Вы находитесь на странице: 1из 10

Journal of Neuroimmunology 320 (2018) 133–142

Contents lists available at ScienceDirect

Journal of Neuroimmunology
journal homepage: www.elsevier.com/locate/jneuroim

Sex influences in behavior and brain inflammatory and oxidative alterations T


in mice submitted to lipopolysaccharide-induced inflammatory model of
depression
Bruna Stefânia Ferreira Melloa, Adriano José Maia Chaves Filhoa, Charllyany Sabino Custódioa,
Rafaela Carneiro Cordeiroa, Fabio Miyajimaa, Francisca Cléa Florenço de Sousaa,

Silvânia Maria Mendes Vasconcelosa, David Freitas de Lucenaa, Danielle Macedoa,b,
a
Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do
Ceará, Fortaleza, CE, Brazil
b
National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil

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

Keywords: Peripheral inflammation induced by lipopolysaccharide (LPS) causes a behavioral syndrome with translational
Depression relevance for depression. This mental disorder is twice more frequent among women. Despite this, the majority
Lipopolysaccharide of experimental studies investigating the neurobiological effects of inflammatory models of depression have been
Inflammation performed in males. Here, we sought to determine sex influences in behavioral and oxidative changes in brain
Sex
regions implicated in the pathophysiology of mood disorders (hypothalamus, hippocampus and prefrontal cortex
Oxidative stress
- PFC) in adult mice 24 h post LPS challenge. Myeloperoxidase (MPO) activity and interleukin (IL)-1β levels were
Lipid peroxidation
measured as parameters of active inflammation, while reduced glutathione (GSH) and lipid peroxidation as
parameters of oxidative imbalance. We observed that male mice presented behavioral despair, while females
anxiety-like alterations. Both sexes were vulnerable to LPS-induced anhedonia. Both sexes presented increased
MPO activity in the PFC, while male only in the hippocampus. IL-1β increased in the PFC and hypothalamus of
animals of both sexes, while in the hippocampus a relative increase of this cytokine in males compared to
females was detected. GSH levels were decreased in all brain areas investigated in animals of both sexes, while
increased lipid peroxidation was observed in the hypothalamus of females and in the hippocampus of males after
LPS exposure. Therefore, the present study gives additional evidence of sex influence in LPS-induced behavioral
alterations and, for the first time, in the oxidative changes in brain areas relevant for mood regulation.

1. Introduction more recurrent episodes (Schuch et al., 2014).


In the last decades, compelling evidences revealed an important
Depression is a common chronic-recurrent mental disorder. contribution of immune-inflammatory alterations to the pathophy-
Recently, the World Health Organization reported that globally > 300 siology of depression (Miller et al., 2009; Rosenblat et al., 2014). In-
millions of people of all ages suffer from depression, highlighting de- deed, recent meta-analyses have confirmed that depressed patients
pression as the leading cause of disability worldwide (WHO | present increased serum levels of inflammatory markers, such as in-
Depression, 2015). Depression causes great social and economic costs. terleukin-1β (IL-1β), IL-6, tumor necrosis factor (TNF)-alpha and IL-10,
For example, in the United States a survey revealed that 8.3% of all and markers of cell activation, such IL-2 receptors (sIL-2Rs) and
years lived with disability (YLDs) were associated to depression, with neopterin (Farooq et al., 2016; Köhler et al., 2017). Furthermore, the
an enormous economic burden in the order of 210.5 billions of dollars systemic injection of cytokines or of the bacterial endotoxin lipopoly-
each year (Greenberg et al., 2015). Furthermore, there is a well-es- saccharide (LPS) induce depressive symptoms in health humans and
tablished overall gender difference in the prevalence of depression, depressive-like behavior in rodents (Suarez et al., 2004; Vogelzangs
with women outnumbering men 2:1. Additionally, the onset of de- et al., 2016).
pression in women occurs at younger ages with women also presenting In preclinical approaches, LPS-based models of depressive-like


Corresponding author at: Drug Research and Development Center, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Rua Cel. Nunes de Melo 1000,
Fortaleza 60431-270, CE, Brazil.
E-mail address: danielle.macedo@ufc.br (D. Macedo).

https://doi.org/10.1016/j.jneuroim.2018.04.009
Received 21 November 2017; Received in revised form 12 April 2018; Accepted 12 April 2018
0165-5728/ © 2018 Elsevier B.V. All rights reserved.
B.S.F. Mello et al. Journal of Neuroimmunology 320 (2018) 133–142

behavior are accompanied by time-dependent alterations related to 2.2. Animals


sickness behaviors (behavioral inhibition, anorexia, weight loss, fa-
tigue, hyperalgesia, malaise symptoms, neurocognitive impairment and Male and female adult Swiss mice (weighing 25 to 30 g) obtained
anxiety) (Dantzer et al., 2008; Maes et al., 2012) and depressive-like from the Animal house of the Federal University of Ceara were used.
behavior. In this regard, some studies (Custódio et al., 2013; Dantzer Animals were housed 10 per cage under standard polycarbonate cages
et al., 2008; Frenois et al., 2007) have demonstrated that most beha- (42 × 20.5 × 20 cm) and at normal environmental conditions
vioral alterations elicited by the acute (1.5–2 h) exposure to LPS com- (22 ± 1 °C, humidity of 60 ± 5%; reversed 12 h light cycle/darkness
pose the spectrum characteristic of sickness behavior, in which a with lights on at 18:00) with access to food (FRILAB Mouse II, FRIRibe)
marked pyrexia, anorexia and locomotor inhibition occurs. On the and water ad libitum. All experimental procedures were performed
other hand, 24 h later, the emergence of depressive-like behavior between 8:00 AM and 02:00 PM and were conducted in accordance
characterized by despair-, anhedonia- and anxiety-like behaviors takes with the NIH Guide for the Care and Use of Laboratory Animals (NIH,
place. Twenty-four h after LPS exposure motor activity, food and drink 2011) and the Brazilian law for scientific use of animals from the
consumption returns to normal (Dantzer et al., 2008). Brazilian College of Animal Experimentation (COBEA). This research
The influence of sex in the behavioral responses to LPS is not fully protocol was approved by the local ethics committee of the Federal
understood. In this regard, there are studies pointing to a greater sen- University of Ceara.
sibility of female animals to LPS and cytokine effects in some behavioral
aspects, such as sexual activity as well as sucrose and food consumption 2.3. Experimental protocol
(Avitsur and Yirmiya, 1999; Merali et al., 2003). On the other hand,
other studies reported better coping strategies of females in despair-like The animals were randomly divided into four experimental groups:
conditions, such as forced swimming test (Pitychoutis et al., 2009). male LPS-challenged group, male control-group, female LPS-challenged
Furthermore, the findings in the literature about sex influences in LPS- group, female control-group (n = 24 mice/group). LPS-challenged
induced behavioral effects changes markedly accordingly to the dose of groups received an intraperitoneal injection of 0.5 mg/kg LPS dissolved
LPS used, time of assessment and murine specie tested (Badalà et al., in 0.2 ml sterile endotoxin-free SF 0.9% - saline (as vehicle). Control
2008; Cai et al., 2016). animals received saline, 0.1 ml/10 g weight. Twenty-four hours after
It is well estabished that depression is accompanied by increased LPS or saline exposure, the behavioral tests and sample collection were
levels of reactive oxygen- (ROS) and nitrogen- (RNS) species, such as performed. The behavioral tests were conducted in the order of the less
superoxide, peroxynitrite and hydrogen peroxide, and oxidative da- stressful to the more stressful, i.e., open field, elevated plus maze and
mage (Maes et al., 2011a). These pro-oxidative alterations are triggered forced swimming test. A different set of animals was used to perform
in some patients by inflammation and mitochondrial metabolic pro- sucrose preference test due the potential stressful conditions of food and
cesses. In clinical samples, some studies pointed that sex could be a water deprivation involved in this test. In all behavioral determina-
determining factor for ROS and RNS levels in health subjects (Bellanti tions, two raters blinded to the experimental treatment performed the
et al., 2013; Massafra et al., 2002), however little is known about the tests. Two independent experiments were performed to ensure the re-
influence of sex in a depressive subset of patients, that is, those patients producibility of the data. To avoid some potential confounding effect of
with depression induced by inflammatory alterations. Also, regarding the behavioral testing in the neurochemical parameters, a different set
animal models, several studies demonstrated the contribution of oxi- of animals was used to the removal of brain structures. The animals
dative stress in the molecular signature of depressive-like alterations were killed by decapitation, and the brain areas were quickly dissected
(Lucca et al., 2009; Zhang et al., 2009). However, most of them were namely prefrontal cortex (PFC), hippocampus and hypothalamus. All
conducted in male animals, thus not assessing the possible influence of samples were immediately stored −70 °C until assay. The dose of LPS
sex in the oxidative response triggered by depressive-like conditions used and the time of assessment was based on previous studies de-
induced by inflammatory challenge. This issue regarding sex influences monstrating depressive-like behavior and neuroinflammatory altera-
in depression deserves a better comprehension, since it will contribute tions induced by LPS in mice (Custódio et al., 2013; Frenois et al.,
to the development of personalized treatments against this mental 2007).
disorder.
Therefore, taking into account the important and poorly understood 2.4. Behavioral determinations
influence of sex in the manifestation of depressive-like phenomenology
by LPS immune challenge, in this study, we investigated the behavioral 2.4.1. Forced swimming test (FST)
(despair-, anhedonia- and anxiety-like) alterations in male and female The animals were placed individually in an acrylic cylinder (25 cm
mice 24 h post-LPS administration, a time related to the emergence of of height × 10 cm of diameter) containing 8 cm of water at 24 °C. After
depressive-like phenotype without locomotor bias (Custódio et al., a habituation period of 1 min, the immobility time (in seconds) of the
2013). Also, we evaluated the influence of sex in brain inflammatory animals was evaluated for 5 min. Immobility was defined as the absence
alterations, namely myeloperoxidase (MPO) activity and IL-1β, and, to of targeted escape behavior, such as swimming, jumping, lifting, smell
our knowledge for the first time, in the oxidative stress parameters or diving (Porsolt et al., 1977). Any mouse seeming to have trouble
reduced glutathione (GSH) and lipid peroxidation in relevant brain keeping its head out of the water was removed from the cylinder and
areas for mood regulation: prefrontal cortex (PFC), hippocampus and excluded from the analysis. In this study, two experienced evaluators
hypothalamus. blinded to the treatment group independently assessed the mice beha-
vior.

2. Materials and methods 2.4.2. Sucrose preference test (SPT)


The test was performed as described previously (Mao et al., 2014).
2.1. Drugs Briefly 72 h before testing, rats were trained to adapt sucrose solution at
10% (w/v). Two sucrose solution bottles were placed in each cage, and
Lipopolysaccharide (LPS) from Escherichia coli, strain 055:B5 (Sigma 24 h later one of them was replaced by a bottle with water for 24 h.
e Aldrich Corp., St Louis, (USA) was used. The drugs were made up After adaptation, mice were deprived of food and water for 24 h. The
freshly for the study. All other chemicals used were of analytical grade. test section was conducted to 9:00 AM in which the rats were housed in

134
B.S.F. Mello et al. Journal of Neuroimmunology 320 (2018) 133–142

individual cages and had free access to two bottles containing 100 ml of 2.6. Statistical analyses
sucrose solution (10% w/v) and 100 ml of water, respectively. After 1 h,
the volumes of sucrose solution and water consumed were recorded and Data are present in scatter dot plot with lines representing
preferably sucrose consumption was calculated by the following for- mean ± S.E.M. (standard errors of the mean) and were compared by
mula: regular two-way ANOVA with Tukey's as post hoc test. The factors used
were “sex” (male and female) and “LPS treatment” (LPS and saline
Sucrose preference = (sucrose consumption ×100%) treatment). The significance level was set at P ≤ 0.05. The statistical
/(water consumption + sucrose consumption) program used was GraphPad Prism 6.0 Version for Windows, San
Diego, CA, USA.

2.4.3. Open field test (OFT)


The open field arena was made of acrylic (30 × 30 × 15 cm) with 3. Results
transparent walls and a black floor, divided into nine squares of equal
areas. The open field test was used to evaluate the locomotor and ex- 3.1. Male mice are more susceptible to despair-like behavior while both
ploratory activity of the animal during 5 min (Archer, 1973). The sexes present anhedonia without motor alterations 24 h post-LPS
parameters number of squares crossed by the animal (crossings
number), number of rearings (vertical locomotion), number of self- In the evaluation of despair-like behavior using the FST, two-way
cleaning grooming behavior, and total time in the center of arena were ANOVA revealed a significant interaction between the factors “sex” and
observed over 5 min and were registered in seconds. “LPS challenge” [F(1, 27) = 4.989, P = 0.0340], with a significant
main effect “sex” [F(1, 27) = 59.21, P < 0.0001] and “LPS challenge”
2.4.4. Elevated plus maze test (EPM) [F(1, 27) = 8.867, P = 0.0061]. Post hoc test revealed a significant
The elevated plus-maze consisted of two open (30 × 5 cm) and two increase in the immobility time in LPS-challenged male mice when
darkened, closed arm (30 × 5 × 15 cm) emanating from a common compared to male controls (P < 0.0001). No significant alterations
central platform (5 × 5 cm), to form a plus shape (Pellow and File, were observed in female animals exposed to LPS. We also observed
1986). The entire apparatus was raised 45 cm above the base, and the increased immobility time in male control animals in relation to female
test was performed under dim red light (60 W × 2). The test started by ones (P < 0.0001), being this sex-related increase in immobility time
placing a mouse on the central platform, facing an open arm. An ob- also observed in LPS-challenged males when compared to females
servation period of 5 min was used, during which the total arms entries, (P < 0.0001) (Fig. 1A).
and the amount of time spent by animals in the open and closed arms of
the maze was measured. These data were used to calculate the number
of total entries, % of open entries (i.e., open entries/total entries × 100)
and % of time in the open arms (time in open arms/total time of the
test × 100).

2.5. Neurochemical determinations

2.5.1. Analysis of myeloperoxidase (MPO) activity


Myeloperoxidase is a highly oxidative enzyme. The extracellular
activity of this enzyme gives an estimate of the oxidative stress in in-
flammatory conditions (Pulli et al., 2013). Peroxidase activity with
3,30,5,50-Tetramethylbenzidine (TMB) was measured as described
elsewhere (Suzuki et al., 1983). Absorbance was determined at 450 nm
in two time-points, 0 and 3 min, to estimate MPO activity (U MPO/min/
mg tissue).

2.5.2. Immunoassay for IL-1β


The brain areas were homogenized in 8 volumes of PBS buffer with
protease (EMD Biosciences) inhibitor and centrifuged (10.000 rpm,
5 min). The concentration of the cytokines in 50 ml samples was de-
termined by the immunoenzymatic assay ELISA (R&D systems,
Minneapolis, MN, USA) according to the manufacturer's protocol and
expressed in pg/g tissue.

2.5.3. Determination of reduced glutathione (GSH) levels


The levels of GSH were evaluated to estimate endogenous defenses
against oxidative stress. The method was based on Ellman's reagent
(DTNB) reaction with free thiol groups (Sedlak and Lindsay, 1968). The
reaction was read in the absorbance of 412 nm, and the product was
Fig. 1. Effect of LPS (0.5 mg/kg, i.p.) in the duration of immobility (in seconds)
expressed as ng of GSH/mg wet tissue. in the forced swimming test (A) and in the % of sucrose preference in the su-
crose consumption test (B). Male and female mice were injected with LPS
2.5.4. Measurement of lipid peroxidation (0.5 mg/kg, i.p) and, 24 h later, the behavioral tests were performed. Each dot
Lipid peroxides formation was analyzed by measuring the thio- represents individual values, n = 7–8/group, with horizontal and vertical lines
barbituric-acid reacting substances (TBARS) in the brain homogenates representing mean ± S.E.M., respectively. Data are representative of two in-
(Mihara and Uchiyama, 1978), as an index of reactive oxygen species dependent experiments and were analyzed by two-way ANOVA followed by
(ROS) production. Lipid peroxidation was assessed by the absorbance at Tukey's post hoc test. ***P < 0.001, ****P < 0.0001. Abbreviations:
532 nm and expressed as μmol of malonaldehyde (MDA)/mg of tissue. LPS = Lipopolysaccharide, Saline = vehicle.

135
B.S.F. Mello et al. Journal of Neuroimmunology 320 (2018) 133–142

Fig. 2. Effect of LPS (0.5 mg/kg, i.p.) in the number


of crossings (A), number of rearings (B), number of
groomings (C) and time in the center of arena (in
seconds) (D) in the open field test. Male and female
mice were injected with LPS (0.5 mg/kg, i.p) and,
24 h later, the behavioral tests were performed. Each
dot represents individual values, n = 7–8/group,
with horizontal and vertical lines representing
mean ± S.E.M., respectively. Data are re-
presentative of two independent experiments and
were analyzed by two-way ANOVA followed by
Tukey's post hoc test. *P < 0.05. Abbreviations:
LPS = Lipopolysaccharide, Saline = vehicle.

In the evaluation of anhedonia with the SPT (Fig. 1B), we detected a 3.3. Male and female mice present brain alterations in MPO activity, IL-1β
significant main effect of “LPS challenge” in the percent of sucrose levels and oxidative parameters 24 h post-LPS
consumption [F(1, 25) = 29.05, P < 0.0001], without significant in-
teraction between factors. Post hoc analysis revealed a significant de- Regarding MPO activity in the PFC (Fig. 4A), two-way ANOVA
crease in the percent of sucrose consumption in LPS-challenged male demonstrated a significant main effect of each factor alone ([F(1,
(P < 0.001) and female (P < 0.001) mice in relation to sex-matched 26) = 23.26, P < 0.0001], for “sex”; [F(1, 26) = 36.48, P < 0.0001],
controls. for “LPS challenge”), without significant interaction between factors.
The number of crossings in the OFT was used as a parameter of Post hoc comparisons revealed a significant increase in MPO activity in
horizontal locomotor activity. As shown in Fig. 2A, LPS immune chal- the PFC of both male and female subjects challenged with LPS com-
lenge did not induced significant alterations in the number of crossings pared to their sex-matched controls (P < 0.0001 for males and fe-
in both male and female animals when compared to their sex-matched males). Furthermore, male control animals presented higher MPO ac-
controls, as well no significant difference was noted when comparing tivity when compared to female control ones (P < 0.05). In the
each treatment group in relation to sex. In the exploratory behavior, we hippocampus (Fig. 4B), ANOVA analysis revealed a significant inter-
did not observe significant differences in the number of rearings post- action between factors [F(1, 24) = 8.064, P = 0.0091], with a sig-
LPS challenge in animals of both sexes (Fig. 2B). nificant main effect of each one alone: [F(1, 24) = 8.900, P = 0.0065],
Regarding grooming behavior and the time spent in the center of the for “sex”; [F(1, 24) = 8.523, P = 0.0075], for “LPS challenge”. In the
arena, both parameters related to anxiety-like behavior, we observed in post hoc comparisons, we evidenced a significant increase in MPO ac-
grooming behavior (Fig. 2C) a significant main effect of “LPS challenge” tivity in the hippocampus of female mice after LPS challenge
[F(1, 27) = 5.984, P = 0.0212] without significant interaction between (P < 0.01). We also observed higher MPO activity in the hippocampus
factors. In this behavioral parameter, we observed that LPS-challenged of male control mice when compared to female controls (P < 0.01). In
female mice presented increased grooming behavior in relation to fe- the hypothalamus ANOVA analysis showed no significant interaction
male controls. On the other hand, in the analysis of the time spent in the nor main effect of any of the factors analyzed (Fig. 4C).
center of the arena, two-way ANOVA demonstrated no significant al- The levels of IL-1β in the PFC (Fig. 4D) were significantly increased
terations (Fig. 2D). in both male (P < 0.05) and female (P < 0.01) mice challenged with
LPS in relation to sex-matched controls (two-way ANOVA: main effect
of “LPS challenge” [F(1, 24) = 24.11, P < 0.0001]. In the hippo-
3.2. Female mice seem to be more susceptible to anxiety-like behavior in the campus (Fig. 4E), two-way ANOVA revealed no interaction between
EPM test 24 h post-LPS factors, but a significant main effect of “sex” [F(1, 23) = 39.30,
P < 0.0001] and “LPS challenge” [F(1, 23) = 9.269, P = 0.0058]. Post
In the EPM test, we did not detect significant differences in the total hoc comparisons showed a significant increase in IL-1β levels in male
number of entries (Fig. 3A) as well as in the percent of time in the open controls when compared to female controls (P < 0.01) and in LPS-
arms (Fig. 3C). Nevertheless, in the percent of entries in the open arms, challenged males when compared to LPS-challenged females
a significant main effect of “LPS challenge” [F(1, 21) = 7.863, (P < 0.01). In the hypothalamus (Fig. 4F), ANOVA analysis demon-
P = 0.0106], without a significant interaction between factors was strated main effect of “sex” [F(1, 24) = 27.34, P < 0.0001] and “LPS
observed. In the post hoc test, a significant decrease in the percentage challenge” [F(1, 24) = 30.16, P < 0.0001], without significant inter-
of open arms entries was noted in LPS-challenged female mice when action between factors. In the post hoc comparisons, it was evidenced a
compared to female controls (P < 0.05) (Fig. 3B). significant increase in IL-1β levels in LPS-challenged mice of both sexes

136
B.S.F. Mello et al. Journal of Neuroimmunology 320 (2018) 133–142

when compared to their same sex controls (P < 0.0001, for males;
P < 0.001, for females) (Fig. 5B). In the hypothalamus, there was a
significant main effect of “LPS challenge” [F(1, 25) = 88.34,
P < 0.0001]. Post hoc comparisons demonstrated a significant de-
crease in GSH levels in the hypothalamus of animals of both sexes
challenged with LPS compared to sex-matched saline controls
(P < 0.0001, for males; P < 0.0001, for females) (Fig. 5C).
Lipid peroxidation represents the damage of lipid membranes
caused by oxidative imbalance (Ramos-Loyo et al., 2013). In this con-
text, we observed no lipid peroxidation in the PFC of LPS-challenged
mice when compared to controls (Fig. 5D). In the hippocampus, two-
way ANOVA analysis demonstrated a significant interaction between
“sex” and “LPS challenge” [F(1, 23) = 6.131, P = 0.0211]. In the post
hoc test, we evidenced a significant increase in MDA levels in the
hippocampus of LPS-challenged male mice in relation to their same sex
controls (P < 0.05) (Fig. 5E). In the hypothalamus (Fig. 5F), a sig-
nificant increase in MDA levels was evidenced in LPS-challenged female
mice in relation to their same sex controls (P < 0.05) (two-way
ANOVA: main effect of “LPS challenge” [F(1, 21) = 8.211,
P = 0.0093].

4. Discussion

In this study, we demonstrated that at the time-point of 24 h post


systemic challenge with LPS, male and female mice present a distinctive
spectrum of behavioral accompanied by proinflammatory and oxidative
alterations. Of note, male animals presented behavioral despair and
anhedonia, while females presented anhedonia and anxiety-like al-
terations. Both sexes presented brain proinflammatory and pro-oxida-
tive alterations. The main strengths of the present study were de-
termining sex influences in anxiety-like behavior as well as
proinflammatory and pro-oxidative alterations in brain areas relevant
to mood regulation in mice submitted to an inflammatory (LPS) model
of depressive-like behavior.
Men and women differ in the occurrence and presentation of several
psychiatric disorders. This is specifically relevant for major depressive
disorder (MDD), which is twice as common in females than in males.
Interestingly, prior to puberty, subjects of both genders are equally
affected by depression (Marcus et al., 2005). Many biological, psycho-
social, and sociological theories attempted to explain this dramatic in-
Fig. 3. Effect of LPS (0.5 mg/kg, i.p.) in the number of total entries (A), in the %
crease in the prevalence of depression among women, but none of them
of open entries (B) and in the % of time in open arms (C) in the elevated plus
is fully satisfactory. In this context, animal models despite their lim-
maze test. Male and female mice were injected with LPS (0.5 mg/kg, i.p.) and,
24 h later, the behavioral tests were performed. Each dot represents individual itations, represent a useful tool for the investigation of sex differences in
values, n = 7–8/group, with horizontal and vertical lines representing the neurobiology of depression and antidepressant response (Dalla
mean ± S.E.M., respectively. Data are representative of two independent ex- et al., 2010).
periments and were analyzed by two-way ANOVA followed by Tukey's post hoc In line with the limited knowledge regarding sex influences in de-
test. *P < 0.05. Abbreviations: LPS = Lipopolysaccharide, Saline = vehicle. pression, only few studies were conducted aiming to determine sex
influences in the behavioral and neurochemical alterations induced by
in relation to sex-matched controls (P < 0.001 for males and P < 0.05 LPS challenge. In this context, Pitychoutis et al., 2009 reported that
for females). Also, male control mice presented increased levels of IL-1β male and female rats presented different vulnerability to sickness be-
in relation to female ones (P < 0.05). havior at the time-point of 2 h post-LPS administration. In their study,
Regarding the levels of the main endogenous antioxidant, GSH, we LPS-challenged female rats showed a better coping strategy, evidenced
observed in the PFC, a significant main effect of both factors alone: [F by the increased time of swimming, while males presented a more ro-
(1, 26) = 10.44, P = 0.0033], for “sex” and [F(1, 26) = 26.93, bust locomotor suppression. The other behavioral parameters eval-
P < 0.0001], for “LPS challenge”, without significant interaction. In uated, such as sucrose consumption, social exploration and food intake
the post hoc test, we identified a significant decrease in GSH contents in were equally affected in animals of both sexes (Pitychoutis et al., 2009).
the PFC of both male (P < 0.05) and female (P < 0.01) mice chal- Additionally, a recent study focusing on the sex-specific behavioral
lenged with LPS when compared to their sex-matched controls. Female effects of LPS at the time-points of 6 and 24 h post LPS (Sens et al.,
control mice presented higher levels of GSH when compared to control 2017) found that the immobility duration was equally affected in ani-
male mice (P < 0.05) (Fig. 5A). In the hippocampus, two-way ANOVA mals of both sexes. The same results were obtained when mice were
revealed a significant main effect of “sex” [F(1, 27) = 1.626, tested for anhedonia by sucrose consumption. Notably, they found that
P = 0.2131] and “LPS challenge” [F(1, 27) = 54.69, P < 0.0001], and the grooming behavior in the splash test, an index of motivation and
a significant interaction between factors [F(1, 27) = 5.290, self-care behavior, was persistently impaired in female mice, while
P = 0.0294]. In post hoc test, we detected a significant decrease in GSH anorexic behavior showed to be more pronounced in male counterparts
levels in the hippocampus of animals of both sexes challenged with LPS (Sens et al., 2017).
In our experimental conditions, male and female mice showed a

137
B.S.F. Mello et al. Journal of Neuroimmunology 320 (2018) 133–142

Fig. 4. Effect of LPS (0.5 mg/kg, i.p.) in MPO activity


and IL-1β levels in the prefrontal cortex (A, D), hip-
pocampus (B, E) and hypothalamus (C, F). Male and
female mice were injected with LPS (0.5 mg/kg, i.p.)
and, 24 h later, the brain areas were collected for
neurochemical analysis. Each dot represents individual
values, n = 7–8/group, with horizontal and vertical
lines representing mean ± S.E.M., respectively. Data
are representative of two independent experiments
and were analyzed by two-way ANOVA followed by
Tukey's post hoc test. *P < 0.05, **P < 0.01,
***P < 0.001, ****P < 0.0001. Abbrevia-
tions: LPS = Lipopolysaccharide, Saline = vehicle,
IL = interleukin, MPO = myeloperoxidase.

distinct behavioral pattern in the forced swimming test 24 h post LPS effects of LPS in male rats in several doses and in several behavioral
challenge. Our results pointed to an increased vulnerability of LPS tests (elevated plus maze, black-white box, open field, elevated T maze,
challenged male animals to the development of despair behavior in the emission of ultrasonic vocalizations) 3–4 h after administration. They
FST. Of note, we also observed that male control animals presented found that LPS induced a dose-dependent increase in anxiety-like be-
increased immobility time in relation to female ones. Taken together, haviors, forming an inverted U curve which peaks in 200 μg/kg dose
these results obtained in male animals corroborates previous findings (Bassi et al., 2012). Several other studies demonstrated the anxiogenic-
showing the ability of female animals in the development of coping like effects of LPS in the sickness spectrum of alterations (few hours
strategies in the stressful FST (Pitychoutis et al., 2009). It is important after injection) as well as in depressive-like ones (24 h after) (Li et al.,
to highlight that this previous study was conducted in Sprague–Dawley 2017; Salazar et al., 2012; Savignac et al., 2016). We also observed, in a
(SD) rats with LPS serotype 026:B6, i.e., different from the serotype previous study, that LPS 0.5 mg/kg caused anxiety-like behavior in
used in the present study, and that the enhanced coping abilities ob- male animals at the time-point of 1.5 h post LPS, while at the time-point
served in females were detected at the time-point of 2 h post LPS of 24 h no alteration was observed (Custódio et al., 2013). Furthermore,
(Pitychoutis et al., 2009). This sex influence in the immobility time in the intravenous administration of Salmonella abortus equi endotoxin to
the FST observed in our results was not observed at the time-point of 20 healthy male volunteers caused a transient significant increase in the
24 h post LPS serotype 026:B6 (Sens et al., 2017), revealing, thus, that levels of anxiety and depressive mood at the time-points of 1, 3 and 9 h
the effects of LPS are not only time, but also specie and serotype de- post administration. To the best of our knowledge, no previous study
pendent (Felgner et al., 2017). evaluated sex influences in anxious-like behavior in LPS-challenged
In addition, we observed that anhedonia assessed by SPT was animals (Reichenberg et al., 2001).
equally established in both male and female animals. As expected, 24 h Our results showed that LPS-challenged female mice presented with
post-LPS administration, no locomotor impairment was observed in a more robust anxiety-like phenotype. This evidence came from two
male and female mice. This result is in line with previous ones showing different behavioral tasks, i.e., increased grooming behavior in the OFT,
that both sexes are vulnerable to anhedonia (Sens et al., 2017). and reduced percentage of open entries in the EPM. In general, anxiety
A well-established concept is that anxiety and inflammation walk disorders are much more prevalent in women than in men (McLean
together. In this regard, Bassi et al., 2012. described the anxiogenic-like et al., 2011). Indeed, in animal studies females usually demonstrate less

138
B.S.F. Mello et al. Journal of Neuroimmunology 320 (2018) 133–142

Fig. 5. Effect of LPS (0.5 mg/kg, i.p.) in reduced


glutathione (GSH) levels and lipid peroxidation in
the prefrontal cortex (A, D), hippocampus (B, E) and
hypothalamus (C, F). Male and female mice were
injected with LPS (0.5 mg/kg, i.p.) and, 24 h later,
the brain areas were collected for neurochemical
analysis. Each dot represents individual values,
n = 7–8/group, with horizontal and vertical lines
representing mean ± S.E.M., respectively. Data are
representative of two independent experiments and
were analyzed by two-way ANOVA followed by
Tukey's post hoc test. *P < 0.05,
**P < 0.01, ****P < 0.0001. Abbreviat-
ions: LPS = lipopolysaccharide, Saline = vehicle,
MDA = malonaldehyde.

anxiety-like behavior in basal situations (Rodgers and Cole, 1993; IL-1β acts as a pleotropic proinflammatory cytokine, inducing its own
Võikar et al., 2001). However, when exposed to stressful-aversive sti- synthesis and the synthesis of other cytokines that potentiate its effect,
muli, remarkable anxiety-like alterations are observed. such as IL-18 and IL-6 (Weber et al., 2010). Mitochondria ROS is both a
It is worth mentioning that adrenocorticotropic hormone (ACTH) and major trigger and product of NLRP3 inflammasome activation (Han
corticosterone responses are greater in female subjects than in male ones et al., 2015).
during the exposure to a variety of acute stressors (Doremus-Fitzwater Oxidative stress and inflammation are closely related pathophysio-
et al., 2009; Iwasaki-Sekino et al., 2009). More recently, Vieira et al., logical processes. Inflammatory cells release reactive species at the site
2017, confirmed this proposition by demonstrating that female rodents of inflammation leading to exaggerated oxidative stress. On the other
showed increased anxiety-like behavior and plasma corticosterone levels hand, a number of reactive species can initiate intracellular signaling
when exposed to chronic unpredictable stress and chronic homotypic cascades that enhances proinflammatory gene expression (Biswas,
stress (repeated restraint stress) (Vieira et al., 2017). In this context, our 2016). In line with this evidence, MPO is an important enzyme of innate
results revealing enhanced vulnerability of females to anxiety-like phe- immune system that is activated in phagocytic cells during in-
notype in LPS inflammatory model of depressive-like behavior are in flammatory conditions (Arnhold and Flemmig, 2010). MPO generates
accordance with the previous findings of female susceptibility to anxiety- strong oxidant radicals, in special, hypochlorous acid (HOCl), that can
like behavior against environmental stressful situations. It is important to covalently modify lipids and/or proteins causing local damage and
mention that there is a discussion in the literature about the possible amplification of inflammatory response (Arnhold and Flemmig, 2010;
relation of inflammatory models of depression with one specific subtype Pattison et al., 2012).
of depression, namely atypical depression (Remus and Dantzer, 2016). In Relevant studies reported increased MPO expression and activity in
fact, atypical depression is the most common form of depression, four serum samples of depressive patients. For example, Vaccarino et al.,
times more prevalent in women presenting high comorbidity with an- 2008, demonstrated in 178 monozygotic and dizygotic twins, a strong
xiety disorder (Singh and Williams, 2006). association between serum MPO levels and depression scores in stan-
It is consistently recognized that LPS activates toll-like receptor 4 dard scales (Vaccarino et al., 2008). Additionally, it was found that
(TLR4). The TLR4 pathway is coupled to the activation inflammasome MPO mRNA and protein levels are markedly enhanced in peripheral
signaling, in special nucleotide-binding domain leucine-rich repeat blood cells of patients with recurrent depression, and present a positive
(NLRP) 3 (Scholz and Eder, 2017). NLRP3 mediates, in large part, the correlation with the impaired performance in several cognitive tasks
processing and maturation of IL-1β (Cullen et al., 2015; Jo et al., 2016). (Talarowska et al., 2015).

139
B.S.F. Mello et al. Journal of Neuroimmunology 320 (2018) 133–142

In the present study, we observed a brain region-dependent increase our study gives the first evidence of sex influences in brain oxidative
in MPO activity in animals submitted to LPS immune challenge. Indeed, status in LPS-induced depressive-like model in mice.
MPO activity was increased in the PFC of male and female mice, while It is an important to mention that the neural circuitries related to
in the hippocampus this increase was observed only in female animals. anxiety situations or reward-pleasure stimuli involves basolateral
Animal submitted to chronic stress presented higher MPO activity in amygdala and bed nucleus of the stria terminalis (BNST) (Duval et al.,
selected brain areas, namely hippocampus and cortex, which was ac- 2015; Russo and Nestler, 2013). BNST recruits projections from the
companied by additional proinflammatory alterations such as micro- ventral hippocampus and hypothalamic nuclei, promoting the pitui-
glial activation and NO release (Kasımay Çakır et al., 2017; Lisowski tary-adrenal response (Duval et al., 2015). In reward-promoting situa-
et al., 2013). Currently, as far as we know there is no available data tions, orbitofrontal cortex (OFT) projects reward information to ante-
about brain MPO activity in rodents submitted to LPS-based depressive- rior cingulate cortex, that, in turn, send projections to anterior
like model. Therefore, our results demonstrated not only the effects of ventromedial PFC, dorsolateral PFC and ventral hippocampus (Russo
LPS as an activator of MPO activity in the brain, as well as advocate for and Nestler, 2013). These nuclei are key regulators of decision-making
a possible sex related brain region-dependent response. based on reward value and the reinforcement for future actions
Still regarding inflammatory alterations triggered by LPS challenge, (Gorwood, 2008).
in the present study we assessed the levels of IL-1β in relevant brain Taking this above mecioned cirtuitry into consideration, it is pos-
areas related to mood regulation. We found 24 h post LPS increased sible to suggest, in our experimental condition, that the presence of a
levels of IL-1β in the PFC and hypothalamus of male and female mice. pronounced inflammatory-oxidative damage in hippocampal-hypotha-
We also observed higher levels of IL-1β in the hippocampus of LPS- lamic areas of female mice is in accordance with the emergence of an
challenged male mice when compared to female ones, although this increased anxiety-like phenotype, since these neurocircuits are involved
increase was not significant in relation to male control mice. It is im- in the regulation of anxiety and stress response (Duval et al., 2015). On
portant to highlight that despite being classified as a cytokine, IL-1β is the other hand, in male animals, the predominance of oxidative damage
also a neuroactive molecule that interferes with several mechanisms in the PFC and hippocampal regions, which are key areas for the reg-
related to depression: i) decreases hippocampi cell proliferation, being ulation of brain reward circuitry (Russo and Nestler, 2013), can be a
an antineurogenic mediator (Koo and Duman, 2008) and ii) is a potent plausible factor for the development of a robust despair-like/anhedonia
stimulant of the serotonin metabolism (Ramamoorthy et al., 1995). behavior in these animals. Finally, the hippocampus, as a region in-
Considering sex influences in LPS-induced inflammatory alterations, trinsically involved in the regulation of both circuits, was impaired in
some authors reported an increased inflammatory response in females animals of both sexes. On the other hand, in our experimental condition
24 h post nasal LPS administration (Badalà et al., 2008), while others it seems that hippocampus was more affected in males than in females.
showed increased sickness-behavior and serum cytokine levels in male This study presents some limitations. Firstly, we did not assess the
mice 10 h post intraperitoneal LPS injection (Cai et al., 2016). influence of estrous cycle changes in the behavioral and neurochemical
Regarding oxidative alterations in depression, considerable evi- effects of LPS in females. This is an interesting point for future research,
dence reported diminished levels of GSH and/or impaired activity of since ovarian hormones, in special estrogens, are known to modulate
the enzymes involved in GSH synthesis and restoration in the serum and not only female behavior, as well as the activity of several cellular
post-mortem brain of depressed patients (Gawryluk et al., 2011; Maes antioxidant systems (Bellanti et al., 2013). Additionally, we decided to
et al., 2011b). Among the endogenous antioxidant systems, GSH is the measure the parameters of oxidative damage: MPO, GSH, and lipid
most abundant thiol antioxidant present in mammalian cells protecting peroxidation, and not others, for example, catalase and superoxide
cells against ROS generated by the mitochondrial respiratory chain dismutase enzymes, that despite being relevant for redox status, present
(Dringen and Hirrlinger, 2003). Accordingly, several preclinical studies controversial evidence in depressive patients (Maes et al., 2011a).
showed that rodents submitted to environmental or pharmacological In conclusion, this study provides additional evidence of a sex-
models of stress presented a marked reduction in total GSH levels and specific pattern of behavior in adult mice submitted to LPS-induced
glutathione peroxidase activity (GPX) in brain areas related to mood inflammatory model of depression. Notably, in our results, male ani-
regulation (Silva et al., 2016; Tao et al., 2016; Todorović and Filipović, mals presented a greater vulnerability to despair-like behavior, while
2017). females developed anxiety-related alterations. Both sexes were equally
Lipid peroxidation is an autoxidation process promoted by radical vulnerable to LPS-induced anhedonia. The behavioral alterations in
species against phospholipids and unsaturated fatty acids of the cellular females were followed by increased oxidative changes in all brain areas
membranes. This process severely hampers the membrane functions, investigated here, but especially in the hypothalamus, a brain area as-
increasing membrane rigidity and allowing the leakage of calcium ions sociated to stress response and anxiety. Male animals presented a pre-
(Fuchs et al., 2014). Malondialdehyde (MDA) is a byproduct of poly- ponderance for the involvement of oxidative damage in the PFC and
unsaturated fatty acid and arachidonic acid peroxidation consistently hippocampal regions, which are key areas for the regulation of reward.
employed as a measure of lipid peroxidation (Gaweł et al., 2004). There Therefore, this study advances the knowledge about sex influence in
are numerous studies supporting the involvement of lipid peroxidation inflammatory-based model of depression by presenting evidences of
in depression, and a lot of them demonstrating increased MDA levels in sex-specific pattern of behavioral alterations and, for the first time, of
biological samples of depressive patients [for a detailed Review, see brain oxidative changes.
(Maes et al., 2011a)].
In our study, we found that male and female mice exposed to LPS
immune challenge equally presented impaired GSH levels in all brain Disclosure
areas investigated, and a sex- and brain region-dependent increase in
lipid peroxidation. Notably, male mice showed increased levels of MDA The authors declare no conflict of interests.
in the hippocampus, while females showed increased levels of MDA
only in the hypothalamus. Despite previous studies have demonstrated
important oxidative damage (lipid peroxidation, increased nitrite and Acknowledgements
PGE2 levels) and impairment in antioxidant systems (reduced GSH le-
vels) in the brain of mice submitted to LPS challenge (Custódio et al., The authors thank the Brazilian Institutions CAPES, FUNCAP and
2013; Mello et al., 2013; Sayd et al., 2014), all of them investigated CNPq for the financial support. The authors are grateful for the tech-
oxidative changes only in male rodents. Therefore, as far as we know, nical assistance of Maria Vilani Bastos.

140
B.S.F. Mello et al. Journal of Neuroimmunology 320 (2018) 133–142

References psychological. Psychoneuroendocrinology 34, 226–237. http://dx.doi.org/10.1016/


j.psyneuen.2008.09.003.
Jo, E.-K., Kim, J.K., Shin, D.-M., Sasakawa, C., 2016. Molecular mechanisms regulating
Archer, J., 1973. Tests for emotionality in rats and mice: a review. Anim. Behav. 21, NLRP3 inflammasome activation. Cell. Mol. Immunol. 13, 148–159. http://dx.doi.
205–235. org/10.1038/cmi.2015.95.
Arnhold, J., Flemmig, J., 2010. Human myeloperoxidase in innate and acquired im- Kasımay Çakır, Ö., Ellek, N., Salehin, N., Hamamcı, R., Keleş, H., Kayalı, D.G., Akakın, D.,
munity. Arch. Biochem. Biophys. 500, 92–106. http://dx.doi.org/10.1016/j.abb. Yüksel, M., Özbeyli, D., 2017. Protective effect of low dose caffeine on psychological
2010.04.008. stress and cognitive function. Physiol. Behav. 168, 1–10. http://dx.doi.org/10.1016/
Avitsur, R., Yirmiya, R., 1999. The immunobiology of sexual behavior: gender differences j.physbeh.2016.10.010.
in the suppression of sexual activity during illness. Pharmacol. Biochem. Behav. 64, Köhler, C.A., Freitas, T.H., Maes, M., de Andrade, N.Q., Liu, C.S., Fernandes, B.S., Stubbs,
787–796. B., Solmi, M., Veronese, N., Herrmann, N., Raison, C.L., Miller, B.J., Lanctôt, K.L.,
Badalà, F., Nouri-mahdavi, K., Raoof, D.A., 2008. Intranasal immune challenge induces Carvalho, A.F., 2017. Peripheral cytokine and chemokine alterations in depression: a
sex-dependent depressive-like behavior and cytokine expression in the brain leo- meta-analysis of 82 studies. Acta Psychiatr. Scand. 135, 373–387. http://dx.doi.org/
nardo. Neuropsychopharmacology 144, 724–732. http://dx.doi.org/10.1038/jid. 10.1111/acps.12698.
2014.371. Koo, J.W., Duman, R.S., 2008. IL-1 is an essential mediator of the antineurogenic and
Bassi, G.S., Kanashiro, A., Santin, F.M., de Souza, G.E.P., Nobre, M.J., Coimbra, N.C., anhedonic effects of stress. Proc. Natl. Acad. Sci. 105, 751–756. http://dx.doi.org/10.
2012. Lipopolysaccharide-induced sickness behaviour evaluated in different models 1073/pnas.0708092105.
of anxiety and innate fear in rats. Basic Clin. Pharmacol. Toxicol. 110, 359–369. Li, C., Li, M., Yu, H., Shen, X., Wang, J., Sun, X., Wang, Q., Wang, C., 2017. Neuropeptide
http://dx.doi.org/10.1111/j.1742-7843.2011.00824.x. VGF C-terminal peptide TLQP-62 alleviates lipopolysaccharide-induced memory
Biswas, S.K., 2016. Does the interdependence between oxidative stress and inflammation deficits and anxiety-like and depression-like behaviors in mice: the role of BDNF/
explain the antioxidant paradox? Oxidative Med. Cell. Longev. 2016, 17–19. http:// TrkB signaling. ACS Chem. Neurosci. 8, 2005–2018. (acschemneuro.7b00154).
dx.doi.org/10.1155/2016/5698931. https://doi.org/10.1021/acschemneuro.7b00154.
Bellanti, F., Matteo, M., Rollo, T., De Rosario, F., Greco, P., Vendemiale, G., Serviddio, G., Lisowski, P., Wieczorek, M., Goscik, J., Juszczak, G.R., Stankiewicz, A.M., Zwierzchowski,
2013. Sex hormones modulate circulating antioxidant enzymes: impact of estrogen L., Swiergiel, A.H., 2013. Effects of chronic stress on prefrontal cortex transcriptome
therapy. Redox Biol. 1, 340–346. http://dx.doi.org/10.1016/j.redox.2013.05.003. in mice displaying different genetic backgrounds. J. Mol. Neurosci. 50, 33–57. http://
Cai, K.C., van Mil, S., Murray, E., Mallet, J.-F., Matar, C., Ismail, N., 2016. Age and sex dx.doi.org/10.1007/s12031-012-9850-1.
differences in immune response following LPS treatment in mice. Brain Behav. Lucca, G., Comim, C.M., Valvassori, S.S., Réus, G.Z., Vuolo, F., Petronilho, F., Dal-Pizzol,
Immun. 58, 327–337. http://dx.doi.org/10.1016/j.bbi.2016.08.002. F., Gavioli, E.C., Quevedo, J., 2009. Effects of chronic mild stress on the oxidative
Cullen, S.P., Kearney, C.J., Clancy, D.M., Martin, S.J., 2015. Diverse activators of the parameters in the rat brain. Neurochem. Int. 54, 358–362. http://dx.doi.org/10.
NLRP3 inflammasome promote IL-1beta secretion by triggering necrosis. Cell Rep. 1016/j.neuint.2009.01.001.
11, 1535–1548. http://dx.doi.org/10.1016/j.celrep.2015.05.003. Maes, M., Galecki, P., Chang, Y.S., Berk, M., 2011a. A review on the oxidative and ni-
Custódio, C.S., Mello, B.S.F., Cordeiro, R.C., de Araújo, F.Y.R., Chaves, J.H., Vasconcelos, trosative stress (O&NS) pathways in major depression and their possible contribution
S.M.M., Nobre Júnior, H.V., de Sousa, F.C.F., Vale, M.L., Carvalho, A.F., Macêdo, to the (neuro)degenerative processes in that illness. Prog. Neuro-Psychopharmacol.
D.S., 2013. Time course of the effects of lipopolysaccharide on prepulse inhibition Biol. Psychiatry 35, 676–692. http://dx.doi.org/10.1016/j.pnpbp.2010.05.004.
and brain nitrite content in mice. Eur. J. Pharmacol. 713, 31–38. http://dx.doi.org/ Maes, M., Mihaylova, I., Kubera, M., Uytterhoeven, M., Vrydags, N., Bosmans, E., 2011b.
10.1016/j.ejphar.2013.04.040. Lower whole blood glutathione peroxidase (GPX) activity in depression, but not in
Dalla, C., Pitychoutis, P.M., Kokras, N., Papadopoulou-Daifoti, Z., 2010. Sex differences in myalgic encephalomyelitis/chronic fatigue syndrome: another pathway that may be
animal models of depression and antidepressant response. Basic Clin. Pharmacol. associated with coronary artery disease and neuroprogression in depression. Neuro
Toxicol. 106, 226–233. http://dx.doi.org/10.1111/j.1742-7843.2009.00516.x. Endocrinol. Lett. 32, 133–140.
Dantzer, R., O'Connor, J.C., Freund, G.G., Johnson, R.W., Kelley, K.W., 2008. From in- Maes, M., Berk, M., Goehler, L., Song, C., Anderson, G., Ga, P., 2012. Depression and
flammation to sickness and depression: when the immune system subjugates the Sickness Behavior Are Janus-Faced Responses to Shared Inflammatory Pathways
brain. Nat. Rev. Neurosci. 9, 46–56. http://dx.doi.org/10.1038/nrn2297. 1–19. http://dx.doi.org/10.1186/1741-7015-10-66.
Doremus-Fitzwater, T.L., Varlinskaya, E.I., Spear, L.P., 2009. Social and non-social an- Mao, Q.-Q., Huang, Z., Zhong, X.-M., Xian, Y.-F., Ip, S.-P., 2014. Brain-derived neuro-
xiety in adolescent and adult rats after repeated restraint. Physiol. Behav. 97, trophic factor signalling mediates the antidepressant-like effect of piperine in
484–494. http://dx.doi.org/10.1016/j.physbeh.2009.03.025. chronically stressed mice. Behav. Brain Res. 261, 140–145. http://dx.doi.org/10.
Dringen, R., Hirrlinger, J., 2003. Glutathione pathways in the brain. Biol. Chem. 384, 1016/j.bbr.2013.12.020.
505–516. http://dx.doi.org/10.1515/BC.2003.059. Marcus, S.M., Young, E.A., Kerber, K.B., Kornstein, S., Farabaugh, A.H., Mitchell, J.,
Duval, E.R., Javanbakht, A., Liberzon, I., 2015. Neural circuits in anxiety and stress Wisniewski, S.R., Balasubramani, G.K., Trivedi, M.H., Rush, A.J., 2005. Gender dif-
disorders: a focused review. Ther. Clin. Risk Manag. 11, 115–126. http://dx.doi.org/ ferences in depression: findings from the STAR*D study. J. Affect. Disord. 87,
10.2147/TCRM.S48528. 141–150. http://dx.doi.org/10.1016/j.jad.2004.09.008.
Farooq, R., Asghar, K., Kanwal, S., Zulqernain, A., 2016. Role of inflammatory cytokines Massafra, C., Gioia, D., De Felice, C., Muscettola, M., Longini, M., Buonocore, G., 2002.
in depression: focus on interleukin-1β (review). Biomed. Rep. http://dx.doi.org/10. Gender-related differences in erythrocyte glutathione peroxidase activity in healthy
3892/br.2016.807. subjects. Clin. Endocrinol. 57, 663–667.
Felgner, J., Jain, A., Nakajima, R., Liang, L., Jasinskas, A., Gotuzzo, E., Vinetz, J.M., McLean, C.P., Asnaani, A., Litz, B.T., Hofmann, S.G., 2011. Gender differences in anxiety
Miyajima, F., Pirmohamed, M., Hassan-Hanga, F., Umoru, D., Jibir, B.W., Gambo, S., disorders: prevalence, course of illness, comorbidity and burden of illness. J.
Olateju, K., Felgner, P.L., Obaro, S., Davies, D.H., 2017. Development of ELISAs for Psychiatr. Res. 45, 1027–1035. http://dx.doi.org/10.1016/j.jpsychires.2011.03.006.
diagnosis of acute typhoid fever in Nigerian children. PLoS Negl. Trop. Dis. 11. Merali, Z., Brennan, K., Brau, P., Anisman, H., 2003. Dissociating anorexia and anhedonia
http://dx.doi.org/10.1371/journal.pntd.0005679. elicited by interleukin-1beta: antidepressant and gender effects on responding for
Frenois, F., Moreau, M., O'Connor, J., Lawson, M., Micon, C., Lestage, J., Kelley, K.W., "free chow" and "earned" sucrose intake. Psychopharmacology 165, 413–418. http://
Dantzer, R., Castanon, N., 2007. Lipopolysaccharide induces delayed FosB/DeltaFosB dx.doi.org/10.1007/s00213-002-1273-1.
immunostaining within the mouse extended amygdala, hippocampus and hypotha- Mihara, M., Uchiyama, M., 1978. Determination of malonaldehyde precursor in tissues by
lamus, that parallel the expression of depressive-like behavior. thiobarbituric acid test. Anal. Biochem. 86, 271–278.
Psychoneuroendocrinology 32, 516–531. http://dx.doi.org/10.1016/j.psyneuen. Miller, A.H., Maletic, V., Raison, C.L., 2009. Inflammation and its discontents: the role of
2007.03.005. cytokines in the pathophysiology of major depression. Biol. Psychiatry 65, 732–741.
Fuchs, P., Perez-Pinzon, M.A., Dave, K.R., 2014. Chapter 2 – Cerebral ischemia in dia- http://dx.doi.org/10.1016/j.biopsych.2008.11.029.
betics and oxidative stress. In: Diabetes: Oxidative Stress and Dietary Antioxidants, Mello, B.S.F., Monte, A.S., McIntyre, R.S., Soczynska, J.K., Custódio, C.S., Cordeiro, R.C.,
pp. 15–23. http://dx.doi.org/10.1016/B978-0-12-405885-9.00002-4. Chaves, J.H., Vasconcelos, S.M.M., Nobre, H.V., Florenço de Sousa, F.C., Hyphantis,
Gaweł, S., Wardas, M., Niedworok, E., Wardas, P., 2004. Malondialdehyde (MDA) as a T.N., Carvalho, A.F., Macêdo, D.S., 2013. Effects of doxycycline on depressive-like
lipid peroxidation marker. Wiad. Lek. 57, 453–455. behavior in mice after lipopolysaccharide (LPS) administration. J. Psychiatr. Res. 47,
Gawryluk, J.W., Wang, J.-F., Andreazza, A.C., Shao, L., Young, L.T., 2011. Decreased 1521–1529. http://dx.doi.org/10.1016/j.jpsychires.2013.06.008.
levels of glutathione, the major brain antioxidant, in post-mortem prefrontal cortex NIH, N.R.C. (US) C. for the U. of the G. for the C. and U. of L, 2011. Guide for the Care and
from patients with psychiatric disorders. Int. J. Neuropsychopharmacol. 14, 123–130. Use of Laboratory Animals. National Academies Press, Washington, D.C.. http://dx.
http://dx.doi.org/10.1017/S1461145710000805. doi.org/10.17226/12910.
Gorwood, P., 2008. Neurobiological mechanisms of anhedonia. Dialogues Clin. Neurosci. Pattison, D.I., Davies, M.J., Hawkins, C.L., 2012. Reactions and reactivity of myeloper-
10, 291–299. oxidase-derived oxidants: differential biological effects of hypochlorous and hy-
Greenberg, P.E., Fournier, A.-A., Sisitsky, T., Pike, C.T., Kessler, R.C., 2015. The economic pothiocyanous acids. Free Radic. Res. 46, 975–995. http://dx.doi.org/10.3109/
burden of adults with major depressive disorder in the United States (2005 and 10715762.2012.667566.
2010). J. Clin. Psychiatry 76, 155–162. http://dx.doi.org/10.4088/JCP.14m09298. Pitychoutis, P.M., Nakamura, K., Tsonis, P.A., Papadopoulou-Daifoti, Z., 2009.
Han, S., Cai, W., Yang, X., Jia, Y., Zheng, Z., Wang, H., Li, J., Li, Y., Gao, J., Fan, L., Hu, D., Neurochemical and behavioral alterations in an inflammatory model of depression:
2015. ROS-mediated NLRP3 inflammasome activity is essential for burn-induced sex differences exposed. Neuroscience 159, 1216–1232. http://dx.doi.org/10.1016/j.
acute lung injury. Mediat. Inflamm. 2015. http://dx.doi.org/10.1155/2015/720457. neuroscience.2009.01.072.
Iwasaki-Sekino, A., Mano-Otagiri, A., Ohata, H., Yamauchi, N., Shibasaki, T., 2009. Pellow, S., File, S.E., 1986. Anxiolytic and anxiogenic drug effects on exploratory activity
Gender differences in corticotropin and corticosterone secretion and corticotropin- in an elevated plus-maze: a novel test of anxiety in the rat. Pharmacol. Biochem.
releasing factor mRNA expression in the paraventricular nucleus of the hypothalamus Behav. 24, 525–529.
and the central nucleus of the amygdala in response to footshock stress or

141
B.S.F. Mello et al. Journal of Neuroimmunology 320 (2018) 133–142

Porsolt, R.D., Le Pichon, M., Jalfre, M., 1977. Depression: a new animal model sensitive to induces sex-dependent behavioural and serotonergic neurochemical signatures in
antidepressant treatments. Nature 266, 730–732. mice. Pharmacol. Biochem. Behav. 153, 168–181. http://dx.doi.org/10.1016/j.pbb.
Pulli, B., Ali, M., Forghani, R., Schob, S., Hsieh, K.L.C., Wojtkiewicz, G., Linnoila, J.J., 2016.12.016.
Chen, J.W., 2013. Measuring myeloperoxidase activity in biological samples. PLoS Singh, T., Williams, K., 2006. Atypical depression. Psychiatry (Edgmont). 3, 33–39.
One 8, e67976. http://dx.doi.org/10.1371/journal.pone.0067976. Silva, M.C.C., de Sousa, C.N.S., Gomes, P.X.L., de Oliveira, G.V., Araújo, F.Y.R., Ximenes,
Ramamoorthy, S., Ramamoorthy, J.D., Prasad, P.D., Bhat, G.K., Mahesh, V.B., Leibach, N.C., da Silva, J.C., Vasconcelos, G.S., Leal, L.K.A.M., Macêdo, D., Vasconcelos,
F.H., Ganapathy, V., 1995. Regulation of the human serotonin transporter by inter- S.M.M., 2016. Evidence for protective effect of lipoic acid and desvenlafaxine on
leukin-1 beta. Biochem. Biophys. Res. Commun. http://dx.doi.org/10.1006/bbrc. oxidative stress in a model depression in mice. Prog. Neuro-Psychopharmacol. Biol.
1995.2659. Psychiatry 64, 142–148. http://dx.doi.org/10.1016/j.pnpbp.2015.08.002.
Ramos-Loyo, J., Medina-Hernández, V., Estarrón-Espinosa, M., Canales-Aguirre, A., Suarez, E.C., Lewis, J.G., Krishnan, R.R., Young, K.H., 2004. Enhanced expression of
Gómez-Pinedo, U., Cerdán-Sánchez, L.F., 2013. Sex differences in lipid peroxidation cytokines and chemokines by blood monocytes to in vitro lipopolysaccharide sti-
and fatty acid levels in recent onset schizophrenia. Prog. Neuro-Psychopharmacol. mulation are associated with hostility and severity of depressive symptoms in healthy
Biol. Psychiatry 44, 154–161. http://dx.doi.org/10.1016/j.pnpbp.2013.02.007. women. Psychoneuroendocrinology 29, 1119–1128. http://dx.doi.org/10.1016/j.
Reichenberg, A., Yirmiya, R., Schuld, A., Kraus, T., Haack, M., Morag, A., Pollmächer, T., psyneuen.2004.01.002.
2001. Cytokine-associated emotional and cognitive disturbances in humans. Arch. Suzuki, K., Ota, H., Sasagawa, S., Sakatani, T., Fujikura, T., 1983. Assay method for
Gen. Psychiatry 58, 445. http://dx.doi.org/10.1001/archpsyc.58.5.445. myeloperoxidase in human polymorphonuclear leukocytes. Anal. Biochem. 132,
Remus, J.L., Dantzer, R., 2016. Inflammation models of depression in rodents: relevance 345–352. http://dx.doi.org/10.1016/0003-2697(83)90019-2.
to psychotropic drug discovery. Int. J. Neuropsychopharmacol. http://dx.doi.org/10. Tao, W., Dong, Y., Su, Q., Wang, H., Chen, Y., Xue, W., Chen, C., Xia, B., Duan, J., Chen,
1093/ijnp/pyw028. G., 2016. Liquiritigenin reverses depression-like behavior in unpredictable chronic
Rodgers, R.J., Cole, J.C., 1993. Influence of social isolation, gender, strain, and prior mild stress-induced mice by regulating PI3K/Akt/mTOR mediated BDNF/TrkB
novelty on plus-maze behaviour in mice. Physiol. Behav. 54, 729–736. pathway. Behav. Brain Res. 308, 177–186. http://dx.doi.org/10.1016/j.bbr.2016.04.
Rosenblat, J.D., Cha, D.S., Mansur, R.B., McIntyre, R.S., 2014. Inflamed moods: a review 039.
of the interactions between inflammation and mood disorders. Prog. Neuro- Talarowska, M., Szemraj, J., Gałecki, P., 2015. Myeloperoxidase gene expression and
Psychopharmacol. Biol. Psychiatry 53, 23–34. http://dx.doi.org/10.1016/j.pnpbp. cognitive functions in depression. Adv. Med. Sci. 60, 1–5. http://dx.doi.org/10.1016/
2014.01.013. j.advms.2014.06.001.
Russo, S.J., Nestler, E.J., 2013. The brain reward circuitry in mood disorders. Nat. Rev. Todorović, N., Filipović, D., 2017. Prefrontal cortical glutathione-dependent defense and
Neurosci. 14, 609–625. http://dx.doi.org/10.1038/nrn3381. proinflammatory mediators in chronically isolated rats: modulation by fluoxetine or
Savignac, H.M., Couch, Y., Stratford, M., Bannerman, D.M., Tzortzis, G., Anthony, D.C., clozapine. Neuroscience 355, 49–60. http://dx.doi.org/10.1016/j.neuroscience.
Burnet, P.W.J., 2016. Prebiotic administration normalizes lipopolysaccharide (LPS)- 2017.04.044.
induced anxiety and cortical 5-HT2A receptor and IL1-β levels in male mice. Brain Vaccarino, V., Brennan, M.-L., Miller, A.H., Bremner, J.D., Ritchie, J.C., Lindau, F.,
Behav. Immun. 52, 120–131. http://dx.doi.org/10.1016/j.bbi.2015.10.007. Veledar, E., Su, S., Murrah, N.V., Jones, L., Jawed, F., Dai, J., Goldberg, J., Hazen,
Sayd, A., Antón, M., Alén, F., Caso, J.R., Pavón, J., Leza, J.C., Rodríguez de Fonseca, F., S.L., 2008. Association of major depressive disorder with serum myeloperoxidase and
García-Bueno, B., Orio, L., 2014. Systemic administration of oleoylethanolamide other markers of inflammation: a twin study. Biol. Psychiatry 64, 476–483. http://dx.
protects from neuroinflammation and anhedonia induced by LPS in rats. Int. J. doi.org/10.1016/j.biopsych.2008.04.023.
Neuropsychopharmacol. 18. http://dx.doi.org/10.1093/ijnp/pyu111. Vieira, J.O., Duarte, J.O., Costa-Ferreira, W., Morais-Silva, G., Marin, M.T., Crestani, C.C.,
Salazar, A., Gonzalez-Rivera, B.L., Redus, L., Parrott, J.M., O'Connor, J.C., 2012. 2017. Sex differences in cardiovascular, neuroendocrine and behavioral changes
Indoleamine 2,3-dioxygenase mediates anhedonia and anxiety-like behaviors caused evoked by chronic stressors in rats. Prog. Neuro-Psychopharmacol. Biol. Psychiatry.
by peripheral lipopolysaccharide immune challenge. Horm. Behav. 62, 202–209. http://dx.doi.org/10.1016/j.pnpbp.2017.08.014.
http://dx.doi.org/10.1016/j.yhbeh.2012.03.010. Vogelzangs, N., de Jonge, P., Smit, J.H., Bahn, S., Penninx, B.W., 2016. Cytokine pro-
Scholz, H., Eder, C., 2017. Lysophosphatidylcholine activates caspase-1 in microglia via a duction capacity in depression and anxiety. Transl. Psychiatry 6, e825. http://dx.doi.
novel pathway involving two inflammasomes. J. Neuroimmunol. 310, 107–110. org/10.1038/tp.2016.92.
http://dx.doi.org/10.1016/j.jneuroim.2017.07.004. Võikar, V., Kõks, S., Vasar, E., Rauvala, H., 2001. Strain and gender differences in the
Schuch, J.J.J., Roest, A.M., Nolen, W.A., Penninx, B.W.J.H., de Jonge, P., 2014. Gender behavior of mouse lines commonly used in transgenic studies. Physiol. Behav. 72,
differences in major depressive disorder: results from the Netherlands study of de- 271–281.
pression and anxiety. J. Affect. Disord. 156, 156–163. http://dx.doi.org/10.1016/j. Weber, A., Wasiliew, P., Kracht, M., 2010. Interleukin-1 (IL-1) pathway. Sci. Signal. 3
jad.2013.12.011. (cm1-cm1). https://doi.org/10.1126/scisignal.3105cm1.
Sedlak, J., Lindsay, R.H., 1968. Estimation of total, protein-bound, and nonprotein sulf- WHO | Depression [WWW Document] (Ed.), 2015. Depress, Fact sheet [Internet].
hydryl groups in tissue with Ellman's reagent. Anal. Biochem. 25, 192–205. Zhang, D., Wen, X., Wang, X., Shi, M., Zhao, Y., 2009. Antidepressant effect of
Sens, J., Schneider, E., Mauch, J., Schaffstein, A., Mohamed, S., Fasoli, K., Saurine, J., Shudihuang on mice exposed to unpredictable chronic mild stress. J.
Britzolaki, A., Thelen, C., Pitychoutis, P.M., 2017. Lipopolysaccharide administration Ethnopharmacol. 123, 55–60. http://dx.doi.org/10.1016/j.jep.2009.02.029.

142

Вам также может понравиться