ArticlePDF Available

Extremely Low Frequency Magnetic Fields Inhibit Adipogenesis of Human Mesenchymal Stem Cells

Authors:

Abstract and Figures

It was reported that obese (Ob/Ob) mice lose their weight and fat when treated with 0.5 T direct current electromagnetic fields. We also observed that 7.5 Hz, 0.4 T rotation of extremely low frequency magnetic fields (ELF-MF) has an inhibitory effect on obesity. Mesenchymal stem cells (MSCs) are multi-potent cells capable of differentiating to different MSC lineages, including adipose. We hypothesized that inhibitory effects of ELF-MF on obesity may be related to the differentiation of MSCs to adipocytes. In the present study, we investigated the effects of 7.5 Hz, 0.4 T ELF-MF on differentiation of human umbilical cord MSCs. We found that ELF-MF inhibited adipogenic differentiation (exposed 2 h/day for 15 days) of MSCs but had no effect on osteogenic differentiation (exposed 2 h/day for 21 days). Moreover, ELF-MF inhibited adipocyte-specific expression of peroxisome proliferator-activated receptor 2 (PPARγ2). ELF-MF promoted c-Jun N-terminal kinase (JNK)-dependent intracellular signaling in MSCs. Furthermore, activation of the non-canonical Wnt pathway provoked the inhibition of PPARγ2 expression resulting in suppression of adipogenic differentiation. In addition, the effects of ELF-MF on growth and apoptosis of MSCs were not observed. Our data indicated that ELF-MF of 7.5 Hz, 0.4 T inhibited the adipogenic differentiation of MSCs via JNK-dependent Wnt signaling pathway, but had no effect on the growth and function of MSCs, suggesting the inhibitory effect of ELF-MF on obesity may be attributed to the inhibition of differentiation of MSCs into adipocytes. This study may provide a potential approach for the treatment of obesity. Bioelectromagnetics © 2014 Wiley Periodicals, Inc.
Content may be subject to copyright.
Extremely Low Frequency Magnetic Fields
Inhibit Adipogenesis of Human Mesenchymal
Stem Cells
Leilei Du,
1
Hongye Fan,
1
Huishuang Miao,
1
Guangfeng Zhao,
2
and Yayi Hou
1
*
1
The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology,
Medical Scho ol, Nanjing Universi ty, Nanjing, P.R. China
2
Department of Obstetrics and Gynecology, NanjingDrumTower Hospital,
NanjingUniversity Medical School, Nanjing, P.R. China
It was reported that obese (Ob/Ob) mice lose their weight and fat when treated with 0.5 T direct
current electromagnetic fields. We also observed that 7.5 Hz, 0.4 T rotation of extremely low
frequency magnetic fields (ELF-MF) has an inhibitory effect on obesity. Mesenchymal stem cells
(MSCs) are multi-potent cells capable of differentiating to different MSC lineages, including
adipose. We hypothesized that inhibitory effects of ELF-MF on obesity may be related to the
differentiation of MSCs to adipocytes. In the present study, we investigated the effects of 7.5 Hz,
0.4 T ELF-MF on differentiation of human umbilical cord MSCs. We found that ELF-MF inhibited
adipogenic differentiation (exposed 2 h/day for 15 days) of MSCs but had no effect on osteogenic
differentiation (exposed 2 h/day for 21 days). Moreover, ELF-MF inhibited adipocyte-specific
expression of peroxisome proliferator-activated receptor 2 (PPARg2). ELF-MF promoted c-Jun N-
terminal kinase (JNK)-dependent intracellular signaling in MSCs. Furthermore, activation of the
non-canonical Wnt pathway provoked the inhibition of PPARg2 expression resulting in suppression
of adipogenic differentiation. In addition, the effects of ELF-MF on growth and apoptosis of MSCs
were not observed. Our data indicated that ELF-MF of 7.5 Hz, 0.4 T inhibited the adipogenic
differentiation of MSCs via JNK-dependent Wnt signaling pathway, but had no effect on the growth
and function of MSCs, suggesting the inhibitory effect of ELF-MF on obesity may be attributed to
the inhibition of differentiation of MSCs into adipocytes. This study may provide a potential
approach for the treatment of obesity. Bioelectromagnetics. © 2014 Wiley Periodicals, Inc.
Key words: ELF-MF; adipogenic differentiation; MSCs; obesity; PPARg2
INTRODUCTION
Obesity is a major health concern worldwide
[Friedman, 2004] and is associated with the develop-
ment of a number of pathological disorders including
certain cancers, heart disease, stroke, and diabetes
[Cornier et al., 2008; Attie and Scherer, 2009; Pi-
Sunyer, 2009; Nichols, 2012]. Mesenchymal stem
cells (MSCs) are multi-potent cells capable of differen-
tiating in vitro and in vivo to different MSC lineages,
including adipose, bone and cartilage [Haynesworth
et al., 1992; Pittenger et al., 1999]. Recent studies
suggested that the inverse relationship between bone
and fat mass might be caused by enhanced differentia-
tion of MSC into either the osteoblastic or adipocytic
lineages [Takeda et al., 2003; Gimble et al., 2006].
Therefore, it is important to maintain the balance of
osteogenic differentiation and adipogenic differentia-
tion of MSCs to prevent obesity.
Electromagnetic fields have been studied with
great interest due to the possible effects they have on
human health. Pulsed electromagnetic fields (PEMF)
enhance osteogenic effects of bone morphogenetic
Grant sponsor: National Natural Science Foundation of China;
grant number: 31370899; grant sponsor: Ministry of Science and
Technology Twelfth Five-YearNational Scientific and Techno-
logical Support for Major Projects (China); grant number:
2012BA/15B03.
*Correspondence to: Yayi Hou, Medical School, Nanjing Universi-
ty, Hankou Road 22, Nanjing 210093, P.R. China.
E-mail: yayihou@nju.edu.cn
Received for review 1 January 2014; Accepted 14 July 2014
DOI: 10.1002/bem.21873
Published online XX Month Year in Wiley Online Library
(wileyonlinelibrary.com).
Bioelectromagnetics
2014 Wiley Periodicals, Inc.
protein 2 (BMP-2) on MSCs cultured on calcium
phosphate substrates, suggesting that PEMF will
improve MSC response to BMP-2 in vivo in bones
[Schwartz et al., 2008]. It was reported that 15 Hz,
1 mT magnetic fields can directly regulate the rat bone
marrow MSCs, promoting their differentiation to
osteoblasts and inhibiting differentiation to adipocytes
[Yang et al., 2010]. A recent study found that
alternating electric current promoted the differentiation
of adult human MSCs toward the osteogenic pathway
[Creecy et al., 2013], and the high-frequency (200 Hz)
vibratory stimulation, when combined with a synthetic
fibrous scaffold, served as a potent modulator of MSC
functions [Tong et al., 2013].
Moreover, frequencies below 300 Hz are known
as extremely low frequency magnetic fields (ELF-
MF). Interestingly, ELF-MFs do not have enough
energy to break molecular bonds, for example, they
cause no direct damage to DNA. ELF-MF is also non-
invasive and non-ionizing and may have non-thermal
effects on cells and tissues. These properties have led
to studies of ELF-MF influence on the development of
various diseases. It has been found that ELF-MF might
be a way to stimulate and maintain chondrogenesis of
MSCs and provide a new step in regenerative medi-
cine regarding tissue [Mayer-Wagner et al., 2011]. We
previously found that ELF-MF of 7.5 Hz, 0.4 T can
inhibit tumor cell proliferation and disrupt the cell
cycle [Wang et al., 2011]. It was reported that when
obese (Ob/Ob) mice were treated with 0.5 T direct
current electromagnetic fields, the mice increased their
activity, lost weight and fat in a 6-day period
[Nichols, 2012]; electromagnetic fields also reduced
human abdominal obesity [Beilin et al., 2012]. We
hypothesized that inhibitory effect of ELF-MF on
obesity may be related to the differentiation of MSCs
to adipocytes.
Adipogenesis consists of two related steps: the
determination of MSCs into preadipocytes and the
differentiation of preadipocytes into mature fat cells
[Bowers and Lane, 2007]. Because the number of
preadipocytes and mature fat cells has been shown to
be different between lean and obese human adult
subjects [Tchoukalova et al., 2007], variations in the
determination process in early stages of adipose tissue
development might be important in the pathogenesis
of obesity. However, the regulation of adipogenesis
(adipocyte differentiation) is complex and this process
includes alteration of the sensitivity to hormones and
the expression of a number of genes in response to
various stimuli including lipid mediators.
Interestingly, several transcriptional factors and
intracellular signaling pathways have been demonstrated
to control the differentiation of MSCs into osteoblastic
or adipocyte cells, for example, peroxisome proliferator-
activated receptor (PPAR) g2 [Akune et al., 2004] and
canonical Wnt-b-catenin and non-canonical Wnt signal-
ing pathways [Taipaleenmaki et al., 2011]. PPARgis a
ligand-activated transcription factor [Mangelsdorf
and Evans, 1995; Kersten et al., 2000; Rosen and
Spiegelman, 2001]. It is believed that PPAR gand
CCAAT/enhancer-binding proteins (C/EBPs) are the
most important factors involved in the activation of
adipogenesis, and they induce the expression of a
number of adipogenic genes that participate in the
control of adipogenesis [Lefterova and Lazar, 2009;
Rosen et al., 2009]. PPARg2 overexpression in fibro-
blast cell lines can initiate adipogenesis [Tontonoz et al.,
1994] and PPARgdefect in ES cells and embryonic
fibroblastic cells from mice were unable to differentiate
into adipocytes [Barak et al., 1999; Kubota et al., 1999;
Rosen et al., 1999]. Canonical Wnt/b-catenin signaling
is a key regulator of bone formation and MSC differenti-
ation to either the osteogenic or chondrogenic lineage
[Church et al., 2002; Day et al., 2005; Hill et al., 2005;
Holmen et al., 2005]. In addition, non-canonical Wnt
signaling is also a regulator of cell differentiation [He
et al., 2008].
Therefore, in the present study, we attempted to
explore the effect of 7.5 Hz, 0.4 T rotation of ELF-MF
on the differentiation of human umbilical cord MSCs
(UC-MSCs) to adipocytes. Furthermore, we tried to
investigate whether ELF-MF affects the differentiation
of MSCs through regulating the expression of PPAR g
2 and Wnt signaling pathways.
MATERIALS AND METHODS
Culture of Human Umbilical Cords
MSCs ( UC -MSC)
Human umbilical cords were obtained from full-
term caesarian section births in a sterile manner at the
time of delivery at the Department of Gynecology and
Obstetrics, the Affiliated Drum Tower Hospital of
Nanjing University Medical School (Nanjing, China).
The hospital ethics committee approved the consent
forms and the protocol for evaluation of the tissue.
Umbilical arteries and veins were removed, and the
remaining tissue was transferred to a sterile container
in DMEM/F12 (Gibco, Carlsbad, CA) with antibiotics
(penicillin 100 mg/ml, streptomycin 10 mg/ml; Life
Sciences, Carlsbad, CA) and was diced into 12mm
3
fragments. The tissue was incubated in an enzyme
cocktail (hyaluronidase 5 U/ml, collagenase 125 U/ml,
and dispase 50 U/ml; Sigma, St. Louis, MO) for 45
60 min with gentle agitation at 37 8C. The cells were
pelleted by low-speed centrifugation (250gfor 5 min),
2Duetal.
Bioelectromagnetics
suspended in fresh medium, and transferred to cell
culture flasks containing DMEM/F12 supplemented
with 20% fetal bovine serum (Gibco).
Cells were incubated with 5% CO
2
at saturating
humidity. When cells reached 7080% confluence or
when numerous colonies were observed, the cells were
detached with 0.25% trypsin-EDTA (Sigma); the trypsin
was inactivated with fresh media. The culture medium
was replaced every 3 or 4 days. After the 2nd to 4th cell
passages, the adherent cells were symmetric, with
phenotypic surface antigens CD105
þ
,CD73
þ
,CD90
þ
,
HLA-ABC
þ
,CD29
þ
,CD44
þ
, CD106
,HLA-DR
,
CD19
,CD11b
,CD14
,CD34
,CD31
. The cells
of the 3rd passage were used in the experiments.
Flow Cytometry
The specific surface antigens of UC-MSCs in the
cultures, after passages 24, were characterized by
flow cytometry analysis. The following murine
monoclonal antibodies, purified or directly conjugated
with fluorescein isothiocyanate (FITC), phycoerythrin
(PE or R-PE), allophycocyanin (APC) were used in
fluorescence-activated cell sorting (FACS) analysis:
anti-CD105, anti-CD73, anti-CD90, anti-HLA-ABC,
anti-CD29, anti-CD44, anti-CD106, anti-HLA-DR, anti-
CD19, anti-CD11b, anti-CD14, anti-CD34, anti-CD31,
anti-CD45, and IgG/IgM isotype controls (all from BD
Biosciences, San Jose, CA). For fluorescence measure-
ments only, data from 10000 single cell events were
collected using a standard FACScalibur flow cytometer
(Becton Dickinson, San Jose, CA). Data were analyzed
using CELLQuest (Becton Dickinson) or FlowJo soft-
ware (Treestar, San Carlos, CA).
Experimental Magnetic Field
The construction of experimental magnetic
fields has been described previously [Wang
et al., 2011; Nie et al., 2013]. Two pairs of fan-
shaped NdFeB permanent magnets (N45; Innuovo,
Dongyang, China; Fig. 1A) were attached to a
circular iron plate and arranged to produce ELF-MF
(Fig. 1B). The black arrow indicated site is the place
where MSCs were exposed. The bottom two magnets
rotated at certain frequency driven by a step motor,
which was controlled using a functional signal
generator. The top two magnets rotated synchronous-
ly due to the strong magnetic interaction. Magnetic
flux density was measured at the target site using a
gauss meter (HT201; Hengtong, Shanghai, China).
ELF-MF at the target site is a series of alternate
pulses with a maximum flux density of 0.4 T (6%
variation). The frequency of ELF-MF could be
varied from 0 to 7.5 Hz, but all experiments were
conducted at 7.5 Hz. This instrument was fabricated
by the National Laboratory of Solid Micro-struc-
tures, Nanjing University (Nanjing, China). Control
cells were placed in a similar apparatus except that
there were two rotating iron plates instead of
magnets, thus lacking an ELF-MF. The entire
magnetic apparatus was located in a hood with
controlled humidity and temperature.
MSCs Osteogenesis and Adipogenesis
Differentiation
For differentiation experiments, the methods
were used as Bilkovski et al. [2010] described. MSCs
were transferred to six-well plates. Two days post-
confluence, adipogenesis was induced by adding
adipogenesis medium (Dulbeccos modified Eagles
medium, 10% FBS, 1% penicillin/streptomycin, 1 mM
dexamethasone, 5 mg/ml insulin, 0.5 mM isobutylme-
thylxanthine, and 50 mM indomethacin). Osteogenesis
was induced at 80% confluency by adding osteogenesis
medium (Dulbeccos modified Eagles medium,
10%FBS, 1% penicillin/streptomycin, 100 nM dexa-
methasone, 10 mM ascorbic acid, and 10 mM
b-glycero-phosphate). Before performing the experi-
ments, the two primary cell populations were cultured
for at least 3 weeks under standard conditions.
Alizarin Red S Stain Analysis
After 21 days of differentiating condition, media
was removed from the six-well plate and rinsed once
with PBS. Cells were fixed with 4% formaldehyde
solution for 30 min. After fixation, wells were rinsed
twice with distilled water and cells were stained with
2% Alizarin Red S solution (pH 4.2; Sigma) for 2
3 min. Wells were rinsed three times with distilled
water, visualized under a light microscope and images
were captured for analysis.
Oil Red O Stain Analysis
After 21 days of differentiating condition, media
from six-well plate was removed and rinsed once with
PBS. Cells were fixed with 4% formaldehyde solution
for 30 min. After fixation, wells were rinsed twice with
distilled water and 1 ml of 60% isopropanol was added
for 30 min. Cells were then stained with Oil Red O
solution for 30 min. Wells were rinsed three times with
distilled water, visualized under a light microscope
and images were captured for analysis.
Reverse Transcription and Real-Time
PolymeraseChainReaction(PCR)
Total RNA was extracted from the cultured cells
using Trizol reagent (Invitrogen, Carlsbad, CA)
according to the manufacturers instructions. For
quantitative RT-PCR analysis of genes FABP4,
ELF-MF Inhibits Adipogenesis of Human MSCs 3
Bioelectromagnetics
PPARg2, LPL, ALP, Runnx2, OCN, Col1a1 and
GAPDH, 1 mg of total RNA was reverse transcribed to
cDNA with oligdT and Thermoscript (TaKaRa, Dalian,
China). Real-time PCR for these genes was performed
on a tepOne Sequence Detection System (Applied
Biosystems, Foster City, CA) using SYBR green dye
(Invitrogen). A 10 ml PCR reaction was used and
included 1 ml RT product, 5 ml2QuantiTect SYBR
green PCR Master Mix, and 0.5 ml forward and
reverse primers. The forward and reverse PCR oligo-
nucleotide primers were designed with the Primer
premier 5.0 software (Premier Biosoft, Palo Alto, CA),
and the primer sequences were blasted to exclude the
nonspecific sequences. The primer sequences are
shown in Table 1. The reactions were incubated in a
96-well plate at 95 8C for 10 min, followed by 40
cycles of 95 8C for 15 s, 60 8C for 30 s and 72 8C for
30 s. The housekeeping gene GAPDH was used as
endogenous control for RNA normalization. All
experiments were done in triplicate. The level of
expression was calculated based on the PCR cycle
number (C
t
) and the relative gene expression level was
determined using the DDC
t
method.
Western Blot Analysis
Whole-cell lysates for Western blotting were
extracted with lysis buffer containing 50 mM Tris
(pH 8), HEPES (pH 7.5), 150 mM NaCl, 1.5 mM
MgCl
2
, 1 mM EDTA, 0.1% Triton X-100, 0.25%
sodium deoxycholate and protease inhibitor (Roche,
Basel, Switzerland). Protein samples were resolved
by 10% SDS/PAGE, and gels were transferred to
polyvinylidene difluoride membranes (Roche). Mem-
branes were blocked using 5% bovine serum albumin
Fig. 1. Magnetic field exp osure system. A: The magnetic field exposure system instrument was
fabricated by the National Laboratory of Solid Micro-structures, Nanjing University (Nanjing,
China). Internal schematic diagram of the instrument.The black arrow indicated site where MSCs
were exposed. B: Two pairs of fan-shaped NdFeB permanent magnets were arranged to estab-
lish magnetic fields.ELF- MF at the target site is alternative pulses with a maximum flux density of
about 0.4 T.
4Duetal.
Bioelectromagnetics
(BSA) for 12h,at258C and subsequently incubated
overnight at 4 8C with diluted primary monoclonal
antibodies against PPARg2 (1:1000 dilution; Santa
Cruz, Dallas, TX), JNK (1:1000 dilution), p-JNK
(1:1000 dilution), b-catenin (1:1000 dilution; Santa
Cruz), P-CamKII (1:1000 dilution), CamKII (1:1000
dilution), and GAPDH (1:1000 dilution; Cell Signal-
ing Technology, Boston, MA). Signals were detected
TABLE 1. Primers Used for Real-Time Quantitative PCR Analysis
SANGER_NAME Forward primer (5030) Reverse primer (5030)
HUMAN-PPARg2 CTCCTATTGACCCAGAAAGCG CAAAGTTGGTGGGCCAGAAT
HUMAN-FABP4 GGAGTGGGCTTTGCCACCAGG GCACATGTACCAGGACACCCCC
HUMAN-LPL GTGGACTGGCTGTCACGGGC GCCAGCAGCATGGGCTCCAA
HUMAN-Runx2 GATGTCCGTAAGGTCTTGCCA TGCAGTCTCCATCACGAAATG
HUMAN-Col1a1 CTTGGTCTCGTCACAGATCA TGTTCAGCTTTGTGGACCTC
HUMAN-OCN CGGTGCAGAGTCCAGCAAAG TACAGGTAGCGCCTGGGTCTCT
HUMAN-ALP TACCCAGATGACTACAGCCAA TCGGTGGATCTCGTATTTCATGT
HUMAN-GAPDH AGAAGGCTGGGGCTCATTTG AGGGGCCATCCACAGTCTTC
Fig. 2. Isolation and characterization of UC-MSCs derived from human umbilical cord tissue. A:
Morphology of MSCs isolate after 4, 6, and 8 days. B: Flow cy tometry characterization of human
MSC passage 3. Phenotypic surface antigens of MSCs (CD105, CD73, CD90, HLA-ABC, CD29,
CD44, HLA-DR, CD19,CD11b,CD14,CD34, CD45, CD106, and CD31) were detected using FACS.
ELF-MF Inhibits Adipogenesis of Human MSCs 5
Bioelectromagnetics
using the appropriate HRP-conjugated secondary
antibody (Cell Signaling Technology). The blots
were visualized using an enhanced Immobilon West-
ern chemiluminescent HRP substrate (Millipore,
Billerica, MA), according to the manufacturers
instructions, and the relative intensity of the specific
bands was quantified using the FluorChem FC2
system (Alpha Innotech, San Jose, CA).
Immunofluorescence
The effects of ELF-MF on expression of PPARg2
and cytoskeleton of MSCs could be detected by
immunofluorescence. Briefly, exponentially growing
cells were seeded onto a six-well plate (Costar,
Carlsbad, CA; 1 10
5
cells/well). The cells were
adherent overnight. Media was removed from six-well
plates and rinsed once with PBS. Cells were fixed with
4% formaldehyde solution for 30 min. After fixation,
wells were rinsed twice with PBS and 0.1% Triton-100
was added for 15 min; wells were then rinsed twice
with PBS and 3% BSA was added for 1 h; wells were
rinsed twice with PBST, after adding PPARg2Ab
(Santa Cruz) (1:2000) for PPARg2 protein detect or
mouse monoclonal vinculin Ab (Santa Cruz) (1:2000)
for cytoskeleton, 4 8C overnight. Wells were rinsed
fourth with PBST, adding again Alexa Fluor 594
donkey anti-mouse IgG (Abcam, Cambridge, England;
1:1000) was added for PPARg2 protein detect or Alexa
Fluor 594 donkey anti-mouse IgG (Abcam; 1:1000)
and Phalloidin-FITC (Santa Cruz; 1:1000) for 2 h.
Wells were rinsed four times with PBST, DAPI was
added for 10 min. Using a laser scanning confocal
microscope (Olympus FluoView FV10i, Osaka, Japan)
cells were observed and a photograph was taken.
Cell Proliferation, Cell Cycle, and Apoptosis
Assay
The effect of ELF-MF on cell viability was
determined using the Cell Counting Kit-8 (CCK-8)
assay (Dojindo, Kumamoto, Japan). Briefly, exponen-
Fig. 3. The effects of ELF-MF on osteogenic differentiation of MSCs. A: The cells were cultured
in osteogenesisinducingmedium andexposed to ELF-MF (ELF group) orsham exposure (CON).
The cellswithout adding osteogenesis mediumwerea s NC group.Osteoblast-specific mRNAex -
pression of ALP, COL1a1, OCN and Runx2 and were measured by the real-time PCR. The real-
time PCR results are expressed as means SEM. B:AlizarinRedSstainingwasperformedto
detect the osteogenic differentiationat day 21.
6Duetal.
Bioelectromagnetics
tially growing cells were seeded onto a 24-well plate
(Costar; 2 10
4
cells/well). Growing cells were exposed
to ELF of 7.5 Hz, 0.4 T with different exposure times.
Identical, non-energized exposure chambers were used
for sham exposure of control cells in the same room.
Twenty microliters of CCK-8 solution was added to
each well, and the cells were further incubated at 37 8C
for another 3.5 h. The absorbance values (A) at 450 nm
were measured on an ELx-800 Universal Microplate
Reader (BioTek, Winooski, VT). All data are expressed
as mean values from three independent studies. For the
apoptosis assay, the cells were harvested, stained with
propidium iodide and anti-Annexin-V antibody, and
then analyzed by a fluorescence-activated cell-sorting
(FACS) Calibur (BD Biosciences). For cell cycle
experiment, the treated cells were harvested, washed
once with PBS, and fixed in 70% ethanol overnight.
Staining of DNA content was performed with 50 mg/ml
propidium iodide and 1 mg/ml RNase A for 30 min.
Analysis was performed with Cell Quest Pro software.
Cell-cycle modeling was performed with Modfit 3.0
software (Verity Software House, Topsham, ME).
Statistical Analysis
All values were expressed as mean SEM. The
analyses were conducted with the SPSS 11.5 software
(SPSS, Chicago, IL). Statistical significance was
assessed by Studentst-test. For PCR analysis, statistical
analyses were conducted according to instructions for
the Gel-Pro Analyzer (Alpha Innotech). For all statistical
tests, P<0.05 was considered significant.
RESULTS
Morphology of Human Umbilical Cords MSCs
in Culture
The cultures of primary UC-MSCs underwent an
initial lag phase of about 48 days, adherent cells with
fibroblastic morphology could be observed as early as
24 h. The cells formed a monolayer of homogenous
Fig. 4. The effects of ELF-MF on adipogenic differentiation of Human umbilical cords MSCs. A:
The cells were cultured in adipogenesis inducing medium and exposed to (ELF group) or sham
exposure (CON).The cells without adding adipogenesis medium were as NC group. adipocyte-
specific mRNAexpression of PPARg2, FABP4, and LPL was measured by the real-time PCR.The
real-time PCR results are expressed as means SEM. P<0.05 versus CON.B:Oil RedOstain-
ingwas performed detect the adipogenic differentiationat day 21.
ELF-MF Inhibits Adipogenesis of Human MSCs 7
Bioelectromagnetics
bipolar spindle-like cells with a whirl pool like array
within 1 week (Fig. 2A). After 3 cell passages, the
adherent cells were symmetric with phenotypic surface
antigens. Results showed that UC-MSCs shared most
of their immunophenotype with bone marrow-derived
MSCs (BM-MSCs) as reported, including positivity
for CD29, CD44, CD90, CD105 (SH2), CD73 (SH3),
and HLA-ABC, negativity for CD19, CD11b, CD14,
CD34, and CD31 (endothelial cell marker) and HLA-
DR (Fig. 2B).
Effects of ELF-MF on Osteogenesis by Human
UC-MSCs
To test the effect of ELF-MF on osteogenic
differentiation of human UC-MSCs, osteoblast-spe-
cific mRNA expression of ALP, COL1a1, Runx2
and OCN was measured by the real-time PCR. The
adherent MSCs with plates (ELF group) were
exposed for 2 h/day for 15 days. Control treatments
(CON group) were placed in a similar apparatus but
without ELF-MF. The cells without added osteogen-
esis medium were the NC group. Real-time PCR
results showed that ELF-MF had no effects on the
expression of ALP, COL1a1, Runx2, and OCN
(Fig. 3A). Furthermore, we analyzed the osteogenic
differentiation of MSCs using Alizarin Red S Stain
analysis. After 21 days of ELF-MF exposure (2 h/
day), there was no change on osteogenic differentia-
tion compared with control samples (Fig. 3B).
ELF-MF Exposure Inhibits Adipocyte Master
Gene Expression of Human UC-MSCs
To test the effect of ELF-MF on adipogenic
differentiation of human UC-MSCs, adipocyte-specific
mRNA expression of PPARg2, FABP4, and LPL was
measured by the real-time PCR. The adherent MSCs
with plates (ELF group) were exposed for 2 h/day for
15 days. Control treatments (CON group) were placed
in a similar apparatus without the ELF-MF. The cells
without added adipogenesis medium were the NC
group. ELF-MF exposure for 15 days resulted in a
decrease in PPARg2 and FABP4 over the non-treated
control (P<0.01; Fig. 4A). To further confirm the effect
of ELF-MF on MSCs adipogenesis, cells were incubated
in adipogenic induction medium with ELF-MF (2 h/day)
or sham exposure for 21 days and then stained with Oil
Red O. In Figure 4B, we can see that the control has
more lipid droplets than the ELF-MF exposed group.
ELF-MF Downregulates the PPARg2
Expression of Human UC-MSCs
To further study whether expression of PPARg2
proteinwasdownregulatedbyELF-MF,protein
expression analysis was performed using Western
blot and immunofluorescence methods. MSCs were
cultured in adipogenic induction medium with ELF-
MF or sham exposure for 2 h/day for 21 days. Then
the cells were stained with PPARg2 for immunofluo-
rescence assay or the cells were lysed to PPARg2
protein level with immunoblot analysis. As shown in
Figure 5A, after treatment with ELF-MF for 21 days,
PPARg2 expression in MSCs was decreased. West-
ern blotting results also showed that the level of
PPARg2 protein was reduced after stimulation with
ELF-MF (Fig. 5B).
Effect of ELF-MF on Adipogenic Differentiation
of Human UC-MSCs Is Mediated via the
JNK-Dependent Noncanonical Wnt
Signaling Pathway
We next aimed to investigate how ELF-MF
affected MSCs adipogenic differentiation. We detected
several central signaling molecules (b-catenin, Cam-
KII, JNK, and p-JNK) in Wnt signaling pathway.
MSCs were cultured in adipogenic induction medium
with ELF-MF or sham exposure for 2 h/day for
Fig. 5. The effect of ELF-MF exposure on PPARg2 protein ex-
pression of human umbilical cords MSCs. A:MSCscellswere
cultured on coverslips in adipogenic induction medium. After
treatment with ELF-MF (ELF) or sham exposure (CON) for
21days, MSCs were fixed and immunostained with specific Abs
for rat PPARg2(60). B: MSCs cells treated with were ELF-MF
(ELF) or sham exposure (CON) for 21 days. Representative
Western blots PPARg2 and GAPDH in MSCs were shown and
the relative expression of PPARg2 to GAPDH was calculated.
The results are shown as mean SE from three representative
independent experiments. P<0.05, compared with CON.
8Duetal.
Bioelectromagnetics
21 days. Then the cells were lysed and the immunoblot
analysis was performed. ELF-MF had no effects on
the expression of b-catenin, CamKII, and p-CamKII.
However, the level of p-JNK was increased after ELF-
MF exposure (Fig. 6).
ELF-MF Has No Ef fect on Morphology, Cell
Cycle, Apoptosis, and Cell Proliferation of
Human UC-MSCs
Our studies have shown that ELF-MF was able to
inhibit adipogenic differentiation of MSCs. We further
examined the effects of ELF-MF on cytoskeleton, cell
viability, cell cycle and apoptosis of MSCs. F-actin and
vinculin are cell cytoskeleton proteins. After exposure
with ELF-MF of 7.5 Hz, 0.4 T for 14 days, 2 h/day,
ELF had no effects on the expression of F-actin and
vinculin in MSCs (Fig. 7A). In addition, the cell
viability was analyzed after treatment with ELF-MF
using CCK-8 assay kit. As shown in Figure 7B, ELF-
MF had no effects on MSCs cell viability. Then the cell
cycle was measured after treatment with ELF-MF. The
results showed that the cell cycle of ELF-MF treated
cells was similar to that of control cells (Fig. 7C).
Assessment of cell apoptosis of MSC showed that
treatment of ELF-MF of 7.5 Hz, 0.4 T for 14 days,
2 h/day had no effects on cell apoptosis (Fig. 7D).
DISCUSSION
Bassett [1982] used a pair of Helmholtz coils to
produce a magnetic field across a fracture site and
enhance osteogenesis. Since then, several experimental
studies have examined the influence of EMF on
osteoporosis. The effect appears to vary with the
waveform of magnetic field used [Bassett et al., 1981;
Brighton, 1984; Rubin et al., 1989; Skerry et al., 1991].
The effect was variable, but in some cases, osteoporo-
sis was prevented or even reversed. However, the exact
mechanism by which EMF stopped bone loss has not
been defined. In our study, we found that 7.5 Hz, 0.4 T
ELF-MF inhibit adipogenic differentiation of MSCs
while they have no affect on osteogenic differentiation.
Furthermore, we found that ELF-MF of 7.5 Hz, 0.4 T
may inhibit adipogenic differentiation of MSCs via
JNK-dependent Wnt signaling pathway.
ELF-MF inhibited adipocyte-specific expression
of PPARgand C/EBPs, but had no effects on the
expression of ALP, Runx2, OCN, and Col1a1. Several
transcriptional factors and intracellular signaling path-
ways have been demonstrated to control the differenti-
ation of MSCs into osteoblastic or adipocytic cells.
Generally, it is believed that PPAR gand C/EBPs are
the most important factors involved in the activation
Fig. 6. Effect of ELF-MF on Wnt signaling pathway. MSCs cells treated with were ELF-MF (ELF)
or sham exposure (CON) for 21days.Representative Western blots b-catenin,JNK, p -JNK,Cam -
KII, p-CamKII, and GAPDH in MSCs were shown and the relative expression of b-catenin to
GAPDH, p -JNK t o JNK and p- CamKIIto CamKIIwas calculated.The results are shownas mean
SE from three representativeindependent experiments. P<0.05, compared with CON.
ELF-MF Inhibits Adipogenesis of Human MSCs 9
Bioelectromagnetics
of adipogenesis [Noer et al., 2007; Hasegawa
et al., 2008; Lefterova and Lazar, 2009; Rosen et al.,
2009]. ALP, Runx2, OCN, and Col1a1 are osteogene-
sis markers of osteoblast differentiation [Glimcher
et al., 2007; Holleville et al., 2007]. Esposito et al.
[2013] reported that pulsed electromagnetic field
(PEMF) increased the division of MSCs and reduced
the time to obtain chondrocyte cell differentiation.
Ceccarelli et al. [2013] also found that PEMF
exposure promoted the cell proliferation of BM-MSCs
and increased ALK protein level and activity. Our
results demonstrated that ELF-MF inhibited adipo-
genesis of the stem cells but had no effect on
osteogenesis. This indicates that ELF-MF and PEMF
have different effects on MSCs differentiation. The
present findings that ELF-MF concomitantly corrected
osteoblastogenesis and adipogenesis suggests that
ELF-MF may act directly on the common precursor
cell to inhibit its commitment in the adipocyte lineage.
This may be one of the mechanisms by which ELF-
MF stops obesity.
ELF-MF was found to promote JNK-dependent
intracellular signaling in MSCs. Wnt pathway is
considered to be important in regulating mechanisms
for the proliferation, development, differentiation of
cells and organisms and can be divided into canonical
and noncanonical Wnt pathway. In the noncanonical
Wnt pathway, Wnt ligand such as Wnt5a regulates
target gene expression. JNK is a downstream effector
of Wnt5a that activates activator protein 1 (AP-1),
thereby regulating planar cell polarity (PCP) signaling
[Ling et al., 2009; Rao and Kühl, 2010]. Several recent
Fig. 7. Effects of ELF-MF on cytoskeleton, cell viability, cell cycle and apoptosis of MSCs.
A: MSCs cells were cultured on coverslips. After treatment with 7.5 Hz, 0.5 T ELF-MF (ELF) or
sham exposure (CON) for 21days, MSCs were fixed and immunostained with specific Abs forrat
F-actin (Green) and Vinculin (Red) or DAPI for nucleus (60). B: Cell viability analysis of MSCs
treated with ELF-MF using CCK- 8 a ssay kit. C: Representative data of flow cytometry analysis of
cell cycle. After 4 days of treatment, cells were analyzed and the percentage of cells in different
cell cycle interphases G0/G1, S, and G2 are indicated. D: Repre sentative data of flow cytometry
analysis ofapoptosis. D1,D2, D3,D 4 refers to cellswere treatedwith1- 4 days of ELF-MFexposure
(7.5 Hz, 0.4 T, 2 h/day). Data from onerepresentative experiment performedin triplicate.
10 Du et al.
Bioelectromagnetics
studies have shown that noncanonical Wnt signaling
has critical effects on the differentiation of MSCs,
which express a number of ligands, receptors and
pathway inhibitors [Etheridge et al., 2004]. In this
study, we found that ELF-MF activates JNK nonca-
nonical signaling in human MSCs. Interestingly it has
been shown that JNK signaling is essential in the
regulation of bone formation and inactivation of JNK
signaling impairs osteogenesis, although adipogenesis
is promoted [Tominaga et al., 2005].
There are some limitations to our study. First,
most studies show that in the adipogenic and osteo-
genic differentiation of MSCs there exists a reciprocal
relationship. This means that if adipogenic differentia-
tion was promoted, osteogenic differentiation will be
inhibited. In our study, ELF-MF inhibited adipogenic
differentiation, but had no effect on osteogenic differ-
entiation; a point to be further explored in the future.
Our study revealed that ELF-MF acted on MSCs to
inhibit differentiation to adipocytes. However, it is not
clear if this effect is direct or indirect.
In conclusion, the adipogenic differentiation of
MSCs could be inhibited by ELF-MF of 7.5 Hz, 0.4 T,
suggesting the inhibitory effect of ELF-MF on obesity
may be attributed to the inhibition of differentiation of
MSCs into adipocytes. Despite the limitations of our
study, our findings have several implications for the
biology and therapy of human diseases associated with
adipogenesis and will provide a possible approach for
the treatment of obesity.
REFERENCES
Akune T, Ohba S, Kamekura S, Yamaguchi M, Chung UI, Kubota
N, Terauchi Y, Harada Y, Azuma Y, Nakamura K, Kadowaki
T, Kawaguchi H. 2004. PPARgamma insufficiency enhan-
ces osteogenesis through osteoblast formation from bone
marrow progenitors. J Clin Invest 113:846855.
Attie AD, Scherer PE. 2009. Adipocyte metabolism and obesity.
J Lipid Res 50:S395399.
Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz-Lozano P, Chien
KR, Koder A, Evans RM. 1999. PPAR gis required for
placental, cardiac, and adipose tissue development. Mol
Cell 4:585595.
Bassett CA. 1982. Claims for magnetotherapy. Can Med Assoc J
127:10791080.
Bassett CA, Mitchell SN, Gaston SR. 1981. Treatment of ununited
tibial diaphyseal fractures with pulsing electromagnetic
fields. J Bone Joint Surg Am 63:511523.
Beilin G, Benech P, Courie R, Benichoux F. 2012. Electromagnetic
fields applied to the reduction of abdominal obesity.
J Cosmet Laser Ther 14:2442.
Bilkovski R, Schulte DM, Oberhauser F, Gomolka M, Udelhoven
M, Hettich MM, Roth B, Heidenreich A, Gutschow C,
Krone W, Laudes M. 2010. Role of WNT-5a in the
determination of human mesenchymal stem cells into
preadipocytes. J Biol Chem 285:61706178.
Bilkovski R, Schulte DM, Oberhauser F, Gomolka M, Udelhoven
M, Hettich MM, Roth B, Heidenreich A, Gutschow C,
Krone W, Laudes M. 2010. Role of WNT-5a in the
determination of human mesenchymal stem cells into
preadipocytes. J Biol Chem 285:61706178.
Bowers RR, Lane MD. 2007. A role for bone morphogenetic
protein-4 in adipocyte development. Cell Cycle 6:385
389.
Brighton CT. 1984. The semi-invasive method of treating non-
union with direct current. Orthop Clin North Am 15:3345.
Ceccarelli G, Bloise N, Mantelli M, Gastaldi G, Fassina L, De
Angelis MGC, Ferrari D, Imbriani M, Visai L. 2013. A
comparative analysis of the in vitro effects of pulsed
electromagnetic field treatment on osteogenic differentiation
of two different mesenchymal cell lineages. Biores Open
Access 2:283294.
Church V, Nohno T, Linker C, Marcelle C, Francis-West P. 2002.
Wnt regulation of chondrocyte differentiation. J Cell Sci
115:48094818.
Cornier MA, Dabelea D, Hernandez TL, Lindstrom RC, Steig AJ,
Stob NR, Van Pelt RE, Wang H, Eckel RH. 2008. The
metabolic syndrome. Endocr Rev 29:777822.
Creecy CM, ONeill CF, Arulanandam BP, Sylvia VL, Navara CS,
Bizios R. 2013. Mesenchymal stem cell osteodifferentiation
in response to alternating electric current. Tissue Eng Part A
19:467474.
Day TF, Guo X, Garrett-Beal L, Yang Y. 2005. Wnt/beta-catenin
signaling in mesenchymal progenitors controls osteoblast
and chondrocyte differentiation during vertebrate skeleto-
genesis. Cell 8:739750.
Esposito M, Lucariello A, Costanzo C, Fiumarella A, Giannini A,
Riccardi G, Riccion I. 2013. Differentiation of human
umbilical cord-derived mesenchymal stem cells, WJ-MSCs,
into chondrogenic cells in the presence of pulsed electro-
magnetic fields. In Vivo 27:495500.
Etheridge SL, Spencer GJ, Heath DJ, Genever PG. 2004. Expres-
sion profiling and functional analysis of wnt signaling
mechanisms in mesenchymal stem cells. Stem Cells
22:849860.
Friedman JM. 2004. Modern science versus the stigma of obesity.
Nat Med 10:563569.
Gimble JM, Zvonic S, Floyd ZE, Kassem M, Nuttall ME. 2006.
Playing with bone and fat. J Cell Biochem 98:251266.
Glimcher LH, Jones DC, Wein MN. 2007. Control of postnatal
bone mass by the zinc finger adapter protein Schnurri-3.
Ann NY Acad Sci 1116:174181.
Hasegawa S, Yamasaki M, Inage T, Takahashi N, Fukui T. 2008.
Transcriptional regulation of ketone body-utilizing enzyme,
acetoacetyl-CoA synthetase, by C/EBPalpha during adipo-
cyte differentiation. Biochim Biophys Acta 1779:414419.
Haynesworth SE, Goshima J, Goldberg VM, Caplan AI. 1992.
Characterization of cells with osteogenic potential from
human marrow. Bone 13:8188.
He S, Pant D, Schiffmacher A, Meece A, Keefer CL. 2008.
Lymphoid enhancer factor 1-mediated Wnt signaling pro-
motes the initiation of trophoblast lineage differentiation in
mouse embryonic stem cells. Stem Cells 26:842849.
Hill TP, Spater D, Taketo MM, Birchmeier W, Hartmann C. 2005.
Canonical Wnt/beta-catenin signaling prevents osteoblasts
from differentiating into chondrocytes. Dev Cell 8:727738.
Holleville N, Matéos S, Bontoux M, Bollerot K, Monsoro-Burq
AH. 2007. Dlx5 drives Runx2 expression and osteogenic
differentiation in developing cranial suture mesenchyme.
Dev Biol. 304:860874.
ELF-MF Inhibits Adipogenesis of Human MSCs 11
Bioelectromagnetics
Holmen SL, Zylstra CR, Mukherjee A, Sigler RE, Faugere MC,
Bouxsein ML, Deng L, Clemens TL, Williams BO. 2005.
Essential role of beta-catenin in postnatal bone acquisition.
J Biol Chem 280:2116221168.
Kersten S, Desvergne B, Wahli W. 2000. Roles of PPARs in health
and disease. Nature 405:421424.
Kubota N, Terauchi Y, Miki H, Tamemoto H, Yamauchi T,
Komeda K, Satoh S, Nakano R, Ishii C, Sugiyama T, Eto K,
Tsubamoto Y, Okuno A, Murakami K, Sekihara H, Hase-
gawa G, Naito M, Toyoshima Y, Tanaka S, Shiota K,
Kitamura T, Fujita T, Ezaki O, Aizawa S, Kadowaki T.
1999. PPAR gmediates high-fat diet-induced adipocyte
hypertrophy and insulin resistance. Mol Cell 4:597609.
Lefterova MI, Lazar MA. 2009. New developments in adipo-
genesis. Trends Endocrinol Metab 20:107114.
Ling L, Nurcombe V, Cool SM. 2009. Wnt signaling controls the
fate of mesenchymal stem cells. Gene 433:17.
Mangelsdorf DJ, Evans RM. 1995. The RXR heterodimers and
orphan receptors. Cell 83:841850.
Mayer-Wagner S, Passberger A, Sievers B, Aigner J, Summer B,
Schiergens TS, Jansson V, Müller PE. 2011. Effects of low
frequency electromagnetic fields on the chondrogenic
differentiation of human mesenchymal stem cells. Bioelec-
tromagnetics 32:283290.
Nichols TW, Jr. 2012. Mitochondria of mice and men: Moderate
magnetic fields in obesity and fatty liver. Med Hypotheses
79:287293.
Nie Y, Du L, Mou Y, Xu Z, Weng L, Du Y, Zhu Y, Hou Y, Wang T.
2013. Effect of low frequency magnetic fields on melanoma:
Tumor inhibition and immune modulation. BMC Cancer 13:582.
Noer A, Boquest AC, Collas P. 2007. Dynamics of adipogenic
promoter DNA methylation during clonal culture of human
adipose stem cells to senescence. BMC Cell Biol 8:18.
Pi-Sunyer X. 2009. The medical risks of obesity. Postgrad Med
121:2133.
Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R,
Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak
DR. 1999. Multilineage potential of adult human mesenchy-
mal stem cells. Science 284:143147.
Rao TP, Kühl M. 2010. An updated overview on Wnt signaling
pathways: A prelude for more. Circ Res 106:17981806.
Rosen E, Eguchi J, Xu Z. 2009. Transcriptional targets in
adipocyte biology. Expert Opin Ther Targets 13:975986.
Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS,
Spiegelman BM, Mortensen RM. 1999. PPAR gis required
for the differentiation of adipose tissue in vivo and in vitro.
Mol Cell 4:611617.
Rosen ED, Spiegelman BM. 2001. PPARg: A nuclear regulator of
metabolism, differentiation, and cell growth. J Biol Chem
12:3773137734.
Rubin CT, McLeod KJ, Lanyon LE. 1989. Prevention of osteopo-
rosis by pulsed electromagnetic fields. J Bone Joint Surg
Am 71:411417.
Schwartz Z, Simon BJ, Duran MA, Barabino G, Chaudhri R,
Boyan BD. 2008. Pulsed electromagnetic fields enhance
BMP-2 dependent osteoblastic differentiation of human
mesenchymal stem cells. J Orthop Res 26:12501255.
Skerry TM, Pead MJ, Lanyon LE. 1991. Modulation of bone loss
during disuse by pulsed electromagnetic fields. J Orthop
Res 9:600608.
Taipaleenmaki H, Abdallah BM, AlDahmash A, Saamanen AM,
Kassem M. 2011. Wnt signalling mediates the cross-talk
between bone marrow derived pre-adipocytic and
pre-osteoblastic cell populations. Exp Cell Res 317:745
756.
Takeda S, Elefteriou F, Karsenty G. 2003. Common endocrine
control of body weight, reproduction, and bone mass. Annu
Rev Nutr 23:403411.
Tchoukalova Y, Koutsari C, Jensen M. 2007. Committed subcuta-
neous preadipocytes are reduced in human obesity. Diabeto-
logia 50:151157.
Tominaga S, Yamaguchi T, Takahashi S, Hirose F, Osumi T. 2005.
Negative regulation of adipogenesis from human mesenchy-
mal stem cells by Jun N-terminal kinase. Biochem Biophys
Res Commun 326:499504.
Tong Z, Duncan RL, Jia X. 2013. Modulating the behaviors of
mesenchymal stem cells via the combination of high-
frequency vibratory stimulations and fibrous scaffolds.
Tissue Eng Part A 19:18621878.
Tontonoz P, Hu E, Spiegelman BM. 1994. Stimulation of adipo-
genesis in fibroblasts by PPAR g2, a lipid-activated
transcription factor. Cell 79:11471156.
Wang T, Nie Y, Zhao S, Han Y, Du Y, Hou Y. 2011. Involvement
of midkine expression in the inhibitory effects of low-
frequency magnetic fields on cancer cells. Bioelectromag-
netics 32:443452.
Yang Y, Tao C, Zhao D, Li F, Zhao W, Wu H. 2010. EMF acts on
rat bone marrow mesenchymal stem cells to promote
differentiation to osteoblasts and to inhibit differentiation to
adipocytes. Bioelectromagnetics 31:277285.
12 Du et al.
Bioelectromagnetics
... In recent times, there have been several studies focusing on the effects of electromagnetic waves on cell differentiation, specifically in the inhibition of adipogenesis for the treatment of obesity. In vitro research conducted by Du et al. (2014) Concluded that electromagnetic wave exposure can induce osteogenesis and inhibit the differentiation of mesenchymal stem cells to become adipocyte cells (Du et al., 2014). Other studies conducted by Lu et al. (2015) Also support the same thing, namely exposure to electromagnetic waves can suppress the expression of transcription factors that support adipogenesis (Lu et al., 2015). ...
... In recent times, there have been several studies focusing on the effects of electromagnetic waves on cell differentiation, specifically in the inhibition of adipogenesis for the treatment of obesity. In vitro research conducted by Du et al. (2014) Concluded that electromagnetic wave exposure can induce osteogenesis and inhibit the differentiation of mesenchymal stem cells to become adipocyte cells (Du et al., 2014). Other studies conducted by Lu et al. (2015) Also support the same thing, namely exposure to electromagnetic waves can suppress the expression of transcription factors that support adipogenesis (Lu et al., 2015). ...
... The inhibition of adipogenesis due to PEMF exposure is supported by research conducted by Du et al. (2014) in this study, the results were obtained that PEMF exposure with a frequency of 7.5 Hz, magnetic field magnitude 0.4 T and exposure time of 2 h/day for 15 days of adipogenesis can inhibit the expression of adipogenic genes. However, in this study, checking of gene expression was only carried out on the last day of exposure, i.e., on the 15 th day so it could not be seen how the pattern of gene expression occurred during the 15 days of the exposure time. ...
... Extremely low frequency magnetic fields (ELF-MF) also is capable to inhibit the effect of obesity. Inhibitory effect of ELF-MF on obesity may be related to the differentiation of MSCs to adipocytes [5]. Adipogenesis of MSCs expressed lipoprotein lipase (LPL). ...
... Our previous study showed that PEMF exposure may have potent effect to inhibit adipogenic differentiation indicated by PPARγ and ADIPOQ mRNA expression [9-10]. Extremely low frequency magnetic fields (ELF-MF) also inhibited adipogenic differentiation of ASCs showed by decreasing proliferator-activated receptor 2 (PPARγ2) expression but had no effect on osteogenic differentiation [5]. ELF-MF regulates JNK-dependent intracellular signaling in MSCs [18]. ...
... ELF-MF regulates JNK-dependent intracellular signaling in MSCs [18]. JNK signaling is essential in the bone formation [5]. However, no data of osteogenesis were recorded in this study. ...
Article
Full-text available
Pulsed electromagnetic fields (PEMFs) have an important role in cell differentiation. Previous study reported that PEMFs had positive and negative effect towards cell differentiation that depends on their frequencies applied to the cells. Human adipose-derived stem cells (ASCs) are mesenchymal stem cells that have an ability to differentiate into several types of cell including adipocytes, chondrocytes and osteocytes. This study aimed to evaluate the effect of human ASCs towards their adipogenic differentiation during PEMFs exposure. Human ASCs were isolated from adipose tissue. The cells then cultured in specific medium of adipocyte that induced ASCs differentiation along with PEMFs exposure. The maximum magnetic field used is 2 mT with a frequency of 75 Hz. To confirm the effect of PEMFs exposure towards adipogenic differentiation, mRNA expression of lipo protein lipase (LPL) was measured in mRNA expression level. The results showed that ASCs cultured on adipogenic differentiation without PEMFs exposure gradually increased LPL expression until day 14 of observation, while ASCs with PEMFs exposure significantly decreased LPL expression from day 2 to day 14. Based on the results, we concluded that PEMFs exposure can inhibit LPL expression that suppressed adipogenic differentiation.
... It was also shown that exposure to a rotating MF with a frequency of 7.5 Hz and an induction of 400 mT 2 hours a day for 15-21 days suppresses the differentiation of MSC along the adipogenic pathway through the JNK/Wnt/PPARγ2 signaling pathway, but MF does not affect osteogenic differentiation of MSC [52]. However, other researchers have not found a noticeable effect of a rotating MF with similar parameters on the processes of regeneration and remodeling of bone tissue [53]. ...
Article
Full-text available
Relevance. Magnets have long been used to treat various diseases, especially in inflammatory processes. According to existing historical data, magnetotherapy was already used in ancient times by the Chinese, Egyptians and Greeks. Different magnetic field strengths affect cells in different ways, with medium-strength magnetic fields being the most widely used. The review presents a brief history and current state of the issue of using a magnetic field in bone tissue regeneration. Modern knowledge about the mechanisms of physiological and reparative regeneration, restoration of bone tissue is clarified, and modern areas of bone tissue engineering are considered, taking into account the characteristics of microcirculation and the effect of a magnetic field on the physiology of bone tissue and reparative regeneration. One of the key findings of the review is that the magnetic field improves bone tissue repair by influencing the metabolic behavior of cells. Studies show that magnetotherapy promotes the activation of cellular processes, accelerates the formation of new bone tissue and improves its quality. It is also noted that the magnetic field has a positive effect on microcirculation, improving the blood supply to tissues and facilitating a better supply of nutrients to the site of injury. This contributes to faster wound healing and early rehabilitation of patients. Conclusion. Magnetotherapy is one of the effective physical and rehabilitation methods of treatment that will become increasingly important in modern medicine. However, further research is needed to better understand the mechanisms of action of a magnetic field on bone tissue and to determine the optimal parameters for its application.
... Consistent with our study it has been found that EMFs of 7.5 Hz, 0.4 T inhibited the adipogenic differentiation of MSCs via JNKdependent Wnt signaling pathway [27].Our findings were consistent with the results of a study by Shahbazi et al.; it was concluded that ELF-EMF increases the survival and proliferation of mesenchymal stem cells with intensities of 0.5 and 1 ms Tesla [28]. Consistent with the present study results, it was found that the proliferation of neuronal stem cells under magnetic field irradiation increased by 1 mT and 50 Hz for 6 to 24 h [29]. ...
Article
Full-text available
Introduction Exposure to pulsed electromagnetic field (PEMF) has been revealed to affect the differentiation and proliferation of human mesenchymal stem cells derived from dental pulp multipotent stromal stem cells (DP-MSCs). This study aimed to investigate the differentiation effect of electromagnetic fields (EMFs) on the DP-MSC. Materials and Methods PEMF was produced by a system comprising a multi-meter autotransformer, solenoid coils, and teslameter. This study included 10 groups of DP-MSCs which underwent different electromagnetic radiation time and beam intensity. Three samples tested for each group. The effect of PEMF with the intensity of 0.5 and 1 mT (mili Tesla) and 50 Hz on the proliferation rate of DP-MSC was evaluated at 20 and 40 minutes per day for seven days. MTT assay was applied to determine the growth and proliferation of DP-MSC. Gene expression of DMP1 for differentiation of DPSCs to odontoblasts was confirmed by Real Time PCR., ANOVA statistical analysis and Kruskal-Wallis test were used to analyze the data. Results The survival in all exposure groups was significantly higher than that in control except in the group of 40 minutes, 1 mT (P<0.05). In 20 minutes, 0.5 mT exposure, the survival intensity is significantly more than others (P<0.05). In general, the intensity of survival was recorded, 20, 0.5 mT≥20, 1 mT≥40, 0.5 mT≥40, 1 mT respectively. Therefore, according to the obtained results, ELF-EMF increases the survival of cells except for one case (40 minutes, 1 mT), even though the effective underlying mechanisms in this process are still unclear. Conclusions The results obtained promise that in the future, by placing an important part of the pulp next to the electromagnetic field, the lost part of the pulp can be reconstructed and the dentin barrier can be created.
... inhibition of adipogenic differentiation of mesenchymal stem cells, but there is no effect on osteogenic differentiation with ELF-EMF (Du et al., 2014). The ELF-EMFs can affect the expression of some genes while exerting no obvious influence on others. ...
Article
Full-text available
During the past decades, the potential effects of extremely low-frequency electromagnetic fields (ELF-EMFs) on human health have gained great interest all around the world. Though the International Commission on Non-Ionizing Radiation Protection recommended a 100 μT, and then a 200 μT magnetic field limit, the long-term effects of ELF-EMFs on organisms and systems need to be further investigated. It was reported that both electrotherapy and possible effects on human health could be induced under ELF-EM radiation with varied EM frequencies and fields. This present article intends to systematically review the in vivo experimental outcome and the corresponding mechanisms to shed some light on the safety considerations of ELF-EMFs. This will further advance the subsequent application of electrotherapy in human health.
... (Carpenter, 2019;Lee et al., 2012;Maffei, 2022). To date, ELF-MF have been shown to alter some cell functions such as redox homeostasis, proliferation, differentiation, and adhesion (Baek et al., 2014;Du et al., 2014;Nezamtaheri et al., 2022;Ross, 2017;Sulpizio et al., 2011;Wolf et al., 2005), but the molecular mechanism regulating the pathogenic effects remains elusive. ...
Article
Full-text available
Extremely low-frequency electromagnetic fields (ELF-MF) can modify the cell viability and regulatory processes of some cell types, including breast cancer cells. Breast cancer is a multifactorial disease where a role for ELF-MF cannot be excluded. ELF-MF may influence the biological properties of breast cells through molecular mechanisms and signaling pathways that are still unclear. This study analyzed the changes in the cell viability, cellular morphology, oxidative stress response and alteration of proteomic profile in breast cancer cells (MDA-MB-231) exposed to ELF-MF (50 Hz, 1 mT for 4 h). Non-tumorigenic human breast cells (MCF-10A) were used as control cells. Exposed MDA-MB-231 breast cancer cells increased their viability and live cell number and showed a higher density and length of filopodia compared with the unexposed cells. In addition, ELF-MF induced an increase of the mitochondrial ROS levels and an alteration of mitochondrial morphology. Proteomic data analysis showed that ELF-MF altered the expression of 328 proteins in MDA-MB-231 cells and of 242 proteins in MCF-10A cells. Gene Ontology term enrichment analysis demonstrated that in both cell lines ELF-MF exposure up-regulated the genes enriched in "focal adhesion" and "mitochondrion". The ELF-MF exposure decreased the adhesive properties of MDA-MB-231 cells and increased the migration and invasion cell abilities. At the same time, proteomic analysis, confirmed by Real Time PCR, revealed that transcription factors associated with cellular reprogramming were upregulated in MDA-MB-231 cells and downregulated in MCF-10A cells after ELF-MF exposure. MDA-MB-231 breast cancer cells exposed to 1 mT 50 Hz ELF-MF showed modifications in pro-teomic profile together with changes in cell viability, cellular morphology, oxidative stress response, adhesion, migration and invasion cell abilities. The main signaling pathways involved were relative to focal adhesion, mitochondrion and cellular reprogramming.
... As one of the common variable magnetic fields, a rotating magnetic field (RMF) is a magnetic field that changes the direction of the magnetic field at a constant angular rate and period, which is typically generated by the circling motion of permanent magnets driven by a motor [46]. Until now, RMF has rarely been used in biomedical research, but various bioeffects of RMF have been discovered, including adjusting cell differentiation [47], regulating cytokine production [48], increasing bone mineral density [49], ameliorating oxidative stress in the heart [50,51], and improving the heat distribution of the body surface [52]. Currently, there is less data on the impact of MFs on AS disease in the available literature. ...
Article
Full-text available
Ankylosing spondylitis (AS) is clinically characterized by bone fusion that is induced by the pathological formation of extra bone. Unfortunately, the fundamental mechanism and related therapies remain unclear. The loss of SHP-2 (encoded by Ptpn11) in CD4-Cre;Ptpn11f/f mice resulted in the induction of AS-like pathological characteristics, including spontaneous cartilage and bone lesions, kyphosis, and arthritis. Hence, this mouse was utilized as an AS model in this study. As one of the basic physical fields, the magnetic field (MF) has been proven to be an effective treatment method for articular cartilage degeneration. In this study, the effects of a rotating magnetic field (RMF; 0.2 T, 4 Hz) on an AS-like mouse model were investigated. The RMF treatment (2 h/d, 0.2 T, 4 Hz) was performed on AS mice from two months after birth until the day before sampling. The murine specimens were subjected to transcriptomics, immunomics, and metabolomics analyses, combined with molecular and pathological experiments. The results demonstrated that the mitigation of inflammatory deterioration resulted in an increase in functional osteogenesis and a decrease in dysfunctional osteolysis due to the maintenance of bone homeostasis via the RANKL/RANK/OPG signaling pathway. Additionally, by regulating the ratio of CD4+ and CD8+ T-cells, RMF treatment rebalanced the immune microenvironment in skeletal tissue. It has been observed that RMF interventions have the potential to alleviate AS, including by decreasing pathogenicity and preventing disease initiation. Consequently, RMF, as a moderately physical therapeutic strategy, could be considered to alleviate the degradation of cartilage and bone tissue in AS and as a potential option to halt the progression of AS.
... Although the investigation showed no visible morphological differences were observed between the DE, IE and control (NC) groups, there was noticeable improvement in the speed at which confluence is achieved. In addition, the MF treated cells showed similar morphology to untreated cells (NC), as they maintained the typical appearance and morphology of healthy hUC-MSCs (fibroblast-like and spindle-like shaped) appearance which is in agreement with the studies reported by Marędziak et al. 47 , and Du et al. 48 . This, therefore, suggests that direct exposure to MF may provide a potent method for increasing propagation speed whilst maintaining a normal morphological structures. ...
Article
Full-text available
Despite the extensive reports on the potential hazard of magnetic field (MF) exposures on humans, there are also concurrently reported on the improved proliferative property of stem cells at optimum exposure. However, the effect on mesenchymal stem cells (MSCs) remains unknown. Therefore, we aimed to investigate the impact of induced static MF (SMF) on human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) using Samarium Cobalt (SmCO5). At passage 3, hUC-MSCs (1 × 104) were exposed to 21.6 mT SMF by a direct exposure (DE) showed a significantly higher cell count (p < 0.05) in the growth kinetics assays with the shortest population doubling time relative to indirect exposure and negative control. The DE group was committed into the cell cycle with increased S phase (55.18 ± 1.38%) and G2/M phase (21.75 ± 1.38%) relative to the NC group [S-phase (13.54 ± 2.73%); G2/M phase (8.36 ± 0.28%)]. Although no significant changes were observed in the immunophenotype, the DE group showed an elevated expression of pluripotency-associated markers (OCT4, SOX2, NANOG, and REX1). These results suggest that the MFs could potentially induce proliferation of MSCs, a promising approach to promote stem cells propagation for clinical therapy and research without compromising the stemness of hUC-MSCs.
Article
Extremely low-frequency electromagnetic fields (ELF-MF) can modify the cell viability and regulatory processes of some cell types, including breast cancer cells. Breast cancer is a multifactorial disease where a role for ELF-MF cannot be excluded. ELF-MF may influence the biological properties of breast cells through molecular mechanisms and signaling pathways that are still unclear. This study analyzed the changes in the cell viability, cellular morphology, oxidative stress response and alteration of proteomic profile in breast cancer cells (MDA-MB-231) exposed to ELF-MF (50 Hz, 1 mT for 4 h). Non-tumorigenic human breast cells (MCF-10A) were used as control cells. Exposed MDA-MB-231 breast cancer cells increased their viability and live cell number and showed a higher density and length of filopodia compared with the unexposed cells. In addition, ELF-MF induced an increase of the mitochondrial ROS levels and an alteration of mitochondrial morphology. Proteomic data analysis showed that ELF-MF altered the expression of 328 proteins in MDA-MB-231 cells and of 242 proteins in MCF-10A cells. Gene Ontology term enrichment analysis demonstrated that in both cell lines ELF-MF exposure up-regulated the genes enriched in "focal adhesion" and "mitochondrion". The ELF-MF exposure decreased the adhesive properties of MDA-MB-231 cells and increased the migration and invasion cell abilities. At the same time, proteomic analysis, confirmed by Real Time PCR, revealed that transcription factors associated with cellular reprogramming were upregulated in MDA-MB-231 cells and downregulated in MCF-10A cells after ELF-MF exposure. MDA-MB-231 breast cancer cells exposed to 1 mT 50 Hz ELF-MF showed modifications in proteomic profile together with changes in cell viability, cellular morphology, oxidative stress response, adhesion, migration and invasion cell abilities. The main signaling pathways involved were relative to focal adhesion, mitochondrion and cellular reprogramming.
Article
As one of the common variable magnetic fields, rotating magnetic field (RMF) plays a crucial role in modern human society. The biological effects of RMF have been studied for over half a century, and various results have been discovered. Several reports have shown that RMF can inhibit the growth of various types of cancer cells in vitro and in vivo and improve clinical symptoms of patients with advanced cancer. It can also affect endogenous opioid systems and rhythm in central nerve systems, promote nerve regeneration and regulate neural electrophysiological activity in the human brain. In addition, RMF can influence the growth and metabolic activity of some microorganisms, alter the properties of fermentation products, inhibit the growth of some harmful bacteria and increase the susceptibility of antibiotic-resistant bacteria to common antibiotics. Besides, there are other biological effects of RMF on blood, bone, prenatal exposure, enzyme activity, immune function, aging, parasite, endocrine, wound healing, and plants. These discoveries demonstrate that RMF have great application potential in health care, medical treatment, fermentation engineering, and even agriculture. However, in some cases like pregnancy, RMF exposure may need to be avoided. Finally, the specific mechanisms of RMF's biological effects remain unrevealed, despite various hypotheses and theories. It does not prevent us from using it for our good.
Article
Full-text available
We previously found that the low frequency magnetic fields (LF-MF) inhibited gastric and lung cancer cell growth. We suppose that exposure to LF-MF may modulate immune function so as to inhibit tumor. We here investigated whether LF-MF can inhibit the proliferation and metastasis of melanoma and influence immune function. The effect of MF on the proliferation, cell cycle and ultrastracture of B16-F10 in vitro was detected by cell counting Kit-8 assay, flow cytometry, and transmission electron microscopy. Lung metastasis mice were prepared by injection of 2 x 105 B16-F10 melanoma cells into the tail vein in C57BL/6 mice. The mice were then exposed to an LF-MF (0.4 T, 7.5 Hz) for 43 days. Survival rate, tumor markers and the innate and adaptive immune parameters were measured. The growth of B16-F10 cells was inhibited after exposure to the LF-MF. The inhibition was related to induction of cell cycle arrest and decomposition of chromatins. Moreover, the LF-MF prolonged the mouse survival rate and inhibited the proliferation of B16-F10 in melanoma metastasis mice model. Furthermore, the LF-MF modulated the immune response via regulation of immune cells and cytokine production. In addition, the number of Treg cells was decreased in mice with the LF-MF exposure, while the numbers of T cells as well as dendritic cells were significantly increased. LF-MF inhibited the growth and metastasis of melanoma cancer cells and improved immune function of tumor-bearing mice. This suggests that the inhibition may be attributed to modulation of LF-MF on immune function and LF-MF may be a potential therapy for treatment of melanoma.
Article
Full-text available
Human mesenchymal stem cells (MSCs) are a promising candidate cell type for regenerative medicine and tissue engineering applications. Exposure of MSCs to physical stimuli favors early and rapid activation of the tissue repair process. In this study we investigated the in vitro effects of pulsed electromagnetic field (PEMF) treatment on the proliferation and osteogenic differentiation of bone marrow MSCs (BM-MSCs) and adipose-tissue MSCs (ASCs), to assess if both types of MSCs could be indifferently used in combination with PEMF exposure for bone tissue healing. We compared the cell viability, cell matrix distribution, and calcified matrix production in unstimulated and PEMF-stimulated (magnetic field: 2 mT, amplitude: 5 mV) mesenchymal cell lineages. After PEMF exposure, in comparison with ASCs, BM-MSCs showed an increase in cell proliferation (p<0.05) and an enhanced deposition of extracellular matrix components such as decorin, fibronectin, osteocalcin, osteonectin, osteopontin, and type-I and -III collagens (p<0.05). Calcium deposition was 1.5-fold greater in BM-MSC-derived osteoblasts (p<0.05). The immunofluorescence related to the deposition of bone matrix proteins and calcium showed their colocalization to the cell-rich areas for both types of MSC-derived osteoblast. Alkaline phosphatase activity increased nearly 2-fold (p<0.001) and its protein content was 1.2-fold higher in osteoblasts derived from BM-MSCs. The quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis revealed up-regulated transcription specific for bone sialoprotein, osteopontin, osteonectin, and Runx2, but at a higher level for cells differentiated from BM-MSCs. All together these results suggest that PEMF promotion of bone extracellular matrix deposition is more efficient in osteoblasts differentiated from BM-MSCs.
Article
Full-text available
We are interested in the in vitro engineering of artificial vocal fold tissues via the strategic combination of multipotent mesenchymal stem cells (MSCs), physiologically-relevant mechanical stimulations and biomimetic artificial matrices. We have constructed a vocal fold bioreactor capable of imposing vibratory stimulations on the cultured cells at human phonation frequencies. Separately, fibrous poly (ε-caprolactone) (PCL) scaffolds emulating the ligamentous structure of the vocal fold were prepared by electrospinning and were incorporated in the vocal fold bioreactor and were driven into a wave-like motion in an axisymmetrical fashion by the oscillating air. MSC-laden PCL scaffolds were subjected to vibrations at 200 Hz with a normal center displacement of ~40 μm for a total of 7 days. A continuous (CT) or a 1h-on-1h-off (OF) regime with a total dynamic culture time of 12 h per day were applied. The dynamic loading did not cause any physiological trauma to the cells. Immunohistotochemical staining revealed the reinforcement of the actin filament and the enhancement of α5β1 integrin expression under selected dynamic culture conditions. Cellular expression of essential vocal fold extracellular matrix (ECM) components, such as elastin, hyaluronic acid (HA) and matrix metalloproteinase-1 (MMP-1), was significantly elevated as compared to the static controls and the OF regime is more conducive to matrix production than the CT vibration mode. Analyses of genes of typical fibroblast hallmarks (tenascin-C, collagen III and procollagen I) as well as markers for MSC differentiation into non-fibroblastic lineages confirmed MSCs' adaptation of fibroblastic behaviors. Overall, the high frequency vibratory stimulation, when combined with a synthetic fibrous scaffold, serves as a potent modulator of MSC functions. The novel bioreactor system presented herein, as a versatile, yet well-controlled model, offers an in vitro platform for understanding vibration-induced mechanotransduction and for engineering of functional vocal fold tissues.
Article
Full-text available
In developed societies, chronic diseases such as diabetes, obesity, atherosclerosis and cancer are responsible for most deaths. These ailments have complex causes involving genetic, environmental and nutritional factors. There is evidence that a group of closely related nuclear receptors, called peroxisome proliferator-activated receptors (PPARs), may be involved in these diseases. This, together with the fact that PPAR activity can be modulated by drugs such as thiazolidinediones and fibrates, has instigated a huge research effort into PPARs1. Here we present the latest developments in the PPAR field, with particular emphasis on the physiological function of PPARs during various nutritional states, and the possible role of PPARs in several chronic diseases.
Article
One hundred and twenty-five patients with one hundred and twenty-seven ununited fractures of the tibial diaphysis were treated exclusively with pulsing electromagnetic fields. The over-all success rate in healing of the fracture with this surgically non-invasive out-patient method was 87 per cent. The success rate was not materially affected by the age or sex of the patient, the length of prior disability, the number of previous failed operations, or the presence of infection or metal fixation.
Article
The process of adipogenesis is known to involve the interplay of several transcription factors. Activation of one of these factors, the nuclear hormone receptor PPARγ, is known to promote fat cell differentiation in vitro. Whether PPARγ is required for this process in vivo has remained an open question because a viable loss-of-function model for PPARγ has been lacking. We demonstrate here that mice chimeric for wild-type and PPARγ null cells show little or no contribution of null cells to adipose tissue, whereas most other organs examined do not require PPARγ for proper development. In vitro, the differentiation of ES cells into fat is shown to be dependent on PPARγ gene dosage. These data provide direct evidence that PPARγ is essential for the formation of fat.
Article
During cartilage regeneration, proliferation and differentiation of new chondrocytes are required and towards this goal, in humans electromagnetic stimulation has been used in order to increase the spontaneous regenerative capacity of bone and cartilage tissue. In vivo tissue engineering has pointed out that the absence of an abundant source of cells accelerating the healing process is a limiting factor in the ability to repair articular cartilage. Considering that the umbilical cord is a viable alternative source of mesenchymal stem cells (MSC), our study evaluated the possibility of a combined use of Wharton's jelly - mesenchymal stem cells (WJ-MSCs) and pulsed electromagnetic field (PMEF). The first effect observed was that compared with the untreated cells, when the WJ-MSCs were treated with PMEF, there was an increase in the division of cells and a rapid increase in cell density and the morphological and biochemical data showed that the treatment with PMEF reduced the time to obtain chondrocyte cell differentiation and deposition of extracellular matrix. Taken together these data indicate the capacity of PEMF to induce early differentiation of WJ-MSCs cells towards cartilaginous tissue.
Article
The nuclear hormone receptor PPARγ promotes adipogenesis and macrophage differentiation and is a primary pharmacological target in the treatment of type II diabetes. Here, we show that PPARγ gene knockout results in two independent lethal phases. Initially, PPARγ deficiency interferes with terminal differentiation of the trophoblast and placental vascularization, leading to severe myocardial thinning and death by E10.0. Supplementing PPARγ null embryos with wild-type placentas via aggregation with tetraploid embryos corrects the cardiac defect, implicating a previously unrecognized dependence of the developing heart on a functional placenta. A tetraploid-rescued mutant surviving to term exhibited another lethal combination of pathologies, including lipodystrophy and multiple hemorrhages. These findings both confirm and expand the current known spectrum of physiological functions regulated by PPARγ.
Article
The present study addressed adult human mesenchymal stem cell (MSC) differentiation towards the osteoblastic lineage in response to alternating electric current, a biophysical stimulus. For this purpose, MSCs (chosen because of their proven capability for osteodifferentiation in the presence of select bone morphogenetic proteins) were dispersed and cultured within electric-conducting type I collagen hydrogels, in the absence of supplemented exogenous dexamethasone and/or growth factors, and were exposed to either 10 or 40 microampere alternating electric current for six hours per day. Under these conditions MSCs expressed both early (such as Runx-2 and osterix) and late (specifically, osteopontin and osteocalcin) osteogenic genes as a function of level, and duration of exposure to, alternating electric current. Compared to results obtained after 7 days, gene expression of osteopontin and osteocalcin (late-osteogenic genes) increased at day 14. In contrast, expression of these osteogenic markers from MSCs cultured under similar conditions and time periods, but not exposed to alternating electric current, did not increase as a function of time. Most importantly, expression of genes pertinent to the either the adipogenic (specifically, Fatty Acid Binding Protein-4) or chondrogenic (specifically, type II collagen) pathways was not detected when MSCs were exposed to the aforementioned alternating electric current conditions tested in the present study. The present research study was the first to provide evidence that alternating electric current promoted the differentiation of adult human mesenchymal stem cells towards the osteogenic pathway. Such an approach has the yet untapped potential to provide critically-needed differentiated cell supplies for cell-based assays and/or therapies for various biomedical applications.