ArticlePDF Available

Real-time measurement of cytosolic free calcium concentration in Jurkat cells during ELF magnetic field and evaluation of the role of cell cycle

Authors:

Abstract and Figures

Extremely low frequency magnetic fields (ELF MF) have been reported to alter a number of cell signaling pathways, including those involved in proliferation, differentiation and apoptosis where cytosolic free calcium ([Ca(2+)](c)) plays an important role. To better understand the biological conditions under which ELF MF exposure might alter [Ca(2+)](c), we measured [Ca(2+)](c) by ratiometric fluorescence spectrophotometry during exposure to ELF MF in Jurkat E6.1 cells synchronized to different phases of the cell cycle. Suspensions of cells were exposed either to a near zero MF (Null) or a 60 Hz, 100 microT sinusoidal MF superimposed upon a collinear 78.1 microT static MF (AC + DC). An initial series of experiments indicated that the maximum increase in [Ca(2+)](c) above baseline after stimulation with anti-CD3 was significantly higher in samples exposed to AC + DC (n = 30) compared to Null (n = 30) with the largest difference in G2-M enriched samples. However, in a second study with G2-M enriched cells, samples treated with AC + DC (n = 17) were not statistically different from Null-treated samples (n = 27). Detailed analysis revealed that the dynamics in [Ca(2+)](c) before and after stimulation with anti-CD3 were dissimilar between Null samples from each study. From the results, we concluded (i) that the ELF MF increased [Ca(2+)](c) during an antibody-induced signaling event, (ii) that the ELF MF effect did not depend to a large degree on cell cycle, and (iii) that a field-related change in [Ca(2+)](c) signaling appeared to correlate with features in the [Ca(2+)](c) dynamics. Future work could evaluate [Ca(2+)](c) dynamics in relation to the phase of the cell cycle and inter-study variation, which may reveal factors important for the observation of real-time effects of ELF MF on [Ca(2+)](c).
Content may be subject to copyright.
Bioelectromagnetics 27:354^ 364 (2006)
Real-Time Measurement of Cytosolic Free
Calcium Concentration in Jurkat Cells During
ELF Magnetic Field Exposure and Evaluation
of the Role of Cell Cycle
Cheryl R. McCreary,
1,4
S. Jeffrey Dixon,
5
Laurence J. Fraher,
3,6
Jeffrey J.L. Carson,
1,4
and Frank S. Prato
1,2,4
*
1
Imaging Program, Lawson Health Research Institute, London, Ontario, Canada
2
Department of Nuclear Medicine, St. Josephs Health Care, London, Ontario, Canada
3
Departments of MedicalandBiochemistry, University of Western Ontario,
London, Ontario, Canada
4
Medical Biophysics, University of Western Ontario, London, Ontario, Canada
5
Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
6
Metabolism and Diabetes Program, Lawson Health Research Institute, London,
Ontario, Canada
Extremely low frequency magnetic fields (ELF MF) have been reported to alter a number of cell
signaling pathways, including those involved in proliferation, differentiation and apoptosis where
cytosolic free calcium ([Ca
2þ
]
c
) plays an important role. Tobetter understand the biological conditions
under which ELF MF exposure might alter [Ca
2þ
]
c
, we measured [Ca
2þ
]
c
by ratiometric fluorescence
spectrophotometry during exposure to ELF MF in Jurkat E6.1 cells synchronized to different phases of
the cell cycle. Suspensions of cells were exposed either to a near zero MF (Null) or a 60 Hz, 100 mT
sinusoidal MF superimposed upon a collinear 78.1 mT static MF (AC þDC). An initial series of
experiments indicated that the maximum increase in [Ca
2þ
]
c
above baseline after stimulation with
anti-CD3 was significantly higher in samples exposed to AC þDC (n¼30) compared to Null (n¼30)
with the largest difference in G2-M enriched samples. However, in a second study with G2-M enriched
cells, samples treated with AC þDC (n¼17) were not statistically different from Null-treated
samples (n¼27). Detailed analysis revealed that the dynamics in [Ca
2þ
]
c
before and after stimulation
with anti-CD3 were dissimilar between Null samples from each study. From the results, we concluded
(i) that the ELF MF increased [Ca
2þ
]
c
during an antibody-induced signaling event, (ii) that the ELF
MF effect did not depend to a large degree on cell cycle, and (iii) that a field-related change in [Ca
2þ
]
c
signaling appeared to correlate with features in the [Ca
2þ
]
c
dynamics. Future work could evaluate
[Ca
2þ
]
c
dynamics in relation to the phase of the cell cycle and inter-study variation, which may reveal
factors important for the observation of real-time effects of ELF MF on [Ca
2þ
]
c
. Bioelectromagnetics
27:354– 364, 2006. 2006 Wiley-Liss, Inc.
Key words: 60 Hz AC; DC; fluorescence; ratiometric measurement; indo-1
INTRODUCTION
At the cellular level, weak (<1 mT), extremely
low frequency (<300 Hz) magnetic fields (ELF MF)
have been shown to alter immune function, cell
proliferation, gene expression, DNA repair, and
second messenger pathways such as calcium signaling
(reviewed in Lacy-Hulbert et al., 1998; Loscher and
Liburdy, 1998). Calcium is a ubiquitous second
messenger that is involved in the regulation of many
physiological functions. Measurement of cytosolic free
calcium concentration ([Ca
2þ
]
c
) after or during ELF
MF exposure led investigators to suggest that field-
induced alterations in [Ca
2þ
]
c
might lead to down-
stream physiological effects. For example, Carson et al.
[1990] were the first to report an increase in [Ca
2þ
]
c
in
HL60 cells during ELF MF exposure. Lindstrom et al.
[1993, 1995b] found that exposure of quiescent Jurkat
2006 Wiley-Liss, Inc.
—————
Grant sponsor: Canadian Institutes of Health Research (CIHR).
*Correspondence to: Frank S. Prato, Imaging Program, Lawson
Health Research Institute, 268 Grosvenor Street, London, Ont.,
Canada. E-mail: prato@lawsonimaging.ca
Received for review 12 September 2005; Final revision received
10 March 2006
DOI 10.1002/bem.20248
Published online 19 May 2006 in Wiley InterScience
(www.interscience.wiley.com).
E6.1 cells to a 50 Hz MF induced [Ca
2þ
]
c
oscillations.
In another study with Jurkat cells, the power spectral
density of [Ca
2þ
]
c
oscillations was reduced in cells
exposed to ELF MF [Galvanovskis et al., 1999]. While
the literature contains many positive findings, negative
results also have been reported. In one case, [Ca
2þ
]
c
oscillations recorded in Jurkat cells were unaffected
during exposure to 50 Hz MF [Wey et al., 2000]. Other
laboratories have observed calcium signaling events in
a number of cell types and found that exposure to ELF
MF had no effect [Lyle et al., 1997; Sisken and
DeRemer, 2000; Shahidain et al., 2001; Craviso et al.,
2002; Madec et al., 2003]. Differences in study outcome
may be related to a number of methodological differ-
ences between laboratories: (i) MF exposure conditions
were often different between studies and between
laboratories, which complicated interpretation of
results even on the same biological endpoint. (ii) There
were often inconsistencies in the biological protocol.
For instance, two laboratories used different criteria
to select individual cells for inclusion during [Ca
2þ
]
c
measurements [Lindstrom et al., 1993; Wey et al.,
2000]. (iii) Differences in biochemical conditions could
lead to different experimental outcomes. For example,
intracellular and extracellular calcium levels were
shown to influence cellular functions such as apoptosis,
differentiation, and secretion during MF exposure
[Karabakhtsian et al., 1994; Morgado-Valle et al.,
1998; Fanelli et al., 1999; Zhou et al., 2002; Gobba et al.,
2003]. (iv) Finally, the physiological state of the cell at
the time of MF exposure may have been important
[Walleczek and Liburdy, 1990; Walleczek and
Budinger, 1992; Nindl et al., 1997; Diniz et al., 2002].
Data from our lab also support this last possibility. In
previous work, we identified a number of factors that
obscured the effect of ELF MF exposure on [Ca
2þ
]
c
in
Jurkat cells, one of which included the distribution of
cells within the cell cycle [McCreary et al., 2002].
The purpose of the work described here was to
extend our previous findings with Jurkat E6.1 cells by
measurement of [Ca
2þ
]
c
dynamics during ELF MF
exposure of samples enriched at one of three phases of
the cell cycle. We exposed cell samples enriched in G0/
G1, S, and G2-M phases to either a 60 Hz, 100 mT
1
sinusoidal MF collinear with a 78.1 mT static MF
(AC þDC) or a control condition in which the ambient
static MF was zeroed to a tolerance of 0.5 mT (Null).
These MF conditions were selected since in our
laboratory they have been associated with effects on
[Ca
2þ
]
c
in vitro [McCreary et al., 2002] and on the
response to a thermal stimulus in vivo [Prato et al.,
1995, 2000]. In addition, the combination of time-
varying and static MF satisfied the theoretical predic-
tions of the ion resonance model and its variants
[Lednev, 1991; Blanchard and Blackman, 1994], which
have shown consistency with experimental results from
other laboratories [Blackman et al., 1995; Baureus
Koch et al., 2003; Sarimov et al., 2005].
MATERIALS
The Jurkat E6.1 clonal cell line (TIB-152) was
purchased from the American Type Culture Collection
(ATCC, Manassas, VA). Fetal bovine serum and
antibiotic/antimycotic solution (10 000 units/ml pen-
icillin, 10 000 mg/ml streptomycin, and 25 mg/ml
amphotericin B) were purchased from Invitrogen
(Burlington, ON). The acetoxymethyl ester form of
the fluorescent calcium indicator indo-1 (indo-1 AM)
was purchased from Molecular Probes (Eugene, CA,
USA). The monoclonal antibody, anti-CD3 (UCHT-1
mouse anti-human), was purchased from ID Labs
(London, ON, Canada). Modified RPMI 1640 medium
(with L-glutamine and without phenol red or sodium
bicarbonate) and all other chemicals were obtained
from Sigma-Aldrich Canada (Oakville, ON, Canada).
Buffered saline solution (BSS) was prepared in our
laboratory and contained (in mM): 135 NaCl, 5 KCl,
1 MgCl
2
, 1 CaCl
2
, 10 glucose, and 10 HEPES (pH 7.30).
Conditioned RPMI 1640 medium (cRPMI) was pre-
pared in our laboratory by culturing Jurkat E6.1 cells
(67 10
5
cells per ml) in modified RPMI 1640 with
10% fetal bovine serum, 1% antibiotic/antimycotic
solution, 10 mM HEPES, and 23.8 mM sodium
bicarbonate (supplemented RPMI) for 24 h. The cells
were pelleted by centrifugation (300gfor 5 min), the
supernatant collected and the conditioned medium was
stored at 70 8C.
METHODS
Overview
The potential effect of exposure to ACþDC MF
on [Ca
2þ
]
c
homeostasis and [Ca
2þ
]
c
signaling was
examined by comparing MF-exposed samples to
samples treated with a Null MF condition, which
represented our standard control.
2
Normalized ratios
—————
1
AC magnetic field strength here and throughout the text reported
as peak amplitude, that is, one half of the peak to trough difference
in the waveform.
—————
2
In the natural environment biological systems are exposed to the
earth’s magnetic field. For this reason, it could be argued that a
Null MF may not be a suitable control condition; however,
calcium signaling measurements performed in Jurkat cells in our
lab indicated that there was no difference between samples
exposed to a DC MF similar to that of the earth’s MF and samples
exposed to a Null MF.
Real-Time Measurement of [Ca
2þ
]
c
355
from synchronized Jurkat cells loaded with indo-1 were
obtained from fluorescence measurements on different
samples that contained cells at one of three phases of the
cell cycle. This approach resulted in six experimental
groups: three distributions of cells in the cell cycle and
two MF conditions. In each experiment, the dynamics
of [Ca
2þ
]
c
during MF was monitored at three different
stages of activation by anti-CD3: resting or basal
[Ca
2þ
]
c
prior to anti-CD3, the initial [Ca
2þ
]
c
response
after anti-CD3, and the sustained [Ca
2þ
]
c
response after
anti-CD3.
Cell Culture
Cells were grown in RPMI, passaged twice
weekly, and diluted with fresh medium once between
passages. Cultures were kept at 37 8C inside a vented
Mu-metal box within an incubator that contained a
humidified atmosphere of 5% CO
2
and 95% air. The
Mu-metal box (33 38 20 cm) was constructed of
1 mm thick nickel iron alloy with four 2.5 cm diameter
circular vents on both the top and bottom of the box. The
vents were topped with 2.5 cm long Mu-metal cylinders
to improve the degree of ambient MF shielding. This
ensured that the cell cultures were exposed to low static
(<0.3 mT) and ELF (<0.001 mT) MF during culturing
and that the MF exposure prior to the experiments
was standardized. The cell culture from ATCC was
expanded through four passages to form a bank of stock
cells frozen with 7% dimethyl sulfoxide (DMSO) and
kept in a liquid nitrogen cryotank. Stock cells were then
thawed, washed with RPMI and grown for subsequent
experimentation. All of the cells used in these experi-
ments were passaged fewer than 12 times after arrival in
the laboratory.
Synchronization and DNA Analysis
Jurkat E6.1 cells were reversibly arrested in
S phase by treatment with 200 ng/ml aphidicolin for
1819 h [Merrill, 1998; Kai et al., 1999]. Cell density
(by Neubaum hemocytometer), viability (by trypan
blue exclusion), and DNA content (by flow cytometry)
were determined every 2– 5 h thereafter for 30 h. For
flow cytometry, a sample of approximately 1 10
6
aphidicolin-treated cells was taken from the culture
flask and centrifuged at 500g. The cells were resus-
pended in 250 ml of phosphate-buffered saline (PBS)
with 0.1% bovine albumin and fixed with 750 ml of 70%
cold ethanol. Samples were cooled on ice and stored at
48C for up to 2 weeks. The fixed Jurkat cells were
washed twice with PBS, treated with 1 mg/ml RNase A,
then labeled with propidium iodide (27 mg/ml) for at
least 1 h at 4 8C.
A Coulter XL-MCL (Beckman-Coulter, Miami,
FL, USA) was used for flow cytometry of DNA content
(excitation with 15 mW Argon laser at 488 nm with
fluorescence detection at 620 nm). At least 10 000
events were collected for each determination of the
distribution of cells within the cell cycle. Multicycle
software (Phoenix Flow Systems, San Diego, CA,
USA) was used to fit the ungated data. The signal from
debris and clumping was fit using a non-linear, least
squares fitting algorithm that assumed the probability of
aggregation between any two cells was independent of
phase of the cell cycle. The clumping fraction of the
signal was subtracted from the total signal before the
distribution of cells within the cell cycle was deter-
mined. To allow sufficient time for more than one
experiment to be performed at a given time point, that is,
both Null and AC þDC from the same preparation,
progression of the cell cycle was slowed by incubation
at 4 8C for up to 3.5 h. From the cell density and flow
cytometry data, it was determined that G0/G1, S, and
G2-M phases were maximally enriched 1418, 14,
and 6-8 h after aphidicolin treatment, respectively (data
not shown).
Sample Preparation and
Fluorescence Measurement
Cell culture, sample handling, indo-1 AM loading
and fluorescence measurement have been described
previously [Carson and Prato, 1996; McCreary et al.,
2002]. Briefly, aliquots of cells were taken from the
suspension cultures, washed, and resuspended in 1 ml
BSS. A sample of 1.5 10
6
cells was treated with 6 ml
DMSO. Three more samples each of 1.5 10
6
cells was
taken from the same flask and treated with 1 mM indo-1
AM in 6 ml DMSO. The four samples were incubated in
an open circulating water bath at 37 8C for 30 min the
pump motor was 2 m distant from the bath to minimize
possibility of stray MF. After incubation, the cells were
washed three times to remove residual extracellular
indo-1 AM. Both the DMSO and the indo-1 AM treated
samples were resuspended in cRPMI with a final cell
density of 2.5 10
5
cells/ml.
Aliquots of cell suspension (3 ml each) were
transferred to one of four quartz cuvettes: one for
autofluorescence measurement (i.e., DMSO-treated
only; cuvette 1) and three for indo-1-loaded cells
(cuvettes 2, 3, and 4). Fluorescence excitation was at
352 nm (6 nm slit) with emission detection at 398 nm
(10 nm slit) and 490 nm (10 nm slit). For each
time point t(j), fluorescence was recorded from each
cuvette at 398 nm (F
398,i
(j)) and 490 nm (F
490,i
(j)),
where irepresented the cuvette number {1, 2, 3, 4}
and jthe time index {1, 2, 3,...,N}. These data were
used to form three ratios by the relation R
k
(j)¼
(F
398,kþ1
(j)F
398,1
(j))/(F
490,kþ1
(j)F
490,1
(j)), where
k¼{1, 2, 3}. The ratios were normalized to form three
356 McCreary et al.
normalized ratios by the relation NR
k
(j)¼
R
k
(j)/R
k
(t(j)¼300), where R
k
(t(j)¼300) was estimated
from linear regression of the ratio data acquired during
first 5 min (indicated as Pre in Fig. 1B). Although
sufficient data was collected to permit calibration of the
ratios into [Ca
2þ
]
c
, averaged results based on values
from the calculated [Ca
2þ
]
c
showed greater variability
than results based on normalized ratios. Therefore,
results have been presented as normalized ratios instead
of [Ca
2þ
]
c
where appropriate. If excessive scatter from
dust particles was observed or the agonist injection
failed to reach the sample directly, for example, it hit the
inside surface of the cuvette, then the data from the
affected sample was excluded from the triplicate
average. Triplicate determinations were obtained in
92% of the experiments. Duplicate averages were
obtained in 8% of the experiments.
Magnetic Field Exposure System
Stray MF artifacts were minimized by a periscopic
optical system attached to the fluorescence spectropho-
tometer. This modification allowed the four samples to
be monitored and isolated from stray MF associated
with electronic equipment. A static MF was generated
by a triaxial mutually orthogonal square Helmholtz coil
system (with diameters of 0.8, 0.9, and 1.0 m). The
static field was monitored with a digital magnetometer
(Model DM 2220, Schonstedt Instrument Company,
Reston, VA, USA) and the input current was adjusted on
each of the three DC power supplies to achieve the
desired static field magnitude and direction. The ELF
MF was produced by a Helmholtz coil (0.208 m
diameter) and a computer controlled waveform gen-
erator (Model 75, Wavetek, San Diego, CA, USA). The
signal from the waveform generator was amplified
(Model 7570, Techron Inc., Elkhart, IN, USA) and then
fed to the coil. The ELF MF exposure system was able
to generate an ELF waveform on top of a DC offset,
which permitted exposure of samples to a collinear
AC þDC MF combination. Details of the apparatus
have been described elsewhere [Carson and Prato, 1996].
Time Course for Each Experiment
Samples were allowed to equilibrate to 37.0 8C for
5 min before fluorescence measurements were started.
Temperature variation within and between experiments
was less than 0.1 8C. During the first 5 min of
fluorescence data collection, that is, t(j)¼0300 s, a
Null MF was applied. The ELF MF exposure started at
t(j)¼300 s, which consisted of either a Null condition
(ambient static fields 0 0.5 mT) or an AC þDC
condition (78.1 0.5 mT static MF superimposed
collinearly with a 100 0.5 mT 60 Hz sinusoidal MF).
The ELF MF was applied for the duration of the
Fig.1. Time course fluorescence data showing raw and derived
data fromcuvettes1 and 2 for a typicalexperiment.Data from cuv-
ettes 3 and 4 are omitted for clarity, but resemble results
from cuvette 2. A: Raw PMT voltage representing the fluores-
cence signal from DMSO-treated Jurkat cells and indo-1-loaded
Jurkat cells. B: Autofluorescence-corrected fluorescence ratio
derived from the data in panel A. Monoclonal antibody (anti-
CD3, 0.67 mg/ml final concentration), digitonin, and EGTA were
added at 1200, 2400, and 30 00 s, respectively. C:Normalized
ratio derived fromdata in panel B. See methods for notation and
analysis.
Real-Time Measurement of [Ca
2þ
]
c
357
experiment. The sequence of Null versus ELF MF
exposure was randomized.
At t¼1200 s, anti-CD3 at the final concentration
of 0.67 mg/ml, was injected simultaneously into each
cuvette in a 50 ml saline vehicle via an automated
injection system. The calcium response was followed
for 20 min (t(j)¼12002400 s). Thereafter, samples
were treated in succession with digitonin (50 mM),
which permeabilized the cell membrane and saturated
indo-1 with Ca
2þ
(maximum fluorescence signal), and
EGTA (13 mM), which chelated Ca
2þ
(minimum
fluorescence signal; Fig. 1A). Throughout each experi-
ment a gas mixture containing 5% CO
2
and 95% air was
blown through a diffuser over the samples to maintain
the pH of the conditioned medium. The data acquis-
ition, agonist injection system, non-magnetic mixing
system, and AC MF generation were computer
controlled.
Statistical Analysis
Before statistical analysis was performed,
the normalized ratios within each experiment
were averaged according to the relation NR(j)¼
(NR
1
(j)þNR
2
(j)þNR
3
(j))/3. Each time course of the
average normalized ratio was then characterized by a
number of descriptive statistics. These included the
slope, intercept and average over the 15 min prior the
introduction of anti-CD3 (basal); the slope, intercept
and average over the 15 min starting 5 min after the
introduction of anti-CD3 (active); and the rate of initial
increase (rise), rate of recovery (recovery) to a new
steady state level, the maximum gain in the normalized
ratio above baseline (peak), and the time to reach the
peak after the introduction of anti-CD3 (as shown in
Fig. 1C).
In the first study (Study 1), cell samples for
experiments conducted on the same day came from the
same parent culture and each combination of exposure
condition and cell cycle distribution was performed on a
single day. Repeated measures multivariate analysis of
variance was used for statistical comparison. The ten
descriptive statistics of the average normalized ratios
were compared within the three cell cycle distributions
and two exposure conditions. The distribution (skew
and kurtosis) and variance of the descriptive statistics
were examined for each group to establish if the
assumptions of normality and variance were met.
Where significant main effects were found, post-hoc
comparisons were performed with a paired t-test
between exposure groups. Multivariate analysis of
variance was used to analyze the descriptive data from
the second study (Study 2). It was also used to compare
the results between Study 1 and 2. Differences
were considered statistically significant at the P.05
level.
RESULTS
Synchronization
The cell distributions within the cell cycle after
aphidicolin treatment were summarized in Table 1. For
S phase enrichment, the number of cells in S phase was
82% compared to 31% in an asynchronous culture. For
experiments with G2-M phases enriched, the propor-
tion of cells in G2-M in comparison to the asynchronous
culture increased from 12% to 50%, whereas for G0/G1
enrichment, the distribution of G0/G1 cells increased
from 56% to 65%. The cell viability was 9397% for up
to 30 h after aphidicolin treatment. The doubling time of
the population was approximately 24 h, similar to
untreated, asynchronous Jurkat E6.1 cultures. There
were no significant differences in cell cycle distribution
between samples that were exposed to the Null or the
AC þDC condition.
Dependence of Descriptive Statistics
on Cell Cycle
The average normalized ratios for G0/G1, S, and
G2-M enriched distributions are shown in Figure 2. The
corresponding descriptive statistics describing the
normalized ratios are given in Table 2. Dependence of
the descriptive statistics on cell cycle distribution was
observed primarily after the introduction of anti-CD3
and was independent of magnetic field condition. The
active slope and intercept were significantly greater in
TABLE 1. Distributions of Synchronized Jurkat E6.1 Cells for G0/G1, S, and G2-M Enriched
Phases (Average SE)
Enriched phase
G0/G1 (%) S (%) G2-M (%)
Null AC þDC Null AC þDC Null AC þDC
G0/G1 (n¼10) 65 2642232222122143
S(n¼10) 9 11018228139192
G2-M (n¼10) 31 2313192202503492
358 McCreary et al.
G0/G1 enriched cultures than in G2-M enriched
cultures (P<.03 for each descriptive statistic). The
active intercept and active average were significantly
greater in G0/G1 enriched cultures than in S enriched
cultures (P<.05 for each descriptive statistic). No
significant interaction between magnetic field condition
and phase of the cell cycle was found.
ELF MF Effects
In Study 1, an effect of ELF MF exposure was
observed in the peak transient response to anti-CD3 in
aphidicolin-treated cells. There was a significant effect
of MF exposure on the peak increase in normalized
ratio above baseline after anti-CD3 [F
(1,9)
¼11.68,
P¼.008], which indicated that the peak [Ca
2þ
]
c
in cells
exposed to AC þDC was greater than the peak [Ca
2þ
]
c
in cells exposed to the Null condition over all cell cycle
distributions. Although, no significant interaction
between cell cycle distribution and magnetic field
exposure was detected, it was decided that investigative
post-hoc analyses should be performed to compare
exposure groups that represented each enriched phase
of the cell cycle. It was determined that the largest
difference in peak normalized ratio between exposure
groups was observed for G2-M enriched samples.
However, the difference was not statistically significant
(P¼.057, Fig. 2C). Likewise, comparison of the
descriptive statistics between the Null and AC þDC
groups representative of each cell cycle distribution
revealed no statistically significant differences
(Table 2).
To further investigate the possibility that [Ca
2þ
]
c
signaling was altered by exposure to an ACþDC MF
during the G2-M phase of the cell cycle, the MF
condition was repeated in Study 2 using a protocol
similar to Study 1. Twenty-seven samples were exposed
to the Null condition and seventeen samples were
exposed to the AC þDC condition. The distribution of
cells within the cell cycle for each MF exposure
condition in Study 2 was similar to the distribution of
cells for G2-M enriched samples in Study 1 (Compare
Table 3 to Table 1). In Study 2, the average normalized
ratios for the Null and AC þDC exposure groups were
similar with significant overlap of the SE at each time
point (Fig. 3). Analysis of the descriptive data revealed
no statistically significant difference between the two
exposure groups (Table 4).
However, when the normalized ratios between
Studies 1 and 2 were compared, differences were
found (Fig. 4). Qualitative differences between the rate
of increase before anti-CD3 and the shape of the
recovery to the steady state level after anti-CD3
between Study 1 and 2 were apparent. Statistical
analysis of the descriptive data validated the qualitative
findings (Table 4). Statistically significant differences
between the Null groups from each study were obtained
for the basal slope, basal intercept, basal average,
time to peak, recovery rate, and active average.
Statistically significant differences between AC þDC
groups from Studies 1 and 2 were also found and
indicated in Table 4.
Fig. 2. The effect of MF exposure on normalized ratios in
synchronized cells from Study1.The average and standard error
ofthe normalizedratioat eachtimepoint wascalculatedforindivid-
ualexperimentalgroupsandplottedasindicatedinthelegend.Null
and AC þDC exposed groups for G 0/G1, S, and G2 -M enriched
samplesareshownin panels A,B,andC,respectively.The number
of experimentsin eachgroupappearsin the legend.
Real-Time Measurement of [Ca
2þ
]
c
359
DISCUSSION
ELF MF Effects
In Study 1, we examined the dependence of
calcium signaling on ELF MF exposure in Jurkat cells
synchronized with aphidicolin. Analysis of the pooled
data revealed a statistically significant increase in the
peak-normalized ratio after anti-CD3 stimulation dur-
ing ELF MF exposure. Post-hoc analysis of ELF MF
experiments with samples enriched to different phases
of the cell cycle indicated that the largest change in
[Ca
2þ
]
c
signaling occurred in G2-M enriched samples.
However, the increase in the peak normalized ratio after
anti-CD3 in G2-M enriched samples failed to achieve
statistical significance. The result suggested that ELF
MF influenced calcium signaling in Jurkat cells, but in a
manner that was only weakly dependent on the phase of
the cell cycle.
It is unlikely that the difference obtained with the
pooled data could be attributed to selection bias or
experimental artifact. In both studies, large numbers of
replicate experiments were done, groups were properly
randomized on each day, and treatment conditions were
balanced between each day of experiments. Addition-
ally, experimental artifact could be ruled out since the
apparatus was thoroughly characterized for artifacts
that could have arisen from vibration, heating effects,
electronic interference, magnetic interference, and
optical issues [Carson and Prato, 1996].
Possible Mechanisms Underlying
ELF MF Effect
It is now well understood in many cell types,
including Jurkat, that calcium signaling involves
calcium release from intracellular calcium stores
followed by capacitative calcium entry via channels in
the plasma membrane [Putney, 2001, 2005]. Given
that the peak normalized ratio after stimulation in ELF
MF-exposed samples was the only descriptive statistic
to be measurably different from Null-exposed samples,
we concluded that ELF MF exposure affected the
calcium-release dependent pathway. Although the
exact molecular target could not be determined
from our data, several candidates were hypothesized,
which were supported by data from other laboratories.
TABLE 2. Descriptive Statistics from Study 1 (Average SE)
Descriptive statistic
G0/G1 S G2-M
Null AC þDC Null AC þDC Null AC þDC
Basal slope (10
4
/s)
b
1.2 0.1 1.2 0.1 0.9 0.2 0.8 0.2 1.0 0.2 1.0 0.1
Basal intercept 0.969 0.004 0.976 0.006 0.982 0.008 0.989 0.004 0.976 0.006 0.969 0.005
Basal average 1.06 0.01 1.06 0.01 1.05 0.01 1.05 0.01 1.05 0.01 1.05 0.01
Rise (10
2
/s) 1.5 0.1 1.5 0.1 1.3 0.1 1.5 0.1 1.2 0.1 1.4 0.1
Peak
a
1.04 0.05 1.07 0.05 0.92 0.06 1.03 0.04 0.88 0.04 1.00 0.05
Time to peak (s) 80.9 4.7 83.7 3.6 86.5 4.4 80.9 3.7 85.1 4.5 88.0 5.9
Recovery (10
3
/s) 4.2 0.4 4.5 0.6 4.3 0.6 3.6 0.5 3.3 0.4 3.8 0.4
Active slope
a
(10
5
/s) 10 210 210 2924272
Active intercept
a,b
2.04 0.06 2.01 0.03 1.83 0.06 1.89 0.08 1.81 0.05 1.89 0.07
Active average
b
1.82 0.03 1.82 0.03 1.65 0.04 1.71 0.05 1.73 0.03 1.75 0.06
a
Post-hoc comparison of samples grouped by cell cycle show a significant difference between G0/G1 and G2-M enriched phases (indicated
by bolded text).
b
Post-hoc comparison of samples grouped by cell cycle show a significant difference between G0/G1 and S enriched phases (indicated by
bolded text).
TABLE 3. Distribution of Synchronized Jurkat E6.1 Cells in
Study 2 (Average SE)
ELF MF exposure % G0-G1 % S % G2-M
Null (n¼27) 27 1191541
AC þDC (n¼17) 26 1191542
Fig. 3. Results from Study 2 for samples of G2- M enrichment.
Each symbolrepresents the normalized ratio obtained by averag-
ing experiments where the exposure condition was similar. The
exposure condition and number of experiments are indicated in
the legend. Error bars represent standard errors of the mean.
360 McCreary et al.
These supporting data included an ELF MF influence
on (i) the binding of anti-CD3 to membrane-
bound receptors [Nindl et al., 2000], (ii) inositol
trisphosphate levels [Korzh-Sleptsova et al., 1995],
and (iii) CD45 phosphatase activity [Lindstrom et al.,
1995a].
Cell Cycle Dependence
We were unable to determine any dependence of
ELF MF modulated calcium signaling on cell cycle.
Our result was consistent with another study where c-
myc transcript levels were measured in G0/G1 enriched
lymphoid cells, but no changes were observed after
exposure to ELF MF [Desjobert et al., 1995]. Other
studies found ELF MF effects on the cell cycle, but only
when the exposure was combined with radiation [Harris
et al., 2002; Tian et al., 2002]. It is largely unknown
how relevantthese reports were to our findings, since it is
unclear if any of the cited reports could be related to
direct or indirect ELF MF-related alterations in [Ca
2þ
]
c
.
Although we could not detect ELF MF-related
changes in [Ca
2þ
]
c
that depend on cell cycle, the basal
and stimulated [Ca
2þ
]
c
response were found to depend
on phase of the cell cycle independent of ELF MF.
Pande et al. [1996] found that [Ca
2þ
]
c
depended on the
phase of the cell cycle with the lowest level at the
beginning of G1, gradually increasing to a maximum at
the G1-S border, then decreasing during S and slightly
increasing again at G2. Although their results provided
plausibility for our findings, our results were not
directly comparable since we only reported relative
changes in fluorescence ratio and not absolute measures
of [Ca
2þ
]
c
. Karas et al. [1999] also measured the
[Ca
2þ
]
c
response to activation of the T-cell receptor
by anti-CD3 monoclonal antibody, OKT-3, in Jurkat
cells with G0/G1, S, and G2-M enriched phases. Phases
of the cell cycle were enriched using counterflow
centrifugal elutriation rather than cell cycle arrest by
chemical treatment. In contrast to our results and those
of Pande and coworkers, they were unable to detect
any cell cycle dependence of the [Ca
2þ
]
c
response.
However, the sample size was low (two independent
samples for each experimental group), which would
make it difficult to detect small changes in a variable
response.
Possible Mechanisms Underlying
[Ca
2þ
]
c
Dynamics
The results from Study 1 indicated the possibility
of a weak dependence of ELF-MF effects on calcium
signaling in G2-M enriched Jurkat cells. We examined
this apparent phenomenon in greater detail in Study 2,
but found no difference between the peak-normalized
ratios after anti-CD3 stimulation from the two exposure
groups. Although this was statistically consistent with
our Study 1 finding, the close proximity to statistical
significance of Study 1 (i.e., P¼.057) was not observed
in Study 2. Therefore, the result of Study 2 might best be
TABLE 4. Descriptive Statistics for G2-M-Enriched Samples
From Study 2 (Average SE)
Descriptive statistic Null (n¼27) AC þDC (n¼17)
Basal slope (10
4
/s) 1.8 0.1
a
1.7 0.1
a
Basal intercept 0.947 0.003
a
0.951 0.006
a
Basal average 1.08 0.01
a
1.08 0.01
a
Rise (10
2
/s) 1.5 0.1 1.6 0.1
Peak 0.91 0.06 0.94 0.05
Time to peak (s) 105 3
a
105 5
Recovery (10
3
/s) 1.1 0.2
a
0.9 0.1
a
Active slope (10
5
/s) 3112
a
Active intercept 1.99 0.06 1.94 0.05
Active average 1.92 0.05
a
1.92 0.04
a
a
Significantly different from same descriptive statistic in Study 1.
Fig. 4. Comparison of the average normalized ratios between
Study1 and Study 2.The average normalized ratios from samples
of G2-M enrichment andexposedto the Nullconditionare shownin
panel A. Similarly, results from samples exposed to the AC þDC
MF are shown in panel B.
Real-Time Measurement of [Ca
2þ
]
c
361
explained by an as yet unknown experimental variable
upon which the ELF MF influence on calcium signaling
depended. One explanation may be related to the
mechanism(s) that underlie the differences in calcium
dynamics between the two studies. Perhaps one or more
of these mechanisms rendered cells insensitive to ELF
MF in Study 2.
Mechanistic explanation of the observed [Ca
2þ
]
c
dynamics include the following possibilities.
(i) The increased rate of [Ca
2þ
]
c
rise in resting
cells from Study 2 compared to Study 1 suggested that
indo-1 was leaking from the cells into the extracellular
environment where Ca
2þ
is typically 10 000-fold more
concentrated and leads to higher fluorescence ratios.
Alternatively, the basal permeability of the plasma
membrane and/or intracellular Ca
2þ
store membranes
may have been higher in cells from Study 2 compared
to Study 1.
(ii) Differences in the response to anti-CD3
stimulation may indicate a subtle difference in the
potency of the anti-CD3 between Studies 1 and 2. This
may have been complicated by differences in CD3
receptor expression, co-stimulation by cytokines in
different batches of fetal bovine serum, and extrac-
ellular calcium concentration between batches of
conditioned medium.
(iii) The [Ca
2þ
]
c
response to stimulation in
individual T-lymphocytes and in suspensions of Jurkat
cells was shown to be highly variable [Wulfing et al.,
1997; McCreary et al., 2002]. The proportion of
individual cells that responded to anti-CD3 with an
initial transient increase followed by a small undershoot
and sustained high [Ca
2þ
]
c
may be reduced in Study 2
compared to Study 1. Possibly, a greater proportion of
the cells in Study 2 had a greater response to the same
concentration of anti-CD3. In Study 2, the difference
between the peak [Ca
2þ
]
c
and the elevated steady state
[Ca
2þ
]
c
after activation was smaller and the undershoot
was not observed (Fig. 4).
(iv) Numerous in vitro studies have suggested that
the effects of weak ELF magnetic fields depend on the
metabolic status of the cells [Walleczek and Liburdy,
1990; Muehsam and Pilla, 1999], particularly activation
state [Reinbold and Pollack, 1997; Campbell-Beachler
et al., 1998; Tuinstra et al., 1998; Richard et al., 2002],
differentiation state [Loschinger et al., 1999], and
growth stage [Nindl et al., 1997; Felaco et al., 1999;
Diniz et al., 2002]. Given that the two studies were
separated by over a year it is not unreasonable to suggest
that slight differences in handling and culture con-
ditions could have existed between the two studies, in
turn leading to the differences in [Ca
2þ
]
c
dynamics
between Studies 1 and 2 by one or more of the above-
mentioned mechanisms.
Implications of the Work
We observed an overall 9% increase in the peak-
normalized ratio in ELF-MF exposed Jurkat cells
over and above the response of Null-exposed cells
after stimulation with anti-CD3. This apparent small
increase in the assay metric could have corresponded to
a 40% increase in [Ca
2þ
]
c
after calibration of the
ratiometric data. If all cells in the cuvette responded
with the same magnitude, then it remained debatable
whether such a small change could lead to downstream
biological consequences. However, if only a small
subpopulation of cells responded with a large change in
[Ca
2þ
]
c
with the balance of cells unresponsive to ELF
MF, then large downstream biological effects might be
expected in the subpopulation. At least one report lends
some support to this possibility in Jurkat cells
[Lindstrom et al., 1993].
Future Work
The results from this work led us to suggest
several avenues for future study. First, it would be
worthwhile to explore the reasons for the differences in
[Ca
2þ
]
c
dynamics between the two studies. Specifically,
the roles of anti-CD3 concentration and extracellular
Ca
2þ
should be studied systematically for both Null and
ELF MF exposure conditions. Second, although we
chose ELF MF conditions that followed the predictions
of a theoretical model, the applicability of the model is
still unclear and stronger ELF MF fields might elicit
larger, more reliable changes in [Ca
2þ
]
c
signals after
anti-CD3. Last, ratiometric fluorescence microscopy of
the [Ca
2þ
]
c
response to anti-CD3 in individual Jurkat
cells might reveal the existence of a small population of
cells that are strongly affected by ELF MF.
ACKNOWLEDGMENTS
The authors thank Mr. Mike Keeney and
Ms. Wendy Brown for assistance with the flow
cytometry measurements and analysis; Mr. Larry Stitt,
Drs. Yves Bureau and Alex Thomas for advice on the
statistical analysis of the data; Mr. Lynn Keenliside for
technical assistance with the fluorescence spectropho-
tometer; and Dr. Michelle Belton for assistance with the
manuscript preparation.
REFERENCES
Baureus Koch CL, Sommarin M, Persson BR, Salford LG,
Eberhardt JL. 2003. Interaction between weak low frequency
magnetic fields and cell membranes. Bioelectromagnetics
24(6):395–402.
Blackman CF, Blanchard JP, Benane SG, House DE. 1995.
The ion parametric resonance model predicts magnetic
362 McCreary et al.
field parameters that affect nerve cells. Faseb J 9(7):547
551.
Blanchard JP, Blackman CF. 1994. Clarification and application of
an ion parametric resonance model for magnetic field
interactions with biological systems. Bioelectromagnetics
15(3):217– 238.
Campbell-Beachler M, Ishida-Jones T, Haggren W, Phillips JL.
1998. Effect of 60 Hz magnetic field exposure on c-fos
expression in stimulated PC12 cells. Mol Cell Biochem
189(1– 2):107 111.
Carson JJL, Prato FS. 1996. A fluorescence spectrophotometer for
the real time detection of cytosolic free calcium from cell
suspensions during exposure to extremely low frequency
magnetic fields. Rev Sci Instrum 67:4336– 4346.
Carson JJ, Prato FS, Drost DJ, Diesbourg LD, Dixon SJ. 1990. Time-
varying magnetic fields increase cytosolic free Ca2þin HL-
60 cells. Am J Physiol 259(4 Pt 1):C687C692.
Craviso GL, Poss J, Lanctot C, Lundback SS, Chatterjee I,
Publicover NG. 2002. Intracellular calcium activity in
isolated bovine adrenal chromaffin cells in the presence and
absence of 60 Hz magnetic fields. Bioelectromagnetics
23(8):557– 567.
Desjobert H, Hillion J, Adolphe M, Averlant G, Nafziger J. 1995.
Effects of 50 Hz magnetic fields on C-myc transcript levels in
nonsynchronized and synchronized human cells. Bioelec-
tromagnetics 16(5):277– 283.
Diniz P, Shomura K, Soejima K, Ito G. 2002. Effects of pulsed
electromagnetic field (PEMF) stimulation on bone tissue like
formation are dependent on the maturation stages of the
osteoblasts. Bioelectromagnetics 23(5):398–405.
Fanelli C, Coppola S, Barone R, Colussi C, Gualandi G, Volpe P,
Ghibelli L. 1999. Magnetic fields increase cell survival by
inhibiting apoptosis via modulation of Ca2þinflux. Faseb J
13(1):95– 102.
Felaco M, Reale M, Grilli A, De Lutiis MA, Barbacane RC, Di Luzio
S, Conti P. 1999. Impact of extremely low frequency electro-
magnetic fields on CD4 expression in peripheral blood
mononuclear cells. Mol Cell Biochem 201(12):49– 55.
Galvanovskis J, Sandblom J, Bergqvist B, Galt S, Hamnerius Y.
1999. Cytoplasmic Ca
2þ
oscillations in human leukemia T-
cells are reduced by 50 Hz magnetic fields. Bioelectromag-
netics 20(5):269–276.
Gobba F, Malagoli D, Ottaviani E. 2003. Effects of 50 Hz magnetic
fields on fMLP-induced shape changes in invertebrate
immunocytes: The role of calcium ion channels. Bioelec-
tromagnetics 24(4):277– 282.
Harris PA, Lamb J, Heaton B, Wheatley DN. 2002. Possible
attenuation of the G2 DNA damage cell cycle checkpoint in
HeLa cells by extremely low frequency (ELF) electro-
magnetic fields. Cancer Cell Int 2(1):3.
Kai Y, Miyako K, Muta T, Umeda S, Irie T, Hamasaki N, Takeshige
K, Kang D. 1999. Mitochondrial DNA replication in human T
lymphocytes is regulated primarily at the H-strand termi-
nation site. Biochim Biophys Acta 1446(12):126– 134.
Karabakhtsian R, Broude N, Shalts N, Kochlatyi S, Goodman R,
Henderson AS. 1994. Calcium is necessary in the cell
response to EM fields. FEBS Lett 349(1):1–6.
Karas M, Zaks TZ, Liu JL, LeRoith D. 1999. T cell receptor-induced
activation and apoptosis in cycling human T cells occur
throughout the cell cycle. Mol Biol Cell 10(12):4441–4450.
Korzh-Sleptsova IL, Lindstrom E, Mild KH, Berglund A, Lundgren
E. 1995. Low frequency MFs increased inositol 1,4,5-
trisphosphate levels in the Jurkat cell line. FEBS Lett
359(2– 3):151 154.
Lacy-Hulbert A, Metcalfe JC, Hesketh R. 1998. Biological
responses to electromagnetic fields. Faseb J 12(6):395– 420.
Lednev VV. 1991. Possible mechanism for the influence of weak
magnetic fields on biological systems. Bioelectromagnetics
12(2):71–75.
Lindstrom E, Lindstrom P, Berglund A, Mild KH, Lundgren E.
1993. Intracellular calcium oscillations induced in a T-cell
line by a weak 50 Hz magnetic field. J Cell Physiol 156(2):
395–398.
Lindstrom E, Berglund A, Mild KH, Lindstrom P, Lundgren E.
1995a. CD45 phosphatase in Jurkat cells is necessary for
response to applied ELF magnetic fields. FEBS Lett 370
(1–2):118 122.
Lindstrom E, Lindstrom P, Berglund A, Lundgren E, Mild KH.
1995b. Intracellular calcium oscillations in a T-cell line after
exposure to extremely-low-frequency magnetic fields with
variable frequencies and flux densities. Bioelectromagnetics
16(1):41–47.
Loscher W, Liburdy RP. 1998. Animal and cellular studies on
carcinogenic effects of low frequency (50/60-Hz) magnetic
fields. Mutat Res 410(2):185220.
Loschinger M, Thumm S, Hammerle H, Rodemann HP. 1999.
Induction of intracellular calcium oscillations in human skin
fibroblast populations by sinusoidal extremely low-fre-
quency magnetic fields (20 Hz, 8 mT) is dependent on the
differentiation state of the single cell. Radiat Res 151(2):
195–200.
Lyle DB, Fuchs TA, Casamento JP, Davis CC, Swicord ML. 1997.
Intracellular calcium signaling by Jurkat T-lymphocytes
exposed to a 60 Hz magnetic field. Bioelectromagnetics
18(6):439–445.
Madec F, Billaudel B, Charlet de Sauvage R, Sartor P, Veyret B.
2003. Effects of ELF and static magnetic fields on calcium
oscillations in islets of Langerhans. Bioelectrochemistry
60(1– 2):73 80.
McCreary CR, Thomas AW, Prato FS. 2002. Factors confounding
cytosolic calcium measurements in Jurkat E6.1 cells during
exposure to ELF magnetic fields. Bioelectromagnetics
23(4):315–328.
Merrill GF. 1998. Cell synchronization. Methods Cell Biol 57:229–
249.
Morgado-Valle C, Verdugo-Diaz L, Garcia DE, Morales-Orozco C,
Drucker-Colin R. 1998. The role of voltage-gated Ca2þ
channels in neurite growth of cultured chromaffin cells
induced by extremely low frequency (ELF) magnetic field
stimulation. Cell Tissue Res 291(2):217– 230.
Muehsam DJ, Pilla AA. 1999. The sensitivity of cells and tissues to
exogenous fields: Effects of target system initial state.
Bioelectrochem Bioenerg 48(1):35–42.
Nindl G, Swez JA, Miller JM, Balcavage WX. 1997. Growth stage
dependent effects of electromagnetic fields on DNA syn-
thesis of Jurkat cells. FEBS Lett 414(3):501506.
Nindl G, Balcavage WX, Vesper DN, Swez JA, Wetzel BJ,
Chamberlain JK, Fox MT. 2000. Experiments showing that
electromagnetic fields can be used to treat inflammatory
diseases. Biomed Sci Instrum 36:7–13.
Pande G, Kumar NA, Manogaran PS. 1996. Flow cytometric study
of changes in the intracellular free calcium during the cell
cycle. Cytometry 24(1):55–63.
Prato FS, Carson JJ, Ossenkopp KP, Kavaliers M. 1995. Possible
mechanisms by which extremely low frequency magnetic
fields affect opioid function. Faseb J 9(9):807–814.
Prato FS, Kavaliers M, Thomas AW. 2000. Extremely low frequency
magnetic fields can either increase or decrease analgaesia in
Real-Time Measurement of [Ca
2þ
]
c
363
the land snail depending on field and light conditions.
Bioelectromagnetics 21(4):287– 301.
Putney JW, Jr. 2001. Pharmacology of capacitative calcium entry.
Mol Interv 1(2):84–94.
Putney JW, Jr. 2005. Capacitative calcium entry: Sensing the
calcium stores. J Cell Biol 169(3):381382.
Reinbold KA, Pollack SR. 1997. Serum plays a critical role in
modulating [Ca2þ]c of primary culture bone cells exposed to
weak ion-resonance magnetic fields. Bioelectromagnetics
18(3):203– 214.
Richard D, Lange S, Viergutz T, Kriehuber R, Weiss DG, Myrtill S.
2002. Influence of 50 Hz electromagnetic fields in combina-
tion with a tumour promoting phorbol ester on protein kinase
C and cell cycle in human cells. Mol Cell Biochem 232(1
2):133–141.
Sarimov R, Markova E, Johansson F, Jenssen D, Belyaev I. 2005.
Exposure to ELF magnetic field tuned to Zn inhibits growth
of cancer cells. Bioelectromagnetics 26(8):631638.
Shahidain R, Mullins RD, Sisken JE. 2001. Calcium spiking activity
and baseline calcium levels in ROS 17/2.8 cells exposed to
extremely low frequency electromagnetic fields (ELF EMF).
Int J Radiat Biol 77(2):241–248.
Sisken JE, DeRemer D. 2000. Power-frequency electromagnetic
fields and the capacitative calcium entry system in SV40-
transformed Swiss 3T3 cells. Radiat Res 153(5 Pt 2):699
705.
Tian F, Nakahara T, Yoshida M, Honda N, Hirose H, Miyakoshi J.
2002. Exposure to power frequency magnetic fields sup-
presses X-ray-induced apoptosis transiently in Ku80-defi-
cient xrs5 cells. Biochem Biophys Res Commun 292(2):
355–361.
Tuinstra R, Goodman E, Greenebaum B. 1998. Protein kinase C
activity following exposure to magnetic field and phorbol
ester. Bioelectromagnetics 19(8):469– 476.
Walleczek J, Budinger TF. 1992. Pulsed magnetic field
effects on calcium signaling in lymphocytes: Dependence
on cell status and field intensity. FEBS Lett 314(3):351–
355.
Walleczek J, Liburdy RP. 1990. Nonthermal 60 Hz sinusoidal
magnetic-field exposure enhances 45Ca2þuptake in rat
thymocytes: Dependence on mitogen activation. FEBS Lett
271(1–2):157 160.
Wey HE, Conover DP, Mathias P, Toraason M, Lotz WG. 2000. 50-
Hertz magnetic field and calcium transients in Jurkat cells:
Results of a research and public information dissemination
(RAPID) program study. Environ Health Perspect 108(2):
135–140.
Wulfing C, Rabinowitz JD, Beeson C, Sjaastad MD, McConnell
HM, Davis MM. 1997. Kinetics and extent of T cell
activation as measured with the calcium signal. J Exp Med
185(10):1815–1825.
Zhou J, Yao G, Zhang J, Chang Z. 2002. CREB DNA binding
activation by a 50-Hz magnetic field in HL60 cells is
dependent on extra- and intracellular Ca(2þ) but not PKA,
PKC, ERK, or p38 MAPK. Biochem Biophys Res Commun
296(4):1013–1018.
364 McCreary et al.
... The common denominator of these studies is modulation of calcium homeostasis by LF MF. Cells from primary cultures or permanent cell lines (Simkó and Mattsson, 2004;McCreary et al., 2006;Carson et al., 1990;Conti et al., 1985a) are studied to explain or predict the mechanistic aspects of the observed interactions (Pilla et al., 2011;Pall, 2013;Gartzke and Lange, 2002). For example, it has been suggested that the cationic nature of the calcium ion might make it susceptible to the induced electric fields (IEF) generated by LF MF in solution (Gartzke and Lange, 2002;Lednev, 1991). ...
... However, if outcomes were determined at different time points using separate independent samples, all time points were included. When the intracellular calcium concentration of resting cells, and subsequently the concentration during stimulation of a calcium influx, was measured under sham and LF MF conditions, both outcomes were noted (McCreary et al., 2006;Luo et al., 2014). From these double datasets, only data from the stimulated sample were used for the overall analysis. ...
... From these, 1490 articles could already be excluded based on title and abstract, since they did not describe the exposure of cells to magnetic fields. Further investigation of the 227 remaining papers based on full text led to inclusion of 42 studies (McCreary et al., 2006;Conti et al., 1985a;Liburdy, 1992;Galvanovskis et al., 1996;Walleczek and Liburdy, 1990;Lisi et al., 2006;Pilger et al., 2004;Sakurai et al., 2005;Liu et al., 2014;Tonini et al., 2001;Garciasancho et al., 1994;Lee et al., 2002;Hwang et al., 2011;Lindstrom et al., 1995;Nishimura et al., 1999;Grande et al., 1991;Oh et al., 2001;Conti et al., 1985b;Luo et al., 2014;Mattsson et al., 2001;Wey et al., 2000;Lindstrom et al., 1998;Craviso et al., 2003;Fitzsimmons et al., 1994;Fixler et al., 2012;Gaetani et al., 2009;Bernabo et al., 2007;Morabito et al., 2010a;Liburdy et al., 1993;Kim et al., 2013;Piacentini et al., 2008;Loschinger et al., 1999;Yamaguchi et al., 2002;Craviso et al., 2002;Lyle et al., 1997;Coulton and Barker, 1993;Morabito et al., 2010b;McCreary et al., 2002;de Groot et al., 2014;Wei et al., 2014;Wu et al., 2014;Lyle et al., 1991). ...
Article
Low frequency magnetic field (LF MF) exposure is recurrently suggested to have the ability to induce health effects in society. Therefore, in vitro model systems are used to investigate biological effects of exposure. LF MF induced changes of the cellular calcium homeostasis are frequently hypothesised to be the possible target, but this hypothesis is both substantiated and rejected by numerous studies in literature. Despite the large amount of data, no systematic analysis of in vitro studies has been conducted to address the strength of evidence for an association between LF MF exposure and calcium homeostasis. Our systematic review, with inclusion of 42 studies, showed evidence for an association of LF MF with internal calcium concentrations and calcium oscillation patterns. The oscillation frequency increased, while the amplitude and the percentage of oscillating cells remained constant. The intracellular calcium concentration increased (SMD 0.351, 95% CI 0.126, 0.576). Subgroup analysis revealed heterogeneous effects associated with the exposure frequency, magnetic flux density and duration. Moreover, we found support for the presence of MF-sensitive cell types. Nevertheless, some of the included studies may introduce a great risk of bias as a result of uncontrolled or not reported exposure conditions, temperature ranges and ambient fields. In addition, mathematical calculations of the parasitic induced electric fields (IEFs) disclosed their association with increased intracellular calcium. Our results demonstrate that LF MF might influence the calcium homeostasis in cells in vitro, but the risk of bias and high heterogeneity (I2 > 75%) weakens the analyses. Therefore any potential clinical implications await further investigation.
... Additionally, satisfying results were obtained in chronic patients as most of them was able to interrupt periodic infiltration therapy thanks to the possibility of managing pain and improving articular motion with a limited number of effective HIRO TT treatment sessions. This is the first report on clinical experience with HIRO TT and the result of this study are consistent with Hilterapia ® safety and clinical outcome in pain treatment related to neuromuscular and osteoarticular conditions (6)(7)(8)(9)(10)(11)(12)(13). In summary, the thermal gradients, created amongst tissues at different depths after the alternating application of SmartCooler and Hilterapia ® , generate a thermal exchange that: emphasises stimulation of the microcirculation function and lymphatic drainage, resulting in edema resorption, enhances reactivation of the biological repair processes, restores tissue homeostasis, prolongs muscle relaxation effect, speeds up functional recovery. ...
... A lot of assumptions have been made to explain how physical stimulus is converted to chemical signal, but the transduction pathway by which EMF acts on cells, has not been elucidated. Calcium signaling plays an important role in regulating proliferation, differentiation and apoptosis and it was reported that ELF EMFs can alter the levels of cytosolic Ca2+ [13], although this effect seems to be specific to the cell type and the EMFs parameters applied. Another important molecular response to ELF EMFs is represented by an increasing level of intracellular reactive oxygen species (ROS) in cells exposed to EMFs [14]. ...
Article
Full-text available
In the last decades, electromagnetotherapy generated an intense interest for the medical treatment of some pathological states related to the musculoskeletal system. In particular, extremely low frequency (ELF) electromagnetic fields (EMFs) are used to improve tissue regeneration in bone non- union fractures, to facilitate skin wound healing and to reduce pain symptomatology. This therapy represents a valid and non- invasive approach widely used to treat the area of interest limiting the adverse effects related to drug administration. The molecular mechanisms by which ELF EMFs act on cell behavior is still not completely known, but numerous and heterogeneous effects have been observed on a very large number of biological processes. These effects vary in relation to the treatment parameters and intrinsic susceptibility of specific cell lines. In order to study the molecular mechanism by which ELF EMFs act on fibroblasts, the mouse-derived NIH3T3 cell line was chosen as the experimental model to carry out some biochemical investigations. After EMF exposure, the cells showed an increased level of ROS and a decreasing activity of the enzyme pyruvate kinase (PK), leading to a slowdown of the glycolytic flux and a redirection of glycolytic metabolites towards the pentose phosphate pathway (PPP). Hence, the results of the study define a coherent biochemical mechanism by which ELF EMFs are able to promote a shift of cell metabolism from catabolic to anabolic processes. Additional investigations such as the evaluation of reduced glutathione levels, are however necessary to confirm the mechanism.
... However, our primary data from real-time LF EMF exposure of neutrophil-like cells in a flow cytometric setup did not show any indications for LF EMF modulation of the calcium influx. Furthermore, the potential sensitivity to LF EMFs may be influenced by cell cycle [McCreary et al., 2006], cell activation [McCreary et al., 2006;Hwang et al., 2011], cell morphology Fig. 6. mRNA expression of calcium influx genes after five days exposed to LF EMFs. PLB-985 cells were exposed for a period of five days during neutrophilic differentiation to (a) 50 Hz sine LF EMF of 500 mT (stripped bars) and sham (white bars) or (b) 300 mT Immunent LF EMF exposure (black bars) and sham (white bars). ...
... However, our primary data from real-time LF EMF exposure of neutrophil-like cells in a flow cytometric setup did not show any indications for LF EMF modulation of the calcium influx. Furthermore, the potential sensitivity to LF EMFs may be influenced by cell cycle [McCreary et al., 2006], cell activation [McCreary et al., 2006;Hwang et al., 2011], cell morphology Fig. 6. mRNA expression of calcium influx genes after five days exposed to LF EMFs. PLB-985 cells were exposed for a period of five days during neutrophilic differentiation to (a) 50 Hz sine LF EMF of 500 mT (stripped bars) and sham (white bars) or (b) 300 mT Immunent LF EMF exposure (black bars) and sham (white bars). ...
Article
We are increasingly exposed to low-frequency electromagnetic fields (LF EMFs) by electrical devices and power lines, but if and how these fields interact with living cells remains a matter of debate. This study aimed to investigate the potential effect of LF EMF exposure on calcium signalling in neutrophils. In neutrophilic granulocytes, activation of G-protein coupled receptors leads to efflux of calcium from calcium stores and influx of extracellular calcium via specialised calcium channels. The cytoplasmic rise of calcium induces cytoskeleton rearrangements, modified gene expression patterns, and cell migration. If LF EMF modulates intracellular calcium signalling, this will influence cellular behaviour and may eventually lead to health problems. We found that calcium mobilisation upon chemotactic stimulation was not altered after a short 30 min or long-term LF EMF exposure in human neutrophil-like cell lines HL-60 or PLB-985. Neither of the two investigated wave forms (Immunent and 50 Hz sine wave) at three magnetic flux densities (5 μT, 300 μT, and 500 μT) altered calcium signalling in vitro. Gene-expression patterns of calcium-signalling related genes also did not show any significant changes after exposure. Furthermore, analysis of the phenotypical appearance of microvilli by scanning electron microscopy revealed no alterations induced by LF EMF exposure. The findings above indicate that exposure to 50 Hz sinusoidal or Immunent LF EMF will not affect calcium signalling in neutrophils in vitro. Bioelectromagnetics. 2015;9999:XX-XX. © 2015 Wiley Periodicals, Inc. © 2015 Wiley Periodicals, Inc.
... Exposure to ELF-EMF can also modify the biophysical properties of cell membranes, including their permeability to Ca 2+ ions (21). Exposure to ELF-EMF reportedly modifies intra cellular Ca 2+ levels in rat thymic lymphocytes, human T-lymphocytes, and Jurkat cells (22)(23)(24). Depending on the dose (field induction and frequency) and duration of treatment, and the type of inflamed tissue, exposure to EMF can be harmful or may induce a cytoprotective cellular response (25). ...
Article
Full-text available
Background: Activation of immunologically competent cells results in the overproduction of pro-inflammatory factors, and causes progression of nerve tissue damage. However, the potential neuroprotective effects of these factors in brain damage have not been well investigated. Objective: To evaluate the effect of extremely low frequency electromagnetic field (ELF-EMF) treatment on the molecular mechanism of inflammatory cytokine activity in post-stroke patients. Methods: All patients underwent the same programme of physical therapy, but the ELF-EMF group were also given ELF-EMF treatment. In order to determine the plasma level of cytokines, the levels of interleukin 1β (IL-1β), interleukin 2 (IL-2), interferon γ (INFγ) and transforming growth factor β (TGF-β) were evaluated, and the level of IL-1β mRNA expression was determined. Results: After ELF-EMF treatment, both IL-1β plasma level and IL-1β mRNA expression level, as well as IL-2 plasma level increased, while IFN-γ and TGF-β levels did not change. Conclusion: The increased expression of IL-1β found in this study may be a response to ELF-EMF stimulation. It is hypothesized that a neuroprotective role of this cytokine may occur due to IL-1β-dependent regulation of neurotrophic factors. Further research is needed to explore this hypothesis.
... [13]. There are other studies that investigated the possible hazardous effects of ELF -EMFs on cytosolic calcium concentration which plays substantial role as a ubiquitous second messenger in several key biochemical pathways and is thought to be an important mechanism for controlling and synchronizing physiological responses [14]. Effects of electromagnetic radiation exposure on bone mineral density and oxidative stress index in electrical workers were studied [15,16]. ...
... The biological effects of electromagnetic fields, especially the extremely low frequency fields, have been studied for more than fifty years and a huge amount of evidence has accumulated regarding the possible effects of ELF-EMF on living system 9 , including cancer 50-52 , immune cells 53,54 , bone cells 55 , and nerve cells 30,56,57 . However, there is still no general agreement on the relevant underlying mechanisms. ...
Article
Full-text available
Accumulating evidence suggests significant biological effects caused by extremely low frequency electromagnetic fields (ELF-EMF). Although exo-endocytosis plays crucial physical and biological roles in neuronal communication, studies on how ELF-EMF regulates this process are scarce. By directly measuring calcium currents and membrane capacitance at a large mammalian central nervous synapse, the calyx of Held, we report for the first time that ELF-EMF critically affects synaptic transmission and plasticity. Exposure to ELF-EMF for 8 to 10 days dramatically increases the calcium influx upon stimulation and facilitates all forms of vesicle endocytosis, including slow and rapid endocytosis, endocytosis overshoot and bulk endocytosis, but does not affect the RRP size and exocytosis. Exposure to ELF-EMF also potentiates PTP, a form of short-term plasticity, increasing its peak amplitude without impacting its time course. We further investigated the underlying mechanisms and found that calcium channel expression, including the P/Q, N, and R subtypes, at the presynaptic nerve terminal was enhanced, accounting for the increased calcium influx upon stimulation. Thus, we conclude that exposure to ELF-EMF facilitates vesicle endocytosis and synaptic plasticity in a calcium-dependent manner by increasing calcium channel expression at the nerve terminal.
... Changes in intracellular Ca 2þ in response to magnetic fields have been demonstrated in a range of cells [18,66,67]. We show here for the first time that in neurons LI-rMS releases Ca 2þ from intracellular stores. ...
Article
Full-text available
Background: Repetitive transcranial magnetic stimulation is increasingly used as a treatment for neurological dysfunction. Therapeutic effects have been reported for low intensity rTMS (LI-rTMS) although these remain poorly understood. Objective: Our study describes for the first time a systematic comparison of the cellular and molecular changes in neurons in vitro induced by low intensity magnetic stimulation at different frequencies. Methods: We applied 5 different low intensity repetitive magnetic stimulation (LI-rMS) protocols to neuron-enriched primary cortical cultures for 4 days and assessed survival, and morphological and biochemical change. Results: We show pattern-specific effects of LI-rMS: simple frequency pulse trains (10 Hz and 100 Hz) impaired cell survival, while more complex stimulation patterns (theta-burst and a biomimetic frequency) did not. Moreover, only 1 Hz stimulation modified neuronal morphology, inhibiting neurite outgrowth. To understand mechanisms underlying these differential effects, we measured intracellular calcium concentration during LI-rMS and subsequent changes in gene expression. All LI-rMS frequencies increased intracellular calcium, but rather than influx from the extracellular milieu typical of depolarization, all frequencies induced calcium release from neuronal intracellular stores. Furthermore, we observed pattern-specific changes in expression of genes related to apoptosis and neurite outgrowth, consistent with our morphological data on cell survival and neurite branching. Conclusions: Thus, in addition to the known effects on cortical excitability and synaptic plasticity, our data demonstrate that LI-rMS can change the survival and structural complexity of neurons. These findings provide a cellular and molecular framework for understanding what low intensity magnetic stimulation may contribute to human rTMS outcomes.
Article
Full-text available
The geomagnetic field plays an important role in the existence of life on Earth. The study of the biological effects of (hypomagnetic conditions) HMC is an important task in magnetobiology. The fundamental importance is expanding and clarifying knowledge about the mechanisms of magnetic field interaction with living systems. The applied significance is improving the training of astronauts for long-term space expeditions. This review describes the effects of HMC on animals and plants, manifested at the cellular and organismal levels. General information is given about the probable mechanisms of HMC and geomagnetic field action on living systems. The main experimental approaches are described. We attempted to systematize quantitative data from various studies and identify general dependencies of the magnetobiology effects’ value on HMC characteristics (induction, exposure duration) and the biological parameter under study. The most pronounced effects were found at the cellular level compared to the organismal level. Gene expression and protein activity appeared to be the most sensitive to HMC among the molecular cellular processes. The nervous system was found to be the most sensitive in the case of the organism level. The review may be of interest to biologists, physicians, physicists, and specialists in interdisciplinary fields.
Article
The telegraph plant (Codariocalyx motorius) has drawn much interest among plant physiologists because of its peculiar movements of the leaflets. While the terminal leaflets move from a horizontal position during the day and downward during the night, the lateral leaflets display rhythmic up and down movements in the minute range. The period length of the lateral leaflets is temperature dependent, while that of the terminal leaflet is temperature compensated. The movements of both the leaflets are regulated in the pulvini, a flexible organ between the leaflets and the stalk. Electrophysiological recordings using microelectrodes have revealed the physiological mechanisms underlying the leaflet movements. Early experiments related to effect of mechanical load, light, electric and magnetic fields on the leaflet oscillations by the Indian physicist Bose, and followed up by others, are presented. Experimental approaches are discussed and indicate, that Ca2+, various membrane channels, electric and osmotic mechanisms participate in the oscillating system. Modelling the pulvinus tissue would certainly aid in understanding the signal transduction during the movements. New approaches of modelling the mechanisms could further help in understanding the oscillations in the leaflet movements. Such oscillations might be of much broader relevance than known so far, although not as conspicuous as in the leaflet movements. © 2012 Springer-Verlag Berlin Heidelberg. All rights are reserved.
Article
Full-text available
Although extremely low frequency (ELF, <300 Hz) magnetic fields exert a variety of biological effects, the magnetic field sensing/transduction mechanism (or mechanisms) remain to be identified. Using the well-defined inhibitory effects that magnetic fields have on opioid peptide mediated antinociception or "analgesial' in the land snail Cepaea nemoralis, we show that these actions only occur for certain frequency and amplitude combinations of time-varying sinusoidal magnetic fields in a manner consistent with a direct influence of these fields. We exposed snails with augmented opioid activity to ELF magnetic fields, which were varied in both amplitude and frequency, along with a parallel static magnetic field, When the peak amplitude (0-547 mu T) of a magnetic field of 60 Hz was varied systematically, we observed a nonlinear response, i.e., a nonlinear reduction in analgesia as measured by the latency of a defined response by the snails to a thermal stimulus. When frequency (10-240 Hz) was varied, keeping the amplitude constant (141 mu T), we saw significant inhibitory effects between 30 and 35 Hz, 60 and 90 Hz and at 120 and 240 Hz. Finally, when the static field was varied but the amplitude and frequency of the time-varying field were held constant, we observed significant inhibition at almost all amplitudes. This amplitude/frequency "resonance-like" dependence of the magnetic field effects suggests that the mechanism (or mechanisms) of response to weak ELF fields likely involves a direct magnetic field detection mechanism rather than an induced current phenomenon. We examined the implications of our findings for several models proposed for the direct sensing of ELF magnetic fields.
Article
Full-text available
An ion parametric resonance (IPR) model recently developed by Blanchard and Blackman predicts distinct magnetic field interactions with biological systems based on a selective relation among four factors: the flux density of the static magnetic field, the frequency and flux density (Bac) of the parallel ac magnetic field, and the charge-to-mass ratio of ions of biological relevance. To test this model, PC-12 cells stimulated by nerve growth factor to produce neurites were exposed for 23 h in a 5% CO2 incubator using a multiple-coil exposure system to produce 45 Hz ac and dc (366 mG parallel to ac; less than 2 mG perpendicular to ac) magnetic fields. Our earlier work showed a cycle of inhibition/no inhibition of neurite outgrowth consistent with the IPR model predictions for Bac exposures between 0 and 468 mG rms. The work described here tests neurite outgrowth over a broader range of Bac (233-1416 mG rms). The experimental results remain consistent with earlier results, and with IPR model predictions of a second cycle of inhibition, return to control values, followed by a third cycle of inhibition of neurite outgrowth. These responses support the fundamental relationships predicted by the IPR model. The results have broad significance for biology.
Article
Electromagnetic fields have been reported to cause a variety of biological effects. It has been hypothesized that many of these phenomena are mediated by a primary effect on the concentration of cytosolic free calcium ((Ca2+)i). We investigated the effects of exposure to electromagnetic fields on (Ca2+)i in HL-60 cells using the Ca2(+)-sensitive fluorescent indicator indo-1. Indo-1-loaded cell samples were exposed to a radiofrequency electromagnetic field, a static magnetic field, and a time-varying magnetic field, which were generated by a magnetic resonance imaging (MRI) unit. We found that a 23-min exposure to all three fields, in combination, induced a significant increase in (Ca2+)i of 31 +/- 8 (SE) nM (P less than 0.01, n = 13) from a basal level of 121 +/- 8 nM. Also, cells exposed to only the time-varying magnetic field had a mean (Ca2+)i that was 34 +/- 10 nM (P less than 0.01, n = 11) higher than parallel control samples. Separate exposure to the radio-frequency (6.25 MHz) or static field (0.15 T) had no detectable effects. These results demonstrate that time-varying magnetic fields alter (Ca2+)i and suggest that at least some of the reported biological effects of time-varying magnetic fields may arise from elevation of (Ca2+)i.
Article
This chapter describes the methods of analyzing cell-cycle kinetics, methods for synchronizing mammalian cells, and caveats in interpreting cell synchrony experiments. Many cell-synchronization protocols involve placing cells under growth inhibitory conditions for periods of time related to the duration of specific cell-cycle phases. The synchronization methods include release from GO arrest, release from M- and S-phase blocking agents, mitotic detachment, and centrifugal elutriation. To distinguish between growth-dependent and cell cycle-dependent regulation, cells are generally synchronized using more than one method. In some cases, the use of two synchronization methods reveals that all or part of an observed biochemical change is not cell-cycle dependent. The chapter also describes methods for analyzing the cell-cycle kinetics of asynchronous mammalian cells. The asynchronous cell methods include the determination of mean generation time and proliferative fraction, the determination of mitotic and [3H]thymidine-labeling indices, and the estimation of cell-cycle phase durations by the labeled mitoses procedure and by flow cytometry.
Article
Results of prior investigations with opioid peptide mediated antinociception or analgaesia have suggested that these extremely low frequency (ELF) magnetic field effects are described by a resonance mechanism rather than mechanisms based on either induced currents or magnetite. Here we show that ELF magnetic fields (141–414 μT peak) can, in a manner consistent with the predictions of Lednev's parametric resonance model (PRM) for the calcium ion, either (i) reduce, (ii) have no effect on, or (iii) increase endogenous opioid mediated analgaesia in the land snail, Cepaea nemoralis. When the magnetic fields were set to parameters for the predictions of the PRM for the potassium ion, opioid-peptide mediated analgaesia increased and there was evidence of antagonism by the K+ channel blocker, glibenclamide. Furthermore, these effects were dependent on the presence of light; the effects were absent in the absence of light. These observed increases and decreases in opioid analgaesia are largely consistent with the predictions of Lednev's PRM. Bioelectromagnetics 21:287–301, 2000. © 2000 Wiley-Liss, Inc.
Article
The ion Ca2+ has been shown to play an important role in a wide variety of cellular functions, one of them being related to cell differentiation in which nerve growth factor (NGF) is involved. Chromaffin cells obtained from adrenals of 2- to 3-day-old rats were cultured for 7 days. During this time, these cells were subjected to the application of either NGF or extremely low frequency magnetic fields (ELF MF). Since this induced cell differentiation toward neuronal-like cells, the mechanism by which this occurred was studied. When the L-Ca2+ channel blocker nifedipine was applied simultaneously with ELF MF, this differentiation did not take place, but it did when an N-Ca2+ channel blocker was used. In contrast, none of the Ca2+ channel blockers prevented differentiation in the presence of NGF. In addition, Bay K-8644, an L-Ca2+ channel agonist, increased both the percentage of differentiated cells and neurite length in the presence of ELF MF. This effect was much weaker in the presence of NGF. [3H]-noradrenaline release was reduced by nifedipine, suggesting an important role for L-Ca2+ channels in neurotransmitter release. Total high voltage Ca2+ currents were significantly increased in ELF MF-treated cells with NGF, but these currents in ELF MF-treated cells were more sensitive to nifedipine. Amperometric analysis of catecholamine release revealed that the KCl-induced activity of cells stimulated to differentiate by ELF MF is highly sensitive to L-type Ca2+ channel blockers. A possible mechanism to explain the way in which the application of magnetic fields can induce differentation of chromaffin cells into neuronal-like cells is proposed.
Article
We have previously observed that cytosolic free calcium ([Ca<sup>2+</sup>] i ) is increased in HL‐60 cells after exposure to an extremely low frequency (ELF) magnetic field. To facilitate the investigation of the time dependence of this effect, we built a magnetic field exposure system in which four samples of cells could be kept at constant temperature and maintained in suspension with constant mixing. Fluorimetric measurements of [Ca<sup>2+</sup>] i from these samples were made in real time using a periscopic optical system attached to a fluorescence spectrophotometer. This design feature eliminated possible confounding influences of the applied magnetic field on the light detection electronics, and isolated the cell samples against stray magnetic fields from electronic equipment. The operation of the instrument was computerized. This permitted the precise definition and replication of experimental conditions such as the properties of the applied magnetic field, the rate of sample mixing, and the instant at which an agonist was added to the cells. It also provided a time‐resolved record of sample temperature and fluorescence for each experiment. Characterization of the instrument enabled us to rule out potential confounding influences on fluorimetric [Ca<sup>2+</sup>] i measurements, such as temperature and mechanical vibration, and to have confidence in the accuracy and precision of the applied ELF and static magnetic fields. Experiments with HL‐60 and Jurkat cells verified that the instrument could measure [Ca<sup>2+</sup>] i in live cells and follow large rapid changes in [Ca<sup>2+</sup>] i after activation with an agonist. This instrument will provide an ideal tool with which to investigate in greater detail the effect of ELF magnetic fields on [Ca<sup>2+</sup>] i in HL‐60 cells. © 1996 American Institute of Physics.
Article
Electrification in developed countries has progressively increased the mean level of extremely low-frequency electromagnetic fields (ELF-EMFs) to which populations are exposed; these humanmade fields are substantially above the naturally occurring ambient electric and magnetic fields of approximately 10(-4) Vm(-1) and approximately 10(-13) T, respectively. Several epidemiological studies have concluded that ELF-EMFs may be linked to an increased risk of cancer, particularly childhood leukemia. These observations have been reinforced by cellular studies reporting EMF-induced effects on biological systems, most notably on the activity of components of the pathways that regulate cell proliferation. However, the limited number of attempts to directly replicate these experimental findings have been almost uniformly unsuccessful, and no EMF-induced biological response has yet been replicated in independent laboratories. Many of the most well-defined effects have come from gene expression studies; several attempts have been made recently to repeat these key findings. This review analyses these studies and summarizes other reports of major cellular responses to EMFs and the published attempts at replication. The opening sections discuss quantitative aspects of exposure to EMFs and the incidence of cancers that have been correlated with such fields. The concluding section considers the problems that confront research in this area and suggests feasible strategies.
Article
The effect of 3-Hz, monopolar, quasi-rectangular magnetic field pulses on 45Ca2+ uptake in resting and mitogen-treated rat thymic lymphocytes was evaluated. A 30-min, non-thermal exposure to the pulsed magnetic field (Bpeak = 6.5 mT, Emax = 0.69 mV/cm, Jmax = 2.6 microA/cm2) reduced Concanavalin A-induced 45Ca2+ uptake by 45%. It was observed that (i) the induction of the 3-Hz field response depended on Ca2+ signal transduction activation; (ii) the response direction (stimulation or inhibition) depended on the level of lymphocyte mitogen responsiveness, and (iii) the field response magnitude increased with increasing magnetic field flux densities (Bpeak = 0, 1.6, 6.5 and 28 mT). Our results demonstrate field effects at Bmax nearly 10(4) greater than that of the average human environment for low-frequency magnetic fields and they are consistent with the independent results from other 3-Hz pulsed magnetic field studies with lymphocytes.