In-vitro analysis of Quantum Molecular Resonance effects on human mesenchymal stromal cells

Sabrina Sella, Valentina Adami, Eliana Amati, Martina Bernardi, Katia Chieregato, Pamela Gatto, Martina Menarin, Alessandro Pozzato, Gianantonio Pozzato, Giuseppe Astori, Sabrina Sella, Valentina Adami, Eliana Amati, Martina Bernardi, Katia Chieregato, Pamela Gatto, Martina Menarin, Alessandro Pozzato, Gianantonio Pozzato, Giuseppe Astori

Abstract

Electromagnetic fields play an essential role in cellular functions interfering with cellular pathways and tissue physiology. In this context, Quantum Molecular Resonance (QMR) produces waves with a specific form at high-frequencies (4-64 MHz) and low intensity through electric fields. We evaluated the effects of QMR stimulation on bone marrow derived mesenchymal stromal cells (MSC). MSC were treated with QMR for 10 minutes for 4 consecutive days for 2 weeks at different nominal powers. Cell morphology, phenotype, multilineage differentiation, viability and proliferation were investigated. QMR effects were further investigated by cDNA microarray validated by real-time PCR. After 1 and 2 weeks of QMR treatment morphology, phenotype and multilineage differentiation were maintained and no alteration of cellular viability and proliferation were observed between treated MSC samples and controls. cDNA microarray analysis evidenced more transcriptional changes on cells treated at 40 nominal power than 80 ones. The main enrichment lists belonged to development processes, regulation of phosphorylation, regulation of cellular pathways including metabolism, kinase activity and cellular organization. Real-time PCR confirmed significant increased expression of MMP1, PLAT and ARHGAP22 genes while A2M gene showed decreased expression in treated cells compared to controls. Interestingly, differentially regulated MMP1, PLAT and A2M genes are involved in the extracellular matrix (ECM) remodelling through the fibrinolytic system that is also implicated in embryogenesis, wound healing and angiogenesis. In our model QMR-treated MSC maintained unaltered cell phenotype, viability, proliferation and the ability to differentiate into bone, cartilage and adipose tissue. Microarray analysis may suggest an involvement of QMR treatment in angiogenesis and in tissue regeneration probably through ECM remodelling.

Conflict of interest statement

Competing Interests: I have read the journal's policy and the authors of this manuscript have the following competing interests: GP is the cofounder and AP is an employee of Telea Electronic Engineering srl, Sandrigo, Italy.This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. QMR stimulation protocol.
Fig 1. QMR stimulation protocol.
A) Image of the exposure system. B) Scheme of QMR treatment. Cells were seeded on day 0, harvested and reseeded on day 7. The first cycle of treatment started after media renewal on day 3 (black arrows) and the second one on day 10 (blue arrows). Cultures were stimulated 10 minutes/day for 4 consecutive days at 40 or 80 nominal powers. Sham-exposed controls were kept in parallel.
Fig 2. MSC morphology and flow cytometry…
Fig 2. MSC morphology and flow cytometry analysis after QMR stimulation.
A) The images were obtained after 10 minutes of QMR stimulation at Day 5 (first cycle of treatment) and at B) Day 12 (second cycle of treatment). Scale bar = 100 μm. Total magnification = 100X. One representative experiment was shown. C) Five colour combination of monoclonal antibodies was used to verify MSC identity according to the above listed surface markers of a representative sample. Grey line = unstained control (CTL-). Blue line = sham-exposed control (CTL). Green line = QMR setting 80. Red line = QMR setting 40.
Fig 3. Adipogenic, osteogenic and chondrogenic differentiation…
Fig 3. Adipogenic, osteogenic and chondrogenic differentiation after QMR cycles of stimulation.
Panels display one representative experiment showing the final outcome in MSC multilineage differentiation after 21 days of induction. QMR-treated (at 40 and 80 nominal powers) and untreated samples (CTL+) were induced to differentiation. Osteogenic differentiation after one cycle (A) and two cycles (B) of QMR stimulation was assessed using Alizarin Red. Adipogenic (C, D) and chondrogenic (E, F) differentiation were detected using Oil Red O and Alcian Blue stainings, respectively. Scale bar = 100 μm. Total magnification = 100x.
Fig 4. Cellular viability and proliferation after…
Fig 4. Cellular viability and proliferation after QMR treatment.
A) Histograms represent the % of cellular viability after two cycles of QMR treatment at the different settings compared to the sham-exposed controls determined by flow cytometry. Data were shown as mean ± SD of three independent experiments; B) Percentages of cellular proliferation on the controls were obtained by WST-1 assay after 72 hours. Data were represented as mean ± SD of n = 6 independent experiments. No statistical differences were found between conditions.
Fig 5. Dot plots of differentially expressed…
Fig 5. Dot plots of differentially expressed genes associated with QMR treatment at 40 and 80 nominal powers.
Images illustrated the distribution of (A) 40 and (B) 80 up- and down-regulated genes (Benjamini & Hochberg correction at p

Fig 6. Best enrichment gene lists.

Analysis…

Fig 6. Best enrichment gene lists.

Analysis of functional gene enrichment using ToppFun tool (application…

Fig 6. Best enrichment gene lists.
Analysis of functional gene enrichment using ToppFun tool (application of ToppGene Suite) for A) up-regulated and B) down-regulated DEG between 40 QMR setting and control with significant enrichment (dotted line) for FDR B&H q-value 2.

Fig 7. Relative gene expressions using quantitative…

Fig 7. Relative gene expressions using quantitative real-time PCR.

Expression of 8 genes selected by…

Fig 7. Relative gene expressions using quantitative real-time PCR.
Expression of 8 genes selected by cDNA microarray was illustrated after n = 6 independent experiments using TBP as representative reference gene; mean ± SD; * p
All figures (7)
Similar articles
References
    1. Liu Q, Song B. Electric field regulated signaling pathways. The international journal of biochemistry & cell biology. 2014. October;55:264–8. - PubMed
    1. Taghian T, Narmoneva DA, Kogan AB. Modulation of cell function by electric field: a high-resolution analysis. J R Soc Interface. 2015. June;12(107). - PMC - PubMed
    1. Costin GE, Birlea SA, Norris DA. Trends in wound repair: cellular and molecular basis of regenerative therapy using electromagnetic fields. Curr Mol Med. 2012. January;12(1):14–26. - PubMed
    1. Fan W, Qian F, Ma Q, Zhang P, Chen T, Chen C, et al. 50 Hz electromagnetic field exposure promotes proliferation and cytokine production of bone marrow mesenchymal stem cells. Int J Clin Exp Med. 2015;8(5):7394–404. - PMC - PubMed
    1. Zimmerman JW, Pennison MJ, Brezovich I, Yi N, Yang CT, Ramaker R, et al. Cancer cell proliferation is inhibited by specific modulation frequencies. Br J Cancer. 2012. January;106(2):307–13. doi: 10.1038/bjc.2011.523 - DOI - PMC - PubMed
Show all 52 references
Publication types
MeSH terms
Grant support
Telea Electronic Engineering provided support in the form of salaries for SS, AP and GP but did not have any additional role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. The specific roles of these authors are articulated in the ‘author contributions’ section.
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig 6. Best enrichment gene lists.
Fig 6. Best enrichment gene lists.
Analysis of functional gene enrichment using ToppFun tool (application of ToppGene Suite) for A) up-regulated and B) down-regulated DEG between 40 QMR setting and control with significant enrichment (dotted line) for FDR B&H q-value 2.
Fig 7. Relative gene expressions using quantitative…
Fig 7. Relative gene expressions using quantitative real-time PCR.
Expression of 8 genes selected by cDNA microarray was illustrated after n = 6 independent experiments using TBP as representative reference gene; mean ± SD; * p
All figures (7)

References

    1. Liu Q, Song B. Electric field regulated signaling pathways. The international journal of biochemistry & cell biology. 2014. October;55:264–8.
    1. Taghian T, Narmoneva DA, Kogan AB. Modulation of cell function by electric field: a high-resolution analysis. J R Soc Interface. 2015. June;12(107).
    1. Costin GE, Birlea SA, Norris DA. Trends in wound repair: cellular and molecular basis of regenerative therapy using electromagnetic fields. Curr Mol Med. 2012. January;12(1):14–26.
    1. Fan W, Qian F, Ma Q, Zhang P, Chen T, Chen C, et al. 50 Hz electromagnetic field exposure promotes proliferation and cytokine production of bone marrow mesenchymal stem cells. Int J Clin Exp Med. 2015;8(5):7394–404.
    1. Zimmerman JW, Pennison MJ, Brezovich I, Yi N, Yang CT, Ramaker R, et al. Cancer cell proliferation is inhibited by specific modulation frequencies. Br J Cancer. 2012. January;106(2):307–13. doi:
    1. Stacey M, Fox P, Buescher S, Kolb J. Nanosecond pulsed electric field induced cytoskeleton, nuclear membrane and telomere damage adversely impact cell survival. Bioelectrochemistry. 2011. October;82(2):131–4. doi:
    1. Yoon YJ, Li G, Kim GC, Lee HJ, Song K. Effects of 60-Hz time-varying electric fields on DNA damage and cell viability support negligible genotoxicity of the electric fields. JEES. 2015;15(3):134–41.
    1. Serena E, Figallo E, Tandon N, Cannizzaro C, Gerecht S, Elvassore N, et al. Electrical stimulation of human embryonic stem cells: cardiac differentiation and the generation of reactive oxygen species. Exp Cell Res. 2009. December;315(20):3611–9. doi:
    1. Rouabhia M, Park H, Meng S, Derbali H, Zhang Z. Electrical stimulation promotes wound healing by enhancing dermal fibroblast activity and promoting myofibroblast transdifferentiation. PLoS One. 2013;8(8):e71660 doi:
    1. Maioli M, Rinaldi S, Santaniello S, Castagna A, Pigliaru G, Gualini S, et al. Radiofrequency energy loop primes cardiac, neuronal, and skeletal muscle differentiation in mouse embryonic stem cells: a new tool for improving tissue regeneration. Cell Transplant. 2012;21(6):1225–33. doi:
    1. Zhao Z, Watt C, Karystinou A, Roelofs AJ, McCaig CD, Gibson IR, et al. Directed migration of human bone marrow mesenchymal stem cells in a physiological direct current electric field. Eur Cell Mater. 2011. November;22:344–58.
    1. Tandon N, Goh B, Marsano A, Chao PH, Montouri-Sorrentino C, Gimble J, et al. Alignment and elongation of human adipose-derived stem cells in response to direct-current electrical stimulation. Conf Proc IEEE Eng Med Biol Soc. 2009;2009:6517–21. doi:
    1. Zhang J, Ren R, Luo X, Fan P, Liu X, Liang S, et al. A small physiological electric field mediated responses of extravillous trophoblasts derived from HTR8/SVneo cells: involvement of activation of focal adhesion kinase signaling. PLoS One. 2014;9(3):e92252 doi:
    1. Gómez-Ochoa I, Gómez-Ochoa P, Gómez-Casal F, Cativiela E, Larrad-Mur L. Pulsed electromagnetic fields decrease proinflammatory cytokine secretion (IL-1β and TNF-α) on human fibroblast-like cell culture. Rheumatol Int. 2011. October;31(10):1283–9. doi:
    1. Vianale G, Reale M, Amerio P, Stefanachi M, Di Luzio S, Muraro R. Extremely low frequency electromagnetic field enhances human keratinocyte cell growth and decreases proinflammatory chemokine production. Br J Dermatol. 2008. June;158(6):1189–96. doi:
    1. Jennings J, Chen D, Feldman D. Transcriptional response of dermal fibroblasts in direct current electric fields. Bioelectromagnetics. 2008. July;29(5):394–405. doi:
    1. Lee HC, Hong MN, Jung SH, Kim BC, Suh YJ, Ko YG, et al. Effect of extremely low frequency magnetic fields on cell proliferation and gene expression. Bioelectromagnetics. 2015. October;36(7):506–16. doi:
    1. Messerli MA, Graham DM. Extracellular electrical fields direct wound healing and regeneration. Biol Bull. 2011. August;221(1):79–92. doi:
    1. D'Eredità R, Bozzola L. Molecular resonance vs. coblation tonsillectomy in children. Laryngoscope. 2009. October;119(10):1897–901. doi:
    1. Chang H, Hah JH. Comparison of post-tonsillectomy pain with two different types of bipolar forceps: low temperature quantum molecular resonance device versus high temperature conventional electrocautery. Acta Otolaryngol. 2012. June;132 Suppl 1:S130–3.
    1. Schiavon M, Calabrese F, Nicotra S, Marulli G, Pozzato G, Giacometti C, et al. Favorable tissue effects of quantum molecular resonance device (Vesalius) compared with standard electrocautery. A novel paradigm in lung surgery. Eur Surg Res. 2007;39(4):222–8. doi:
    1. Dal Maschio M, Canato M, Pigozzo FM, Cipullo A, Pozzato G, Reggiani C. Biophysical effects of high frequency electrical field (4–64 MHz) on muscle fibers in culture. Basic Applied Miology. 2009;19(1):49–56.
    1. Lopresti M, Tomba A, Caserta A, Di Dimenica F. Effectiveness of molecular quantum resonance in reducing edema after total knee arthroplasty: a clinical trial. Arch Ortop Reumatol. 2011;122(1):34–5.
    1. Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 2013. November;45:e54 doi:
    1. Rubio-Azpeitia E, Andia I. Partnership between platelet-rich plasma and mesenchymal stem cells: in vitro experience. Muscles Ligaments Tendons J. 2014. January;4(1):52–62.
    1. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315–7. doi:
    1. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005. February 15;105(4):1815–22. doi:
    1. Uccelli A, Moretta L, Pistoia V. Immunoregulatory function of mesenchymal stem cells. Eur J Immunol. 2006. October;36(10):2566–73. doi:
    1. Chen Y, Xiang L- X, Shao J- Z, Pan R- L, Wang Y- X, Dong X- J, et al. Recruitment of endogenous bone marrow mesenchymal stem cells towards injured liver. Journal of Cellular and Molecular Medicine. 2010;14(6b):1494–508. doi:
    1. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015. April;43(7):e47 doi:
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001. December;25(4):402–8. doi:
    1. Sundelacruz S, Levin M, Kaplan DL. Membrane potential controls adipogenic and osteogenic differentiation of mesenchymal stem cells. PLoS One. 2008;3(11):e3737 doi:
    1. 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. April;31(3):209–19. doi:
    1. 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 2012. Mar-Apr;26(2):299–304.
    1. Teven CM, Greives M, Natale RB, Su Y, Luo Q, He BC, et al. Differentiation of osteoprogenitor cells is induced by high-frequency pulsed electromagnetic fields. J Craniofac Surg. 2012. March;23(2):586–93. doi:
    1. Kučera O, Cifra M. Radiofrequency and microwave interactions between biomolecular systems. Journal of Biological Physics. 2016;42(1):1–8. doi:
    1. Sheikh AQ, Taghian T, Hemingway B, Cho H, Kogan AB, Narmoneva DA. Regulation of endothelial MAPK/ERK signalling and capillary morphogenesis by low-amplitude electric field. J R Soc Interface. 2013. Jan;10(78):20120548 doi:
    1. Marinković G, Heemskerk N, van Buul JD, de Waard V. The Ins and Outs of Small GTPase Rac1 in the Vasculature. J Pharmacol Exp Ther. 2015. August;354(2):91–102. doi:
    1. Mori M, Saito K, Ohta Y. ARHGAP22 localizes at endosomes and regulates actin cytoskeleton. PLoS One. 2014;9(6):e100271 doi:
    1. Cifra M, Fields JZ, Farhadi A. Electromagnetic cellular interactions. Prog Biophys Mol Biol. 2011. May;105(3):223–46. doi:
    1. Heissig B, Dhahri D, Eiamboonsert S, Salama Y, Shimazu H, Munakata S, et al. Role of mesenchymal stem cell-derived fibrinolytic factor in tissue regeneration and cancer progression. Cell Mol Life Sci. 2015. December;72(24):4759–70. doi:
    1. Neuss S, Schneider RK, Tietze L, Knüchel R, Jahnen-Dechent W. Secretion of fibrinolytic enzymes facilitates human mesenchymal stem cell invasion into fibrin clots. Cells Tissues Organs. 2010;191(1):36–46. doi:
    1. Roy R, Zhang B, Moses MA. Making the cut: protease-mediated regulation of angiogenesis. Exp Cell Res. 2006. March;312(5):608–22. doi:
    1. Pepper MS. Role of the matrix metalloproteinase and plasminogen activator-plasmin systems in angiogenesis. Arterioscler Thromb Vasc Biol. 2001. July;21(7):1104–17.
    1. Nam HS, Kwon I, Lee BH, Kim H, Kim J, An S, et al. Effects of Mesenchymal Stem Cell Treatment on the Expression of Matrix Metalloproteinases and Angiogenesis during Ischemic Stroke Recovery. PLoS One. 2015;10(12):e0144218 doi:
    1. Park JE, Keller GA, Ferrara N. The vascular endothelial growth factor (VEGF) isoforms: differential deposition into the subepithelial extracellular matrix and bioactivity of extracellular matrix-bound VEGF. Mol Biol Cell. 1993. December;4(12):1317–26.
    1. Whitelock JM, Murdoch AD, Iozzo RV, Underwood PA. The degradation of human endothelial cell-derived perlecan and release of bound basic fibroblast growth factor by stromelysin, collagenase, plasmin, and heparanases. J Biol Chem. 1996. April;271(17):10079–86.
    1. Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol. 2014. December;15(12):786–801. doi:
    1. Fredriksson L, Li H, Fieber C, Li X, Eriksson U. Tissue plasminogen activator is a potent activator of PDGF-CC. EMBO J. 2004. October;23(19):3793–802. doi:
    1. Kim IS, Song JK, Zhang YL, Lee TH, Cho TH, Song YM, et al. Biphasic electric current stimulates proliferation and induces VEGF production in osteoblasts. Biochim Biophys Acta. 2006. September;1763(9):907–16. doi:
    1. Bai H, Forrester JV, Zhao M. DC electric stimulation upregulates angiogenic factors in endothelial cells through activation of VEGF receptors. Cytokine. 2011. July;55(1):110–5. doi:
    1. Asadi MR, Torkaman G, Hedayati M, Mofid M. Role of sensory and motor intensity of electrical stimulation on fibroblastic growth factor-2 expression, inflammation, vascularization, and mechanical strength of full-thickness wounds. J Rehabil Res Dev. 2013;50(4):489–98.

Source: PubMed

3
Abonnieren