You are on page 1of 15

Electromagnetic Biology and Medicine

ISSN: (Print) (Online) Journal homepage: www.tandfonline.com/journals/iebm20

Role of NADPH oxidase in MAPK signaling


activation by a 50 Hz magnetic field in human
neuroblastoma cells

María Antonia Martínez, Alejandro Úbeda & María Ángeles Trillo

To cite this article: María Antonia Martínez, Alejandro Úbeda & María Ángeles Trillo
(2021) Role of NADPH oxidase in MAPK signaling activation by a 50 Hz magnetic field in
human neuroblastoma cells, Electromagnetic Biology and Medicine, 40:1, 103-116, DOI:
10.1080/15368378.2020.1851250

To link to this article: https://doi.org/10.1080/15368378.2020.1851250

© 2020 The Author(s). Published with View supplementary material


license by Taylor & Francis Group, LLC.

Published online: 19 Dec 2020. Submit your article to this journal

Article views: 1449 View related articles

View Crossmark data Citing articles: 3 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iebm20
ELECTROMAGNETIC BIOLOGY AND MEDICINE
2021, VOL. 40, NO. 1, 103–116
https://doi.org/10.1080/15368378.2020.1851250

Role of NADPH oxidase in MAPK signaling activation by a 50 Hz magnetic field in


human neuroblastoma cells
María Antonia Martínez, Alejandro Úbeda, and María Ángeles Trillo
Servicio BEM, Dept. Investigación, Hosp, Univ. Ramón Y Cajal– IRYCIS, Madrid, Spain

ABSTRACT ARTICLE HISTORY


Our previous studies have shown that intermittent exposure to a 50-Hz, 100-µT sine wave magnetic field Received 30 April 2020
(MF) promotes human NB69 cell proliferation, mediated by activation of the epidermal growth factor Accepted 1 November 2020
receptor (EGFR) and pathways MAPK-ERK1/2 and p38; being the effects on proliferation and p38 KEYWORDS
activation blocked by the chelator N-acetylcysteine. The present work investigates the MF effects on NB69; ELF; magnetic field;
free radical (FR) production, and the potential involvement of NADPH oxidase, the main source of MAPK; p67phox; ROS
reactive oxygen species (ROS), in the MF-induced activation of MAPK pathways. To this end, the field
effects on MAPK-ERK1/2, -p38 and -JNK activation in the presence or absence of the NADPH oxidase
inhibitor, diphenyleneiodonium chloride (DPI), as well as the expression of the p67phox subunit, were
analyzed. The results revealed that field exposure increases FR production and induces early, transient
expression of the cytosolic component of the NADPH oxidase, p67phox. Also, the MF-induced activation
of the MAPK-JNK pathway, but not that of -ERK1/2 or -p38 pathways, was prevented in the presence of
the DPI, which has been shown to significantly reduce p67phox expression. These data, together with
those from previous studies, identify various, FR-dependent or -independent mechanisms, involved in
the MF-induced proliferative response mediated by MAPK signaling activation.

Introduction
2007; Mattsson and Simkó 2014; Wang and Zhang 2017;
Although the epidemiological evidence of an association Yakymenko et al. 2016). For instance, in vitro exposure to
between chronic exposure to extremely low frequency 50 Hz MF at 0.1–0.4 mT induces rapid (15–30 min)
(ELF) magnetic fields (MF) and the incidence of cancer is increases of total ROS or mitochondrial ROS in human
limited (Carlberg et al. 2017; De Roos et al. 2001; Turner amniotic epithelial cells (Feng et al. 2016; Sun et al. 2018),
et al. 2017), expert groups have considered such fields as and activates cytoprotective mechanisms related to redox
possible cancer promoters (IARC 2002; SCENIR 2009). systems, promoting cell proliferation and malignancy in
Also, there is a large body of experimental evidence that the SH-SY5Y human neuroblastoma line (Falone et al.
various human cell types are responsive to short-term 2017). Although most studies describe an increase in ROS
exposure to weak ELF MF, having been the power fre­ levels in response to MF, others report decreases or no
quency (50–60 Hz) fields and their potential effects on changes in ROS content (Hong et al. 2012; Song et al.
DNA damage (Ruiz-Gomez and Martinez-Morillo 2009), 2018). It has been proposed that this variety of responses
cell proliferation (Sulpizio et al. 2011; Trillo et al. 2012), cell could be due to differences in the experimental procedures
differentiation (Ayşe et al. 2010) or apoptosis (Basile et al. applied, including, among other factors, the MF frequency
2011) extensively investigated. Nevertheless, other studies or flux density, the exposure cycle or interval, the cell type,
have not obtained similar results (Santini et al. 2009) and, the biological parameters examined or the presence of
in any case, the physical and biological mechanisms under­ exogenous stressors (Mattsson and Simkó 2014).
lying the bioeffects induced by these MFs have not yet been Previous studies by our group have shown that 50 Hz
described and characterized sufficiently. MF at magnetic flux densities (B) of 10 or 100 µT, applied
Oxidative stress due to disturbances in redox home­ intermittently (3 h On/3 h Off or 5 min On/10 min Off)
ostasis that increases the levels of reactive oxygen species during 24–63 h hours, significantly increase DNA synthesis
(ROS) has been proposed as one of the potential mechan­ and cell proliferation in two human cancer lines: hepato­
isms through which radiofrequency/microwave (RF/MW) cellular carcinoma HepG2 and neuroblastoma NB69
radiation and ELF-MF exposure affect cellular behaviour (Martínez et al. 2012; Trillo et al. 2012). In NB69 cells the
(Consales et al. 2012; Falone et al. 2018; Friedman et al. reported effects were mediated by MF-induced alterations

CONTACT María Antonia Martínez. m.antonia.martinez@hrc.es Servicio BEM, Dept. Investigación, Hosp, Univ. Ramón Y Cajal– IRYCIS, Madrid, Spain.
Supplemental data for this article can be accessed on the publisher’s website.
© 2020 The Author(s). Published with license by Taylor & Francis Group, LLC.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd
/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon
in any way.
104 M. A. MARTÍNEZ ET AL.

in the activation of pathways MAPK-ERK1/2 and p38 strongly involved in the regulation of cell proliferation, the
(Martínez et al. 2012, 2016). The results also showed that DPI inhibitor was used in the present study to investigate
the free radical (FR) chelator N-acetylcysteine (NAC) a potential, NADPH-mediated mechanism, through which
blocks the MF effects on p38 activation and cell prolifera­ a weak, power frequency MF could activate the MAPK
tion, but not those on ERK1/2 activation, which reveals (ERK, p38 and JNK) pathway.
that FR intervene in some of these processes. More recent
results have shown that the MF-induced proliferative
response is mediated by the epidermal growth factor recep­ Methods
tor (EGFR) through the field effects on the aforementioned Cell culture
signaling pathways (Martínez et al. 2019).
Reactive oxygen species are ubiquitous in mamma­ The neuroblastoma cell line NB69 (lot No. 03I019/2008,
lian cells, and it is widely accepted that low levels of ROS item No. 99072802) was purchased from the European
can act as subcellular messengers in regulatory routes of Collection of Authenticated Cell Cultures (ECACC,
gene expression and signal transduction, such as the Salisbury, UK). The cells were periodically tested for myco­
EGFR pathway (Tseng et al. 2012), which is involved plasma contamination (PCR) and response to chemical and
in relevant physiological processes, such as cell prolif­ physical treatments. Cells were cultured for four days in
eration and differentiation or apoptosis. On the other a cell incubator (Thermo Fisher Scientific, Waltham, MA,
hand, high levels of ROS can alter cellular physiological USA) set at 37°C, 5% CO2 in moist air, using Dulbecco’s
processes by attacking membrane phospholipids, dis­ Minimum Essential Medium (DMEM, Lonza, Verviers,
rupting mitochondrial functions or damaging proteins Belgium) supplemented with 10%, heat inactivated fetal
and DNA (Schieber and Chandel 2014). bovine serum (FBS, Gibco, Thermo Fisher Scientific),
ROS are generated by both mitochondrial and non- 2 mM L-glutamine and 100 U/ml Antibiotic-Antimycotic
mitochondrial pathways. In the latter, NOX, the oxidase (Gibco, Thermo Fisher Scientific). In each experimental
of the nicotinamide adenine dinucleotide phosphate run the cell suspension was seeded (4.5 × 104 cells ml−1)
(NADPH), is the main contributor to the production of either directly on the bottom of the Petri dishes (Nunc,
ROS. ROS derived from NOX activity have been reported LabClinics, Barcelona, Spain) or on 12-mm diameter glass
to be closely related to mechanisms involved in cancerous coverslips placed inside the dishes. When needed, black 96-
processes (Paletta-Silva et al. 2013). The NOX complex is well microplates were used. A pre-incubation of 1 hour
formed by a membranal and a cytosolic component. The before the MF exposure was carried out using the cell-
membranal one, called cytochrome b558, is made up of permeable fluorogenic ROS probe, 2´,7´-dichlorodihydr
subunits p22phox and gp91phox (NOX2) and is the cata­ ofluorescein diacetate (DCFH-DA, Sigma-Aldrich, St. Lou
lytic part of the complex. The cytosolic component is is, MO, USA), the ROS scavenger, N-acetyl-L-cysteine
formed by three subunits: p40phox, p47phox and p67phox (NAC, Sigma-Aldrich), and the NADPH oxidase (NOX)
that, together with the GTPase Rac, regulate the activity of inhibitor, diphenyleneiodonium chloride (DPI, Santa Cruz
the catalytic complex. When activation takes place, the Biotechnology, Dallas, Texas, USA.). Stock compounds of
cytosolic components migrate to the membranes, where DCFH-DA, NAC and the NOX inhibitor DPI were dis­
they associate with the membrane-bound components to solved in dimethyl sulfoxide (DMSO, Sigma-Aldrich),
assemble the catalytically active oxidase responsible for milliQ water and ethanol, respectively. A volume of the
superoxide production (Babior et al. 1999). For the latter vehicle (DMSO, milliQ water-, or ethanol-only) equivalent
to occur, the p67phox cytoplasmic subunit must interact to that added to the experimental samples, was added to
with NOX2 (Italiano et al. 2012). It has been proposed that control samples in each experimental run. The final con­
NOX could amplify the receptor-mediated tyrosine kinase centration of vehicle in the culture was less than 0.1%.
signal, contributing to carcinogenesis through regulation of
cell signaling pathways such as MAPK-p38, -ERK1/2 or -
Magnetic field exposure protocol
JNK (Paletta-Silva et al. 2013; Rezatabar et al. 2019).
Diphenyleneiodonium (DPI), one of the most com­ The cultures were exposed to a 50 Hz, sine wave, vertically
monly used inhibitors of NOX activity, has been reported polarized MF, at a magnetic flux density BAC = 100 µT rms.
to inhibit cytoplasmic and mitochondrial ROS production The exposure set-up has been described elsewhere (Trillo
(Altenhöfer et al. 2015; Li and Trush 1998) and to limit et al. 2012). Briefly, the current flow was supplied by a wave
tumor cell growth in vitro when administered at nanomo­ generator (Newtronic Model 200MSTPC, Madrid, Spain)
lar concentrations (Doroshow et al. 2013). Thus, on the having a 3.53 mA DC offset (BDC = 15 µT rms). The
basis of our previous results on NB69 response to MF, and generator was connected to a pair of coils set in Helmholtz
since the NADPH oxidase complex has been shown to be configuration. One Helmholtz pair was placed inside each
ELECTROMAGNETIC BIOLOGY AND MEDICINE 105

of two magnetically shielded chambers (co-netic metal; analysis. At least three independent replicates were per­
Amuneal Corp., Philadelphia, PA, USA) located within formed per experimental condition.
two identical CO2 incubators (Thermo Fisher Scientific).
The electric current in the coils was monitored using
a multimeter (Hewlett Packard, model 974A, Loveland, NADPH oxidase inhibition
CO, USA) and the induced MF was routinely checked
On day four after seeding, one hour prior to MF- or sham-
using two magnetometers (EFA-3, Wandel and Golterma
exposure, the media were renewed and supplemented with
nn, Eningen, Germany, and EMDEX II, Enertech
1 µM DPI, a concentration previously tested by other
Consultants, Campbell, CA, USA). The background MF
authors (Alvarez-Maqueda et al. 2004). After 20 minutes
inside the shielded chambers was BAC: 0.05 ± 0.02 µT (rms);
of MF exposure, the levels of phosphorylation of the
BDC: 0.04 ± 0.04 µT (rms). In each experimental run Petri
Mitogen-Activated Protein Kinases (MAPK-p38, -ERK1
dishes (at least three per experimental group) or multiwells,
/2 and -JNK) were assessed by Western blotting and
containing the cell samples, were stacked in the central
immunofluorescence analysis. In preliminary experiments
region of the Helmholtz coil gap to ensure uniformity of
aiming to select optimal intervals for DPI treatment and
MF exposure. In each experimental run only one set of coils
MF exposure, tests were performed to analyze, indepen­
was energized, the samples in the unenergized set being
dently and at different time intervals, the DPI effects on
considered sham-exposed controls. Following a random
p67phox expression and MAPK activation in the presence
sequence, both coil sets and incubators were alternatively
of MF (supplementary files 1 and 2).
used for MF- or sham-exposure. NB69 cultures were MF-
or sham-exposed for 5 to 30-minute intervals at day 4
postplating.
Western blotting
To assess the expression levels of subunit p67phox and the
Free radical detection phosphorylated proteins p-p38, p-ERK1/2 and p-JNK,
The determination and quantification of FR production Western blotting was conducted as described elsewhere
were carried out using the fluorescent probe 2′,7-Dichloro (Martínez et al. 2012). Briefly, after MF- or sham-
dihydrofluorescein diacetate (5 µM DCFH-DA, Sigma- exposure the cells were harvested in hypotonic lysis buffer
Aldrich), which is routinely used for the identification of and proteins were separated and transferred to nitrocellu­
ROS (Piras et al. 2018). ROS production was assessed by lose membranes (Hybond ECL, GE Healthcare, Little
quantification of the intracellular oxidative transformation Chalfont, Buckinghamshire, UK). The membranes were
of the oxidation-sensitive probe DCFH-DA into the fluor­ blocked and incubated overnight at 4°C with mouse mono­
escent dye dichlorofluorescein (DCF). On day four post- clonal antibody against p-p38 (1:1000 dilution, 44–684 G,
plating the induction assay was performed. Cells were Thermo Fisher Scientific), rabbit polyclonal antibodies
treated with or without 1 mM NAC and 5 µM DCFH- against p-ERK1/2 (1:1000 dilution; 44–680 G, Thermo
DA and maintained in the dark at 37°C and 5% CO2 Fisher Scientific), p-SAPK/JNK (1:1000 dilution; Cell
during 1-hour before MF- or sham-exposure. The used Signaling Technology, Inc, 4668, Danvers, MA, USA) or
DCFH-DA and NAC concentrations were selected after goat polyclonal antibody against p67phox (1:500 dilution,
comprehensive literature search (Martínez et al. 2016; Piras sc-7662, Santa Cruz Biotechnology). Monoclonal mouse
et al. 2018). The samples were distributed in each of the two anti- β-Actin (1:5000; A5441, Sigma-Aldrich) was used as
sets of coils placed inside the MF-shielded chambers and loading control. After incubation with the indicated pri­
submitted to 10 minutes of MF- or sham-treatment fol­ mary antibodies, the proteins of interest were detected
lowed by 30 minutes of postincubation (10 min On/30 min using peroxidase-conjugated secondary antibodies (GE
Off). Four different experimental conditions were assayed: Healthcare) or fluorescently labeled secondary antibody
sham-exposure in the absence of NAC (controls); NAC IRdye (LI-COR, Bioscience, Lincoln, NE, USA). ProXima
only; MF exposure only; NAC + MF. For DCFH-DA- Imaging system (Isogen Life Science, Utrecht, Netherlands)
fluorescence spectroscopy analysis, cell cultures incubated was used to detect ECL-chemiluminescence in the blots,
with DCFH-DA were established in black 96-well micro­ and fluorescence signal was revealed by infrared imaging
plates (3 × 103 cells/well). After treatment, the samples Odyssey (LI-COR). The bands obtained were evaluated by
were washed with PBS and read on a microplate reader densitometry (PDI Quantity One 4.5.2 software, BioRad,
(TECAN SpectraFluor, Gödrig, Austria; λexc 490, λemi Munich, Germany) and normalized against the corre­
535). The fluorescence values vs. background (fluorescence sponding β-Actin band. At least four experimental repli­
in the absence of DCFH-DA) were processed for statistical cates were conducted for each protein.
106 M. A. MARTÍNEZ ET AL.

Immunofluorescence intervals (Figure 1(a, b)). In turn, the immunocyto­


chemical study revealed a significantly increased rate of
After MF- or sham-exposure of the samples cultured on
cells expressing p67phox (p67phox+) after 10 minutes of
coverslips, ERK1/2, p38 and JNK activation and p67phox
MF exposure (Figure 1(c, d)). The same increase in
expression were characterized by indirect immunofluores­
p67phox+ cells was observed at 15 minutes of exposure,
cence and computer-assisted image analysis. Cells were
although the effect was not accompanied by an increase
fixed with 4% formaldehyde, permeabilized with ethanol/
in the protein expression, which could be indicative of
acetic acid for 20 min at −4°C and blocked with PBS
a rapid drop in the amount of p67phox per cell. No
containing 10% goat serum for 1 hour at room tempera­
significant changes were found in the rate of p67phox+
ture. Coverslips were incubated overnight with goat pri­
cells in samples exposed for shorter or longer intervals.
mary antibody against p67phox (1:50; Santa Cruz
Biotechnology), with mouse monoclonal antibody against
p-p38 (1:100 dilution, Cell Signaling Technology), or with MF effects on free radical levels
rabbit antibody against p-ERK1/2 (1:100, Thermo Fisher
NB69 cells were MF- or sham-exposed and treated
Scientific) or p-JNK (1:100; Abcam, Cambridge, UK).
with DCFH-DA in the presence or absence of the
Secondary anti-goat conjugated with the fluorophore
free radical chelating agent NAC. This chelator has
Alexa Fluor 488 (Thermo Fisher Scientific), anti-mouse-
been shown adequate for assessment of the MF-
IgG conjugated to AlexaFluor® 546 or anti-rabbit IgG con­
induced response of the MAPK pathway in NB69
jugated to AlexaFluor® 488 (Molecular Probes, Eugene, OR,
cells (Martínez et al. 2016) and, apart from having
USA) were used to reveal the proteins of interest. The cell
a scavenging role, NAC can suppress superoxide gen­
nuclei were counterstained with Hoechst 33342 (Bisbenzi
eration in human neutrophils by restraining the trans­
mide, Sigma-Aldrich). Images were captured with a Nikon
location of p47phox or p67phox to the cell membrane
microscopy (Eclipse TE300, 1076 ER Amsterdam,
(Kitaoka et al. 2005). The fluorescent DCF due to
Netherlands) and analyzed by computer imaging Analy-
intracellular oxidation of DCFH-DA was analyzed by
SIS software (GMBH, Munich, Germany). In each of at
fluorescence spectroscopy and expressed in terms of
least three experimental runs, three coverslips were studied
ROS production. After 10 minutes of MF exposure
per experimental condition: sham-exposed controls, MF
followed by a 30-minute post-exposure incubation,
only and, when needed, the NOX inhibitor DPI + MF.
the intensity of fluorescent DCF and therefore, of FR
Fifteen microscope-fields per coverslip were randomly
production, were significantly increased (26.67% ±
selected for analysis. The total number of nuclei and
5.9% over controls, Figure 2). Such an increase was
the percent of p-p38, p-ERK1/2, or p67phox positive cells
not observed in samples exposed to the MF in the
per microscope-field were recorded.
presence of NAC.

Statistical analysis
MF effect on MAPK-p38 activation in the presence of
All experimental procedures and analysis were con­ DPI
ducted blindly for treatment. Data were normalized
The potential role of NADPH activation in the MF-
and expressed as means ± standard error (SEM) of at
induced expression of phosphorylated MAPK was inves­
least three independent experimental runs. Statistical
tigated at 20 minutes of field exposure. This time-lapse
analyses were performed with Graph-Pad Prism 6.01
was selected on the basis of the results of preliminary
software (GraphPad Software, Inc., La Jolla, CA, USA).
Western blotting tests, which revealed a simultaneous
The two-tailed Student’s t-test was used when compar­
and statistically significant activation peak for the three
ing two samples and the one-way ANOVA was applied
MAPK proteins studied (p38, ERK1/2 and JNK) at
when comparing multiple samples. The limit of statisti­
20 minutes of MF exposure, but not at shorter or longer
cal significance was set at p < .05.
exposure intervals (supplementary file, Fig. S1). As
described in the methodological section, the DPI inhi­
Results bitor was added to the culture media 60 minutes before
the MF exposure onset. Therefore, although here we will
MF effects on p67phox expression
refer to the 20-minute treatments corresponding to the
The Western blotting analysis of the chronology of interval of simultaneous exposure to DPI + MF, in fact,
p67phox expression showed significant protein overex­ the total period of treatment with the inhibitor, whether
pression at 10 minutes of MF exposure and no effects at applied independently or in combination with the MF,
shorter (5 min) or longer (15, 20 or 30 min) exposure was 80 minutes.
ELECTROMAGNETIC BIOLOGY AND MEDICINE 107

MF C 5 min 10 min 15 min 20 min

Figure 1. Expression of protein p67phox and rate of p67phox+ cells after short-term MF exposure. (a) Western blotting quantification
of p67phox expression at different MF- or sham-exposure intervals (5–20 min). The data, normalized over the corresponding sham-
exposed controls, are means ± SEM of at least 4 experimental replicates per time interval, with 6 samples (3 MF- and 3 sham-exposed)
per replicate. The average raw values at t = 0 min (5521.0 ± 430.75) did not differ from those in controls sham-exposed during the
studied time interval (5876.0 ± 215.4 at 5–20 min). (b) Representative blots of p67phox at the different exposure (MF+) or sham-
exposure (MF-) intervals, using β-Actin as loading control. (c) Immunocytochemical and computer-assisted image quantification of the
rate of p67phox+ cells. The data, normalized over controls (line 100%), are means ± SEM of 4 experimental replicates. (d) Upper panel:
representative images of p67phox labeling (green) at the corresponding exposure intervals. Lower panel: Hoechst-stained nuclei (blue)
of the cells in the corresponding upper micrographs. Scale bar = 50 µm. **: 0.05 ≤p < .01 (ANOVA and Student’s t-test).

The results confirmed the preliminary observations of NADPH oxidase does not intervene in the MF-induced
significant p-p38 overexpression in cells exposed to MF activation of the p38 pathway.
only (28.3% ± 5.9% above controls, p < .01, Figure 3(a, b)).
A similar overexpression of p-p38 (24.1% ± 10.3%, p < .05)
MF effect on MAPK-ERK1/2 activation in the
was observed in samples exposed to the MF in the presence
presence of DPI
of DPI, while when DPI was applied in sham-exposure
conditions, a not statistically significant subexpression of The Western blotting data summarized in Figure 4(a, b)
p38 was observed (16.11% ± 7.9% below controls). These confirm preliminary data on significant p-ERK1/2 over­
results are consistent with those from the immunocyto­ expression in response to the exposure to MF only. Such
chemical analysis (Figure 3(c, d)), which revealed signifi­ increase with respect to the expression levels in
cant increases in p-p38+ cell rate, induced by MF both in untreated controls (MF-/DPI-) was not observed when
the presence and absence of DPI (10.7% ± 3.4% and 10.9% the MF was applied in the presence of DPI. This could be
± 5.4% over controls, respectively; p < .05), while the indicative that the inhibitor, which in the absence of MF
treatment with the inhibitor alone significantly decreased induced significant p-ERK1/2 subexpression with respe
the p-p38+ cell rate (11.44% ± 3.7% below controls; ct to untreated controls (−25.40% ± 4.8%; p < .001),
p < .05). Taken together, these results indicate that could partially block the field-induced overexpression.
108 M. A. MARTÍNEZ ET AL.

Figure 2. Impact of MF exposure (10 min On/30 min Off) on ROS production in the presence or absence of NAC (40 min), determined by
DCF-fluorescence microscopy. The data, normalized over sham-exposed controls (line 100%), are means ± SEM of 3 experimental
replicates, with 120 samples (30 sham-exposed, 30 MF, 30 NAC and 30 NAC+MF) per replicate. *: 0.01 ≤ p < .05; ***: p < .001 (ANOVA
and Student´s t-test).

However, the complementary immunocytochemical Discussion


study (Figure 4(c, d)) did not reveal significant changes
Previous studies by our group (Martínez et al. 2012,
with respect to untreated controls in the rate of p-ERK1/
2016, 2019) have shown that exposure to a 50 Hz, 100
2+ cells when DPI was administered in the absence of
µT MF activates the MAPK ERK1/2, p38 and JNK sig­
MF. On the other hand, MF exposure, both in the pre­
naling pathways in human neuroblastoma cells NB69.
sence and in the absence of the inhibitor, significantly
Indeed, the activation of p38 and ERK1/2, as well as that
increased the rate of cells expressing p-ERK1/2 com­
of the EGF receptor, are known to mediate the MF-
pared to untreated controls and to samples treated
induced proliferation promotion in NB69, and the fact
with DPI only. Therefore, taken together, these data do
that such proliferative response is blocked by NAC sug­
not support a potential implication of NADPH oxidase
gests that free radicals can be involved in the field’s
in the MF-induced activation of ERK1/2.
effect. Evidence exists that NADPH oxidase, an enzy­
matic system involved in ROS generation, could be
a common mediator for various biological processes
MF effect on MAPK-JNK activation in the presence of
including antioxidation/oxidative stress, various mito­
DPI
chondrial functions, calcium homeostasis, gene expres­
The Western blotting results illustrated in Figure 5(a, b) sion, immunological functions or cell aging and death
confirmed previously reported data (Martínez et al. (Nauseef 2008). Protein p67phox, one of the cytosolic
2016) that, compared to untreated controls, MF expo­ subunits of NADPH oxidase, regulates the activity of
sure induces significant overexpression of p-JNK. this enzyme complex through its interaction with the
However, the field effect was prevented by DPI, so that membrane component of the complex (Italiano et al.
under these conditions the p-JNK expression levels did 2012). The present study investigates the field effects
not differ from those of untreated controls nor from on free radical production and p67phox protein expres­
those of samples treated with the inhibitor only. These sion, as well as the potential involvement of NADPH
data were supported by the corresponding immunocy­ oxidase in the MF-induced activation of MAPK-p38, -
tochemical study (Figure 5(c, d)) and are indicative that ERK1/2 and -JNK signal transduction pathways.
the NADPH oxidase complex is involved in the MF- The obtained results show that the MF induces early
induced activation of the MAPK-JNK pathway. overexpression of the p67phox protein, which peaks at
ELECTROMAGNETIC BIOLOGY AND MEDICINE 109

Figure 3. Expression of p-p38 and rate of p-p38+ cells after 20 min of MF exposure in the presence or absence of DPI. (a) Western blot
quantification of p-p38. The data, normalized over the corresponding controls (line 100%), are means ± SEM of 4 experimental
replicates with 12 samples (3 sham-exposed, 3 DPI, 3 MF and 3 DPI + MF) per replicate. (b) Representative blots of p-p38 at the
different exposure conditions (MF+) or sham-exposure (MF-), using β-Actin as loading control. (c). Immunofluorescence quantification
of p-p38+ cells by computer-assisted image analysis. The data, normalized over the corresponding controls (line 100%), are means ±
SEM of 3 experimental replicates. (d) Representative images of p-p38 labeling (upper panel) and Hoechst-stained nuclei of the cells in
the corresponding upper micrographs (lower panel). Scale bar = 50 µm. *: 0.01 ≤ p < .05; **: 0.05 ≤ p < .01 (ANOVA and Student’s
t-test).

10 minutes of exposure. The mechanism through which at such level, both at 10 and 15 minutes of MF exposure.
the MF causes such a transient effect on this component In fact, as proposed in previous works (Martínez et al.
of the NADPH oxidase is unknown. Although some 2019), the AC frequency (f = 50 Hz) and DC flux density
studies, generally using chemical agents, have reported (BDC = 15 µT rms) parameters applied in this study
translocation of the NADPH oxidase subunits at short would meet the resonance conditions for the sulfur
times similar to those reported here (Touyz et al. 2002; ion, according to the ion parametric resonance model
Wang and Lou 2009), in most studies de novo protein IPR (Blackman et al. 1995; Blanchard and Blackman
synthesis has been observed at longer times (Seo et al. 1994). Therefore, it can be hypothesized that under
2012; Touyz et al. 2002). Therefore, it is conceivable the these conditions, a potential, MF-induced perturbation
effect described in the present study corresponds to the on this ion could change the reactivity of the thiol or
initial phases of the MF response, involving thiolate groups present in the cysteine residues of the
a translocation of preexisting p67phox from the cyto­ p67phox subunit (Bizouarn et al. 2016; Fradin et al.
plasm to the plasma membrane, rather than a de-novo 2018). Such an effect could promote the binding of
expression of the protein. Although additional studies p67phox to NOX2 and/or a translocation of p67phox
would be necessary to test this hypothesis and determine to the plasma membrane, and subsequently activate the
the location where the effect takes place, the possibility NADPH oxidase.
of a membrane location would obtain partial support On the other hand, increased expression of p67phox
from the immunocytochemical data illustrated in Figure does not necessarily imply activation of the NADPH
1(d), which showed a slight increase in p67phox labeling oxidase complex, which is made up of a set of subunits
110 M. A. MARTÍNEZ ET AL.

Figure 4. Expression of p-ERK1/2 and rate of p-ERK1/2+ cells after 20 min of MF exposure in the presence or absence of DPI. (a) Western
blot quantification of pERK1/2 expression. The data, normalized over the corresponding controls (line 100%) are means ± SEM of 4
experimental replicates with 12 samples (3 sham-exposed, 3 DPI, 3 MF and 3 DPI + MF) per replicate. (b) Representative blots of
p-ERK1/2, at the different exposure conditions (MF+) or sham-exposure (MF-), using β-Actin as loading control. (c)
Immunofluorescence quantification of p-ERK1/2+ cells by computer-assisted image analysis. Bars are means ± SEM of 4 experimental
replicates. Normalized values respect to sham-exposed controls (line 100%). (d) Representative images of p-ERK1/2 labeling (upper
panel) and Hoechst-stained nuclei of the cells in the corresponding upper micrographs (lower panel). Scale bar = 50 µm. *:
0.01 ≤ p < .05; ***: p < .001 (ANOVA and Student’s t-test).

whose specific functions and their interactions within types exposed to RF/MW (Durdik et al. 2019; Friedman
the complex have not been sufficiently elucidated yet. In et al. 2007; Yakymenko et al. 2016) or DC – ELF MF
what concerns subunit p67phox, it is the key component (Mattsson and Simkó 2014; Merla et al. 2019;
in the causation of a conformational remodeling of Poniedziałek et al. 2013; Rollwitz et al. 2004). In some of
NOX2 (Gorzalczany et al. 2000), and an activation these ELF studies, increases in ROS and cytoplasmic super­
domain in p67phox has been found essential for the oxides occurring after 5–45 minutes of exposure to flux
oxidase activation, but not for the actual p67phox- densities B ≥ 0.4 mT were inhibited by NADPH oxidase
NOX2 interaction (Sumimoto 2008). Also, experimental inhibitors (Feng et al. 2016; Lupke et al. 2004; Merla et al.
evidence exists supporting the hypothesis that the NOX2 2019). Although the present work does not address directly
dehydrogenase region (DHR) acts as a protein disulfide the effects of DPI on ROS production, other studies have
isomerase (PDI) that leads to formation of disulfide shown that DPI concentrations similar to those used in the
bonds with p67phox and ensures the stabilization of present work do not cause a reduction in free radical levels.
the NADPH oxidase complex (Bechor et al. 2015; Such studies show that DPI reverses the generation of
Fradin et al. 2018). reactive oxygen species induced by ELF MF in SH-SY5Y
The present results also reveal that ROS production is neuroblastoma cells (Merla et al. 2019) and does not inhibit
significantly increased by a 10-minute exposure to the MF the increased production of free radicals or superoxide
followed by a 30-minute post-exposure interval, this effect radical anions induced by MF in non-stimulated Mono-
being prevented by NAC. This supports observations by Mac-6 cells (Lupke el al. 2004). Overall, these MF-induced
other authors reporting ROS level increases in various cell increases in ROS levels were moderate and similar to those
ELECTROMAGNETIC BIOLOGY AND MEDICINE 111

Figure 5. Expression of p-JNK and rate of p-JNK+ cells after 20-minute MF exposure in the presence or absence of DPI. (a) Western
blotting quantification of p-JNK expression. Each bar, normalized over the corresponding controls (line 100%), are means ± SEM of 4
experimental replicates with 12 samples (3 sham-exposed, 3 DPI, 3 MF and 3 DPI + MF) per replicate. (b) Representative blots of p-JNK
at the different exposure conditions (MF+) or sham-exposure (MF-), using β-Actin as loading control. (c) Immunofluorescence
quantification of p-JNK+ cells by computer-assisted image analysis. The data, normalized over the corresponding controls (line
100%), are means ± SEM of 3 experimental replicates. (d) Representative images of p-JNK labeling (upper panel) and Hoechst-stained
nuclei of the cells in the corresponding upper micrographs (lower panel). Scale bar = 50 µm. *: 0.01 ≤ p < .05; **: 0.05 ≤ p < .01; ***:
p < .001 (ANOVA and Student’s t-test).

obtained in the present study (about 26%). While high ROS activation mediates the stimulation or blockade of cell
levels can cause severe cell dysfunction through DNA, cycle progression in various cell types (Tormos et al.
RNA and protein damage, moderate levels can act 2013; Venkatachalam et al. 2008). Concerning p67phox
as second messengers, activating signaling cascades and expression, although no evidence of direct effects of DPI
triggering cellular responses affecting proliferation, apop­ has been reported so far, the possibility cannot be dis­
tosis or gene expression (Schieber and Chandel 2014). In regarded that DPI may induce indirect effects through
any case, the mechanism or mechanisms through which an action on the p67phox-regulated flavin adenine dinu­
MF-induced ROS could affect specific biological systems cleotide (FAD) redox center of NADPH oxidase (Han
are not yet sufficiently characterized. and Lee 2000). From this, it can be stated that the field-
The present study applied inhibitor DPI to investigate induced activation of ERK1/2 and p38 at 20 minutes of
the potential involvement of NADPH oxidase in the exposure was not affected by the presence of DPI, which
field-induced activation of MAPK-ERK1/2, -p38 and - indicates that such activation would not be mediated by
JNK. In the absence of MF, DPI caused significant sub­ NADPH oxidase.
expression of the cytosolic component of NADPH oxi­ In the presence of the DPI inhibitor only, the phos­
dase, p67phox (supplementary file 2, Fig. S2), as well as phorylation levels of MAPK-p38 did not decrease sig­
subexpression of p-ERK1/2 (Figure 4(a)) and decreased nificantly with respect to those in controls. However,
rate of cells showing p38 activation (Figure 3(c)). These after a 20-minute interval, during which the MF signifi­
data are consistent with those reporting that, through cantly increased phosphorylation of this pathway (sup­
their involvement in the regulation of p38 and ERK plementary Figure 1), the expression of the p67phox
activation, the ROS generated by NADPH oxidase subunit of NADPH oxidase was significantly reduced
112 M. A. MARTÍNEZ ET AL.

by DPI (supplementary Figure 2). These results are in the NB69 cell line (Martínez et al. 2019). However, the
indicative that a critical treatment time is necessary for possibility that a direct relationship exists between ROS
the response of each of the different MAPKs to DPI to be induction and EGFR activation in NB69 has not yet been
detected. Indeed, while the field-induced phosphoryla­ investigated. On the other hand, although the NADPH
tion in pathways p38 and JNK declines rapidly, that oxidase complex (different NOX isoforms) has been
induced in pathway ERK1/2 lasts longer ( described to be involved in the regulation of EGFR activa­
Supplementary Figure 1), so a significant response of tion in various cell types (Heppner and Van der Vliet
this pathway to DPI was observed at 20 minutes of 2016), such activation may also be mediated by mechan­
treatment (Figure 2). It could be argued that since DPI isms independent of ROS, including metalloproteinases or
has been reported to have potential pro-oxidant effects G proteins capable of releasing ligands that are specific to
(Kučera et al. 2016), the possibility that such effects EGFR, tyrosine kinases or intracellular Ca2+ (Forrester
could have increased the cell sensitivity to the MF in et al. 2016; George et al. 2013).
our experiments cannot be ruled out. However, the As for the activation of JNK by the MF, our results are
obtained results showing non-significant differences compatible with those showing that short-term exposure
with respect to controls indicate that the effect, if any, (15–30 min) to 0.4 or 0.8 mT fields promotes activation of
would be rather weak. the stress-activated protein kinase pathway (SAPK/JNK) in
In contrast to the above-described results, NADPH the hamster lung fibroblast cell line CHL (Sun et al. 2001).
oxidase has been reported to be involved in ERK1/2 activa­ Nevertheless, the potential cellular responses deriving from
tion in immortalized monkey fibroblasts (COS7) and in these changes occurring at the molecular level have not yet
human epithelial cells (HeLa) exposed in vitro to a 50 Hz, 1 been elucidated. Our previous studies on pharmacological
mT MF (Kapri-Pardes et al. 2017). These cell types, on the inhibition of the SAPK/JNK pathway have ruled out an
other hand, responded to the MF with early ERK or p38 association between its activation and NB69 cell prolifera­
activation peaks (at 5–17 min of exposure), which is con­ tion, both induced by the MF (Martínez et al. 2016).
sistent with the herein reported early ERK and p38 activa­ However, the possibility cannot be ruled out that SAPK/
tions in response to the 0.1 mT MF. Similar effects on ERK JNK activation may contribute to the proliferative response
have also been observed in response to short-term expo­ through a potential increase in cell survival. In fact, JNK
sure to RF electromagnetic fields. For instance, subthermal pathway activation has been shown to be involved in
exposure to an 875 MHz signal has been reported to induce increased survival of human neuroblastoma cells and in
rapid ERK phosphorylation in isolated plasma membranes resistance to the treatment with chemical oncostatics, and
from Rat1 and HeLa cells, this effect being mediated by has been associated with poor prognosis in oncology
field-induced activation of NADH oxidase, which gener­ (Sheikh et al. 2016; Wu et al. 2019).
ates ROS at the plasma membrane (Friedman et al. 2007). As regards the MAPK-JNK pathway activation, the
Regarding the activation of the MAPK-p38 pathway by ROS generated by the activity of NADPH oxidase is
the MF, previous studies by our group had shown that this known to be involved in the activation of JNK in different
pathway is inhibited by NAC (Martínez et al. 2016), which cell types, including liver or nervous system cells (Li et al.
together with the present results, suggests that the p38 2017; Yan et al. 2013). The present results showing that,
activation by the MF would be mediated by a source of although the NADPH oxidase inhibitor alone does not
FR, perhaps of mitochondrial origin, other than NADPH. affect the expression of p-JNK at 20 min of treatment, it is
In fact, several studies have provided evidence of potential capable of preventing the MF-induced JNK activation,
involvement of mitochondrial electron transfer processes due perhaps to underexpression of the p67phox subunit
in the RF/ELF-induced ROS generation (Consales et al. at this time interval (Figure 2). These results suggest that
2012; Destefanis et al. 2015; Feng et al. 2016; Kesari et al. the field effect on the JNK pathway activation would be
2016; Luukkonen et al. 2014; Wang and Zhang 2017). For mediated by NADPH oxidase.
instance, Feng et al. (2016) reported that exposure to
a 50 Hz, 0.4 mT MF promotes ROS production in amniotic
Conclusion
cells through two pathways: the NADPH oxidase complex
and the mitochondria, the production of cytoplasmic per­ In conclusion, the present results reveal that in vitro expo­
oxides (from 5 min of exposure) being earlier than that of sure to a 50 Hz, 100 µT MF increases the total ROS levels in
mitochondrial ROS (at 15 or 30 min). Besides, ROS inhibi­ NB69 cells and induces early, transient expression of the
tion by DPI prevented the MF-induced formation of EGFR cytosolic component of the NADPH oxidase, p67phox.
clusters. Recent studies by our group have revealed that Also, the MF-induced activation of the MAPK-JNK path­
EGFR is involved in the MF-induced proliferative response way, but not that of -ERK1/2 or -p38 pathways, was pre­
and activation of the MAPK pathways ERK, p38 and JNK vented in the presence of the used NADPH inhibitor,
ELECTROMAGNETIC BIOLOGY AND MEDICINE 113

Figure 6. Schematic representation, based on the present (boxed texts) and previously published results, of the molecular mechanism
that triggers the MF-induced proliferative response of the NB69 cell line, and of the involvement of free radicals (FR) in said mechanism.
Our previous studies reported that activation of the EGF receptor (EGFR) would trigger cell proliferation through phosphorylation of
pathways ERK1/2 (FR-independent) and p38 (FR-dependent). EGFR also activated the JNK pathway, which did not intervene in the
proliferative response to the MF (see the Discussion for detailed explanation). The present results show that the MF increases both the
expression levels of the p67phox protein, a component of the NADPH oxidase complex, and the total production of intracellular FR.
However, the analysis of the involvement of NOX in the activation of MAPK pathways revealed that only the JNK pathway was
prevented by DPI in the presence of MF. These data show that the NOX complex is not involved in the MF-induced proliferative effect
mediated by EGFR-p38-pERK.

which has been shown to significantly reduce p67phox assembly by human neutrophils. Effects on MAPK activa­
expression. Taken together with previously published tion and neutrophil migration. Atherosclerosis 172:229–38.
data (Martínez et al. 2016, 2019), the present results suggest doi:10.1016/j.atherosclerosis.2003.11.005.
that the proliferative response of NB69 to the MF exposure Ayşe, I. G., A. Zafer, O. Sule, I.-T. Işil, and T. Kalkan. 2010.
Differentiation of K562 cells under ELF-EMF applied at
is mediated by free radicals originated from sources other different time courses. Electromagn. Biol. Med. 29:122–30.
than NOX (Figure 6). On the other hand, since the JNK doi:10.3109/15368378.2010.502451.
pathway is known to be strongly involved in the regulation Babior, B. M. 1999. NADPH oxidase: An update. Blood
of cell survival and apoptosis (Wu et al. 2019), NADPH is 93:1464–76. https://www.ncbi.nlm.nih.gov/pubmed/
likely to mediate the field effects on such processes, thus 10029572.
Basile, A., R. Zeppa, N. Pasquino, C. Arra, M. Ammirante,
contributing to the proliferative response of NB69 to
M. Festa, A. Barbieri, A. Giudice, M. Pascale, M. C. Turco,
the MF. et al. 2011. Exposure to 50 Hz electromagnetic field raises
the levels of the anti-apoptotic protein BAG3 in melanoma
cells. J Cell. Physiol. 226:2901–07. doi:10.1002/jcp.22641.
Competing interests Bechor, E., I. Dahan, T. Fradin, Y. Berdichevsky, A. Zahavi,
A. F. Gross, M. Rafalowski, and E. Pick. 2015. The dehydro­
The authors declare that they have no competing interests. genase region of the NADPH oxidase component Nox2 acts
as a protein disulfide isomerase (PDI) resembling PDIA3
with a role in the binding of the activator protein p67
References (phox.). Front. Chem. 3:3. doi:10.3389/fchem.2015.00003.
Bizouarn, T., G. Karimi, R. Masoud, H. Souabni, P. Machillot,
Altenhöfer, S., K. A. Radermacher, P. W. M. Kleikers, X. Serfaty, F. Wien, M. Réfrégiers, C. Houée-Levin, and
K. Wingler, and H. H. H. W. Schmidt. 2015. Evolution of L. Baciou. 2016. Exploring the arachidonic acid-induced
NADPH oxidase inhibitors: Selectivity and mechanisms for structural changes in phagocyte NADPH oxidase p47
target engagement. Antioxid. Redox Signal. 23:406–27. (phox) and p67(phox) via thiol accessibility and SRCD
doi:10.1089/ars.2013.5814. spectroscopy. Febs J 283:2896–910. doi:10.1111/febs.13779.
Alvarez-Maqueda, M., R. El Bekay, J. Monteseirín, G. Alba, Blackman, C. F., J. P. Blanchard, S. G. Benane, and
P. Chacón, A. Vega, C. Santa María, J. R. Tejedo, D. E. House. 1995. The ion parametric resonance model
J. Martín-Nieto, F. J. Bedoya, et al. 2004. Homocysteine predicts magnetic field parameters that affect nerve cells.
enhances superoxide anion release and NADPH oxidase Faseb J 9:547–51. doi:10.1096/fasebj.9.7.7737464.
114 M. A. MARTÍNEZ ET AL.

Blanchard, J. P., and C. F. Blackman. 1994. Clarification and the 369 Cys-Gly-Cys 371 triad in Nox2 and in p67 phox.
application of an ion parametric resonance model for magnetic J. Leukoc. Biol. 104:1023–39. doi:10.1002/jlb.4a0418-173r.
field interactions with biological systems. Bioelectromagnetics Friedman, J., S. Kraus, Y. Hauptman, Y. Schiff, and R. Seger.
15:217–382. doi:10.1002/bem.2250150306. 2007. Mechanism of short-term ERK activation by electro­
Carlberg, M., T. Koppel, M. Ahonen, and L. Hardell. 2017. magnetic fields at mobile phone frequencies. Biochem. J.
Case-control study on occupational exposure to extremely 405:559–68. doi:10.1042/bj20061653.
low frequency electromagnetic fields and glioma risk. Am. George, A. J., R. D. Hannan, and W. G. Thomas. 2013.
J. Ind. Med. 60:494–503. doi:10.1002/ajim.22707. Unravelling the molecular complexity of GPCR-mediated
Consales, C., C. Merla, C. Marino, and B. Benassi. 2012. EGFR transactivation using functional genomics
Electromagnetic fields, oxidative stress, and neurodegenerati approaches. Febs J 280:5258–68. doi:10.1111/febs.12509.
on. Int. J. Cell. Biol. (2012:683897. doi:10.1155/2012/683897. Gorzalczany, Y., N. Sigal, M. Itan, O. Lotan, and E. Pick. 2000.
De Roos, A. J., K. Teschke, D. A. Savitz, C. Poole, S. Grufferman, Targeting of Rac1 to the phagocyte membrane is sufficient
B. H. Pollock, and A. F. Olshan. 2001. Parental occupational for the induction of NADPH oxidase assembly. J. Biol.
exposures to electromagnetic fields and radiation and the inci­ Chem. 275:40073–81. doi:10.1074/jbc.m006013200.
dence of neuroblastoma in offspring. Epidemiol 12:508–17. Han, C. H., and M. H. Lee. 2000. Activation domain in
doi:10.1097/00001648-200109000-00008. p67phox regulates the steady state reduction of FAD in
Destefanis, M., M. Viano, C. Leo, G. Gervino, A. Ponzetto, and gp91phox. J. Vet. Sci. 1:27–31.
F. Silvagno. 2015. Extremely low frequency electromagnetic Heppner, D. E., and A. Van der Vliet. 2016. Redox-dependent
fields affect proliferation and mitochondrial activity of regulation of epidermal growth factor receptor signaling.
human cancer cell lines. Int. J. Radiat. Biol. 91:964–72. Redox Biol 8:24–27. doi:10.1016/j.redox.2015.12.002.
doi:10.3109/09553002.2015.1101648. Hong, M. N., N. K. Han, H. C. Lee, Y.-K. Ko, S.-G. Chi, Y.-
Doroshow, J. H., S. Gaur, S. Markel, J. Lu, J. van Balgooy, S. Lee, Y.-M. Gimm, S.-H. Myung, and J.-S. Lee. 2012.
T. W. Synold, B. Xi, X. Wu, and A. Juhasz. 2013. Effects of Extremely low frequency magnetic fields do not elicit oxi­
iodonium-class flavin dehydrogenase inhibitors on growth, dative stress in MCF10A cells. J. Radiat. Res. 53:79–86.
reactive oxygen production, cell cycle progression, NADPH doi:10.1269/jrr.11049.
oxidase 1 levels, and gene expression in human colon can­ [IARC] International Agency for Research of Cancer. IARC
cer cells and xenografts. Free Radic. Biol. Med. 57:162–75. monograph on the evaluation of carcinogenic risks to
doi:10.1016/j.freeradbiomed.2013.01.002. humans, Vol. 80. Non-ionizing radiation, Part 1: Static
Durdik, M., P. Kosik, E. Markova, A. Somsedikova, and extremely low-frequency (ELF) electric and magnetic
B. Gajdosechova, E. Nikitina, E. Horvathova, K. Kozics, fields. Lyon, France: IARC Press. (2002). Accessed on 15
D. Davis, and I. Belyaev. 2019. Microwaves from mobile July 2020: http://publications.iarc.fr/Book-And-Report-
phone induce reactive oxygen species but not DNA damage, Series/Iarc-Monographs-On-The-Identification-Of-
preleukemic fusion genes and apoptosis in hematopoietic Carcinogenic-Hazards-To-Humans/Non-ionizing-
stem/progenitor cells. Sci. Rep. 9:16182. doi:10.1038/ Radiation-Part-1-Static-And-Extremely-Low-frequency-
s41598-019-52389-x. ELF-Electric-And-Magnetic-Fields-2002
Falone, S., S. Jr, S. V. Cordone, P. Cesare, A. Bonfigli, Italiano, D., A. M. Lena, G. Melino, and E. Candi. 2012.
M. Grannonico, G. Di Emidio, C. Tatone, M. Cacchio, Identification of NCF2/p67phox as a novel p53 target
and F. Amicarelli. 2017. Power frequency magnetic field gene. Cell Cycle 11:4589–96. doi:10.4161/cc.22853.
promotes a more malignant phenotype in neuroblastoma Kapri-Pardes, E., T. Hanoch, G. Maik-Rachline, M. Murbach,
cells via redox-related mechanisms. Sci. Rep. 7:11470. P. L. Bounds, N. Kuster, and R. Seger. 2017. Activation of
doi:10.1038/s41598-017-11869-8. signaling cascades by weak extremely low frequency elec­
Falone, S., S. Jr, S. V. Cordone, G. Di Emidio, C. Tatone, tromagnetic fields. Cell. Physiol. Biochem. 43:1533–46.
M. Cacchio, and F. Amicarelli. 2018. Extremely doi:10.1159/000481977.
low-frequency magnetic fields and redox-responsive path­ Kesari, K. K., J. Juutilainen, J. Luukkonen, and J. Naarala.
ways linked to cancer drug resistance: Insights from 2016. Induction of micronuclei and superoxide production
co-exposure-based in vitro studies. Front. Public Health. in neuroblastoma and glioma cell lines exposed to weak 50
6:33. doi:10.3389/fpubh.2018.00033. Hz magnetic fields. J. R. Soc. Interface 13:20150995.
Feng, B., A. Dai, L. Chen, L. Qiu, Y. Fu, and W. Sun. 2016. doi:10.1098/rsif.2015.0995.
NADPH oxidase-produced superoxide mediated a 50-Hz Kitaoka, N., G. Liu, N. Masuoka, K. Yamashita, M. Manabe,
magnetic field-induced epidermal growth factor receptor and H. Kodama. 2005. Effect of sulfur amino acids on
clustering. Int. J. Radiat. Biol. 92:596–602. doi:10.1080/0955 stimulus-induced superoxide generation and translocation
3002.2016.1206227. of p47phox and p67phox to cell membrane in human neu­
Forrester, S. J., T. Kawai, S. O’Brien, W. Thomas, R. C. Harris, trophils and the scavenging of free radical. Clin. Chim. Acta.
and S. Eguchi. 2016. Epidermal growth factor receptor 353:109–16. doi:10.1016/j.cccn.2004.10.011.
transactivation: Mechanisms, pathophysiology, and poten­ Kučera, J., L. Binó, K. Štefková, J. Jaroš, O. Vašíček, J. Večeřa,
tial therapies in the cardiovascular system. Annu. Rev. L. Kubala, and J. Pacherník. 2016. Apocynin and dipheny­
Pharmacol. Toxicol. 56:627–53. doi:10.1146/annurev- leneiodonium induce oxidative stress and modulate PI3K/
pharmtox-070115-095427. Akt and MAPK/Erk activity in mouse embryonic stem cells.
Fradin, T., E. Bechor, Y. Berdichevsky, W. Thomas, Oxid. Med. Cell. Longev. (2016:7409196. doi:10.1155/2016/
R. C. Harris, and S. Eguchi. 2018. Binding of p67 phox to 7409196.
Nox2 is stabilized by disulfide bonds between cysteines in Li, Y., and M. A. Trush. 1998. Diphenyleneiodonium, an
NAD(P)H oxidase inhibitor, also potently inhibits
ELECTROMAGNETIC BIOLOGY AND MEDICINE 115

mitochondrial reactive oxygen species production. Rezatabar, S., A. Karimian, V. Rameshknia, H. Parsian,
Biochem. Biophys. Res. Commun. 253:295–99. doi:10.1006/ M. Majidinia, T. A. Kopi, A. Bishayee, A. Sadeghinia,
bbrc.1998.9729. M. Yousefi, M. Monirialamdari, et al. 2019. RAS/MAPK
Li, Y. Y., Z. M. Shi, X. T. Yu, P. Feng, and X. J. Wang. 2017. signaling functions in oxidative stress, DNA damage
The effects of urotensin II on migration and invasion are response and cancer progression. J. Cell. Physiol.
mediated by NADPH oxidase-derived reactive oxygen spe­ 234:14951–65. doi:10.1002/jcp.28334.
cies through the c-Jun N-terminal kinase pathway in Rollwitz, J., M. Lupke, and M. Simkó. 2004. Fifty-hertz magnetic
human hepatoma cells. Peptides 88:106–14. doi:10.1016/j. fields induce free radical formation in mouse bone
peptides.2016.12.005. marrow-derived promonocytes and macrophages. Biochim.
Lupke, M., J. Rollwitz, and M. Simkó. 2004. Cell activating Biophys. Acta. 1674:231–38. doi:10.1016/j.bbagen.2004.06.024.
capacity of 50 Hz magnetic fields to release reactive oxygen Ruiz-Gómez, M. J., and M. Martínez-Morillo. 2009.
intermediates in human umbilical cord blood-derived Electromagnetic fields and the induction of DNA strand
monocytes and in Mono Mac 6 cells. Free Radic. Res. breaks. Electromagn. Biol. Med 28:201–14. doi:10.1080/
38:985–93. doi:10.1080/10715760400000968. 15368370802608696.
Luukkonen, J., A. Liimatainen, J. Juutilainen, and J. Naarala. Santini, M. T., G. Rainaldi, and P. L. Indovina. 2009. Cellular
2014. Induction of genomic instability, oxidative processes, effects of extremely low frequency (ELF) electromagnetic
and mitochondrial activity by 50Hz magnetic fields in fields. Int. J. Radiat. Biol. 85:294–313. doi:10.1080/0955300
human SH-SY5Y neuroblastoma cells. Mutat. Res. 0902781097.
760:33–41. doi:10.1016/j.mrfmmm.2013.12.002. [SCENIHR]Scientific Committee on Emerging and Newly
Martínez, M. A., A. Úbeda, M. A. Cid, and M. A. Trillo. 2012. Identified Health Risks: Health effects of exposure to
The proliferative response of NB69 human neuroblastoma EMF. (2009). Accessed on 15 July 2020: https://ec.europa.
cells to a 50 Hz magnetic field is mediated by ERK1/2 eu/health/ph_risk/committees/04_scenihr/docs/scenihr_o_
signaling. Cell. Physiol. Biochem. 29:675–86. doi:10.1159/ 022.pdf
000178457. Schieber, M., and N. S. Chandel. 2014. ROS function in redox
Martínez, M. A., A. Úbeda, J. Moreno, and M. A. Trillo. 2016. signaling and oxidative stress. Curr. Biol. 24:R453–462.
Power frequency magnetic fields affect the p38 MAPK doi:10.1016/j.cub.2014.03.034.
mediated regulation of NB69 cell proliferation implication of Seo, J. S., J. Y. Park, J. Choi, T. K. Kim, J. H. Shin, J. K. Lee, and
free radicals. Int. J. Mol. Sci. 17:510. doi:10.3390/ijms17040510. P. L. Han. 2012. NADPH oxidase mediates depressive beha­
Martínez, M. A., A. Úbeda, and M. A. Trillo. 2019. Involvement of vior induced by chronic stress in mice. NADPH oxidase
the EGF receptor in MAPK signaling activation by a 50 Hz mediates depressive behavior induced by chronic stress in
magnetic field in human neuroblastoma cells. Cell. Physiol. mice. J. Neurosci 32:9690–99.
Biochem. 52:893–907. doi:10.33594/000000062. Sheikh, A., A. Takatori, M. S. Hossain, M. K. Hasan, M. Tagawa,
Mattsson, M. O., and M. Simkó. 2014. Grouping of experi­ H. Nagase, and A. Nakagawara. 2016. Unfavorable neuroblas­
mental conditions as an approach to evaluate effects of toma prognostic factor NLRR2 inhibits cell differentiation by
extremely low-frequency magnetic fields on oxidative transcriptional induction through JNK pathway. Cancer Sci
response in vitro studies. Front. Public. Health 2:132. 107:1223–32. doi:10.1111/cas.13003.
doi:10.3389/fpubh.2014.00132. Song, K., S. H. Im, Y. J. Yoon, H. M. Kim, H. J. Lee, and
Merla, C., M. Liberti, C. Consales, A. Denzi, F. Apollonio, G. S. Park. 2018. A 60 Hz uniform electromagnetic field
C. Marino, and B. Benassi. 2019. Evidences of plasma promotes human cell proliferation by decreasing intracel­
membrane-mediated ROS generation upon ELF exposure lular reactive oxygen species levels. PLoS One 13.
in neuroblastoma cells supported by a computational multi­ doi:10.1371/journal.pone.0199753.
scale approach. Biochim. Biophys. Acta Biomembr. Sulpizio, M., S. Falone, F. Amicarelli, M. Marchisio, F. Di
1861:1446–57. doi:10.1016/j.bbamem.2019.06.005. Giuseppe, E. Eleuterio, C. Di Ilio, and S. Angelucci. 2011.
Nauseef, W. M. 2008. Biological roles for the NOX family Molecular basis underlying the biological effects elicited by
NADPH oxidases. J. Biol. Chem 283:16961–65. https:// extremely low-frequency magnetic field (ELF-MF) on neuro­
www.ncbi.nlm.nih.gov/pubmed/18420576. blastoma cells. J. Cell. Biochem. 112:3797–806. doi:10.1002/
Paletta-Silva, R., N. Rocco-Machado, and J. R. Meyer- jcb.23310.
Fernandes. 2013. NADPH oxidase biology and the regula­ Sumimoto, H. 2008. Structure, regulation and evolution of
tion of tyrosine kinase receptor signaling and cancer drug nox-family NADPH oxidases that produce reactive oxygen
cytotoxicity. Int. J. Mol. Sci. 14:3683–704. doi:10.3390/ species. Febs J 275:3249–77. doi:10.1111/j.1742-4658.2008.
ijms14023683. 06488.x.
Piras, S., A. L. Furfaro, R. Caggiano, L. Brondolo, S. Garibaldi, Sun, L., L. Chen, L. Bai, Y. Xia, X. Yang, W. Jiang, and W. Sun.
C. Ivaldo, U. M. Marinari, M. A. Pronzato, R. Faraonio, and 2018. Reactive oxygen species mediates 50-Hz magnetic
M. Nitti. 2018. MicroRNa-494 favors ho-1 expression in field-induced EGF receptor clustering via acid sphingomye­
neuroblastoma cells exposed to oxidative stress in a linase activation. Int. J. Radiat. Biol. 94:678–84. doi:10.1080/
Bach1-independent way. Front. Oncol. 8:199. doi:10.3389/ 09553002.2018.1466208.
fonc.2018.00199. Sun, W. J., H. Chiang, Y. T. Fu, Y. N. Yu, H. Y. Xie, and
Poniedziałek, B., P. Rzymski, J. Karczewski, F. Jaroszyk, and D. Q. Lu. 2001. Exposure to 50 Hz electromagnetic fields
K. Wiktorowicz. 2013. Reactive oxygen species (ROS) pro­ induces the phosphorylation and activity of stress-activated
duction in human peripheral blood neutrophils exposed protein kinase in cultured cells. Electro- and Magnetobiol
in vitro to static magnetic field. Electromagn. Biol. Med. 20:415–23. doi:10.1081/JBC-100108579.
32:560–68. doi:10.3109/15368378.2013.773910.
116 M. A. MARTÍNEZ ET AL.

Tormos, A. M., R. Taléns-Visconti, A. R. Nebreda, and Venkatachalam, K., S. Mummidi, D. M. Cortez, S. D. Prabhu,
J. Sastre. 2013. p38 MAPK: A dual role in hepatocyte pro­ A. J. Valente, and B. Chandrasekar. 2008. Resveratrol inhi­
liferation through reactive oxygen species. Free Radic. Res. bits high glucose-induced PI3K/Akt/ERK-dependent
47:905–16. doi:10.3109/10715762.2013.821200. interleukin-17 expression in primary mouse cardiac
Touyz, R. M., X. Chen, F. Tabet, G. Yao, G. He, M. T. Quinn, fibroblasts. Am. J. Physiol. Heart. Circ. Physiol. 294:
P. J. Pagano, and E. L. Schiffrin. 2002. Expression of H2078–2087. doi:10.1152/ajpheart.01363.2007.
a functionally active gp91phox-containing neutrophil-type Wang, H., and X. Zhang. 2017. Magnetic fields and reactive
NAD(P)H oxidase in smooth muscle cells from human oxygen species. Int. J. Mol. Sci. 18:2175. doi:10.3390/
resistance arteries: Regulation by angiotensin II. Circ. Res. ijms18102175.
90:1205–13. doi:10.1161/01.res.0000020404.01971.2f. Wang, Y., and M. F. Lou. 2009. The regulation of NADPH
Trillo, M. A., M. A. Martínez, M. A. Cid, J. Leal, and A. Úbeda. oxidase and its association with cell proliferation in human
2012. Influence of a 50 Hz magnetic field and of all-trans- lens epithelial cells. Invest. Ophthalmol. Vis. Sci.
retinol on the proliferation of human cancer cell lines. Int. 50:2291–300. doi:10.1167/iovs.08-2568.
J. Oncol. 40:1405–13. doi:10.3892/ijo.2012.1347. Wu, Q., W. Wu, B. Fu, L. Shi, X. Wang, and K. Kuca. 2019.
Tseng, H. Y., Z. M. Liu, and H. S. Huang. 2012. NADPH JNK signaling in cancer cell survival. Med. Res. Rev.
oxidase-produced superoxide mediates EGFR transactiva­ 39:2082–104. doi:10.1002/med.21574.
tion by c-Src in arsenic trioxide-stimulated human Yakymenko, I., O. Tsybulin, E. Sidorik, D. Henshel,
keratinocytes. Arch. Toxicol. 86:935–45. doi:10.33594/ O. Kyrylenko, and S. Kyrylenko. 2016. Oxidative mechan­
000000062. isms of biological activity of low-intensity radiofrequency
Turner, M. C., G. Benke, J. D. Bowman, J. Figuerola, radiation. Electromagn. Biol. Med. 35:186–202. doi:10.3109/
S. Fleming, M. Hours, L. Kincl, D. Krewski, D. McLean, 15368378.2015.1043557.
M.-E. Parent, et al. 2017. Interactions between occupational Yan, L., S. Liu, C. Wang, F. Wang, Y. Song, N. Yan, S. Xi, Z. Liu,
exposure to extremely low frequency magnetic fields and and G. Sun. 2013. JNK and NADPH oxidase involved in
chemicals for brain tumour risk in the INTEROCC study. fluoride-induced oxidative stress in BV-2 microglia cells.
Occup. Environ. Med. 74:802–09. doi:10.1136/oemed-2016- Mediators Inflamm 2013:895975. doi:10.1155/2013/895975.
104080.

You might also like