Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Original Article
  • Published:

Role of TGF-β1 and nitric oxide in the bystander response of irradiated glioma cells

Abstract

The radiation-induced bystander effect (RIBE) increases the probability of cellular response and therefore has important implications for cancer risk assessment following low-dose irradiation and for the likelihood of secondary cancers after radiotherapy. However, our knowledge of bystander signaling factors, especially those having long half-lives, is still limited. The present study found that, when a fraction of cells within a glioblastoma population were individually irradiated with helium ions from a particle microbeam, the yield of micronuclei (MN) in the nontargeted cells was increased, but these bystander MN were eliminated by treating the cells with either aminoguanidine (an inhibitor of inducible nitric oxide (NO) synthase) or anti-transforming growth factor β1 (anti-TGF-β1), indicating that NO and TGF-β1 are involved in the RIBE. Intracellular NO was detected in the bystander cells, and additional TGF-β1 was detected in the medium from irradiated T98G cells, but it was diminished by aminoguanidine. Consistent with this, an NO donor, diethylamine nitric oxide (DEANO), induced TGF-β1 generation in T98G cells. Conversely, treatment of cells with recombinant TGF-β1 could also induce NO and MN in T98G cells. Treatment of T98G cells with anti-TGF-β1 inhibited the NO production when only 1% of cells were targeted, but not when 100% of cells were targeted. Our results indicate that, downstream of radiation-induced NO, TGF-β1 can be released from targeted T98G cells and plays a key role as a signaling factor in the RIBE by further inducing free radicals and DNA damage in the nontargeted bystander cells.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6

Similar content being viewed by others

References

  • Albertini RJ, Anderson D, Douglas GR, Hagmar L, Hemminki K, Merlo F et al. (2000). IPCS guidelines for the monitoring of genotoxic effects of carcinogens in humans. International Programme on Chemical Safety. Mutat Res 463: 111–172.

    Article  CAS  Google Scholar 

  • Arnold SF, Tims E, Bluman EM, McGrath BE . (1999). Regulation of transforming growth factor beta1 by radiation in cells of two human breast cancer cell lines. Radiat Res 152: 487–492.

    Article  CAS  Google Scholar 

  • Ayache N, Boumediene K, Mathy-Hartert M, Reginster JY, Henrotin Y, Pujol JP . (2002). Expression of TGF-betas and their receptors is differentially modulated by reactive oxygen species and nitric oxide in human articular chondrocytes. Osteoarthritis Cartilage 10: 344–352.

    Article  CAS  Google Scholar 

  • Azzam EI, de Toledo SM, Gooding T, Little JB . (1998). Intercellular communication is involved in the bystander regulation of gene expression in human cells exposed to very low fluences of alpha particles. Radiat Res 150: 497–504.

    Article  CAS  Google Scholar 

  • Azzam EI, de Toledo SM, Little JB . (2001). Direct evidence for the participation of gap junction-mediated intercellular communication in the transmission of damage signals from alpha-particle irradiated to nonirradiated cells. Proc Natl Acad Sci USA 98: 473–478.

    CAS  Google Scholar 

  • Barcellos-Hoff MH . (2005). Integrative radiation carcinogenesis: interactions between cell and tissue responses to DNA damage. Semin Cancer Biol 15: 138–148.

    Article  CAS  Google Scholar 

  • Bishayee A, Rao DV, Howell RW . (1999). Evidence for pronounced bystander effects caused by nonuniform distributions of radioactivity using a novel three-dimensional tissue culture model. Radiat Res 152: 88–97.

    Article  CAS  Google Scholar 

  • Blanco FJ, Geng Y, Lotz M . (1995). Differentiation-dependent effects of IL-1 and TGF-beta on human articular chondrocyte proliferation are related to inducible nitric oxide synthase expression. J Immunol 154: 4018–4026.

    CAS  Google Scholar 

  • Boerma M, Bart CI, J W . (2002). Effects of ionizing radiation on gene expression in cultured rat heart cells. Int J Radiat Biol 78: 219–225.

    Article  CAS  Google Scholar 

  • Burdak-Rothkamm S, Short SC, Folkard M, Rothkamm K, Prise KM . (2007). ATR-dependent radiation-induced gamma H2AX foci in bystander primary human astrocytes and glioma cells. Oncogene 26: 993–1002.

    Article  CAS  Google Scholar 

  • Chesrown SE, Monnier J, Visner G, Nick HS . (1994). Regulation of inducible nitric oxide synthase mRNA levels by LPS, INF-gamma, TGF-beta, and IL-10 in murine macrophage cell lines and rat peritoneal macrophages. Biochem Biophys Res Commun 200: 126–134.

    Article  CAS  Google Scholar 

  • Dedon PC, Tannenbaum SR . (2004). Reactive nitrogen species in the chemical biology of inflammation. Arch Biochem Biophys 423: 12–22.

    Article  CAS  Google Scholar 

  • Deshpande A, Goodwin EH, Bailey SM, Marrone BL, Lehnert BE . (1996). Alpha-particle-induced sister chromatid exchange in normal human lung fibroblasts: evidence for an extranuclear target. Radiat Res 145: 260–267.

    Article  CAS  Google Scholar 

  • Folkard M, Vojnovic B, Hollis KJ, Bowey AG, Watts SJ, Schettino G et al. (1997a). A charged-particle microbeam: II. A single-particle micro-collimation and detection system. Int J Radiat Biol 72: 387–395.

    Article  CAS  Google Scholar 

  • Folkard M, Vojnovic B, Prise KM, Bowey AG, Locke RJ, Schettino G et al. (1997b). A charged-particle microbeam: I. Development of an experimental system for targeting cells individually with counted particles. Int J Radiat Biol 72: 375–385.

    Article  CAS  Google Scholar 

  • Gizatullina ZZ, Grapengiesser E, Shabalina IG, Nedergaard J, Heldin CH, Aspenstrom P . (2003). Effect of transforming growth factor-beta on calcium homeostasis in prostate carcinoma cells. Biochem Biophys Res Commun 304: 643–649.

    Article  CAS  Google Scholar 

  • Hamby ME, Hewett JA, Hewett SJ . (2006). TGF-beta1 potentiates astrocytic nitric oxide production by expanding the population of astrocytes that express NOS-2. Glia 54: 566–577.

    Article  Google Scholar 

  • Han J, Hendzel MJ, Allalunis-Turner J . (2006). Quantitative analysis reveals asynchronous and more than DSB-associated histone H2AX phosphorylation after exposure to ionizing radiation. Radiat Res 165: 283–292.

    Article  CAS  Google Scholar 

  • Iyer R, Lehnert BE . (2000). Factors underlying the cell growth-related bystander responses to alpha particles. Cancer Res 60: 1290–1298.

    CAS  Google Scholar 

  • Jobling MF, Mott JD, Finnegan MT, Jurukovski V, Erickson AC, Walian PJ et al. (2006). Isoform-specific activation of latent transforming growth factor (LTGF-beta) by reactive oxygen species. Radiat Res 166: 839–848.

    Article  CAS  Google Scholar 

  • Keefer LK, Nims RW, Davies KM, Wink DA . (1996). ‘NONOates’ (1-substituted diazen-1-ium-1,2-diolates) as nitric oxide donors: convenient nitric oxide dosage forms. Methods Enzymol 268: 281–293.

    Article  CAS  Google Scholar 

  • Lewis DA, Mayhugh BM, Qin Y, Trott K, Mendonca MS . (2001). Production of delayed death and neoplastic transformation in cgl1 cells by radiation-induced bystander effects. Radiat Res 156: 251–258.

    Article  CAS  Google Scholar 

  • Limoli CL, Ponnaiya B, Corcoran JJ, Giedzinski E, Kaplan MI, Hartmann A et al. (2000). Genomic instability induced by high and low LET ionizing radiation. Adv Space Res 25: 2107–2117.

    Article  CAS  Google Scholar 

  • Little JB . (2000). Radiation carcinogenesis. Carcinogenesis 21: 397–404.

    Article  CAS  Google Scholar 

  • Lyng FM, Maguire P, McClean B, Seymour C, Mothersill C . (2006). The involvement of calcium and MAP kinase signaling pathways in the production of radiation-induced bystander effects. Radiat Res 165: 400–409.

    Article  CAS  Google Scholar 

  • Matsumoto H, Hayashi S, Hatashita M, Ohnishi K, Shioura H, Ohtsubo T et al. (2001). Induction of radioresistance by a nitric oxide-mediated bystander effect. Radiat Res 155: 387–396.

    Article  CAS  Google Scholar 

  • Matsumoto H, Hayashi S, Hatashita M, Shioura H, Ohtsubo T, Kitai R et al. (2000). Induction of radioresistance to accelerated carbon-ion beams in recipient cells by nitric oxide excreted from irradiated donor cells of human glioblastoma. Int J Radiat Biol 76: 1649–1657.

    Article  CAS  Google Scholar 

  • Morgan WF . (2003a). Non-targeted and delayed effects of exposure to ionizing radiation: I. Radiation-induced genomic instability and bystander effects in vitro. Radiat Res 159: 567–580.

    Article  CAS  Google Scholar 

  • Morgan WF . (2003b). Non-targeted and delayed effects of exposure to ionizing radiation: II. Radiation-induced genomic instability and bystander effects in vivo, clastogenic factors and transgenerational effects. Radiat Res 159: 581–596.

    Article  CAS  Google Scholar 

  • Mothersill C, Seymour C . (1997). Medium from irradiated human epithelial cells but not human fibroblasts reduces the clonogenic survival of unirradiated cells. Int J Radiat Biol 71: 421–427.

    Article  CAS  Google Scholar 

  • Mothersill C, Seymour CB . (1998). Cell-cell contact during gamma irradiation is not required to induce a bystander effect in normal human keratinocytes: evidence for release during irradiation of a signal controlling survival into the medium. Radiat Res 149: 256–262.

    Article  CAS  Google Scholar 

  • Nagasawa H, Little JB . (1992). Induction of sister chromatid exchanges by extremely low doses of alpha-particles. Cancer Res 52: 6394–6396.

    CAS  Google Scholar 

  • Nagasawa H, Little JB . (1999). Unexpected sensitivity to the induction of mutations by very low doses of alpha-particle radiation: evidence for a bystander effect. Radiat Res 152: 552–557.

    Article  CAS  Google Scholar 

  • Narayanan PK, Goodwin EH, Lehnert BE . (1997). Alpha particles initiate biological production of superoxide anions and hydrogen peroxide in human cells. Cancer Res 57: 3963–3971.

    CAS  Google Scholar 

  • Nesti LJ, Caterson EJ, Li WJ, Chang R, McCann TD, Hoek JB et al. (2007). TGF-beta1 calcium signaling in osteoblasts. J Cell Biochem 101: 348–359.

    Article  CAS  Google Scholar 

  • Newman E, Spratt DE, Mosher J, Cheyne B, Montgomery HJ, Wilson DL et al. (2004). Differential activation of nitric oxide synthase isozymes by calmodulin-troponin C chimeras. J Biol Chem 279: 33547–33557.

    Article  CAS  Google Scholar 

  • Prise KM, Belyakov OV, Folkard M, Michael BD . (1998). Studies of bystander effects in human fibroblasts using a charged particle microbeam. Int J Radiat Biol 74: 793–798.

    Article  CAS  Google Scholar 

  • Sawant SG, Randers-Pehrson G, Geard CR, Brenner DJ, Hall EJ . (2001). The bystander effect in radiation oncogenesis: I. Transformation in C3 H 10T1/2 cells in vitro can be initiated in the unirradiated neighbors of irradiated cells. Radiat Res 155: 397–401.

    Article  CAS  Google Scholar 

  • Schmidt A, Geigenmueller S, Voelker W, Seiler P, Buddecke E . (2003). Exogenous nitric oxide causes overexpression of TGF-beta1 and overproduction of extracellular matrix in human coronary smooth muscle cells. Cardiovasc Res 58: 671–678.

    Article  CAS  Google Scholar 

  • Seymour CB, Mothersill C . (1997). Delayed expression of lethal mutations and genomic instability in the progeny of human epithelial cells that survived in a bystander-killing environment. Radiat Oncol Investig 5: 106–110.

    Article  CAS  Google Scholar 

  • Shao C, Aoki M, Furusawa Y . (2003a). Bystander effect on cell growth stimulation in neoplastic HSGc cells induced by heavy-ion irradiation. Radiat Environ Biophys 42: 183–187.

    Article  CAS  Google Scholar 

  • Shao C, Aoki M, Furusawa Y . (2004a). Bystander effect in lymphoma cells vicinal to irradiated neoplastic epithelial cells: nitric oxide is involved. J Radiat Res (Tokyo) 45: 97–103.

    Article  CAS  Google Scholar 

  • Shao C, Folkard M, Michael BD, Prise KM . (2004b). Targeted cytoplasmic irradiation induces bystander responses. Proc Natl Acad Sci USA 101: 13495–13500.

    Article  CAS  Google Scholar 

  • Shao C, Folkard M, Michael BD, Prise KM . (2005). Bystander signaling between glioma cells and fibroblasts targeted with counted particles. Int J Cancer 116: 45–51.

    Article  CAS  Google Scholar 

  • Shao C, Lyng FM, Folkard M, Prise KM . (2006). Calcium fluxes modulate the radiation-induced bystander responses in targeted glioma and fibroblast cells. Radiat Res 166: 479–487.

    Article  CAS  Google Scholar 

  • Shao C, Stewart V, Folkard M, Michael BD, Prise KM . (2003b). Nitric oxide-mediated signaling in the bystander response of individually targeted glioma cells. Cancer Res 63: 8437–8442.

    CAS  Google Scholar 

  • Sokolov MV, Smilenov LB, Hall EJ, Panyutin IG, Bonner WM, Sedelnikova OA . (2005). Ionizing radiation induces DNA double-strand breaks in bystander primary human fibroblasts. Oncogene 24: 7257–7265.

    Article  CAS  Google Scholar 

  • Tartier L, Gilchrist S, Burdak-Rothkamm S, Folkard M, Prise KM . (2007). Cytoplasmic irradiation induces mitochondrial-dependent 53BP1 protein relocalization in irradiated and bystander cells. Cancer Res 67: 5872–5879.

    Article  CAS  Google Scholar 

  • Vodovotz Y, Chesler L, Chong H, Kim SJ, Simpson JT, DeGraff W et al. (1999). Regulation of transforming growth factor beta1 by nitric oxide. Cancer Res 59: 2142–2149.

    CAS  Google Scholar 

  • Yang H, Asaad N, Held KD . (2005). Medium-mediated intercellular communication is involved in bystander responses of X-ray-irradiated normal human fibroblasts. Oncogene 24: 2096–2103.

    Article  CAS  Google Scholar 

  • Zhou H, Randers-Pehrson G, Waldren CA, Vannais D, Hall EJ, Hei TK . (2000). Induction of a bystander mutagenic effect of alpha particles in mammalian cells. Proc Natl Acad Sci USA 97: 2099–2104.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to Stuart Gilchrist and Bob Sunderland for assistance with the microbeam irradiation. We acknowledge the support of Cancer Research UK (CUK) grant number C1513/A7047, the European Commission (NOTE, FI6R 036465) and the Gray Cancer Institute in these studies.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to K M Prise.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Shao, C., Folkard, M. & Prise, K. Role of TGF-β1 and nitric oxide in the bystander response of irradiated glioma cells. Oncogene 27, 434–440 (2008). https://doi.org/10.1038/sj.onc.1210653

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/sj.onc.1210653

Keywords

This article is cited by

Search

Quick links