Tibolone Reduces Oxidative Damage and Inflammation in Microglia Stimulated With Palmitic Acid Through Mechanisms Involving Estrogen Receptor Beta.

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Mol Neurobiol

DOI 10.1007/s12035-017-0777-y

Tibolone Reduces Oxidative Damage and Inflammation


in Microglia Stimulated with Palmitic Acid through Mechanisms
Involving Estrogen Receptor Beta
Oscar Hidalgo-Lanussa 1 & Marco Ávila-Rodriguez 2 & Eliana Baez-Jurado 1 &
Jairo Zamudio 1 & Valentina Echeverria 3,4 & Luis Miguel Garcia-Segura 5,6 &
George E. Barreto 1

Received: 5 July 2017 / Accepted: 15 September 2017


# Springer Science+Business Media, LLC 2017

Abstract High concentrations of palmitic acid in plasma in- membrane potential. These effects were accompanied by re-
crease both the inflammation associated with obesity and the duced nuclear translocation of NF-κB p65, upregulation of
susceptibility to develop a neurodegenerative event. In the neuroglobin, and improved antioxidant defense.
brain, the inflammatory response is mediated by activated Furthermore, estrogen receptor beta (ERβ) inhibition partially
microglial cells, which undergo morphological and biochem- dampened tibolone’s protective actions in BV-2 cells stimu-
ical changes and can directly affect cell viability. Recent evi- lated with palmitic acid. In conclusion, tibolone protects BV-2
dence shows that the use of estrogenic compounds can control cells by a mechanism involving ERβ and upregulation of
microglia-induced inflammation with promising results. In neuroglobin.
this study, we explored the actions of the synthetic steroid
tibolone on BV-2 microglia cells stimulated with palmitic ac- Keywords Palmitic acid . Obesity . Neurodegenerative
id. Our results demonstrated that tibolone increased cell via- diseases . Microglia . Neuroglobin . Estrogen receptors .
bility and reduced nuclear fragmentation and the production Tibolone . Neuroinflammation
of reactive oxygen species, as well as preserved mitochondrial

Electronic supplementary material The online version of this article Introduction


(https://doi.org/10.1007/s12035-017-0777-y) contains supplementary
material, which is available to authorized users.
High-fat diets significantly increase plasma-free fatty acid
* Luis Miguel Garcia-Segura
levels and are directly associated with the expression of genes
lmgs@cajal.csic.es that interfere with cell metabolism, growth, and differentiation
* George E. Barreto
[1, 2], as well as in the susceptibility to develop chronic in-
gsampaio@javeriana.edu.co; gesbarreto@gmail.com flammatory processes [3]. Recent studies have found that a
diet rich in simple sugars and saturated fatty acids is associated
1
Departamento de Nutrición y Bioquímica, Facultad de Ciencias, with decreased neurotrophic protection [4] and increased sus-
Pontificia Universidad Javeriana, Bogotá D.C., Colombia ceptibility to cognitive pathologies such as Alzheimer’s dis-
2
Departamento de Ciencias Clínicas, Facultad de Ciencias de la Salud, ease [5, 6]. Elevated levels of palmitic acid, a saturated fatty
Universidad del Tolima, Ibagué, Colombia acid, induce the expression of markers for the activation in
3
Research & Development Service, Bay Pines VA Healthcare System, glial cells and the expression of the pro-inflammatory cyto-
Bay Pines, FL 33744, USA kine TNF-α mRNA in the brain of ob/ob mice [7]. Similarly,
4
Fac. Cs de la Salud, Universidad San Sebastián, Lientur 1457, fatty acids disrupt the uptake of glucose in cells and at high
4080871 Concepción, Chile concentrations can trigger metabolic disorders, including
5
Instituto Cajal, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain dyslipidemias, obesity, and diabetes [8]. Nowadays, obesity
6
CIBER de Investigación Biomédica en Red de Fragilidad y
has become a major public health problem and represents
Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos a common pathophysiological basis for several chronic
III, Madrid, Spain diseases [9].
Mol Neurobiol

The inflammatory processes underlying cerebral patholo- plates for flow cytometry determinations, and in 24-well
gies [10, 11] may lead to a continuous and systematic deteri- plates for tetramethyl rhodamine methyl ester (TMRM) and
oration of the brain tissue [12–14] and activation of different immunofluorescence measurements.
inflammation-related cells such as astrocytes and microglia
[15]. These cells may have a dual-side role, acting as protec- Drug Treatment
tors or worsening the damage depending upon the microenvi-
ronment and type of injury [16–18]. As therapeutic ap- Once the cells were seeded in multi-well plates, the culture
proaches, the administration of neuroactive steroids such as medium was replaced with serum-free medium in order to
17ß-estradiol [19, 20], testosterone [21], and the synthetic decrease trophic stimulation that could affect the results of
steroid tibolone [22] has been proven to downregulate the the study. Twelve hours after palmitic acid, cells were treated
inflammatory processes by acting on glial cell activation. with different concentrations of tibolone (Sigma-Aldrich,
Previous studies have reported the action of tibolone on the T0827); 10 nM–10 μM of 1,3,5-Tris(4-hydroxyphenyl)-4-
preservation of mitochondrial membrane potential [22], im- propyl-1H-pyrazole (PPT; ERα agonist; Sigma-Aldrich,
proved cell survival, and reduced inflammation in neural cells H6036) or 2,3-bis(4-hydroxyphenyl)propionitrile (DPN;
[23, 24]. It is noteworthy that many of these mechanisms ERβ agonist; Sigma-Aldrich, H5915). In addition, 3 or 6 h
associated with tibolone are mediated by the activation of prior incubation with tibolone, PPT, or DPN, cell cultures
estrogen receptors and by regulating the expression of survival were treated with 100 nM 1,3-bis(4-hydroxyphenyl)-4-meth-
proteins such as Bcl-2 [25] or neuroglobin (Ngb1) [26]. Ngb1 yl-5-[4-(2-piperidinyl-ethoxy)phenyl]dihydrochloride-1H-
is involved in several critical processes, such as oxygen trans- pyrazole (MPP; ERα antagonist, Sigma-Aldrich, M7068) or
port, the control of reactive oxygen species, and the preven- 100 nM 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-
tion of apoptosis [27–29]. Nevertheless, the effects and mech- a]pyrimidin-3-yl]phenol (Phtpp; ERβ antagonist; Sigma-
anisms of action of tibolone have not been explored in Aldrich, SML1355). Tibolone, PPT, DPN, MPP, or Phtpp
microglial cells. In this study, we have addressed whether (i) were dissolved in 0.01% dimethyl sulfoxide (DMSO) as stock
tibolone has an effect on the inflammatory response induced solution at 5 mM, and further dilutions were made using
by palmitic acid on microglial cells and (ii) its mechanism of serum-free culture medium and without phenol red and L-
action involve the activation of estrogen receptors and/or the glutamine.
expression of Ngb1.

Palmitic Acid
Materials and Methods
We prepared a mixture containing 5% bovine serum albumin
BV-2 Cell Culture (BSA) (Sigma, St Louis, MO, USA), 2 mM palmitic acid
(Sigma, P0500), and 2 mM carnitine (Sigma, C0283) in
BV-2 cell line (ATCC CRL-2469) was used as a microglial RPMI with L-glutamine and serum-free conditions.
cell model. It is a mouse cell line with macrophage morphol- Carnitine was used to allow palmitic acid entry to the mito-
ogy derived from murine neonates and was generated by in- chondria. We tested the following concentrations of palmitic
fecting primary microglial cell cultures with a v-raf/v-myc acid: 100, 250, 500, 750 μM, and 1 mM for 12, 18, and 24 h.
oncogenic retrovirus. BV-2 cells express non-specific esterase The maximum concentration of BSA used was 2.5%, and in
activity, phagocytosis capacity, and a low peroxidase. The all experiments, we used 2 mM carnitine. Control group
BV-2 cell line has been used in numerous studies and has been consisted in 2.5% BSA and 2 mM carnitine dissolved in
shown to share structural and functional similarities with pri- serum-free conditions.
mary microglia [30] and to retain most of the morphological,
phenotypic, and functional properties described for newly iso- Determination of Viability and Nuclear Fragmentation
lated microglial cells [31].
Cells were maintained under exponential growth on RPMI After the experimental treatments, cells were incubated for 4 h
1640 medium (Lonza, Walkersville, USA, Cat.12-702Q), at 37 °C with 5 mg/mL MTT (3-(4,5-dimethylthiazol-2-yl)-
containing 10% fetal bovine serum (Lonza, Walkersville, 2,5-diphenyltetrazolium; Sigma M2128). Cells were then
USA), and penicillin 10 U/Streptomycin 10 mg/amphotericin lysed by the addition of DMSO. The blue formazan product
25 ng (Lonza, Walkersville, USA). Cultures were incubated at was evaluated in a plate reader at 595 nm (spectrophotometer).
37 °C in a humidified atmosphere containing 5% carbon di- The values were then normalized to the value of control
oxide. The medium was changed three times per week. Cells cultures without palmitate, which was considered to be
were seeded in 96-well plates for death cell measurement, in 100% survival. Each trial was performed with a minimum of
6-well plates for Western blot determinations, in 12-well three replicates per experiment.
Mol Neurobiol

Cell viability was also determined with propidium iodide Determination of Mitochondrial Membrane Potential
(Santa Cruz, CAS 25535-16-4); cells were stained for 15 min (ΔΨm)
with 10 μg/mL propidium iodide dissolved in PBS 1X.
Propidium iodide positive cells were quantified by fluores- Mitochondrial membrane potential was assessed by
cence microscopy. Alternatively, cells were washed in PBS tetramethyl rhodamine methyl ester (TMRM, Sigma T5428)
and detached with trypsin (trypsin/EDTA 500:200 mg/L; staining. Cells were incubated at 37 °C for 20 min in the dark
Lonza, Walkersville, USA). Propidium iodide positive cells with 500 nM TMRM. The results were quantified by flow
were then quantified by flow cytometry (Guava EasyCyte ™ cytometry or fluorescence microscopy. As an experimental
Millipore cytometer, Billerica, Massachusetts, USA). Each control, carbonylcyanide-m-chlorophenylhydrazone (CCCP)
trial was performed with a minimum of three replicates for (Sigma-Aldrich) was used. This allowed to dissipate the mem-
each condition. brane potential and define the baseline for mitochondrial po-
In order to confirm cell viability, we determined frag- tential analysis.
mentation and nuclear condensation by Hoechst 33258
staining. Cells were washed three times in PBS 1X and Determination of Mitochondrial Mass
fixed for 20 min in a solution of 4% paraformaldehyde at
room temperature. Subsequently, cells were washed and Mitochondrial mass was evaluated by staining 10-N-nonyl
labeled with Hoechst 33258 (5 mg/mL; Invitrogen) for orange acridine (NAO, Invitrogen). This probe is primar-
15 min. Cell nuclei were observed and photographed using ily used to determine non-peroxidated cardiolipin that is
an inverted fluorescence microscope Olympus IX-53 (UV present mainly in active mitochondria. Cells were stained
excitation, filter CKX-NU N1157600) (excitation 352 nm/ at 37 °C for 20 min in the dark with 200 nM NAO, after
emission 461 nm spectra) with an exposure time set be- washing twice with PBS 1X. The results were quantified
tween 80 and 100 ms to avoid the saturation of the pixels. by flow cytometry or fluorescence microscopy.
The number of fragmented/condensation nuclei was deter-
mined in at least eight randomly selected areas (0.03 mm2) Immunocytochemistry
from each experimental group. The number of fragmented
and condensed nuclei was determined in at least eight ran- Cells were washed in PBS 1X and fixed with 4% parafor-
domly selected areas of each experimental group. Data maldehyde for 20 min at room temperature. Subsequently,
were expressed as a percentage of fragmented and con- cells were permeabilized for 10 min in 0.1% Triton X-100
densed nuclei relative to the total number of cells. in PBS 1X. Then, cells were washed with PBS 1X, blocked
with 2% BSA + Tris-buffered saline (TBS). Subsequently,
cells were incubated overnight with the primary antibodies
Determination of Reactive Oxygen Species for 4-hydroxynonenal (ab46545) (1:500), neuroglobin
(ab37258) (1:50), anti-NF-κB p65 (ab16502) (1:500), 3-
Evaluation of Superoxide Levels nitrotyrosine (ab61392) (1:500), and anti-8-
hydroxyguanosine (8-OGN) (15A3) (ab62623) (1:500).
Cells were treated in the dark for 30 min at 37 °C with 10 μM After incubation with primary antibody, cells were washed
dihydroethidium (DHE; Sigma, D7008) to evaluate the three times for 5 min with TBS and incubated for 45 min at
superoxide radical O2−. Cells were then washed in PBS and room temperature with the appropriate secondary antibody
detached with trypsin (trypsin/EDTA 500:200 mg/L; Lonza, goat anti-mouse IgG (H + L), DyLight 488 conjugate, or
Walkersville, USA) for flow cytometry analysis or maintained goat anti-rabbit for 1 h. Finally, cells were washed with
on the plate to be evaluated by fluorescence microscopy. As a TBS, stained with Hoechst 33258 to label the nuclei, and
positive control, we used 50 mM rotenone to induce the photographed on an inverted Olympus IX-53 fluorescence
production of reactive oxygen species (ROS). The microscope. Quantification was performed using ImageJ
experiments were repeated in three different cultures. software.

Estimation of Cellular Mean Fluorescence Intensity


Evaluation of Hydrogen Peroxide Radical
The calculation of mean fluorescence intensity of the cells,
Production of hydrogen peroxide radical was evaluated by for the determination of ROS, mitochondrial membrane po-
staining with 2,7-dichlorofluorescein diacetate (DCFDA; tential, and immunocytochemistry, was assessed using
Sigma 35845) at a final concentration of 10 μM, in the dark ImageJ version 1.47v. The microphotographs were opened
at 37 °C for 20 min. The results were quantified by flow in the software and pre-processed eliminating the back-
cytometry or fluorescence microscopy. ground. Subsequently, 20 cells were randomly selected
Mol Neurobiol

using a numbered grid in each microphotograph. The mean UGUGAUUUAUGGUGCAGUAACCAAC-3′.


fluorescence value of the 20 cells was determined in 8 mi- Oligofectamine and oligonucleotides (400 pM) were mixed
crophotographs for each treatment using the measure algo- with Optimem. The mixture was incubated for 20 min at room
rithm of ImageJ and selecting each cell manually via ROI’s temperature, diluted with Optimem, and added to the cell me-
Management. There were no variations in the conditions of dium for 4 h at 37 °C. The medium was added to cells to reach
the image processing. Thus, gain of the mercury lamp, time the growing conditions (i.e., 10% (v/v) serum).
of exposure, and fluorochrome bleach were maintained
constant. Each assay was performed with a minimum of 6 Co-localization Analysis
replicate wells for each condition. The experiments were
repeated three times. For the analysis of the co-localization of p65 and Hoechst,
cells were incubated overnight with the primary antibody
Protein Extraction and Western Blotting anti-NF-κB p65 (ab16502), which was recognized using
the corresponding goat anti-rabbit secondary antibody in-
BV-2 cells were lysed on ice with RIPA Lysis and cubated for 1 h. Then, cells were co-stained with Hoechst
Extraction Buffer Thermo Scientific™ supplemented with 33258. The percentage of co-localization was determined
Halt™ Protease Inhibitor Cocktail, EDTA-free (100X) in for cells positive for both markers. Cells were
(Roche). Protein content was estimated using the photographed in 4 different microscope fields accounting
Pierce™ BCA Protein Assay Kit. Equal amounts of protein for an area of 0.03 mm 2 . The co-localization plugin
were dissolved in sample buffer containing 5% β- Intensity Correlation Analysis 6.0 of ImageJ was used to
mercaptoethanol and boiled. Then, proteins were separated obtain the percentage of co-localization in each field. Four
by electrophoresis in SDS–PAGE at 90 V for 90 min at microscope fields in 10 images for each condition were
25 °C, transferred onto PVDF membranes for 90 min at analyzed. The results were plotted as percentage of co-
350 mA and 4 °C and blocked in 5% skim milk dissolved localization.
in Tris-buffered saline containing 0.05% Tween 20 (TBS-
T), at RT for 1 h. The membranes were incubated at 4 °C Statistical Analysis
overnight with antibodies against neuroglobin (Ngb1)
(Abcam) (1:500), β-actin (Thermo Fisher) (1:3000), super- Data obtained from this study were tested for normal dis-
oxide dismutase 2 (Thermo Fisher, PA5-30604) (1:1000), tribution with the Kolmogorov–Smirnov test and for ho-
Catalase (Thermo Fisher, PA5-29183) (1:1000), GPX1 mogeneity of variances with the Levene’s test. Then, data
(Thermo Fisher, PA5-30593) (1:1500), NF-κB p65 were examined by analysis of variance (ANOVA), follow-
(abcam) (1.500), IkBα (Thermo Fisher, PA5-12186) ed by Dunnett’s post hoc test for comparisons between
(1:1000), estrogen receptor beta (Thermo Fisher, PA1- controls and treatments and Tukey’s post hoc test for mul-
311) (1:1000), and estrogen receptor alpha (Thermo tiple comparisons between the means of treatments and
Fisher, MA1-310) (1:1000). The immunoreactivity was vi- time points. Data are presented as mean ± SEM of three
sualized by incubating the membrane with specific second- independent experiments. A statistically significant differ-
ary antibody (IRDye® Antibodies) for 1 h and detected ence was defined at P < 0.05.
using Odyssey CLx Imaging System Specifications (LI-
COR Biosciences). The intensity of each band was quanti-
fied using ImageJ software (National Institutes of Health, Results
Bethesda, MD, USA). All data were normalized to control
values on each gel. The experiments were repeated in three Tibolone Promoted the Survival and Preserved
different cultures. the Morphology of BV-2 Cells Treated with Palmitic Acid

Neuroglobin Silencing The analysis of time and dose-dependent curves of the ac-
tion of palmitic acid on BV-2 cells (Fig. S1) showed that
Cells were transfected in serum-free conditions with either 250 μM palmitic acid for 12 h reduced cell survival by
Stealth RNAi™ Ngb1 siRNA (siNgb1; Invitrogen, Carlsbad, ~ 50% (Fig. S1D). This was accompanied by increased
CA, USA) or a mismatch sequence in accordance with the fragmented nuclei (Fig. S1E), diminished mitochondrial
manufacturer ’s instructions, using oligofectamine membrane potential (Fig. S1F), and augmented ROS pro-
(Invitrogen) as the transfection reagent. The sequence used duction (Fig. S1G, H). Next, we evaluated the actions of
for Ngb1 oligonucleotides was 5′- tibolone in our model. Cells were pre-treated or co-treated
CGUGAUUGAUGCUGCAGUGACCAAU-3′. The control with different concentrations of tibolone for 12, 18, or 24 h
m i s m a t c h s e q u e n c e w a s 5 ′ - and 250 μM palmitic acid (Fig. S2). Under these
Mol Neurobiol

Fig. 1 Effects of tibolone (Tibo) and palmitic acid (Pal) on cell viability 0.01 μM tibolone was able to attenuate the number of condensed nuclei
and nuclear fragmentation. a–b MTT and PI assays revealed that 250 μM (P < 0.0001 Pal vs. Pal + Tibo 0.01 μM). The panel shows representative
palmitic acid for 12 h reduced cell viability (P < 0.0001, Vh vs. Pal 12 h) examples of Hoechst 33258, propidium iodide (PI) stained nuclei of BV-2
and that post-treatment with 0.01 μM tibolone for 12 h significantly cells treated with 250 μM palmitic acid for 12 h, and 0.01 μM tibolone for
increased cell survival under palmitic acid insult (P < 0.001, Pal vs. Pal 12 h. Cells were photographed at magnification of ×20. Vh, vehicle. Scale
+ Tibo 0.01 μM). c Palmitic acid increased the number of cells with bar, 20 μm. Significant differences: ****P < 0.0001
fragmented/condensed nucleus (P < 0.0001, Vh vs. Pal 12 h), and

conditions, tibolone was unable to protect from palmitic Tibolone Preserved Mitochondrial Functions
acid. However, our results demonstrated that post- and Reduced Oxidative Stress in BV-2 Cells Exposed
treatment with 0.01 μM tibolone was able to preserve the to Palmitic Acid
viability of cells treated with 250 μM palmitic acid (Fig. 1).
Post-treatment with 0.01 μM tibolone for 12 h significantly Palmitic acid significantly reduced in 50% the mitochondrial
increased cell viability in 25% compared to the group treat- membrane potential compared to vehicle (Fig. 2a, P < 0.0001,
ed with palmitic acid alone (Fig. 1a–b, P < 0.01, Pal vs. Pal Vh vs. Pal). Conversely, tibolone significantly increased by
+ Tibo 0.01 μM at 12 h). We used these parameters in next 40% the mitochondrial membrane potential in palmitic acid-
experiments. Figure 1c shows the number of fragmented/ treated cells (Fig. 2a, P < 0.0001, Pal vs. Pal + Tibo). Similar
condensed nuclei [22, 32]. Cells treated with palmitic acid results were observed in NAO fluorescence. Palmitic acid
had a significant increase by 75% in the number of reduced mitochondrial mass in 20%, but treatment with
fragmented/condensed nuclei in comparison with vehicle tibolone preserved it at levels similar to vehicle (Fig. 2b).
(P < 0.0001, Pal vs. Vh). On the contrary, tibolone reduced Regarding ROS production, we observed that palmitic acid
by 30% the number of fragmented/condensed nuclei in increased by 37% the production of superoxide (Fig. 2c,
palmitic acid-treated cells (Fig. 1c, P < 0.0001, Pal vs. Pal P < 0.0001, Vh vs. Pal) and peroxide by 15% (Fig. 2d,
+ Tibo). P < 0.001, Vh vs. Pal). On the contrary, tibolone attenuated
Mol Neurobiol

Fig. 2 Effects of post-treatment with 0.01 μM tibolone (Tibo) and NAO fluorescence (b). Tibolone preserved mitochondrial mass and vol-
250 μM palmitic acid (Pal) on mitochondrial membrane potential, mito- ume in cells upon treatment with palmitic acid (P < 0.001, Pal vs. Pal +
chondrial mass, and ROS production in BV-2 cells by flow cytometry. Tibo). The levels of superoxide ion (c) were found decreased in palmitic
Microphotographs illustrate cells stained for TMRM (Δψm, mitochon- acid-treated cells following administration of tibolone (P < 0.01, Pal vs.
drial membrane potential), NAO (mitochondrial mass), and DHE (super- Pal + Tibo). On the contrary, a post-treatment with tibolone augmented
oxide levels) in cells exposed to both palmitic acid and tibolone; flow the levels of hydrogen peroxide (d) in cells insulted with palmitic acid
cytometry plots indicate DCFDA (peroxide levels). a Palmitic acid damp- (P < 0.0001, Pal vs. Pal + Tibo). Cells were photographed at ×20 mag-
ened Δψm (P < 0.0001, Vh vs. Pal) and tibolone preserved mitochon- nification. Vh, vehicle. Scale bar, 20 μm. Significant differences:
drial potential in palmitic acid-treated cells at 12 h (P < 0.0001, Pal vs. Pal **P < 0.01, ***P < 0.001, ****P < 0.0001
+ Tibo). Similar effects of tibolone and palmitic acid were observed on

superoxide production by 15% (Fig. 2c, P < 0.01 Pal vs. Pal + tibolone or palmitic acid treatments (P > 0.05; Fig. 3a).
Tibo) and increased hydrogen peroxide by 60% (Fig. 2d, Likewise, no significant alterations in ERβ expression were
P < 0.0001, Pal vs. Pal + Tibo) in cells under palmitic acid. observed when estrogen receptor agonists were administered
(DPN and PPT, Fig. 3a). Next, we treated palmitic acid-
Estrogen Receptor Beta Is Involved in the Protective insulted cells with either DPN (ERβ agonist) or PPT (ERα
Effects of Tibolone on BV-2 Cells agonist) and assessed cell viability by PI. Our findings indi-
cated that 0.1 μM DPN was able to increase by 20% BV-2 cell
Previous studies have shown that estrogen receptor beta plays survival against palmitic acid (Fig. 3b, P ˂ 0.001, Pal vs. Pal +
a role in mitochondrial protection under different stimuli [26, DPN). On the contrary, PPT was unable to improve cell via-
33–35]. First, we determined the protein expression of both bility in cells treated with palmitic acid (Fig. 3c). Finally, we
receptors in BV-2 cells by Western blot. Although we did not investigated whether the pharmacologic blockade of ERβ or
find expression of ERα, BV-2 cells did express ERβ. ERα affects the actions of tibolone, DPN, or PPT in BV-2
However, no significant differences were observed in ERβ cells. We observed that ERβ blockade significantly dimin-
expression, regardless whether cells were subjected to ished by 60% cell viability of microglia subjected to palmitic
Mol Neurobiol

Fig. 3 Implication of estrogen and androgen receptors in BV-2 cells. a cells treated with palmitic acid. d Blocking ERβ with 0.1 μM Phtpp for 3
Western blot analysis showed that these cells do not express ERα even and 6 h, in the presence of tibolone or DPN, reduced cell viability in
under different experimental paradigms. On the contrary, ERβ expression palmitic acid-treated cells. e Blocking ERα with MPP did not exert any
was detected. However, ERβ expression was not modulated when cells significant effect on cell viability. f Blocking androgen receptor with
were treated with vehicle (Vh), palmitic acid (Pal), tibolone (Tibo), Phtpp different flutamide concentrations did not affect the actions of tibolone
(ERβ antagonist), DPN (ERβ agonist), or PPT (ERα agonist). b Post- on cell survival. Western blot lanes: 1 Vh, 2 Pal, 3 Pal + Tibo, 4 Phtpp +
treatment with 0.1 μM DPN for 12 h significantly increased cell viability Pal + Tibo, 5 Pal + DPN, 6 Phtpp + Pal + Dpn, 7 Pal + PPT. Significant
in cells exposed to palmitic acid (P < 0.001, Pal vs. Pal + 0.1 μM DPN). c differences: **P < 0.01, ***P < 0.001, ****P < 0.0001
Different PPT concentrations had no significant effects on cell viability in

acid and exposed to tibolone or DPN (Fig. 3d, P < 0.0001, Pal acid decreased by 20% the SOD expression compared
+ Tibo vs. Phtpp + Pal + Tibo; P < 0.0001, Pal + DPN vs. to control (Fig. S4B, P < 0.01 Vh vs. Pal). In contrast,
Phtpp + Pal + DPN). In contrast, blocking ERα with MPP did both tibolone and DPN increased SOD expression to
not significantly affect cell viability (Fig. 3e). Finally, we also control values in palmitic acid-treated cells (Fig. S4B,
assessed the role of androgen receptor in mediating the effect P < 0.05 Vh vs. Pal + Tibo; P < 0.01, Vh vs. Pal +
of tibolone on BV-2 cells. Our findings showed that the an- DPN). Upon ERβ blockade using Phtpp, tibolone and
drogen receptor antagonist flutamide did not alter the protec- DPN effects on SOD were dampened (Fig. S4B,
tive effect of tibolone on palmitic acid-treated cells (Fig. 3f). P < 0.01, Pal + Tibo vs. Phtpp + Pal + Tibo;
P < 0.01, Pal + DPN vs. Phtpp + Pal + DPN).
Tibolone Reduced Oxidation of Lipids, Proteins, Further, we investigated the actions of tibolone and
and Nucleic Acids in BV-2 Cells Treated with Palmitic DPN on catalase and GPX-1 expression in palmitic
Acid acid-treated cells. Similar to SOD, palmitic acid reduced
by 10% the expression of catalase (Fig. S4C, P < 0.01,
In this aim, we evaluated the actions of tibolone on DNA, Vh vs. Pal), but not of GPX-1 (Fig. S4D). Catalase
protein, and cell membrane oxidation in cells treated with expression was also found decreased when cells were
palmitic acid. We observed that tibolone reduced by 40% treated with either tibolone or DPN in the presence of
DNA oxidation (Fig. 4a, P < 0.0001, Pal vs. Pal + Tibo), by palmitic acid (Fig. S4C, P < 0.01, Pal vs. Pal + Tibo;
60% protein oxidation (Fig. 4b, P < 0.0001, Pal vs. Pal + P < 0.05, Pal vs. Pal + DPN). When the ERβ was
Tibo), and by 50% lipid peroxidation in cells treated with antagonized, the actions of tibolone and DPN on cata-
palmitic acid (Fig. 4c, P < 0.0001, Pal vs. Pal + Tibo). lase expression were blunted (Fig. S4C). GPX-1 expres-
Similar effects on all three analyzed parameters were observed sion was not significantly affected by palmitic acid,
when cells were treated with the ERβ agonist DPN. tibolone, PPT, or DPN (Fig. S4D).

Tibolone Regulated the Expression of Antioxidant Tibolone and DPN Prevented the Effect of Palmitic Acid
Enzymes on the Expression of Neuroglobin in BV-2 Cells

Figure S4A shows representative Western blots for each Ngb1 expression was assessed by immunocytochemistry
protein under different experimental paradigms. Palmitic and Western blotting (Fig. 5). Interestingly, palmitic acid
Mol Neurobiol

Fig. 4 Effects of tibolone (Tibo), ERβ agonist (DPN), and ERα agonist similar to that exerted by DPN (P < 0.0001, Pal vs. Pal + DPN). Tibolone
(PPT) on oxidative stress markers and ROS production in cells exposed to and DPN had similar effects by attenuating the levels of both 3-
palmitic acid (Pal). Microphotographs showed cells stained for 8OHdG, nitrotyrosine (b), a marker of nitrated proteins, and 4-HNE (c), a marker
3-NT, and 4-HNE in the presence of Vh (vehicle), Pal, DPN, or Tibo by of lipid peroxidation (P < 0.0001, Pal vs. Pal + Tibo) under palmitic acid
immunocytochemistry. a Tibolone decreased 8OHdG levels (P < 0.0001, exposure. Cells were photographed at ×20 magnification. Scale bar,
Pal vs. Pal + Tibo) in cells stimulated with palmitic acid, with an effect 20 μm. Significant differences: ***P < 0.001, ****P < 0.0001

significantly reduced by 30% neuroglobin immunofluo- Tibolone and Palmitic Acid Modulated p65 Expression
rescence in BV-2 cells in comparison to vehicle (Fig. and Nuclear Translocation
5a, P ˂ 0.0001, Vh vs. Pal). On the contrary, tibolone
and DPN increased by 50% Ngb1 immunoreactivity in Recent studies showed that tibolone regulates the NF-κB
palmitic acid-treated cells, a level that was above to pathway via AKT activation [36]. Next, we investigated the
control values (Fig. 5a, P ˂ 0.0001, Pal vs. Pal + effect of palmitic acid and tibolone on p65 nuclear expression
Tibo and Pal vs. Pal + DPN). The ERβ antagonist subunit at different times using immunofluorescence (Fig. 6a).
Phtpp caused a significant reduction of 27 and 22% in Our findings indicated that palmitic acid induced an increase
Ngb1 immunoreactivity in cells treated either with of ~ 60% in expression of p65 at 12 h when compared to
tibolone (Fig. 5a, P ˂ 0.05, Pal + Tibo vs. Phtpp + baseline. In contrast, tibolone decreased the expression of
Pal + Tibo) or DPN (Fig. 5a, P ˂ 0.05, Pal + DPN p65 when administered along with palmitic acid (Fig. 6a,
vs. Phtpp + Pal + DPN), respectively. This finding sug- P < 0.0001, Pal vs. Pal + Tibo) in this same time period. Co-
gests potential implication of ERβ in the upregulation localization studies using ImageJ co-localization plugin at
of Ngb1 levels by tibolone. These observations were 12 h (Fig. 6b–e) were performed and corroborated our previ-
corroborated by Ngb1 protein expression (Fig. 5b). ous findings.
Since we observed that tibolone induced the expres- The effect of tibolone, reducing p65 nuclear translocation
sion of Ngb1 in BV-2 cells, next we evaluated the role in cells treated with palmitic acid, was imitated by the ERβ
played by this protein by using siRNA (Fig. S3). Ngb1 agonist DPN and prevented by the ERβ antagonist Phtpp
silencing did not affect the protective effect of tibolone (Fig. 7a). Similar results were observed in Western blot
on the viability of cells treated with palmitic acid (Fig. (WB) analysis (Fig. 7b). We investigated the expression of
S3A). In contrast, Ngb1 silencing prevented the effect IκBα in BV-2 cells. Palmitic acid significantly decreased
of tibolone on superoxide ion production (Fig. S3B) and IκBα expression in comparison to controls (Fig. 7c,
mitochondrial membrane potential (Fig. S3C) in cells P ˂ 0.001, Vh vs. Pal). DPN and tibolone positively modulat-
treated with palmitic acid. ed by 60% the IκBα expression in the cells treated with
Mol Neurobiol

Fig. 5 Expression of neuroglobin (Ngb1) in BV-2 cells stimulated with showed reduction in Ngb1 expression. b Western blot quantification of
vehicle (Vh), palmitic acid (Pal), tibolone (Tibo), ERβ antagonist Ngb1 under different conditions corroborated the evidences observed in
(Phtpp), and ERβ agonist (DPN) and subjected to palmitic acid. immunostaining. Cells were photographed at ×20 magnification; scale
Microphotographs showed cells stained for Ngb1 after different treat- bar, 20 μm. Western blot lanes: 1 Vh, 2 Pal, 3 Pal + Tibo, 4 Phtpp +
ments. a Immunofluorescence for Ngb1 demonstrated increased expres- Pal + Tibo, 5 Pal + DPN, 6 Phtpp + Pal + DPN. Scale bar, 20 μm.
sion when cells were treated with tibolone 0.01 or 0.1 μM DPN, in the Significant differences: *P < 0.05, **P < 0.01, ***P < 0.001,
presence of palmitic acid. On the contrary, cells treated with Phtpp ****P < 0.0001

palmitic acid (Fig. 7c, P ˂ 0.01, Pal vs. Pal + Tibo and Pal vs. indicated that tibolone improved cell viability, attenuated nu-
Pal + DPN). The ERβ antagonist Phtpp prevented the effects clear condensation, reduced lipid and protein oxidation, and
of tibolone and DPN on IκBα expression in palmitic acid- preserved mitochondrial functionality. These actions were ac-
treated cells (Fig. 7c, P ˂ 0.01, Pal + Tibo vs. Phtpp + Pal + companied by a positive regulation of Ngb1 and diminished
Tibo; P ˂ 0.01, Pal + DPN vs. Phtpp + Pal + DPN). translocation of p65 subunit of NF-κB. Our results also sug-
gest that tibolone protective effects are in part mediated by
estrogen receptor beta.
Discussion Upon increased concentration of palmitic acid, it is expect-
ed that the glycolytic and oxidative pathways result in an
Palmitic Acid Actions on BV-2 Cells imbalance of metabolic intermediates that migrate to alterna-
tive pathways to complete their metabolism. For example, a
Our study for the first time characterized the effects of tibolone dysregulation in the activity of serine palmitoyltransferase
on palmitic acid-treated microglial cells in vitro. Our findings may lead to an increase of dehydrosphingosine and ceramide
Mol Neurobiol

Fig. 6 Expression of the NF-kB p65 subunit at different times of expo- acid alone (c), or palmitic acid and tibolone (d). e Co-localization studies
sure to palmitic acid (Pal), vehicle (Vh), and tibolone (Tibo). a Palmitic using ImageJ plugin showed the percentage of double-labeled cells for
acid-treated cells exposed to tibolone show a reduced expression of p65 at p65 and Hoechst at 12 h. Tibolone reduced the percentage of palmitic
4 h (P < 0.0001, Pal vs. Pal + Tibo), 8 h (P < 0.0001, Pal vs. Pal + Tibo), acid-stimulated cells positive for both p65 and Hoechst (P < 0.0001, Pal
and 12 h (P < 0.0001, Pal vs. Pal + Tibo). Microphotographs showing p65 vs. Pal + Tibo). Cells were photographed in magnification of ×20. Scale
nuclear translocation at 12 h when cells are exposed to Vh (b), palmitic bar, 20 μm. Significant differences: ****P < 0.0001

synthesis [37, 38], which is suggested to decrease the produc- to their protonophoric action on the inner mitochondrial mem-
tion of Bcl-2, an anti-apoptotic protein [39, 40]. On the other brane [44].
hand, saturated fatty acids, such as palmitic acid, may impair
the mitochondrial ATP production and increase ROS produc- Tibolone Diminished the Detrimental Effect of Palmitic
tion [37, 40, 41]. Previously, Lumbertucci et al. (2008) dem- Acid in BV-2 Cells
onstrated that 100 μM palmitic acid can increase ROS pro-
duction by inducing mitochondrial electron transport chain Tibolone has been described as a protective compound with
deterioration [16, 30, 42], inducing cytochrome C reduction the ability to counteract detrimental effects of several dam-
and increased production of the superoxide ion in primary aging conditions [22, 45, 46]. In our model, we observed
culture of rat skeletal muscle cells [43]. Indeed, Shonfeld that BV-2 cells mainly expressed estrogen receptor beta,
and Wojtczak (2008) proposed that palmitic acid may act as suggesting a possible implication of this receptor in medi-
a modulator of ROS production—superoxide and peroxide ating tibolone’s actions. It is reported that the activation of
species—mainly by (i) slowing down the rate of electron flow estrogenic response via estrogen receptor beta is linked to
through Complexes I, III, and IV of the respiratory chain due protective functions in neurons [24], astrocytes [35], and
to interaction within the complex subunit structure and (ii) due microglia [47, 48]. It has been also shown that the
Mol Neurobiol

Fig. 7 Effect of vehicle (Vh), palmitic acid (Pal), tibolone (Tibo), ERβ that Phtpp was able to partially dampen this effect (b). c Quantification of
antagonist (Phtpp), and ERβ agonist (DPN) on p65 expression in cells IkBα expression by WB. Although palmitic acid reduced IkBα expres-
subjected to palmitic acid. a Immunofluorescence for p65 showed re- sion in BV-2 cells, tibolone and DPN preserved its expression in cells
duced expression when cells are treated with tibolone or DPN in the exposed to this fatty acid. A reduced expression of IkBα was observed
presence of palmitic acid (P < 0.0001, Pal vs. Pal + Tibo; P < 0.0001, when cells are treated with Phtpp. Western blot lanes: 1 Vh, 2 Pal, 3 Pal +
Pal vs. Pal + DPN). On the contrary, upon blockade of ERβ with Phtpp, Tibo, 4 Phtpp + Pal + Tibo, 5 Pal + DPN, 6 Phtpp + Pal + Dpn. Cells were
p65 expression was found augmented. These results were corroborated photographed in magnification of ×20. Scale bar, 20 μm. Significant
by WB, showing that tibolone and DPN attenuated p65 expression and differences: *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001

estrogenic response includes the activation of the PI3K-Akt Previous evidences support the actions of estrogen recep-
pathway [49], expression of anti-apoptotic Bcl-2 [25], and tors to counteract oxidative damage [35, 56, 57]. Similar to
increased expression of SOD-2 [50] and Gpx1 [51]. The our study, upon blockade of ERβ, the protective effects of
reduction in oxidative stress induced by palmitic acid in tibolone and DPN were dampened, suggesting a possible im-
cells treated with tibolone suggests a protective effect of plication of ERβ in our model. It has been also reported that
this drug on microglia. Indeed, the regulation of antioxidant estrogen receptor beta is localized in extranuclear compart-
pathways and the maintenance of the mitochondrial func- ments [58], including the mitochondria [58]. For example,
tion evidenced in our study are directly related to the Yang et al. (2009) demonstrated that knockdown of the ERβ
cytoprotective effect of tibolone on mitochondrial mem- results in a lower resting mitochondrial membrane potential in
brane potential and preservation of mitochondrial function- a model of hippocampal cells and primary hippocampal neu-
ality. Additionally, our results are in accordance with these rons [58]. Similarly, ERs mediate innate immune responses
previous studies, as DPN, an ERβ agonist, modulated ROS and microglial activation, as well as IL-6 production, and the
production and improved the mitochondrial membrane po- loss or ER deficiency associated with aging can lead to dereg-
tential [52–55] in BV-2 cells treated with palmitic acid. ulation of the inflammatory response and increase
Mol Neurobiol

Fig. 8 Main effects of palmitic acid and tibolone in our model. In our Interestingly, tibolone was able to counteract the detrimental effects of
study, BV-2 microglial cells were subjected to palmitic acid stimulation. palmitic acid by eliciting an increased viability and diminishing ROS
The basal conditions of BV-2 cells included cytoplasmic p50/p65 local- production. Tibolone also modulated the levels of key antioxidant pro-
ization, preserved mitochondrial functions, and a non-reactive phenotype. teins such as SOD and catalase. More importantly, tibolone upregulated
After palmitic acid stimulation, BV-2 cells undergo detrimental effects as neuroglobin in palmitic acid-treated cells, and upon ERβ blockade,
reduced viability, increased ROS production, and ROS-dependent cell neuroglobin expression was found reduced and this partially attenuated
component degradation. Other detrimental effects included neuroglobin tibolone’s protective action on BV-2 cells
downregulation and increased p50/p65 nuclear translocation.

vulnerability to ischemic injury in female mice brains [59]. It Role of Tibolone in the Inflammatory Response
seems like that ERβ activity is associated with metabolic and in the Model
mitochondrial functions in microglia [58, 60–63]. ERβ is lo-
cated in the cytoplasm of microglia, suggesting a protective It is important to point out that microglia have the ability to
mechanism associated with the non-classic effects of estro- induce an inflammatory response that includes the release of
gens, which inhibit the activation of microglia [30]. In our cytokines and chemokines, and activation of NF-κB down-
results, the stimulation of BV-2 cells with palmitic acid result- stream pathway [57, 73]. Other inflammatory mediators in-
ed in a detrimental mitochondrial function, which was pre- clude growth factors, apoptotic modulators, NLRP3, and
served following tibolone administration. Aim2 [74–76]. Free fatty acids can increase the permeability
It is noteworthy that palmitic acid dampened mitochondrial of the blood–brain barrier allowing the infiltration and activa-
membrane potential, and that tibolone preserved mitochondri- tion of inflammatory modulators. These modulators can reg-
al function through activation of estrogen receptor beta. ulate the activation of microglia by conditioning their re-
Mitochondrial preservation by estrogens, upon different dam- sponse and altering metabolic processes in the brain, thus
aging stimuli, is well documented [64–67]. Indeed, recent increasing the oxidative damage [75, 77]. The inflammatory
studies have shown that estrogenic activity via estrogen recep- response can be modulated by various factors or molecules,
tor beta was involved in the regulation of the protective factor including steroid hormones such as estradiol or SERMs [78].
Ngb1 in astrocytes and neurons [29, 68]. Ngb1 has been Several studies have confirmed the modulation of the immune
shown to be expressed in neurons, astrocytes [68], microglia response by steroid metabolites as estrogen and progesterone
[35, 69], and the endocrine system [70]. Other reports have [14, 69, 78–80].
detected the upregulation of Ngb1 in peri-infarct area, demon- Since some secreted pro-inflammatory factors are under
strating a protective reaction of the tissue against a detrimental control of upstream NF-κB pathway, a critical signal transduc-
condition [69, 71]. Our results extend previous findings show- tion pathway that regulates pro-inflammatory responses [30,
ing increased Ngb1 expression in cultured astrocytes after 57], we determined the response of NF-κB signaling transduc-
LPS stimulation or in an in vivo model of inflammation in- tion by palmitic acid and tibolone. Our results demonstrated
duced by H2O2 toxicity [68, 72]. that NF-κB p65 nuclear translocation was stimulated by
Mol Neurobiol

palmitic acid insult, and this action was reversed by tibolone References
administration, which is in agreement with previous studies
[81–83]. We demonstrated that palmitic acid induced a detri- 1. Sampath H, Ntambi JM (2005) Polyunsaturated fatty acid regula-
mental oxidative stress that involved the NF-κB pathway. This tion of genes of lipid metabolism. Annu Rev Nutr 25:317–340.
https://doi.org/10.1146/annurev.nutr.25.051804.101917
result is in accordance with a previous study suggesting that
2. Jump DB (2004) Fatty acid regulation of gene transcription. Crit
ROS and oxidative damage induce NF-κB activation [76]. Rev Clin Lab Sci 41(1):41–78. https://doi.org/10.1080/
The estrogenic action of tibolone associated with ERβ reduces 10408360490278341
the expression of inflammatory factors [26], as p65 expression 3. Karelis AD, Faraj M, Bastard JP, St-Pierre DH, Brochu M,
was also modulated by the activation of ERβ. In our study, Prud'homme D, Rabasa-Lhoret R (2005) The metabolically healthy
but obese individual presents a favorable inflammation profile. J
blocking ERβ increased the expression of p65, thus notably Clin Endocrinol Metab 90(7):4145–4150. https://doi.org/10.1210/
inhibiting the action of tibolone (Fig. 8). On the other hand, jc.2005-0482
we observed that DPN and tibolone induced similar actions on 4. Mattson MP, Chan SL, Duan W (2002) Modification of brain aging
p65 expression, and upon blocking ERβ, their action was and neurodegenerative disorders by genes, diet, and behavior.
found reduced. These results are supported by Frasor and Physiol Rev 82(3):637–672. https://doi.org/10.1152/physrev.
00004.2002
colleagues (2009), who described an association between 5. Colton CA (2009) Heterogeneity of microglial activation in the
ERs and NF-κB suggesting a co-repression activity [73]. innate immune response in the brain. J NeuroImmune Pharmacol
Our findings also demonstrated that palmitic acid increased 4(4):399–418. https://doi.org/10.1007/s11481-009-9164-4
the expression of p65 subunit and decreased the expression of 6. Tracy LM, Bergqvist F, Ivanova EV, Jacobsen KT, Iverfeldt K
(2013) Exposure to the saturated free fatty acid palmitate alters
IkBα, an upstream regulator of NF-κB, suggesting a possible
BV-2 microglia inflammatory response. J Mol Neurosci 51(3):
pro-inflammatory preconditioning of palmitic acid in BV-2 805–812. https://doi.org/10.1007/s12031-013-0068-7
cells as indicated by Acton (2012) in an astrocytic model 7. Kappe C, Tracy LM, Patrone C, Iverfeldt K, Sjoholm A (2012)
stimulated with LPS [84]. Interestingly, all the pro- GLP-1 secretion by microglial cells and decreased CNS expression
inflammatory activity was reversed in part by tibolone admin- in obesity. J Neuroinflammation 9:276. https://doi.org/10.1186/
1742-2094-9-276
istration, suggesting a novel role of tibolone in modulating the
8. Miller AA, Spencer SJ (2014) Obesity and neuroinflammation: a
inflammatory response via ERβ. Finally, Biswas et al. (2005) pathway to cognitive impairment. Brain Behav Immun 42:10–21.
described that while anti-inflammatory effects of ER activa- https://doi.org/10.1016/j.bbi.2014.04.001
tion have been observed, a non-classical and non-genomic 9. De Zwaan M, Mitchell JE, Howell LM, Monson N, Swan-
effect of estrogen receptors has also been elucidated. This Kremeier L, Roerig JL, Kolotkin RL, Crosby RD (2002) Two mea-
sures of health-related quality of life in morbid obesity. Obes Res
includes the activation of NF-κB and its inflammatory down- 10(11):1143–1151. https://doi.org/10.1038/oby.2002.155
stream response, suggesting that molecules that bind ERs and 10. Pekny M, Pekna M (2014) Astrocyte reactivity and reactive
selectively block NF-κB activation may have a potential ther- astrogliosis: costs and benefits. Physiol Rev 94(4):1077–1098.
apeutic effect [85]. https://doi.org/10.1152/physrev.00041.2013
In conclusion, our results demonstrated the protective ef- 11. Amor S, Peferoen LA, Vogel DY, Breur M, van der Valk P, Baker D,
van Noort JM (2014) Inflammation in neurodegenerative diseases–
fects of tibolone in BV-2 cells against palmitic acid-induced an update. Immunology 142(2):151–166. https://doi.org/10.1111/
oxidative stress and inflammation. Tibolone activates estro- imm.12233
genic activity via ERβ and modulates the expression of 12. Pedraza-Alva G, Perez-Martinez L, Valdez-Hernandez L, Meza-
Ngb1 and NF-κB components in order to reduce mitochon- Sosa KF, Ando-Kuri M (2015) Negative regulation of the
inflammasome: keeping inflammation under control. Immunol
drial damage and inflammation. However, further experi- Rev 265(1):231–257. https://doi.org/10.1111/imr.12294
ments are needed to confirm the involvement of tibolone in 13. Sochocka M, Diniz BS, Leszek J (2016) Inflammatory response in
regulating other inflammatory mediators on in vitro and the CNS: friend or foe? Mol Neurobiol. https://doi.org/10.1007/
in vivo models. s12035-016-0297-1
14. Arevalo MA, Santos-Galindo M, Acaz-Fonseca E, Azcoitia I,
Garcia-Segura LM (2013) Gonadal hormones and the control of
reactive gliosis. Horm Behav 63(2):216–221. https://doi.org/10.
1016/j.yhbeh.2012.02.021
Acknowledgments We acknowledge support from Ministerio de
15. Karve IP, Taylor JM, Crack PJ (2016) The contribution of astro-
Economía y Competitividad (MINECO; grant number BFU2014-
cytes and microglia to traumatic brain injury. Br J Pharmacol
51836-C2-1-R), from CIBERFES, and Fondos FEDER.
173(4):692–702. https://doi.org/10.1111/bph.13125
16. Haversen L, Danielsson KN, Fogelstrand L, Wiklund O (2009)
Induction of proinflammatory cytokines by long-chain saturated
fatty acids in human macrophages. Atherosclerosis 202(2):382–
Compliance with Ethical Standards 393. https://doi.org/10.1016/j.atherosclerosis.2008.05.033
17. Nilsen J, Brinton RD (2004) Mitochondria as therapeutic targets of
Conflict of Interest The authors declare that they have no conflict of estrogen action in the central nervous system. Curr Drug Targets
interest. CNS Neurol Disord 3(4):297–313
Mol Neurobiol

18. Ransohoff RM (2016) How neuroinflammation contributes to neu- 34. Acaz-Fonseca E, Sanchez-Gonzalez R, Azcoitia I, Arevalo MA,
rodegeneration. Science 353(6301):777–783. https://doi.org/10. Garcia-Segura LM (2014) Role of astrocytes in the neuroprotective
1126/science.aag2590 actions of 17beta-estradiol and selective estrogen receptor modula-
19. Said MM, Bosland MC (2016) The anti-inflammatory effect of tors. Mol Cell Endocrinol 389(1–2):48–57. https://doi.org/10.1016/
montelukast, a cysteinyl leukotriene receptor-1 antagonist, against j.mce.2014.01.009
estradiol-induced nonbacterial inflammation in the rat prostate. 35. Acaz-Fonseca E, Avila-Rodriguez M, Garcia-Segura LM, Barreto
Naunyn Schmiedeberg's Arch Pharmacol. https://doi.org/10.1007/ GE (2016) Regulation of astroglia by gonadal steroid hormones
s00210-016-1325-4 under physiological and pathological conditions. Prog Neurobiol
20. Sribnick EA, Ray SK, Banik NL (2006) Estrogen prevents 144:5–26. https://doi.org/10.1016/j.pneurobio.2016.06.002
glutamate-induced apoptosis in C6 glioma cells by a receptor- 3 6 . S u n H Z , Ya n g T W, Z a n g W J , W u S F ( 2 0 1 0 )
mediated mechanism. Neuroscience 137(1):197–209. https://doi. Dehydroepiandrosterone-induced proliferation of prostatic epithe-
org/10.1016/j.neuroscience.2005.08.074 lial cell is mediated by NFKB via PI3K/AKT signaling pathway. J
21. Toro-Urrego N, Garcia-Segura LM, Echeverria V, Barreto GE Endocrinol 204(3):311–318. https://doi.org/10.1677/JOE-09-
(2016) Testosterone protects mitochondrial function and regulates 0270
neuroglobin expression in astrocytic cells exposed to glucose dep- 37. Randle PJ, Garland PB, Hales CN, Newsholme EA (1963) The
rivation. Front Aging Neurosci 8:152. https://doi.org/10.3389/ glucose fatty-acid cycle. Its role in insulin sensitivity and the met-
fnagi.2016.00152 abolic disturbances of diabetes mellitus. Lancet 1(7285):785–789
22. Avila Rodriguez M, Garcia-Segura LM, Cabezas R, Torrente D, 38. Gries FA, Oberdisse K (1970) Lipid metabolism disorders and di-
Capani F, Gonzalez J, Barreto GE (2014) Tibolone protects T98G abetes mellitus. Dtsch Med Wochenschr 95(14):727–734. https://
cells from glucose deprivation. J Steroid Biochem Mol Biol 144 Pt doi.org/10.1055/s-0028-1108532
B:294–303. doi:https://doi.org/10.1016/j.jsbmb.2014.07.009 39. El Akoum S, Lamontagne V, Cloutier I, Tanguay JF (2011) Nature
23. Guzman CB, Zhao C, Deighton-Collins S, Kleerekoper M, of fatty acids in high fat diets differentially delineates obesity-linked
Benjamins JA, Skafar DF (2007) Agonist activity of the 3- metabolic syndrome components in male and female C57BL/6J
hydroxy metabolites of tibolone through the oestrogen receptor in mice. Diabetol Metab Syndr 3:34. https://doi.org/10.1186/1758-
the mouse N20.1 oligodendrocyte cell line and normal human as- 5996-3-34
trocytes. J Neuroendocrinol 19(12):958–965. https://doi.org/10. 40. Little JP, Madeira JM, Klegeris A (2012) The saturated fatty acid
1111/j.1365-2826.2007.01611.x palmitate induces human monocytic cell toxicity toward neuronal
24. Farfan-Garcia ED, Castillo-Hernandez MC, Pinto-Almazan R, cells: exploring a possible link between obesity-related metabolic
Rivas-Arancibia S, Gallardo JM, Guerra-Araiza C (2014) impairments and neuroinflammation. J Alzheimers Dis 30(Suppl
Tibolone prevents oxidation and ameliorates cholinergic deficit in- 2):S179–S183. https://doi.org/10.3233/JAD-2011-111262
duced by ozone exposure in the male rat hippocampus. Neurochem 41. Kingsbury KJ, Heyes TD, Morgan DM, Aylot C, Burton PA,
Res 39(9):1776–1786. https://doi.org/10.1007/s11064-014-1385-0 Emmerson R, Robinson PJ (1962) The effect of dietary changes
25. Gompel A, Chaouat M, Jacob D, Perrot JY, Kloosterboer HJ, on the fatty acid composition of normal human depot fat. Biochem
Rostene W (2002) In vitro studies of tibolone in breast cells. J 84:124–133
Fertil Steril 78(2):351–359 42. Maloney E, Sweet IR, Hockenbery DM, Pham M, Rizzo NO,
26. Avila-Rodriguez M, Garcia-Segura LM, Hidalgo-Lanussa O, Baez E, Tateya S, Handa P, Schwartz MW et al (2009) Activation of NF-
Gonzalez J, Barreto GE (2016) Tibolone protects astrocytic cells from kappaB by palmitate in endothelial cells: a key role for NADPH
glucose deprivation through a mechanism involving estrogen recep- oxidase-derived superoxide in response to TLR4 activation.
tor beta and the upregulation of neuroglobin expression. Mol Cell Arterioscler Thromb Vasc Biol 29(9):1370–1375. https://doi.org/
Endocrinol 433:35–46. https://doi.org/10.1016/j.mce.2016.05.024 10.1161/ATVBAHA.109.188813
27. Baez E, Echeverria V, Cabezas R, Avila-Rodriguez M, Garcia- 43. Lambertucci RH, Hirabara SM, Silveira Ldos R, Levada-Pires AC,
Segura LM, Barreto GE (2016) Protection by neuroglobin expres- Curi R, Pithon-Curi TC (2008) Palmitate increases superoxide pro-
sion in brain pathologies. Front Neurol 7:146. https://doi.org/10. duction through mitochondrial electron transport chain and
3389/fneur.2016.00146 NADPH oxidase activity in skeletal muscle cells. J Cell Physiol
28. Fan R, Yu T, Lin JL, Ren GD, Li Y, Liao XX, Huang ZT, Jiang CH 216(3):796–804. https://doi.org/10.1002/jcp.21463
(2016) Remote ischemic preconditioning improves post resuscita- 44. Schonfeld P, Wojtczak L (2008) Fatty acids as modulators of the
tion cerebral function via overexpressing neuroglobin after cardiac cellular production of reactive oxygen species. Free Radic Biol Med
arrest in rats. Brain Res 1648 (Pt A):345–355. doi:https://doi.org/ 45(3):231–241. https://doi.org/10.1016/j.freeradbiomed.2008.04.
10.1016/j.brainres.2016.08.002 029
29. Amri F, Ghouili I, Amri M, Carrier A, Masmoudi-Kouki O (2016) 45. Kloosterboer HJ (2004) Tissue-selective effects of tibolone on the
Neuroglobin protects astroglial cells from hydrogen peroxide- breast. Maturitas 49(1):S5–S15. https://doi.org/10.1016/j.maturitas.
induced oxidative stress and apoptotic cell death. J Neurochem. 2004.06.022
https://doi.org/10.1111/jnc.13876 46. Reed MJ, Kloosterboer HJ (2004) Tibolone: a selective tissue es-
30. Wang Z, Liu D, Wang F, Liu S, Zhao S, Ling EA, Hao A (2012) trogenic activity regulator (STEAR). Maturitas 48(Suppl 1):S4–S6.
Saturated fatty acids activate microglia via toll-like receptor 4/NF- https://doi.org/10.1016/j.maturitas.2004.02.013
kappaB signalling. Br J Nutr 107(2):229–241. https://doi.org/10. 47. Wu SY, Chen YW, Tsai SF, Wu SN, Shih YH, Jiang-Shieh YF,
1017/S0007114511002868 Yang TT, Kuo YM (2016) Estrogen ameliorates microglial activa-
31. Blasi E, Barluzzi R, Bocchini V, Mazzolla R, Bistoni F (1990) tion by inhibiting the Kir2.1 inward-rectifier K(+) channel. Sci Rep
Immortalization of murine microglial cells by a v-raf/v-myc carry- 6:22864. doi:https://doi.org/10.1038/srep22864
ing retrovirus. J Neuroimmunol 27(2–3):229–237 48. Ishihara Y, Itoh K, Ishida A, Yamazaki T (2015) Selective estrogen-
32. Takuma K, Baba A, Matsuda T (2004) Astrocyte apoptosis: impli- receptor modulators suppress microglial activation and neuronal
cations for neuroprotection. Prog Neurobiol 72(2):111–127. https:// cell death via an estrogen receptor-dependent pathway. J Steroid
doi.org/10.1016/j.pneurobio.2004.02.001 Biochem Mol Biol 145:85–93. https://doi.org/10.1016/j.jsbmb.
33. Arevalo MA, Azcoitia I, Garcia-Segura LM (2015) The neuropro- 2014.10.002
tective actions of oestradiol and oestrogen receptors. Nat Rev 49. Yang L, Huang F, Mei J, Wang X, Zhang Q, Wang H, Xi M, You Z
Neurosci 16(1):17–29. https://doi.org/10.1038/nrn3856 (2017) Posttranscriptional control of PD-L1 expression by 17beta-
Mol Neurobiol

estradiol via PI3K/Akt signaling pathway in ER alpha-positive can- 65. Richardson TE, Yu AE, Wen Y, Yang SH, Simpkins JW (2012)
cer cell lines. Int J Gynecol Cancer 27(2):196–205. https://doi.org/ Estrogen prevents oxidative damage to the mitochondria in
10.1097/IGC.0000000000000875 Friedreich’s ataxia skin fibroblasts. PLoS One 7(4):e34600.
50. Skiljic D, Nilsson S, Petersen A, Karlsson JO, Behndig A, https://doi.org/10.1371/journal.pone.0034600
Kalaboukhova L, Zetterberg M (2016) Oestradiol levels and super- 66. Simpkins JW, Yang SH, Sarkar SN, Pearce V (2008) Estrogen
oxide dismutase activity in age-related cataract: a case-control actions on mitochondria–physiological and pathological implica-
study. BMC Ophthalmol 16(1):210. https://doi.org/10.1186/ tions. Mol Cell Endocrinol 290(1–2):51–59. https://doi.org/10.
s12886-016-0392-0 1016/j.mce.2008.04.013
51. Priyanka HP, Singh RV, Pratap UP, Thyaga Rajan S (2014) 67. Simpkins JW, Wang J, Wang X, Perez E, Prokai L, Dykens JA
Estrogen modulates beta2-adrenoceptor-induced cell-mediated (2005) Mitochondria play a central role in estrogen-induced neuro-
and inflammatory immune responses through ER-alpha involving protection. Curr Drug Targets CNS Neurol Disord 4(1):69–83
distinct intracellular signaling pathways, antioxidant enzymes, and 68. De Marinis E, Acaz-Fonseca E, Arevalo MA, Ascenzi P, Fiocchetti
nitric oxide. Cell Immunol 292(1–2):1–8. https://doi.org/10.1016/j. M, Marino M, Garcia-Segura LM (2013) 17beta-Oestradiol anti-
cellimm.2014.08.001 inflammatory effects in primary astrocytes require oestrogen recep-
52. Feng L, Li W, Liu Y, Jiang WD, Kuang SY, Jiang J, Tang L, Zhang tor beta-mediated neuroglobin up-regulation. J Neuroendocrinol
YA et al (2016) Protective role of phenylalanine on the ROS-induced 25(3):260–270. https://doi.org/10.1111/jne.12007
oxidative damage, apoptosis and tight junction damage via Nrf2, 69. Acaz-Fonseca E, Duran JC, Carrero P, Garcia-Segura LM, Arevalo
TOR and NF-kappaB signalling molecules in the gill of fish. Fish MA (2015) Sex differences in glia reactivity after cortical brain
Shellfish Immunol. https://doi.org/10.1016/j.fsi.2016.11.048 injury. Glia. https://doi.org/10.1002/glia.22867
53. Giatti S, Romano S, Pesaresi M, Cermenati G, Mitro N, Caruso D, 70. Sandhir R (2014) Neuroglobin increases brain fitness: commentary
Tetel MJ, Garcia-Segura LM et al (2015) Neuroactive steroids and to: BNeuroglobin overexpression improves sensorimotor outcomes
the peripheral nervous system: an update. Steroids 103:23–30. in a mouse model of traumatic brain injury^. Neurosci Lett 577:
https://doi.org/10.1016/j.steroids.2015.03.014 123–124. https://doi.org/10.1016/j.neulet.2014.06.005
54. Madan K, Bhardwaj P, Thareja S, Gupta SD, Saraya A (2006) 71. Jin K, Mao Y, Mao X, Xie L, Greenberg DA (2010) Neuroglobin
Oxidant stress and antioxidant status among patients with nonalco- expression in ischemic stroke. Stroke 41(3):557–559. https://doi.
holic fatty liver disease (NAFLD). J Clin Gastroenterol 40(10): org/10.1161/STROKEAHA.109.567149
930–935. https://doi.org/10.1097/01.mcg.0000212608.59090.08 72. De Marinis E, Ascenzi P, Pellegrini M, Galluzzo P, Bulzomi P,
55. Yuan L, Wu Y, Ren X, Liu Q, Wang J, Liu X (2014) Isoorientin Arevalo MA, Garcia-Segura LM, Marino M (2010) 17beta-
attenuates lipopolysaccharide-induced pro-inflammatory responses estradiol–a new modulator of neuroglobin levels in neurons: role
through down-regulation of ROS-related MAPK/NF-kappaB sig- in neuroprotection against H(2)O(2)-induced toxicity. Neurosignals
naling pathway in BV-2 microglia. Mol Cell Biochem 386(1–2): 18(4):223–235. https://doi.org/10.1159/000323906
153–165. https://doi.org/10.1007/s11010-013-1854-9 73. Frasor J, Weaver A, Pradhan M, Dai Y, Miller LD, Lin CY,
56. Baulieu EE, Robel P, Schumacher M (2001) Neurosteroids: begin- Stanculescu A (2009) Positive cross-talk between estrogen receptor
ning of the story. Int Rev Neurobiol 46:1–32 and NF-kappaB in breast cancer. Cancer Res 69(23):8918–8925.
57. Deshpande R, Khalili H, Pergolizzi RG, Michael SD, Chang MD https://doi.org/10.1158/0008-5472.CAN-09-2608
(1997) Estradiol down-regulates LPS-induced cytokine production 74. Heese K, Fiebich BL, Bauer J, Otten U (1997) Nerve growth factor
and NFkB activation in murine macrophages. Am J Reprod (NGF) expression in rat microglia is induced by adenosine A2a-
Immunol 38(1):46–54 receptors. Neurosci Lett 231(2):83–86
58. Simpkins JW, Yi KD, Yang SH, Dykens JA (2010) Mitochondrial 75. Leszek J, Barreto GE, Gasiorowski K, Koutsouraki E, Avila-
mechanisms of estrogen neuroprotection. Biochim Biophys Acta Rodrigues M, Aliev G (2016) Inflammatory mechanisms and oxi-
1800(10):1113–1120. https://doi.org/10.1016/j.bbagen.2009.11. dative stress as key factors responsible for progression of neurode-
013 generation: role of brain innate immune system. CNS Neurol
59. Cordeau P Jr, Lalancette-Hebert M, Weng YC, Kriz J (2016) Disord Drug Targets 15(3):329–336
Estrogen receptors alpha mediates postischemic inflammation in 76. Strle K, McCusker RH, Tran L, King A, Johnson RW, Freund GG,
chronically estrogen-deprived mice. Neurobiol Aging 40:50–60. Dantzer R, Kelley KW (2007) Novel activity of an anti-
https://doi.org/10.1016/j.neurobiolaging.2016.01.002 inflammatory cytokine: IL-10 prevents TNFalpha-induced resis-
60. Sukocheva OA, Yang Y, Gierthy JF (2009) Estrogen and proges- tance to IGF-I in myoblasts. J Neuroimmunol 188(1–2):48–55.
terone interactive effects in postconfluent MCF-7 cell culture. https://doi.org/10.1016/j.jneuroim.2007.05.003
Steroids 74(4–5):410–418. https://doi.org/10.1016/j.steroids.2008. 77. Valavanidis A, Vlachogianni T, Fiotakis C (2009) 8-hydroxy-2′ -
12.010 deoxyguanosine (8-OHdG): a critical biomarker of oxidative stress
61. Barreto GE, Santos-Galindo M, Garcia-Segura LM (2014) and carcinogenesis. J Environ Sci Health C Environ Carcinog
Selective estrogen receptor modulators regulate reactive microglia Ecotoxicol Rev 27(2):120–139. https://doi.org/10.1080/
after penetrating brain injury. Front Aging Neurosci 6:132. https:// 10590500902885684
doi.org/10.3389/fnagi.2014.00132 78. Bruce-Keller AJ, Keeling JL, Keller JN, Huang FF, Camondola S,
62. Wu WF, Tan XJ, Dai YB, Krishnan V, Warner M, Gustafsson JA Mattson MP (2000) Antiinflammatory effects of estrogen on
(2013) Targeting estrogen receptor beta in microglia and T cells to microglial activation. Endocrinology 141(10):3646–3656. https://
treat experimental autoimmune encephalomyelitis. Proc Natl Acad doi.org/10.1210/endo.141.10.7693
Sci U S A 110(9):3543–3548. https://doi.org/10.1073/pnas. 79. Fumagalli S, Perego C, Pischiutta F, Zanier ER, De Simoni MG
1300313110 (2015) The ischemic environment drives microglia and macrophage
63. Wood H (2011) Neuroimmunology: estrogen receptor ligands sup- function. Front Neurol 6:81. https://doi.org/10.3389/fneur.2015.
press inflammatory responses in astrocytes and microglia. Nat Rev 00081
Neurol 7(7):355. https://doi.org/10.1038/nrneurol.2011.87 80. Ganter S, Northoff H, Mannel D, Gebicke-Harter PJ (1992) Growth
64. Zhou Z, Zhou J, Du Y (2012) Estrogen receptor beta interacts and control of cultured microglia. J Neurosci Res 33(2):218–230.
colocalizes with HADHB in mitochondria. Biochem Biophys Res https://doi.org/10.1002/jnr.490330205
Commun 427(2):305–308. https://doi.org/10.1016/j.bbrc.2012.09. 81. Mizoguchi S, Mori K, Wang Z, Liu T, Funahashi Y, Sato F,
047 DeFranco DB, Yoshimura N et al (2017) Effects of estrogen
Mol Neurobiol

receptor beta stimulation in a rat model of non-bacterial prostatic dependent inflammation in brain endothelium without interfering
inflammation. Prostate 77(7):803–811. https://doi.org/10.1002/ with I kappa B degradation. Neuroreport 13(11):1469–1472
pros.23320 84. Acton QA (2012) Brain ischemia: new insights for the healthcare
82. Pozzi S, Benedusi V, Maggi A, Vegeto E (2006) Estrogen action in professional: 2012 edition: ScholarlyBrief. ScholarlyEditions,
neuroprotection and brain inflammation. Ann N Y Acad Sci 1089: Atlanta
302–323. https://doi.org/10.1196/annals.1386.035 85. Biswas DK, Singh S, Shi Q, Pardee AB, Iglehart JD (2005)
83. Galea E, Santizo R, Feinstein DL, Adamsom P, Greenwood J, Crossroads of estrogen receptor and NF-kappaB signaling. Sci
Koenig HM, Pelligrino DA (2002) Estrogen inhibits NF kappa B- STKE 2005(288):pe27. https://doi.org/10.1126/stke.2882005pe27

You might also like