Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Increased Frequency of Spontaneous Neoplastic Transformation in Progeny of Bystander Cells from Cultures Exposed to Densely Ionizing Radiation

  • Manuela Buonanno,

    Affiliations Department of Radiology, University of Medicine and Dentistry of New Jersey (UMDNJ), New Jersey Medical School Cancer Center, Newark, New Jersey, United States of America, University of Medicine and Dentistry of New Jersey (UMDNJ), Graduate School of Biomedical Sciences, Newark, New Jersey, United States of America

  • Sonia M. de Toledo,

    Affiliation Department of Radiology, University of Medicine and Dentistry of New Jersey (UMDNJ), New Jersey Medical School Cancer Center, Newark, New Jersey, United States of America

  • Edouard I. Azzam

    azzamei@umdnj.edu

    Affiliations Department of Radiology, University of Medicine and Dentistry of New Jersey (UMDNJ), New Jersey Medical School Cancer Center, Newark, New Jersey, United States of America, University of Medicine and Dentistry of New Jersey (UMDNJ), Graduate School of Biomedical Sciences, Newark, New Jersey, United States of America

Abstract

An increased risk of carcinogenesis caused by exposure to space radiation during prolonged space travel is a limiting factor for human space exploration. Typically, astronauts are exposed to low fluences of ionizing particles that target only a few cells in a tissue at any one time. The propagation of stressful effects from irradiated to neighboring bystander cells and their transmission to progeny cells would be of importance in estimates of the health risks of exposure to space radiation. With relevance to the risk of carcinogenesis, we investigated, in model C3H 10T½ mouse embryo fibroblasts (MEFs), modulation of the spontaneous frequency of neoplastic transformation in the progeny of bystander MEFs that had been in co-culture 10 population doublings earlier with MEFs exposed to moderate doses of densely ionizing iron ions (1 GeV/nucleon) or sparsely ionizing protons (1 GeV). An increase (P<0.05) in neoplastic transformation frequency, likely mediated by intercellular communication through gap junctions, was observed in the progeny of bystander cells that had been in co-culture with cells irradiated with iron ions, but not with protons.

Introduction

The experience gained from space missions over the past 50 years has highlighted the necessity of humans for the exploration of deep space [1]. However, the health risks, in particular the increased risk of cancer, that may be associated with exposure to space radiation represents a major limitation to this activity [2]. To control and mitigate these risks, the National Aeronautics and Space Agency (NASA) has emphasized the need for ground-based characterization of the biological effects of space radiation and elucidation of the mediating mechanisms. These studies must be investigated in the context of low dose, low fluence, and low dose-rate effects.

During interplanetary missions, astronauts are exposed to different types of radiation [3]. Galactic cosmic rays ranging from energetic protons of low linear energy transfer (LET), to helium particles and particles of high charge and high energy (HZE) with high LET, as well as transient radiation from solar particle events (protons and small percentage of energetic ions) are the primary source of ionizing radiation in outer space.

Though less abundant than sparsely ionizing protons, HZE particles are highly ionizing with much greater potential to induce cell and tissue injury. The chemical reactants that they produce at high concentration along their trajectory interact efficiently with cellular molecules causing both covalent modifications and changes in cell signaling [4]. The net effect is clustered damage in DNA and other molecules [5] that occur shortly after irradiation and can extend along a long column of cells in tissue [6], and perturbation of oxidative metabolism that results in delayed damage [7]. Clustered DNA damage challenges repair systems and often is misrepaired (reviewed in [8]) leading to chromosomal changes that may predispose to cancer. Therefore, the risk of carcinogenesis from exposure to low fluence HZE particles may be greater in magnitude than caused by low doses of common terrestrial forms of radiation, such as γ rays [9], [10]. Epidemiological studies in humans would be ideal to evaluate the health risks induced by exposure to space radiation. However, such studies are likely to remain limited for years to come, given the relatively insignificant number of humans exposed to HZE particles. Thus, experimental studies in animal and cell culture models are essential to understand biological effects and their underlying mechanisms, which may help estimate corresponding risks for humans [11]. Such investigations would complement modeling studies based on extrapolation of epidemiological data obtained from the survivors of the A-bomb, nuclear accidents, and industrial activities involving exposure to radiation [12].

Risk estimates of carcinogenesis induced by space radiation, particularly at low doses and low fluences, are further complicated by accumulating experimental evidence indicating that the deleterious effects of certain radiation types (e.g. alpha or HZE particles) may manifest also in non-irradiated bystander cells in the exposed population (reviewed in [13]). Important biological responses, including genetic changes, have been shown to arise in non-irradiated cells that were in the vicinity of directly targeted cells or recipient of growth medium from irradiated cell cultures [14]. Recently, stressful bystander effects were shown to be induced by the HZE component of space radiation [15], [16]. Furthermore, such effects are propagated in progeny cells [17]. Progeny of bystander cells from normal human cell cultures exposed to HZE particles (1 GeV/nucleon iron ions or 600 MeV/nucleon silicon ions) harbored higher levels of chromosomal damage and oxidative stress, as judged by protein carbonylation and lipid peroxidation, than respective control cells [17]. DNA damage and oxidative stress act at all stages of carcinogenesis [18]. However, the risk of neoplastic transformation in bystander cells remains unclear; if it exists, it would have profound implications for the health risks associated with human space exploration. It would show that the risk is greater than predicted solely from the biological responses triggered in the targeted cells.

Using a model cell culture system to investigate malignant transformation [19], here we show that 1 GeV/nucleon iron ions induce a significant increase in the frequency of spontaneous neoplastic transformation in the progeny of bystander C3H 10T½ mouse embryo fibroblasts (MEFs),when assayed 10 generations after they had been in co-culture with irradiated C3H 10T½ MEFs.

Results and Discussion

Using a layered tissue culture system [17], we measured the frequency of spontaneous neoplastic transformation in the progeny of bystander C3H 10T½ MEFs that had been in co-culture 10 population doublings earlier with confluent MEFs that were irradiated with energetic iron ions (1 GeV/nucleon). Briefly, C3H 10T½ cells destined to be bystanders were seeded onto inverted Transwell® inserts with 3 µm pores. Following attachment, the inserts were inverted and placed into the wells of plates and cultured to form an adherent confluent monolayer. Irradiated or sham-treated cells, derived from confluent cultures maintained in flasks, were harvested within 10 min after exposure to a 25 cGy dose from energetic iron ions. At this dose, most cells (87%) in the exposed population were traversed through the nucleus on average by 2 iron ion tracks, which would result in an absorbed dose of ∼30 cGy/cell [20]. The harvested cells were then seeded at confluent density on top of the insert with confluent bystander cells growing at the bottom of it. Within 2 h after plating, irradiated cells adhered and formed functional junctional channels with bystander cells, as assessed by the transfer of calcein dye [21]. Irradiated and bystander cells may also communicate with each other through diffusible factors transferred across the pores of the membrane. The directly irradiated and bystander cells were left in co-culture for a total of 5 h. Subsequently, bystander cells were harvested, grown, and serially subcultured for progeny studies upon reaching the confluent state. Control experiments whereby the irradiated cells were labeled with a cell tracker dye (Cell Tracker Orange CMTMR) confirmed that these cells do not move through the 3 ìm pores of the membrane, and the purity of the bystander population was confirmed by microscopy and flow cytometry analyses. In order to include all the progeny cells derived from the initial bystander cell population in the analyses, at each passage, the cells were subcultured in increasingly larger size flasks upon reaching the confluent state. Following 10 population doublings that occurred in ∼2 weeks, the cells were harvested and assayed for clonogenic survival and neoplastic transformation. When scoring transformed foci, type II or III foci according to the classification of Reznikoff et al. [19] were considered in the analyses.

The frequency of neoplastic transformation was evaluated by three different methods, namely transformation frequency per viable cell, per cell at risk, and by applying the null method [22] to the combined results of 3 independent experiments (Table 1). In every case, relative to control, a significant increase in the spontaneous neoplastic transformation frequency was observed. This increase was associated with an increase (P<0.01) in cell viability (Table 1, 14.1±0.6% in bystander cells co-cultured with irradiated cells vs. 8.2±1.0% in bystander cell co-cultured with sham-irradiated cells). When assessed by the null method, the progeny of bystander cells expressed a 2-fold increase (within 95% confidence interval) in transformation frequency [(2.7±0.5)×10−3 vs. (1.3±0.2) x 10−3 in control] (Figure 1). Similarly, the increase in spontaneous neoplastic potential in bystander cells was significant when the data were analyzed in terms of the number of foci per flask (P<0.0001) or the number of flasks without foci (P<0.005) (Table 2).

thumbnail
Figure 1. Spontaneous neoplastic transformation frequency in the progeny of bystander C3H 10T½ MEFs.

Co-culture of bystander C3H 10T½ MEFs with MEFs exposed to 0 or 25 cGy of 1 GeV/nucleon iron ions results in increased frequency of spontaneous neoplastic transformation in the distant progeny of the bystander cells. Neoplastic transformation frequencies and standard errors of the mean (SEM) were calculated with the null method applied to combined data from 3 independent experiments.

https://doi.org/10.1371/journal.pone.0021540.g001

thumbnail
Table 1. Frequency of spontaneous neoplastic transformation in bystander cells.

https://doi.org/10.1371/journal.pone.0021540.t001

thumbnail
Table 2. Neoplastic transformation in bystander cells expressed as number of foci/flask or flasks without foci.

https://doi.org/10.1371/journal.pone.0021540.t002

Traditionally, the important biological effects of radiation were assumed to arise in directly irradiated cells; bystander cells in the vicinity of irradiated cells would be unaffected [23]. Using an endpoint that is relevant to the risk of carcinogenesis, we show that the stressful effects of energetic iron ions are greater than predicted by traditional assumptions. The above results are opposite to the observations that exposure to low doses of γ rays, a radiation with low LET character, induces protective effects that reduce the transformation frequency to below the spontaneous level [24], [25]. In this context, during space travel or in the course of radiotherapy with HZE particles, low LET δ rays generated from HZE particle traversals may induce signaling events that trigger protective effects that may reduce the risk of neoplastic transformation [26]. Further, during space travel, δ rays are thought to traverse each cell in a human body about once per day [27]. However, when co-cultured with irradiated cells in the layered tissue culture system used in the present study, bystander cells did not receive δ rays. Investigating the effect of sequential exposure to low LET protons or γ rays followed by high LET HZE particles on the frequency of neoplastic transformation in irradiated and bystander cells would complement these studies and is pertinent to radiation protection and to radiotherapy, as patients receive diagnostic procedures prior to treatment [28].

Consistent with the lack of induction of DNA damage and oxidative stress in progeny of bystander cells from cultures exposed to energetic protons, a low LET radiation [17], co-culture of bystander C3H 10T½ MEFs with MEFs exposed to 100 cGy of 1 GeV protons (LET ∼0.2 keV/µm), did not increase the frequency of spontaneous neoplastic transformation above background level. Rather, the transformation frequency calculated by the null method was slightly lower than control [(6.6±0.4)×10−3 vs. (7.3±0.2)×10−3 in control]. These results are not likely due to alterations in the cloning efficiency of the bystander cells; the cloning efficiency was similar to that of progeny of bystander cells that were co-cultured with iron ion-irradiated cells. These data expand our previous studies showing that the induction of bystander stressful effects strongly depends on the rate of energy deposition per unit length along the particle trajectory (i.e. LET). Doses ranging from 100 to 400 cGy of low LET protons or γ rays did not result in the propagation of stressful bystander effects as assessed by the endpoints of cloning efficiency, micronucleus formation and markers of protein or lipid oxidation [29], [30].

Several mechanisms have been implicated in the propagation of radiation-induced bystander effects. They include oxidative metabolism, direct and indirect modes of intercellular communication, physical contact and likely other factors [31], [32]. Gap junctions linking contiguous cells were shown, by direct approaches, to mediate the propagation of signaling events that result in DNA damage and oxidative stress from cells exposed to α particles (a high LET radiation) to neighboring bystander cells [33]. Similar to irradiation with iron ions, we have also observed that co-culture of C3H 10T½ MEFs exposed to a mean dose of 80 cGy from 3.2 MeV α particles (LET ∼122 keV/µm) with bystander MEFs resulted in increased (P<0.05) frequency of spontaneous neoplastic transformation in the progeny of bystander cells [(3.9±0.4)×10−3 vs. (0.4±0.3)×10−3 in control, calculated by the null method]. Consistent with a prominent role for junctional communication in mediating the observed effect [34], the frequency of transformed cells was reduced [(3.9±0.4 vs. 2.3±0.4)×10−3] when the irradiated and bystander cells were co-cultured in the presence of á-glycyrrhetinic acid (AGA), a gap junction inhibitor.

Although it was shown that cells derived from type II and III C3H 10T½ foci form tumors when injected subcutaneously in syngeneic mice [35], [36], the use of results from tissue culture experiments in human health risk assessment must be done with caution. Nevertheless, neoplastic transformation experiments have been useful to predict radiation-induced carcinogenesis in humans exposed to domestic radon [37], and were consistent with risk estimations for leukemia and breast cancer derived by epidemiological findings [38]. Quantitative oncogenic transformation experiments in normal human cells would be preferred to those in MEFs, however, these are difficult to achieve [39].

In summary, our results demonstrate by a biological endpoint that is of relevance to carcinogenesis that stressful bystander effects are induced by the high LET component of space radiation and persist in progeny cells. They extend those of others which suggested that cellular exposure to low fluence á particles induces neoplastic transformation in bystander cells [40]. They show a 2 to 3 fold-increase in neoplastic transformation in progeny of bystander cells (Table 1). This increase is similar to that induced in C3H 10T½ cells targeted by one or two á particles [41], and greater than that observed in HeLa x skin fibroblast cell line CGL1 directly targeted by 1 GeV/nucleon iron ions [26]. In the latter cells, an increase in the transformation frequency occurred at doses greater than 20 cGy to the exposed cell cultures.

In addition to their relevance in estimates of health risks during extended space missions, the results are also pertinent to radiotherapy, as particle therapy with energetic protons or heavy ions (e.g. carbon ions) is increasingly being used in cancer treatment [42], [43]. Neoplastic transformation of progeny of bystander normal cells surrounding the irradiated tumor may increase the risk of second malignancies after treatment. In this respect, it would be informative to expand the current studies with HZE particles and investigate the effect of higher doses relevant to cancer therapy.

Materials and Methods

Cells

Mouse embryo fibroblasts, C3H 10T½ clone 8, obtained from the American Type Tissue Culture Collection, were cultured as previously described [44]. Briefly, cells at passage 8–10 were grown in Dulbecco Minimum Essential Medium (DMEM)/F-12 (Mediatech) supplemented with 10% heat-inactivated FCS (CellGro), 2 mM alanyl-L-glutamine, 100 U/mL penicillin and 100 µg/mL streptomycin (Sigma). Cells were routinely maintained in a 37° C humidified incubator in an atmosphere of 5% CO2 in air. Cells destined for irradiation were seeded in tissue culture flasks at a density that allowed them to reach the density-inhibited state within 5 days. They were then fed twice on alternate days, and experiments were initiated 24–48 h after the last feeding. Under these conditions, ∼90% of the cells were in G0/G1 phase of the cell cycle as determined by [3H]-thymidine uptake and/or flow cytometry. Synchronization of the cells in G0/G1 phase by density-inhibition eliminates complications in interpretation of the results, which arises from changes in cellular radiation sensitivity at different phases of the cell cycle [45]. Under these cell culture conditions, the colony forming efficiency of CH3 10 T½ MEFs was ∼15%.

The irradiated cells were co-cultured with confluent bystander cells (∼90% in G0/G1) grown onto the inverted Transwell inserts. Under these conditions, the colony forming efficiency of the bystander cells varied between 6 and 10%. The cells appeared healthy; they harbored similar level of micronuclei and proliferated at the same rate as cells grown in flasks (doubling time ∼ 16 h).

Inhibition of Gap Junction Communication

18-α-glycyrrhetinic acid (AGA) (Sigma), a reversible inhibitor of gap junction communication, was dissolved in 99.5% dimethyl sulfoxide (DMSO) and added to cell cultures at a concentration of 50 µM. At this concentration, AGA was not toxic to C3H 10T½ cells incubated in the presence of the drug for 5 h. Control cell cultures were incubated with the dissolving vehicle (0.25% DMSO).

Irradiation

Iron ion (1 GeV/nucleon 56Fe26+) and proton (1GeV 1H+) irradiations were conducted at the NASA Space Radiation Laboratory (NSRL) located at the Brookhaven National Laboratory (Upton, NY). Description of the facility and radiation beam information can be found at www.bnl.gov/medical/NASA/LTSF.asp. The culture flasks were positioned perpendicular to the beam so that the irradiating particles impacted first the plastic of the culture vessel, followed by the adherent cells and then the growth medium. Confluent cultures were exposed to 25 cGy (50 cGy/min) from iron ions or to 100 cGy (100 cGy/min) from protons. Whereas protons result in uniform irradiation of the cell population, according to Charlton and Sephton [20], exposure to a mean dose of 25 cGy from iron ions results in ∼87% of nuclei (mean nuclear area of a C3H 10T½ cell being ∼203 µm2 [46]) in a confluent population being traversed on average by 2 particles (∼15 cGy/particle hit) and 13% of nuclei being unhit. At the place where the culture flasks were positioned, the LET was estimated to be ∼151 keV/µm for iron ions and 0.2 keV/µm for protons. The flasks were filled to capacity with growth medium that was pH- and temperature-equilibrated, 3 h before the radiation exposure. This ensured that during the irradiation, temperature fluctuations were attenuated and the cells were immersed in medium, which alleviates changes in osmolarity and partial oxygen tension, parameters that greatly affect the cellular response to radiation [47], [48]. Control cells were sham-treated and handled in parallel with the test cultures. The experiments with HZE particles were carried out during different NSRL runs between 2008 and 2010, and dosimetry was performed by the NSRL physics staff.

For irradiation with α particles, cells were exposed at 37 °C in a 5% CO2 atmosphere to a 0.0002 Ci 241Am-collimated source housed in a helium-filled Plexiglas box at a dose rate of 2 cGy/min as we have previously described [21]. Irradiation was carried out from below with α particles with an average energy of 3.2 MeV (LET ∼122 keV/µm) at contact with the cells.

Neoplastic Transformation

Progeny of bystander C3H 10T½ cells were assayed for clonogenic survival and neoplastic transformation as described [36]. Briefly, for each treatment, fifteen 75 cm2 flasks were seeded with ∼1 × 103 cells per flask. The cells were then fed once a week for the subsequent 6–7 weeks with DMEM/F-12 medium supplemented with 5% heat-inactivated FCS. The cultures were then fixed in 95% ethanol and stained with Giemsa (1∶20 in PBS, pH 7.4). Neoplastic transformation frequencies were estimated using morphological criteria. A transformed colony is a focus of highly polar, multilayered criss-crossed array of densely stained cells classified as type II or type III foci [36]. Transformed foci are dense and discrete and can be easily distinguished from the flat background of control cells or type I foci. Further, they induce tumors when injected subcutaneously in mice [35], [36]. To estimate clonogenic survival, three flasks per treatment were fixed following a 9 day-incubation period. Colonies consisting of 50 or more cells were considered in the analyses [49].

Type II and III foci were considered and the data were analyzed by summing the results of 3 separate experiments; pooled data from any condition were compared to pooled data from control samples. Neoplastic transformation frequencies were calculated per viable cell (i.e. colony forming cell) (± the standard error), per cell at risk, and using the null method. The latter calculates the transformation frequency per viable cell, and eliminates possible errors from satellite colony formation as a consequence of the multiple feedings during the 42–49 days post-irradiation expression period. It compares the average number of transformed foci per flask (λ), with the fraction of flasks without foci (f = n/N, where n is the number of flasks without foci and N is the total number of flasks) and assumes that the potential number of foci follows a Poisson distribution (λ =  -ln f = -ln (n/N)). The transformation frequency is given by λ, N−S. = −, ln(,n-N.), N−S.., where S is the total number of viable cells. This method also allows for calculation of the standard error of the mean (SEM) =  ,−(, 1−n.−, 1−N)., and the 95% confidence interval (twice the standard error of the mean) [22].

In Table 2, the Fisher exact test was used to calculate the P values when neoplatic transformation was evaluated by comparing the number of foci per flask or the number of flasks without foci in bystander cells that were co-cultured with sham-irradiated or irradiated cells, respectively. Different approaches were adopted to analyze the frequency of neoplastic transformation as a significant increase may be observed by one method but not the other.

Acknowledgments

We thank Drs. Adam Rusek, Michael Sivertz, I-Hung Chang, Peter Guida, and their colleagues for their support during the experiments at the NASA Space Radiation Laboratory. We also thank Dr. Roger W. Howell for his critical input, and Narongchai Autsavapromporrn, Geraldine Gonon, Min Li, and Jie Zhang for their excellent support.

Author Contributions

Conceived and designed the experiments: SMdT MB EIA. Performed the experiments: SMdT MB. Analyzed the data: MB EIA. Wrote the paper: MB EIA.

References

  1. 1. White RJ, Averner M (2001) Humans in space. Nature 409: 1115–1118.
  2. 2. Committee on the Evaluation of Radiation Shielding for Space Exploration NRC (2008) Managing Space Radiation Risk in the New Era of Space Exploration. The National Academy Press.
  3. 3. Cucinotta FA, Durante M (2006) Cancer risk from exposure to galactic cosmic rays: implications for space exploration by human beings. Lancet Oncol 7: 431–435.
  4. 4. Ding LH, Shingyoji M, Chen F, Hwang JJ, Burma S, et al. (2005) Gene expression profiles of normal human fibroblasts after exposure to ionizing radiation: a comparative study of low and high doses. Radiat Res 164: 17–26.
  5. 5. Sutherland BM, Bennett PV, Saparbaev M, Sutherland JC, Laval J (2001) Clustered DNA damages as dosemeters for ionising radiation exposure and biological responses. Radiat Prot Dosimetry 97: 33–38.
  6. 6. Goodhead DT (1988) Spatial and temporal distribution of energy. Health Phys 55: 231–240.
  7. 7. Spitz DR, Azzam EI, Li JJ, Gius D (2004) Metabolic oxidation/reduction reactions and cellular responses to ionizing radiation: a unifying concept in stress response biology. Cancer Metastasis Rev 23: 311–322.
  8. 8. Georgakilas AG (2008) Processing of DNA damage clusters in human cells: current status of knowledge. Mol Biosyst 4: 30–35.
  9. 9. Sutherland BM, Bennett PV, Schenk H, Sidorkina O, Laval J, et al. (2001) Clustered DNA damages induced by high and low LET radiation, including heavy ions. Phys Med 17: Suppl 1202–204.
  10. 10. Chappell LJ, Whalen MK, Gurai S, Ponomarev A, Cucinotta FA, et al. (2010) Analysis of flow cytometry DNA damage response protein activation kinetics after exposure to x rays and high-energy iron nuclei. Radiat Res 174: 691–702.
  11. 11. Shore RE (2009) Low-dose radiation epidemiology studies: status and issues. Health Phys 97: 481–486.
  12. 12. Preston DL, Shimizu Y, Pierce DA, Suyama A, Mabuchi K (2003) Studies of mortality of atomic bomb survivors. Report 13: Solid cancer and noncancer disease mortality: 1950-1997. Radiat Res 160: 381–407.
  13. 13. Hamada N, Maeda M, Otsuka K, Tomita M (2011) Signaling Pathways Underpinning the Manifestations of Ionizing Radiation-Induced Bystander Effects. Curr Mol Pharmacol 4: 79–95.
  14. 14. Prise KM, O'Sullivan JM (2009) Radiation-induced bystander signalling in cancer therapy. Nat Rev Cancer 9: 351–360.
  15. 15. Shao C, Furusawa Y, Kobayashi Y, Funayama T, Wada S (2003) Bystander effect induced by counted high-LET particles in confluent human fibroblasts: a mechanistic study. FASEB J 17: 1422–1427.
  16. 16. Yang H, Anzenberg V, Held KD (2007) The time dependence of bystander responses induced by iron-ion radiation in normal human skin fibroblasts. Radiat Res 168: 292–298.
  17. 17. Buonanno M, de Toledo SM, Pain D, Azzam EI (2011) Long-term consequences of radiation-induced bystander effects depend on radiation quality and dose and correlate with oxidative stress. Radiat Res 175: 405–415.
  18. 18. Cerutti PA (1985) Prooxidant states and tumor promotion. Science 227: 375–381.
  19. 19. Reznikoff CA, Brankow DW, Heidelberger C (1973) Establishment and characterization of a cloned line of C3H mouse embryo cells sensitive to postconfluence inhibition of division. Cancer Res 33: 3231–3238.
  20. 20. Charlton DE, Sephton R (1991) A relationship between microdosimetric spectra and cell survival for high-LET irradiation. Int J Radiat Biol 59: 447–457.
  21. 21. Azzam EI, de Toledo SM, Little JB (2003) Expression of CONNEXIN43 is highly sensitive to ionizing radiation and other environmental stresses. Cancer Res 63: 7128–7135.
  22. 22. Balcer-Kubiczek EK, Harrison GH, Thompson BW (1987) Repair time for oncogenic transformation in C3H/10T1/2 cells subjected to protracted X-irradiation. Int J Radiat Biol Relat Stud Phys Chem Med 51: 219–226.
  23. 23. Little JB (2003) Genomic instability and bystander effects: a historical perspective. Oncogene 22: 6978–6987.
  24. 24. Azzam EI, de Toledo SM, Raaphorst GP, Mitchel RE (1996) Low-dose ionizing radiation decreases the frequency of neoplastic transformation to a level below the spontaneous rate in C3H 10T1/2 cells. Radiat Res 146: 369–373.
  25. 25. Redpath JL, Antoniono RJ (1998) Induction of an adaptive response against spontaneous neoplastic transformation in vitro by low-dose gamma radiation. Radiat Res 149: 517–520.
  26. 26. Elmore E, Lao XY, Kapadia R, Redpath JL (2009) Threshold-type dose response for induction of neoplastic transformation by 1 GeV/nucleon iron ions. Radiat Res 171: 764–770.
  27. 27. Cucinotta FA, Nikjoo H, Goodhead DT (2000) Model for radial dependence of frequency distributions for energy imparted in nanometer volumes from HZE particles. Radiat Res 153: 459–468.
  28. 28. NCRP (2006) Information needed to make radiation protection recommendations for space missions beyond low-earth orbit. Bethesda: National Council on Radiation Protection and Measurements. Report No. 153 Report No 153.
  29. 29. Autsavapromporn N, de Toledo SM, Buonanno M, Jay-Gerin JP, Harris AL, et al. (2011) Intercellular Communication Amplifies Stressful Effects in High-Charge, High-Energy (HZE) Particle -Irradiated Human Cells. Journal of Radiation Research. In press.
  30. 30. Autsavapromporn N, de Toledo SM, Little JB, Jay-Gerin JP, Harris AL, et al. (2011) The role of gap-junction communication and oxidative stress in the propagation of toxic effects among high dose alpha particle-irradiated human cells Radiat Res 175: 345–357.
  31. 31. Gerashchenko BI, Howell RW (2005) Bystander cell proliferation is modulated by the number of adjacent cells that were exposed to ionizing radiation. Cytometry A 66: 62–70.
  32. 32. Hei TK, Zhou H, Ivanov VN, Hong M, Lieberman HB, et al. (2008) Mechanism of radiation-induced bystander effects: a unifying model. J Pharm Pharmacol 60: 943–950.
  33. 33. 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 U S A 98: 473–478.
  34. 34. Trosko JE, Ruch RJ (2002) Gap junctions as targets for cancer chemoprevention and chemotherapy. Curr Drug Targets 3: 465–482.
  35. 35. Raaphorst GP, Azzam EI (1986) Fixation and repair by anisotonic treatment of radiation damage leading to oncogenic transformation. Int J Radiat Biol Relat Stud Phys Chem Med 49: 383–393.
  36. 36. Reznikoff CA, Bertram JS, Brankow DW, Heidelberger C (1973) Quantitative and qualitative studies of chemical transformation of cloned C3H mouse embryo cells sensitive to postconfluence inhibition of cell division. Cancer Res 33: 3239–3249.
  37. 37. Brenner DJ, Miller RC, Huang Y, Hall EJ (1995) The biological effectiveness of radon-progeny alpha particles. III. Quality factors. Radiat Res 142: 61–69.
  38. 38. Elmore E, Lao XY, Kapadia R, Redpath JL (2006) The effect of dose rate on radiation-induced neoplastic transformation in vitro by low doses of low-LET radiation. Radiat Res 166: 832–838.
  39. 39. Rhim JS (1993) Neoplastic transformation of human cells in vitro. Crit Rev Oncog 4: 313–335.
  40. 40. Sawant SG, Randers-Pehrson G, Geard CR, Brenner DJ, Hall EJ (2001) The bystander effect in radiation oncogenesis: I. Transformation in C3H 10T1/2 cells in vitro can be initiated in the unirradiated neighbors of irradiated cells. Radiat Res 155: 397–401.
  41. 41. Miller RC, Randers-Pehrson G, Geard CR, Hall EJ, Brenner DJ (1999) The oncogenic transforming potential of the passage of single alpha particles through mammalian cell nuclei. Proc Natl Acad Sci U S A 96: 19–22.
  42. 42. Durante M, Loeffler JS (2009) Charged particles in radiation oncology. Nat Rev Clin Oncol 7: 37–43.
  43. 43. Okada T, Kamada T, Tsuji H, Mizoe JE, Baba M, et al. (2010) Carbon ion radiotherapy: clinical experiences at National Institute of Radiological Science (NIRS). J Radiat Res (Tokyo) 51: 355–364.
  44. 44. Azzam EI, Raaphorst GP, Mitchel RE (1994) Radiation-induced adaptive response for protection against micronucleus formation and neoplastic transformation in C3H 10T1/2 mouse embryo cells. Radiat Res 138: S28–31.
  45. 45. Terasima T, Tolmach LJ (1963) X-ray sensitivity and DNA synthesis in synchronous populations of HeLa cells. Science 140: 490–492.
  46. 46. Schillaci ME, Carpenter S, Raju MR, Sebring RJ, Wilder ME, et al. (1989) Radiobiology of ultrasoft X rays. II. Cultured C3H mouse cells (10T1/2). Radiat Res 118: 83–92.
  47. 47. Rueckert RR, Mueller GC (1960) Effect of oxygen tension on HeLa cell growth. Cancer Res 20: 944–949.
  48. 48. Gray LH, Conger AD, Ebert M, Hornsey S, Scott OC (1953) The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. Br J Radiol 26: 638–648.
  49. 49. Puck TT, Marcus PI (1956) Action of x-rays on mammalian cells. J Exp Med 103: 653–666.