ArticlePDF Available

Benzophenone-3 Impairs Autophagy, Alters Epigenetic Status, and Disrupts Retinoid X Receptor Signaling in Apoptotic Neuronal Cells

Authors:
  • Maj Institute of Pharmacology Polish Academy of Sciences

Abstract and Figures

Benzophenone-3 (BP-3) is the most widely used compound among UV filters for the prevention of photodegradation. Population studies have demonstrated that it penetrates through the skin and crosses the blood-brain barrier. However, little is known about the impact of BP-3 on the nervous system and its possible adverse effects on the developing brain. We demonstrated that the neurotoxic effects of BP-3 were accompanied by the induction of apoptosis, as evidenced by apoptosis-related caspase-3 activation and apoptotic body formation as well as the inhibition of autophagy, as determined by the downregulation of autophagy-related genes, decreased autophagosome formation, and reduced LC3B-to-LC3A ratio. In this study, we showed for the first time that the BP-3-induced apoptosis of neuronal cells is mediated via the stimulation of RXRα signaling and the attenuation of RXRβ/RXRγ signaling, as demonstrated using selective antagonist and specific siRNAs as well as by measuring the mRNA and protein expression levels of the receptors. This study also demonstrated that environmentally relevant concentrations of BP-3 were able to inhibit autophagy and disrupt the epigenetic status of neuronal cells, as evidenced by the inhibition of global DNA methylation as well as the reduction of histone deacetylases and histone acetyl transferases activity, which may increase the risks of neurodevelopmental abnormalities and/or neural degenerations.
This content is subject to copyright. Terms and conditions apply.
Benzophenone-3 Impairs Autophagy, Alters Epigenetic Status,
and Disrupts Retinoid X Receptor Signaling in Apoptotic
Neuronal Cells
Agnieszka Wnuk
1
&Joanna Rzemieniec
1
&Władysław Lasoń
1
&Wojciech Krzeptowski
2
&
Małgorzata Kajta
1
Received: 12 May 2017 /Accepted: 1 August 2017 /Published online: 16 August 2017
#The Author(s) 2017. This article is an open access publication
Abstract Benzophenone-3 (BP-3) is the most widely used
compound among UV filters for the prevention of
photodegradation. Population studies have demonstrated that
it penetrates through the skin and crosses the blood-brain bar-
rier. However, little is known about the impact of BP-3 on the
nervous system and its possible adverse effects on the devel-
oping brain. We demonstrated that the neurotoxic effects of
BP-3 were accompanied by the induction of apoptosis, as
evidenced by apoptosis-related caspase-3 activation and apo-
ptotic body formation as well as the inhibition of autophagy,
as determined by the downregulation of autophagy-related
genes, decreased autophagosome formation, and reduced
LC3B-to-LC3A ratio. In this study, we showed for the first
time that the BP-3-induced apoptosis of neuronal cells is me-
diated via the stimulation of RXRαsignaling and the attenu-
ation of RXRβ/RXRγsignaling, as demonstrated using selec-
tive antagonist and specific siRNAs as well as by measuring
the mRNA and protein expression levels of the receptors. This
study also demonstrated that environmentally relevant con-
centrations of BP-3 were able to inhibit autophagy and disrupt
the epigenetic status of neuronal cells, as evidenced by the
inhibition of global DNA methylation as well as the reduction
of histone deacetylases and histone acetyl transferases activity,
which may increase the risks of neurodevelopmental abnor-
malities and/or neural degenerations.
Keywords Benzophenone-3 .BP-3 .Retinoid X receptors .
RXR .Primary neuronal cell cultures .Autophagy
Introduction
Because of public anxiety about skin cancer caused by ultra-
violet light (UV), production and consumption of sunscreen
products are increasing. Nowadays, over 10,000 t of UV filters
are produced annually for the global market [1]. Chemical UV
filters are generally used as a mixture since none of the com-
pounds used individually get sufficient protection against UV.
Among the filters, benzophenones (BPs) are the primary
ingredients in the organic UV filter family. Benzophenone-3
(2-hydroxy-4-methoxybenzophenone, oxybenzone, 2OH-4
MeO-BP or BP-3) is the most widely used compound among
BPs for the skin prevention against photodegradation [2].
Human studies have demonstrated that after topical applica-
tion, BP-3 is absorbed through the skin, partially metabolized,
and is excreted in the urine. BP-3 was detected in almost all
(8096%) urine samples collected from the general population
in the USA [3]; pregnant women in France [4,5]; and Danish
mothers and their children, adolescents, young men, and preg-
nant women [6,7]. BP-3 can be detected in the serum and
urine of adult volunteers shortly after dermal application,
proving that it can pass through the skin into the body [8]. It
has been estimated that 10% of the applied dermal BP-3 pen-
etrates skin into systemic circulation [9].
It is extremely disturbing that BP-3 has been detected in a
large proportion of milk samples indicating that breastfed
babies are exposed to BP-3 [10]. A recent study showed that
maternal exposure to BP-3 is strongly associated with the onset
of Hirschsprungs disease in offspring [11]. However, data on
the effects of BP-3 on the nervous system are scarce. Espe-
cially little is known about the impact of BP-3 on individual
*Małgorzata Kajta
kajta@if-pan.krakow.pl
1
Department of Experimental Neuroendocrinology, Institute of
Pharmacology, Polish Academy of Sciences, Smetna Street 12,
31-343 Krakow, Poland
2
Department of Cell Biology and Imaging, Institute of Zoology,
Jagiellonian University, Gronostajowa Street 9,
30-387 Krakow, Poland
Mol Neurobiol (2018) 55:50595074
DOI 10.1007/s12035-017-0704-2
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
receptors that are strongly associated with brain development
such as retinoid X receptors. The only data on the apoptotic and
neurotoxic effects of BP-3 on the neural cells come from SH-
SY5Y neuroblastoma cells and our recently published original
paper [12,13]. BP-3 has been reported to act as endocrine
disrupting chemicals (EDCs). At present, it is only known that
BP-3 can weakly agonize estrogen signaling and strongly an-
tagonize androgen-related pathways [1416]. An association
between BP-3 exposure and the estrogen-related disease endo-
metriosis has been found [17]. Recently, retinoid X receptors
have been postulated to be a target for EDCs.
The retinoid X receptor (RXR) is a type of nuclear receptor
family that is encoded by three genes: RXRα,RXRβ,and
RXRγ[18]. RXRs heterodimerize with one-third of the 48
human nuclear receptor superfamily members [19]. For most
of them, RXR is an obligatory partner for DNA binding and
transcriptional regulation. In addition, RXRαis able to form
homodimers and homotetramers, which is suggestive of the
self-regulation of specific RXRαsignaling pathways [20].
The diversity of RXRs suggests that they play critical roles
in a wide range of cellular pathways. Recent studies have
shown the prominence of RXR signaling in developing inner-
vation and myelination in health and disease of the central
nervous system [21]. Current studies in our research group
have shown the involvement of RXRs in the effects of EDCs
(specifically the pesticide dichlorodiphenyldichloroethylene
(DDE) and nonylphenol) [2224].
One of the most important ways of regulating gene expres-
sion is the remodeling of chromatin, including post-
translational modifications of histones and DNA methylation.
It has been postulated that low doses of EDCs may cause
epigenetic changes, such as the incomplete methylation of
specific gene regions in the young brain [25,26]. Histone
post-translational modifications include the most studied
modificationsthe acetylation of histones by histone acetyl
transferases (HATs) and the removal of acetyl groups from
histones by histone deacetylases (HDACs). These processes
play important roles in cognition as well as psychiatric and
neurologic diseases such as Alzheimer's disease, Huntingtons
disease, traumatic brain injury, post-traumatic stress disorder,
stress, depression, and addiction [27].
Autophagy is a process that is mainly responsible for elim-
inating the cells or keeping them alive, even in conditions
deprived of trophic factors. Autophagy is postulated to play a
housekeeping role in removing abnormal proteins or clearing
damaged organelles. The formation of autophagosomes de-
pends on several core Atg proteins, such as the following:
ULK1 complex, Beclin1:Vps34/Atg14L complex, and LC3
conjugation systems. During the process of autophagy, LC3
protein is cleaved by Atg4 to LC3A which next is modified
by ubiquitin-like systems to produce LC3B. Thus, LC3A and
LC3B are present in autophagosomes; both the ratio of LC3B
to LC3A and the amount of LC3B only can be used to estimate
the level of autophagy. Recent studies have proposed that gen-
erally autophagy is a survival mechanism, although its dysreg-
ulation may lead to non-apoptotic cell death [28].
The present study aimed to investigate the neurotoxic and
apoptotic effects of BP-3 and the impact of this chemical on the
expression and function of RXRs, including RXRα,RXRβ,
and RXRγ. Neurotoxicity was estimated by measuring lactate
dehydrogenase (LDH) release, which was complemented by
an assessment of caspase-3 activity. These data were supported
by Hoechst 33342/calcein acetoxymethyl (AM) staining,
which allowed for the visualization of apoptotic nuclei and cell
survival. The involvement of RXRs in the actions of BP-3 was
verified using selective antagonist and agonist as well as spe-
cific siRNAs. The levels of receptor mRNAs and proteins were
measured with qPCR, western blot, and enzyme-linked immu-
nosorbent assay (ELISA), and the cellular distributions of the
receptors were demonstrated using a confocal microscope. The
process of autophagy was assessed by measuring the expres-
sion of autophagy-specific genes using microarray analysis
and autophagosome detection, and the concentrations of
autophagy-selective proteins were measured by ELISAs.
Results regarding epigenetic modifications such as histone
post-translational modifications and DNA methylation were
complemented by an assessment of HAT and HDAC activity
and the measurement of global DNA methylation.
Materials and Methods
Materials
B27 and neurobasal media were obtained from Gibco (Grand
Island, NY, USA). L-glutamine, fetal bovine serum (FBS), N-
acetyl-Asp-Glu-Val-Asp-p-nitro-anilide (Ac-DEVD-pNA), di-
methyl sulfoxide (DMSO), HEPES, CHAPS, mouse monoclo-
nal anti-MAP2 antibody, ammonium persulfate, TEMED,
TRIZMA base, Tween 20, DL-dithiothreitol, Nonidet NP-40,
sodium deoxycholate, protease inhibitor (EDTA-free),
bromophenol blue, 2,7-dichlorofluorescein diacetate, RIPA
buffer, the Imprint Methylated DNA Quantification Kit, the
Histone Deacetylase Assay Kit, the Histone Acetyltransferase
Activity Assay Kit, the Autophagy Assay Kit, protease inhibitor
cocktail for mammalian tissues, and poly-ornithine were obtain-
ed from Sigma-Aldrich (St. Louis, MO, USA). Bradford re-
agent, SDS, 30% acrylamide, 0.5 M Tris-HCl buffer, 1.5 M
Tris-HCl gel buffer, and Laemmli sample buffer were from
Bio-Rad Laboratories (Munchen, Germany). DHA and HX
531 were from Tocris Bioscience (Minneapolis, MN, USA).
2-Mercaptoethanol was from Carl Roth GmbH + Co. KG
(Karlsruhe, Germany). Immobilon-P membranes were pur-
chased from Millipore (Bedford, MA, USA). Alexa 488-
conjugated anti-goat IgG, calcein AM, and Hoechst 33342 were
purchased from Molecular Probes (Eugene, OR, USA). Cy3-
5060 Mol Neurobiol (2018) 55:50595074
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
conjugated anti-rabbit IgG and Cy5-conjugated anti-mouse
were obtained from Jackson ImmunoResearch, Inc. (West
Grove, PA, USA). The cytotoxicity detection kit and BM
chemiluminescence western blotting substrate (POD) were pur-
chased from Roche Diagnostics GmbH (Mannheim, Germany).
ELISA kits for RXRα,RXRβ,RXRγ, LC3A, and LC3B were
purchased from Shanghai Sunred Biological Technology Co.
(Sunred, China). The culture dishes were obtained from TPP
Techno Plastic Products AG (Trasadingen, Switzerland). The
rabbit polyclonal anti-RXRαantibody (sc-774), mouse mono-
clonal anti-RXRβantibody (sc-56869), mouse monoclonal
anti-RXRγantibody (sc-514134), mouse monoclonal anti-β-
actin antibody (sc-47778), as well as RXRαsiRNA (sc-
36448), RXRβsiRNA (sc-36446) and RXRγsiRNA (sc-
38879) were purchased from Santa Cruz Biotechnology, Inc.
(Santa Cruz, CA, USA). AllStars Negative Control siRNA AF
488, the RNeasy Mini Kit, RT
2
First Strand Kit, and RT
2
Profiler PCR Autophagy Array were obtained from Qiagen
(Valencia, CA, USA). INTERFERin was obtained from
PolyPlus Transfection (Illkirch, France), and the High
Capacity cDNA-Reverse Transcription Kit, the TaqMan Gene
Expression Master Mix, and TaqMan probes for specific genes
encoding hypoxanthine phosphoribosyltransferase coding gene
(Hprt), Rxrα,Rxrβ,andRxrγwere obtained from Life
Technologies Applied Biosystems (Foster City, CA, USA).
Quick-gDNAMicroPrep was obtained from Zymo
Research (Irvine, CA, USA).
Primary Neocortical Cell Cultures
Neocortical tissue for primary cultures was prepared from
Swiss mouse embryos (Charles River, Germany) at 15
17 days of gestation and cultured as previously described
[22,29]. All procedures were performed in accordance with
the National Institutes of Health Guidelines for the Care and
Use of Laboratory Animals and were approved by the
Bioethics Commission in compliance with Polish Law (21
August 1997). Animal care followed official governmental
guidelines, and all efforts were made to minimize suffering
as well as the number of animals used. The cells were
suspended in estrogen-free neurobasal medium supplemented
with B27 on poly-ornithine (0.01 mg/ml)-coated multi-well
plates at a density of 2.0 × 10
5
cells per cm
2
. The cultures
were maintained at 37 °C in a humidified atmosphere contain-
ing 5% CO
2
for 7 days in vitro (DIV) prior to experimentation.
The number of astrocytes, as determined by the content of
intermediate filament glial fibrillary acidic protein (GFAP),
did not exceed 10% for all cultures [22,30].
Treatment
Primary neuronal cell cultures were exposed to BP-3 (10
100 μM) for 6 or 24 h. The involvement of RXR signaling
in BP-3-induced effects was verified with the high-
affinity RXR antagonist HX 531 (0.1 μM) and the
RXR agonist DHA (1 μM) as previously described
[22]. Specific ligands were added to the culture media
4560 min before BP-3. To avoid nonspecific effects in
our study, agonist and antagonist of RXRs were used at
concentrations that did not affect the levels of caspase-3
activity or LDH release. All the compounds were orig-
inally dissolved in DMSO and were then further diluted
in culture medium to maintain DMSO concentrations
below 0.1%. The control cultures were supplemented
with DMSO in concentrations that were equal to those
used in the experimental groups.
Identification of Apoptotic Cells
Apoptotic cells were detected via Hoechst 33342 stain-
ing at 24 h after the initial treatment as previously de-
scribed [22,29]. Neocortical cells that were cultured on
glass coverslips were washed with 10 mM phosphate-
buffered saline (PBS) and stained with Hoechst 33342
(0.6 mg/ml) at room temperature (RT) for 5 min. The
cells containing bright blue fragmented nuclei, which
was indicative of condensed chromatin, were identified
as apoptotic cells. Qualitative analysis was performed
using a fluorescence microscope (NIKON Eclipse 80i,
NIKON Instruments Inc., Melville, New York, USA)
equipped with a camera with the BCAM Viewer©
Basler AG software. The level of cellular fluorescence
from fluorescence microscopy images was determined
using ImageJ software. To calculate the corrected total
cell fluorescence (CTCF), the following equation was
used: CTCF = Integrated density (Area of selected
cell × Mean fluorescence of background).
Staining with Calcein AM
Intracellular esterase activity in the neocortical cultures
was measured by calcein AM staining at 24 h after the
initial treatment with BP-3 as previously described [22,
29]. To avoid the esterase activity present in the growth
media, the cells were washed with PBS and incubated in
2μM calcein AM in PBS at RT for 10 min. The cells
displaying bright green cytoplasm were identified as live
cells. Fluorescence intensity was monitored at Ex/Em 494/
520 nm using a fluorescence microscope (NIKON Eclipse
80i, NIKON Instruments Inc., Melville, New York, USA)
equipped with a camera with the BCAM Viewer© Basler
AG software. The level of cellular fluorescence from fluo-
rescence microscopy images was determined using ImageJ
software. To calculate the CTCF, the following equation
was used: CTCF = Integrated density (Area of selected
cell × Mean fluorescence of background).
Mol Neurobiol (2018) 55:50595074 5061
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Assessment of Caspase-3 Activity
Caspase-3 activity was determined according to the protocol de-
scribed by Nicholson (1995) using samples treated for 6 or 24 h
with BP-3 alone or in combination with the test compounds. The
assessment of caspase-3 activity was performed as previously
described [22,30,31]. Cell lysates from neocortical cultures were
incubatedat3CusingAc-DEVD-pNA, a colorimetric sub-
strate that is preferentially cleaved by caspase-3. The levels of p-
nitroanilide were continuously monitored for 60 min using a
Multimode Microplate Reader Infinite M200PRO (Tecan,
Mannedorf, Switzerland). The data were analyzed using the
Magellan software, normalized to the absorbance of vehicle-
treated cells, and expressed as a percentage of control ± SEM
from three to four independent experiments. The absorbance of
blanks, which acted as our enzyme-less control, was subtracted
from each value.
Measurement of Lactate Dehydrogenase Activity
To quantify cell death, lactate dehydrogenase (LDH) that was
released from damaged cells into the cell culture media was
measured 6 or 24 h after treatment with BP-3. LDH release
was measured as previously described [22,32]. Cell-free su-
pernatants from neocortical cultures were collected from each
well and incubated at room temperature for 30 to 60 min with
the appropriate reagent mixture according to the manufac-
turers instructions (Cytotoxicity Detection Kit) depending
on the reaction progress. The intensity of the red color that
formed in the assay was measured at a wavelength of 490 nm
(Infinite M200pro microplate reader, Tecan Mannedorf,
Switzerland) and was proportional to both LDH activity as
well as the number of damaged cells. The data were analyzed
using the Magellan software, normalized to the color intensity
from vehicle-treated cells (100%), and expressed as a percent-
age of the control value from three to four independent exper-
iments. Theabsorbance of blanks, which acted as ourenzyme-
less control, was subtracted from each value.
Silencing of RXRα,RXRβ, and RXRγ
Specific siRNAs were used to inhibit RXRα,RXRβ,and
RXRγexpression in neocortical cells. Each siRNA was
applied separately for 6 h at 50 nM in antibiotic-free me-
dium containing the siRNA transfection reagent
INTERFERinas previously described [22]. After trans-
fection,theculturemediawerechanged,andthecellswere
incubated for 12 h before starting the experiment. Positive
and negative siRNAs containing a scrambled sequence that
did not lead to the specific degradation of any known cel-
lular mRNA were used as controls. The effectiveness of
mRNA silencing was verified through the measurement
of specific mRNAs using qPCR.
qPCR Analysis of mRNAs Encoding the Receptors Rxrα,
Rxrβ,andRxrγ
Total RNA was extracted from neocortical cells that were cul-
tured for 7 DIV (approx. 1.5 × 10
6
cells per sample) using the
RNeasy Mini Kit (Qiagen, Valencia, CA) according to the man-
ufacturers instructions. The quantity of RNA was spectropho-
tometrically determined at 260 and 260/280 nm (ND/1000 UV/
Vis; Thermo Fisher NanoDrop, USA). Two-step real-time
quantitative polymerase chain reaction (qPCR) was performed
as previously described [22]. Both the reverse transcription re-
action and qPCR were run on a CFX96 Real-Time System
(BioRad, USA). The products of the reverse transcription reac-
tion were amplified using TaqMan Gene Expression Master
Mix containing TaqMan primer probes specific to the genes
encoding Hprt,Rxrα,Rxrβ,andRxrγ.Amplification was per-
formed in a total volume of 20 μl containing 10 μlofTaqMan
Gene Expression Master Mix and 1.0 μl of reverse transcription
product as the PCR template. A standard qPCR procedure was
utilized: 2 min at 50 °C and 10 min at 95 °C followed by 40
cycles of 15 s at 95 °C and 1 min at 60 °C. The threshold value
(Ct) for each sample was set during the exponential phase, and
the delta Ct method was used for data analysis. Hprt was used
as a reference gene.
Mouse Autophagy RT
2
Profiler PCR Array
Total RNA was extracted from neocortical cells cultured for
7 DIV (approx. 1.5 × 10
6
cells per sample) using the RNeasy
Mini Kit (Qiagen, Valencia, CA) according to the manufac-
turers instructions. The quantity of RNA was spectrophoto-
metrically determined at 260 and 260/280 nm (ND/1000 UV/
Vis; Thermo Fisher NanoDrop, USA). A total of 1 μgof
mRNA was reverse-transcribed to cDNA using the RT
2
First
Strand Kit (Qiagen, Valencia, CA) and suspended in a final
volume of 20 μl as previously described [13]. Each cDNAwas
prepared for further use in qPCR. To analyze the signaling
pathway, the RT
2
ProfilerPCR Array System (Qiagen,
Valencia, CA) was used according to the manufacturerspro-
tocol. The Ct values for all wells were exported to a blank
Excel spreadsheet and were analyzed with the Web-based
software (www.SABiosciences.com/pcrarraydataanalysis.
php).
Western Blot Analy si s
The cells exposed to BP-3 for 24 h were lysed in ice-cold RIPA
lysis buffer containing a protease inhibitor cocktail. The lysates
were sonicated and centrifuged at 15,000×g for 20 min at 4 °C.
The protein concentrations in the supernatants were determined
using Bradford reagent (Bio-Rad Protein Assay) with bovine se-
rum albumin (BSA) as the standard. Samples containing 40 μgof
total protein were reconstituted in the appropriate amount of
5062 Mol Neurobiol (2018) 55:50595074
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
sample buffer comprised of 125 mM Tris, pH 6.8, 4% SDS, 25%
glycerol, 4 mM EDTA, 20 mM DTT, and 0.01% bromophenol
blue, denatured, and separated on a 7.5% SDS-polyacrylamide
gel using a Bio-Rad Mini-Protean II Electrophoresis Cell as pre-
viously described [22,33,34]. After electrophoretic separation,
the proteins were electrotransferred to PVDF membranes
(Millipore, Bedford, MA, USA) using the Bio-Rad Mini Trans-
Blot apparatus. Following the transfer, the membranes were
washed, and the nonspecific binding sites were blocked with
5% dried milk and 0.2% Tween-20 in 0.02 M Tris-buffered saline
(TBS) for 2 h with shaking. The membranes were incubated
overnight (at 4 °C) with one of the following primary antibodies
(Santa Cruz Biotechnology) diluted in TBS/Tween: anti-RXRα
rabbit polyclonal antibody (diluted 1:150), anti-RXRβmouse
monoclonal antibody (diluted 1:100), anti-RXRγmouse mono-
clonal antibody (diluted 1:100), or anti-β-actin mouse monoclo-
nal antibody (diluted 1:3000). The signals were developed by
chemiluminescence (ECL) using BM Chemiluminescence
Blotting Substrate (Roche Diagnostics GmBH) and visualized
using a Luminescent Image Analyzer Fuji-Las 4000 (Fuji,
Japan). Immunoreactive bands were quantified using a
MultiGauge V3.0 image analyzer.
Enzyme-Linked Immunosorbent Assays for RXRα,
RXRβ, and RXRγ
The levels of RXRα,RXRβ,RXRγ, LC3A, and LC3B were
determined in neocortical cells 24 h after treatment with BP-3
as previously described [22]. Specific detection of these pro-
teins was achieved using ELISAs and the quantitative sandwich
enzyme immunoassay technique. A 96-well plate was precoat-
ed with monoclonal antibodies that were specific for RXRα,
RXRβ,RXRγ, LC3A, and LC3B. The standards and non-
denatured cell extracts were added to the wells with biotin-
conjugated polyclonal antibodies specific for RXRα,RXRβ,
RXRγ, LC3A, and LC3B. Therefore, all native RXRα,RXRβ,
RXRγ, LC3A, and LC3B proteins were captured using the
immobilized antibodies. The plates were washed to remove
any unbound substances, and horseradish peroxidase-
conjugated avidin was added to interact with the biotin bound
to RXRα,RXRβ,RXRγ, LC3A, and LC3B. After washing,
the substrate solution was added to the wells. The enzymatic
reaction yielded a blue product. The absorbance was measured
at 450 nm and was proportional to the amount of RXRα,
RXRβ,RXRγ, LC3A, and LC3B in the sample. The protein
concentration was determined in each sample using Bradford
reagentBio-Rad Protein Assay [22,35].
Immunofluorescence Labeling of RXRα,RXRβ,
and RXRγand Confocal Microscopy
For immunofluorescence detection of RXRα,RXRβ,and
RXRγ, neocortical cells were cultured on glass coverslips
and subjected to immunofluorescence double-labeling as
previously described [22,36].After1hofincubationina
blocking buffer (5% normal donkey serum and 0.3%
TritonX-100in0.01MPBS),thecellsweretreatedfor
24 h (at 4 °C) using four primary antibodies: rabbit poly-
clonal anti-RXRαantibody (1:50), mouse monoclonal
anti-RXRβantibody (1:50), mouse monoclonal anti-
RXRγantibody (1:50), and anti-MAP2 mouse monoclo-
nal antibody (1:100) followed by a 24-h incubation in a
mixture of secondary antibodies, including Cy3-
conjugated anti-rabbit IgG (1:300) and Cy5-conjugated
anti-mouse IgG (1:300). The samples were subsequently
washed, mounted, coverslipped, and analyzed using an
LSM510 META, Axiovert 200M confocal laser scanning
microscope (Carl Zeiss MicroImaging GmbH, Jena,
Germany) under a Plan-Neofluor 40×/1.3 Oil DIC objec-
tive. A He/Ne laser and an argon laser, with two laser
lines emitting at 514 and 633 nm, were used to excite
the Cy3-, and Cy5-conjugated antibodies, respectively.
The fluorescence signal was enhanced after combining
four scans per line. A pinhole value of 1 airy unit was
used to obtain flat images.
Measurement of Global DNA Methylation
Genomic DNA was extracted from neocortical tissues
using the Quick-gDNAMicroPrep kit (Zymo
Research, Irvine, CA) according to the manufacturersin-
structions. The quantity of DNA was spectrophotometri-
cally determined at 260 and 260/280 nm (ND/1000 UV/
Vis; Thermo Fisher NanoDrop, USA). Global DNA meth-
ylation changes were measured in neocortical cells at 24 h
after treatment using a specific ELISA-based kit
(Imprint® Methylated DNA QuantificationSigma-
Aldrich; St. Louis, MO, USA) as previously described
[22]. This kit contained all the reagents required to deter-
mine the relative levels of methylated DNA. The methyl-
ated DNA was detected using the capture and detection
antibodies and quantified colorimetrically using an
Infinite M200pro microplate reader (Tecan, Austria). The
amount of methylated DNA present in the sample was
proportional to the absorbance measured.
Detection of Autophagosomes
Cultured cells on 96-well plates were treated according
to the manufacturers instructions. The Autophagy
Assay kit provided a simple and direct procedure for
measuring autophagy in a variety of cell types using a
proprietary fluorescent autophagosome marker
(λ
ex
=333/λ
em
= 518 nm). The autophagosomes were
detected using an Infinite M200pro microplate reader
(Tecan, Austria).
Mol Neurobiol (2018) 55:50595074 5063
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Measurement of HDAC and HAT Activity
The HDAC and HAT activities were detected using the Histone
Deacetylase Assay Kit and the Histone Acetyltransferase
Activity Fluorometric Assay Kit (Sigma-Aldrich, St. Louis,
MO, USA) according to the manufacturers instructions.
Regarding HDAC kit, the measured fluorescence at
λ
ex
=365nm/λ
em
= 460 nm was proportional to the
deacetylation activity. In the HAT assay, the generated product
of histone acetyltransferase activity was detected fluorimetrically
at λ
ex
= 535/λ
em
= 587 nm. The kit included an active nuclear
extract to be used as a positive control. The abovementioned
assays provided positive and negative controls.
Data Analysis
Statistical tests were performed on raw data that were expressed
as the mean arbitrary absorbance or as the fluorescence units
per well containing 50,000 cells (measurements of caspase-3,
LDH, autophagosomes; the fluorescence units per 1.5 million
cells (qPCR, global DNA methylation, and HDAC and HAT
activity); the mean optical density per 40 μg of protein (western
blotting); or picograms of RXRα,RXRβ,RXRγ,LC3A,and
LC3B per micrograms of total protein (ELISA). Statistical anal-
ysis of cellular fluorescence related to Hoechst 33342 and
calcein AM staining was performed on CTCF data using 40
counts per image. One-way analysis of variance (ANOVA) was
preceded by the Levenes test of homogeneity of variances and
was used to determine overall significance. Differences be-
tween the control and experimental groups were assessed using
a post hoc NewmanKeuls test, and significant differences
were designated as
*
p< 0.05,
**
p<0.01,and
***
p<0.001
versus control cultures;
#
p<0.05,
##
p<0.01,and
###
p< 0.001 versus the cultures exposed to BP-3; and
$
p<0.05and
$$$
p< 0.001 versus the siRNA-transfected control
cultures. The results were expressed as the mean ± SEM of
three to four independent experiments. The number of repli-
cates in each experiment ranged from 2 to 3, except for the
measurements of caspase-3 activity and LDH release, which
contained five to eight replicates. To compare the effects of
BP-3 in different treatment paradigms, the results for the cas-
pase-3, LDH, ELISA, and western blot analyses were presented
as a percentage of the control.
Results
Effects of BP-3 on Caspase-3 Activity and LDH Release
in Neocortical Cultures at 7 DIV
In neocortical cultures at 7 DIV, BP-3 (25100 μM) induced
an increase in caspase-3 levels to 170% of the control level at
6 h, which were further enhanced to 196% at 24-h post-
treatment (Fig. 1a). In these cells, LDH release from neocor-
tical cells increased in a time-dependent manner to 150180%
of the control value at 6 h and to 200290% at 24 h (Fig. 1b).
Effects of BP-3 Alone or in Combination with HX 531
on Hoechst 33342 and Calcein AM Staining in Neocortical
Cultures
In the present study, a 24-h exposure to BP-3 (25 μM) was
necessary to develop an apoptotic morphology in cell nuclei.
Apoptotic cells were detected by Hoechst 33342 staining as for-
mation of bright blue fragmented nuclei containing condensed
chromatin (Fig. 2). Furthermore, treatment with BP-3 reduced
the density of calcein AM-stained living cells at 7 DIV, as indi-
cated by the decreased number of cells exhibiting light-colored
cytoplasm. Co-treatment with RXR antagonist-HX 531
(0.1 μM) inhibited the BP-3-induced effects. Quantitative analy-
sis of relevant fluorescence signals showed that at 7 DIV, 25 μM
BP-3 caused an increase in formation of condensed chromatin by
386% of the control level and reduced number of live cells by
56%. Treatment with HX 531 (0.1 μM) inhibited the effect of
BP-3 in respect to the apoptotic fragmentation of cell nuclei by
231% and enhanced cell viability by 35% (Fig. 2).
Effects of BP-3 Alone or in Combination with DHA
and HX 531 on BP-3-Induced Caspase-3 Activity
and LDH Release in Neocortical Cultures
Neocortical cultures exposed to BP-3 (25 μM) for 24-h caused
a greater than 50% increase in caspase-3 activity in the neu-
ronal cells. Co-treatment with the selective RXR agonist DHA
(1 μM) did not change the effect of BP-3 on caspase-3 activity.
The RXR antagonist HX 531 (0.1 μM) inhibited the BP-3
(25 μM)-induced caspase-3 activity by 40% (Fig. 3a).
The selective RXR agonist DHA (1 μM) did not affect BP-
3-induced LDH release. However, the high-affinity RXR an-
tagonist HX 531 (0.1 μM) diminished BP-3-induced LDH
release by 25% (Fig. 3b).
Effect of BP-3 on the mRNA Levels of Rxrα,Rxrβ,
and Rxrγ
According to our data, treatment with BP-3 (25 μM) affected
the mRNA levels of Rxrα,Rxrβ,andRxrγ. A 3-h exposure of
the neocortical cultures to BP-3 caused a 25% decrease in Rxrβ
and a 55% decrease in Rxrγbut caused a 100% increase in
RxrαmRNA compared with the control (Fig. 4a). The pattern
of mRNA expression was changed after prolonged exposure to
BP-3. After 6 h of treatment, BP-3 decreased the mRNA ex-
pression level of Rxrα(26%) but did not change the expression
level of Rxrβin neocortical cells (Fig. 4b). After 24 h of expo-
sure, BP-3 did not change the mRNA expression levels of any
Rxrs (Fig. 4c). These data were normalized to Hprt.
5064 Mol Neurobiol (2018) 55:50595074
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
control
25 μM BP-3
Hoechst 33342 calcein AM
25 μM BP-3
+ HX 531
Corrected total cell
fluorescence
(CTCF)
control 25 μM BP-3 25 μM BP-3
+ HX 531
Hoechst 33342 0.153 (100 %) 0.591 (386 %) 0.238 (155 %)
calcein AM 49.328 (100 %) 21.609 (44 %) 38.802 (79%)
***
***
*
*
Fig. 2 Influence of BP-3
(25 μM) and HX 531 (0.1 μM) on
Hoechst 33342 (first column)and
calcein AM (second column)
staining in mouse neocortical
cultures at 7 DIV, examined 24 h
post-treatment. Cells with bright
fragmented nuclei with
condensed chromatin were
identified as cells undergoing
apoptosis, whereas cells with
light-colored cytoplasm were
identified as live cells. Statistical
analysis of relevant fluorescence
signals was performed on CTCF
data using 40 counts per image.
*
p<0.05and
***
p< 0.001 versus
control cultures
0
50
100
150
200
250
caspase-3 activity
[% of control]
neocortical neurons 6h
24h
*** ***
*** ***
***
***
*
control
0
50
100
150
200
250
300
350
10 μM 25 μM 50 μM 75 μM 100 μM
control 10 μM 25 μM 50 μM 75 μM 100 μM
LDH release
[% of control]
neocortical neurons 6h
24h
***
*** ***
***
***
***
**
b
a
Fig. 1 Time-course effects of
BP-3 (10, 25, 50, 75, and
100 μM) on caspase-3 activity (a)
and LDH release (b)inprimary
cultures of mouse neocortical
cells at 7 DIV. The cells were
treated with BP-3 for 6 and 24 h.
The results are presented as a
percentage of the control. Each
bar represents the mean of three
to four independent
experiments ± SEM. The number
of replicates in each experiment
ranged from 5 to 8.
*
p<0.05,
**
p<0.01,and
***
p<0.001
versus control cultures
Mol Neurobiol (2018) 55:50595074 5065
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
0
50
100
150
200
250
25 μM BP-3
caspase-3 acttivity
[% of control]
25 μM BP-3
+DHA
+HX 531
***
###
a
control
0
50
100
150
200
250
25 μM BP-3
LDH release
[% of control]
25 μM BP-3
+ DHA
+HX 531
control
***
##
b
Fig. 3 Impact of the RXR
agonist and antagonist on BP-3-
induced caspase-3 activity (a)and
LDH release (b)inneocortical
cultures at 7 DIV. The primary
neocortical cultures were treated
with BP-3 (25 μM) for 24 h. The
results were normalized to the
absorbance of vehicle-treated
cells and are expressed as a
percentage of control. Each bar
represents the mean of three to
four independent
experiments ± SEM. The number
of replicates in each experiment
ranged from 5 to 8.
***
p<0.001
versus control cultures;
##
p<0.01
and
###
p< 0.001 versus the
cultures exposed to BP-3
0
0.5
1
1.5
2
2.5
RxrRxrRxr
RxrRxrRxr
RxrRxrRxr
mRNA
[folds Hprt normalized]
3h control
25 μM BP3
***
***
**
a
0
0.5
1
1.5
2
2.5
mRNA
[folds Hprt normalized]
6h control
25 μM BP3
***
**
b
0
0.5
1
1.5
2
2.5
mRNA
[folds Hprt normalized]
24h control
25 μM BP3
c
Fig. 4 Effect of BP-3 (25 μM) on
the mRNA expression levels of
Rxrα,Rxrβ,andRxrγin
neocortical cultures at 7 DIV.
Each bar represents the
mean ± SEM of three independent
experiments. The number of
replicates for each experiment
ranged from 2 to 3.
**
p<0.01and
***
p< 0.001 versus control
cultures
5066 Mol Neurobiol (2018) 55:50595074
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Effects of BP-3 on the Protein Expression Levels of RXRα,
RXRβ, and RXRγin Mouse Neocortical Cells
A 24-h exposure to BP-3 was necessary to detect changes
in the protein levels of the receptors. In the cultures treat-
ed with BP-3, RXRαexpression enhanced and reached
1.43 pg/μg of total protein (134% of the control). In cul-
tures exposed to BP-3 (25 μM) for 24 h, the concentration
of RXRβwas 0.76 pg/μg of total protein, and it was 56%
less than that in control cultures. In the cultures exposed
to BP-3, the level of RXRγreached 0.38 pg/μg, which
was 49% less than controls (Fig. 5a, b).
Western blot analysis determined the relative protein
expression levels of RXRα,RXRβ,andRXRγin mouse
neocortical cells at 7 DIV. Exposure to BP-3 (25 μM) for
24 h decreased the relative RXRβand RXRγprotein
levels by 61 and 56%, respectively. Treatment with BP-3
(25 μM) increased the relative RXRαprotein level by
49% (Fig. 5c, d).
Effect of BP-3 on the Distribution of RXRα,RXRβ,
RXRγ, and MAP2 Staining in Neocortical Cells
Immunofluorescence labeling was performed in parallel with
the measurements of receptor protein levels. Confocal micros-
copy revealed that RXRα,RXRβ, and RXRγwere localized
to neocortical cells at 7 DIV. A 24-h exposure to BP-3
(25 μM) increased RXRαstaining but reduced RXRβ-and
RXRγ-specific immunofluorescence. MAP2 staining con-
firmed the neural localization of the receptors and revealed
the BP-3-induced inhibition of neurite outgrowth (Fig. 6).
Influence of BP-3 on Caspase-3 Activity and LDH Release
in Neocortical Cells Transfected with RXRα,RXRβ,
and RXRγsiRNAs
A24-hexposuretoBP-3(25μM) only slightly reduced
caspase-3 activity and LDH release in the RXRβand RXRγ
siRNA-transfected cells, suggesting that the transfected cells
relative density of
bands [% control]
control BP-3
(25 μM)
RXRα 100±5.7 149±5.3***
RXRβ 100±6.4 39±6.8***
RXRγ 100±6.6 44±7.1***
BP-3
(25 μM)
control
24 h treatment
RXRα
RXRβ
RXRγ
β-actin
b
a
c
d
0
50
100
150
200
RxrRxrRxr
receptor protein
[% of control]
control
25 μM BP3
***
*** ***
pg/ug protein RxrRxrRxr
control 1,07157 1,67567 0,74483
BP-3 1,43959 0,76412 0,37776
Fig. 5 Effects of BP-3 on the
protein levels of RXRα,RXRβ,
and RXRγin mouse neocortical
cultures at 7 DIV. The neocortical
cells were cultured for 7 DIV and
then treated for 24 h with BP-3
(25 μM). The concentrations of
the receptors were measured
using specific ELISAs and are
presented as a percentage of the
control (a) and as picogram of
specific protein, i.e., RXRα,
RXRβ, and RXRγ,per
microgram of total protein (b).
For the western blot analyses,
protein samples were denatured,
electrophoretically separated,
transferred to PVDF membranes,
and subjected to immunolabeling
(c). The relative protein levels of
RXRα,RXRβ, and RXRγwere
presented as a percentage of the
control (d). Each bar or value
represents the mean of three
independent experiments ± SEM.
The number of replicates in each
experiment ranged from 2 to 3.
***
p< 0.001 versus control
cultures
Mol Neurobiol (2018) 55:50595074 5067
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
control
BP-3
(25 μM)
RXRα+RXRγ bright field
control
BP-3
(25 μM)
MAP2 dleifthgirb2PAM+αRXRαRXR
control
BP-3
(25 μM)
RXRα RXRγ
RXRαRXRβRXRα+ RXRβbright field
Fig. 6 Influence of BP-3 on the cellular distributions of RXRα(red),
RXRβ(blue), RXRγ(blue), and MAP2 (blue) in mouse neocortical
cultures at 7 DIV. The overlay of RXRα/RXRβ, RXRα/RXRγ, and
RXRα/MAP2 (red plus blue) staining with the bright field images are
shown. The primary neocortical cultures were treated with BP-3 (25 μM)
for 24 h. Analyzed using an LSM510 META, Axiovert 200M confocal
laser scanning microscope under a Plan-Neofluor 40×/1.3 Oil DIC
objective
5068 Mol Neurobiol (2018) 55:50595074
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
were slightly less vulnerable to BP-3 than the non-transfected
cells. In comparison to the non-transfected cells, the effects of
BP-3 were reduced by 20% with respect to caspase-3 levels
andby30%withrespecttoLDHreleaseinthesiRNA-
transfected cells (Fig. 7a, b).
A 24-h exposure of RXRαsiRNA-transfected cells to
25 μM BP-3 reduced caspase-3 activity and LDH release to
55 and 50% of the control values, respectively (Fig. 7a, b).
These cells were much less vulnerable to BP-3 than the non-
siRNA-treated wild-type cells.
The effectiveness of mRNA silencing was verified by qPCR.
In this study, siRNA treatment decreased the RxrαmRNA level
by 83% (equal to 0.17-fold), the RxrβmRNA level by 64%
(equal to 0.36-fold), and the RxrγmRNA level by 68% (equal
to 0.32-fold) compared to the non-transfected wild-type cells.
Influence of BP-3 on Global DNA Methylation
in Neocortical Cultures
A 24-h exposure of neocortical cells to BP-3 (25 μM) caused
changes in the level of global DNA methylation. The treat-
ment with BP-3 reduced the methylation level by 55% of the
control value (Fig. 8).
Effects of BP-3 on HDAC and HATActivity in Neocortical
Cultures
A 24-h exposure of neocortical cultures to BP-3 reduced the
levels of HDAC and HAT activities. Treatment with BP-3
decreased the activities of HDAC and HAT by 32 and 17%
of the control value, respectively (Fig. 9a, b).
0
50
100
150
200
250
caspase-3 activity
[% of control]
$
non-transfected cells
$$$ ***
$$$
RXRγ
RXRβ
siRNA-transfected cells
RXRα
a
0
50
100
150
200
250
negative control BP-3 control BP-3 control BP-3 control BP-3
negative control BP-3 control BP-3 control BP-3 control BP-3
LDH release
[% of control]
$
$$$
***
$$$
non-transfected cells
siRNA-transfected cells
RXRα RXRβ RXRγ
b
Fig. 7 Effect of BP-3 (25 μM) on
caspase-3 activity (a) and LDH
release (b)inRXRα,RXRβ,and
RXRγsiRNA-transfected
neocortical cells. Each bar
represents the mean ± SEM of
three to four independent
experiments. The number of
replicates in each experiment
ranged from 5 to 8.
***
p<0.001
versus the non-transfected control
cultures;
$
p<0.05and
$$$
p< 0.001 versus the siRNA-
transfected control cultures
0
20
40
60
80
100
120
control 25 μM BP-3
Global DNA methylation [% of control]
***
Fig. 8 Influence of BP-3 on
global DNA methylation in
neocortical cultures at 7 DIV.
Primary neocortical cultures were
treated with BP-3 (25 μM) for
24 h. Total DNA was extracted
from cells followed by ELISA.
Each bar represents the mean of
three independent
experiments ± SEM. The number
of replicates in each experiment
ranged from 2 to 3.
***
p<0.001
versus control cultures
Mol Neurobiol (2018) 55:50595074 5069
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Effects of BP-3 on the Expression Profiles of Genes
Involved in Autophagy Using a Mouse Autophagy RT
2
Profiler PCR Array
To validate that BP-3 impairs autophagy in neuronal
cells, we analyzed a total number of 84 key genes that
are involved in this process. Among them, 71 genes
were differentially expressed in response to BP-3 treat-
ment: 60 were downregulated (green color) and 11 were
upregulated (red color) in the BP-3-treated samples. The
downregulated genes were Akt1,Ambra1,App,Atg10,
Atg16l1,Atg16l2,Atg3,Atg4b,Atg4d,Atg7,Atg9a,
Atg9b,Bcl2,Bid,Cdkn1b,Cdkn2a,Cln3,Ctsb,Ctsd,
Ctss,Cxcr4,Dram1,Eif2ak3,Eif4g1,Esr1,Gaa,
Gabarapl1,Gabarapl2,Hdac1,Hdac6,Hgs,
Hsp90aa1,Hspa8,Htt,Igf1,Irgm1,Lamp1,Map1lc3a,
Map1lc3b,Mapk14,Mapk8,Mtor,Npc1,Pik3c3,
Pik3r4,Prkaa1,Pten,Rab24,Rb1,Rgs19,Rps6kb1,
Snca,Sqstm1,Tgfb1,Tgm2,Tmem74,Ulk1,Ulk2,
Uvrag,andWip i1. The upregulated genes were Bad,
Bak1,Bax,Bcl2l1,Bnip3,Casp3,Casp8,Dapk1,Fas,
Nfkb1,andTr p53 (Fig. 10).
Effects of BP-3 on Autophagosome Detection
A 24-h exposure of neocortical cultures to BP-3 reduced the
level of autophagosomes in mouse neuronal cell cultures.
Treatment with BP-3 decreased autophagosome level by
29% compared to the control value (Fig. 11).
Effects of BP-3 on the Protein Expression Levels of LC3A
and LC3B in Mouse Neocortical Cells
InculturesexposedtoBP-3(25μM) for 24 h, the concentration
of LC3A was 1.23 pg/μg of total protein, which was 126%
higher than that in control cultures. In the cultures exposed to
BP-3, the level of LC3B reached 0.21 pg/μg, which was re-
duced by 75% compared to controls (Fig. 12a, b).
Discussion
The primary aim of the present study was to evaluate the
neurotoxic effects of BP-3 with an emphasis on apoptosis,
autophagy, the epigenetic status of neuronal cells, and the mo-
lecular mechanisms involving RXRs. The results of the study
demonstrated that BP-3 caused neurotoxicity, as evidenced by
the concentration-dependent activation of caspase-3 and LDH
release in mouse neocortical cells. In the present study, neo-
cortical cells responded to 25100 μM BP-3. This was in line
with our previous study in which 25 μM BP-3 was determined
to be the lowest effective concentration at 24 h of exposure
[13]. The used concentration is environmentally relevant since
BP-3 has been found in human adipose tissue at concentra-
tions up to 5 mg/kg (~22 μM) [37]. The ability of BP-3 to
cross the blood-brain barrier has been shown; after it was
applied by gavage, the Erαand ErβmRNA expression levels
were changed in the rat pituitary gland [38]. Moreover, a BP-3
analogue (BP-4) changed the expression levels of many genes
0
20
40
60
80
100
120
control 25 µM BP-3 positive control negative control
Histone Deacetylase
(HDAC) Activity
[% of control]
control
25 µM BP-3
positive control
negative control
***
a
0
50
100
150
200
250
control BP-3 25 uM
p
ositive
Histone Acetyltransferase
(HAT) Activity
[% of control]
control
BP-3 25 uM
positive
**
b
Fig. 9 Effects of BP-3 (25 μM)
on HDAC (a)andHAT(b)
activity in the primary cultures of
mouse neocortical cells at 7 DIV.
The cells were treated with BP-3
for 24 h. The results are presented
as a percentage of the control.
Each bar represents the mean of
three to four independent
experiments ± SEM. The number
of replicates in each experiment
ranged from 5 to 8.
**
p<0.01and
***
p< 0.001 versus control
cultures
5070 Mol Neurobiol (2018) 55:50595074
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
in the Danio rerio brain when it was added to water [39]. In the
present study, the neurotoxic effects of BP-3 involved en-
hanced LDH release and impaired cell survival (calcein AM
staining), which were accompanied by the induction of apo-
ptosis as estimated by caspase-3 activation and increased ap-
optotic body formation (Hoechst 33342 staining).
In this study, both the neurotoxic and apoptotic effects of BP-3
were inhibited by HX 531, which is a potent RXR antagonist.
These data suggest that RXR receptors are involved in BP-3-
induced effects in neuronal cells. Previously, we demonstrated
an important role of RXRα-andRXRβ-intracellular signaling in
the propagation of DDE- and nonylphenol-induced apoptosis
during the early stages of neural development [2224].
Recently, the involvement of RXRs in apoptotic signaling in
retina pigment epithelial cells and gastrointestinal cancer cells
has been shown [40,41]. The role of RXRs in neuronal survival
and neurotoxicity is complex and depends on RXR
heterodimerization partners. Interestingly, A/B domain region
of RXR receptors was found to cause growth inhibition or
rexinoid-induced apoptosis [42]. Elevated levels of RXRαgene
and protein expression have been found in individuals suffering
from dementia [43], and knockout of RXRγimpairs the working
memory in mice [44]. In the present study, BP-3 altered the
mRNA expression levels of Rxrα,Rxrβ,andRxrγin a time-
dependent manner. Increased expression of RxrαmRNA and
reduced expression levels of Rxrβand RxrγmRNA were ob-
served at 3 h of exposure, which mirrored the alterations in the
estimated protein levels of the receptors at 24 h of exposure.
These profiles were also concurred with the immunofluorescent
labeling of RXRα,RXRβ,andRXRγfollowingBP-3treat-
ment. Based on these data, we hypothesize that the BP-3-
Fig. 10 Gene expression patterns of autophagy showing the 71 genes
that were significantly differentially expressed between the control and
BP-3-treated groups. Among these genes, 60 genes were downregulated
(green color) and 11 genes were upregulated (red color) in the BP-3-
treated samples compared to the control
0
20
40
60
80
100
120
control 25 μM BP-3
fluorescent autophagosome marker
[% of control]
Autophagosome detection
**
Fig. 11 The effect of 25 μM BP-3 on autophagosome levels at 24 h. The
data are expressed as the mean ± SEM of four independent experiments,
consisting of eight replicates per treatment group.
**
p< 0.001 versus the
control
b
a
0
50
100
150
200
250
300
B3CLA3CL
protein level [% of control]
control
25 µM BP-3
***
***
pg/μg protein LC3A LC3B
control 0.544655 0.887605
25 μM BP-3 1.235620 0.217725
Fig. 12 Effects of BP-3 on the protein levels of LC3A and LC3B in
mouse neocortical cultures at 7 DIV. The neocortical cells were cultured
for 7 DIV and then treated for 24 h with BP-3 (25 μM). The
concentrations of the receptors were measured using ELISA and are
presented as a percentage of the control (a) and as picogram of the
specific protein (LC3A or LC3B) per microgram of total protein (b).
***p< 0.001 versus control cultures
Mol Neurobiol (2018) 55:50595074 5071
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
induced apoptosis of neuronal cells is mediated by the attenua-
tion of RXRβ/RXRγand the stimulation of RXRαsignaling
pathways. To test this hypothesis, we used gene-specific
siRNAs. Compared to non-transfected wild-type cells, silencing
of RXRαcaused a substantial reduction of BP-3-induced
caspase-3 activity and LDH release; however, silencing of
RXRβand RXRγdid not affect the BP-3-induced effects.
Therefore, we suggest that the BP-3-induced apoptosis of neuro-
nal cells is mediated by the stimulation of RXRαsignaling and
the attenuation of RXRβ/RXRγsignaling, which is in line with
the BP-3-induced alterations in RXR expression levels.
Previously, we demonstrated that stimulation of ERβ/GPR30
and impairment of ERα/PPARγsignaling were involved in
propagation of BP-3-induced apoptosis [13]. Taking into account
our previous and present data, one may assume that in neuronal
cells BP-3 stimulates RXRα/ERβ/GPR30 and inhibits
RXRβ/RXRγ/ERα/PPARγintracellular pathways. It has been
documented that ERβmay downregulate ERα/PPARγand dis-
rupt RXRα/PPARγsignaling [45]. RXRαis an obligatory het-
erodimer partner of PPARγas well as RXRβ/RXRγ[46]. We
suggest that in our study, BP-3 by upregulation of ERβimpaired
RXRβ/RXRγ/ERα/PPARγsignaling. We also postulate that
BP-3 was able to destroy RXRβand RXRγheterodimers but
not RXRαhomodimers, possibly due to stronger covalent bond-
ing in homodimers than in heterodimers.
In addition to the demonstration that the BP-3-induced apo-
ptosis involves the activation of RXRαsignaling and the impair-
ment of RXRβ/RXRγsignaling, we showed that BP-3 inhibited
global DNA methylation as well as reduced HDAC and HAT
activities in mouse embryonic neuronal cells. Aberrant DNA
methylation and other epigenetic modifications have been found
to be associated with development and normal cellular homeo-
stasis as well as growing number of human diseases. Low doses
of a pesticide DDT have been postulated to cause hypomethyla-
tion of specific gene regions in the young brain and impaired
hippocampal neurogenesis [25]. Recently, we showed evidence
of the involvement of global DNA hypomethylation in DDT-
induced depressive-like effects and the DDE-induced apoptosis
of primary neuronal cells [22,47]. Exposures to xenobiotics such
as tributyltin (TBT) and triphenyltin (TPT) have been shown to
alter HDAC and HAT activity [48]. The global DNA hypome-
thylation as well as diminished HDAC and HAT activity suggest
of chromosomal instability thus can cause inappropriate gene
expression pattern. We postulate that global DNA hypomethyla-
tion and diminished HDAC activity are responsible for the BP-3-
induced increase in the RXRαexpression level, whereas dimin-
ished HAT activity corresponds to reduced expression of RXRβ/
RXRγin response to BP-3 treatment in our study.
Based upon our data, we suggest that altered epigenetic
status caused by BP-3 treatment may not only be involved
in apoptosis and the disruption of RXR signaling but may also
affect autophagy. Autophagy is neuroprotective and is respon-
sible for degrading damaged organelles and misfolded
proteins. Dysregulation of autophagy has been linked to neu-
ral degenerative diseases such as Alzheimersdisease,
Parkinsons disease, Huntingtons disease, amyotrophic later-
al sclerosis, and encephalopathy. In the present study, the at-
tenuation of the autophagic process was confirmed by the
downregulation of genes involved in autophagy as detected
by the microarray analysis, decreased autophagosome forma-
tion, and the reduced ratio of LC3B to LC3A. We hypothesize
that BP-3 induced the downregulation of genes related to au-
tophagy through decreased HAT activity in mouse neurons.
Conclusions
In summary, we showed for the first time that the BP-3-
induced apoptosis of neuronal cells is mediated via the stim-
ulation of RXRαsignaling and the attenuation of RXRβ/
RXRγsignaling, as demonstrated by the use of selective an-
tagonist and specific siRNAs as well as by measuring the
mRNA and protein expression levels (qPCR, ELISA, western
blot, and immunofluorescent labeling) of the receptors. This
study also demonstrated that the use of BP-3 at environmen-
tally relevant concentrations was able to inhibit autophagy and
disrupt the epigenetic status of neuronal cells, which may
increase the risk of neurodevelopmental abnormalities and/or
neural degeneration.
Abbreviations
AM acetoxymethyl
ANOVA analysis of variance
BP-3 benzophenone-3
BPA bisphenol A
BPs benzophenones
DDE dichlorodiphenyldichloroethylene
DIV days in vitro
DMSO dimethyl sulfoxide
EDCs endocrine disrupting chemicals
ELISA enzyme-linked immunosorbent assay
FBS fetal bovine serum
GFAP glial fibrillary acidic protein
HAT histone acetyltransferase
HDAC histone deacetylase
Hprt hypoxanthine-guanine phosphoribosyltransferase
LDH lactate dehydrogenase
PBS phosphate-buffered saline
qPCR quantitative polymerase chain reaction
RT room temperature
RXR retinoid X receptor
TBT tributyltin
TPT triphenyltin
UV ultraviolet light
5072 Mol Neurobiol (2018) 55:50595074
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Acknowledgments This study was financially supported by grant no.
2014/13/N/NZ4/04845 from the National Science Centre of Poland and
the statutory fund of the Institute of Pharmacology at the Polish Academy
of Sciences in Krakow, Poland.
Agnieszka Wnuk and Joanna Rzemieniec received scholarships from
the KNOW, which was sponsored by the Ministry of Science and Higher
Education in Poland.
This publication was also supported by funding from the Jagiellonian
University within the SET project, which was co-financed by the
European Union. The authors thank Professor Elżbieta Pyza of the
Department of Cell Biology and Imaging at the Institute of Zoology of
Jagiellonian University in Krakow for suggestions and for kindly provid-
ing access to the LSM 510 META, Axiovert 200M, ConfoCor 3 confocal
microscope (Carl Zeiss MicroImaging GmbH, Jena Germany).
The manuscript has been edited by the American Journal Experts for
English language and grammar - C2A7-BFBD-03E0-6E26-A2D8.
The publication charge was supported by KNOW funds MNiSW-DS-
6002-4693-26/WA/12.
Compliance with Ethical Standards
Conflict of Interest The authors declare that they have no conflict of
interest.
Research Involving Human Participants and/or Animals This arti-
cle does not contain any studies with human participants performed by
any of the authors.
All procedures performed in studies involving animals were in accor-
dance with the ethical standards of the institution or practice at which the
studies were conducted.
All members of the research team received approval from the local
ethical committee on animal testing. Animal care followed official gov-
ernmental guidelines, and all efforts were made to minimize suffering and
the number of animals used.
Open Access This article is distributed under the terms of the Creative
Commons Attribution 4.0 International License (http://
creativecommons.org/licenses/by/4.0/), which permits unrestricted use,
distribution, and reproduction in any medium, provided you give appro-
priate credit to the original author(s) and the source, provide a link to the
Creative Commons license, and indicate if changes were made.
References
1. Danovaro R, Bongiorni L, Corinaldesi C, Giovannelli D, Damiani
E, Astolfi P, Greci L, Pusceddu A (2008) Sunscreens cause coral
bleaching by promoting viral infections. Environ Health Perspect
116:441447. doi:10.1289/ehp.10966
2. Zhang T, Sun H, Kannan K (2013) Blood and urinary bisphenol A
concentrations in children, adults, and pregnant women from
China: partitioning between blood and urine and maternal and fetal
cord blood. Environ Sci Technol 47:46864694. doi:10.1021/
es303808b
3. Calafat AM, Wong LY, Ye X, Reidy JA, Needham LL (2008)
Concentrations of the sunscreen agent benzophenone-3 in residents
of the United States. Environ Health Perspect 116:893897. doi:10.
1289/ehp.11269
4. Philippat C, Mortamais M, Chevrier C, Petit C, Calafat AM, Ye X,
Silva MJ, Brambilla C et al (2012) Exposure to phthalates and
phenols during pregnancy and offspring size at birth. Environ
Health Perspect 120:464470. doi:10.1289/ehp.1103634
5. Wolff MS, Engel SM, Berkowitz GS, Ye X, Silva MJ, Zhu C,
Wetmur J, Calafat AM (2008) Prenatal phenol and phthalate expo-
sures and birth outcomes. Environ Health Perspect 116:10921097.
doi:10.1289/ehp.11007
6. Frederiksen H, Jensen TK, Jørgensen N, Kyhl HB, Husby S,
Skakkebæk NE, Main KM, Juul A et al (2014) Human urinary
excretion of non-persistent environmental chemicals: an overview
of Danish data collected between 2006 and 2012. Reproduction
147:555565. doi:10.1530/REP-13-0522. Print 2014
7. Frederiksen H, Nielsen JK, Mørck TA, Hansen PW, Jensen JF,
Nielsen O, Andersson AM, Knudsen LE(2013b) Urinary excretion
of phthalate metabolites, phenols and parabens in rural and urban
Danish mother-child pairs. Int J Hyg Environ Health 216(6):772
783. doi:10.1016/j.ijheh.2013.02.006
8. Janjua NR, Kongshoj B, Andersson AM, Wulf HC (2008)
Sunscreens in human plasma and urine after repeated whole-body
topical application. J Eur Acad Dermatol Venereol 22(4):456461.
doi:10.1111/j.1468-3083.2007.02492.x
9. Jiang R, Roberts MS, Collins DM, Benson HA (1999) Absorption
of sunscreens across human skin: an evaluation of commercial
products for children and adults. Br J Clin Pharmacol 48:635637
10. Schlumpf M, Kypke K, Wittassek M, Angerer J, Mascher H,
Mascher D, Vökt C, Birchler M et al (2010) Exposure patterns of
UV-filters, fragrances, parabens, phthalates, organochlor pesticides,
PBDEs, and PCBs in human milk: correlation of UV-filters with
use of cosmetics. Chemosphere 81:11711183. doi:10.1016/j.
chemosphere.2010.09.079
11. Huo W, Cai P, Chen M, Li H, Tang J, Xu C, Zhu D, Tang W et al
(2016) The relationship between prenatal exposure to BP-3 and
Hirschsprung's disease. Chemosphere 144:10911097. doi:10.
1016/j.chemosphere.2015.09.019
12. Broniowska Ż, Pomierny B, Smaga I, Filip M, Budziszewska B
(2016) The effect of UV-filters on the viability of neuroblastoma
(SH-SY5Y) cell line. Neurotoxicology May 54:4452. doi:10.
1016/j.neuro.2016.03.003
13. Wnuk A, Rzemieniec J, LasońW, Krzeptowski W, Kajta M
(2017) Apoptosis induced by the UV filter benzophenone-3
in mouse neuronal cells is mediated via attenuation of Erα/
Pparγand stimulation of Erβ/Gpr30 signaling. Mol
Neurobiol. doi:10.1007/s12035-017-0480-z
14. Ma R, Cotton B, Lichtensteiger W, Schlumpf M (2003) UV filters
with antagonistic action at androgen receptors in the MDA-kb2 cell
transcriptional-activation assay. Toxicol Sci 74(1):4350
15. Schlumpf M, Cotton B, Conscience M, Haller V, Steinmann B,
Lichtensteiger W (2001) In vitro and in vivo estrogenicity of UV
screens. Environ Health Perspect 109(3):239244
16. Schreurs RH, Sonneveld E, Jansen JH, Seinen W, van der Burg B
(2005) Interaction of polycyclic musks and UV-filters with the es-
trogen receptor (ER), androgen receptor (AR), and progesterone
receptor (PR) in reporter gene bioassays. Toxicol Sci 83:264272
17. Kunisue T, Chen Z, Buck Louis GM, Sundaram R, Hediger ML,
Sun L, Kannan K (2012) Urinary concentrations of benzophenone-
type UV filters in US women and their association with endometri-
osis. Environ Sci Technol 46:46244632. doi:10.1021/es204415a
18. Evans, R.M., Mangelsdorf, D.J. (2014) Nuclear receptors, RXR,
and the big bang. Cell 2014;157(1):255266. doi: 10.1016/j.cell.
2014.03.012
19. Rőszer T, Menéndez-Gutiérrez MP, Cedenilla M, Ricote M (2013)
Retinoid X receptors in macrophage biology. Trends Endocrinol
Metab 24(9):460468. doi:10.1016/j.tem.2013.04.004
20. Zhang H, Chen L, Chen J, Jiang H, Shen X (2011) Structural basis
for retinoic X receptor repression on the tetramer. J Biol Chem
286(28):2459324598. doi:10.1074/jbc.M111.245498
21. Huang JK, Jarjour AA, Nait Oumesmar B, Kerninon C, Williams
A, Krezel W, Kagechika H, Bauer J et al (2011) Retinoid X receptor
Mol Neurobiol (2018) 55:50595074 5073
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
gamma signaling accelerates CNS remyelination. Nat Neurosci
14(1):4553. doi:10.1038/nn.2702
22. Wnuk A, Rzemieniec J, Litwa E, LasońW, Krzeptowski W,
Wójtowicz AK, Kajta M (2016) The crucial involvement of reti-
noid X receptors in DDE neurotoxicity. Neurotox Res 29(1):155
172. doi:10.1007/s12640-015-9572-6
23. Litwa E, Rzemieniec J, Wnuk A, Lason W, Krzeptowski W, Kajta
M (2014) Apoptotic and neurotoxic actions of 4-para-nonylphenol
are accompanied by activation of retinoid X receptor and impair-
ment of classical estrogen receptor signaling. J Steroid Biochem
Mol Biol 144(Pt B):334347. doi:10.1016/j.jsbmb.2014.07.014
24. Litwa E, Rzemieniec J, Wnuk A, LasońW, Kr z e pto w s ki W, Ka j t a
M(2016)RXRα, PXR and CAR xenobiotic receptors mediate the
apoptotic and neurotoxic actions of nonylphenol in mouse hippo-
campal cells. J Steroid Biochem Mol Biol 156:4352. doi:10.1016/
j.jsbmb.2015.11.018
25. Shutoh Y, Takeda M, Ohtsuka R, Haishima A, Yamaguchi S, Fujie
H, Komatsu Y, Maita K et al (2009) Low dose effects of dichloro-
diphenyltrichloroethane (DDT) on gene transcription and DNA
methylation in the hypothalamus of young male rats: implication
of hormesis-like effects. J Toxicol Sci 34(5):469482
26. Huen K, Yousefi P, Bradman A, Yan L, Harley KG, Kogut K,
Eskenazi B, Holland N (2014) Effects of age, sex, and persistent
organic pollutants on DNA methylation in children. Environ Mol
Mutagen 55(3):209222. doi:10.1002/em.21845
27. Volmar CH, Wahlestedt C (2015) Histone deacetylases (HDACs) and
brain function. Neuroepigenetics Volume 1(January 2015):2027
28. Glick D, Barth S, Macleod KF (2010) Autophagy: cellular
and molecular mechanisms. J Pathol 221(1):312. doi:10.
1002/path.2697
29. Kajta M, Domin H, Grynkiewicz G, LasońW (2007) Genistein
inhibits glutamate-induced apoptotic processes in primary neuronal
cell cultures: an involvement of aryl hydrocarbon receptor and es-
trogen receptor/glycogen synthase kinase-3beta intracellular signal-
ing pathway. Neuroscience 145(2):592604
30. Kajta M, Makarewicz D, Ziemińska E, Jantas D, Domin H, Lasoń
W, Kutner A, Łazarewicz JW (2009) Neuroprotection by co-
treatment and post-treating with calcitriol following the ischemic
and excitotoxic insult in vivo and in vitro. Neurochem Int 55(5):
265274. doi:10.1016/j.neuint.2009.03.010
31. Nicholson DW, Ali A, Thornberry NA, Vaillancourt JP, Ding CK,
Gallant M, Gareau Y, Griffin PR et al (1995) Identification and
inhibition of the ICE/CED-3 protease necessary for mammalian
apoptosis. Nature 376:3743
32. Kajta M, Trotter A, LasońW, Beyer C (2005) Effect of NMDA on
staurosporine-induced activation of caspase-3 and LDH release in
mouse neocortical andhippocampal cells. Brain Res Dev Brain Res
160(1):4052
33. Wójtowicz AK, Kajta M, Gregoraszczuk EŁ(2007) DDT- and
DDE-induced disruption of ovarian steroidogenesis in prepubertal
porcine ovarian follicles: a possible interaction with the main ste-
roidogenic enzymes and estrogen receptor beta. J Physiol
Pharmacol 58(4):873885
34. Rzemieniec J, Litwa E, Wnuk A, LasońW, Go łas A. Krzeptowski
W, Kajta M (2015) Neuroprotective action of raloxifene against
hypoxia-induced damage in mouse hippocampal cells depends on
ERαbut not ERβor GPR30 signalling. J steroid Biochem Mol
Biol. 146:26-37. doi: 10.1016/j.jsbmb.2014.05.005.
35. Rzemieniec J, Litwa E, Wnuk A, Lason W, Krzeptowski W, Kajta
M (2016) Selective aryl hydrocarbon receptor modulator 3,3-
diindolylmethane impairs AhR and ARNT signaling and protects
mouse neuronal cells against hypoxia. Mol Neurobiol 53(8):5591
5606. doi:10.1007/s12035-015-9471-0
36. Kajta M, Rzemieniec J, Litwa E, LasońW, Le nart o wic z M ,
Krzeptowski W, Wójtowicz AK (2013) The key involvement of
estrogen receptor βand G-protein-coupled receptor 30 in the neu-
roprotective action of daidzein. Neuroscience 238:345360. doi:10.
1016/j.neuroscience.2013.02.005
37. Wang L, Asimakopoulos AG, Kannan K (2015) Accumulation of
19 environmental phenolic and xenobiotic heterocyclic aromatic
compounds in human adipose tissue. Environ Int 78:4550. doi:
10.1016/j.envint.2015.02.015
38. Schlecht C, Klammer H, Jarry H, Wuttke W (2004) Effects
of estradiol, benzophenone-2 and benzophenone-3 on the
expression pattern of the estrogen receptors (ER) alpha and
beta, the estrogen receptor-related receptor 1 (ERR1) and the
aryl hydrocarbon receptor (AhR) in adult ovariectomized
rats. Toxicology 205(12):123130
39. Zucchi S, Blüthgen N, Ieronimo A, Fent K (2010) TheUV-absorber
benzophenone-4 alters transcripts of genes involved in hormonal
pathways in zebrafish (Danio rerio) eleuthero-embryos and adult
males. Toxicol Appl Pharmacol 250(2):137146. doi:10.1016/j.
taap.2010.10.001
40. Ayala-Peña VB, Pilotti F, Volonté Y, Rotstein NP, Politi LE,
German OL (2016) Protective effects of retinoid x receptors on
retina pigment epithelium cells. Biochim Biophys Acta 1863(6 Pt
a):11341145. doi:10.1016/j.bbamcr.2016.02.010
41. Papi A, Govoni M, Ciavarella C, Spisni E, Orlandi M, Farabegoli F
(2016) Epigallocatechin-3-gallate increases RXRγ-mediated pro-
apoptotic and anti-invasive effects in gastrointestinal cancer cell
lines. Curr Cancer Drug Targets 16(4):373385
42. Qin S, Okawa Y, Atangan LI, Brown G, Chandraratna RA,
Zhao Y (2008) Integrities of A/B and C domains of RXR
are required for rexinoid-induced caspase activations and ap-
optosis. J Steroid Biochem Mol Biol 112(13):2531. doi:
10.1016/j.jsbmb.2008.08.001
43. Akram A, Schmeidler J, Katsel P, Hof PR, Haroutunian V (2010)
Increased expression of RXRαin dementia: an early harbinger for
the cholesterol dyshomeostasis? Mol Neurodegener 15(5):36. doi:
10.1186/1750-1326-5-36
44. Wietrzych M, Meziane H, Sutter A, Ghyselinck N, Chapman PF,
Chambon P, Krezel W (2005) Working memory deficits in retinoid
X receptor gamma-deficient mice. Learn Mem 12(3):318326
45. Wang X, Liu J, Long Z, Sun Q, Liu Y, Wang L, Zhang X, Hai C
(2015) Effect of diosgenin on metabolic dysfunction: role of ERβ
in the regulation of PPARγ. Toxicol Appl Pharmacol 289(2):286
296. doi:10.1016/j.taap.2015.09.015
46. Lefebvre P, Benomar Y, Staels B (2010) Retinoid X receptors:
common heterodimerization partners with distinct functions.
Trends Endocrinol Metab 21(11):676683. doi:10.1016/j.tem.
2010.06.009
47. Kajta M, Wnuk A, Rzemieniec J, Litwa E, Lason W, Zelek-Molik
A, Nalepa I, RogóżZet al (2017) Depressive-like effect of prenatal
exposure to DDT involves global DNA hypomethylation and im-
pairment of GPER1/ESR1 protein levels but not ESR2 and AHR/
ARNT signaling. J Steroid Biochem Mol Biol. doi:10.1016/j.
jsbmb.2017.03.001
48. Osada S, Nishikawa J, Nakanishi T, Tanaka K, Nishihara T (2005)
Some organotin compounds enhance histone acetyltransferase ac-
tivity. Toxicol Lett 155(2):329335
5074 Mol Neurobiol (2018) 55:50595074
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... After hypoxia/ischemia and amorfrutin B post-treatment, the total RNA was extracted from neocortical cells using the RNeasy Mini Kit (Qiagen, Hilden, Germany), as previously described [30]. The amount of RNA was assessed at 260 nm and 260/280 nm with a NanoDrop ND-1000 UV-Vis Spectrophotometer (Thermo Fisher Scientific, Waltham, MA, USA). ...
... Sets were designed for the bisulfite-converted DNA sequences and represented: (i) fully methylated and fully unmethylated probes for the Bax, Bcl2, Ambra1, Map1lc3b, Atg7, and Becn1 promoters, (ii) the internal reference set for the Hprt1 gene to control for input DNA. Quantitative real-time polymerase chain reaction (MethyLight) was performed as previously described [30], and the EpiTect MethyLight assays enabled the calculation of the degree of methylation according to the following formula: ΔΔCt = methylated signal (Ct target gene − Ct Hprt1) − unmethylated signal (Ct target gene − Ct Hprt1). The results are presented as the methylation rate ± SEM. ...
... Previously, we showed altered methylation levels of Bcl2 and Bax genes that were correlated with an increased BAX/BCL2 ratio in response to neurotoxic action of triclocarban [26]. Another study from our group showed that benzophenone-3-induced neurotoxicity involved Bax hypomethylation and Bcl2 hypermethylation, which confirmed a key role of DNA methylation in the regulation of the expression levels of apoptosis-related factors [30]. ...
Article
Full-text available
Amorfrutin B is a selective modulator of the PPARγ receptor, which has recently been identified as an effective neuroprotective compound that protects brain neurons from hypoxic and ischemic damage. Our study demonstrated for the first time that a 6-h delayed post-treatment with amorfrutin B prevented hypoxia/ischemia-induced neuronal apoptosis in terms of the loss of mitochondrial membrane potential, heterochromatin foci formation, and expression of specific genes and proteins. The expression of all studied apoptosis-related factors was decreased in response to amorfrutin B, both during hypoxia and ischemia, except for the expression of anti-apoptotic BCL2, which was increased. After post-treatment with amorfrutin B, the methylation rate of the pro-apoptotic Bax gene was inversely correlated with the protein level, which explained the decrease in the BAX/BCL2 ratio as a result of Bax hypermethylation. The mechanisms of the protective action of amorfrutin B also involved the inhibition of autophagy, as evidenced by diminished autophagolysosome formation and the loss of neuroprotective properties of amorfrutin B after the silencing of Becn1 and/or Atg7. Although post-treatment with amorfrutin B reduced the expression levels of Becn1, Nup62, and Ambra1 during hypoxia, it stimulated Atg5 and the protein levels of MAP1LC3B and AMBRA1 during ischemia, supporting the ambiguous role of autophagy in the development of brain pathologies. Furthermore, amorfrutin B affected the expression levels of apoptosis-focused and autophagy-related miRNAs, and many of these miRNAs were oppositely regulated by amorfrutin B and hypoxia/ischemia. The results strongly support the position of amorfrutin B among the most promising anti-stroke and wide-window therapeutics.
... Among female partners, n = 62 (15.4%) were ≥35 years compared to n = 115 (29.8%) of male partners. The median ages at study entry were 29 years (IQR = 6) for females and 32 years (IQR = 6) for their male partners and 2.5th and 97.5th percentiles for age was (23,38) and (24,42) for females and males, respectively (Table 1). Median BMI was 25.8 kg/m 2 (IQR = 8.6) for females and 28.8 kg/m 2 (IQR = 5.9) for males. ...
... Biological aging may affect endometrial receptivity and, therefore, could modify the effect of BP-type filters on fecundability. Epigenetic changes occurred following BP-type filter exposure [42,43] and they are reported to increase with age [44]. Therefore, this suggests a possible mechanism by which benzophenones may affect fecundability. ...
Article
Full-text available
Urinary concentrations of several endocrine disrupting chemicals, including phthalate metabolites, bisphenol A (BPA), and benzophenone (BP)-type ultraviolet (UV) filters, have been associated with a longer time-to-pregnancy (TTP). Potential modification of these associations by couple’s age has not been studied. TTP was defined as the number of prospectively observed menstrual cycles a couple attempted pregnancy until the occurrence of a human chorionic gonadotropic-detected pregnancy. Urinary concentrations of two BP-type UV filters and three phthalate metabolites were measured at baseline. Fecundability odds ratios (FORs) and 95% confidence intervals (CIs) were estimated for each chemical adjusting for age, body mass index, serum cotinine, creatinine, and accounting for right censoring and left truncation. Models evaluated effect modification between EDC concentrations and TTP by partner’s age, dichotomized at 35 years. Separate models were run for male and female partners. No significant effect modification was observed for any EDC for either partner, but data were suggestive of a longer TTP among females aged ³35 years, particularly for BP-2 (FOR = 0.61, 95% CI 0.36, 1.05) and 4-hydroxybenzophenone (FOR = 0.71, 95% CI: 0.46, 1.09) reflecting 39% and 29% reductions in fecundability, respectively. We saw no evidence of effect modification by couples’ age on associations between TTP and urinary phthalate or BPA metabolite concentrations. Across the EDCs we examined, we found little evidence that age modifies TTP-exposure associations.
... For example, a recent population study based on a large birth cohort reported a significant negative correlation between maternal exposure to benzophenone, in particular that of 4-hydroxybenzophenone (4HBP), and child cognitive development at 2 years (Jiang et al., 2019). The toxicology of benzophenone-3 (BP-3) exposure includes apoptosis in mouse mature neurons and rat frontal cortex and hippocampus, which was accompanied by impaired autophagy and altered epigenetics (Krzyzanowska et al., 2018;Wnuk et al., 2018b;Wnuk et al., 2018c). Importantly, benzophenones have been shown to be able to cross the placental and blood-brain barriers (Pomierny et al., 2019;van der Meer et al., 2017), leading to potential neurodevelopmental toxicity. ...
... Several recent studies have profiled the neurotoxicity of benzophenones in mouse and rat models. BP-3 treatment induced apoptosis in mature neurons in vitro and in vivo, which may be explained by the impaired autophagy and altered epigenetic modifications (Krzyzanowska et al., 2018;Wnuk et al., 2018aWnuk et al., , 2018b. Likewise, BP-3 caused similar apoptotic effects in neocortical culture isolated from mouse embryos, accompanied by disrupted estrogen receptor and RXRs/PPARγ signaling pathways, altered activities of epigenetic modulators, and repressed post-translational modifications (Wnuk et al., 2018c;Wnuk et al., 2019). ...
Article
Benzophenones are widely used in industry and commonly added in many personal care products. However, the neurotoxicity, in particular neurodevelopmental toxicity, of benzophenone family chemicals and metabolites has not been fully elucidated. Our recent mechanistic study in mice showed that early life exposure to a major benzophenone metabolite, 4-hydroxybenzophenone (4HBP), disrupted endoplasmic reticulum (ER) proteostasis and evoked inflammatory response in hippocampal neural stem cells (NSCs), leading to cognitive dysfunction. Despite so, detailed inflammatory cytokine(s) that possibly mediate this toxicity remains to be defined and validated. In this study, we confirmed that 4HBP treatment inhibited the viability and sphere growth of mouse NSCs in vitro. Importantly, re-interrogation of the transcriptomic data of NSCs treated with 4HBP identified the top upregulated genes, wherein the chemokine Cxcl1 ranked first. Immunofluorescent staining and qRT-PCR validated the robust induction of Cxcl1 on the protein and mRNA levels upon 4HBP treatment. Furthermore, siRNA-mediated knockdown of Cxcl1 transiently blocked its expression and led to enhanced NSCs viability in the presence of 4HBP. Together, these in vitro results indicated that the adverse effect of benzophenones on NSCs is mediated, at least in part, by induction of the chemokine Cxcl1.
... Toxicopathological processes and multiple etiological mechanisms caused by oxybenzone are only now being characterized with audacity and rigor, though rodent-model studies dating back to 1992 demonstrated the occurrence of histopathological lesions to liver and kidneys, as well as a decrease in sperm density and duration of the estrous cycle (French, 1992). Work by Wnuk and co-workers has elucidated the neuropathological pathways of oxybenzone exposure, generating several distinct pathways of neuronal cell death, as well as inducing epigenetic changes in affected neurons (Pomierny et al., 2019;Wnuk et al., 2018aWnuk et al., , 2018bWnuk et al., , 2019Wnuk and Kajta, 2021). Oxybenzone is argued to play a role in Hirschsprung's Disease, exhibiting a neuropathological role similarly described by Wnuk et al. that induces cell death and prevents neuro-stem cell migration during development via modification of cellular signal pathways (DiNardo and Downs, 2019;Huo et al., 2016;Viola and Grant-Kels, 2021;Wang et al., 2021). ...
Article
A preponderance of recent evidence indicates that oxybenzone and other personal-care product chemicals threaten the biota inhabiting various ecological niches. What is understudied is the ecotoxicological impact of oxybenzone, a UV filter in sunscreens and anti-aging products, to terrestrial/soil organisms that are keystone species in these habitats. In the present study, acute exposure (14-day) to oxybenzone resulted in earthworm mortality (LC 50 of 364 mg/kg) and growth rate inhibition. Environmentally relevant concentration of oxy-benzone (3.64, 7.28 and 36.4 mg/kg) at exposures of 7-day, 14-day, 28-day induced oxidative stress and neurotoxicity followed by perturbations in reproduction processes and changes in vital organs. Decreased levels of superoxide dismutase (SOD) and catalase (CAT) activity were statistically lower than controls (p < 0.05) on day 14 for all three concentrations, while glutathione-s-transferase (GST) activity was significantly elevated from controls on days 7 and 14. On day 28, SOD and CAT activities were either not significantly different from the control or were higher, demonstrating a temporal multiphasic response of anti-oxidant enzymes. GST activity on day 28 was significantly reduced compared to controls. Acetylcholinesterase levels across the three-time points exhibited a complicated behaviour, with every exposure concentration being significantly different from the control. Chronic exposure negatively influences earthworm health status with elevated biomarker values analysed using IBRv2 index. This, in turn, impacted higher levels of hierarchical organization, significantly impairing reproduction and organismal homeostasis at the histological level and manifesting as decreasing cocoon formation and successful hatching events. Thus, the overall findings demonstrate that oxybenzone is toxic to Eisenia fetida at low-level, long-term exposure. Based on the concentration verification analysis and application of the EPA PestDF tool, oxybenzone undergoes single first-order kinetics degradation in OECD soil with DT50 and DT90 as 8.7-28.9 days, respectively.
... The apoptotic and neurotoxic effects of BP-3 on SH-SY5Y neuroblastoma cells was later confirmed by Broniowska et al. [82]. Wnuk et al. [83] showed that this compound is capable of inhibiting an autophagy and disturbing the epigenetic state of primary neuronal cell cultures by the inhibition of global DNA methylation as well as reduction of the activity of histone deacetylases (HDACs) and histone acetyl transferases (HATs). The global DNA hypomethylation and diminished activity of enzymes related to this process suggest the chromosomal instability and inappropriate gene expression pattern, leading to an increased risk of neurodevelopmental disorders or nerve degenerations. ...
Article
Full-text available
Children and adolescents are particularly vulnerable to skin damage caused by ultraviolet radiation and require intensified photoprotection. Benzophenone-3 (BP-3) belongs to the organic sunscreens, which are widely used in personal care and cosmetic products. However, the impact of BP-3 on human health requires a careful assessment. This review focuses on potentially harmful effect of this compound in relation to the developing organism. Studies show that BP-3, after topical application, can penetrate into bloodstream, blood-brain barrier and blood-placental barrier and may induce the reproductive toxicity and abnormal development of the foetus, endocrine system disruption and neurotoxicity in experimental animal models. So far, human studies have been scarce and controversial, therefore the cosmetics containing BP-3 should be carefully used by the pregnant women, children and adolescents.
Article
Full-text available
Background Benzophenone-3 (BP-3) and its major metabolite benzophenone-1 (BP-1) are widely used as UV filters in sunscreens and cosmetics to prevent sunburn and skin damage, or as stabilizers to prevent photodegradation in many commercial products. As a result, their presence is ubiquitous in the environment, wildlife and humans. Based on endocrine disruption concerns, international regulatory agencies are performing a closer evaluation. Objective and Methods: This work aimed to comprehensively review the available human relevant evidence for safety issues in MEDLINE/PubMed in order to create a structured database of studies, as well as to conduct an integrative analysis as part of the Human Biomonitoring for Europe (HBM4EU) Initiative. Results A total of 1,635 titles and abstracts were screened and 254 references were evaluated and tabulated in detail, and classified in different categories: i) exposure sources and predictors; ii) human biomonitoring (HBM) exposure levels to perform a meta-analysis; iii) toxicokinetic data in both experimental animals and humans; iv) in vitro and in vivo rodent toxicity studies; and v) human data on effect biomarkers and health outcomes. Our integrative analysis showed that internal peak BP-3 concentrations achieved after a single whole-body application of a commercially available sunscreen (4% w/w) may overlap with concentrations eliciting endocrine disrupting effects in vitro, and with internal concentrations causing in vivo adverse female reproductive effects in rodents that were supported by still limited human data. The adverse effects in rodents included prolonged estrous cycle, altered uterine estrogen receptor gene expression, endometrium hyperplasia and altered proliferation and histology of the mammary gland, while human data indicated menstrual cycle hormonal alterations and increased risk of uterine fibroids and endometriosis. Among the modes of action reported (estrogenic, anti-androgenic, thyroid, etc.), BP-3 and especially BP-1 showed estrogenic activity at human-relevant concentrations, in agreement with the observed alterations in female reproductive endpoints. The meta-analysis of HBM studies identified a higher concern for North Americans, showing urinary BP-3 concentrations on average 10 and 20 times higher than European and Asian populations, respectively. Discussion and Conclusions: Our work supports that these benzophenones present endocrine disrupting properties, endorsing recent European regulatory efforts to limit human exposure. The reproducible and comprehensive database generated may constitute a point of departure in future risk assessments to support regulatory initiatives. Meanwhile, individuals should not refrain from sunscreen use. Commercially available formulations using inorganic UV filters that are practically not absorbed into systemic circulation may be recommended to susceptible populations.
Article
Benzophenones (BPs) are endocrine disruptors frequently used in sunscreens and food packaging as UV blockers. Our goal was to assess the effect of benzophenone 2 (BP2) and 3 (BP3) on gene expression related to autophagy process and ER stress response in pancreatic beta cells. To that end, the mouse pancreatic beta cell line MIN6B1 was treated with 10 µM BP2 or BP3 in the presence or absence of the autophagy-inhibitor chloroquine (CQ, 10 µM) or the autophagy-inducer rapamycin (RAPA, 50 nM) during 24 h. BP3 inhibited the expression of the autophagic gene Ulk1, and additional effects were uncovered when autophagy was modified by CQ and RAPA. BP3 counteracted CQ-induced Lamp2 expression but did not compensate CQ-induced Sqstm1/p62 gene transcription, neither BP2. Nevertheless, the BPs did not alter the autophagic flux. In relation to ER stress, BP3 inhibited unspliced and spliced Xbp1 mRNA levels in the presence or absence of CQ, totally counteracted CQ-induced Chop gene expression, and partially reverted CQ-induced Grp78/Bip mRNA levels, while BP2 also partially inhibited Grp78/Bip mRNA induction by CQ. In conclusion, BPs, principally BP3, affect cellular adaptive responses related to autophagy, lysosomal biogenesis, and ER stress in pancreatic beta cells, indicating that BP exposure could lead to beta cell dysfunction.