ArticlePDF Available

Abstract

Low frequency (LF) electromagnetic fields (EMFs) are abundantly present in modern society and in the last 20 years the interest about the possible effect of extremely low frequency (ELF) EMFs on human health has increased progressively. Epidemiological studies, designed to verify whether EMFs exposure may be a potential risk factor for health, have led to controversial results. The possible association between EMFs and an increased incidence of childhood leukemia, brain tumors or neurodegenerative diseases was not fully elucidated. On the other hand, EMFs are widely used, in neurology, psychiatry, rheumatology, orthopedics and dermatology, both in diagnosis and in therapy. In vitro studies may help to evaluate the mechanism by which LF-EMFs affect biological systems. As in vitro model of wound healing were used keratinocytes (HaCaT), neuroblastoma cell line (SH-SY5Y) as a model for analysis of differentiation, metabolism and functions related to neurodegenerative processes, monocytic cell line (THP-1) as a model for inflammation and cytokines production, while leukemic cell line (K562) was used as a model of hematopoietic differentiation. MCP-1, a chemokine that regulates the migration and infiltration of memory T cells, natural killer (NK), monocytes and epithelial cells, has been demonstrated to be induced and involved in various diseases. Since, varying the parameters of EMFs different effects may be observed, we have studied MCP-1 expression in HaCaT, SH-SY5Y, THP-1 and K562 exposed to a sinusoidal EMF at 50Hz frequency with a flux density of 1 mT (rms). Our preliminary results showed that EMF-exposure differently modifies the expression of MCP-1 in different cell types. Thus, the MCP-1 expression needs to be better determined, with additional studies, with different parameters and times of exposure to ELF-EMF.
ORIGINAL ARTICLE
Experimental model for ELF-EMF
exposure: Concern for human health
C. D’Angelo
a,*
, E. Costantini
a
, M.A. Kamal
b
, M. Reale
a
a
Dept. Experimental and Clinical Sciences, Immunodiagnostic and Molecular Pathology Section, University ‘‘G. d’Annunzio’’
Chieti-Pescara, Italy
b
King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
Received 2 June 2014; revised 16 July 2014; accepted 17 July 2014
Dedicated to the memory of our dear friend and colleague, Giovina Vianale.
KEYWORDS
ELF-EMF;
HaCaT;
K562;
MCP-1;
SH-SY5Y;
THP-1
Abstract Low frequency (LF) electromagnetic fields (EMFs) are abundantly present in modern
society and in the last 20 years the interest about the possible effect of extremely low frequency
(ELF) EMFs on human health has increased progressively. Epidemiological studies, designed to
verify whether EMF exposure may be a potential risk factor for health, have led to controversial
results. The possible association between EMFs and an increased incidence of childhood leukemia,
brain tumors or neurodegenerative diseases was not fully elucidated. On the other hand, EMFs are
widely used, in neurology, psychiatry, rheumatology, orthopedics and dermatology, both in diag-
nosis and in therapy.
In vitro studies may help to evaluate the mechanism by which LF-EMFs affect biological systems.
In vitro model of wound healing used keratinocytes (HaCaT), neuroblastoma cell line (SH-SY5Y)
as a model for analysis of differentiation, metabolism and functions related to neurodegenerative
processes, and monocytic cell line (THP-1) was used as a model for inflammation and cytokines
production, while leukemic cell line (K562) was used as a model for hematopoietic differentiation.
MCP-1, a chemokine that regulates the migration and infiltration of memory T cells, natural
killer (NK), monocytes and epithelial cells, has been demonstrated to be induced and involved in
various diseases.
Abbreviations: AD, Alzheimer’s disease; ELF, extremely low fre-
quency; EMFs, electromagnetic fields; HD, Huntington disease; LF,
low frequency; MCP-1, monocyte chemoattractant protein-1; PMA,
phorbol-12-myristate-13-acetate; PEMF, pulsed EMF
*Corresponding author.
E-mail address: chiara_dangelo@hotmail.it (C. D’Angelo).
Peer review under responsibility of King Saud University.
Production and hosting by Elsevier
Saudi Journal of Biological Sciences (2014) xxx, xxxxxx
King Saud University
Saudi Journal of Biological Sciences
www.ksu.edu.sa
www.sciencedirect.com
http://dx.doi.org/10.1016/j.sjbs.2014.07.006
1319-562X ª2014 Production and hosting by Elsevier B.V. on behalf of King Saud University.
Please cite this article in press as: D’Angelo, C. et al., Experimental model for ELF-EMF exposure: Concern for human health. Saudi Journal of Biological Sciences
(2014), http://dx.doi.org/10.1016/j.sjbs.2014.07.006
Since, varying the parameters of EMFs different effects may be observed, we have studied MCP-1
expression in HaCaT, SH-SY5Y, THP-1 and K562 exposed to a sinusoidal EMF at 50 Hz fre-
quency with a flux density of 1 mT (rms).
Our preliminary results showed that EMF-exposure differently modifies the expression of MCP-1
in different cell types. Thus, the MCP-1 expression needs to be better determined, with additional
studies, with different parameters and times of exposure to ELF-EMF.
ª2014 Production and hosting by Elsevier B.V. on behalf of King Saud University.
1. Introduction
Exposure to electromagnetic fields (EMF) is a phenomenon
that has always existed, nevertheless, during the 20th century,
this is steadily increasing due to environmental exposure to
man-made electromagnetic fields. Growing electricity demand,
ever-advancing technologies and changes in social behaviors
have created more and more artificial sources. Thus, both at
home and at work everyone are exposed to a complex mix of
weak electric and magnetic fields, arising from the generation
and transmission of electricity by domestic appliances and
industrial equipment, by telecommunications and
broadcasting.
Generally the extremely low frequency (ELF) region of the
electromagnetic spectrum is defined by frequencies from 3 to
3000 Hz (Poole and Ozonoff, 1996). These fields are produced
by electrical devices, high tension electrical distribution net-
works, from residential and occupational sources and by
power lines. 60 Hz (in the USA) and 50 Hz sine wave signals
resemble the household alternating current electrical power
supply in Europe and a large part of the world. Low-frequency
electric fields influence all systems characterized by charged
particles as the human body. In fact tiny electrical currents
exist in the human body due to the chemical reactions that
occur as part of the normal bodily functions, even in the
absence of external electric fields. For example, nerves transmit
signals through electrical impulses. Most biochemical reac-
tions, from digestion to brain activities, are complying with
the rearrangement of charged particles.
For several years, it has been considered that both residen-
tial and occupational exposures to ELF magnetic fields (MF)
could be a possible carcinogen, based on several epidemiolog-
ical studies reporting childhood leukemia and brain tumors in
adult and leukemia following chronic exposure to MF (IARC
2002). Epidemiologic studies on EMF effect, reported evidence
of association among childhood leukemia and postnatal expo-
sures above 0.4lT. Previous studies concluded that residential
exposures to EMFs carry an increased risk of leukemia,
although other studies showed that there is no significant risk
(Leitgeb, 2011). In contrast with earlier studies (Wertheimer
and Leeper, 1979; Savitz et al., 1988; London et al., 1991),
but in accord with others (Jirik et al., 2012; Auvinen et al.,
2000) that have shown no significant increase in risk of the
Acute Lymphoblastic Leukemia (ALL) for children exposed
to residential levels of magnetic fields, Linet et al. show a lack
of association between electromagnetic field exposure and
ALL (Linet et al., 1997.
Harmful effect of EMF exposure on living tissue depends
primarily on the frequency (wavelength) and density of the
field and on the exposure time. Further important risk factors
are the functional state and the sensibility of the exposed
organism. The vascularization of the irradiated parts and the
distance from the radiation source must be considered, too.
EMFs of the magnitude to which we are now regularly
exposed, have been implicated as a contributory factor to the
childhood cancer incidence, particularly leukemia and brain
cancer.
There are numerous publications describing various in vitro
effects of EMF exposure, although the significance of these
observations for clinical interpretation is unsubstantiated. A
fundamental interaction mechanism between weak ELF mag-
netic fields and cells is also lacking, although several candidate
mechanisms have been proposed. Numerous hypotheses have
been suggested (IARC 2002; Davanipour et al., 2007; Draper
et al., 2005; Gottwald et al., 2007), although none is convincingly
supported by experimental data. A large number of cellular
components, systems and processes such as proliferation, (Tsai
et al., 2007) morphology, (Noriega-Luna et al., 2011) apoptosis,
(Grassi et al., 2004) gene expression (Mayer-Wagner et al., 2011)
and differentiation (Piacentini et al., 2008), can conceivably be
affected by EMF exposure (Simko
`and Mattsson, 2004).
Although the role of increased intracellular Ca
2+
was already
well documented more than 20 years ago (Walleczek, 1992),
recent studies have confirmed the role of increased intracellular
Ca
2+
following EMF exposure. Recently, it was suggested that a
possible early biological response to EMF exposure, is the for-
mation and prolonged survival of reactive oxygen species and
other free radicals (Mannerling et al., 2010).
Different types of magnetic and electromagnetic fields are
now used effectively in medicine (Markov 2007), such as in
diagnostic (e.g. magnetic resonance imaging-MRI, scanner
and microwave imaging) or therapy (Consales et al., 2012).
Electromagnetic therapy carries the promise to be used in dif-
ferent diseases, in fact magnetotherapy provides an easy and
non invasive method to treat the site of injury (Markov
2007). Pulsed electromagnetic fields in low frequency and
intensity range (Gauss or micro-Tesla) increase oxygenation
to the blood, improve circulation and cell metabolism, improve
function, pain and fatigue from fibromyalgia (Sutbeyaz et al.,
2009), help patients with treatment-resistant depression
(Martiny et al., 2010), and may reduce symptoms from multi-
ple sclerosis (Lappin et al., 2003). EMFs have been commonly
used for the treatment of some pathological conditions to stim-
ulate tissue regeneration and repair (Bertolino et al., 2006).
Application in the area of orthopedics for the treatment of
non-union fractures and failed fusions, takes advantage of
the evidence that pulsed EMF (PEMF) accelerates the re-
establishment of normal potentials in damaged cells (Fiorani
et al., 1997), promotes the proliferation and differentiation
of osteoblasts (Wei et al., 2008) and improves the osteogenic
2 C. D’Angelo et al.
Please cite this article in press as: D’Angelo, C. et al., Experimental model for ELF-EMF exposure: Concern for human health. Saudi Journal of Biological Sciences
(2014), http://dx.doi.org/10.1016/j.sjbs.2014.07.006
phase of the healing process (Cane
`et al., 1993). Long-lasting
relief of pelvic pain of gynecological origin has been obtained
consistently by short exposures of affected areas with the
application of a magnetic induction device, producing short,
sharp, magnetic-field pulses of minimal amplitude (Jorgensen
et al., 1994). EMFs improve cell survival and reduce ischemic
damage (Grant et al., 1994).
2. EMFs and skin injury
Being the skin the largest organ that covers the surface of our
body, it is frequently subject to the action of non ionizing MF.
Keratinocytes, those are able to release immunomodulators
and to play a key role in immune system function may be used
as an in vitro model to evaluate the biological effects of non
ionizing electromagnetic field on the skin.
Wound healing is a highly coordinated and complex pro-
cess involving the proliferation and migration of various cell
types (epidermal, dermal as well as inflammatory cell), chemi-
cal mediators and the surrounding extracellular matrix, result-
ing in a tightly orchestrated re-establishment of tissue integrity
by specific cytokines. Wounds can be categorized as acute or
chronic according to their healing time-frame. Acute wounds
repair themselves and heal normally following the correct
pathway. The chronic condition derives from non-healing
wounds in a timely and orderly manner, that determines ulcers
(Lazarus et al., 1994). Ischemia, diabetes mellitus, venous sta-
sis and pressure can be at the root of the majority of non-heal-
ing wounds that are prone to complications including
functional limitations, infections and malignant transforma-
tion (Eltorai et al., 2002; Chraibi et al., 2004).
Although there are many experimental and clinical evidences
supporting the use of magnetic fields to help bone healing, its
application for soft tissue healing, including skin and tendons
is still ambiguous. Several authors, however, showed the ability
of PEMFs in reducing the wound healing durations (Cheing
et al., 2014; Athanasiou et al., 2007; Strauch et al., 2007) and
improving tensile strength of scars (Goudarzi et al., 2010).
Roland et al. used pulsed magnetic energy to stimulate neovas-
cularization in a rat model (Roland et al., 2000). Weber et al.
showed that rat groin composite flap survival increases when
supported by an arterial loop, thus confirming that PEMFs pro-
mote neovascularization (Weber et al., 2004). The most rapid
wound healing exposed to EMF may be dependent on the
anti-inflammatory effects caused by the change in the coagula-
tion system, in the improvement of microcirculation and in
immunological reactiveness (Matic et al., 2009). Conversely,
Milgram found that PEMF did not produce any beneficial
effects on wound healing. So the effects of PEMF on wound clo-
sure varied among the studies, possibly due to different treat-
ment protocols that were applied (Milgram et al., 2004).
Callaghan et al. (2008) confirmed the results of Tepper et al.
(2004), that demonstrated the increase in proliferation and tubu-
lization of endothelial cell cultures and the increase in the expres-
sion of fibroblast growth factor 2 (FGF-2), a potent stimulator
of angiogenesis, after exposure to electromagnetic field.
Vianale et al. showed that ELF-EMF modulates produc-
tion of RANTES, MCP-1, MIP-1aand IL-8, and keratinocyte
growth through the inhibition of the NF-kB signaling pathway
and they hypothesized that ELF-EMF may inhibit inflamma-
tory processes (Vianale et al., 2008).
Recently, several reports have supported the anti-inflamma-
tory effects of EMFs on tissue repair. Pesce et al. reviewed the
effect of EMFs on cytokines that drive the transition from a
chronic pro-inflammatory to an anti-inflammatory state of the
healing process (Pesce et al., 2013). Patruno’s results showed
the ability of ELF-EMF to induce keratinocyte proliferation
and to up modulate NOS activities and to down-regulate
COX-2 expression and PGE-2 production, involved in the mod-
ulation of inflammatory reaction (Patruno et al., 2010). In vitro
study of Huo et al. showed that the noninvasive EMFs have a
strong effect on normal human keratinocytes and fibroblast
migration while only weakly promote keratinocyte proliferation
(Huo et al., 2009). The observations of Manni et al. confirm the
hypothesis that ELF-EMF (50 Hz) may modify cell membrane
morphology and interfere with initiation of the signal cascade
pathway and cellular adhesion (Manni et al., 2002). ELF-
EMF application modifies the biochemical properties of human
keratinocytes (HaCaT) associated with different actin distribu-
tions as demonstrated by Lisi et al. (2006).
3. EMFs and neurodegenerative diseases
The term neurodegeneration indicates the progressive loss of
neuronal function and structure until the neuron death. Many
neurodegenerative diseases such as Parkinson disease (PD),
Alzheimer’s disease (AD), Huntington disease (HD), and
Amyotrophic Lateral Sclerosis (ALS) result from neurodegen-
erative processes and many of these are classified as patholo-
gies due to the aggregation of misfolded proteins. PD is a
disorder of the central nervous system resulting from the death
of dopaminergic cells in the substantia nigra. The basis of this
mechanism may consist of an abnormal accumulation of the
protein alpha-synuclein that forms insoluble fibrils, in the
damaged cells. The beta-amyloid peptide (Ab)is a small pep-
tide that comes from the cleavage of a larger transmembrane
protein called amyloid precursor protein (APP). Abis the
major component of plaques in the cerebral cortex of AD
and is critical to neuron growth, survival and is involved in
the loss of synapses and the neuron death, as well as hyper-
phosphorylated tau protein, the main component of neurofi-
brillary tangles in AD brain. Sobel and Davanipour
hypothesized the ability of the ELF-EMFs to increase the
intracellular calcium concentration levels that are positively
correlated with the cleavage of the APP to give the soluble
Ab(Sobel and Davanipour, 1996). Several studies seem to sug-
gest a potential association between occupational exposure to
ELF-EMFs (typical of electric power installers and repairers,
power plant operators, electricians, telephone line technicians,
welders, carpenters, and machinists) and AD onset (Garcı
`a
et al., 2008; Ro
¨o
¨sli, 2008), although their biological nexuses
remain unknown.
HD is a progressive neurodegenerative disorder whose
underlying genetic defect lies in expanded trinucleotide
(CAG)n of the Huntington ubiquitous protein. In HD, an
autosomal dominant disease, the mutated gene leads neuronal
dysfunction and degeneration, even though the mechanisms by
which it acts are not fully understood. The potential correla-
tion between EMF exposure and HD pathogenesis is not sus-
tained by epidemiological evidence, while there is evidence that
the improvement in behavior and the neuroprotective effect of
ELF-EMF exposure may be due to enhanced neurotrophic
Experimental model for ELF-EMF exposure 3
Please cite this article in press as: D’Angelo, C. et al., Experimental model for ELF-EMF exposure: Concern for human health. Saudi Journal of Biological Sciences
(2014), http://dx.doi.org/10.1016/j.sjbs.2014.07.006
factor levels, and reduced both oxidative damage. The Amyo-
trophic Lateral Sclerosis is a fatal neurodegenerative disorder
characterized by progressive degeneration of motor neurons
in the spinal cord, motor cortex, and brainstem. Approxi-
mately 20% of patients were found to show mutation of the
gene encoding the antioxidant Cu
2+
/Zn
2+
SOD (SOD1)
(Julien and Kriz 2006), confirming the central role of oxidative
stress in neurodegenerative diseases (Chang et al., 2008). On
the basis of epidemiologic findings, evidence shows an associ-
ation between amyotrophic lateral sclerosis and occupational
EMF exposure although there is confounding (Davanipour
et al., 1997; Savitz et al., 1998). The investigations of EMF
effects on neurodegenerative diseases are now very interesting,
although not well developed, in fact the experimental findings
supporting this link are still controversial due to the field fre-
quency applied and the disease investigated (Consales et al.,
2012). Crasson et al. indicated that 50 Hz EMF may have
slight influence on event-related potential and reaction time
under specific circumstances of sustained attention in healthy
male volunteers. (Crasson et al., 1999). Trimmel’s study shows
a reduction of cognitive performances in attention, perception
and memory performances by a 50 Hz EMF of 1 mT (Trimmel
and Schweiger, 1998).
Sulpizio et al. have demonstrated that ELF-MF exposure
triggers significant changes in the protein global profile of
SH-SY5Y cell line, experimental model for neurodegenerative
disorders. In particular, the expression levels of common pro-
tein spots involved in cellular defense mechanisms, organiza-
tion, and biogenesis increased as a consequence of ELF-
EMF treatment. In ELF-EMF treated samples was observed
the over-expression of proteins related to a high malignant
potential, drug resistance, cytoskeleton re-arrangement, and
enhanced defense against oxidative stress, in association with
higher proliferative activity (Sulpizio et al., 2011; Xie et al.,
2010). In vivo study showed that exposure to environmental
ELF-EMF did not change the expression of a3, a5 and a7 nic-
otinic cholinergic receptors impaired in AD (Antonini et al.,
2006). Falone et al. have demonstrated that a 50 Hz magnetic
field induced a significant enhancement of the antioxidant
defenses together with a major shift of redox homeostasis
and they previously established that ELF-MF exposure
improves cellular viability and induces significant adaptations
in the redox-related biochemical machinery of the human neu-
roderived SH-SY5Y cell line (Falone et al., 2007).
4. EMFs and immune cells
Cells of the immune system regulate health on a systemic level,
thus are plausible study targets. .In response to a pathogen chal-
lenge they must respond in a very sensitive, swift and effective
way. Immune cells produce cytokines, important signaling mol-
ecules, which are key regulators of cell activation and inhibition.
Monocytes and macrophages have an important function
as the first line of defense against pathogens and can act as
antigen-presenting cells to trigger a specific response from lym-
phocytes, and are capable of producing several cytokines
including interleukin-1 beta (IL-1b), tumor necrosis factor-
alpha (TNF-a), and interleukin-10 (IL-10). Their recruitment
to inflammatory sites and neoplastic tissues and their activa-
tion induce a wide range of intracellular signaling pathways
and are crucial to the success of an immune reaction. Cytokine
production and secretion patterns are modified upon differen-
tiation of monocytes into macrophages (Bouwens et al., 2012).
Several papers have demonstrated that the in vitro exposure of
immune cells to nonthermal ELF-EMF can elicit molecular
and cellular changes that might be relevant to the activity of
the immune system in vivo. Years ago, it was demonstrated
that only mutagen-activated lymphocytes are responsive to
EMF exposure, in fact EMF do not interfere with activation
and committeemen of cells (Cadossi et al., 1992). Nindl et al.
have showed that 60 Hz sinusoidal EMFs induce an increase
in anti-CD3 binding to T cell receptors (TcRs) of Jurkat cells,
a T lymphocyte cell line, and that can regulate lymphocyte
proliferation in vitro and in vivo (Nindl et al., 2000). Reale
et al. have demonstrated that upon ELF-EMF activation,
monocytes/macrophages increase the production of chemo-
kines, peroxidases, cytolytic proteases, and nitric oxide (NO)
enhancing their microbicidal/tumoricidal capacity (Reale
et al., 2006). Akan et al. have demonstrated that field applica-
tion increased NO, cGMP, and HSP levels, and caused a slight
decrease in apoptosis (Akan et al., 2010; Frahm et al., 2010).
Many in vitro and in vivo studies have evaluated the expression
of free radicals in human monocytes and mouse macrophages
after exposure to 50 Hz, 1 mT ELF-EMF (Simko
`et al., 2001;
Lupke et al., 2004; Rollwitz et al., 2004). NO, a free radical, is
an important intracellular and intercellular signaling molecule,
and an important host defense effector for the phagocytic cells
of the immune system (Fo
¨rstermann and Kleinert, 1995).
Several studies have been conducted to evaluate the effect of
ELF-EMF exposure on cytokine profiles, consistent and inde-
pendently replicated laboratory evidence to support modula-
tion of cytokines expression and production has not been
obtained (Ikeda et al., 2003; Luceri et al., 2005; Miller et al.,
1999; Natarajan et al., 2006; Reale et al., 2006; Lupke et al.,
2006; Murabayashi et al., 2004). Ays
ße et al. demonstrated that
in vitro effect of ELF-EMF on the differentiation of K562 cells
is time dependent. In fact single exposure to ELF-EMF resulted
in a decrease in differentiation; ELF-EMF applied everyday for
1 h caused an increase in differentiation. These results imply
that the time-course of application is an important parameter
determining the physiological response of cells to ELF-EMF
(Ays
ße et al., 2010) and other authors have supported the
hypothesis that the effect of ELF-EMF on biological
systems depends on the conditions of the cell (Garip and
Akan, 2010).
Numerous studies are still underway to try to understand
the mechanism behind these alterations investigated as the
gaming is very complex. Conflicting conclusion were showed
in many EMF in vivo studies, due to small numbers of subject,
distance from EMF source, time exposure or concomitant
environmental risks. The in vivo study of Boscolo did find dif-
ferences in cytokine levels in serum of subject exposed to ELF-
EMF (Boscolo et al., 2001). In a set of experiments evaluating
time courses for immediate early genes, stress response, cell
proliferation and apoptotic genes, Kirschenlohr et al. showed
no consistent response profiles after repeated ELF-EMF expo-
sures (Kirschenlohr et al. 2012).
5. Cell line in vitro models
Cell lines have some advantages over human primary cells
such as (a) homogeneous genetic background that minimizes
4 C. D’Angelo et al.
Please cite this article in press as: D’Angelo, C. et al., Experimental model for ELF-EMF exposure: Concern for human health. Saudi Journal of Biological Sciences
(2014), http://dx.doi.org/10.1016/j.sjbs.2014.07.006
the degree of variability in the cell phenotype, a trait particu-
larly important when studying the biological function with
high variability; (b) ability to be stored indefinitely in liquid
nitrogen to guarantee sufficient cells for DNA, RNA and pro-
tein; (c) reduced variability compared to primary cells; and (d)
reproducibility of the results obtained.
SH-SY5Y cells were derived from immature neoplastic neu-
ral crest cells that exhibit properties of stem cells. The SH-
SY5Y cell line is a thrice-cloned subline of SK-N-SH cells that
were originally established from a bone marrow biopsy of a
neuroblastoma patient and were widely used as model of neu-
rons since the early 1980’s (Biedler et al., 1973). These cells
possess the capability of proliferating in culture for long peri-
ods without contamination, a prerequisite for the development
of an in vitro cell model, posses many biochemical and func-
tional properties of neurons, exhibits neuronal marker enzyme
activity, express neurofilament proteins and also express opi-
oid, muscarinic, and nerve growth factor receptors
(Ciccarone et al., 1989). Consequently, the SH-SY5Y cell line
has been widely used in experimental neurological studies,
including analysis of processes related to neurodegeneration,
neuroprotection and neurotoxicity. The processes of keratino-
cyte proliferation and differentiation represent the central and
final event in tissue regeneration leading to the formation of a
massive bulk of cells, necessary to cover the wounded area. It
is widely accepted that in vitro keratinocyte model systems,
such as HaCaT cell line, at low and high density can be com-
pared with early and late phases of the re-epithelialization pro-
cess. HaCaT cells are in vitro spontaneously transformed
keratinocytes from histologically normal skin. Thus keratino-
cytes are the most likely cells to be impacted by electromag-
netic radiation.
THP-1 is single, round suspension cells that after exposure
to phorbol-12-myristate-13-acetate (PMA) or 1a,25-
dihydroxyvitamin D3 (1a,25(OH)2D3) may start to adhere
to culture plates accompanied by phenotype change into a
macrophage. Based on phenotypic and functional features
with human microglial cells, human monocyte-derived macro-
phages were called brain macrophages (Ulvestad et al., 1994).
THP-1 cells, due to their functional and morphological simi-
larities, have been widely used as a model of human mono-
cytes/macrophages or microglia (Tsuchiya et al., 1982;
Tsuchiya et al., 1980; McDonald et al., 1998) or as a valid
model to mimic proliferation, adhesion and migration of
monocytes and macrophages in the vasculature.
The human K562 cell line has been isolated and character-
ized by Lozzio (Lozzio and Lozzio, 1975) from a patient with
chronic myelogenous leukemia (CML) in blast crisis. K562
has been used as a model of common progenitor of erythro-
blasts and megakaryocytes and can be differentiated into ery-
throid and megakaryocytic lineages thus has been used
extensively as a model for the study of leukemia differentiation,
molecular mechanism(s) regulating the expression of genes
(Iyamu et al., 2000), as well as to determine the therapeutic
potential of new differentiation-inducing compounds (Bianchi
et al., 2001).
6. Effects of ELF-EMF exposure on MCP-1
Chemokines are low molecular weight chemotactic cytokines
that have been shown to play a relevant role in inflammatory
events, such as transendothelial migration and accumulation
of leucocytes at the site of damage. In addition, they modulate
a number of biological responses, including enzyme secretion,
cellular adhesion, cytotoxicity and T-cell activation and tissue
regeneration (Vianale et al. 2008).
The monocyte chemoattractant protein-1 (MCP-1/CCL2)
is a member of the C–C chemokine family and is a potent che-
motactic factor for monocytes. Located on chromosome 17
(chr.17, q11.2), human MCP-1 is composed of 76 amino acids
and is 13 kDa in size (Van Coillie et al., 1999). A variety of cell
types including endothelial, fibroblasts, epithelial, smooth
muscle, mesangial, astrocytic, monocytic, and microglial cells
(Cushing et al., 1990; Standiford et al., 1991; Brown et al.,
1992; Barna et al., 1994), are able to produce MCP-1, either
constitutively or after induction by oxidative stress, cytokines,
or growth factors. Rolling of monocytes on endothelial cells is
dependent on the binding of E-selectin and sialyl Lewis X, and
adhesion to the endothelium is dependent on the interaction of
integrin on monocytes and adhesion molecules on the endothe-
lial cells. Although leukocytes have been considered the main
targets for chemokines, recent evidence indicates that the
actions of these proteins are not restricted to these cell types.
The main function of MCP-1 consists of the establishment of
chemotaxis driving the recruitment of cells at sites of inflam-
mation, by integrin activation. Specifically MCP-1 attracts
monocytes, natural killer cell and memory T cells, and influ-
ences expression of cytokines related to T helper responses.
Its expression occurs in a variety of diseases characterized by
mononuclear cell infiltration, and there is substantial biologi-
cal and genetic evidence suggesting that it may contribute to
the inflammatory component of diseases such as atherosclero-
sis, multiple sclerosis, Alzheimer’s disease, or rheumatoid
arthritis. In the central nervous system (CNS), MCP-1 is
involved in the recruitment of the main resident immune cell
types of the brain (astrocytes and microglia) and of infiltrating
monocytes from the systemic bloodstream. There is strong evi-
dence that MCP-1 plays a major role in myocarditis, ischemia/
reperfusion injury in the heart, in transplant rejection, and in
cardiac repair. After 24 h of chronic exposure to 50 Hz,
1 mT EMF, MCP-1 levels were reduced significantly in
PHA-stimulated cells, while in non-stimulated cells no signifi-
cant differences in MCP-1 levels were observed. The authors
speculate the anti-inflammatory potency of electromagnetic
fields and suggest that the inhibitory effect on MCP-1 release,
evaluated by the ELISA assay, could be one of the mechanisms
by which ELF-EMF is therapeutic in inflammatory diseases
(Di Luzio et al., 2001).
Previous studies have suggested that magnetic field is
involved in NO production. Thus, Reale et al. exposed LPS-
stimulated peripheral blood adherent mononuclear cells to
50 Hz EMF. Results of RT-PCR showed that, while both
mRNA and protein levels of MCP-1 were up-regulated, iNOS
was down-regulated. The increase in MCP-1 is related to NF-
kB protein expression and in agreement with previous results
showing that the inhibition of nitric oxide production in endo-
thelial cells increased the expression of MCP-1. The changes in
MCP-1 and iNOS expression, evaluated through RT-PCR,
after ELF-EMF are very interesting for their roles in the devel-
opment of inflammatory responses. The authors suggest a non
pharmacological role of EMF in maintaining the balance
between MCP-1 and NO in inflammatory reaction (Reale
et al., 2006).
Experimental model for ELF-EMF exposure 5
Please cite this article in press as: D’Angelo, C. et al., Experimental model for ELF-EMF exposure: Concern for human health. Saudi Journal of Biological Sciences
(2014), http://dx.doi.org/10.1016/j.sjbs.2014.07.006
Since the EMF effect is cell type-dependent and MCP-1 is
produced which acts on different cell types, and very little is
known about the influence of ELF-EMF on MCP-1 expression
in different cell types, we studied the effect of ELF-EMF on
MCP-1 expression and production in HaCaT, SH-5YSY,
THP-1 and K562 cells.
In our ELF-EMF the flux density of 1 mT (rms) was pro-
duced by an electromagnetic generator (Agilent Technologies,
Santa Clara, CA, mod. 33220A) with stability higher than 1%
both in frequency and in amplitude. The generator was con-
nected to a power amplifier (Nad Electronics Ltd, London,
U.K., mod. 216). An oscilloscope (ISO-TECH mod. ISR658,
Vicenza, Italy) was dedicated to the monitoring of output sig-
nals from the Gaussmeter (MG-3D, Walker Scientific Inc.,
Worcester, MA). A current flux passed through a 160 turns
solenoid (22 cm length, 6 cm radius, copper wire diameter of
1.25 ·10
5
cm) generating a horizontal magnetic field. The
achieved MF intensity (1 mT/rms) was measured continuously
during exposure using a Hall-effect probe connected to the
Gaussmeter. The solenoid was then placed inside the incuba-
tor. The environmental magnetic noise inside the incubator
was related to the geomagnetic field (40 mT), and to the
50 Hz disturbance associated with the working incubator
[7 mT (rms)]. The built-in digital thermometer of the incuba-
tor monitored the internal temperature, which resulted con-
stant at 37 ± 0.38 C. In addition, another digital
thermometer (HD 2107.2; Delta OHM, Padova, Italy) was
placed inside the solenoid and near the cell cultures to record
local temperature variations. No significant temperature
change related to applied ELF fields was observed
(DT0.18C). However, no thermal effect on cells can be hypoth-
esized for temperatures around 37.8 C, because EMF interac-
tions with biological molecules are known to be non thermal in
nature. Low-level Joule heating was dissipated inside the incu-
bator by a fan system. In all experiments cells were placed in
the central part of the solenoid, which presented the highest
degree of the field homogeneity (98%).
All experiments are performed at the same conditions of
EMF intensity, frequency, chronical exposure, and tempera-
ture. In Table 1, we report the effects of the ELF-EMF expo-
sure on MCP-1 expression in different cell lines.
In HaCaT cells, using RT-PCR we have evidenced a
decrease in MCP-1 expression from 4 to 72 h in EMF-exposed
cells with respect to non-exposed cells. This decrease was con-
firmed by additional Real Time PCR (basal exposed
0.9 ± 0.02 vs. basal non-exposed 1.6 ± 0.05). Also the ELISA
immunoassay, performed to evaluate the release of MCP-1,
confirmed the expression results. Since it is well accepted that
an excessive or prolonged inflammatory response may interfere
with wound healing and cause reduction of the inflammatory
chemokines by ELF-EMF exposure, represents an interesting
and new therapeutic approach in delayed healing.
In SH-SY5Y cell cultures exposed to ELF-EMF, genes
involved in the stress response, cell growth and differentiation
or protein metabolism have been reported to be generally
down-regulated. Genes involved in Ca
2+
metabolism, the
PI3-kinase pathway are up-regulated. Likewise, key mediators
of the inflammatory response appear susceptible to swift mod-
ulation, in SH-SY5Y.
MCP-1 is involved in the neuroinflammatory processes
associated with diseases characterized by neuronal degenera-
tion. To characterize the impact of ELF-EMF on early ongo-
ing cellular processes, MCP-1 gene expression in SH-SY5Y,
was evaluated in the presence and absence of ELF-EMF expo-
sure by RT-PCR. After 24 h of ELF-EMF exposure MCP-1
expression was not significantly affected. Albeit our results
on MCP-1 expression, despite differences in experimental con-
ditions, are in line with several other ELF-EMF exposure
results, while they are not in accord with a study reporting that
ELF-EMF promotes cellular neurodifferentiation, as exempli-
fied by neurite extension and number (Falone et al., 2007). In
conclusion, our results showed that ELF-EMF exposure is well
tolerated and has no relevant impact on MCP-1 gene
expression.
The role of MCP-1 in human disease has been demon-
strated by immunohistochemical studies in fact the adhesion
of cells to the endothelium was induced by expression of adhe-
sion molecules and chemotactic proteins, such as MCP-1. We
have analyzed the effects of EMF on the expression of MCP-1
also in THP-1 cells. Since in THP-1 exposed to ELF-EMF no
increase in basal levels of MCP-1 was observed, cells were trea-
ted or not with LPS and exposed to 50 Hz, 1 mT EMF for
24hr. Our data indicate that the presence of 10 lg/ml of LPS
leads to an increase in expression of MCP-1 in both THP-1
cells non exposed or exposed to EMF. Thus, we hypothesized
that MCP-1 mediated THP-1 migration is not affected by
EMF exposure, and consequently the exposure to the fields
is not a risk factor in diseases in which microglial migration
plays a crucial role, such as atherosclerosis, multiple sclerosis
and other neuroinflammatory diseases.
Although it is known that intracellular redox status modu-
lates MCP-1 expression and that ELF-EMF exposure can act
on redox state of K562 cells. No studies have evaluated the
influence of EMF exposure on MCP-1 expression in K562 cell
line. In K562 exposed to the ELF-EMF spontaneous expres-
sion of MCP-1, detected by RT-PCR, was not modulated in
comparison to cells not exposed. PMA induces monocytic or
megakaryocytic differentiation of K562 cells through the acti-
vation of MAP kinases. When PMA-stimulated cells were
exposed to the field, we noticed a slight increase in the expres-
sion of the chemokines and particularly the increase in MCP-1.
Next step of this study will be to evaluate if ELF-EMF expo-
sure is able to modulate the activation of MAP kinases in com-
parison with PMA.
Table 1 Effects of the ELF-EMF exposure on MCP-1 in
different cell types.
Cell lines Effect on MCP-1 expression
HaCaT
Basal Decreased
SH-SY5Y
Basal Decreased
THP-1
Basal No effect
LPS-stimulated No effect
K562
Basal No effect
PMA-stimulated Increased
6 C. D’Angelo et al.
Please cite this article in press as: D’Angelo, C. et al., Experimental model for ELF-EMF exposure: Concern for human health. Saudi Journal of Biological Sciences
(2014), http://dx.doi.org/10.1016/j.sjbs.2014.07.006
7. Conclusions
The results of in vivo and in vitro studies suggest that EMF
may modulate the expression of some inflammatory molecules.
The understanding of the influences of EMF on transcriptional
events will lead to a better understanding of their mechanisms
and to therapeutic interventions for diseases in which these
inflammatory molecules play a key role. In spite of the fact
that the mechanisms of action of EMF are still under investi-
gation, some authors have supposed that exposure to ELF-
EMF affects cell function through mechanical action on both
intracellular and membrane proteins, which includes ion chan-
nels, membrane receptors and enzymes. All studies agree that
the effect of the sinusoidal ELF-EMF varies in relation to cell
type and other parameters, such as frequency, flux density and
time exposure.
Our data confirm the cell-type dependent effects; in fact we
observed increase, decrease or no effect on the MCP-1 expres-
sion in different cell lines grown under the same conditions
(sinusoidal 50 Hz, 1 mT, 37 C, 5% CO
2
).
In order to assess if ELF-EMFs, associated with both indus-
trial and domestic use, may play a role as adjuvant or causative
factor in disease development or may play a role as therapeutic
and diagnostic tool, further studies to evaluate a more complete
list of genes that may be up- or down-regulated by ELF-EMF
exposure must be preformed. New studies designed to evaluate
the actions of ELF-EMF under multiple conditions, including
chronic or sporadic exposure, in combination with common
stressors pertinent to real life, appear warranted and may aid
our understanding of their true biological impact.
References
Akan, Z., Aksu, B., Tulunay, A., Bilsel, S., Inhan-Garip, A., 2010.
Extremely low-frequency electromagnetic fields affect the immune
response of monocyte-derived macrophages to pathogens. Bioelec-
tromagnetics 31 (8), 603–612.
Antonini, R.A., Benfante, R., Gotti, C., Moretti, M., Kuster, N.,
Schuderer, J., Clementi, F., Fornasari, D., 2006. Extremely low-
frequency electromagnetic field (ELF-EMF) does not affect the
expression of alpha3, alpha5 and alpha7 nicotinic receptor subunit
genes in SH-SY5Y neuroblastoma cell line. Toxicol. Lett. 164 (3),
268–277.
Athanasiou, A., Karkambounas, S., Batistatou, A., Lykoudis, E.,
Katsaraki, A., Kartsiouni, T., Papalois, A., Evangelou, A., 2007.
The effect of pulsed electromagnetic fields on secondary skin
wound healing: an experimental study. Bioelectromagnetics 28,
362–368.
Auvinen, A., Linet, M.S., Hatch, E.E., Kleinerman, R.A., Robison,
L.L., Kaune, W.T., Misakian, M., Niwa, S., Wacholder, S.,
Tarone, R.E., 2000. Extremely low-frequency magnetic fields and
childhood acute lymphoblastic leukemia: an exploratory analysis of
alternative exposure metrics. Am. J. Epidemiol. 152, 20–31.
Aysße, I.G., Zafer, A., Sule, O., Isßil, I.T., Kalkan, T., 2010. Differen-
tiation of K562 cells under ELF-EMF applied at different time
courses. Electromagn. Biol. Med. 29 (3), 122–130.
Barna, B.P., Pettay, J., Barnett, G.H., Zhou, P., Iwasaki, K., Estes,
M.L., 1994. Regulation of monocyte chemoattractant protein-1
expression in adult human non-neoplastic astrocytes is sensitive to
tumor necrosis factor (TNF) or antibody to the 55-kDa TNF
receptor. J. Neuroimmunol. 50, 101–107.
Bertolino, G., de Freitas Braga, A., de Oliveira Lima do Couto, K., de
Brito Junior, L.C., de Araujo, J.E., 2006. Macroscopic and
histological effects of magnetic field exposition in the process of
tissue reparation in Wistar rats. Arch. Dermatol. Res. 298 (3), 121–
126.
Bianchi, N., Chiarabelli, C., Borgatti, M., Mischiati, C., Fibach, E.,
Gambari, R., 2001. Accumulation of gamma-globin mRNA and
induction of erythroid differentiation after treatment of human
leukaemic K562 cells with tallimustine. Br. J. Haematol. 113, 951–
956.
Biedler, J.L., Helson, L., Spengler, B.A., 1973. Morphology and
growth, tumorigenicity and cytogenetics of human neuroblastoma
cells in continuous culture. Cancer Res. 33, 2643–2652.
Boscolo, P., Bergamaschi, A., Di Sciascio, M.B., Benvenuti, F., Reale,
M., Di Stefano, F., Conti, P., Di Gioacchino, M., 2001. Effects of
low frequency electromagnetic fields on expression of lymphocyte
subsets and production of cytokines of men and women employed
in a museum. Sci. Total Environ. 270 (1–3), 13–20.
Bouwens, M., de Kleijn, S., Ferwerda, G., Cuppen, J.J., Savelkoul,
H.F., Kemenade, B.M., 2012. Low-frequency electromagnetic
fields do not alter responses of inflammatory genes and proteins
in human monocytes and immune cell lines. Bioelectromagnetics 33
(3), 226–237.
Brown, Z., Strieter, R.M., Neild, G.H., Thompson, R.C., Kunkel,
S.L., Westwick, J., 1992. IL-1 receptor antagonist inhibits mono-
cyte chemotactic peptide 1 generation by human mesangial cells.
Kidney Int. 42, 95–101.
Cadossi, R., Bersani, F., Cossarizza, A., Zucchini, P., Emilia, G.,
Torelli, G., Franceschi, C., 1992. Lymphocytes and low-frequency
electromagnetic fields. FASEB J. 6 (9), 2667–2674.
Callaghan, M.J., Chang, E.I., Seiser, N., Aarabi, S., Ghali, S.,
Kinnucan, E.R., Simon, B.J., Gurtner, G.C., 2008. Pulsed electro-
magnetic fields accelerate normal and diabetic wound healing by
increasing endogenous FGF-2 release. Plast. Reconstr. Surg. 121,
130–141.
Cane
`, V., Botti, P., Soana, S., 1993. Pulsed magnetic fields improve
osteoblast activity during the repair of an experimental osseous
defect. J. Orthop. Res. 11 (5), 664–670.
Chang, Y., Kong, Q., Shan, X., Tian, G., Ilieva, H., Cleveland, D.W.,
Rothstein, J.D., Borchelt, D.R., Wong, P.C., Lin, C.L., 2008.
Messenger RNA oxidation occurs early in disease pathogenesis and
promotes motor neuron degeneration in ALS. PLoS ONE 3 (8),
e2849.
Cheing, G.L., Li, X., Huang, L., Kwan, R.L., Cheung, K.K., 2014.
Pulsed electromagnetic fields (PEMF) promote early wound
healing and myofibroblast proliferation in diabetic rats. Bioelec-
tromagnetics 35 (3), 161–169.
Chraibi, H., Dereure, O., Te
´ot, L., Guillot, B., 2004. The diagnosis and
treatment of carcinomas occurring at the sites of chronic pressure
ulcers. J. Wound Care 13, 447–448.
Ciccarone, V., Spengler, B.A., Meyers, M.B., Biedler, J.L., Ross, R.A.,
1989. Phenotypic diversification in human neuroblastoma cells:
expression of distinct neural crest lineages. Cancer Res. 49, 219–
225.
Consales, C., Merla, C., Marino, C., Benassi, B., 2012. Electromag-
netic fields, oxidative stress and neurodegeneration. Int J. Cell Biol.
2012, 683897.
Crasson, M., Legros, J.J., Scarpa, P., Legros, W., 1999. 50 Hz
magnetic field exposure influence on human performance and
psychophysiological parameters: two double-blind experimental
studies. Bioelectromagnetics 20 (8), 474–486.
Cushing, S.D., Berliner, J.A., Valente, A.J., Territo, M.C., Navab, M.,
Parhami, F., Gerrity, R., Schwartz, C.J., Fogelman, A.M., 1990.
Minimally modified low density lipoprotein induces monocyte
chemotactic protein 1 in human endothelial cells and smooth
muscle cells. Proc. Natl. Acad. Sci. USA 87, 5134–5138.
Davanipour, Z., Sobel, E., Bowman, J.D., Qian, Z., Will, A.D., 1997.
Amyotrophic lateral sclerosis and occupational exposure to elec-
tromagnetic fields. Bioelectromagnetics 18, 28–35.
Davanipour, Z., Tseng, C.C., Lee, P.J., Sobel, E., 2007. A case-control
study of occupational magnetic field exposure and Alzheimer’s
Experimental model for ELF-EMF exposure 7
Please cite this article in press as: D’Angelo, C. et al., Experimental model for ELF-EMF exposure: Concern for human health. Saudi Journal of Biological Sciences
(2014), http://dx.doi.org/10.1016/j.sjbs.2014.07.006
disease: results from the California Alzheimer’s disease diagnosis
and treatment centers. BMC Neurol. 7, 13.
Di Luzio, S., Felaco, M., Barbacane, R.C., Frydas, S., Grilli, A.,
Castellani, M.L., Macrı
`, M.A., Di Gioacchino, M., Merlitti, D., De
Lutiis, M.A., Masci, S., Di Giulio, C., Cacchio, M., Reale, M.,
2001. Effects of 50 Hz sinusoidal electromagnetic fields on MCP-1
and RANTES generated from activated human macrophages. Int.
J. Immunopathol. Pharmacol. 14 (3), 169–172.
Draper, G., Vincent, T., Kroll, M.E., Swanson, J., 2005. Childhood
cancer in relation to distance from high voltage power lines in
England and Wales: a case-control study. BMJ 330 (7503), 1290.
Eltorai, I.M., Montroy, R.E., Kobayashi, M., Jakowatz, J., Guttierez,
P., 2002. Marjolin’s ulcer in patients with spinal cord injury. J.
Spinal Cord Med. 25 (3), 191–196.
Falone, S., Grossi, M.R., Cinque, B., D’Angelo, B., Tettamanti, E.,
Cimini, A., Di Ilio, C., Amicarelli, F., 2007. Fifty hertz extremely
low-frequency electromagnetic field causes changes in redox and
differentiative status in neuroblastoma cells. Int. J. Biochem. Cell
Biol. 39 (11), 2093–2106.
Fiorani, M., Biagiarelli, B., Vetrano, F., Guidi, G., Dacha
`,M.,
Stocchi, V., 1997. In vitro effects of 50 Hz magnetic fields on
oxidatively damaged rabbit red blood cells. Bioelectromagnetics 18
(2), 125–131.
Fo
¨rstermann, U., Kleinert, H., 1995. Nitric oxide synthase: expression
and expressional control of the three isoforms. Naunyn Schmiede-
bergs Arch. Pharmacol. 352 (4), 351–364.
Frahm, J., Mattsson, M.O., Simko
´, M., 2010. Exposure to ELF
magnetic fields modulate redox related protein expression in mouse
macrophages. Toxicol. Lett. 192 (3), 330–336.
Garcı
`a, A.M., Sisternas, A., Hoyos, S.P., 2008. Occupational exposure
to extremely low frequency electric and magnetic fields and
Alzheimer disease: a meta-analysis. Int. J. Epidemiol. 37, 329–340.
Garip, A.I., Akan, Z., 2010. Effect of ELF-EMF on number of
apoptotic cells; correlation with reactive oxygen species and HSP.
Acta Biol. Hung. 61 (2), 158–167.
Gottwald, E., Sontag, W., Lahni, B., Weibezahn, K.F., 2007.
Expression of HSP72 after ELF-EMF exposure in three cell lines.
Bioelectromagnetics 28 (7), 509–518.
Goudarzi, I., Hajizadeh, S., Salmani, M.E., Abrari, K., 2010. Pulsed
electromagnetic fields accelerate wound healing in the skin of
diabetic rats. Bioelectromagnetics 31, 318–323.
Grant, G., Cadossi, R., Steinberg, G., 1994. Protection against focal
cerebral ischemia following exposure to a pulsed electromagnetic
field. Bioelectromagnetics 15 (3), 205–216.
Grassi, C., D’Ascenzo, M., Torsello, A., Martinotti, G., Wolf, F.,
Cittadini, A., Azzena, G.B., 2004. Effects of 50 Hz electromagnetic
fields on voltage-gated Ca
2+
channels and their role in modulation
of neuroendocrine cell proliferation and death. Cell Calcium 35 (4),
307–315.
Huo, R., Ma, Q., Wu, J.J., Chin-Nuke, K., Jing, Y., Chen, J., Miyar,
M.E., Davis, S.C., Li, J., 2009. Noninvasive electromagnetic fields
on keratinocyte growth and migration. J. Surg. Res. 162 (2), 299–
307.
IARC, 2002. Monographs on the evaluation of carcinogenic risks to
humans. Non-ionizing radiation, Part 1:static and extremely low-
frequency (ELF) electric and magnetic fields, 429 ISBN 92 832 1280
0.
Ikeda, K., Shinmura, Y., Mizoe, H., Yoshizawa, H., Yoshida, A.,
Kanao, S., Sumitani, H., Hasebe, S., Motomura, T., Yamakawa,
T., Mizuno, F., Otaka, Y., Hirose, H., 2003. No effects of
extremely low frequency magnetic fields found on cytotoxic
activities and cytokine production of human peripheral blood
mononuclear cells in vitro. Bioelectromagnetics 24 (1), 21–31.
Iyamu, W.E., Adunyah, S.E., Fasold, H., Horiuchi, K., Elford, H.L.,
Asakura, T., Turner, E.A., 2000. Enhancement of hemoglobin and
F-cell production by targeting growth inhibition and differentiation
of K562 cells with ribonucleotide reductase inhibitors (didox and
trimidox) in combination with streptozotocin. Am. J. Hematol. 63
(4), 176–183.
Jirik, V., Pekarek, L., Janout, V., Tomaskova, H., 2012. Association
between childhood leukaemia and exposure to power-frequency
magnetic fields in Middle Europe. Biomed. Environ. Sci. 25 (5),
597–601.
Jorgensen, W.A., Frome, B.M., Wallach, C., 1994. Electrochemical
therapy of pelvic pain: effects of pulsed electromagnetic fields
(PEMF) on tissue trauma. Eur. J. Surg. Suppl. 574, 83–86.
Julien, J.P., Kriz, J., 2006. Transgenic mouse models of amyotrophic
lateral sclerosis. Biochim. Biophys. Acta 1762 (11–12),
1013–1024.
Kirschenlohr, H., Ellis, P., Hesketh, R., Metcalfe, J., 2012. Gene
expression profiles in white blood cells of volunteers exposed to a
50 Hz electromagnetic field. Radiat. Res. 178 (3), 138–149.
Lappin, M.S., Lawrie, F.W., Richards, T.L., Kramer, E.D., 2003.
Effects of a pulsed electromagnetic therapy on multiple sclerosis
fatigue and quality of life: a double-blind, placebo controlled trial.
Altern. Ther. Health Med. 9 (4), 38–48.
Lazarus, G.S., Cooper, D.M., Knighton, D.R., Percoraro, R.E.,
Rodeheaver, G., Robson, M.C., 1994. Definitions and guidelines
for assessment of wounds and evaluation of healing. Wound Repair
Regen. 2 (3), 165–170.
Leitgeb, N., 2011. Comparative health risk assessment of electromag-
netic fields. Wien. Med. Wochenschr. 161 (9–10), 251–262.
Linet, M.S., Hatch, E.E., Kleinerman, R.A., Robison, L.L., Kaune,
W.T., Friedman, D.R., Severson, R.K., Haines, C.M., Hartsock,
C.T., Niwa, S., Wacholder, S., Tarone, R.E., 1997. Residential
exposure to magnetic fields and acute lymphoblastic leukemia in
children. N. Engl. J. Med. 337 (1), 1–7.
Lisi, A., Foletti, A., Ledda, M., Rosola, E., Giuliani, L., D’Emilia, E.,
Grimaldi, S., 2006. Extremely low frequency 7 Hz 100 microT
electromagnetic radiation promotes differentiation in the human
epithelial cell line HaCaT. Electromagn. Biol. Med. 25 (4), 269–
280.
London, S.J., Thomas, D.C., Bowman, J.D., Sobel, E., Cheng, T.C.,
Peters, J.M., 1991. Exposure to residential electric and magnetic
fields and risk of childhood leukemia. Am. J. Epidemiol. 134, 923–
937.
Lozzio, C.B., Lozzio, B.B., 1975. Human chronic myelogenous
leukemia cell-line with positive Philadelphia-chromosome. Blood
45, 321–334.
Luceri, C., De Filippo, C., Giovannelli, L., Blangiardo, M., Cavalieri,
D., Aglietti, F., Pampaloni, M., Andreuccetti, D., Pieri, L., Bambi,
F., Biggeri, A., Dolara, P., 2005. Extremely low-frequency electro-
magnetic fields do not affect DNA damage and gene expression
profiles of yeast and human lymphocytes. Radiat. Res. 164 (3),
277–285.
Lupke, M., Rollwitz, J., Simko
´, M., 2004. Cell activating capacity of
50 Hz magnetic fields to release reactive oxygen intermediates in
human umbilical cord blood-derived monocytes and in Mono Mac
6 cells. Free Radic. Res. 38 (9), 985–993.
Lupke, M., Frahm, J., Lantow, M., Maercker, C., Remondini, D.,
Bersani, F., Simko, M., 2006. Gene expression analysis of ELFMF
exposed human monocytes indicating the involvement of the
alternative activation pathway. Biochim. Biophys. Acta 1763 (4),
402–412.
Mannerling, A.C., Simko
´, M., Mild, K.H., Mattsson, M.O., 2010.
Effects of 50-Hz magnetic field exposure on superoxide radical
anion formation and HSP70 induction in human K562 cells.
Radiat. Environ. Biophys. 49 (4), 731–741.
Manni, V., Lisi, A., Pozzi, D., Rieti, S., Serafino, A., Giuliani, L.,
Grimaldi, S., 2002. Effects of extremely low frequency (50 Hz)
magnetic field on morphological and biochemical properties of
human keratinocytes. Bioelectromagnetics 23 (4), 298–305.
Markov, M.S., 2007. Expanding use of pulsed electromagnetic field
therapies. Electromagn. Biol. Med. 26 (3), 257–274.
8 C. D’Angelo et al.
Please cite this article in press as: D’Angelo, C. et al., Experimental model for ELF-EMF exposure: Concern for human health. Saudi Journal of Biological Sciences
(2014), http://dx.doi.org/10.1016/j.sjbs.2014.07.006
Martiny, K., Lunde, M., Bech, P., 2010. Transcranial low voltage
pulsed electromagnetic fields in patients with treatment-resistant
depression. Biol. Psychiatry 68 (2), 163–169.
Matic, M., Lazetic, B., Poljacki, M., Djuran, V., Matic, A., Gajinov,
Z., 2009. Influence of different types of electromagnetic fields on
skin reparatory processes in experimental animals. Lasers Med. Sci.
24, 321–327.
Mayer-Wagner, S., Passberger, A., Sievers, B., Aigner, J., Summer, B.,
Schiergens, T.S., Jansson, V., Mu
¨ller, P.E., 2011. Effects of low
frequency electromagnetic fields on the chondrogenic differentia-
tion of human mesenchymal stem cells. Bioelectromagnetics 32,
283–290.
McDonald, D.R., Bamberger, M.E., Combs, C.K., Landreth, G.E.,
1998. Beta-amyloid fibrils activate parallel mitogen-activated pro-
tein kinase pathways in microglia and THP1 monocytes. J.
Neurosci. 18, 4451–4460.
Milgram, J., Shahar, R., Levin-Harrus, T., Kass, P., 2004. The effect
of short, high intensity magnetic field pulses on the healing of skin
wounds in rats. Bioelectromagnetics 25, 271–277.
Miller, S.C., Haberer, J., Venkatachalam, U., Furniss, M.J., 1999. NF-
kappaB or AP-1-dependent reporter gene expression is not altered
in human U937cells exposed to power-line frequency magnetic
fields. Radiat. Res. 151 (3), 310–318.
Murabayashi, S., Yoshikawa, A., Mitamura, Y., 2004. Functional
modulation of activated lymphocytes by time-varying magnetic
fields. Ther. Apher. Dial. 8 (3), 206–211.
Natarajan, M., Nayak, B.K., Galindo, C., Mathur, S.P., Roldan,
F.N., Meltz, M.L., 2006. Nuclear translocation and DNA-binding
activity of NFKB (NF-kappaB) after exposure of human mono-
cytes to pulsed ultra-wideband electromagnetic fields (1 kV/cm)
fails to transactivate kappaB-dependent gene expression. Radiat.
Res. 165 (6), 645–654.
Nindl, G., Balcavage, W.X., Vesper, D.N., Swez, J.A., Wetzel, B.J.,
Chamberlain, J.K., Fox, M.T., 2000. Experiments showing that
electromagnetic fields can be used to treat inflammatory diseases.
Biomed. Sci. Instrum. 36, 7–13.
Noriega-Luna, B., Sabanero, M., Sosa, M., Avila-Rodriguez, M.,
2011. Influence of pulsed magnetic fields on the morphology of
bone cells in early stages of growth. Micron 42 (6), 600–607.
Patruno, A., Amerio, P., Pesce, M., Vianale, G., Di Luzio, S., Tulli, A.,
Franceschelli, S., Grilli, A., Muraro, R., Reale, M., 2010.
Extremely low frequency electromagnetic fields modulate expres-
sion of inducible nitric oxide synthase, endothelial nitric oxide
synthase and cyclooxygenase-2 in the human keratinocyte cell line
HaCat: potential therapeutic effects in wound healing. Br. J.
Dermatol. 162 (2), 258–266.
Pesce, M., Patruno, A., Speranza, L., Reale, M., 2013. Extremely low
frequency electromagnetic field and wound healing: implication of
cytokines as biological mediators. Eur. Cytokine Netw. 24 (1), 1–
10.
Piacentini, R., Ripoli, C., Mezzogori, D., Azzena, G.B., Grassi, C.,
2008. Extremely low-frequency electromagnetic fields pro-
mote in vitro neurogenesis via upregulation of Ca(v)1-channel
activity. J. Cell. Physiol. 215, 129–139.
Poole, C., Ozonoff, D., 1996. Magnetic fields and childhood cancers.
IEEE Eng. Med. Biol. 15, 41–49.
Reale, M., De Lutiis, M.A., Patruno, A., Speranza, L., Felaco, M.,
Grilli, A., Macrı
`, M.A., Comani, S., Conti, P., Di Luzio, S., 2006.
Modulation of MCP-1 and iNOS by 50-Hz sinusoidal electromag-
netic field. Nitric Oxide 15 (1), 50–57.
Roland, D., Ferder, M., Kothuru, R., Faierman, T., Strauch, B., 2000.
Effects of pulsed magnetic energy on a microsurgically transferred
vessel. Plast. Reconstr. Surg. 105, 1371–1374.
Rollwitz, J., Lupke, M., Simko
´, M., 2004. Fifty-hertz magnetic fields
induce free radical formation in mouse bone marrow-derived
promonocytes and macrophages. Biochim. Biophys. Acta 1674 (3),
231–238.
Ro
¨o
¨sli, M., 2008. Commentary: epidemiological research on extremely
low frequency magnetic fields and Alzheimer’s disease-biased or
informative? Int. J. Epidemiol. 37, 341–343.
Savitz, D.A., Wachtel, H., Barnes, F.A., John, E.M., Tvrdik, J.G.,
1988. Case-control study of childhood cancer and exposure to 60-
Hz magnetic fields. Am. J. Epidemiol. 128, 21–38.
Savitz, D.A., Checkoway, H., Loomis, D.P., 1998. Magnetic field
exposure and neurodegenerative disease mortality among electric
utility workers. Epidemiology 9, 398–404.
Simko
`, M., Mattsson, M.O., 2004. Extremely low frequency electro-
magnetic fields as effectors of cellular responses in vitro: possible
immune cell activation. J. Cell. Biochem. 93, 83–92.
Simko
`, M., Droste, S., Kriehuber, R., Weiss, D.G., 2001. Stimulation
of phagocytosis and free radical production in murine macro-
phages by 50 Hz electromagnetic fields. Eur. J. Cell. Biol. 80, 562–
566.
Sobel, E., Davanipour, Z., 1996. Electromagnetic field exposure may
cause increased production of amyloid beta and eventually lead to
Alzheimer’s disease. Neurology 47 (6), 1594–1600.
Standiford, T.J., Kunkel, S.L., Phan, S.H., Rollins, B.J., Strieter,
R.M., 1991. Alveolar macrophage-derived cytokines induce
monocyte chemoattractant protein-1 expression from human
pulmonary type II-like epithelial cells. J. Biol. Chem. 266, 9912–
9918.
Strauch, B., Patel, M.K., Navarro, J.A., Berdichevsky, M., Yu, H.L.,
Pilla, A.A., 2007. Pulsed magnetic fields accelerate cutaneous
wound healing in rats. Plast. Reconstruct. Surg. 120, 425–430.
Sulpizio, M., Falone, S., Amicarelli, F., Marchisio, M., Di Giuseppe,
F., Eleuterio, E., Di Ilio, C., Angelucci, S., 2011. Molecular basis
underlying the biological effects elicited by extremely low-fre-
quency magnetic field (ELF-MF) on neuroblastoma cells. J. Cell.
Biochem. 112 (12), 3797–3806.
Sutbeyaz, S.T., Sezer, N., Koseoglu, F., Kibar, S., 2009. Low-
frequency pulsed electromagnetic field therapy in fibromyalgia: a
randomized, double-blind, sham-controlled clinical study. Clin. J.
Pain 25 (8), 722–728.
Tepper, O.M., Callaghan, M.J., Chang, E.I., Galiano, R.D., Bhatt,
K.A., Baharestani, S., Gan, J., Simon, B., Hopper, R.A., Levine,
J.P., Gurtner, G.C., 2004. Electromagnetic fields increase in vitro
and in vivo angiogenesis through endothelial release of FGF-2.
FASEB J. 18, 1231–1233.
Trimmel, M., Schweiger, E., 1998. Effects of an ELF (50 Hz, 1 mT)
electromagnetic field (EMF) on concentration in visual attention,
perception and memory including effects of EMF sensitivity.
Toxicol. Lett. 96–97, 377–382.
Tsai, M.T., Chang, W.H., Chang, K., Hou, R.J., Wu, T.W., 2007.
Pulsed electromagnetic fields affect osteoblast proliferation and
differentiation in bone tissue engineering. Bioelectromagnetics 28,
519–528.
Tsuchiya, S., Yamabe, M., Yamaguchi, Y., Kobayashi, Y., Konno, T.,
Tada, K., 1980. Establishment and characterization of a human
acute monocytic leukemia cell line (THP-1). Int. J. Cancer 26 (2),
171–176.
Tsuchiya, S., Kobayashi, Y., Goto, Y., Okumura, H., Nakae, S.,
Konno, T., Tada, K., 1982. Induction of maturation in cultured
human monocytic leukemia cells by a phorbol diester. Cancer Res.
42 (4), 1530–1536.
Ulvestad, E., Williams, K., Bjerkvig, R., Tiekotter, K., Antel, J.,
Matre, R., 1994. Human microglial cells have phenotypic and
functional characteristics in common with both macrophages and
dendritic antigen-presenting cells. J. Leukoc. Biol. 56, 732–740.
Van Coillie, E., Van Damme, J., Opdenakker, G., 1999. The MCP/
eotaxin subfamily of CC chemokines. Cytokine Growth Factor
Rev. 10, 61–86.
Vianale, G., Reale, M., Amerio, P., Stefanachi, M., Di Luzio, S.,
Muraro, R., 2008. Extremely low frequency electromagnetic field
enhances human keratinocyte cell growth and decreases proinflam-
Experimental model for ELF-EMF exposure 9
Please cite this article in press as: D’Angelo, C. et al., Experimental model for ELF-EMF exposure: Concern for human health. Saudi Journal of Biological Sciences
(2014), http://dx.doi.org/10.1016/j.sjbs.2014.07.006
matory chemokine production. Br. J. Dermatol. 158 (6), 1189–
1196.
Walleczek, J., 1992. Electromagnetic field effects on cells of the
immune system: the role of calcium signaling. FASEB J. 6 (13),
3177–3185.
Weber, R.V., Navarro, A., Wu, J.K., Yu, H.L., Strauch, B., 2004.
Pulsed magnetic fields applied to a transferred arterial loop support
the rat groin composite flap. Plast. Reconstr. Surg. 114, 1185–1189.
Wei, Y., Xiaolin, H., Tao, S., 2008. Effects of extremely low-
frequency-pulsed electromagnetic field on different-derived osteo-
blast-like cells. Electromagn. Biol. Med. 27 (3), 298–311.
Wertheimer, N., Leeper, E., 1979. Electrical wiring configurations and
childhood cancer. Am. J. Epidemiol. 109, 273–284.
Xie, H.R., Hu, L.S., Li, G.Y., 2010. SH-SY5Y human neuroblastoma
cell line: in vitro cell model of dopaminergic neurons in Parkinson’s
disease. Chin. Med. J. 123 (8), 1086–1092.
10 C. D’Angelo et al.
Please cite this article in press as: D’Angelo, C. et al., Experimental model for ELF-EMF exposure: Concern for human health. Saudi Journal of Biological Sciences
(2014), http://dx.doi.org/10.1016/j.sjbs.2014.07.006
... Furthermore, ELF-EMF has been observed to affect immune system cells, leading to molecular and cellular alterations [10]. The effects of EMF on the immune system appears to be dependent on factors such as frequency, amplitude, exposure time, and the specific biological characteristics of the cells being tested [11]. ...
... Most research in this area has focused on cytokine serum levels [13]. Previous studies have indicated that ELF-EMF exerts its antitumor activity by reducing the levels of interleukin 9 (IL-9), which is associated with inflammatory diseases and autoimmunity, and tumor necrosis factor α (TNF-α), a cytokine involved in coordinating cellular responses and local inflammation [10,13]. Considering the crucial role of cytokines in controlling cancer development, it is essential to examine the impact of ELF-EMF on the immune function of cancer cells without stimulating the immune system. ...
Article
Full-text available
Background: The study aimed to investigate the influence of extremely low-frequency electromagnetic fields (ELF-EMF) on clear cell renal cell carcinoma (ccRCC) by assessing alterations in gene expression and the secretion of cytokines and chemokines. Material and methods: Three ccRCC cell lines (786-O, 769-P, and CAKI-1) and a healthy HEK293 cell line were subjected to ELF-EMF exposure (frequency 50 Hz, magnetic field strength 4.5 mT) for 30 min daily for 5 days. The study examined the expression of ADAM28, NCAM1, and VEGFC genes, along with the secretion of 30 cytokines and chemokines. Results: Notably, primary tumor-derived cell lines, but not those from metastatic sites, exhibited ADAM28 gene expression, which increased following ELF-EMF exposure. A statistically significant reduction in VEGFC gene expression was observed in 769-P cells after ELF-EMF exposure. Additionally, NCAM1 gene expression was upregulated in HEK293, 769-P, and 786-O cells, representing normal embryonic kidney cells and primary tumor cells, but not in CAKI-1 cells, which model metastatic sites. After EMF exposure, there was a statistically significant decrease in transforming growth factor β1 (TGF-β1) concentration in the cell culture supernatants of HEK293 and CAKI-1 cell lines, with no other significant changes in the secretion of tested cytokines. Conclusions: Given the study's findings and available research, caution is warranted when drawing conclusions about the potential inhibitory effect of ELF-EMF on ccRCC progression. Standardization of experimental models is imperative when assessing the effects of EMF in a human context. Med Pr Work Health Saf. 2024;75(2):133-141.
... Electrical lines, transmission towers, telecommunications, household appliances, mobile phones, Wi-Fi, and base stations all generate EMFs (Moon, 2020). Although EMF exposure has always existed, it has been steadily increasing throughout the 21st century due to ambient exposure to artificial EMFs (D'Angelo et al., 2015). The proliferation of electrical and electronic equipment such as personal computers and domestic appliances has made ELF-EMFs increasingly common in the environment (Wang et al., 2016). ...
Article
Full-text available
The widespread use of electrical and electronic devices in the 21st century has been attributed to human exposure to electromagnetic fields (EMFs), and potential public health threats. Amongst other electrical devices, indoor transformer stations have exposed both general public and workers to potentially high levels of extremely low-frequency electromagnetic fields (ELF-EMFs), resulting in acute and severe health implications. In this review, the health effects resulting from occupational and residential exposure to ELF-EMFs from indoor transformer stations were assessed by synthesizing evidence from published studies. Population, Exposure, Comparison and Outcome (PECO) framework was used as a guide in documenting the evidence, and Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) to select studies that match the inclusion criteria. Following the PRISMA guide, a total of 13 studies formed part of this review. Highest magnetic field (MF) exposure level found from a residential exposure assessment study was 11.60 µT, 0.2 m above the transformer station, with the lowest at 0.1 µT due to household electrical appliances. Occupational exposure assessment revealed the highest exposure level of 4.67 µT in the transformer room and lowest at >0.05 µT. Cancer was mostly prevalent in residential studies with longer exposure period (>34 years), and with non-specific exposure symptoms (3 years and 18 months exposure period) within occupational exposure studies. This review found insufficient evidence to suggest the average exposure levels of ELF EMF from indoor transformers, which could pose significant health risks. However, these findings have a noteworthy implication for environmental health and occupational safety.
... There is evidence that EMFs effectively reduce cellular inflammation and pain; indeed, ELF-EMFs have been shown to impact pain and inflammation by modulating G-protein coupling receptors (GPCRs), lowering cyclooxygenase-2 (Cox-2) and nuclear factor kappa B (NF-κB) necessary to induce inflammatory mediators [92]. ELF-EMFs have the ability to shift the transition from a pro-inflammatory to an anti-inflammatory state, upregulate nitric oxide synthase (NOS) activity, and downregulate Cox-2 expression and Prostaglandin E2 (PGE-2) production, which are involved in the modulation of inflammatory reactions [9,93]. ...
Article
Full-text available
A general theory explaining how electromagnetic waves affect cells and biological systems has not been completely accepted yet; nevertheless, extremely low-frequency electromagnetic fields (ELF-EMFs) can interfere with and modify several molecular cellular processes. The therapeutic effect of EMFs has been investigated in several clinical conditions with promising results: in this context a better understanding of mechanisms by which ELF-EMF influences cellular events is necessary and it could lead to more extended and specific clinical applications in different pathological conditions. This paper develops a thermodynamic model to explain how ELF-EMF directly interferes with the cellular membrane, inducing a biological response related to a cellular energy conversion and modification of flows across cell membranes. Indeed, energy, irreversibly consumed by cellular metabolism, is converted into entropy variation. The proposed thermodynamic model views living systems as adaptative open systems, analysing the changes in energy and matter moving in and out of the cell.
... In [4] experimental model for ELF-EMF exposure: Human Health Concerns was the title of an essay that was written. Low frequency (LF) electromagnetic fields (EMFs) are prevalent in modern society, according to their research, and interest in the probable effects of extremely low frequency (ELF) EMFs on human health has risen steadily over the last 20 years. ...
Article
Public Health awareness of the exposure to radiofrequency electromagnetic fields (Rf-EMF) is necessary for epidemiological studies on possible bio-effects. The Knowledge will promote good health, reduce health risk factors, lower morbidity and mortality rates often associated with public health environment (PHE). Modeling EMF radiation variability and its application is vital given the prospective growth in both the number of mobile devices and equipment radiating electromagnetic fields (EMF) and the increasing concerns in the general public. The main goal of this work is to develop a framework based on Machine Learning (ML) approach to determine the exposure level on time-based variances within and during operational procedures of frequency bands used in mobile telecommunication, digital devices, transmitters, broadcasting terminals etc. The system adopts the measurements and instrumentation methodology, using a handheld Poniie pn8000 multi-field EMF metre and EME Spy exposimeter with a detection limit of 0.0066mW/m (2) with sampling at certain interval in a specific frequency ensemble. The measurements were synthesized as exposure coefficients with assessment based on EMF radiation from emitting devices and matching with International Commission on Non-Ionizing Radiation Protection (ICNIRP) standards endorsed by WHO. Consequently, obtained data was processed and segmented into training and test sets on 70:30 ratios respectively. Computing modeling of the system was performed with R programming language using data obtained from field measurements and records. The prosed system used Ensemble ML-Random Forest Algorithm and Decision Tree algorithm (DTA). Performance evaluation and results obtained clearly demonstrates 87% based on the Confusion Matrix (CM) outcomes for Random Forest and Decision Tree presents 56%. This justifies the capability of ML techniques in accurately classifying EMF radiation to mitigate exposure rate and improve healthcare throughput. The model was transformed and deployed in a production system API environment with Graphical User Interface (GUI) for flexibility. This work revealed relating potential health bio-effects associated with EMF exposure and the different parameters that are currently used for evaluating, limiting, and mitigating the impact of the exposure on the general public.
... Depending on frequency, exposure time or type of electromagnetic wave used in experiment as well as type of cells tested, carcinogenic, proliferative and antiproliferative effects were distinguished. Several studies established a connection between ELF magnetic field exposure and leukemia and brain tumors [6] while others discarded such influence [7]. The possible carcinogenic influence caused by increase in the oxidative stress in cells exposed to ELF-EMF was also shown in studies [8]. ...
Chapter
Full-text available
Extremely low frequency electromagnetic field (ELF-EMF) is generated among others by power lines and electrical devices; therefore, most people are exposed to its effects each day. This experiment models a typical week of workers' exposure to ELF-EMF with consideration of a two-day weekend break and analyzes its effect on human cells. Determination of proliferation potential was performed on HeLa cells with intermittent ELF-EMF of 50 Hz generated by a coil particularly measured for this experiment. Cytotoxicity was examined by a clonogenic assay that permits to inspect the ability of formation clonogenic colonies by tested cells. For this purpose, cells were cultivated in three groups: control one (cultivated in normal conditions), sham one (cultivated in an unwired coil in normal conditions) and exposed one (cultivated for 5 days in conditions of intermittent irradiation followed by two days of relaxation). To analyze the experiment's results appropriate devices and statistical tools have been used. Keywords: electromagnetic field, HeLa cells, cancer, proliferation, cytotoxicity
... 3 As a lateral effect, continuous exposure to EMF influences the human body's delicate and sensitive biological system, leading to further complications. 4 The most sensitive organ to EMF is sought to be the nervous system 5,6 Particularly, with the constant use of mobile phones and exposure to cellular antennas, there is a growing concern and interest in the effects of EMF exposure on central nervous system (CNS) functionality. However, the exact mechanisms and interactions between EMF and biological systems are poorly understood. ...
Article
Full-text available
Introduction Radiofrequency electromagnetic radiation (RF-EMR) and extremely low-frequency electromagnetic fields (ELF-EMF) have emerged as noteworthy sources of environmental pollution in the contemporary era. The potential biological impacts of RF-EMR and ELF-EMF exposure on human organs, particularly the central nervous system (CNS), have garnered considerable attention in numerous research studies. Methods This article presents a comprehensive yet summarized review of the research on the explicit/implicit effects of RF-EMR and ELF-EMF exposure on CNS performance. Results Exposure to RF-EMR can potentially exert adverse effects on the performance of CNS by inducing changes in the permeability of the blood-brain barrier (BBB), neurotransmitter levels, calcium channel regulation, myelin protein structure, the antioxidant defense system, and metabolic processes. However, it is noteworthy that certain reports have suggested that RF-EMR exposure may confer cognitive benefits for various conditions and disorders. ELF-EMF exposure has been associated with the enhancement of CNS performance, marked by improved memory retention, enhanced learning ability, and potential mitigation of neurodegenerative diseases. Nevertheless, it is essential to acknowledge that ELF-EMF exposure has also been linked to the induction of anxiety states, oxidative stress, and alterations in hormonal regulation. Moreover, ELF-EMR exposure alters hippocampal function, notch signaling pathways, the antioxidant defense system, and synaptic activities. Conclusion The RF-EMR and ELF-EMF exposures exhibit both beneficial and adverse effects. Nevertheless, the precise conditions and circumstances under which detrimental or beneficial effects manifest (either individually or simultaneously) remain uncertain.
Article
Objective PEMF is an emerging technique in the treatment of Parkinson's disease (PD) due to its potential improvement of movement speed. The aim of this study was to investigate the metabolic profiles of pulsed electromagnetic fields (PEMFs) in an SH-SY5Y cell model of PD. Methods The SH-SY5Y cell model of PD was induced by 1-methyl-4-phenylpyridinium (MPP⁺). Liquid chromatography mass spectrometry (LC‒MS)-based untargeted metabolomics was performed to examine changes in the PD cell model with or without PEMF exposure. We conducted KEGG pathway enrichment analysis to explore the potentially related pathways of the differentially expressed metabolites. Results A total of 275 metabolites were annotated, and 27 significantly different metabolites were found between the PEMF treatment and control groups (VIP >1, P < 0.05), mainly including 4 amino acids and peptides, 4 fatty acid esters, 2 glycerophosphoethanolamines, 2 ceramides and 2 monoradylglycerols; among them, 12 metabolites were upregulated, and 15 were downregulated. The increased expression levels of glutamine, adenosine monophosphate and taurine were highly associated with PEMF stimulation in the PD model. The enrichment results of differentially abundant metabolite functional pathways showed that biological processes such as the mTOR signaling pathway, PI3K-Akt signaling pathway, and cAMP signaling pathway were significantly affected. Conclusion PEMFs affected glutamine, adenosine monophosphate and taurine as well as their functional pathways in an in vitro model of PD. Further functional studies regarding the biological effect of these changes are required to evaluate the clinical efficacy and safety of PEMF treatment in PD.
Article
Purpose The present study aimed to assess the effects of extremely low-frequency electromagnetic fields (ELF-MF) on structural changes of human osteosarcoma cells by analyzing the stained cytoskeleton for assessing the relationship between the fractal dimension parameter and proliferation rate of radiation-induced cells. Materials and Methods In this study, 2-mT magnetic fields with various waveforms, including sinusoidal, triangular, and pulsed shapes, were employed to determine the biological effects of ELF-EMF on the human osteosarcoma MG-63 cell line. All experiments were performed in two modes: continuous exposure at 3 h and fractionated irradiations at 3 consecutive days. Afterward, the proliferation assay was implemented for assessing the cell proliferation in each group. Moreover, immunofluorescence staining and confocal imaging were performed to determine the cell shape index. Furthermore, fractal dimension analysis was carried out by processing morphological images. Results The proliferation and shape index parameters of radiation-induced osteosarcomas significantly decreased compared with non-irradiated cells. In addition, fractal dimensions significantly increased following fractionated exposure at 3 consecutive days. Conclusions Assessing the fractal dimensions can be considered as a new morphological index for the prognosis of the structural remodeling of human osteosarcoma cells in response to fractionated irradiation of ELF-MF. In addition, various waveforms induce a similar effect on morphological remodeling and cell proliferation.
Article
Full-text available
Previous investigations have demonstrated the therapeutic advantages of extremely low-frequency electromagnetic fields (ELF-EMFs) in mitigating inflammation and influencing biological processes. We aimed to shed light on the effects of ELF-EMF on recovery rate following high-intensity exercise. Nine male athletes (26.7 ± 6.0 years; 69.6 ± 7.7 kg, VO2peak 57.3 ± 6.8 mL/kg/min) completed five visits in a double-blinded crossover design, performing two consecutive testing days, following a ventilatory thresholds assessment. Following 62 min of high-intensity cycling, participants lay on an ELF-EMF mattress under active (A) and non-active (NA) conditions, immediately post protocol and during the night. Physical performance and blood markers were assessed at baseline and at 60 min (60 P) and 24 h (24 H) post-protocol. The A-condition demonstrated a notable reduction in interleukin-10 (IL-10) concentrations (mean difference = −88%, p = 0.032) and maximal isometric strength of the quadriceps muscles (mean difference = ~8%, p = 0.045) compared to the NA-condition between 60 P and 24 H. In a sensitivity analysis, the A-condition revealed that younger athletes who possessed lower fat mass experienced attenuated inflammation and biochemical responses and improved physical performance. In conclusion, ELF-EMF showed no significant overall effects on performance and inflammation after intense cycling among athletes. Post-hoc analysis revealed modest benefits of ELF-MLF, suggesting a context-dependent impact. Further research with a larger sample size and multiple sessions is needed to confirm the recovery potential of ELF-EMF.
Article
Full-text available
Objectives Leukaemia is the most common cancer in children, but its aetiology is still poorly understood. We tested the hypothesis that traffic-related air pollution is associated with paediatric leukaemia because of chronic exposure to several potential carcinogens. Methods The Italian SETIL study (Study on the aetiology of lymphohematopoietic malignancies in children) was conducted in 14 Italian regions. All incident cases of leukaemia in children aged ≤10 years from these regions (period 1998–2001) were eligible for enrolment. Two controls per case, matched on birth date, gender and region of residence were randomly selected from the local population registries. Exposure assessment at birth residence included traffic indicators (distance to main roads and length of main roads within 100 m) and estimates of pollutants concentrations (particulate matter -PM2.5 and PM10- and gases -NO2 and O3-) from national dispersion model and land use regression models. The association between the exposure variables and leukaemia was assessed by logistic regression analyses. Results Participation rates were 91.4% among cases and 69.2% in controls; 620 cases (544 acute lymphocytic and 76 acute non-lymphocytic leukaemia) and 957 controls were included. Overall, when considering the residence at birth, 35.6% of cases and 42.4% of controls lived along busy roads, and the mean annual PM10 levels were 33.3 (SD=6.3) and 33.4 µg/m3 (SD=6.5), respectively. No association was found, and all ORs, independent of the method of assessment and the exposure windows, were close to the null value. Conclusions Using various exposure assessment strategies, air pollution appears not to affect the incidence of childhood leukaemia.
Article
Full-text available
Wound healing is a highly coordinated and complex process involving various cell types, chemical mediators and the surrounding extracellular matrix, resulting in a tightly orchestrated re-establishment of tissue integrity by specific cytokines. It consists of various dynamic processes including a series of overlapping phases: inflammation, proliferation, re-epithelialization and remodeling. One of the underlying mechanisms responsible for the disturbances in wound healing is an out-of-control inflammatory response that can cause pathological consequences, such as hypertrophic scars, keloids or chronic wounds and ulcers. Recently, several reports have evaluated the effects of extremely low frequency electromagnetic fields (EMFs) on tissue repair. In particular, the data analysis supports an anti-inflammatory effect of EMFs by the modulation of cytokine profiles that drive the transition from a chronic pro-inflammatory state to an anti-inflammatory state of the healing process. In this review, we focus on the effect of EMFs on skin wound healing showing emerging details of the anti-inflammatory effects of EMFs, with a view to cytokines as candidate biomarkers. Molecular clarification of the mechanisms involved in the modulation of inflammatory factors following exposure to EMFs will provide a better understanding of the cellular responses induced by EMFs and a potential, additional treatment in non-responding, chronic wounds.
Article
A cell-line derived from a patient with chronic myelogenous leukemia (CML) is described. The new cell-line, which has over 175 serial passanges in a 3 1/2-yr period, has the following characteristics: (1) CML cells started to proliferate actively since they were first incubated in culture media. A threefold increase in the total number of cells was observed during the first seven passages; the cell population increased by a factor of 10 to 20 every 7 days from passage 8 through 85; from 20 to 40 times from passage 86 through 150, and more than 40 times after 150 passages. (2) The majority of the nononucleated cells are undifferentiated blasts. (3) The karyotype of all the cells examined show the Philadelphia (Ph1) chromosome and a long acrocentric marker plus aneuploidy. The Giemsa-banding studies identified the Ph1 chromosome as a terminal deletion of the long arm of chromosome 22:del(22)(q12) and the long acrocentric marker as an unbalanced reciprocal translocation of one chromosome 17 and the long arm of one chromosome 15. (4) The CML cells do not produce immunoglobulins, are free of mycoplasma, Epstein-Barr virus, and herpes-like virus particles. (5) CML cells have no alkaline phosphatase and myeloperoxidase activities and did not engulf inert particles. (6) Cultured CML cells provide a constant source of a specific antigen. This CML cell-line represents a unique source of CML cells with meaningful indicators of malignancy for clinical and experimental studies.
Article
Background: Chronic wounds represent a worldwide problem. For laboratory and clinical research to adequately address this problem, a common language needs to exist. Observation: This language should include a system of wound classification, a lexicon of wound descriptors, and a description of the processes that are likely to affect wound healing and wound healing end points. Conclusions: The report that follows defines wound, acute wound, chronic wound, healing and forms of healing, wound assessment, wound extent, wound burden, and wound severity. The utility of these definitions is demonstrated as they relate to the healing of a skin wound, but these definitions are broadly applicable to all wounds.
Article
Reduced collagen deposition possibly leads to slow recovery of tensile strength in the healing process of diabetic cutaneous wounds. Myofibroblasts are transiently present during wound healing and play a key role in wound closure and collagen synthesis. Pulsed electromagnetic fields (PEMF) have been shown to enhance the tensile strength of diabetic wounds. In this study, we examined the effect of PEMF on wound closure and the presence of myofibroblasts in Sprague-Dawley rats after diabetic induction using streptozotocin. A full-thickness square-shaped dermal wound (2 cm × 2 cm) was excised aseptically on the shaved dorsum. The rats were randomly divided into PEMF-treated (5 mT, 25 Hz, 1 h daily) and control groups. The results indicated that there were no significant differences between the groups in blood glucose level and body weight. However, PEMF treatment significantly enhanced wound closure (days 10 and 14 post-wounding) and re-epithelialization (day 10 post-wounding), although these improvements were no longer observed at later stages of the wound healing process. Using immunohistochemistry against α-smooth muscle actin (α-SMA), we demonstrated that significantly more myofibroblasts were detected on days 7 and 10 post-wounding in the PEMF group when compared to the control group. We hypothesized that PEMF would increase the myofibroblast population, contributing to wound closure during diabetic wound healing. Bioelectromagnetics. © 2014 Wiley Periodicals, Inc.
Article
IL-1 receptor antagonist inhibits monocyte chemotactic peptide 1 generation by human mesangial cells. The elicitation of neutrophils and monocytes from the circulation into the inflamed glomerulus is a key process in the pathogenesis of proliferative glomerulonephritis. The aim of this study was to determine the factors which regulate the expression and synthesis of the monocyte specific chemotaxin, monocyte chemotactic peptide 1 (MCP-1). Mesangial cells in culture did not constitutively express MCP-1, but could be induced to express both MCP-1 mRNA and antigenic MCP-1 by either stimulation with IL-1α or TNFα, which are also stimuli for interleukin 8 (IL-8/NAP-1) expression and release. Pre-treatment of mesangial cells with the IL-1 receptor antagonist (IL-1ra) induced dose-dependent inhibition of both the expression of MCP-1 and IL-8 mRNA as well as the release of both chemotactic peptides in response to IL-1α, while the receptor antagonist had no significant effect on TNFα induced MCP-1 and IL-8 generation. This study demonstrates that the IL-1 receptor antagonist was four times more effective at inhibiting the IL-1 induced expression and release of IL-8 compared to that of MCP-1. These results suggest that mesangial cell-derived MCP-1 may play an important role in the recruitment of monocytes in glomerular inflammation and that an IL-1 receptor antagonist may have therapeutic potential for the treatment of glomerulonephritis.
Article
This study was conducted on 232 cases of childhood leukemia occurring in children age 10 and under between 1980 and 1987 in Los Angeles County. Two hundred thirty-two controls were selected from the same geographic area and were matched on sex, age and race. The parents of the 464 subjects were interviewed by telephone to elicit information on medical histories of the parents and child, residential histories of the subjects, occupational histories of both parents, environmental chemical histories, personal histories including drug use and smoking habits, and time and space occupancy of subjects, including exposures to electrical appliances. An extensive assessment of exposure to electric and magnetic fields was made by determining wiring configurations of most subjects (90%), by measuring electric and magnetic fields in various areas of the inside and outside of the home, and by measuring magnetic fields for 24 to 72 hours in the child's sleeping area (66%). We conclude that our data offer no support for a relationship between measured electric field exposure and leukemia risk, little support for the relationship between measured magnetic field exposure and leukemia risk, considerable support for a relationship between wiring configuration and leukemia risk, and considerable support for a relationship between children's electrical appliance use and leukemia risk. The reason(s) why wiring configuration correlates with leukemia risk better than measured exposure are not clear. It is also not clear whether short-term, very high exposure of children to magnetic (or electric) fields from electric appliances are responsible for the observed risk or whether associated exposures or recall biases are responsible. These latter two issues deserve continued research. 41 refs., 31 tabs.