| Peer-Reviewed

The Effect of Radiosensitization of Gemcitabine Related to Suppression of a Repair Pathway: Examination of Mammalian Cells with Therapeutic High Energy X-rays

Received: 11 September 2017    Accepted: 27 September 2017    Published: 7 November 2017
Views:       Downloads:
Abstract

Gemcitabine is used in clinical chemo-radiotherapy; however, the mechanism underlying enhanced radiosensitivity by gemcitabine is not fully elucidated. We evaluated the role of gemcitabine in mammalian cell lines using a therapeutic high energy 10 MeV linac-X-ray irradiation device. Rodent cell lines CHO and xrs5 were used. A total of 5 μM gemcitabine for 24 hours was administered with or without post-X-ray irradiation. DNA double-strand breaks (DSBs) and cell enlargement were observed by using singly gemcitabine. Enhanced cell killing effects by radiotherapy were observed with gemcitabine pre-treatment in both CHO and xrs5 cells. We focused on the dynamics of phosphorylated p53-binding protein 1 (53BP1)-positive foci after irradiation. Significantly higher numbers of 53BP1 foci were observed after irradiation in gemcitabine pre-treated cells than in untreated cells. The radiosensitizing effect of gemcitabine was not suppressed in the non-homologous end joining (NHEJ) deficient xrs5 cells. We confirmed that in rodent cells the radiosensitizing effect of gemcitabine is related to suppression of a repair pathway other than NHEJ.

Published in American Journal of Laboratory Medicine (Volume 2, Issue 6)
DOI 10.11648/j.ajlm.20170206.15
Page(s) 137-143
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2024. Published by Science Publishing Group

Keywords

Radiosensitization, Gemcitabine, Non-homologous End Joining, 53BP1

References
[1] Kwegyir-Afful AK, Murigi FN, Purushottamachar P, Ramamurthy VP, Martin MS, Njar VCO. Galeterone and its analogs inhibit Mnk-eIF4E axis, synergize with gemcitabine, impede pancreatic cancer cell migration, invasion and proliferation and inhibit tumor growth in mice. Oncotarget. 2016, 8(32): 52381-402.
[2] Zhang SH, Liu GF, Li XF, Liu L, Yu SN. Efficacy of different chemotherapy regimens in treatment of advanced or metastatic pancreatic cancer: a network meta-analysis. J Cell Physiol. 2017, Sep 19. doi: 10.1002/jcp.26183.
[3] Rajabpour A, Afgar A, Mahmoodzadeh H, Radfar JE, Rajaei F, Teimoori-Toolabi L. MiR-608 regulating the expression of ribonucleotide reductase M1 and cytidine deaminase is repressed through induced gemcitabine chemoresistance in pancreatic cancer cells. Cancer Chemother Pharmacol. 2017 Sep 8. doi: 10.1007/s00280-017-3418-2.
[4] Schwartzberg LS, Arena FP, Bienvenu BJ, Kaplan EH, Camacho LH, Campos LT, Waymack JP, Tagliaferri MA, Chen MM, Li D. A Randomized, Open-Label, Safety and Exploratory Efficacy Study of Kanglaite Injection (KLTi) plus Gemcitabine versus Gemcitabine in Patients with Advanced Pancreatic Cancer. J Cancer. 2017, 8(10): 1872-83.
[5] Shewach DS, Hahn TM, Chang E, Hertel LW, Lawrence TS. Metabolism of 2',2'-difluoro-2'-deoxycytidine and radiation sensitization of human colon carcinoma cells. Cancer Res. 1994, 54(12): 3218-23.
[6] Lawrence TS, Chang EY, Hahn TM, Hertel LW, Shewach DS. Radiosensitization of pancreatic cancer cells by 2',2'-difluoro-2'-deoxycytidine. Int J Radiat Oncol Biol Phys. 1996, 34(4): 867-72.
[7] Joschko MA, Webster LK, Groves J, Yuen K, Palatsides M, Ball DL, Millward MJ. Enhancement of radiation-induced regrowth delay by gemcitabine in a human tumor xenograft model. Radiat Oncol Investig. 1997, 5(2): 62-71.
[8] Milas L, Fujii T, Hunter N, Elshaikh M, Mason K, Plunkett W, Ang KK, Hittelman W. Enhancement of tumor radioresponse in vivo by gemcitabine. Cancer Res. 1999, 59(1): 107-14.
[9] Wachters FM, van Putten JW, Maring JG, Zdzienicka MZ, Groen HJ, Kampinga HH. Selective targeting of homologous DNA recombination repair by gemcitabine. Int J Radiat Oncol Biol Phys. 2003, 57(2): 553-6.
[10] Wouters A, Pauwels B, Lambrechts HA, Pattyn GG, Ides J, Baay M, Meijnders P, Peeters M, Vermorken JB, Lardon F. Retention of the in vitro radiosensitizing potential of gemcitabine under anoxic conditions, in p53 wild-type and p53-deficient non-small-cell lung carcinoma cells. Int J Radiat Oncol Biol Phys. 2011, 80(2): 558-66.
[11] Shen ZT, Wu XH, Wang L, Li B, Zhu XX. Effects of gemcitabine on radiosensitization, apoptosis, and Bcl-2 and Bax protein expression in human pancreatic cancer xenografts in nude mice. Genet Mol Res. 2015, 14(4): 15587-96.
[12] Kobashigawa S, Morikawa K, Mori H, Kashino G. Gemcitabine Induces Radiosensitization Through Inhibition of RAD51-dependent Repair for DNA Double-strand Breaks. Anticancer Res. 2015, 35(5): 2731-7.
[13] Im MM, Flanagan SA, Ackroyd JJ, Knapp B, Kramer A, Shewach DS. Late DNA Damage Mediated by Homologous Recombination Repair Results in Radiosensitization with Gemcitabine. Radiat Res. 2016, 186(5): 466-77.
[14] van Putten JWG, Groen HJM, Smid K, Peters GJ, Kampinga HH. End-joining deficiency and radiosensitization induced by gemcitabine. Cancer Res. 2001, 61(4): 1585-91.
[15] Lawrence T. S, Eisbruch A, Shewach D. S. Gemcitabine-mediated radiosensitization. Semin. Oncol. 1997, 24(7): 24-8.
[16] Gospodinov A, Herceg Z. Chromatin structure in double strand break repair. DNA Repair (Amst). 2013, 12(10): 800-10.
[17] Kanaar R, Hoeijmakers JH, van Gent DC. Molecular mechanisms of DNA double strand break repair. Trends Cell Biol. 1998, 8: 483-9.
[18] Karran P. DNA double strand break repair in mammalian cells. Curr Opin Genet Dev. 2000, 10: 144-50.
[19] Khanna KK, Jackson SP. DNA double-strand breaks: signaling, repair and the cancer connection. Nat Genet. 2001, 27: 247-54.
[20] Japan society of medical physics, Committee of Dosimetry. Standard Dosimetry of Absorbed in Dose in External Beam Radiotherapy. Tokyo: Tushosangyoukenkyusya; 2006. P. 35-46.
[21] Tashiro S, Walter J, Shinohara A, Kamada N, Cremer T. Rad51 accumulation at sites of DNA damage and in postreplicative chromatin. J Cell Biol. 2000, 150(2): 283-91.
[22] Schy WE1, Hertel LW, Kroin JS, Bloom LB, Goodman MF, Richardson FC. Effect of a template-located 2',2'-difluorodeoxycytidine on the kinetics and fidelity of base insertion by Klenow (3'-->5'exonuclease-) fragment. Cancer Res. 1993, 53(19): 4582-7.
[23] Ross GM, Eady JJ, Mithal NP, Bush C, Steel GG, Jeggo PA, McMillan TJ. DNA strand break rejoining defect in xrs-6 is complemented by transfection with the human Ku80 gene. Cancer Res. 1995, 55(6): 1235-8.
[24] Van Dyck E, Stasiak AZ, Stasiak A, West SC. Binding of double-strand breaks in DNA by human Rad52 protein. Nature. 1999, 398(6729): 728-31.
[25] Singleton BK, Priestley A, Steingrimsdottir H, Gell D, Blunt T, Jackson SP, Lehmann AR, Jeggo PA. Molecular and biochemical characterization of xrs mutants defective in Ku80. Mol Cell Biol. 1997, 17(3): 1264-73.
[26] He DM, Lee SE, Hendrickson EA. Restoration of X-ray and etoposide resistance, Ku-end binding activity and V(D) J recombination to the Chinese hamster sxi-3 mutant by a hamster Ku86 cDNA. Mutat Res. 1996, 363(1): 43-56.
[27] Wang H, Wang X, Chen G, Zhang X, Tang X, Park D, Cucinotta FA, Yu DS, Deng X, Dynan WS, Doetsch PW, Wang Y. Distinct roles of Ape1 protein, an enzyme involved in DNA repair, in high or low linear energy transfer ionizing radiation-induced cell killing. J Biol Chem. 2014 Oct 31;289(44):30635-44. doi: 10.1074/jbc.M114.604959.
[28] Okayasu R, Okada M, Okabe A, Noguchi M, Takakura K, Takahashi S. Repair of DNA damage induced by accelerated heavy ions in mammalian cells proficient and deficient in the non-homologous end-joining pathway. Radiat Res. 2006, 165(1): 59-67.
[29] Wang H1, Wang X, Zhang P, Wang Y. The Ku-dependent non-homologous end-joining but not other repair pathway is inhibited by high linear energy transfer ionizing radiation. DNA Repair (Amst). 2008, 7(5): 725-33.
Cite This Article
  • APA Style

    Keiko Morikawa, Yoshida Yukito, Yuh Sugii, Genro Kashino, Hiromu Mori. (2017). The Effect of Radiosensitization of Gemcitabine Related to Suppression of a Repair Pathway: Examination of Mammalian Cells with Therapeutic High Energy X-rays. American Journal of Laboratory Medicine, 2(6), 137-143. https://doi.org/10.11648/j.ajlm.20170206.15

    Copy | Download

    ACS Style

    Keiko Morikawa; Yoshida Yukito; Yuh Sugii; Genro Kashino; Hiromu Mori. The Effect of Radiosensitization of Gemcitabine Related to Suppression of a Repair Pathway: Examination of Mammalian Cells with Therapeutic High Energy X-rays. Am. J. Lab. Med. 2017, 2(6), 137-143. doi: 10.11648/j.ajlm.20170206.15

    Copy | Download

    AMA Style

    Keiko Morikawa, Yoshida Yukito, Yuh Sugii, Genro Kashino, Hiromu Mori. The Effect of Radiosensitization of Gemcitabine Related to Suppression of a Repair Pathway: Examination of Mammalian Cells with Therapeutic High Energy X-rays. Am J Lab Med. 2017;2(6):137-143. doi: 10.11648/j.ajlm.20170206.15

    Copy | Download

  • @article{10.11648/j.ajlm.20170206.15,
      author = {Keiko Morikawa and Yoshida Yukito and Yuh Sugii and Genro Kashino and Hiromu Mori},
      title = {The Effect of Radiosensitization of Gemcitabine Related to Suppression of a Repair Pathway: Examination of Mammalian Cells with Therapeutic High Energy X-rays},
      journal = {American Journal of Laboratory Medicine},
      volume = {2},
      number = {6},
      pages = {137-143},
      doi = {10.11648/j.ajlm.20170206.15},
      url = {https://doi.org/10.11648/j.ajlm.20170206.15},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.ajlm.20170206.15},
      abstract = {Gemcitabine is used in clinical chemo-radiotherapy; however, the mechanism underlying enhanced radiosensitivity by gemcitabine is not fully elucidated. We evaluated the role of gemcitabine in mammalian cell lines using a therapeutic high energy 10 MeV linac-X-ray irradiation device. Rodent cell lines CHO and xrs5 were used. A total of 5 μM gemcitabine for 24 hours was administered with or without post-X-ray irradiation. DNA double-strand breaks (DSBs) and cell enlargement were observed by using singly gemcitabine. Enhanced cell killing effects by radiotherapy were observed with gemcitabine pre-treatment in both CHO and xrs5 cells. We focused on the dynamics of phosphorylated p53-binding protein 1 (53BP1)-positive foci after irradiation. Significantly higher numbers of 53BP1 foci were observed after irradiation in gemcitabine pre-treated cells than in untreated cells. The radiosensitizing effect of gemcitabine was not suppressed in the non-homologous end joining (NHEJ) deficient xrs5 cells. We confirmed that in rodent cells the radiosensitizing effect of gemcitabine is related to suppression of a repair pathway other than NHEJ.},
     year = {2017}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - The Effect of Radiosensitization of Gemcitabine Related to Suppression of a Repair Pathway: Examination of Mammalian Cells with Therapeutic High Energy X-rays
    AU  - Keiko Morikawa
    AU  - Yoshida Yukito
    AU  - Yuh Sugii
    AU  - Genro Kashino
    AU  - Hiromu Mori
    Y1  - 2017/11/07
    PY  - 2017
    N1  - https://doi.org/10.11648/j.ajlm.20170206.15
    DO  - 10.11648/j.ajlm.20170206.15
    T2  - American Journal of Laboratory Medicine
    JF  - American Journal of Laboratory Medicine
    JO  - American Journal of Laboratory Medicine
    SP  - 137
    EP  - 143
    PB  - Science Publishing Group
    SN  - 2575-386X
    UR  - https://doi.org/10.11648/j.ajlm.20170206.15
    AB  - Gemcitabine is used in clinical chemo-radiotherapy; however, the mechanism underlying enhanced radiosensitivity by gemcitabine is not fully elucidated. We evaluated the role of gemcitabine in mammalian cell lines using a therapeutic high energy 10 MeV linac-X-ray irradiation device. Rodent cell lines CHO and xrs5 were used. A total of 5 μM gemcitabine for 24 hours was administered with or without post-X-ray irradiation. DNA double-strand breaks (DSBs) and cell enlargement were observed by using singly gemcitabine. Enhanced cell killing effects by radiotherapy were observed with gemcitabine pre-treatment in both CHO and xrs5 cells. We focused on the dynamics of phosphorylated p53-binding protein 1 (53BP1)-positive foci after irradiation. Significantly higher numbers of 53BP1 foci were observed after irradiation in gemcitabine pre-treated cells than in untreated cells. The radiosensitizing effect of gemcitabine was not suppressed in the non-homologous end joining (NHEJ) deficient xrs5 cells. We confirmed that in rodent cells the radiosensitizing effect of gemcitabine is related to suppression of a repair pathway other than NHEJ.
    VL  - 2
    IS  - 6
    ER  - 

    Copy | Download

Author Information
  • Department of Radiological Science, Faculty of Health Sciences, Junshin Gakuen University, Fukuoka, Japan

  • Oita University Hopspital, Oita, Japan

  • Department of Radiological Science, Faculty of Health Sciences, Junshin Gakuen University, Fukuoka, Japan

  • Advanced Molecular Imaging Center, Faculty of Medicine, Oita University, Oita, Japan

  • Department of Radiology, Faculty of Medicine, Oita University, Oita, Japan

  • Sections