Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Carcinogenesis

Extremely Low-Dose Ionizing Radiation Causes Activation of Mitogen-activated Protein Kinase Pathway and Enhances Proliferation of Normal Human Diploid Cells

Keiji Suzuki, Seiji Kodama and Masami Watanabe
Keiji Suzuki
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Seiji Kodama
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Masami Watanabe
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI:  Published July 2001
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

We demonstrated here that X-ray irradiation at very low doses of between 2 and 5 cGy stimulated proliferation of normal human diploid cells and human tumor cells. Higher doses of irradiation at >1 Gy accumulated p53 protein and induced phosphorylation of extracellular signal-regulated kinase (ERK) 1/2. Phosphorylation of ERK1/2 decreased with dose down to 50 cGy, however, doses of between 5 cGy and 2 cGy phosphorylated ERK1/2 as efficiently as higher doses of X-rays, whereas the p53 protein level was not changed by doses <50 cGy. We found that mitogen-activated protein /ERK kinase (MEK) 1 was phosphorylated with both 2 cGy and 6 Gy of X-rays, and that activated ERK1/2 augmented phosphorylation of Elk-1 protein. The specific epidermal growth factor receptor tyrosine kinase inhibitor, AG1478, decreased phosphorylation of the ERK1/2 proteins induced by 2 cGy or 6 Gy of X-rays, and similar suppressive effect was observed with MEK inhibitor, PD98059. Suppression of ERK1/2 phosphorylation with these inhibitors alleviated enhanced proliferation of normal human cells by low-dose irradiation. Furthermore, overexpression of ERK2 in NCI-H1299 human lung carcinoma cells potentiated enhanced proliferation, whereas down-regulation of ERK2 using the antisense ERK2 gene abrogated the stimulative effect of low-dose irradiation. These results indicate that a limited range of low-dose ionizing radiation differentially activates ERK1/2 kinases via activation of epidermal growth factor receptor and MEK, which causes enhanced proliferation of cells receiving very low doses of ionizing radiation.

INTRODUCTION

Several reports have indicated that extremely low doses of ionizing radiation cause an unpredicted response in cells (1, 2, 3, 4) . For example, a low dose of ionizing radiation, such as 2 cGy of X-rays, alleviates the lethal and mutagenic effects of subsequently higher doses of radiation (5, 6, 7, 8, 9, 10) . Induction of gene transcriptions or proteins has been found after low-dose irradiation (2 , 11) , indicating that the induction of gene transcription through the activation of signal transduction may be involved in the low-dose effects. Because doses of >10 cGy do not cause the effects (2 , 9 , 12) , only a limited range of low doses may induce the differential stimulation of certain signal transduction pathways. To date, most signal transduction studies have used doses of ionizing radiation >1 Gy, therefore, very little is known about the effect of low-dose ionizing radiation on the activation of signal transduction pathways.

Ionizing radiation induces DNA double-strand breaks in the nucleus. In addition, it causes lipid peroxidation, ceramide generation, and protein oxidation in the membrane, cytoplasm, and nucleus (13) . Therefore, it is very likely that ionizing radiation stimulates multiple signal transduction pathways simultaneously. One such pathway originates in the nucleus and transduces signal to p53 protein. p53 protein is a tumor suppressor gene product of which the function is involved in cell cycle arrest, apoptosis, DNA repair, and senescence (14, 15, 16, 17, 18) . p53 protein has been shown to accumulate after ionizing radiation and to act as a transcription regulator (19, 20, 21, 22) . Several studies have suggested that DNA strand breaks cause accumulation and activation of p53 protein (23 , 24) . Furthermore, studies have shown that phosphorylation plays a crucial role in the regulation of the p53 accumulation and activity (25, 26, 27, 28, 29, 30) , and both ataxia telangiectasia-mutated and CHK2/Cds1 proteins, which are protein kinases, phosphorylate p53 protein at serine 15 and 20, resulting in an accumulation of p53 protein by the inhibition of the interaction between p53 and MDM2 proteins (31, 32, 33, 34, 35, 36) . Thus, DNA strand breaks caused by higher doses of radiation result in activation of the nuclear signal transduction, which is indispensable to the p53 response to ionizing irradiation. Once p53 is activated, it induces a variety of genes of which the products are involved in the induction of apoptosis, cell cycle arrest, and premature senescence. Recently, 0.5 Gy of γ-rays was reported to accumulate p53 protein in human skin diploid fibroblasts (37) . It should therefore be examined whether a very low dose of ionizing radiation can activate p53 function to induce low-dose effects.

Another signal transduction pathway stimulated by ionizing irradiation is mediated by MAPKs. 3 There are several members of MAPK including ERK1/2, JNK1/2, and p38-MAPK (p38; Refs. 38, 39, 40, 41 ). Several studies have reported that high-dose ionizing radiation activates ERK1/2 through the stimulation of tyrosine kinase associated with the membrane (42, 43, 44, 45, 46) , which results in the activation of several transcription factors regulating genes involved in cell growth. Thus, MAPK mediates the transduction of the signal from the membrane to the nucleus, and it may alleviate the lethal effects of radiation. Recently, a study reported that exposure to 1 Gy causes greater alterations in the activities of ERK1/2 than that to 6 Gy (47) , however, it is not known whether very low doses of ionizing radiation cause activation of MAPK.

Although MAPK and p53 are activated by ionizing radiation, these pathways mediate pleiotropic response in cells. For example, p53, JNK1/2, and p38 have been shown to induce cell cycle arrest or apoptosis, whereas ERK1/2 promote cell growth (48 , 49) . These results suggest that the pathways activated differently in response to different doses of ionizing radiation cause differential effects.

In the present study, we examined the effects of low doses of ionizing radiation on cell proliferation of normal human diploid cells and determined whether MAPKs and p53 are activated in a dose-dependent manner at doses between 1 cGy and 6 Gy. The results demonstrated that irradiation of cells with a limited range of low-dose radiation between 2 cGy and 5 cGy stimulated cell proliferation, whereas doses >1 Gy showed lethal effect on cells. Differential activation of ERK1/2 via activation of EGFR and MEK could explain the enhanced cell proliferation of cells receiving low-dose irradiation.

MATERIALS AND METHODS

Antibodies.

The antibodies used in this study were anti-p53 monoclonal antibody and anti p21WAF1/CIP1 monoclonal antibody (Lab Vision Corp.) and anti-phospho-ERK1/2 antibody, anti-ERK1/2 antibody, anti-phospho-JNK1/2 antibody, anti-JNK1/2 antibody, anti-phospho-p38 antibody, anti-p38 antibody, anti-phospho-MEK antibody, anti-MEK antibody, anti-phospho-Elk1 antibody, anti-Elk1 antibody, and antiphosphorylated p53 (New England Biolabs).

Cell Cultures and Reagents.

Normal human diploid (HE49) cells were cultured in Eagle’s MEM supplemented with 10% fetal bovine serum (Trace Bioscience PTY Ltd., ACN, Australia) as described previously (50) . Cells seeded in T25 flasks (25 cm (2) were subcultured every 3–4 days to maintain exponential growth. The specific MEK1 inhibitor PD98059 and EGFR inhibitor AG1478 were obtained from Calbiochem (San Diego, CA).

Construction of the Sense- and Antisense-ERK2-expressing Cells.

NCI-H1299 cells were obtained from American Type Culture Collection. H1299 cells were transfected with pVgRXR plasmid to establish 99V9 cells, which express VgECR and RXR proteins. The 99V9 cells were transfected again with pIND-ERK2 or pIND-ASERK2, which contain the human ERK2 gene in a sense or antisense orientation, respectively. The pIND/GS H-Z11695 plasmid was obtained from Invitrogen, and the pIND-ERK2 and pIND-ASERK2 plasmids were constructed by cutting the pIND/GS H-Z11695 plasmid by Pme I and religated in a sense or antisense orientation.

Irradiation and Cell Survival.

Exponentially growing cells were washed twice with PBS and exposed to a germicidal UV lamp (Model GL-10; Toshiba, Tokyo, Japan) at a dose rate of 0.45 J/m2. For X-irradiation, exponentially growing cells were irradiated with 6 Gy of X-rays from an X-ray generator at 150 kVp and 5 mm with a 0.1-mm copper filter. The dose rate for X irradiation was 0.44 Gy/min and 0.1 Gy/min. After irradiation, the cells were trypsinized, and the number of cells was determined. Cell survival was determined as the plating efficiency by inoculating an appropriate number of cells into 100-mm dishes. The cells were incubated at 37°C for 10 days, fixed with methanol, and then stained with Giemsa stain.

Western Blot Analysis.

Cells were lysed in radioimmunoprecipitation assay buffer [50 mm Tris-HCl (pH 7.2), 150 mm NaCl, 1% NP-40, 1% sodium deoxycholate, and 0.1% SDS] containing 1 mm 4-(2-Aminoethyl)-benzenesulfonyl fluoride hydrochloride as described previously (51) . The cell lysate was cleared by centrifugation at 15,000 rpm for 10 min at 4°C, and the supernatant was used as total cellular protein. The protein concentration was determined by the BCA protein assay (Pierce, Rockford, IL). Protein samples (16 μg) were electrophoresed on a SDS-polyacrylamide gel. The proteins were electrophoretically transferred to polyvinylidene difluoride membrane in transfer buffer (100 mm Tris, 192 mm glycine). After an overnight incubation with blocking solution (10% skim milk), the membrane was incubated with primary antibodies. It was then incubated with a biotinylated secondary antibody and streptavidin-alkaline phosphatase. The bands were visualized after addition of nitroblue tetrazolium/5-bromo-4-chloro-3-indolyl phosphate as substrate.

Assay for ERK1/2 Activity.

To prepare total cell extracts, cells were lysed in lysis buffer [20 mm Tris-HCl (pH 7.5), 150 mm NaCl, 1 mm EDTA, 1 mm EGTA, 1% Triton X-100, 2.5 mm sodium pyrophosphate, 1 mm β-glycerophosphate, 1 mm Na3VO4, 1 μg/ml leupeptin, 1 mm APMSF]. Total protein extracts (200 μg) were mixed with anti-phospho-ERK1/2 in 500 μl of lysis buffer overnight at 4°C. The phosphorylated ERK1/2 proteins were immunoprecipitated by adding protein A-Sepharose beads. The immune complexes were washed twice with lysis buffer and assayed for MAPK activity using a MAPK assay kit (New England Biolabs, Inc.). Briefly, the immunoprecipitates were washed once with Kinase buffer (25 mm Tris-HCl, pH 7.5, 5 mm β-Glycerophosphate, 2 mm DTT, 0.1 mm Na3VO4, 10 mm MgCl2), and were resuspended in 50 μl of kinase buffer containing 1 μl of 10 mm ATP and 2 μg of Elk1-GST fusion protein as a phosphorylation substrate. The reaction mixtures were then incubated for 30 min at 30°C with constant agitation. The reaction was terminated by adding Laemmli’s sample buffer [62.5 mm Tris-HCl (pH 6.8), 10% glycerol, 2% SDS, 5% β-mercaptoethanol]. The samples were analyzed by Western blot analysis using anti-phospho-Elk1 antibody.

RESULTS

Effect of Ionizing Radiation on Cell Survival and Cell Growth.

Lethal effects of X-rays on normal human diploid cells were examined in Fig. 1 ⇓ . Cell killing was detected with doses >1 Gy, and cell survival was decreased in a dose-dependent manner. The effects of low-dose X-rays <1 Gy on cell proliferation were also investigated. Exponentially growing cells were irradiated and counted the number of cells to determine the initial cell number. The rest of the cells were incubated for 24 h before counting cell number. As shown in Fig. 2 ⇓ , cell proliferation was significantly enhanced by low-dose irradiation at doses between 0.02 and 0.05 Gy (P < 0.01 and P < 0.05, respectively).

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Survival curve of normal human diploid cells irradiated with various doses of X-rays. Exponentially growing cells were irradiated with various doses of X-rays. Cell survival was determined as the plating efficiency by inoculating an appropriate number of cells into 100-mm dishes. Cell cultures were incubated for 10 days, fixed with methanol, and stained with Giemsa stain.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Enhanced cell proliferation by low-dose irradiation. Cells (1 × 105) were seeded into T25 culture flasks and incubated for 48 h. Number of cells in T25 flasks was counted to determine the initial cell number. Then, cells were irradiated with various doses of X-rays as indicated and incubated for additional 24 h before counting cell number. Initial number of cells (control) was determined to be 1.0, and relative number of cells after 24 h was calculated. Results are means of three independent experiments; bars, SD. Cell proliferation was significantly stimulated by 0.02 and 0.05 Gy of X-rays (∗∗< 0.01; ∗< 0.05, respectively by t test).

Activation of MAPKs in Normal Human Diploid Cells.

Normal human diploid cells used in this study express detectable levels of all three of the MAPKs (Fig. 3) ⇓ . In the control cells, we detected constitutive phosphorylation of both ERK1 and ERK2 but little or no phosphorylation of JNK1 and JNK2 or p38. To determine the activation of the MAPK families, normal human diploid cells were irradiated with 4 Gy of X-rays and incubated for 5 h (Fig. 3) ⇓ . The amount of phosphorylated ERK1/2 increased 3- to 4-fold compared with the control level; however, no phosphorylation of JNK1/2 and p38 or of c-Jun and ATF-2 was observed for up to 12 h after X-irradiation. In contrast, cells exposed to 10 J/m2 of UV showed phosphorylation of not only ERK1 and ERK2 but also JNK1/2 and p38 (Fig. 3) ⇓ . The activation of JNK1/2 and p38 was confirmed by the phosphorylation of the downstream effectors, c-Jun and ATF-2 transcription factors.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Activation of ERK1/2, JNK1/2, and p38 by X-irradiation and UV in normal human diploid cells. A, Western blot analysis of ERK1 and ERK2. Blots were probed with anti-ERK1/2 antibody (top panel) or with antibody-recognizing phosphorylated forms of ERK1/2 (bottom panel). B, Western blot analysis of JNK1 and JNK2. Blots were probed with anti-JNK1/2 antibody (top panel), with antibody-recognizing phosphorylated forms of JNK1/2 (middle panel), or with antiphosphorylated c-Jun (bottom panel). C, Western blot analysis of p38. Blots were probed with anti-p38 antibody (top panel), with antibody-recognizing phosphorylated forms of p38 (middle panel), or with antiphosphorylated ATF-2 (bottom panel). C, control; X, 5 h after 4 Gy X-rays; U, 1 h after 10 J/m2 of UV.

Dose-dependent Activation of ERK1/2 and p53.

Dose-dependent activation of ERK1/2 and p53 protein is shown in Fig. 4 ⇓ . We found the dose-dependent increase in the phosphorylation of ERK1/2 at doses >2 Gy. The levels decreased with dose down to 1 Gy but increased again at doses between 0.02 Gy and 0.1 Gy. The increases were comparable with those observed at ≥4 Gy. In contrast, p53 accumulation was not detected at doses <0.5 Gy, and between 1 and 6 Gy the level of p53 protein increased in a dose-dependent manner. Phosphorylation of p53 protein at serine 15 was detected at 0.1 Gy, and its level gradually increased as the X-ray dose increased. There was a slight increase of p21WAF1/CIP1 at 0.1 Gy and a dose-dependent increase was observed at ≥1.0 Gy.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Dose-dependent phosphorylation and activation of ERK1/2 and p53. A, exponentially growing cells were irradiated with various doses of X-rays as indicated. To compare the maximum level of each protein, they were incubated for 2 h (for p53 and phosphorylated p53), 4 h (for p21WAF1/CIP1), or 5 h (for ERK1/2 and phosphorylated ERK1/2) before extraction of the proteins, respectively. B, band intensity of the blots was measured densitometrically, and relative amount of each protein was calculated. •, phosphorylated ERK1/2; ⋄, p53; ▵, p21WAF1/CIP1.

Phosphorylation of MEK1/2 and Elk-1.

Because both 2 cGy and 6 Gy stimulated ERK1/2 phosphorylation, we determined if the upstream MEK1/2 and the downstream effector, Elk-1, were activated. Using antibody against phosphorylated MEK1/2 protein, we found that both 2 cGy and 6 Gy of X-rays enhanced phosphorylation of MEK1/2, although phosphorylation was more significant in cells irradiated with 6 Gy (Fig. 5B) ⇓ . IP-kinase assay revealed that the level of Elk-1 protein was significantly increased by 2.5-fold and 4-fold by 2 cGy and 6 Gy irradiation, respectively (Fig. 5, C and D) ⇓ .

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Phosphorylation of MEK1/2 and Elk-1 after X-irradiation with 2 cGy and 6 Gy. A, phosphorylation of ERK1/2 was determined 5 h after X-irradiation at 2 cGy or 6 Gy. B, phosphorylation of MEK1/2 was determined 5 h after X-irradiation at 2 cGy or 6 Gy. Blot was probed with anti-MEK1/2 antibody (top panel) or with anti phosphorylated MEK1/2 (bottom panel). C, phosphorylation of Elk-1 was determined 5 h after X-irradiation at 2 cGy or 6 Gy. Phosphorylated ERK1/2 was immunoprecipitated from the cell lysate, and Elk-1 phosphorylation was assayed in vitro using Elk-1-GST fusion protein as described in “Materials and Methods.” Phosphorylation of ELK-1 was determined by Western blot analysis. Blot was probed with anti-Elk-1 antibody (top panel) or with antiphosphorylated Elk-1 (bottom panel). Elk-1, purified Elk-1-GST fusion protein. D, blot indicated in Fig. 9C was scanned densitometrically, and relative amount of phosphorylated Elk-1 was calculated.

Effects of Inhibitors on ERK1/2 Phosphorylation and Enhanced Cell Proliferation.

The role of MEK1/2 in ERK1/2 activation was examined using a specific inhibitor for MEK, PD98059 (Fig. 6) ⇓ . We found that the administration of 10 μm of PD98059 inhibited the phosphorylation of ERK1/2 in cells irradiated with 6 Gy of X-rays, but its suppressive effect was less effective in cells exposed to 2 cGy of X-rays. The effects of the inhibitor for EGFR tyrosine kinase, AG1478, on ERK1/2 phosphorylation were also examined. Treatment of the cells with 0.2 or 0.4 μm of AG1478 suppressed the phosphorylation of ERK1/2 similarly as in cells irradiated with 2 cGy and 6 Gy. The suppressive effect of the inhibitors on enhanced cell proliferation by 0.02 Gy X-rays is shown in Fig. 7 ⇓ . Both 10 μm of PD98059 and 0.4 μm of AG1478 diminished stimulation of cell proliferation.

Fig. 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 6.

Suppressive effects of the inhibitors PD98059 and AG1478 on ERK1/2 activation by X-irradiation. A, cells were pretreated with or without various doses of PD98059 or AG1478 for 1 h and then irradiated with 2 cGy or 6 Gy of X-rays. Phosphorylation of ERK1/2 was determined by Western blot analysis. B, blot was scanned densitometrically, and relative amount of phosphorylated ERK1/2 was calculated.

Fig. 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 7.

Suppressive effects of the inhibitors PD98059 and AG1478 on enhanced proliferation by 2 cGy of X-rays. Exponentially growing cells were pretreated with 10 μm of PD98059 and 0.4 μm of AG1478 for 1 h and then irradiated with 2 cGy of X-rays. Then cells were incubated for an additional 24 h before counting cell number. ∗∗< 0.01 by t test.

Effects of the Sense- and Antisense-ERK2 Gene Expression on Enhanced Cell Proliferation.

The sense- or antisense-ERK2 gene is induced in human non-small cell lung carcinoma (NCI-H1299) cells by PA. Although PA alone showed no effect on phosphorylation of ERK1/2 and enhanced proliferation of cells by low-dose irradiation, increased expression of ERK2 significantly potentiated the enhanced proliferation of 0.02 Gy-irradiated cells (Fig. 8) ⇓ . Furthermore, the induction of the antisense ERK2 gene decreased the level of ERK2 phosphorylation, and it completely abolished stimulation of cell proliferation by low-dose irradiation.

Fig. 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 8.

Effects of the sense- and antisense-ERK2 gene expression on enhanced cell proliferation of human tumor cells (H1299) by 2 cGy of X-rays. Sense- (E2S) or antisense-ERK2 (E2AS) genes are induced in human non-small cell lung carcinoma (NCI-H1299) cells by PA for 24 h before irradiation. A, phosphorylation of ERK1 and 2 was determined 5 h after X-irradiation at 2 cGy. B, exponentially growing cells were incubated for 24 h with or without PA, irradiated with 2 cGy of X-rays, and then incubated for an additional 24 h before counting cell number. Initial number of cells was given as control. Increased cell proliferation was significantly enhanced by induction of the ERK2 gene compared with the control cells (∗∗∗; P < 0.001 by t test) and completely abolished by induction of the antisense ERK2 gene.

DISCUSSION

In the present study, we demonstrate that low-dose ionizing radiation stimulates proliferation of normal human diploid cells. Because the effect was observed in cells irradiated with very low doses of X-rays, a limited range of low doses was suggested to activate certain signal transduction pathways as described in the rodent cells (52) . We used antibodies specifically recognizing the activated form of MAPKs, in which specific threonine and tyrosine are doubly phosphorylated. Although detectable levels of all three of the MAPKs were observed in normal human diploid cells, X-ray irradiation stimulated phosphorylation of ERK1/2, but it did not induce phosphorylation of JNK1/2 and p38. In contrast, when cells were exposed to 10 J/m2 of UV light, all of these MAPKs were activated and downstream effectors c-Jun and ATF-2 proteins were phosphorylated, indicating that the three pathways are intact in these cells. The results contrast previous studies showing that ionizing radiation causes activation of not only ERK1/2 but also JNK1/2 and p38 (43 , 46 , 47) . However, these studies used tumor-derived cells, and therefore, our study emphasizes that the type of MAPKs activated by ionizing radiation differs depending on the cell type and that ionizing radiation is a strong stimulator for ERK1/2 in normal human diploid cells.

We found that X-ray irradiation at low doses, between 2 cGy and 5 cGy, activated ERK1/2 as efficiently as doses of ≥4 Gy (Fig. 4) ⇓ . The time course experiments showed that phosphorylation of ERK1/2 continued up to 10 h after irradiation (data not shown). Although previous reports have proven that high-dose radiation activates EGFR followed by the stimulation of Ras, Raf, and MEK1/2 (42, 43, 44, 45, 46) , the present results implicated that the same mechanism may be involved in ERK1/2 activation by low-dose radiation. As shown in Fig. 6 ⇓ , both 10 μm of PD98059 and 0.2 and 0.4 μm of AG1478 inhibited the activation of ERK1/2 by 6 Gy of X-rays. Similar suppressive effects of PD 98059 and AG1478 were observed in cells irradiated with 2 cGy, although there was less effect on PD98059 treatment. These results indicated that both 6 Gy and 2 cGy of X-rays transduce the signal to ERK1/2 via activation of EGFR and MEK1/2.

Activated ERK1/2 phosphorylated Elk-1, which involves in the induction of the growth-related genes, suggesting that stimulative effects of low-dose irradiation were mediated by ERK1/2 activation. Involvement of ERK1/2 activation in growth stimulation by low dose was confirmed by the results presented in Figs. 7 ⇓ and 8 ⇓ . First, two kinds of inhibitors, PD98059 and AG1478, were used, and they both suppressed phosphorylation of ERK1/2 and increased cell proliferation. Second, the induced expression of the ERK2 gene potentiated a stimulative effect, whereas the forced expression of the antisense ERK2 gene abrogated enhanced cell growth by low-dose irradiation, completely. Recent studies have reported that ERK1/2 phosphorylate not only transcription factors but also other protein kinases and upstream mediators, such as p90RSK and EGFR and SOS (53) . Thus, our results indicate that activation of ERK1/2 by low-dose radiation result in increased proliferation of cells through phosphorylation and activation of such growth-related factors.

In contrast to ERK1/2, accumulation of p53 did not occur with X-ray doses <50 cGy. The evidence obtained thus far indicates that p53 accumulation is dependent on DNA strand breaks (23 , 24) . An X-ray dose of 10 cGy is estimated to cause approximately five initial double strand breaks per nuclei, and the initial double strand breaks are predicted to be rejoined within ∼10 min of the irradiation (54) . Single strand breaks are repaired much faster than double strand breaks (54) . As shown in Fig. 1 ⇓ , X-rays doses <10 cGy will not kill very many cells, suggesting that most of the damage is repairable. Although it is difficult to estimate the exact number of DNA strand breaks remaining, cells irradiated with ≤10 cGy may not have unrepaired DNA breaks, which are lethal to cells and lead to the accumulation of p53 protein. As shown in Fig. 4 ⇓ , ERK1/2 are activated at these doses. Although previous studies have indicated that MAPKs accumulate and activate p53 protein (55) , our results suggest that ERK1/2 alone are not sufficient to accumulate and activate p53 protein in normal human cells. Accumulated p53 is activated as a transcription factor and stimulates transcription of a group of genes including the p21Waf1/Cip1 gene. The present results confirmed that accumulated p53 induced p21WAF1/CIP1, but it was observed with doses >1 Gy. It can be concluded that very low doses of ionizing radiation stimulate only ERK1/2, and enhance cell proliferation, whereas higher doses activate not only ERK1/2 but also p53, which antagonizes the proliferative effect of ERK1/2 activation and results in cell cycle arrest.

Several studies have shown that ERK1/2 activation leads to an induction of the genes involved in cell proliferation, suggesting that the activation of ERK1/2 has cytoprotective effects. Our previous study found that oncogenic RAF and Ras alleviated the lethal effects of irradiation (56) . A recent study also showed that inhibition of MAPK potentiated the cell killing by ionizing radiation (47) . Although the mechanism of the cytoprotective effect of ERK1/2 is unknown, a recent study has shown that ERK1/2 phosphorylate Histone H3, of which phosphorylation is hypothesized to be involved in the transcriptional activation of immediate-early genes through chromatin remodeling (57, 58, 59, 60) . Therefore, it is very likely that activation of ERK1/2 by a limited range of low doses of X-rays before subsequent higher-dose irradiation may induce gene expression related to DNA damage repair or cell survival or facilitate DNA repair by remodeling the chromatin structure. Thus, the present results provide the possibility that the activation of ERK1/2 may be one mechanism of low-dose effects in normal human diploid cells.

Acknowledgments

We thank Dr. William F. Morgan for valuable discussions and critical review of the manuscript.

Footnotes

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • ↵1 Supported by a Grant for Scientific Research from the Ministry of Education, Science, Sports and Culture, Japan, and by a Grant for the Regional Links Research Program at Nagasaki from the Research Development Corporation of Japan.

  • ↵2 To whom requests for reprints should be addressed, at Laboratory of Radiation and Life Science, School of Pharmaceutical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan. Phone/Fax: 81-958-44-5504; E-mail: kzsuzuki{at}net.nagasaki-u.ac.jp

  • ↵3 The abbreviations used are: MAPK, mitogen-activated protein kinase; ATF, activating transcription factor; EGFR, epidermal growth factor receptor; ERK, extracellular signal-regulated kinase; PA, ponasterone A; JNK, c-Jun N-terminal kinase.

  • Received January 29, 2001.
  • Accepted May 16, 2001.
  • ©2001 American Association for Cancer Research.

References

  1. ↵
    Stecca C., Gerber G. B. Adaptive response to DNA-damaging agents. Biochem. Pharmacol., 55: 941-951, 1998.
    OpenUrlCrossRefPubMed
  2. ↵
    Wolff S. The adaptive response in radiobiology: evolving insights and implications. Environ. Health Perspect., 106: 277-283, 1998.
  3. ↵
    Feinendegen L. E. The role of adaptive responses following exposure to ionizing radiation. Hum. Exp. Toxicol., 18: 426-432, 1999.
    OpenUrlFREE Full Text
  4. ↵
    Skov K. A. Radioresponsiveness at low doses: hyper-radiosensitivity and increased radioresistance in mammalian cells. Mutat. Res., 430: 241-253, 1999.
    OpenUrlPubMed
  5. ↵
    Olivieri G., Bodycote J., Wolff S. Adaptive response of human lymphocytes to low concentrations of radioactive thymidine. Science (Wash. DC), 233: 594-597, 1984.
    OpenUrl
  6. ↵
    Ikushima T. Chromosomal responses to ionizing radiation reminiscent of an adaptive response in cultured Chinese hamster cells. Mutat. Res., 180: 215-221, 1987.
    OpenUrlCrossRefPubMed
  7. ↵
    Kelsey K. T., Memisoglu A., Frenkel D., Liber H. L. Human lymphocytes exposed to low doses of X-rays are less susceptible to radiation-induced mutagenesis. Mutat. Res., 263: 197-201, 1991.
    OpenUrlCrossRefPubMed
  8. ↵
    Sasaki M. S. On the reaction kinetics of the radioadaptive response in cultured mouse cells. Int. J. Radiat. Biol., 68: 281-291, 1995.
    OpenUrlCrossRefPubMed
  9. ↵
    Rigaud O., Moustacchi E. Radioadaptation for gene mutation and the possible molecular mechanisms of the adaptive response. Mutat. Res., 358: 127-134, 1996.
    OpenUrlCrossRefPubMed
  10. ↵
    Suzuki K., Kodama S., Watanabe M. Suppressive effect of low-dose preirradiation on genetic instability induced by X rays in normal human embryonic cells. Radiat. Res., 150: 656-662, 1998.
    OpenUrlPubMed
  11. ↵
    Robson T., Price M. E., Moore M. L., Joiner M. C., McKelvey-Martin V. J., McKeown S. R., Hirst D. G. Increased repair and cell survival in cells treated with DIR1 antisense oligonucleotides: implications for induced radioresistance. Int. J. Radiat. Biol., 76: 617-623, 2000.
    OpenUrlCrossRefPubMed
  12. ↵
    Suzuki K., Kodama S., Watanabe M. Effect of low-dose preirradiation on induction of the HSP70B-LacZ fusion gene in human cells treated with heat shock. Radiat. Res., 149: 195-201, 1998.
    OpenUrlCrossRefPubMed
  13. ↵
    Halliwell B., Gutteridge J. M. C. Oxygen free radicals and iron in relation to biology and medicine: some problems and concepts. Arch. Biochem. Biophys., 246: 501-514, 1986.
    OpenUrlCrossRefPubMed
  14. ↵
    Harris C. C. p53 tumor suppressor gene: from the basic research laboratory to the clinic-an abridged historical perspective. Carcinogenesis (Lond.), 17: 1187-1198, 1996.
    OpenUrlFREE Full Text
  15. ↵
    Prives C. Signaling to p53: breaking the MDM2–p53 circuit. Cell, 95: 5-8, 1998.
    OpenUrlCrossRefPubMed
  16. ↵
    Oren M. Regulation of the p53 tumor suppressor protein. J. Biol. Chem., 274: 36031-36034, 1999.
    OpenUrlFREE Full Text
  17. ↵
    Vogelstein B., Lane D., Levine A. J. Surfing the p53 network. Nature (Lond.), 408: 307-310, 2000.
    OpenUrlCrossRefPubMed
  18. ↵
    Vousden K. H. p53: death star. Cell, 103: 691-694, 2000.
    OpenUrlCrossRefPubMed
  19. ↵
    Kastan M. B., Onyekwere O., Sidrabsky D., Vogelstein B., Craig R. W. Participation of p53 protein in the cellular response to DNA damage. Cancer Res., 51: 6304-6311, 1991.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Amundson S. A., Myers T. G., Fornace A. J., Jr. Roles for p53 in growth arrest and apoptosis: putting on the brakes after genotoxic stress. Oncogene, 17: 3287-3299, 1998.
    OpenUrlCrossRefPubMed
  21. ↵
    Ashcroft M., Vousden K. H. Regulation of p53 stability. Oncogene, 18: 7637-7655, 1999.
    OpenUrlCrossRefPubMed
  22. ↵
    Lavin M. F., Khanna K. K. ATM: the protein encoded by the gene mutated in the radiosensitive syndrome ataxia-telangiectasia. Int. J. Radiat. Biol., 75: 1201-1214, 1999.
    OpenUrlCrossRefPubMed
  23. ↵
    Nelson W. G., Kastan M. B. DNA strand breaks: the DNA template alterations that trigger p53-dependent DNA damage response pathways. Mol. Cell. Biol., 14: 1815-1823, 1994.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Huang L-C., Clarkin K. C., Wahl G. M. Sensitivity and selectivity of the DNA damage sensor responsible for activating p53-dependent G1 arrest. Proc. Natl. Acad. Sci. USA, 93: 4827-4832, 1996.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Kubbutat M. H. G., Vousden K. H. Keeping an old friend under control: regulation of p53 stability. Mol. Med. Today, 4: 250-256, 1998.
    OpenUrlCrossRefPubMed
  26. ↵
    Nakamura Y. ATM: the p53 booster. Nat. Med., 4: 1231-1232, 1998.
    OpenUrlCrossRefPubMed
  27. ↵
    Agarwal M. L., Taylor W. R., Chernov M. V., Chernova O. B., Stark G. R. The p53 network. J. Biol. Chem., 273: 1-4, 1998.
    OpenUrlFREE Full Text
  28. ↵
    Jimenez G. S., Khan S. H., Stommel J. M., Wahl G. M. p53 regulation by post-translational modification and nuclear retention in response to diverse stresses. Oncogene, 18: 7656-7665, 1999.
    OpenUrlCrossRefPubMed
  29. ↵
    Lakin N. D., Jackson S. P. Regulation of p53 in response to DNA damage. Oncogene, 18: 7644-7655, 1999.
    OpenUrlCrossRefPubMed
  30. ↵
    Hupp T. R., Lane D. P., Ball K. L. Strategies for manipulating the p53 pathway in the treatment of human cancer. Biochem. J., 352: 1-17, 2000.
  31. ↵
    Shieh S-Y., Ikeda M., Taya Y., Prives C. DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell, 91: 325-334, 1997.
    OpenUrlCrossRefPubMed
  32. ↵
    Siliciano J. D., Canman C. E., Taya Y., Sakaguchi K., Appella E., Kastan M. B. DNA damage induces phosphorylation of the amino terminus of p53. Genes Dev., 11: 3471-3481, 1997.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Banin S., Moyal L., Shieh S-Y., Taya Y., Anderson C. W., Chessa L., Smorodinsky N. I., Prives C., Reiss Y., Shiloh Y., Ziv Y. Enhanced phosphorylation of p53 by ATM in response to DNA damage. Science (Wash. DC), 281: 1674-1677, 1998.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Shieh S.-Y., Taya Y., Prives C. DNA damage-inducible phosphorylation of p53 at N-terminal sites including a novel site, Ser20, requires tetramerization. EMBO J., 18: 1815-1823, 1999.
    OpenUrlAbstract
  35. ↵
    Chehab N. H., Malikzay A., Appel M., Halazonetis T. D. Chk2/hCds1 functions as a DNA damage checkpoint in G1 by stabilizing p53. Genes Dev., 14: 278-288, 2000.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Hirao A., Kong Y-Y., Matsuoka S., Wakeham A., Ruland J., Yoshida H., Liu D., Elledge S. J., Mak T. W. DNA damage-induced activation of p53 by the checkpoint kinase chk2. Science (Wash. DC), 287: 1824-1827, 2000.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Azzam E. I., de Toledo S. M., Phkett M. J., Nagasawa H., Little J. B. CDC2 is down-regulated by ionizing radiation in a p53-dependent manner. Cell Growth Differ., 8: 1161-1169, 1997.
    OpenUrlAbstract
  38. ↵
    Canman C. E., Kastan M. B. Signal transduction. Three paths to stress relief. Nature (Lond.), 384: 213-214, 1996.
    OpenUrlCrossRefPubMed
  39. ↵
    Kyriakis J. M., Avruch J. Sounding the alarm: protein kinase cascades activated by stress and inflammation. J. Biol. Chem., 271: 24313-24316, 1996.
    OpenUrlFREE Full Text
  40. ↵
    Widmann C., Gibson S., Jarpa M. B., Johnson G. L. Mitogen-activated protein kinase: conservation of a three-kinase module from yeast to human. Physiol. Rev., 79: 143-180, 1999.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Roovers K., Assoian R. K. Integrating the MAP kinase signal into the G1 phase cell cycle machinery. Bioessays, 22: 818-826, 2000.
    OpenUrlCrossRefPubMed
  42. ↵
    Kasid U., Suy S., Dent P., Ray S., Whiteside T. L., Strugill T. W. Activation of Raf by ionizing radiation. Nature (Lond.), 382: 813-816, 1996.
    OpenUrlCrossRefPubMed
  43. ↵
    Verheij M., Bose R., Lin X. H., Yao B., Jarvis W. D., Grant S., Birrer M. J., Szabo E., Zon L. I., Kyriakis J. M., Haimovitz-Friedman A., Fuks Z., Kolesnick R. N. Requirement for ceramide-initiated SAPK/JNK signalling in stress-induced apoptosis. Nature (Lond.), 380: 75-79, 1996.
    OpenUrlCrossRefPubMed
  44. ↵
    Schmidt-Ullrich R. K., Mikkelsen R. B., Dent P., Todd D. G., Valerie K., Kavanagh B. D., Contessa J. N., Rorrer W. K., Chen P. B. Radiation-induced proliferation of the human A431 squamous carcinoma cells is dependent on EGFR tyrosine phosphorylation. Oncogene, 15: 1191-1197, 1997.
    OpenUrlCrossRefPubMed
  45. ↵
    Suy S., Anderson W. B., Dent P., Chang E., Kasid U. Association of Grb2 with Sos and Ras with Raf-1 upon γ irradiation of breast cancer cells. Oncogene, 15: 53-61, 1997.
    OpenUrlCrossRefPubMed
  46. ↵
    Verheij M., Ruiter G. A., Zerp S. F., van Blitterswijk W. J., Fuks Z., Haimovitz-Friedman A., Bartelink H. The role of the stress-activated protein kinase (SAPK/JNK) signaling pathway in radiation-induced apoptosis. Radiother. Oncol., 47: 225-232, 1998.
    OpenUrlCrossRefPubMed
  47. ↵
    Carter S., Auer K. L., Readon D. B., Birrer M., Fisher P. B., Valerie K., Schmidt-Ullrich R., Mikkelsen R., Dent P. Inhibition of the mitogen activated protein (MAP) kinase cascade potentiates cell killing by low dose ionizing radiation in A431 human squamous carcinoma cells. Oncogene, 16: 2787-2796, 1998.
    OpenUrlCrossRefPubMed
  48. ↵
    Ichijo H. From receptors to stress-activated MAP kinases. Oncogene, 18: 6087-6093, 1999.
    OpenUrlCrossRefPubMed
  49. ↵
    Sionov R. V., Haupt Y. The cellular response to p53: the decision between life and death. Oncogene, 18: 6145-6157, 1999.
    OpenUrlCrossRefPubMed
  50. ↵
    Watanabe M., Suzuki M., Suzuki K., Nakano K., Watanabe K. Effect of multiple irradiation with low doses of γ-rays on morphological transformation and growth ability of human embryo cells in vitro. Int. J. Radiat. Biol., 62: 711-718, 1992.
    OpenUrlCrossRefPubMed
  51. ↵
    Suzuki K., Kodama S., Watanabe M. Recruitment of ATM protein to double strand DNA irradiated with ionizing radiation. J. Biol. Chem., 274: 25571-25575, 1999.
    OpenUrlAbstract/FREE Full Text
  52. ↵
    Shimizu T., Kato T., Jr., Tachibana A., Sasaki M. S. Coordinated regulation of radioadaptive response by protein kinase C and p38 mitogen-activated protein kinase. Exp. Cell Res., 251: 424-432, 1999.
    OpenUrlCrossRefPubMed
  53. ↵
    Garrington T. P., Johnson G. L. Organization and regulation of mitogen-activated protein kinase signaling pathways. Curr. Opin. Cell Biol., 11: 211-218, 1999.
    OpenUrlCrossRefPubMed
  54. ↵
    Olive P. L. The role of DNA single- and double-strand breaks in cell killing by ionizing radiation. Radiat. Res., 150: S42-S51, 1998.
    OpenUrlCrossRefPubMed
  55. ↵
    Milne D. M., Campbell D. G., Caudwell F. B., Meek D. W. Phosphorylation of the tumor suppressor protein p53 by mitogen-activated protein kinases. J. Biol. Chem., 269: 9253-9260, 1995.
    OpenUrlAbstract/FREE Full Text
  56. ↵
    Suzuki K., Watanabe M., Miyoshi J. Differences in effects of oncogenes on resistance of γ rays, ultraviolet light, and heat shock. Radiat. Res., 129: 157-162, 1992.
    OpenUrlCrossRefPubMed
  57. ↵
    De Cesare D., Jacquot S., Hanauer A., Sassone-Corsi P. Rsk-2 activity is necessary for epidermal growth factor-induced phosphorylation of CREB protein and transcription of c-fos gene. Proc. Natl. Acad. Sci. USA, 95: 12202-12207, 1998.
    OpenUrlAbstract/FREE Full Text
  58. ↵
    Sassone-Corsi P., Mizzen C. A., Cheung P., Crosio C., Monaco L., Jacquot S., Hnauer A., Allis C. D. Requirement of Rsk-2 for epidermal growth factor-activated phosphorylation of histone H3. Science (Wash. DC), 285: 886-891, 1999.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    Cheung P., Tanner K. G., Cheung W. L., Sassone-Corsi P., Denu J. M., Allis C. D. Synergistic coupling of histone H3 phosphorylation and acetylation in response to epidermal growth factor stimulation. Mol. Cell, 5: 905-915, 2000.
    OpenUrlCrossRefPubMed
  60. ↵
    Clayton A. L., Rose S., Barrett M. J., Mahadevan L. C. Phosphoacetylation of histone H3 on c-fos- and c-jun-associated nucleosomes upon gene activation. EMBO J., 19: 3714-3726, 2000.
    OpenUrlAbstract
View Abstract
PreviousNext
Back to top
Cancer Research: 61 (14)
July 2001
Volume 61, Issue 14
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Extremely Low-Dose Ionizing Radiation Causes Activation of Mitogen-activated Protein Kinase Pathway and Enhances Proliferation of Normal Human Diploid Cells
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Extremely Low-Dose Ionizing Radiation Causes Activation of Mitogen-activated Protein Kinase Pathway and Enhances Proliferation of Normal Human Diploid Cells
Keiji Suzuki, Seiji Kodama and Masami Watanabe
Cancer Res July 15 2001 (61) (14) 5396-5401;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Extremely Low-Dose Ionizing Radiation Causes Activation of Mitogen-activated Protein Kinase Pathway and Enhances Proliferation of Normal Human Diploid Cells
Keiji Suzuki, Seiji Kodama and Masami Watanabe
Cancer Res July 15 2001 (61) (14) 5396-5401;
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • INTRODUCTION
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Abstract LB-089: Defining windows of susceptibility for low-dose exposure to endocrine disruptors in rat mammary development by microRNA profiling
  • Abstract LB-086: Genes targeted by drugs and curcumin in a breast carcinogenesis model
  • Abstract LB-091: Characterization of molecular changes occurring during long-term treatment of human bronchial epithelial cells with cigarette smoke total particulate matter
Show more Carcinogenesis
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement