Int J App Pharm, Vol 16, Issue 3, 2024, 176-181Original Article

SINUSOIDAL ELECTROMAGNETIC FIELD DECREASES OSTEOGENIC DIFFERENTIATION OF RAT BONE MARROW MESENCHYMAL STEM CELLS

DHIYA ALTEMEMY1, MARYAM HAJI GHASEM KASHANI2*, OSAMAH N. WENAS3

1Department of Pharmaceutics, College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq. 2Department of Cellular and Molecular Biology, School of Biology and Institute of Biological Sciences, Damghan University, Damghan, Iran. 3Department of Pharmaceutical Chemistry, College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq. Osama
*Corresponding author: Maryam Haji Ghasem Kashani; *Email: kashani@du.ac.ir

‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬‬Received: 15 Jan 2024, Revised and Accepted: 09 Mar 2024‬‬‬‬‬‬


ABSTRACT

Objective: The widespread use of household electrical appliances generating electric and magnetic fields was a significant focus of WHO attention because of its serious threat to human health, especially osteogenesis. This research investigated the effect of 50 Hz frequency (1 mT intensity) sinusoidal EMF (SEMF) on the osteogenic differentiation of rat bone marrow stem cells (rBMSCs) in vitro.

Methods: Experimental groups were: positive control (cells cultured in osteogenic medium supplemented with 7-10 M Dexamethasone, negative control (cells cultured in α-MEM/10% FBS, 10 mmol Beta-Glycerol-Phosphate, 15% FBS, 50 ug/ml Ascorbic Acid bi-Phosphate, 100 unit/ml Penicillin) and for the EMF group, cells exposed to SEMF (50 Hz, 1 mT, 30 min/day) for 14 and 21 d. Alizarin red staining, Alkaline phosphatase activity, and QRT-PCR were performed.

Results: The EMF group exhibited weaker positive stains for ALP and Alizarin red than the positive control group. The Runx2 and Ocn gene expression levels were significantly decreased compared to negative control at 14 and 21 d of EMF exposure, respectively. After 14 and 21 d of exposure, Runx2 and Ocn gene expression were much lower in the EMF group than in the positive control group.

Conclusion: SEMF (1 mT, 50 Hz, 30 min/day) could retarded osteogenesis and reduce the osteogenic differentiation of rBMSCs.

Keywords: Sinusoidal electromagnetic field, Bone marrow stem cells, Runx2 and Ocn genes


INTRODUCTION

The spectrum of electromagnetic waves with extensive frequency range includes extremely low frequency (ELF), radio frequency, ultraviolet (U. V.), infrared, gamma, and X-rays. Bone marrow mesenchymal stem cells (BMSCs), known as bone marrow stromal cells (BMSCs) [1], increase rapidly in vitro, migrate, and differentiate into different tissues [2]. These cells can differentiate into the osteoblast lineage in vitro [3, 4]. Horwitz et al. showed that allogeneic bone marrow transplantation in kids increases bone mineral density with osteogenesis imperfect [2]. They are also a promising and abundant source for treating inherited diseases and repairing tissues such as cartilage, bone, and myocardium. In animal models, bone marrow cells were transplanted to skeletal and cardiac muscle, vascular endothelium, liver, lung, intestine, nerve tissue, and skin epithelium [5]. The function and Differentiation of MSCs depend on environmental factors and biophysical stimuli [6, 7]. As a biophysical agent, the pulsed electromagnetic field (PEMF) released Ca2+ions from the smooth endoplasmic reticulum [6]. Calcium entry and membrane depolarization lead to the expression of Ca2+-binding proteins, including calmodulin, in MSCs [8]. The rise in cytosolic Ca2+content initiates the wnt/β-catenin signaling pathway, which is essential in osteogenesis and stimulates bone-related genes such as Runx2 and Ocn, this improved MSC osteogenic differentiation [6, 8]. Bassett et al. (1982) devised a magnetic field across the broken site with Helmholtz coils to induce osteogenesis [9]. They suggested that applied external electrical energy could change the behavior of bone cells [10]. Gap junction intercellular communication was inhibited in Chinese lung cells and mouse fibroblasts by 24 h exposure to 50Hz EMF at 0.2-0.8 mT [11, 12].

It could be affected by EMF on plasma membrane gap junction proteins. During the proliferation phase of osteoblasts, ELF inhibited gap junction communication. However, ELF was relatively effective in preventing gap junction communication during the differentiation phase. ELF may only affect pre-osteoblasts or osteoblasts that are not fully developed [13].

In summary, the results of studies showed that EMF with different intensities and frequencies positively affects proliferation without affecting or reducing proliferation and increasing differentiation [14]. Following PEMF treatment, osteoblast proliferation, differentiation, and bone tissue-like formation increased [10]. Aaron et al. (1996) indicated that PEMF stimulates MSC differentiation, increasing bone maturation in the extracellular matrix [15]. Jansen et al. (2010) observed a high level of some signs of bone growth, like BMP2 (3-5 fold), osteoprotegerin (1-7 fold), and osteocalcin (2 fold) in BMSC post-induced by EMF [16]. However, different studies disagree on how PEMF stimulates the development of osteoblasts and the growth of cell lines in vitro [17], which depends on the waveform, frequency, intensity, duration of exposure, type, and cell age [16, 18]. Conflicting reports have been reported, making it difficult to draw any clear conclusion.

Because of the widespread use of electromagnetic devices nowadays, some gaps in knowledge about the bio-environmental effects of EMF have motivated many studies. Although different species are exposed to SEMF, there is no consensus about the effects of EMF on osteogenesis.

This study aimed to determine the effect of SEMF with an intensity of 1 mT and frequency of 50 Hz on the osteogenic differentiation of rat BMSCs.

MATERIALS AND METHODS

Magnetic field generator

This study investigated the effect of a magnetic field with an intensity of 1 mT and a frequency of 50 Hz on cell samples. The magnetic field must be the same over the entire sample space, and the magnetic flux lines must stay parallel. The device was designed according to a recent study [19].

rBMSCs isolation and culture

Femur and tibia bones were collected under sterile conditions from Wistar rats (6-8 w old) and situated in a culture dish that had been sterilized and was under a laminar hood. Research ethics at Damghan University was approved for all tests (IR. DU. REC.1400.014), and this study followed the National Institutes of Health Guide for the Care and Use of Laboratory Animals. (NIH Publication No. 23-80, revised 1996). Bone marrow was flushed into a Falcon tube and centrifuged at 1200 rpm for 5 min. After removing the supernatant, the cell pellets were grown in α-MEM (α-minimal essential medium; Gibco/BRL; cat. No. 52100-0.3) with 10% FBS (Gibco, cat. No. 12203C) added and left to grow. Every three days, the medium was changed. The cells were passaged at 70% to 80% confluency [20]. The stem cells were previously confirmed by Ghorbanian et al. (2012) [21, 22].

Experimental groups

rBMSCs at passage four were examined in three groups: positive control (cultured cells in osteogenic medium containing 7-10 M Dexamethasone, negative control (cultured cells in α-MEM containing 10% FBS), 50 ug/ml Ascorbic Acid bi-Phosphate, 10 mmol Beta-Glycerol-Phosphate, 15% FBS, 100 unit/ml Penicillin) and EMF (cells exposed to SEMF at a frequency of 50 Hz, intensity 1 mT, half an hour daily [23, 24].

Alizarin red staining

On day 14, the cells were stained with Alizarin Red (069K1639, Sigma-Aldrich). After removing the medium, the cells were washed twice with cold PBS. Each sample was set in 4% paraformaldehyde (Merck, Germany) that was kept out for 10 min at 4 °C. Following removing the fixative, the cells underwent two washes with PBS and were subsequently stained with 400 μl of Alizarin Red solution at a pH of 7.2 for 30 min. Later, the cells underwent two rounds of PBS rinsing and were subsequently examined under a microscope [25].

Alkaline phosphatase activity

On day 14 after induction, the cells' total protein was lysed using 30 μl of Triton X-100 lysis solution to assess the activity of alkaline phosphatase (ALP). While the solution was spinning at 2000 rpm and 4 °C for 10 min, the ALP activity of the supernatant was measured at 405 nm with a microplate reader from BioTek Instruments (USA). The phosphatase substrate used was para nitrophenyl phosphate (pNPP). The enzyme activity (I. U.) level was then compared to the amount of protein in the sample [26].

Real-time PCR

The qPCR technique evaluated genes and Runx2 in treated rBMSCs [27]. According to the manufacturer's instructions, The RNX-plus solution (Sinaclon, Iran) was used to extract RNA. The purity of the extracted RNA was examined using a spectrophotometer (Eppendorf Biop Hotometer). Samples with 260/230 ≥ 1.4 adsorbents and 260/280 ≥ 1.8 were analyzed. PrimeScriptTM 1st strand cDNA Synthesis Kit (Takara, Japan) synthesized cDNA. Rotor-Gene 6000 PCR machine with RealQ Plus Master Mix Green (Amplicon, Denmark) was used for real-time PCR. Primer sequences (table 1) were made with AlleleID software version 7.5 (Premierbiosoft, USA). In this study, the housekeeping gene Tbp was used as internal control, and the expression level of each gene was compared with Tbp. The relative expression of genes in the treatment groups compared to the control group was calculated using-2ΔΔct, and the reaction solution volume was tenμl in the end. Finally, the RT-qPCR reaction with the amplicon amplification program includes initial denaturation: 95 °C, 15 min, denaturation: 95 °C, 15 seconds, annealing and extension: 60 °C, 45 seconds, melt: 65-94 °C was performed [28].

Statistical analysis

SPSS software version 16 was used for statistical analysis. A one-way ANOVA test and a Tukey post-hoc test were used to see if there were significant differences between the test groups. The Independent-sample T-test was used to assess the significance of treated cells on days 14 and 21. P<0.05 was considered significant, and each experiment was conducted thrice.

RESULTS

Morphologies of cultured rat bone marrow-derived stem cells

Phase contrast images of rBMSCs are shown in fig. 1. The primary cultured rBMSCs were triangular or polyhedral in shape (A) and then exhibited spindle-shaped morphology. As the passage number increased, the blood cell density decreased, and the rBMSCs gradually changed into flat morphology (B).

Fig. 1: Phase contrast images of rBMSCs at P0 (A) and P3 (B) were shown (×100). The cells appeared flat morphology, and the blood cells were removed with increasing passage number

Fig. 2: Alizarin red staining after 14 d of treatment in experimental groups. A: Undifferentiated cells of the negative control group; B: Positive control group; C: EMF group

Alizarin red staining

Alizarin red staining was used to see how well rBMSCs turned into osteoblasts after 14 d of treatment. According to fig. 2, undifferentiated cells of the negative control group were observed (A). The positively stained regions were more in the positive control (B) than the EMF group (C).

Alkaline phosphatase activity

The results shown in fig. 3 (A-C) indicate higher mineralization in the positive control than in the EMF group.

Alkaline phosphatase activity in experimental groups was evaluated after 14 d of treatment. It can be seen in fig. 4 that the positive control group had more ALP activity than the negative control group (P<0.05). Not so with the EMF group; compared to the positive control, they showed a significant decrease (P<0.05).

Expression of osteogenesis-related genes, including Runx2 and Ocn

After 14 d of treatment, the Runx2 gene expression was analyzed using real-time PCR (fig. 5). The gene expression was significantly decreased in the EMF group compared with the negative and positive controls (P<0.05).

After 21 d of treatment, the positive control group showed a significant increase in gene expression compared to the negative control, and the EMF group showed a significant decrease in that compared to the positive control (P<0.05).

Finally, the expression levels of the Runx2 gene after 14 d were compared with those after 21 d. The Runx2 mRNA level of positive control on day 21 significantly increased compared to the same group on day 14 (P<0.05).

Fig. 3: Mineralization activity of experimental groups. A: Negative control (×400); B: Positive control (×400); C: EMF group (×400). The small calcified particles appeared as white dots

Fig. 4: Alkaline phosphatase activity in negative control, EMF, and positive control groups after 14 d of treatment. *significant increase versus negative control, # significant decrease versus positive control. Values are mean±SD n=3

Fig. 5: Relative expression of Runx2 mRNA levels in experimental groups. *P<0.05 versus negative control and #significant decrease versus positive control at the same time. ***significant difference compared to the same group on day 14. Values are mean±SD n= 3

According to fig. 6, after 14 d of treatment, Ocn gene expression compared to the negative control group increased significantly in the positive control group. It decreased significantly in the EMF group compared to the positive control (P<0.05).

After 21 d of treatment, the positive control group showed a significant increase in Ocn gene expression compared to the negative control, and the EMF group showed a significant decrease in that compared to the negative and positive controls (P<0.05).

Fig. 6: Relative expression of Ocn mRNA levels in experimental groups. *P<0.05 versus negative control and #significant decrease versus positive control at the same time. Values are mean±SD n= 3; finally, we examined how much the Ocn gene was expressed after 14 and 21 d. There wasn't a big difference between the groups in the amount of Ocn mRNA on day 21 compared to day 14 (P>0.05)

DISCUSSION

Devices generating extremely low-frequency fields have found wide applications in daily life. This highlights the need for further research to investigate critical mechanisms involved in osteogenesis. The findings suggest suitable solutions to deal with the destructive effects of EMF on bone formation. Osteogenesis is a complex set of events involving the differentiation of MSCs for new bone production [17, 29]. Lim et al. (2013) showed that extremely low frequency-PEMFs (ELF-PEMFs) at 50 and 100 Hz increased osteogenesis in human alveolar bone mesenchymal stem cells [29]. Also, Zhong et al. (2012) reported that EMF with a frequency of 50 Hz and an intensity of 0.5 mT accelerates cell proliferation and increases cell differentiation [30]. However, there have been adverse reports of the effects of electrical stimuli on cell differentiation proliferation and bone formation in vitro [9].

At the proliferation phase, the proliferation of rBMSCs was enhanced, while their differentiation was inhibited under exposure to EMF. But at the differentiation phase, it was vice versa [31].

This study investigated the effect of a sinusoidal electromagnetic field on extracellular matrix mineralization, alkaline phosphatase activity, and expression of Runx2 and Ocn osteogenic genes in rBMSCs.

The regions that responded positively to Alizarin red staining were lower in the EMF group than in the positive control group. These results contradict the results of Yang et al. (2010), who showed that SEMF with a frequency of 15 Hz and an intensity of 1 mT increased the formation of mineral nodules in rBMSCs [32]. Chang et al. (2004) showed that PEMF with a frequency of 15 Hz and an intensity of 0.1 mT for 14 d did not affect the mineral nodes of rat calvarial osteoblasts [9]. The effects of EMF on the proliferation and the Differentiation of MSCs are very different and sometimes inconsistent [14]. These effects depend on the waveform, frequency, intensity, exposure duration, type, and age of the cell [16, 18]. Our results showed alkaline phosphatase activity in the EMF group was significantly reduced compared to the positive control. This is similar to the results of Yang et al. (2008), who reported that PEMF with a frequency of 48 Hz and an intensity of 1.5 mT for 24 h significantly reduced alkaline phosphatase activity [33]. Schwarts et al. (2007) showed that PEMF at 15 Hz for 24 d did not affect ALP activity in BMSCs [34]. In contrast, Zhou et al. (2014) showed that SEMF with a frequency of 50 Hz, intensities of 1.8 and 3.6 mT, 30 min/day for 15 d increases matrix mineralization and alkaline phosphatase activity in rat osteoblasts [18]. The contrasting effects may be due to the use of different intensities and types of cells.

Runx2 expression level dropped considerably in the EMF group after 14 d of treatment versus the negative and positive controls and after 21 d of treatment compared to the positive control. This is pretty close to the outcomes of Tsai et al. (2009), who reported that PEMF with a frequency of 7.5 Hz, intensity of 0.13 mT, 2 h a day for 10 d leads to a significant reduction of Runx2 in MSCs [35]. In contrast, Yang et al. (2010) reported that SEMF with a frequency of 15 Hz and an intensity of 1 mT increased the expression of the Runx2 gene in rBMSCs [32]. The contrast between our results and the results of Yang et al. is probably due to the use of different frequencies.

Ocn gene expression reduced dramatically in the EMF group after 14 d of treatment compared to the positive control following 21 d of treatment compared to the negative and positive controls. These results are consistent with the results of Junsen et al. (2010), who reported that exposure to BMSCs at a frequency of 15 Hz, the intensity of 0.1 mT for 14 d, reduces the expression of Runx2 and Ocn genes [15] and Liu et al. (2014) stated decreased Runx2 and Ocn genes expression in BMSCs exposed to EMF with a frequency of 50 Hz and an intensity of 1 mT for 2 h/day for up to 14 d [36].

CONCLUSION

However, we could not identify the significance of the disparity between our result and the other findings. The discrepancies in results represent unidentified variations in laboratory techniques and materials. This study used sinusoidal EMF with a frequency of 50 Hz, intensity of 1 mT, and half an hour daily as a stimulus for osteogenic differentiation. SEMF with the mentioned features reduced the osteogenic differentiation of rBMSCs.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

All authors contributed equally to this work regarding the scope of the subject, the literature review and analysis, drafting, revision, editing, and final approval of the manuscript.

CONFLICT OF INTERESTS

The authors have no conflicts of interest.

REFERENCES

  1. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7(2):211-28. doi: 10.1089/107632701300062859, PMID 11304456.

  2. Campagnoli C, Roberts IA, Kumar S, Bennett PR, Bellantuono I, Fisk NM. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood. 2001;98(8):2396-402. doi: 10.1182/blood.v98.8.2396, PMID 11588036.

  3. Mohammed EEA, El-Zawahry M, Farrag ARH, Aziz NNA, Sharaf-ElDin W, Abu-Shahba N. Osteogenic differentiation potential of human bone marrow and amniotic fluid-derived mesenchymal stem cells in vitro and in vivo. Open Access Maced J Med Sci. 2019;7(4):507-15. doi: 10.3889/oamjms.2019.124, PMID 30894903.

  4. Sun LY, Hsieh DK, Yu TC, Chiu HT, Lu SF, Luo GH. Effect of pulsed electromagnetic field on the proliferation and differentiation potential of human bone marrow mesenchymal stem cells. Bioelectromagnetics. 2009;30(4):251-60. doi: 10.1002/bem.20472, PMID 19204973.

  5. Hassan HT, El-Sheemy M. Adult bone-marrow stem cells and their potential in medicine. J R Soc Med. 2004;97(10):465-71. doi: 10.1177/0141076809701003, PMID 15459256, PMCID Pmc1079613.

  6. Jazayeri M, Shokrgozar MA, Haghighipour N, Bolouri B, Mirahmadi F, Farokhi M. Effects of electromagnetic stimulation on gene expression of mesenchymal stem cells and repair of bone lesions. Cell J. 2017;19(1):34-44. doi: 10.22074/cellj.2016.4870, PMID 28367415, PMCID PMC5241516.

  7. Gong T, Lu L, Liu D, Liu X, Zhao K, Chen Y. Dynamically tunable polymer microwells for directing mesenchymal stem cell differentiation into osteogenesis. J Mater Chem B. 2015;3(46):9011-22. doi: 10.1039/c5tb01682g, PMID 32263032.

  8. Leone L, Podda MV, Grassi C. Impact of electromagnetic fields on stem cells: common mechanisms at the crossroad between adult neurogenesis and osteogenesis. Front Cell Neurosci. 2015;9:228. doi: 10.3389/fncel.2015.00228, PMID 26124705, PMCID Pmc4466452.

  9. Chang WH, Chen LT, Sun JS, Lin FH. Effect of pulse-burst electromagnetic field stimulation on osteoblast cell activities. Bioelectromagnetics. 2004;25(6):457-65. doi: 10.1002/bem.20016, PMID 15300732.

  10. Diniz P, Soejima K, Ito G. Nitric oxide mediates the effects of pulsed electromagnetic field stimulation on the osteoblast proliferation and differentiation. Nitric Oxide. 2002;7(1):18-23. doi: 10.1016/s1089-8603(02)00004-6, PMID 12175815.

  11. Zeng Q, Chiang H, Hu G, Mao G, Fu Y, Lu D. ELF magnetic fields induce internalization of gap junction protein connexin 43. In: Chinese hamster lung cells. Bioelectromagnetics: Journal of the Bioelectromagnetics Society. Society for Physical Regulation in Biology and Medicine, The European Bioelectromagnetics Association. 2003;24(2):134-8.

  12. Hu G, Chiang H, Zeng Q, Fu Y. ELF magnetic field inhibits gap junctional intercellular communication and induces hyperphosphorylation of connexin 43. In: NIH3T3 cells. Bioelectromagnetics: Journal of the Bioelectromagnetics Society. Society for Physical Regulation in Biology and Medicine, The European Bioelectromagnetics Association. 2001;22(8):568-73.

  13. Yamaguchi DT, Huang J, Ma D, Wang PK. Inhibition of gap junction intercellular communication by extremely low‐frequency electromagnetic fields in osteoblast‐like models is dependent on cell differentiation. J Cell Physiol. 2002;190(2):180-8. doi: 10.1002/jcp.10047, PMID 11807822.

  14. Safari M, Jadidi M, Baghian A, Hasanzadeh H. Proliferation and differentiation of rat bone marrow stem cells by 400-μT electromagnetic field. Neurosci Lett. 2016;612:1-6. doi: 10.1016/j.neulet.2015.11.044, PMID 26639423.

  15. Jansen JH, van der Jagt OP, Punt BJ, Verhaar JA, van Leeuwen JP, Weinans H. Stimulation of osteogenic differentiation in human osteoprogenitor cells by pulsed electromagnetic fields: an in vitro study. BMC Musculoskelet Disord. 2010;11(1):188. doi: 10.1186/1471-2474-11-188, PMID 20731873, PMCID PMC2936347.

  16. Maziarz A, Kocan B, Bester M, Budzik S, Cholewa M, Ochiya T. How electromagnetic fields can influence adult stem cells: positive and negative impacts. Stem Cell Res Ther. 2016;7(1):54. doi: 10.1186/s13287-016-0312-5, PMID 27086866, PMCID Pmc4834823.

  17. Sun LY, Hsieh DK, Lin PC, Chiu HT, Chiou TW. Pulsed electromagnetic fields accelerate proliferation and osteogenic gene expression in human bone marrow mesenchymal stem cells during osteogenic differentiation. Bioelectromagnetics. 2010;31(3):209-19. doi: 10.1002/bem.20550, PMID 19866474.

  18. Zhou J, Ming LG, Ge BF, Wang JQ, Zhu RQ, Wei Z. Effects of 50 Hz sinusoidal electromagnetic fields of different intensities on proliferation, differentiation and mineralization potentials of rat osteoblasts. Bone. 2011;49(4):753-61. doi: 10.1016/j.bone.2011.06.026. PMID 21726678.

  19. Tabatabai TS, Haji Ghasem Kashani M, Maskani R, Nasiri M, Nabavi Amri SA, Atashi A. Synergic effects of an extremely low-frequency electromagnetic field and betaine on in vitro osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells. In Vitro Cell Dev Biol Animal. 2021;57(4):468-76. doi: 10.1007/s11626-021-00558-6.

  20. Sun X, Su W, Ma X, Zhang H, Sun Z, Li X. Comparison of the osteogenic capability of rat bone mesenchymal stem cells on collagen, collagen/hydroxyapatite, hydroxyapatite and biphasic calcium phosphate. Regen Biomater. 2018;5(2):93-103. doi: 10.1093/rb/rbx018, PMID 29644091, PMCID PMC5888729.

  21. Taghi GM, Ghasem Kashani Maryam H, Taghi L, Leili H, Leyla M. Characterization of in vitro cultured bone marrow and adipose tissue-derived mesenchymal stem cells and their ability to express neurotrophic factors. Cell Biology International. 2012;36(12):1239-49. doi: 10.1042/cbi20110618, PMID 22994924.

  22. Noviantari A, Antarianto RD. Rif’ati L, Rinendyaputri R, Zainuri M, Dany F. The expression of nestin in the induced differentiation into neurons of rat bone marrow mesenchymal stem cells by neurotrophin-3 (NT-3). Int J Appl Pharm. 2020;12:44-9.

  23. Luo F, Hou T, Zhang Z, Xie Z, Wu X, Xu J. Effects of pulsed electromagnetic field frequencies on the osteogenic differentiation of human mesenchymal stem cells. Orthopedics. 2012;35(4):e526-31. doi: 10.3928/01477447-20120327-11, PMID 22495854.

  24. Esposito M, Lucariello A, Riccio I, Riccio V, Esposito V, Riccardi G. Differentiation of human osteoprogenitor cells increases after treatment with pulsed electromagnetic fields. In Vivo. 2012;26(2):299-304. PMID 22351673.

  25. Xu L, Liu Y, Sun Y, Wang B, Xiong Y, Lin W. Tissue source determines the differentiation potentials of mesenchymal stem cells: a comparative study of human mesenchymal stem cells from bone marrow and adipose tissue. Stem Cell Res Ther. 2017;8(1):275. doi: 10.1186/s13287-017-0716-x, PMID 29208029, PMCID PMC5718061.

  26. Jaquiery C, Schaeren S, Farhadi J, Mainil Varlet P, Kunz C, Zeilhofer HF. In vitro osteogenic differentiation and in vivo bone-forming capacity of human isogenic jaw periosteal cells and bone marrow stromal cells. Ann Surg. 2005;242(6):859-67. doi: 10.1097/01.sla.0000189572.02554.2c, PMID 16327496, PMCID Pmc1409890.

  27. Wang L, Dormer NH, Bonewald LF, Detamore MS. Osteogenic differentiation of human umbilical cord mesenchymal stromal cells in polyglycolic acid scaffolds. Tissue Eng Part A. 2010;16(6):1937-48. doi: 10.1089/ten.TEA.2009.0706, PMID 20070186.

  28. Ghali O, Broux O, Falgayrac G, Haren N, van Leeuwen JP, Penel G. Dexamethasone in osteogenic medium strongly induces adipocyte differentiation of mouse bone marrow stromal cells and increases osteoblast differentiation. BMC Cell Biol. 2015;16(1):9. doi: 10.1186/s12860-015-0056-6, PMID 25887471, PMCID PMC4359404.

  29. Lim K, Hexiu J, Kim J, Seonwoo H, Cho WJ, Choung PH. Effects of electromagnetic fields on osteogenesis of human alveolar bone-derived mesenchymal stem cells. BioMed Res Int. 2013;2013:296019. doi: 10.1155/2013/296019, PMID 23862141, PMCID PMC3703802.

  30. Zhong C, Zhang X, Xu Z, He R. Effects of low-intensity electromagnetic fields on the proliferation and differentiation of cultured mouse bone marrow stromal cells. Phys Ther. 2012;92(9):1208-19. doi: 10.2522/ptj.20110224, PMID 22577063.

  31. Wu H, Ren K, Zhao W, Baojian GE, Peng S. Effect of electromagnetic fields on proliferation and differentiation of cultured mouse bone marrow mesenchymal stem cells. J Huazhong Univ Sci Technolog Med Sci. 2005;25(2):185-7. doi: 10.1007/BF02873572, PMID 16116968.

  32. Yang Y, Tao C, Zhao D, Li F, Zhao W, Wu H. EMF acts on rat bone marrow mesenchymal stem cells to promote differentiation to osteoblasts and to inhibit differentiation to adipocytes. Bioelectromagnetics. 2010;31(4):277-85. doi: 10.1002/bem.20560, PMID 20041434.

  33. Wei Y, Xiaolin H, Tao S. Effects of extremely low-frequency-pulsed electromagnetic field on different-derived osteoblast-like cells. Electromagn Biol Med. 2008;27(3):298-311. doi: 10.1080/15368370802289604, PMID 18821205.

  34. Schwartz Z, Simon BJ, Duran MA, Barabino G, Chaudhri R, Boyan BD. Pulsed electromagnetic fields enhance BMP‐2 dependent osteoblastic differentiation of human mesenchymal stem cells. J Orthop Res. 2008;26(9):1250-5. doi: 10.1002/jor.20591, PMID 18404656.

  35. Tsai MT, Li WJ, Tuan RS, Chang WH. Modulation of osteogenesis in human mesenchymal stem cells by specific pulsed electromagnetic field stimulation. J Orthop Res. 2009;27(9):1169-74. doi: 10.1002/jor.20862, PMID 19274753, PMCID PMC2746855.

  36. Yu JZ, Wu H, Yang Y, Liu CX, Liu Y, Song MY. Osteogenic differentiation of bone mesenchymal stem cells regulated by osteoblasts under EMF exposure in a co-culture system. J Huazhong Univ Sci Technolog Med Sci. 2014;34(2):247-53. doi: 10.1007/s11596-014-1266-4, PMID 24710940.