology,vol.,no.,pp.,
.
[] A. L. Choi, E. Budtz-Jørgensen, P. J. Jørgensen et al., “Selenium
as a potential protective factor against mercury developmental
neurotoxicity,” Environmental Research,vol.,no.,pp.,
.
[] A. Fredriksson, A. Teiling Gardlund, K. Bergman et al., “Eects
of maternal dietary supplementation with selenite on the post-
natal development of rat ospring exposed to methyl mercury
in utero,Pharmacology and Toxicology,vol.,no.,pp.
, .
[] D. L. Hateld and V. N. Gladyshev, “How selenium has altered
our understanding of the genetic code,Molecular and Cellular
Biology, vol. , no. , pp. –, .
[] C.L.DallaCorte,F.A.A.Soares,M.Aschner,andJ.B.T.
Rocha, “Diphenyl diselenide prevents methylmercury-induced
mitochondrial dysfunction in rat liver slices,Tet r a h e d r on ,vol.
, no. , pp. –, .
[] D. H. Roos, R. L. Puntel, M. M. Santos et al., “Guano-
sine and synthetic organoselenium compounds modulate
methylmercury-induced oxidative stress in rat brain cortical
slices: involvement of oxidative stress and glutamatergic sys-
tem,Toxicolog y i n Vit r o ,vol.,no.,pp.,.
[] Z. Yin, E. Lee, M. Ni et al., “Methylmercury-induced alterations
in astrocyte functions are attenuated by ebselen,NeuroToxicol-
ogy, vol. , no. , pp. –, .
[] A. S. de Freitas, V. R. Funck, M. D. S. Rotta et al., “Diphenyl
diselenide, a simple organoselenium compound, decreases
methylmercury-induced cerebral, hepatic and renal oxidative
stress and mercury deposition in adult mice,Brain Research
Bulletin,vol.,no.,pp.,.
[] C.W.NogueiraandJ.B.T.Rocha,“Diphenyldiselenideajanus-
faced molecule,Journal of the Brazilian Chemical Society,vol.
, no. , pp. –, .
[] A. S. de Freitas, A. de Souza Prestes, C. Wagner et al., “Reduction
of diphenyl diselenide and analogs by mammalian thioredoxin
reductase is independent of their gluthathione peroxidase-like
activity: a possible novel pathway for their antioxidant activity,
Molecules, vol. , no. , pp. –, .
[] L. Magos, “e absorption, distribution, and excretion of
methylmercury,” in e Toxicity of Methylmercury,C.U.Edccles
andZ.Annau,Eds.,pp.,JohnHopkinsUniversityPress,
Baltimore, Md, USA, .
[] R. Doi and M. Tagawa, “A study on the biochemical and
biological behavior of methylmercury,” Toxicolog y and Appl i e d
Pharmacology,vol.,no.,pp.,.
 BioMed Research International
[] V. B. Brito, V. Folmer, G. O. Puntel et al., “Diphenyl diselenide
and ,-dimercaptopropanol increase the PTZ-induced chemi-
cal seizure and mortality in mice,Brain Research Bulletin,vol.
,no.,pp.,.
[] C. W. Nogueira and J. B. T. Rocha, “Toxicology and phar-
macology of selenium: emphasis on synthetic organoselenium
compounds,Archives of Toxicology,vol.,no.,pp.,
.
[] M. Prigol, R. F. Schumacher, C. WayneNogueira, and G. Zeni,
“Convulsant eect of diphenyl diselenide in rats and mice and
its relationship to plasma levels,Tox i c o l o g y Letters,vol.,no.
,pp.,.
[] C. Paulmier, “Selenoorganic functional groups,” in Selenium
Reagents and Intermediates in Organic Synthesis,pp.,
Pergamon Press, Oxford, UK,  edition, .
[] J. Christinal and T. Sumathi, “Eect of Bacopa monniera extract
on methylmercury-induced behavioral and histopathological
changes in rats,Biological Trace Element Research,vol.,no.
,pp.,.
[] T. Sumathi, C. Shobana, J. Christinal, and C. Anusha, “Pro-
tective eect of Bacopa monniera on methyl mercury-induced
oxidative stress in cerebellum of rats,Cellular and Molecular
Neurobiology,vol.,no.,pp.,.
[] G. R. Bhagure and S. R. Mirgane, “Heavy metal concentrations
in groundwaters and soils of ane Region of Maharashtra,
India,Environmental Monitoring and Assessment,vol.,no.
–, pp. –, .
[] P. L. Broadhurst, “e place of animal psychology in the
development of psychosomatic research,Fortschritte der Psy-
chosomatischen Medizin,vol.,pp.,.
[] R. Lalonde, T. L. Lewis, C. Strazielle, H. Kim, and K. Fukuchi,
“Transgenic mice expressing the 𝛽APPSWE mutation:
eects on exploratory activity, anxiety, and motor coordina-
tion,Brain Research,vol.,no.,pp.,.
[] R. Lalonde, H. D. Kim, and K. Fukuchi, “Exploratory activity,
anxiety, and motor coordination in bigenic APPswe + PS/ΔE
mice,Neuroscience Letters,vol.,no.,pp.,.
[] A. Holmgren and M. Bjornstedt, “ioredoxin and thioredoxin
reductase,Methods in Enzymology, vol. , pp. –, .
[] N. Brustovetsky and J. M. Dubinsky, “Dual responses of CNS
mitochondria to elevated calcium,Journal of Neuroscience,vol.
, no. , pp. –, .
[] G. L. Ellman, “Tissue sulydryl groups,Archives of Biochem-
istry and Biophysics,vol.,no.,pp.,.
[]Q.Guo,S.Christakos,N.Robinson,andM.P.Mattson,
“Calbindin DK blocks the proapoptotic actions of mutant
presenilin : reduced oxidative stress and preserved mitochon-
drial function,Proceedings of the National Academy of Sciences
of the United States of America,vol.,no.,pp.,
.
[] C. Garc´
ıa-Ruiz, A. Colell, M. Mar´
ı, A. Morales, and J. C.
Fern´
andez-Checa, “Direct eect of ceramide on the mitochon-
drialelectrontransportchainleadstogenerationofreactive
oxygen species: role of mitochondrial glutathione,Journal of
Biological Chemistry, vol. , no. , pp. –, .
[] T. Mosmann, “Rapid colorimetric assay for cellular growth and
survival: application to proliferation and cytotoxicity assays,
Journal of Immunological Methods,vol.,no.-,pp.,
.
[] T. V. Votyakova and I. J. Reynolds, “Ca2+-induced permeabiliza-
tion promotes free radical release from rat brain mitochondria
with partially inhibited complex I,Journal of Neurochemistry,
vol. , no. , pp. –, .
[] M. M. Bradford, “A rapid and sensitive method for the quanti-
tation of microgram quantities of protein utilizing the principle
of protein dye binding ,Analytical Biochemistry,vol.,no.-,
pp. –, .
[] P. B. Mello, F. Benetti, M. Cammarota, and I. Izquierdo, “Eects
of acute and chronic physical exercise and stress on dierent
types of memory in rats,Anais da Academia Brasileira de
Ciencias,vol.,no.,pp.,.
[]C.Pinheiro,J.L.M.DoNascimento,L.C.L.Silveira,J.B.
T. Rocha, and M. Aschner, “Mercury and selenium—a review
on aspects related to the health of human populations in the
Amazon,Environmental Bioindicators,vol.,no.,pp.
, .
[] F. E. Huggins, S. A. Raverty, O. S. Nielsen, N. E. Sharp, J. D.
Robertson, and N. V. C. Ralston, “An XAFS investigation of
mercury and selenium in Beluga whale tissues,Environmental
Bioindicators,vol.,no.,pp.,.
[] H. Iwata, T. Masukawa, H. Kito, and M. Hayashi, “Degradation
of methylmercury by selenium,Life Sciences,vol.,no.,pp.
–, .
[] J. H. Palmer and G. Parkin, “-Seleno--alkylbenzimidazoles
and their diselenides: synthesis and structural characterization
of a -seleno--methylbenzimidazole complex of mercury,
Polyhedron,vol.,pp.,.
[] B. Moller-Madsen and G. Danscher, “Localization of mercury
in CNS of the rat: IV. e eect of selenium on orally admin-
istered organic and inorganic mercury,” Toxicolog y and Appl i e d
Pharmacology,vol.,no.,pp.,.
[] J. R. Prohaska and H. E. Ganther, “Interactions between
selenium and methylmercury in rat brain,Chemico-Biological
Interactions,vol.,no.,pp.,.
[] M.O.Dietrich,C.E.Mantese,G.D.Anjos,D.O.Souza,and
M. Farina, “Motor impairment induced by oral exposure to
methylmercury in adult mice,Environmental Toxicology and
Pharmacology,vol.,no.,pp.,.
[] M. Aschner, T. Syversen, D. O. Souza, J. B. T. Rocha, and M.
Farina, “Involvement of glutamate and reactive oxygen species
in methylmercury neurotoxicity,Brazilian Journal of Medical
and Biological Research,vol.,no.,pp.,.
[] D. W. Choi, “Excitotoxic cell death,Journal of Neurobiolo gy,vol.
,no.,pp.,.
[] S. Ceccatelli, E. Dar´
e, and M. Moors, “Methylmercury-induced
neurotoxicity and apoptosis,” Chemico-Biological Interactions,
vol. , no. , pp. –, .
[] N. Mori, A. Yasutake, and K. Hirayama, “Comparative study of
activities in reactive oxygen species production/defense system
in mitochondria of rat brain and liver, and their susceptibility
to methylmercury toxicity,Archives of Toxicology,vol.,no.
, pp. –, .
[] L.V.Papp,A.Holmgren,andK.K.Khanna,“Seleniumand
selenoproteins in health and disease,Antioxidants and Redox
Signaling,vol.,no.,pp.,.
[] W.J.AdamsJr.,J.J.Kocsis,andR.Snyder,“Acutetoxicityand
urinary excretion of diphenyldiselenide,Tox i c o l o g y Let te r s ,vol.
,no.,pp.,.
[] G. Zhang, V. Nitteranon, S. Guo et al., “Organoselenium com-
pounds modulate extracellular redox by induction of extracel-
lular cysteine and cell surface thioredoxin reductase,Chemical
Research in Toxicology, vol. , no. , pp. –, .
... The membrane potential was quickly lost in both groups, and this profile may reflect immature liver mitochondria. However, an absence of membrane disruption was observed for the brain-isolated mitochondria in the presence of TM; a similar behavior was related by Corte et al. (2013) [58] with Wistar rats exposed to methylmercury (5 mg kg -1 i.g.). ...
... The membrane potential was quickly lost in both groups, and this profile may reflect immature liver mitochondria. However, an absence of membrane disruption was observed for the brain-isolated mitochondria in the presence of TM; a similar behavior was related by Corte et al. (2013) [58] with Wistar rats exposed to methylmercury (5 mg kg -1 i.g.). ...
... This profile may be due to the quantity of glutathione because the levels for the liver are around 5 -10 mM [62], while the concentration for the different brain cell types ranges from 1 -3 mM [63]. The mercury species released by TM is EtHg + , which could cross the blood-brain barrier [58], since in low amounts it can bind to the thiol group presented in a reduced glutathione system, decreasing its antioxidant capacity. J o u r n a l P r e -p r o o f Furthermore, cognitive impairment was investigated by examining the loss of acetylcholinesterase enzyme activity, which is the enzyme that regulates acetylcholine dependent synaptic flow [64]. ...
... (PhSe) 2 was demonstrated to decrease cerebral, hepatic and renal content of Hg in adult mice (de Freitas et al. 2009). In contrast, a recent in vivo study demonstrated co-treatment with (PhSe) 2 and MeHg caused an increase hepatic, cerebral and muscle levels of Hg in rats (Dalla Corte et al. 2013). ...
... Rat liver mitochondria were isolated according to Brustovetsky and Dubinsky (2000), with modifications (Dalla Corte et al. 2012, 2013Puntel et al. 2010). The liver slices were quickly weighed and homogenized in 1:5 (w/v) ice-cold buffer containing 225 mM mannitol, 75 mM sucrose, 1 mM K ? ...
... It is well known that (PhSe) 2 is a potent antioxidant being demonstrated to have beneficial effects in several oxidative stress-related disease models (Nogueira and Rocha 2011). Since oxidative stress has been implicated in MeHg toxicity, reflecting its interaction with protein containing thiol and selenol ), (PhSe) 2 could act through its On the other hand, interaction between (PhSe) 2 and MeHg in vivo is much more intricate and there are two studies showing discrepant effects of (PhSe) 2 in change Hg levels (Dalla Corte et al. 2013;de Freitas et al. 2009). In a study with mice, (PhSe) 2 caused a decrease in Hg body burden after a concomitant treatment with MeHg (de Freitas et al. 2009). ...
Article
Full-text available
Organoseleno-compounds have been investigated for its beneficial effects against methylmercury toxicity. In this way, diphenyl diselenide (PhSe)2 was demonstrated to decrease Hg accumulation in mice, protect against MeHg-induced mitochondrial dysfunction, and protect against the overall toxicity of this metal. In the present study we aimed to investigate if co-treatment with (PhSe)2 and MeHg could decrease accumulation of Hg in liver slices of rats. Rat liver slices were co-treated with (PhSe)2 (0.5; 5 µM) and/or MeHg (25 µM) for 30 min at 37 °C and Se and Hg levels were measured by inductively coupled plasma mass spectrometry (ICP-MS) in the slices homogenate, P1 fraction, mitochondria and incubation medium. Co-treatment with (PhSe)2 and MeHg did not significantly alter Se levels in any of the samples when compared with compounds alone. In addition, co-treatment with (PhSe)2 and MeHg did not decrease Hg levels in any of the samples tested, although, co-incubation significantly increased Hg levels in homogenate. We suggest here that (PhSe)2 could exert its previously demonstrated protective effects not by reducing MeHg levels, but forming a complex with MeHg avoiding it to bind to critical molecules in cell.
... Therefore, there is a strong rationale to suggest that this compound provides cerebral protection against mitochondrial impairments induced by hypoxia-related brain diseases, which may extend to recurrent injuries. However, it is important to consider the possibility of adverse effects associated with (PhSe)2, as previously reported (Dalla Corte et al. 2013;Puntel et al. 2013). ...
... However, other studies have reported neurotoxicity resulting from its pro-oxidant action (Souza et al. 2010;Leão et al. 2018). Consequently, the administration of (PhSe) 2 has demonstrated undesirable effects in other dysfunctions (Burger et al. 2004;Dalla Corte et al. 2013), while studies using stroke models have reported a protective action associated with its antioxidant and anti-inflammatory properties (Brüning et al. 2012;Dobrachinski et al. 2014). ...
Article
Full-text available
Hypoxia plays a significant role in the development of various cerebral diseases, many of which are associated with the potential risk of recurrence due to mitochondrial damage. Conventional drug treatments are not always effective for hypoxia-related brain diseases, necessitating the exploration of alternative compounds. In this study, we investigated the potential of diphenyl diselenide [(PhSe)2] to ameliorate locomotor impairments and mitigate brain mitochondrial dysfunction in zebrafish subjected to hypoxia. Additionally, we explored whether these improvements could confer resistance to recurrent hypoxia. Through a screening process, an appropriate dose of (PhSe)2 was determined, and animals exposed to hypoxia received a single intraperitoneal injection of 100 mg/kg of the compound or vehicle. After 1 h from the injection, evaluations were conducted on locomotor deficits, (PhSe)2 content, mitochondrial electron transport system, and mitochondrial viability in the brain. The animals were subsequently exposed to recurrent hypoxia to assess the latency time to hypoxia symptoms. The findings revealed that (PhSe)2 effectively crossed the blood–brain barrier, attenuated locomotor deficits induced by hypoxia, and improved brain mitochondrial respiration by modulating complex III. Furthermore, it enhanced mitochondrial viability in the telencephalon, contributing to greater resistance to recurrent hypoxia. These results demonstrate the beneficial effects of (PhSe)2 on both hypoxia and recurrent hypoxia, with cerebral mitochondria being a critical target of its action. Considering the involvement of brain hypoxia in numerous pathologies, (PhSe)2 should be further tested to determine its effectiveness as a potential treatment for hypoxia-related brain diseases. Graphical Abstract
... 25,306,307 The formation of R-Se-Hg-R adducts such as PhSeHgCH 3 was accompanied by a decrease in the liver, kidney, and brain Hg burden in mice, but not in rats. 308 In contrast to mice, the co-treatment with diphenyl diselenide and methylmercury caused an increase in the deposition of Hg in the brain and liver of rats, 308 suggesting that the formation of the complex PhSeHgCH 3 is followed by its rupture to insoluble mercury selenide (HgSe) (Fig. 12). The formation of different types of Hg-Se complexes (including a variety of insoluble nanoparticles) has been recently reported in several species of animals and plants. ...
... 25,306,307 The formation of R-Se-Hg-R adducts such as PhSeHgCH 3 was accompanied by a decrease in the liver, kidney, and brain Hg burden in mice, but not in rats. 308 In contrast to mice, the co-treatment with diphenyl diselenide and methylmercury caused an increase in the deposition of Hg in the brain and liver of rats, 308 suggesting that the formation of the complex PhSeHgCH 3 is followed by its rupture to insoluble mercury selenide (HgSe) (Fig. 12). The formation of different types of Hg-Se complexes (including a variety of insoluble nanoparticles) has been recently reported in several species of animals and plants. ...
Article
The selenium element is essential of some life forms and its biological-chemistry function is mainly performed by the selenol/selenolate moiety (-SeH/-Se-) in a few selenoproteins. Many synthetic organoselenium compounds (OSeCs)...
... This loss in body and organ weight could be a result of elevated lipid and protein degeneration induced by cadmium and mercury. These findings agree with other reports showing that heavy metals such as cadmium and mercury cause disturbances in the total body and organ weights of rats possibly due to reduced water and food consumption, tissue injury and 19,20 impaired organ function. Diminished cerebellar weight due to the toxicity of heavy metals has also 21,22 been previously reported. ...
Article
Full-text available
Background: The unprecedented increase in metal exposure has been aided by modern industrialization and anthropogenic activities. Cadmium and mercury are recognized as two of the most common heavy metals with destructive impacts on most organ systems. The present study was designed to investigate and improve existing literature on the possible deleterious effects of cadmium and mercury exposure. Methods: Adult Wistar rats were treated with cadmium chloride (5 mg/kg/day) and mercury chloride (4 mg/kg/day) for 14 days. Body, brain and cerebellar weights, motor deficits, antioxidant and lipid peroxidation activities as well as histological alterations to the cerebellum were evaluated at the end of the experiment. Results: Findings showed a significant reduction in body and brain weights, dysregulation of antioxidant enzymes activity and impaired locomotion and exploratory activity in treated rats. Also, an increase in lipid peroxidation and degeneration of Purkinje cells of the cerebellum was observed in treated rats. Conclusion: Overall, these results corroborate previous findings that cadmium and mercury induce deleterious effects on the cerebellum and central nervous system. In addition, this study helps to provide an anatomical perspective and information on the exact cerebellar changes induced by cadmium and mercury in Wistar rats.
... Improvement in mitochondrial function, such as adequate ATP levels, can be achieved through modulation of antioxidant defense by mitochondriatargeted antioxidants (Siegel et al. 2013). Indeed, supplementation with Se in MeHg intoxicated animals has been shown to mitigate mitochondrial dysfunction and other toxic outcomes (Dalla Corte et al. 2013). In addition to the cellular antioxidant defense, a closely related parallel system, namely, the cellular protein quality control, has also been invoked in the defense against MeHg-induced toxicity (Hashimoto-Torii et al. 2014). ...
Article
Full-text available
Human exposure to the neurotoxin methylmercury (MeHg) poses a significant health risk to the development of the nervous system. The mechanisms of MeHg-induced neurotoxicity are associated with the disruption of cellular homeostasis, and include oxidative stress, loss of calcium homeostasis, and impaired protein quality control. The stress inducible protein 1 (STI-1) is involved in the regulation of protein quality control by acting as a protein cochaperone to maintain optimal protein unfolding and refolding. Here, we utilized the Caenorhabditis elegans (C. elegans) model of MeHg toxicity to characterize the role of the sti-1 gene in MeHg-induced toxicity. We showed that lifespan and developmental milestone timings were significantly altered in sti-1 knockout (KO) animals with MeHg exposure. However, knocking down sti-1 by RNAi did not result in an analogous effect for lifespan, but did still sensitize to delays in developmental milestone progression by acute MeHg, suggesting that insufficiency of sti-1 does not recapitulate all phenotypes of the null mutation. Furthermore, inhibition of ATP levels by MeHg exposure was modulated by sti-1. Considering that the skn-1/gst-4 pathway is highly involved in metal’s toxicity, such pathway was also explored in our model. We showed that sti-1 mutant worms exhibited impaired capacity to upregulate the antioxidant genes skn-1/gst-4, highlighting a central role of sti-1 in modulating antioxidant response. Lastly, we showed that loss-of-function mutation in the rrf-3 gene, which encodes a putative RNA-directed RNA polymerase, has significant effect in altering MeHg-induced toxicity by potentiating the animal’s detoxification system. Altogether, our novel data show an indispensable role of protein quality control in the defense against MeHg toxicity.
... Our data show that MeHg-challenged mice exhibited increased oxidative stress. Wistar rats treated with 5 mg/kg/day MeHg and 1 mg/ kg/day of diphenyl diselenide, intragastrically for 21 days, showed increased accumulation of Hg in the liver and the brain, leading to motor deficits and body weight loss (Dalla Corte et al., 2013). Increased MeHg accumulation in the liver may cause liver dysfunction and altered circulating plasma lipids seen in our study. ...
Article
Mercury (Hg) is one of the most toxic environmental pollutants, especially when methylated, forming methylmercury (MeHg). MeHg affects DNA repair, increases oxidative stress, and predisposes to cancer. MeHg neurotoxicity is well-known, but recently MeHg-associated cardiovascular effects were recognized. This study evaluated circulating lipids, oxidative stress, and genotoxicity after MeHg-chronic exposure (20 mg/L in drinking water) in C57BL/6 J wild-type and APOE knockout (ko) mice, the latter, being spontaneously dyslipidemic. Experimental mice were assigned to four groups: non-intoxicated and MeHg-intoxicated wild-type mice and non-intoxicated and MeHg-intoxicated APOE ko mice. Plasma levels of triglycerides, total cholesterol (TC), HDL, and LDL were analyzed. Liver lipid peroxidation and splenic gene expression of xeroderma pigmentosum complementation groups A, C, D, and G (XPA, XPC, XPD, and XPG), X-ray repair cross-complementing protein 1 (XRCC1), and telomerase reverse transcriptase (TERT) were measured. Fur Hg levels confirmed chronic MeHg intoxication. MeHg exposure raises TC levels both in wild-type and APOE ko mice. HDL and LDL-cholesterol levels were increased only in the MeHg-challenged APOE ko mice. MeHg increased liver lipid peroxidation, regardless of the genetic background. Unintoxicated APOE ko mice showed higher expression of TERT than all other groups. APOE dificency increases XPA expression, regardless of MeHg intoxication. Furthermore, MeHg-intoxicated mice had more cytogenetic abnormalities, effect which was independent of APOE deficiency. More studies are needed to dissect the interactions between circulating lipids, MeHg intoxication, and DNA-repair pathways even at young age, interactions that likely play critical roles in cell senescence and the risk for chronic disorders later in life.
... Since selenols have a lower pK a than thiols and are fully ionized to selenolates under physiological conditions [39,41], selenols are more reactive toward Hg [40]. The decrease in TrxR1 activity caused by MeHg exposure has been shown in vitro and in vivo [23,40,42]. The reduction in activity of the cellular Trx system under MeHg exposure has also been demonstrated [40]. ...
Article
Full-text available
Methylmercury (MeHg) is a well-known neurotoxicant that causes severe intoxication in humans. In Japan, it is referred to as Minamata disease, which involves two characteristic clinical forms: fetal type and adult type depending on the exposed age. In addition to MeHg burden level, individual susceptibility to MeHg plays a role in the manifestation of MeHg toxicity. Research progress has pointed out the importance of oxidative stress in the pathogenesis of MeHg toxicity. MeHg has a high affinity for selenohydryl groups, sulfhydryl groups, and selenides. It has been clarified that such affinity characteristics cause the impairment of antioxidant enzymes and proteins, resulting in the disruption of antioxidant systems. Furthermore, MeHg-induced intracellular selenium deficiency due to the greater affinity of MeHg for selenohydryl groups and selenides leads to failure in the recoding of a UGA codon for selenocysteine and results in the degradation of antioxidant selenoenzyme mRNA by nonsense-mediated mRNA decay. The defect of antioxidant selenoenzyme replenishment exacerbates MeHg-mediated oxidative stress. On the other hand, it has also been revealed that MeHg can directly activate the antioxidant Keap1/Nrf2 signaling pathway. This review summarizes the incidence of MeHg-mediated oxidative stress from the viewpoint of the individual intracellular redox system interactions and the MeHg-mediated aforementioned intracellular events. In addition, the mechanisms of cellular stress pathways and neuronal cell death triggered by MeHg-mediated oxidative stress and direct interactions of MeHg with reactive residues of proteins are mentioned.
Article
Full-text available
Here, we addressed the pharmacology and toxicology of synthetic organoselenium compounds and some naturally occurring organoselenium amino acids. The use of selenium as a tool in organic synthesis and as a pharmacological agent goes back to the middle of the nineteenth and the beginning of the twentieth centuries. The rediscovery of ebselen and its investigation in clinical trials have motivated the search for new organoselenium molecules with pharmacological properties. Although ebselen and diselenides have some overlapping pharmacological properties, their molecular targets are not identical. However, they have similar anti-inflammatory and antioxidant activities, possibly, via activation of transcription factors, regulating the expression of antioxidant genes. In short, our knowledge about the pharmacological properties of simple organoselenium compounds is still elusive. However, contrary to our early expectations that they could imitate selenoproteins, organoselenium compounds seem to have non-specific modulatory activation of antioxidant pathways and specific inhibitory effects in some thiol-containing proteins. The thiol-oxidizing properties of organoselenium compounds are considered the molecular basis of their chronic toxicity; however, the acute use of organoselenium compounds as inhibitors of specific thiol-containing enzymes can be of therapeutic significance. In summary, the outcomes of the clinical trials of ebselen as a mimetic of lithium or as an inhibitor of SARS-CoV-2 proteases will be important to the field of organoselenium synthesis. The development of computational techniques that could predict rational modifications in the structure of organoselenium compounds to increase their specificity is required to construct a library of thiol-modifying agents with selectivity toward specific target proteins.
Article
Full-text available
We reevaluate the treatment of mercury poisoning, incorporating recent advances in understanding of mercury toxicity and the mercury:selenium interaction. This review focuses on: 1) the role, limitations and benefits of chelation (Unithiol, succimer and N-Acetylcysteine); 2) the role of selenium supplementation; and 3) how the different forms of mercury are impacted by use of chelation and selenium. Unithiol and succimer produce increases in urinary excretion of mercury and to a lesser degree blood and total body mercury. The primary role of N-acetylcysteine is increasing renal mercury excretion, similar to the thiol-chelators. Additional unique features of acetylcysteine include increased efflux of methylmercury from the brain, and reduced oxidative stress via increased glutathione production. The role of selenium includes: 1) restoration of selenoprotein activity, 2) protection against mitochondrial injury and DNA damage, 3) demethylation of methylmercury, 4) sequestering of mercury via Hg:Se complexes, and 5) redistribution of Hg inside organisms. Selenium may increase blood Hg, via a “sink” effect, causing a redistribution of mercury away from the brain. A combined approach for mercury poisoning treatment was developed focusing on restoration of selenoprotein function, reduction of oxidative stress and increased mercury elimination.
Article
Full-text available
Since the successful use of the organoselenium drug ebselen in clinical trials for the treatment of neuropathological conditions associated with oxidative stress, there have been concerted efforts geared towards understanding the precise mechanism of action of ebselen and other organoselenium compounds, especially the diorganyl diselenides such as diphenyl diselenide, and its analogs. Although the mechanism of action of ebselen and other organoselenium compounds has been shown to be related to their ability to generally mimic native glutathione peroxidase (GPx), only ebselen however has been shown to serve as a substrate for the mammalian thioredoxin reductase (TrxR), demonstrating another component of its pharmacological mechanisms. In fact, there is a dearth of information on the ability of other organoselenium compounds, especially diphenyl diselenide and its analogs, to serve as substrates for the mammalian enzyme thioredoxin reductase. Interestingly, diphenyl diselenide shares several antioxidant and neuroprotective properties with ebselen. Hence in the present study, we tested the hypothesis that diphenyl diselenide and some of its analogs (4,4’-bistrifluoromethyldiphenyl diselenide, 4,4’-bismethoxy-diphenyl diselenide, 4.4’-biscarboxydiphenyl diselenide, 4,4’-bischlorodiphenyl diselenide, 2,4,6,2’,4’,6’-hexamethyldiphenyl diselenide) could also be substrates for rat hepatic TrxR. Here we show for the first time that diselenides are good substrates for mammalian TrxR, but not necessarily good mimetics of GPx, and vice versa. For instance, bis-methoxydiphenyl diselenide had no GPx activity, whereas it was a good substrate for reduction by TrxR. Our experimental observations indicate a possible dissociation between the two pathways for peroxide degradation (either via substrate for TrxR or as a mimic of GPx). Consequently, the antioxidant activity of diphenyl diselenide and analogs can be attributed to their capacity to be substrates for mammalian TrxR and we therefore conclude that subtle changes in the aryl moiety of diselenides can be used as tool for dissociation of GPx or TrxR pathways as mechanism triggering their antioxidant activities.
Article
Excessive formation of reactive oxygen species (ROS) and disruption of glutamate uptake have been pointed as two key mechanisms in methylmercury-toxicity. Thus, here we investigate the involvement of glutamatergic system in methylmercury (MeHg) neurotoxicity and whether diphenyl diselenide, ebselen and guanosine could protect cortical rat brain slices from MeHg-induced ROS generation. MeHg (100 and 200 lM) increased 20 ,70 -dichlorodihydrofluorescin (DCFH) oxidation after 2 h of exposure. At 50 lM, MeHg increased DCFH oxidation only after 5 h of exposure. Guanosine (1 and 5 lM) did not caused any effect per se; however, it blocked the increase in DCFH caused by 200 or 50 lM MeHg. Ebselen (5 and 10 lM) decreased significantly the DCFH oxidation after 2 and 5 h of exposure to MeHg. Diphenyl diselenide (5 lM) did not change the basal DCFH oxidation, but abolished the pro-oxidant effect of MeHg. MK801 also abolished the pro-oxidant effect of MeHg. These results demonstrate for the first time the potential antioxidant properties of organoseleniun compounds and guanosine against MeHg-induced ROS generation after short-term exposure in a simple in vitro model. In conclusion, endogenous purine (guanosine) and two synthetic organoselenium compounds can modulate the pro-oxidant effect of MeHg in cortical brain slices.
Article
Article
A protein determination method which involves the binding of Coomassie Brilliant Blue G-250 to protein is described. The binding of the dye to protein causes a shift in the absorption maximum of the dye from 465 to 595 nm, and it is the increase in absorption at 595 nm which is monitored. This assay is very reproducible and rapid with the dye binding process virtually complete in approximately 2 min with good color stability for 1 hr. There is little or no interference from cations such as sodium or potassium nor from carbohydrates such as sucrose. A small amount of color is developed in the presence of strongly alkaline buffering agents, but the assay may be run accurately by the use of proper buffer controls. The only components found to give excessive interfering color in the assay are relatively large amounts of detergents such as sodium dodecyl sulfate, Triton X-100, and commercial glassware detergents. Interference by small amounts of detergent may be eliminated by the use of proper controls.
Article
Methylmercury (MeHg) is a strong soft electrophile chemical form ubiquitously found in environment and it can exhibit highly toxic effects in multiple organs. Previously, we observed that diphenyl diselenide [(PhSe)2] reduced the deposition and toxicity of MeHg in brain, liver and kidney of mice. The present study was designed to investigate the possible interaction of (PhSe)2 with MeHg-induced mitochondrial dysfunction in rat liver slices in vitro. The liver slices were treated with MeHg (25 μM) and/or (PhSe)2 (0.5, 1, and 5 μM) for 30 min at 37 °C, then mitochondria were isolated from these slices, and the reactive oxygen species (ROS) formation, oxygen consumption, membrane potential (ΔΨm), mitochondrial metabolic function, nonprotein and total thiol content, and Glutathione peroxidase (GPx) activity were assessed. MeHg decreased the mitochondrial function by increasing ROS production, impairing oxygen consumption, and collapsing the ΔΨm. (PhSe)2 protected against the MeHg-induced ROS generation and prevented the decrease in the respiratory rate and in the mitochondrial metabolic function [measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction]. (PhSe)2 (0.5 μM) blunted the MeHg-induced ΔΨm collapse; however at 5 μM, (PhSe)2 alone decreased ΔΨm, yet partially protected the mitochondria from MeHg-induced depolarization. MeHg and (PhSe)2 (0.5 μM) alone or in combination had no effect on nonprotein and total thiol levels and on the GPx activity in mitochondria isolated from liver slices. The protection afforded by (PhSe)2 against the MeHg-induced mitochondrial dysfunction can be associated with (PhSe)2 metabolism to a selenol intermediate, forming inert complex(es) with MeHg, thus effectively decreasing its toxicity. Furthermore, the selenol intermediate of (PhSe)2 (selenophenol) may have direct antioxidant properties against peroxides induced by MeHg. In conclusion, the results demonstrate that low (PhSe)2 concentrations effectively prevent the MeHg-induced mitochondrial dysfunction in vitro.
Article
Methylmercury (MeHg) is a well-recognized environmental contaminant with established health risk to human beings by fish and marine mammal consumption. Bacopa monniera (BM) is a perennial herb and is used as a nerve tonic in Ayurveda, a traditional medicine system in India. This study was aimed to evaluate the effect of B. monniera extract (BME) on MeHg-induced toxicity in rat cerebellum. Male Wistar rats were administered with MeHg orally at a dose of 5 mg/kg b.w. for 21 days. Experimental rats were given MeHg and also administered with BME (40 mg/kg, orally) 1 h prior to the administration of MeHg for 21 days. After treatment period, MeHg exposure significantly decreases the body weight and also caused the following behavioral changes. Decrease tail flick response, longer immobility time, significant decrease in motor activity, and spatial short-term memory. BME pretreatment reverted the behavioral changes to normal. MeHg exposure decreases the DNA and RNA content in cerebellum and also caused some pathological changes in cerebellum. Pretreatment with BME restored all the changes to near normal. These findings suggest that BME has a potent efficacy to alleviate MeHg-induced toxicity in rat cerebellum.