Influence of Powerful Non-Ionizing Terahertz Radiation on Healthy and Tumor Human Cells of Neural Origin
Abstract and keywords
Abstract (English):
Purpose: Study of the influence of high-power pulses of coherent non-ionizing terahertz (THz) radiation on the formation of foci of double-strand DNA breaks and the proliferative activity of human neuronal cells. Material and methods: Irradiated cell cultures are direct reprogramming neural progenitor cells (drNPCs), neuroblastoma cells (SK-N-BE). Cells are irradiated with a sequence of THz radiation pulses with a peak intensity of ~ 20 GW/cm2 and electric field strength of 2.8 MV/cm. Irradiation lasts 30 mins. Results: There is no statistically significant difference in the number of γH2AX histone foci between experimental and control cell groups. Conclusion: It was shown that a short exposure (30 min) of cells to THz radiation with intensity of 20 GW/cm2 does not affect the proliferative activity of both neural progenitor cells and neuroblastoma cells and does not cause a significant increase in γH2AX foci in any of the studied cell lines.

Keywords:
non-ionizing radiation, terahertz radiation, H2AX histone foci, proliferative activity, neural stem cells, SK-N-BE neuroblastoma
References

1. Fröhlich H. Long-range coherence and energy storage in biological systems. Int J Quantum Chem. 1968;2(5):641-9. DOI:https://doi.org/10.1002/qua.560020505.

2. Alexandrov BS, Gelev V, Bishop AR, Usheva A, Rasmussen KØ. DNA breathing dynamics in the presence of a terahertz field. Phys Lett A. 2010;374(10):1214-7. DOI:https://doi.org/10.1016/j.physleta.2009.12.077.

3. Titova LV, Ayesheshim AK, Golubov A, Rodriguez-Juarez R, Woycicki R, Hegmann FA, et al. Intense THz pulses down-regulate genes associated with skin cancer and psoriasis: a new therapeutic avenue? Sci Rep. 2013;3(1):2363. DOI:https://doi.org/10.1038/srep02363.

4. Ol'shevskaya YuS, Kozlov AS, Petrov AK, Zapara TA, Ratushnyak AS. Vliyanie na neyrony in vitro teragercovogo (submillimetrovogo) lazernogo izlucheniya. Zhurnal vysshey nervnoy deyatel'nosti im. I.P. Pavlova. 2009;59(3):353-9.[Olshevskaya YUS, Kozlov AS, Petrov AK, Zapara TA, Ratushnyak AS. Effect of Terahertz (Submillimeter) Laser Radiation on Neurons in Vitro. Journal of Higher Nervous Activity. I.P. Pavlova. 2009; 59 (3): 353-9.]

5. Zapara TA, Treskova SP, Ratushniak AS. Effect of antioxidants on the interaction of terahertz (submillimeter) laser radiation and neuronal membrane. J Surf Investig. 2015;9(5):869-71.

6. Cheon H, Paik JH, Choi M, Yang HJ, Son JH. Detection and manipulation of methylation in blood cancer DNA using terahertz radiation. Sci Rep. 2019;9(1):1-10. DOI:https://doi.org/10.1038/s41598-019-42855-x.

7. Tan SZ, Tan PC, Luo LQ, Chi YL, Yang ZL, Zhao XL, et al. Exposure Effects of Terahertz Waves on Primary Neurons and Neuron-like Cells Under Nonthermal Conditions. Biomed Environ Sci. 2019;32(10):739-54. DOI:https://doi.org/10.3967/bes2019.094.

8. Perera PGT, Appadoo DRT, Cheeseman S, Wandiyanto J V, Linklater D, Dekiwadia C, et al. PC 12 pheochromocytoma cell response to super high frequency terahertz radiation from synchrotron source. Cancers (Basel). 2019;11(2):1-17. DOI:https://doi.org/10.3390/cancers11020162.

9. Maskey D, Pradhan J, Aryal B, Lee C-M, Choi I-Y, Park K-S, et al. Chronic 835-MHz radiofrequency exposure to mice hippocampus alters the distribution of calbindin and GFAP immunoreactivity. Brain Res. 2010;1346(Maskey2010):237-46. DOI:https://doi.org/10.1016/j.brainres.2010.05.045.

10. Rogakou EP, Boon C, Redon C, Bonner WM. Megabase Chromatin Domains Involved in DNA Double-Strand Breaks in Vivo. J Cell Biol. 1999;146(5):905-16. DOI:https://doi.org/10.1083/jcb.146.5.905.

11. Barnes JL, Zubair M, John K, Poirier MC, Martin FL. Carcinogens and DNA damage. Biochem Soc Trans. 2018 Oct 19;46(5):1213-24. DOI:https://doi.org/10.1042/BST20180519.

12. Sitnikov DS, Ilina I V, Pronkin AA. Experimental system for studying bioeffects of intense terahertz pulses with electric field strength up to 3.5 MV/cm. Opt Eng. 2020;59(06):061613. DOI:https://doi.org/10.1117/1.OE.59.6.061613.full

13. Ovchinnikov AV, Chefonov OV, Sitnikov DS, Il'ina IV, Ashitkov SI, Agranat MB, Istochnik teragercevogo izlucheniya s napryazhennost'yu elektricheskogo polya svyshe 1 MV/sm na osnove femtosekundnogo hrom-forsteritovogo lazera s chastotoy sledovaniya impul'sov 100 Gc. Kvantovaya elektronika. 2018;48(6):554-8. [Ovchinnikov AV, Chefonov OV, Sitnikov DS, Il’ina I V, Ashitkov SI, Agranat MB. A source of THz radiation with electric field strength of more than 1 MV cm-1 on the basis of 100-Hz femtosecond Cr : forsterite laser system. Quantum Electron. 2018;48(6):554-8. (In Russian) DOI: 10.1070/ qel16681].

14. Sitnikov DS, Romashevskiy SA, Ovchinnikov A V, Chefonov O V, Savel’ev AB, Agranat MB. Estimation of THz field strength by an electro-optic sampling technique using arbitrary long gating pulses. Laser Phys Lett. 2019;16(11):115302. DOI:https://doi.org/10.1088/1612-202X/ab4d56.

15. Sitnikov DS, Il'ina IV, Gurova SA, Shatalova RO, Revkova VA. Issledovanie indukcii dvunitevyh razryvov v fibroblastah kozhi cheloveka teragercevym izlucheniem vysokoy intensivnosti. Izvestiya Rossiyskoy Akademii Nauk Seriya Fizicheskaya. 2020;84:1605-16. DOI:https://doi.org/10.31857/s0367676520110277. [Sitnikov DS, Ilina I V, Gurova SA, Shatalova RO, Revkova VA. Studying the Induction of Double-Strand Breaks in Human Fibroblasts by High-Intensity Terahertz Radiation. Bull Russ Acad Sci Phys. 2020;84(11):1370-4. (In Russian) DOI:https://doi.org/10.3103/S1062873820 110-246].

16. Dhuppar S, Roy S, Mazumder A. γH2AX in the S Phase after UV Irradiation Corresponds to DNA Replication and Does Not Report on the Extent of DNA Damage. Mol Cell Biol. 2020;40(20). DOI:https://doi.org/10.1128/MCB.00328-20.

17. Bourge M, Fort C, Soler M, Satiat-Jeunemaître B, Brown SC. A pulse-chase strategy combining click-EdU and photoconvertible fluorescent reporter: tracking Golgi protein dynamics during the cell cycle. New Phytol. 2015;205(2):938-50. DOI:https://doi.org/10.1111/nph.13069.

18. Yu T, MacPhail SH, Banáth JP, Klokov D, Olive PL. Endogenous expression of phosphorylated histone H2AX in tumors in relation to DNA double-strand breaks and genomic instability. DNA Repair (Amst). 2006;5(8):935-46. DOI:https://doi.org/10.1016/j.dnarep.2006.05.040.

19. Sitnikov DS, Ilina I V., Revkova VA, Konoplyannikov MA, Kalsin VA, Baklaushev VP. Effect of high-power pulses of terahertz radiation on cell viability. In: 2020 International Conference Laser Optics (ICLO). IEEE; 2020. p. 1. DOI:https://doi.org/10.1109/ICLO48556.2020.9285431.

20. Nagelkerke A, Span PN. Staining Against Phospho-H2AX (γ-H2AX) as a Marker for DNA Damage and Genomic Instability in Cancer Tissues and Cells. In: Koumenis C, Coussens LM, Giaccia A, Hammond E, editors. Tumor Microenvironment. Springer International Publishing; 2016. p. 1-10. PMID: 27325258 DOI:https://doi.org/10.1007/978-3-319-26666-4_1

Login or Create
* Forgot password?