Elsevier

Free Radical Biology and Medicine

Volume 45, Issue 8, 15 October 2008, Pages 1178-1189
Free Radical Biology and Medicine

Original Contribution
Decreasing peroxiredoxin II expression decreases glutathione, alters cell cycle distribution, and sensitizes glioma cells to ionizing radiation and H2O2

https://doi.org/10.1016/j.freeradbiomed.2008.07.015Get rights and content

Abstract

Glioblastomas are notorious for their resistance to ionizing radiation and chemotherapy. We hypothesize that this resistance to ionizing radiation is due, in part, to alterations in antioxidant enzymes. Here, we show that rat and human glioma cells overexpress the antioxidant enzyme peroxiredoxin II (Prx II). Glioma cells in which Prx II is decreased using shRNA exhibit increased hyperoxidation of the remaining cellular Prxs, suggesting that the redox environment is more oxidizing. Of interest, decreasing Prx II does not alter other antioxidant enzymes (i.e., catalase, GPx, Prx I, Prx III, CuZnSOD, and MnSOD). Analysis of the redox environment revealed that decreasing Prx II increased intracellular reactive oxygen species in 36B10 cells; extracellular levels of H2O2 were also increased in both C6 and 36B10 cells. Treatment with H2O2 led to a further elevation in intracellular reactive oxygen species in cells where Prx II was decreased. Decreasing Prx II expression in glioma cells also reduced clonogenic cell survival following exposure to ionizing radiation and H2O2. Furthermore, lowering Prx II expression decreased intracellular glutathione and resulted in a significant decline in glutathione reductase activity, suggesting a possible mechanism for the observed increased sensitivity to oxidative insults. Additionally, decreasing Prx II expression increased cell cycle doubling times, with fewer cells distributed to S phase in C6 glioma cells and more cells redistributed to the most radiosensitive phase of the cell cycle, G2/M, in 36B10 glioma cells. These findings support the hypothesis that inhibiting Prx II sensitizes glioma cells to oxidative stress, presenting Prxs as potential therapeutic targets.

Section snippets

Materials

All chemicals and reagents were purchased from Sigma–Aldrich (St. Louis, MO, USA) unless stated otherwise. Medium and medium components were purchased from Invitrogen (Carlsbad, CA, USA).

Cell cultures

Primary rat astrocytes were isolated from the cerebral cortex of 2- to 3-day-old Sprague–Dawley rat pups as described previously [6]. Type II astrocytes were maintained in Dulbecco’s minimum essential medium (DMEM) supplemented with 10% fetal bovine serum, 4 mM L-glutamine, and 38.9 mM glucose. Medium was

Prx II is overexpressed in human and rat glioma cell lines

As astrocytes, the normal counterpart of glioma cells, express low levels of Prx II, overexpression of Prx II may be important for protecting glioma cells from endogenous and exogenous sources of oxidative stress. Therefore, several rat and human glioma cell lines were examined for expression of Prx II (Figs. 1A and B). When compared with primary rat astrocytes, Prx II levels were at least 4-fold higher in the rat glioma cell lines (Fig. 1A). Similarly, when human glioma cells were compared

Discussion

Gliomas are extremely refractory to IR and chemotherapy. Despite significant advances, median patient survival remains only 14.6 months with optimal therapy [1], [2], [61]. We hypothesized that this resistance is due, in part, to altered levels of antioxidant enzymes. We have previously shown that catalase is overexpressed in glioma cells and that this overexpression contributes to their radioresistance; inhibiting catalase expression sensitized glioma cells to oxidative stress [6]. We now

Acknowledgments

This work was supported by NIH Grants CA110274 (P.S.S.), CA112593 (M.E.R.), GM050389 (L.B.P.), andCA086862 and CA100045 (D.R.S.). We extend special thanks to Mitchell C. Coleman (University of Iowa) for analysis of cellular glutathione content and for technical assistance.

References (88)

  • G.J. DeYulia et al.

    EGF receptor–ligand interaction generates extracellular hydrogen peroxide that inhibits EGFR-associated protein tyrosine phosphatases

    Biochem. Biophys. Res. Commun.

    (2005)
  • S.P. Gabbita et al.

    Redox regulatory mechanisms of cellular signal transduction

    Arch. Biochem. Biophys.

    (2000)
  • C. Quan et al.

    Enhanced expression of peroxiredoxin I and VI correlates with development, recurrence and progression of human bladder cancer

    J. Urol.

    (2006)
  • S.W. Kang et al.

    Characterization of a mammalian peroxiredoxin that contains one conserved cysteine

    J. Biol. Chem.

    (1998)
  • S.W. Kang et al.

    Mammalian peroxiredoxin isoforms can reduce hydrogen peroxide generated in response to growth factors and tumor necrosis factor-alpha

    J. Biol. Chem.

    (1998)
  • A. Matsumoto et al.

    Cloning of the peroxiredoxin gene family in rats and characterization of the fourth member

    FEBS Lett.

    (1999)
  • M.S. Seo et al.

    Identification of a new type of mammalian peroxiredoxin that forms an intramolecular disulfide as a reaction intermediate

    J. Biol. Chem.

    (2000)
  • H. Mizusawa et al.

    Peroxiredoxin I (macrophage 23 kDa stress protein) is highly and widely expressed in the rat nervous system

    Neurosci. Lett.

    (2000)
  • T.D. Oberley et al.

    Localization of the thioredoxin system in normal rat kidney

    Free Radic. Biol. Med.

    (2001)
  • Z.A. Wood et al.

    Structure, mechanism and regulation of peroxiredoxins

    Trends Biochem. Sci.

    (2003)
  • A.B. Fisher et al.

    Phospholipid hydroperoxides are substrates for non-selenium glutathione peroxidase

    J. Biol. Chem.

    (1999)
  • T.S. Chang et al.

    Characterization of mammalian sulfiredoxin and its reactivation of hyperoxidized peroxiredoxin through reduction of cysteine sulfinic acid in the active site to cysteine

    J. Biol. Chem.

    (2004)
  • M.M. Bradford

    A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein–dye binding

    Anal. Biochem.

    (1976)
  • R.F. Beers et al.

    A spectrophotometric method for measuring the breakdown of hydrogen peroxide by catalase

    J. Biol. Chem.

    (1952)
  • R.A. Lawrence et al.

    Glutathione peroxidase activity in selenium-deficient rat liver

    Biochem. Biophys. Res. Commun.

    (1976)
  • L.W. Oberley et al.

    Assay of superoxide dismutase activity in tumor tissue

    Methods Enzymol.

    (1984)
  • O.W. Griffith

    Determination of glutathione and glutathione disulfide using glutathione reductase and 2-vinylpyridine

    Anal. Biochem.

    (1980)
  • O.H. Lowry et al.

    Protein measurement with the Folin phenol reagent

    J. Biol. Chem.

    (1951)
  • F. Denizot et al.

    Rapid colorimetric assay for cell growth and survival: modifications to the tetrazolium dye procedure giving improved sensitivity and reliability

    J. Immunol. Methods

    (1986)
  • F.Q. Schafer et al.

    Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple

    Free Radic. Biol. Med.

    (2001)
  • R. Franco et al.

    SLCO/OATP-like transport of glutathione in FasL-induced apoptosis: glutathione efflux is coupled to an organic anion exchange and is necessary for the progression of the execution phase of apoptosis

    J. Biol. Chem.

    (2006)
  • L.H. Lash

    Role of glutathione transport processes in kidney function

    Toxicol. Appl. Pharmacol.

    (2005)
  • T.H. Lee et al.

    Peroxiredoxin II is essential for sustaining life span of erythrocytes in mice

    Blood

    (2003)
  • X. Wang et al.

    Mice with targeted mutation of peroxiredoxin 6 develop normally but are susceptible to oxidative stress

    J. Biol. Chem.

    (2003)
  • Y.H. Han et al.

    Inhibitory role of peroxiredoxin II (Prx II) on cellular senescence

    FEBS Lett.

    (2005)
  • Y. Takahashi et al.

    Synchronized generation of reactive oxygen species with the cell cycle

    Life Sci.

    (2004)
  • P.A. Savitsky et al.

    Redox regulation of Cdc25C

    J. Biol. Chem.

    (2002)
  • T.S. Chang et al.

    Regulation of peroxiredoxin I activity by Cdc2-mediated phosphorylation

    J. Biol. Chem.

    (2002)
  • G. Draetta

    Cdc2 activation: the interplay of cyclin binding and Thr161 phosphorylation

    Trends Cell Biol.

    (1993)
  • J.C. Moon et al.

    Oxidative stress-dependent structural and functional switching of a human 2-Cys peroxiredoxin isotype II that enhances HeLa cell resistance to H2O2-induced cell death

    J. Biol. Chem.

    (2005)
  • A. Meister

    Glutathione metabolism and its selective modification

    J. Biol. Chem.

    (1988)
  • O.W. Griffith

    Biologic and pharmacologic regulation of mammalian glutathione synthesis

    Free Radic. Biol. Med.

    (1999)
  • D.M. Townsend et al.

    The importance of glutathione in human disease

    Biomed. Pharmacother.

    (2003)
  • M.C. Chamberlain et al.

    Practical guidelines for the treatment of malignant gliomas

    West. J. Med.

    (1998)
  • Cited by (0)

    View full text