Summary

Using Hyperbaric Oxygen to Improve the Radiosensitivity of Human U251 Glioma Cells

Published: October 20, 2022
doi:

Summary

This protocol shows that hyperbaric oxygen can enhance the proliferation inhibition and apoptosis of U251 glioma cells treated with X-ray irradiation by blocking the cells in the G2/M phase. This improves the radiosensitivity of human glioma cell lines.

Abstract

The aim of this study was to explore the use of hyperbaric oxygen to enhance the radiosensitivity of human glioma cells. Sub-cultured U251 human glioma cells were randomly divided into four groups: an untreated control group, cells treated with hyperbaric oxygen (HBO) only, cells treated with X-ray irradiation (X-ray) only, and cells treated with both HBO and X-ray. Cell morphology, cell proliferation activity, cell cycle distribution, and apoptosis were observed in these groups to evaluate the role of HBO in improving the radiosensitivity of glioma cells. With the increase in X-ray doses (0 Gy, 2 Gy, 4 Gy, 6 Gy, 8 Gy), the survival fraction (SF) of glioma cells gradually decreased.

Significantly lower SF was observed for the cells treated with the HBO and X-ray together than in the X-ray group for each dose (all P < 0.05). The proliferation inhibition was significantly higher in the HBO combined with X-ray group than in the X-ray group for each dose (all P < 0.05) for the U251 cell line. The percentage of G2/M phase cells was significantly higher in the HBO combined with X-ray (2 Gy) group (26.70% ± 2.46%) and the HBO group (22.36% ± 0.91%) than in the control group (11.56% ± 2.01%) and X-ray (2 Gy) group (10.35% ± 2.69%) (all P < 0.05). U251 cell apoptosis was significantly higher in the HBO combined with X-ray (2 Gy) group than in the HBO group, the X-ray (2 Gy) group, and the control group (all P < 0.05). We conclude that HBO can enhance the proliferation inhibition and apoptosis of glioma U251 cells by blocking glioma cells in the G2/M phase and improve the radiosensitivity of U251 glioma cells.

Introduction

Glioma is a primary intracranial tumor that originates from central nervous system glial cells1. The current treatment strategy for glioma is surgery combined with radiotherapy and chemotherapy. Postoperative radiotherapy for glioma can provide survival benefits (grade I evidence), and early postoperative radiotherapy can effectively prolong patient survival (grade II evidence)2. For higher-grade gliomas (grade III or IV), especially highly malignant and invasive glioblastoma (grade III evidence)3, postoperative radiotherapy should be performed as early as possible (<6 weeks). However, despite early intervention, glioma still has a high recurrence rate and poor prognosis after comprehensive treatment. These outcomes are mainly associated with the low radiosensitivity of glioma. Factors related to tumor radiosensitivity include the inherent radiosensitivity of tumor cells, hypoxic or non-hypoxic tumor cells, the proportion of hypoxic tumor cells, and the capacity of peritumoral tissue to repair radiation damage4.

Among these factors, hypoxic or non-hypoxic tumor cells and the proportion of hypoxic tumor cells have important effects on tumor radiosensitivity. Hyperbaric oxygen (HBO) can improve tissue oxygen storage by increasing tissue oxygen tension and blood oxygen diffusion. HBO may also produce a series of beneficial biochemical, cytological, and physiological effects5. For example, HBO has a marked reparative effect on radiotherapy-induced radiation damage. Although HBO combined with radiotherapy or chemotherapy is reported to improve the clinical efficacy of radiotherapy or chemotherapy for glioma6, there is considerable debate about how HBO alone affects malignant glioma growth. Ding et al.7 and Wang et al.8 both demonstrated that HBO promotes the growth of in situ glioma in mice via mechanisms that involve the inhibition of apoptosis and the promotion of tumor angiogenesis. Under physiological conditions, HBO is reported to promote tumor angiogenesis by inducing oxidative stress9.

However, one study indicated that short-term HBO exposure promotes tumor cell proliferation, whereas prolonged HBO exposure promotes apoptosis and inhibits proliferation10. Therefore, further studies are needed to explore whether HBO promotes or inhibits the growth of glioma and how HBO combined with radiotherapy or chemotherapy can induce therapeutic sensitization. In particular, mechanistic details about how HBO improves the radiosensitivity of glioma are needed. To explore how HBO improves the radiosensitivity of human U251 glioma cells in this study, we used HBO combined with X-ray irradiation on glioma cell proliferation and observed the effects on cell cycle distribution and apoptosis.

Protocol

All study methods were approved by the Institutional Review Board and Ethics Committee of the Second Hospital Affiliated with Lanzhou University and were performed in accordance with relevant guidelines and regulations. 1. Treatment of glioma cells NOTE: The U251 glioma cell line was used in this experiment. U251 cell culture Seed U251 cells in multiple dishes with DMEM containing 10% fetal bovine serum (FBS) and culture them…

Representative Results

Culture of U251 glioma cells U251 glioma cells had a fusiform shape 24 h to 48 h after culture in DMEM and were adherent. These cells were used for further study (Figure 1). Glioma cell morphology and count The cell counts for the U251 glioma cells in the HBO combined with X-ray (2 Gy) group were significantly lower than that for the X-ray (2 Gy) group after 24 h, 48 h, and 72 h of cell culture (all P < 0….

Discussion

The glioma cell line U251 is one of the most classical human glioma cell lines and is widely used as a glioma model in many studies.

Effects of HBO on U251 glioma cell proliferation
HBO typically refers to breathing pure oxygen (100% oxygen concentration) in a sealed chamber with a pressure 1.5-3-fold higher than normal atmospheric pressure, which can increase oxygen content in microvascular plasma14. For patients with glioma, combining radiothera…

Disclosures

The authors have nothing to disclose.

Acknowledgements

None.

Materials

Binding Buffer   Dickinson and Company RH10 9RR
CCK-8 test kit  DOJINDO  NJ Cell counting assay
CELL FIT  cell cycle analysis (DNA content)
CELLQUEST apoptotic cell analysis
DMEM and Annexin V-FITC Gibco BRL
flow cytometer Dickinson
Glioma U251 and U87 cell line Shanghai Institute of Cell Biology
hyperbaric oxygen chamber Hongyuan Institute
medical linear accelerator Elekta Limited Company
microplate reader
MOD FITLT formac v1.01  cell analysis–cell cycle phase
trypsin Hyclone Laboratories Inc

References

  1. Louis, D. N., et al. The 2016 World Health Organization Classification the Central Nervous System: A summary. Acta Neuropathologica. 131 (6), 803-820 (2016).
  2. Sun, M. Z., et al. Survival impact of time to initiation of chemoradiotherapy after resection of newly diagnosed glioblastoma. Journal of Neurosurgery. 122 (5), 1144-1150 (2015).
  3. Hegi, M. E., et al. MGMT gene silencing and benefit from temoozolomide in glioblastoma. New England Journal of Medicine. 352 (10), 997-1003 (2005).
  4. Zhu, Y., et al. Involvement of decreased hypoxia-inducible factor 1 activity and resultant G1-S cell cycle transition in radioresistance of perinecrotic tumor cells. Oncogene. 32 (16), 2058-2068 (2013).
  5. Kohshi, K., et al. Potential roles of hyperbaric oxygenation in the treatments of brain tumors. Undersea and Hyperbaric Medicine. 40 (4), 351-362 (2013).
  6. Aghajan, Y., Grover, I., Gorsi, H., Tumblin, M., Crawford, J. R. Use of hyperbaric oxygen therapy in pediatric neuro-oncology: a single institutional experience. Journal Neurooncology. 141 (1), 151-158 (2019).
  7. Ding, J. B., Chen, J. R., Xu, H. Z., Qin, Z. Y. Effect of hyperbaric oxygen on the growth of intracranial glioma in rats. Chinese Medical Journal. 128 (23), 3197-3203 (2015).
  8. Wang, Y. G., et al. Hyperbaric oxygen promotes malignant glioma cell growth and inhibits cell apoptosis. Oncology Letters. 10 (1), 189-195 (2015).
  9. Milovanova, T. N., et al. Hyperbaric oxygen stimulates vasculogenic stem cell growth and differentiation in vivo. Journal of Applied Physiology. 106 (2), 711-728 (2009).
  10. Conconi, M. T., et al. Effects of hyperbaric oxygen on proliferative and apoptotic activities and reactive oxygen species generation in mouse fibroblast 3T3/J2 cell line. Journal of Investigative Medicine. 51 (4), 227-232 (2003).
  11. Vermes, I., Haanen, C., Steffiens-Nakken, H., Reutellingsperger, C. A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V. Journal of Immunological Methods. 184 (1), 39-51 (1995).
  12. Cui, W., Niu, F. -. L., He, L. -. Y., W, S. -. R. Comparison of two softwares for analysing apoptosis with flow cytometry. Journal of Beijing University of Traditional Chinese Medicine. 24 (6), 45-47 (2001).
  13. Cecchini, M. J., Amiri, M., Dick, F. A. Analysis of cell cycle position in mammalian cells. Journal of Visualized Experiments. (59), e3491 (2012).
  14. Resanovic, I., et al. Effects of hyperbaric oxygen on inducible nitric oxide synthase activity/expression in lymphocytes of type 1 diabetes patients: A prospective pilot study. International Journal of Endocrinology. 2019, 2328505 (2019).
  15. Stuhr, L. E., et al. Hyperoxia retards growth and induces apoptosis, changes in vascular density and gene expression in transplanted gliomas in nude rats. Journal of Neuro-Oncology. 85 (2), 191-202 (2007).
  16. Biollaz, G., et al. Site-specific anti-tumor immunity: differences in DC function, TGF-beta production and numbers of intratumoral Foxp3+ Treg. European Journal of Immunology. 39 (5), 1323-1333 (2009).
  17. McKenna, F. W., Ahmad, S. Fitting techniques of cell survival curves in high-dose region for use in stereotactic body radiation therapy. Physics in Medicine and Biology. 54 (6), 1593-1608 (2009).
  18. Malaise, E. P., Lambin, P., Joiner, M. C. Radiosensitivity of human cell lines to small doses. Are there some clinical implications. Radiation Research. 138, S25-S27 (1994).
  19. Björk-Eriksson, T., West, C., Karlsson, E., Mercke, C. Tumor radiosensitivity (SF2) is a prognostic factor for local control in head and neck cancers. International Journal of Radiation Oncology Biology Physics. 46 (1), 13-19 (2000).
  20. Bromfield, G. P., Meng, A., Warde, P., Bristow, R. G. Cell death in irradiated prostate epithelial cells: Role of apoptotic and clonogenic cell kill. Prostate Cancer and Prostatic Diseases. 6 (1), 73-85 (2003).
  21. Kalns, J. E., Piepmeier, E. H. Exposure to hyperbaric oxygen induces cell cycle perturbation in prostate cancer cells. In Vitro Cellular & Developmental Biology – Animal. 35 (2), 98-101 (1999).
  22. Lakka, S. S., et al. Inhibition of cathepsin B and MMP-9 gene expression in glioblastoma cell line via RNA interference reduces tumor cell invasion, tumor growth and angiogenesis. Oncogene. 23 (27), 4681-4689 (2004).
  23. Li, S., Shi, D., Zhang, L., Yang, F., Cheng, G. Oridonin enhances the radiosensitivity of lung cancer cells by upregulating Bax and downregulating Bcl-2. Experimental and Therapeutic Medicine. 16 (6), 4859-4864 (2018).
  24. Campbell, K. J., Tait, S. W. G. Targeting BCL-2 regulated apoptosis in cancer. Open Biology. 8 (5), 180002 (2018).
  25. Rengarajan, T., et al. D-pinitol promotes apoptosis in MCF-7 cells via induction of p53 and Bax and inhibition of Bcl-2 and NF-κB. Asian Pacific Journal of Cancer Prevention. 15 (4), 1757-1762 (2014).
  26. Shinagawa, A., et al. The potent peptide antagonist to angiogenesis, C16Y and cisplatin act synergistically in the down-regulation of the Bcl-2/Bax ratio and the induction of apoptosis in human ovarian cancer cells. International Journal of Radiation Oncology Biology Physics. 39 (6), 135-164 (2011).
This article has been published
Video Coming Soon
Keep me updated:

.

Cite This Article
Ma, L., Ye, B., Li, J., Pei, Y., Zhong, J., Mou, F., Sun, P. Using Hyperbaric Oxygen to Improve the Radiosensitivity of Human U251 Glioma Cells. J. Vis. Exp. (188), e62769, doi:10.3791/62769 (2022).

View Video