Effect of Extremely Low Frequency Electromagnetic Field On MAP2 and Nestin Gene Expression of Hair Follicle Dermal Papilla Cells

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

IJAO

Int J Artif Organs 2016; 39(6): 294-299


DOI: 10.5301/ijao.5000512

ISSN 0391-3988 ORIGINAL RESEARCH ARTICLE

Effect of extremely low frequency electromagnetic field


on MAP2 and Nestin gene expression of hair follicle
dermal papilla cells
Marzie Moraveji1,2, Nooshin Haghighipour1, Hamid Keshvari2, Tannaz Nourizadeh Abbariki1, Mohammad Ali Shokrgozar1,
Amir Amanzadeh1
1
National Cell Bank of Iran, Pasteur Institute of Iran, Tehran - Iran
2
Department of Biomedical Engineering, Amirkabir University of Technology, Tehran - Iran

Abstract
Introduction: In recent years, the extremely low frequency electromagnetic field (ELF-EMF) has attracted a great
deal of scientific interest. The ELF-EMF signal is able to control ion transport across ion channels and therefore
induce cell differentiation.
Aim: The purpose of this study was to investigate the effect of ELF-EMF (50 Hz, 1 mT) on MAP2 and Nestin gene
expression of dermal papilla mesenchymal cells (DPCs).
Methods: In order to examine the effect of chemical and electromagnetic factors on gene expression, 4 experi-
mental groups, namely chemical (cell exposure to chemical signals), EMF (exposing cells to ELF-EMF), chemical-
EMF (subjecting cells to chemical signals and ELF-EMF) and control (with no treatment) groups, were prepared,
treated for 5 days, and studied. To assess the effect of extended test time on the expression of neural differentia-
tion markers (Nestin and MAP2), an EMF group was prepared and treated for a period of 14 consecutive days. The
beneficial role of EMF in inducing neural differentiation was shown by real-time PCR analysis.
Results: The higher expression of MAP2 after 14 days compared to that after 5 days and decrease of cell prolifera-
tion on days 5 to 20 were indicative of the positive effect of extending treatment time on neural differentiation
by evaluation of gene expression in EMF group.
Keywords: Chemical signaling, Differentiation, Extremely low frequency electromagnetic field, Necroptosis

Introduction stem cells (DPCs) play major roles in hair formation, growth
and cycling. Ectoderm is one of the germ layers, which de-
Cell sources and signaling factors are 2 main parts of neural velops into neural crest, skin, hair and epidermis. Because of
tissue engineering. A variety of cell types have been examined the common origin of hair and neural cells, they also express
for application in neural tissue engineering and cell therapy, some common genes (10). In this regard, DPCs, which are the
including embryonic stem cells, neural stem/progenitor cells cell source for hair formation and are also easily accessible in
(NSPCs) (1), induced pluripotent stem cells (iPSCs) (2), mes- the body, can be considered as an appropriate cell source for
enchymal stem cells isolated from umbilical cord blood (3), neural differentiation.
bone marrow (4, 5), adipose (6), placenta (7), bulge area (8) Although chemical factors are the most widely studied
as well as olfactory ensheathing cells (OECs) (9). Among these signaling factors in the literature, in recent years electrical
cell types, NSPCs have exhibited the highest potential for dif- and magnetic signals have been also reported to affect cell
ferentiating into functional neurons but they are not easily ac- behavior. EMF has been used to induce cells toward neural
cessible in the body, which limits their application in this field. (11-13), chondrogenic (14), osteogenic (15) and skeletal/
On the other hand, hair follicle dermal papilla mesenchymal cardiac muscle cell (16, 17) differentiation. The membrane
voltage depends on the type of parenchymal cell and the
stage of stem cell differentiation (18). EMF has been sug-
Accepted: July 5, 2016 gested to affect ion channels and the ion flow into or out of
Published online: August 10, 2016 the cell (19) and consequently changing the cell membrane
voltage. Briefly, EMF increases the intracellular calcium level
Corresponding author: which leads cell polarization (19).
Nooshin Haghighipour Among parenchymal cells, skeletal muscle cells, glia and
National Cell Bank of Iran
Pasteur Institute of Iran
neuron cells are associated with the most negative membrane
69 Pasteur Ave voltages, respectively (18), and therefore EMF can be used as
Tehran, Iran a tool to induce neural differentiation. The expression of cal-
haghighipour@pasteur.ac.ir cium channel, voltage-dependent, L type, alpha 1C subunit,

© 2016 Wichtig Publishing


Moraveji et al 295

(Cav1.2) subunit and Cav1.2 channel-mediated Ca2+influx have ELF-EMF device


been reported to be increased during neurogenesis in re-
sponse to the application of EMF (50 Hz) (20). Moreover, a Extremely low frequency-pulsed electromagnetic field
simultaneous activity of 2 subunits of voltage gated K+(Kv3) was used in this study using a device that was previously de-
channel (Kv3.4) and Cav1.2 channels during the formation of signed and fabricated at the National Cell Bank of Iran. It con-
pioneer axons and neuronal network has been reported else- sisted of a multi-turn solenoid, electrical current source, and
where (21). Therefore, EMF is able to induce cell polarization an operative to generate nonsinusoidal, pulsed electromag-
and increase membrane voltage, which will subsequently lead netic fields. This device was placed inside an incubator with
to the differentiation and maturation of neural cells. In one appropriate conditions for cell culture. Cells were exposed to
of the pioneer studies, Piacentini et al (20) reported the ef- EMF, with the frequency and flux density of 50 Hz and 1 mT,
fect of EMF (1 mT; 50 Hz) on inducing neural differentiation respectively (pulse period of 40 ms and Duty cycle (On/Off) of
with an increase in Cav1.2 channel activity, which leads to 25/15), for 7 hours per day in test periods of 5, 14 or 20 days
the expression of MAP2 and β-III-tubulin genes (20). Several (see Supplementary Figure S1, available online as supplemen-
studies have addressed the neuronal effect of 50 Hz ELF-EMF tary material at www.artificial-organs.com).
(11-13, 20, 22), indicating that at such a frequency, lower field
strengths are more beneficial. For instance, as opposed to a Experimental groups
field strength of 20 mT, which shows no neural differentiation
effect (22), low field strengths of 1 or 2 mT are effective in This study was designed to evaluate the expression of neural
inducing such differentiation (12, 13). genes after 5 consecutive days of treatment in 4 experimental
The aim of this research is to assess the neural differentia- groups including the control, chemical, EMF, and chemical-EMF
tion inducing effect of chemical, EMF and chemical-EMF sig- groups. In order to investigate the effect of extended test time
nals on DPCs by quantification and comparison of MAP2 and on mRNA levels of neural differentiation genes, an EMF group
Nestin gene expression levels in these experimental groups was prepared and treated for 14 consecutive days, and the re-
using real-time PCR. sults were compared with those of the control group.
In chemical tests, DPCs were cultured in DMEM supple-
Materials and methods mented with EGF (Gibco), bFGF (Gibco), and DMSO (ICN Bio-
medicals), while in the EMF group, cells were incubated with
Isolation of DP cells DMEM and exposed to an EM field (1 mT, 50 Hz sinusoidal,
7 hours per day). In the chemical-EMF experimental group,
DPCs were isolated from hair follicles of healthy men aged DPCs were incubated with chemical neural differentiation
30 to 35 years, according to a protocol previously described medium and subjected to the EM field. In the EMF and chem-
by Wu et al (23). Briefly, the biopsy obtained from each sub- ical-EMF groups, samples were placed at the center of a uni-
ject was rinsed with D-Hank’s balanced salt solution and then form field area. The percentage of CO2 and the temperature
cut into strips. Dermal subcutaneous fat dissection strips of the incubator were set to 5% and 37°C, respectively. During
were incubated with 0.3% dispase at 4°C for 16 to 18 hours. the test, the cell culture medium was replaced every 2 days.
Epithelial cells were isolated from the dermal sheath. In the Upon completion of tests, real-time PCR method was used to
final enzymatic step, dermal sheathes were incubated with study the gene expression levels of Nestin (neural progenitor
0.1% collagenase I in DMEM (Gibco) medium (supplemented cell marker) and MAP2 (mature neuron marker).
with 10% fetal bovine serum) at 37°C for 8 hours. After the
complete digestion of fibrous sheath, D-Hank’s was added Cell proliferation analysis
to stop the enzymatic process. Following centrifugation the
suspension and the complete separation of DPCs, these iso- Cell proliferation in the EM group was studied using MTT
lated cells were cultured in 0.1% gelatin-coated flasks and in- (3-(4, 5-dimethylthiazol-2yl)-2, 5-diphenyltetrazolium bro-
cubated with DMEM/F12 supplemented with 1% OPI (Gibco) mide) assay at days 5, 14 and 20. Furthermore, the optical
and 10% FBS. density of the colored solution was measured at day 0, which
corresponded to the control group. Upon completion of the
Characterization of DPCs test for each group, after removal of the culture medium and
rinsing once with PBS 1X, 100 μL of MTT solution was added
In order to characterize the isolated DPCs, the produc- to each well. After 4 hours, the MTT solution was replaced
tion of proteoglycans, glycosaminoglycans and alkaline with 100 μL of isopropanol for 20 minutes. Finally, light ab-
phosphatase was examined. Toluidine blue O (24), alcian sorption was measured at 545 and 630 nm wavelengths.
blue (24) and NBT/BCIP (25) were used to stain proteogly-
cans, glycosaminoglycans (GAG) and alkaline phosphatase, RNA isolation and reverse transcription polymerase
respectively. Briefly, for each staining, cells were fixed with chain reaction
0.1% glutaraldehyde in PBS, stained with the stain solution
and then washed with PBS. pH of alcian blue and toluidine The total RNA of each test group was extracted using RNeasy
blue O solutions were 2.5 and 2.0 to 2.5, respectively. In the Plus mini kit (Qiagen), and measured using a nanodrap instru-
case of toluidine blue O staining, after the mentioned steps, ment (NanoDrap). 1 μg of total RNA was utilized to synthesize
dehydration through 95% alcohol and then washing with cDNA by means of a Takara kit (Takara Bio USA). mRNA levels of
PBS were also performed. neural differentiation genes along with GAPDH as the reference

© 2016 Wichtig Publishing


296 Effect of ELF-EMF on neural differentation

Fig. 1 - Effect of ELF-EMF on cell prolif-


eration. Cell proliferation was decreased
over time, due to DPC differentiation
(p<0.05).

gene were evaluated in triplicate utilizing SYBR® Green Mas- measured. Results of this experiment depended on the dif-
ter Mix and ABI Step One real time-PCR instrument, both pur- ference in proliferation rate, as no cell death was observed.
chased from Applied Biosystems. Primer sequences, designed Based on the proliferation rate of cells exposed to EMF after
using Beacon Designer (BD) software and checked by primer ex- days 5, 14 and 20, a decrease in cell proliferation over time
press and gene runner software, are as follows: MAP2, forward compared to the control group was demonstrated (Fig. 1).
5’-ACGGCTGGAGAAGGCTGAA-3’ and reverse 5’-CCACG CT T This observation indicates more differentiation of cells with
GCTGTATTACTGTAGG-3’; Nestin, forward 5’-AAGAGAACC AGGA increasing test time.
GCCACTGAAG-3’ and reverse 5’- ACCTCCGTCGCTGTTGAGTCT-3’
and GAPDH, forward 5’- ACACCC AC T CCTCCACCTTTG-3’ and Morphology of DPCs
reverse 5’ TCCACCACCCTGTTGCTGTAG-3’. mRNA levels of genes
were quantified via comparative Ct method (2–ΔΔCt) and Ct val- Figure 2 shows cell morphology in the experimental
ues of each target marker were normalized relative to GAPDH. groups. As shown in Figures 2B and D, after 5 and 14 days in
Thereafter, resulting values were normalized to the obtained the EMF group the unipolar and multipolar morphologies of
results for the control group. neurons could be observed, respectively. In the chemical-EMF
group, a large number of dead cells were observed, which can
Statistical analysis be related to the death of neurons.

Results of 3 independent tests, carried out in triplicate Expression of neural genes


were expressed in the form of mean ± SD. Student’s t-test
was utilized for comparing the mean values corresponding to The expression of MAP2 and Nestin genes was stud-
experimental groups, by making p<0.05 assumption. ied using real-time PCR technique. According to the results
(Fig. 3A), the highest expression of MAP2 was observed in
Results the chemical group at the day 5, in comparison to the control
group, while no expression of this gene was observed in the
Histological analysis of DPCs chemical-EMF group.
The mRNA level of MAP2 in the EMF group was increased
Results of cell staining with toluidine blue O, alcian blue approximately 2.5- fold after 14 days in comparison to that
and NBT/BCIP demonstrated the production of proteoglycans, after 5 days. The highest expression of Nestin was observed
glycosaminoglycans and alkaline phosphatase, respectively in the chemical-EMF group, compared to the control group
(see Supplementary Figure S2, available online as supple- (Fig. 3B).
mentary material at www.artificial-organs.com). These results
confirmed the dermal papilla cell phenotype of isolated cells, Discussion
and therefore, they could be used in subsequent experiments.
In the present study, we demonstrated the effect of
Effect of ELF-EMF on cell proliferation ELF-EMF on neural differentiation by evaluation of MAP2
and Nestin gene expression. The simultaneous effects of
Using MTT assay, the rate of cell proliferation as well as chemical and electromagnetic signals were also studied
the metabolic events leading to apoptosis and necrosis were for the first time. Briefly, dermal papilla cells were exposed

© 2016 Wichtig Publishing


Moraveji et al 297

Fig. 2 - The morphology of the cells into four groups: (A) Chemical (5 days), (B) EMF (5 days), (C) Chemical-EM (5 days), (D) EM (14 days) and
(E) Control. In the EMF group, the morphologies of cells are unipolar and multipolar after 5 and 14 days, respectively. (Magnification = 200 x).

Fig. 3 - mRNA expression levels in


treatment groups. Real-time PCR data
of MAP2 (A) and Nestin (B) genes ex-
pression in experimental groups.
(*shows p<0.05)

to an electromagnetic field (50 Hz; 1 mT, 7 hours per day) pathway affects the programmed necrosis or necroptosis of
for 5 and 14 days. Meanwhile, the effects of chemical and differentiated neurons (35) (Fig. 4). Neurons are more sensi-
chemical-electromagnetic factors were also studied. The ex- tive to the levels of ROS but neural stem/progenitor cells have
pression levels of MAP2 and Nestin genes were quantified a high capacity of ROS, regulating their self-renewal and neu-
to compare the results of applying such signaling factors. rogenesis through the PI3K/Akt pathway (36). Therefore, cells
The effect of ELF-EMF on cell differentiation (26, 27), in the chemical-EMF may try to survive by transforming into
proliferation (28), migration (29), apoptosis (30) and other neural stem/progenitor cells.
intracellular activities has been previously studied, but the In a previous study, bone marrow-derived mesenchy-
mechanisms underlying such observed cellular responses as mal stem cells (BMSCs) were exposed to EMF (50 Hz;1 mT)
well as the corresponding signaling pathways are still unclear. (12), in which cell type and the time schedule of treatment
Among such studies, neural differentiation using EMF signal (application of EMF for 12 consecutive days), differed from
via reactive oxygen species (ROS)-mediated activation of epi- our study and the expression of MAP2 was increased by
dermal growth factor receptor (EGFR) has been reported (11). almost 1.5 times. The higher expression of MAP2 observed
Recently, the effect of ELF-EMF on cell reprogramming com- in our research compared to that in the mentioned work
bined with the octamer-binding transcription factor 4 (OCT4) may have originated from the duration of cell exposure to
has been proven (31). Thus, depending on the conditions, ELF- EMF, however the exact mechanisms underlying observa-
EMF can play different roles. According to the results of pres- tion of such different results are unclear. The difference
ent study, proliferation of DPCs is decreased over time, which may also arise from the different cell sources used in the
may be due to the differentiation of these cells into neural 2 studies, considering the fact that DPCs express more
cells such as neurons. common active genes with neural cells, and therefore can
Neuronal differentiation was evaluated by quantification exhibit a higher expression of MAP2 gene compared to
of Nestin and MAP2 expression levels. In EMF group, an in- BMSCs.
crease in MAP2 expression and no decrease in Nestin mRNA Cell communication, in particular between neurons,
level were observed at day 14 compared to those at day 5. is of great importance in neural differentiation and tissue
Increase in the expression of MAP2 can be assigned to an im- engineering. Indeed, there has been no successful work in
proved neural differentiation. neuronal differentiation and neuron cell therapy without
Inducing cells toward neural differentiation in the chem- achieving proper neuron-neuron signaling. Neurons can
ical-EMF group resulted in an increase in Nestin expression communicate with each other through chemical synapses
while no expression of MAP2 was observed. Considering the and the release of neurotransmitter molecules by a neuron
intracellular signaling pathway in this group, EMF (11) and EGF in the synaptic cleft space that binds to their receptors on
(32, 33) simultaneously produce ROS, which activate a P13k/ the other cell. Such a release significantly depends on the
AKT/mTOR pathway (34). Being activated by 2 stimulators, this calcium concentration. Briefly, neurotransmitter molecules

© 2016 Wichtig Publishing


298 Effect of ELF-EMF on neural differentation

Disclosures
Financial support: This work was supported by the National Cell
Bank in the Pasteur Institute of Iran.
Conflict of interest: None of the authors has financial interest
related to this study to disclose.

References
1. Marques SA, Almeida FM, Fernandes AM, et al. Predifferenti-
ated embryonic stem cells promote functional recovery after
spinal cord compressive injury. Brain Res. 2010;1349:115-128.
2. Mothe AJ, Tator CH. Review of transplantation of neural stem/
progenitor cells for spinal cord injury. Int J Dev Neurosci. 2013;
31(7):701-713.
3. Hou L, Cao H, Wang D, et al. Induction of umbilical cord blood
mesenchymal stem cells into neuron-like cells in vitro. Int J
Hematol. 2003;78(3):256-261.
4. Naghdi M, Tiraihi T, Namin SA, Arabkheradmand J. Transdiffer-
entiation of bone marrow stromal cells into cholinergic neuro-
nal phenotype: a potential source for cell therapy in spinal cord
injury. Cytotherapy. 2009;11(2):137-152.
5. Yazdani SO, Hafizi M, Zali AR, et al. Safety and possible outcome
assessment of autologous Schwann cell and bone marrow mes-
enchymal stromal cell co-transplantation for treatment of pa-
tients with chronic spinal cord injury. Cytotherapy. 2013;15(7):
782-791.
6. Zhou Z, Chen Y, Zhang H, et al. Comparison of mesenchymal
stromal cells from human bone marrow and adipose tissue for
the treatment of spinal cord injury. Cytotherapy. 2013;15(4):
Fig. 4 - A schematic of the proposed mechanism for neuron necrop- 434-448.
tosis. Effect of EMF and EGF on ROS production and necroptosis of 7. Portmann-Lanz CB, Schoeberlein A, Huber A, et al. Placen-
neuron and consequently no expression of MAP2 gene in chemical- tal mesenchymal stem cells as potential autologous graft for
EMF group. pre- and perinatal neuroregeneration. Am J Obstet Gynecol.
2006;194(3):664-673.
8. Amoh Y, Katsuoka K, Hoffman RM. The advantages of hair
are located inside the vesicles. Calcium ions bind to proteins follicle pluripotent stem cells over embryonic stem cells and
on the surface of vesicles and change their conformation, induced pluripotent stem cells for regenerative medicine. J
Dermatol Sci. 2010;60(3):131-137.
leading to the fusion of vesicles with cell membrane and the 9. Yazdani SO, Pedram M, Hafizi M, et al. A comparison between
release of neurotransmitter molecules (37). Therefore cal- neurally induced bone marrow derived mesenchymal stem
cium play major roles in cell communication and ELF-EMF cells and olfactory ensheathing glial cells to repair spinal cord
can be used as a tool to open calcium channels and facilitate injuries in rat. Tissue Cell. 2012;44(4):205-213.
the entrance of calcium ions. This mechanism implies the 10. Sadler TW. Langman’s medical embryology. 2011: Lippincott
beneficial role of using an electromagnetic wave as a dif- Williams & Wilkins.
ferential factor in neural tissue engineering. ELF-EMF was 11. Park J-E, Seo YK, Yoon HH, Kim CW, Park JK, Jeon S. Electromag-
able to result in cell differentiation and cell communication, netic fields induce neural differentiation of human bone mar-
simultaneously. row derived mesenchymal stem cells via ROS mediated EGFR
activation. Neurochem Int. 2013;62(4):418-424.
12. Cho H, Seo YK, Yoon HH, et al. Neural stimulation on human
Conclusions bone marrow-derived mesenchymal stem cells by extreme-
ly low frequency electromagnetic fields. Biotechnol Prog.
The results of the present paper indicate the significant 2012;28(5):1329-1335.
role of ELF-EMF in MAP2 and Nestin gene expression. Appli- 13. Ma Q, Deng P, Zhu G, et al. Extremely low-frequency electro-
cation of ELF-EMF is a low-cost method compared to the utili- magnetic fields affect transcript levels of neuronal differentia-
zation of chemical signaling factors. Contrary to expectations, tion-related genes in embryonic neural stem cells. PLoS One.
the results showed that the synergistic effect of the 2 signals 2014;9(3):e90041.
leads to no expression of the MAP2 gene. The exact intracel- 14. Chang CH, Loo ST, Liu HL, Fang HW, Lin HY. Can low frequen-
lular mechanisms underlying the results are still unclear and cy electromagnetic field help cartilage tissue engineering? J
require further investigation in the future. The findings of this Biomed Mater Res A. 2010;92(3):843-851.
15. Luo F, Hou T, Zhang Z, Xie Z, Wu X, Xu J. Effects of pulsed
work may be applied in neural tissue engineering to provide electromagnetic field frequencies on the osteogenic differ-
functional neural cells differentiated from suitable cell sourc- entiation of human mesenchymal stem cells. Orthopedics.
es, especially dermal papilla mesenchymal cells (DPCs), which 2012;35(4):e526-e531.
express some common genes with neural cells and are readily 16. Morabito C, Rovetta F, Bizzarri M, Mazzoleni G, Fanò G, Marig-
accessible in the body. giò MA. Modulation of redox status and calcium handling by

© 2016 Wichtig Publishing


Moraveji et al 299

extremely low frequency electromagnetic fields in C2C12 mus- 27. Mayer-Wagner S, Passberger A, Sievers B, et al. Effects of low
cle cells: A real-time, single-cell approach. Free Radic Biol Med. frequency electromagnetic fields on the chondrogenic differen-
2010;48(4):579-589. tiation of human mesenchymal stem cells. Bioelectromagnetics.
17. Bekhite MM, Figulla HR, Sauer H, Wartenberg M. Static mag- 2011;32(4):283-290.
netic fields increase cardiomyocyte differentiation of Flk-1+ 28. Monici M, Cialdai F. The Role of Physical Factors in Cell Differen-
cells derived from mouse embryonic stem cells via Ca2+ influx tiation, Tissue Repair and Regeneration. Rijeka, Croatia: INTECH
and ROS production. Int J Cardiol. 2013;167(3):798-808. Open Access Publisher 2012.
18. Levin M. The Physiology of Bioelectricity in Development, Tissue 29. Komazaki S, Takano K. Induction of increase in intracellular
Regeneration, and Cancer. Boca Raton, Florida: CRC Press 2011. calcium concentration of embryonic cells and acceleration of
19. Levin M. Bioelectric mechanisms in regeneration: unique aspects morphogenetic cell movements during amphibian gastrula-
and future perspectives. In: Seminars in cell & developmental bi- tion by a 50-Hz magnetic field. J Exp Zool A Ecol Genet Physiol.
ology. Amsterdam: Elsevier 2009. 2007;307(3):156-162.
20. Piacentini R, Ripoli C, Mezzogori D, Azzena GB, Grassi C. Ex- 30. Santini MT, Ferrante A, Rainaldi G, Indovina P, Indovina PL. Ex-
tremely low-frequency electromagnetic fields promote in vitro tremely low frequency (ELF) magnetic fields and apoptosis: a
neurogenesis via upregulation of Ca(v)1-channel activity. J Cell review. Int J Radiat Biol. 2005;81(1):1-11.
Physiol. 2008;215(1):129-139. 31. Baek S, Quan X, Kim S, Lengner C, Park JK, Kim J. Electromag-
21. Huang CY, Chu D, Hwang WC, Tsaur ML. Coexpression of high- netic fields mediate efficient cell reprogramming into a plurip-
voltage-activated ion channels Kv3.4 and Cav1.2 in pioneer axons otent state. ACS Nano. 2014;8(10):10125-10138.
during pathfinding in the developing rat forebrain. J Comp Neu- 32. Holmström KM, Finkel T. Cellular mechanisms and physiologi-
rol. 2012;520(16):3650-3672. cal consequences of redox-dependent signalling. Nat Rev Mol
22. Yan J, Dong L, Zhang B, Qi N. Effects of extremely low-frequency Cell Biol. 2014;15(6):411-421.
magnetic field on growth and differentiation of human mesen- 33. Huo Y, Qiu WY, Pan Q, Yao YF, Xing K, Lou MF. Reactive oxygen
chymal stem cells. Electromagn Biol Med. 2010;29(4):165-176. species (ROS) are essential mediators in epidermal growth fac-
23. Wu JJ, Liu RQ, Lu YG, Zhu TY, Cheng B, Men X. Enzyme digestion tor (EGF)-stimulated corneal epithelial cell proliferation, adhe-
to isolate and culture human scalp dermal papilla cells: a more sion, migration, and wound healing. Exp Eye Res. 2009;89(6):
efficient method. Arch Dermatol Res. 2005;297(2):60-67. 876-886.
24. Terry DE, Chopra RK, Ovenden J, Anastassiades TP. Differential 34. Kwon J, Lee SR, Yang KS, et al. Reversible oxidation and inacti-
use of Alcian blue and toluidine blue dyes for the quantification vation of the tumor suppressor PTEN in cells stimulated with
and isolation of anionic glycoconjugates from cell cultures: ap- peptide growth factors. Proc Natl Acad Sci USA. 2004;101(47):
plication to proteoglycans and a high-molecular-weight glyco- 16419-16424.
protein synthesized by articular chondrocytes. Anal Biochem. 35. Liu Q, Qiu J, Liang M, et al. Akt and mTOR mediate programmed
2000;285(2):211-219. necrosis in neurons. Cell Death Dis. 2014;5(2):e1084.
25. Jékely G, Arendt D. Cellular resolution expression profiling us- 36. Le Belle JE, Orozco NM, Paucar AA, et al. Proliferative neural
ing confocal detection of NBT/BCIP precipitate by reflection stem cells have high endogenous ROS levels that regulate self-
microscopy. Biotechniques. 2007;42(6):751-755. renewal and neurogenesis in a PI3K/Akt-dependant manner.
26. Manni V, Lisi A, Rieti S, et al. Low electromagnetic field (50 Hz) Cell Stem Cell. 2011;8(1):59-71.
induces differentiation on primary human oral keratinocytes 37. Neher E, Sakaba T. Multiple roles of calcium ions in the regula-
(HOK). Bioelectromagnetics. 2004;25(2):118-126. tion of neurotransmitter release. Neuron. 2008;59(6):861-872.

© 2016 Wichtig Publishing

You might also like