MORPHOLOGICAL ASSESSMENT OF THE OVARIANS AFTER SINGLE AND FRACTIONAL LOCAL ELECTRON IRADIATION



Cite item

Full Text

Abstract

When malignant neoplasms of the pelvic organs are irradiated, healthy ovarian tissues can get into the irradiation area. So, among all physico-chemical factors, ionizing radiation is the most common cause of ovarian failure, which has a negative impact on fertility. Conducting research in this area is especially important in connection with the active introduction of electron therapy in the protocols for the treatment of malignant neoplasms of the small pelvis with the need to find ways to prevent and treat post-radiation ovarian lesions. In addition, one of the main tasks of modern radiobiology is the creation of experimental animal models in order to reveal the mechanisms of radiation exposure with subsequent extrapolation of the results obtained to humans in order to level the side effects of radiation therapy and select optimal doses.

The aim of the study was a morphofunctional assessment of the ovaries after local electron irradiation in single and fractional modes.

Materials and methods. Wistar rats (n=30) were divided into three groups: I – control (n=10); II (n=10) – subjected to a single local irradiation with electrons at a dose of 2 Gy; III (n=10) – subjected to fractional local irradiation with electrons in a total dose of 20 Gy (in daily fractions of 5 Gy).

Results. After a single local irradiation with electrons at a dose of 2 Gy, multiple hemorrhages and a decrease in the number of growing follicles with a discontinuous theca layer, which were unevenly distributed over its volume, were noted in the ovary. A statistically significant difference in the number of follicles was revealed: a decrease in the number of primordial, primary, secondary and tertiary follicles and an increase in atretic follicles. The most pronounced difference in the number of follicles between the studied groups was found in the group of fractional electron irradiation at a dose of 20 Gy: the smallest number of primordial and the largest number of atretic follicles with signs of post-radiation fibrosis.

Conclusion. The most profound damage to the ovary develops after exposure to fractional electron irradiation at a total dose of 20 Gy compared with a single exposure to ionizing radiation at a dose of 2 Gy: a reduced number of follicles, a decrease in the area and thickness of the cortical substance, as well as the thickness of the ovarian tunica, in combination with the growth of the connective tissue.

Full Text

Restricted Access

About the authors

Grigory Demyashkin

Sechenov University; National Research Center for Radiology

Author for correspondence.
Email: dr.grigdem@gmail.com
ORCID iD: 0000-0001-8447-2600

cand. med. sci., Head of the Laboratory of Histology and Immunohistochemistry, ITMiB, Sechenov University; Head of the Department of Pathomorphology, National Research Center for Radiology

Russian Federation, 119991, Moscow, st. Trubetskaya, d. 8, building 2; 125284, Moscow, 2nd Botkinsky pr., 3

Zaira Murtazalieva

Sechenov University

Email: ZARIA.ALIEVA.90@BK.RU
ORCID iD: 0009-0000-2361-7618

post-graduate student

Russian Federation, 119991, Moscow, st. Trubetskaya, d. 8, building 2

Matvey Vadyukhin

Sechenov University

Email: vma20@mail.ru
ORCID iD: 0000-0002-6235-1020

6th year student of the Institute of Clinical Medicine

Russian Federation, 119991, Moscow, st. Trubetskaya, d. 8, building 2

Makka Bimurzaeva

Sechenov University

Email: bimakka@mail.ru
ORCID iD: 0000-0002-3065-0755

6th year student of the Institute of Clinical Medicine

Russian Federation, 119991, Moscow, st. Trubetskaya, d. 8, building 2

Magomed Lotyrov

Sechenov University

Email: lotyrov_m_i@student.sechenov.ru

6th year student of the Institute of Clinical Medicine

Russian Federation, 119991, Moscow, st. Trubetskaya, d. 8, building 2

References

  1. Gerbi BJ, Antolak JA, Deibel FC, Followill DS, Herman MG, Higgins PD, Huq MS, Mihailidis DN, Yorke ED, Hogstrom KR, et al. Recommendations for clinical electron beam dosimetry: Supplement to the recommendations of Task Group 25. Med. Phys. 2009;36:3239. https://doi.org/10.1118/1.3125820
  2. Reiser E, Bazzano MV, Solano ME, Haybaeck J, Schatz C, Mangesius J, Ganswindt U, Toth B. Unlaid Eggs: Ovarian Damage after Low-Dose Radiation. Cells. 2022;11(7):1219. https://doi.org/10.3390/cells11071219
  3. Immediata V, Ronchetti C, Spadaro D, Cirillo F, Levi-Setti PE. Oxidative Stress and Human Ovarian Response-From Somatic Ovarian Cells to Oocytes Damage: A Clinical Comprehensive Narrative Review. Antioxidants (Basel). 2022;11(7):1335. https://doi.org/10.3390/antiox11071335
  4. Radford IR. DNA lesion complexity and induction of apoptosis by ionizing radiation. Int J Radiat Biol. 2002;78(6):457-66. https://doi.org/10.1080/09553000110120337
  5. Lee CJ, Yoon Y. Gamma-radiation-induced follicular degeneration in the prepubertal mouse ovary. Mutat Res. 2005;578(2):247–255. https://doi.org/10.1016/j.mrfmmm.2005.05.019
  6. Reisz JA, Bansal N, Qian J, Zhao W, Furdui CM. Effects of ionizing radiation on biological molecules--mechanisms of damage and emerging methods of detection. Antioxid Redox Signal. 2014;21(2):260-92. https://doi.org/10.1089/ars.2013.5489
  7. Boots C, Jungheim E. Inflammation and human ovarian follicular dynamics. Semin Reprod Med. 2015;33(4):270–275. https://doi.org/10.1055/s-0035-1554928
  8. Grover AR, Kimler BF, Duncan FE. Use of a Small Animal Radiation Research Platform (SARRP) facilitates analysis of systemic versus targeted radiation effects in the mouse ovary. J Ovarian Res. 2018;11(1):72. https://doi.org/10.1186/s13048-018-0442-8
  9. England K, Cotter TG. Direct oxidative modifications of signalling proteins in mammalian cells and their effects on apoptosis. Redox Rep. 2005;10(5):237–245. https://doi.org/10.1179/135100005X70224
  10. Meirow D, Nugent D. The effects of radiotherapy and chemotherapy on female reproduction. Hum Reprod Update. 2001;7(6):535-43. https://doi.org/10.1093/humupd/7.6.535
  11. Oktem O, Kim SS, Selek U, Schatmann G, Urman B. Ovarian and Uterine Functions in Female Survivors of Childhood Cancers. Oncologist. 2018;23(2):214-224. https://doi.org/10.1634/theoncologist.2017-0201
  12. Gao W, Liang JX, Ma C, Dong JY, Yan Q. The Protective Effect of N-Acetylcysteine on Ionizing Radiation Induced Ovarian Failure and Loss of Ovarian Reserve in Female Mouse. Biomed Res Int. 2017;2017:4176170. https://doi.org/10.1155/2017/4176170
  13. Alesi LR, Nguyen QN, Stringer JM, Winship AL, Hutt KJ. The future of fertility preservation for women treated with chemotherapy. Reprod Fertil. 2023;4(2):e220123. https://doi.org/10.1530/RAF-22-0123
  14. Zhang S, Liu Q, Chang M, Pan Y, Yahaya BH, Liu Y, Lin J. Chemotherapy impairs ovarian function through excessive ROS-induced ferroptosis. Cell Death Dis. 2023;14(5):340. https://doi.org/10.1038/s41419-023-05859-0
  15. Meirow D, Biederman H, Anderson RA, Wallace WH. Toxicity of chemotherapy and radiation on female reproduction. Clin Obstet Gynecol. 2010;53:727–739. https://doi.org/10.1097/GRF.0b013e3181f96b54
  16. Taskin MI, Yay A, Adali E, Balcioglu E, Inceboz U. Protective effects of sildenafil citrate administration on cisplatin-induced ovarian damage in rats. Gynecol Endocrinol. 2015;31(4):272-7. https://doi.org/10.3109/09513590.2014.984679
  17. Land KL, Miller FG, Fugate AC, Hannon PR. The effects of endocrine-disrupting chemicals on ovarian- and ovulation-related fertility outcomes. Mol Reprod Dev. 2022;89(12):608-631. https://doi.org/10.1002/mrd.23652
  18. Kim S, Kim SW, Han SJ, Lee S, Park HT, Song JY, Kim T. Molecular Mechanism and Prevention Strategy of Chemotherapy- and Radiotherapy-Induced Ovarian Damage. Int J Mol Sci. 2021;22(14):7484. https://doi.org/10.3390/ijms22147484
  19. Devine PJ, Perreault SD, Luderer U. Roles of reactive oxygen species and antioxidants in ovarian toxicity. Biol Reprod. 2012;86(2):27. https://doi.org/10.1095/biolreprod.111.095224
  20. Wang W, Craig ZR, Basavarajappa MS, Gupta RK, Flaws JA. Di (2-ethylhexyl phthalate inhibits growth of mouse ovarian antral follicles through an oxidative stress pathway. Toxicology and Applied Pharmacology. 2012;258(2):288–295. https://doi.org/10.1016/j.taap.2011.11.008
  21. Yan F, Zhao Q, Li Y, Zheng Z, Kong X, Shu C, Liu Y, Shi Y. The role of oxidative stress in ovarian aging: a review. J Ovarian Res. 2022;15(1):100. https://doi.org/10.1186/s13048-022-01032-x
  22. Rudnicka E, Kunicki M, Calik-Ksepka A, Suchta K, Duszewska A, Smolarczyk K, Smolarczyk R. Anti-Müllerian Hormone in Pathogenesis, Diagnostic and Treatment of PCOS. Int J Mol Sci. 2021;22(22):12507. https://doi.org/10.3390/ijms222212507
  23. Chatziandreou E, Eustathiou A, Augoulea A, Armeni E, Mili N, Boutas I, Tsoltos N, Kapetanaki A, Kalantaridou S. Antimüllerian Hormone as a Tool to Predict the Age at Menopause. Geriatrics (Basel). 2023;8(3):57. https://doi.org/10.3390/geriatrics8030057
  24. Onder GO, Balcioglu E, Baran M, Ceyhan A, Cengiz O, Suna PA, Yıldız OG, Yay A. The different doses of radiation therapy-induced damage to the ovarian environment in rats. Int J Radiat Biol. 2021;97(3):367-375. https://doi.org/10.1080/09553002.2021.1864497

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) Eco-Vector



СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: серия ПИ № ФС 77 - 74760 от 29.12.2018 г.


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies