Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Dec 23.
Published in final edited form as: J Mammary Gland Biol Neoplasia. 2010 Dec 23;15(4):381–387. doi: 10.1007/s10911-010-9197-6

Stromal Mediation of Radiation Carcinogenesis

Mary Helen Barcellos-Hoff 1,
PMCID: PMC3068291  NIHMSID: NIHMS265729  PMID: 21181431

Abstract

Ionizing radiation is a well-established carcinogen in human breast and rodent mammary gland. This review addresses evidence that radiation elicits the critical stromal context for cancer, affecting not only frequency but the type of cancer. Recent data from the breast tumors of women treated with radiation therapy and the cellular mechanisms evident in experimental models suggest that radiation effects on stromal-epithelial interactions and tissue composition are a major determinant of cancer development.

Keywords: Ionizing radiation, Carcinogenesis, Stromal-epithelial interactions, Mammary gland

Introduction

Many investigators have argued that disruption of the cell interactions and tissue architecture can be a primary driver of carcinogenesis [15]. Recent experiments published from Weinberg [6], Moses [7], Sonnenchein [8] and Coussens [9] offer provocative evidence that composition of the microenvironment is a critical determinant of cancer suppression or promotion and highlight multicellular contributions to the carcinogenic response and cancer progression. Over a decade ago, we hypothesized that modulation of the tissue microenvironment is an additional action of radiation specifically, and of carcinogens in general [10]. The prevailing paradigm of cancer risk following radiation focuses on the probability of DNA damage that can lead to mutations in susceptible cells [11]. An alternative hypothesis is that cancer emerges from irradiated tissues as a result of complex, but ultimately predictable, interactions between mutagenesis from DNA damage and radiation effects on the microenvironment and cell interactions is [12]. Just as DNA damage elicits a dramatic transition in signaling within a cell, each irradiated tissue has its own set of signals and composition, distinct from those of unirradiated tissue and different from other irradiated tissues. Biological responses to radiation damage quickly evolve and amplify, mostly in a non-linear manner, and can alter daughter cell fates such as differentiation and senescence [1316], induce long-range signals that affect non-irradiated cells [1720], or generate a state of chronic genomic instability [1721]. The sum of these events, occurring in different organs and highly modulated by genotype, predicates the health risks. To test this hypothesis we created radiation chimeric tissue by transplanting unirradiated, preneoplastic mammary cells to an irradiated mammary gland [10]. The data from this model and others discussed below supports the hypothesis that non-mutagenic effects of radiation on the stroma can contribute significantly to radiation carcinogenesis in vivo.

Radiation Exposure and Cancer in Humans

Ionizing radiation is a known carcinogen of humans and experimental models (reviewed in [22]). Excess cancers were observed in the Japanese atomic-bomb survivors at acute radiation doses of 10 to 400 cGy, which are 40 to 1600 times the average yearly background levels in the United States. The excess risks vary significantly with gender, attained age, and age at exposure for all solid cancers as a group and many individual sites as a consequence of radiation from the atomic bomb [23]. It has been estimated that if radiation exposure occurs at age 30, the solid cancer rates at age 70 is increased by about 35% per Gy for men and 58% per Gy for women [23].

A pooled analysis by Preston and colleagues of eight populations exposed to ionizing radiation showed that increased risk of breast cancer is inversely correlated with age at irradiation, with the greatest risk conferred by exposure before the age of twenty [24]. In most irradiated populations, which include atomic bomb survivors, girls treated for scoliosis or tuberculosis, and women treated for mastitis, the type of breast cancer was not determined. A recent study from Milan of the molecular and marker analysis of the breast cancers of women exposed to therapeutic radiation for childhood/young adult cancers revealed a high risk of breast cancer diagnosed at an early age (39 compared to 57 in a sporadic consecutive series) and a higher frequency of ER-negative tumors [25]. Moreover half (53%) of the breast carcinomas from irradiated women showed features of basal-like tumors compared to 11% in a consecutive series of breast cancers not preceded by radiation. When compared to age-matched controls, basal-like cancer was significantly more frequent in irradiated women. The breast cancer subtype in young women treated with radiation was much more likely to be ER- and PR-negative and p53 and cytokeratin 5/6 positive and less likely to be HER2+ compared to girls who received radiotherapy [25]. Interestingly a disproportionate frequency of contralateral ER-negative breast cancer has not been noted in older women treated with radiation for breast cancer, suggesting a physiological basis for the shift identified in the Milan study.

Approximately 90% of the breast cancers in women who received irradiation for Hodgkin’s lymphoma can be attributed to their radiation treatment [26]. This population provides a unique opportunity to determine whether radiation alters the course, as well as the frequency, of cancer. Broeks and colleagues used gene expression profiling to assess radiation-associated breast tumors (n=22) compared with a set of control breast tumors (n=20) of women unexposed to radiation, diagnosed at the same age [27]. Unsupervised hierarchical clustering of the profile data resulted in a clustering of the radiation-associated tumors separate from the control tumors. Consistent with the Milan study, tumors from irradiated women were often of the intrinsic basal breast tumor subtype. They also found a chromosomal instability profile and a higher expression of the proliferation marker Ki-67 suggesting a more aggressive tumor type. Since radiation induced DNA damage is random, the biological basis for the prevalence of this tumor type in irradiated women cannot be readily explained on the basis of the mutagenesis per se.

Radiation as a Carcinogen in Experimental Studies

Radiation is considered to be a complete carcinogen. DNA damage produces cells with oncogenic mutations and multicellular responses to DNA damage create a permissive environment for progression [12, 28]. Either microenvironments that promote the malignant phenotype are in a way functionally equivalent to the acquisition of additional mutations in the initiated cell or, the irradiated microenvironment creates novel selective pressures accelerate tumor development, or both. While DNA damage, the DNA damage response, and mutagenic consequences have been intensively characterized and are thought to be the root of radiation’s action as a carcinogen, there are provocative studies that microenvironments induced by the response to radiation can promote neoplastic progression in unirradiated epithelial cells. These events “outside of the box” may significantly increase cancer risk.

A study by Kaplan and colleagues dating back more than 50 years demonstrates that radiation carcinogenesis is complex. These studies used C57BL mice, which are very susceptible to thymic lymphomas after radiation exposure. Young mice underwent thymectomy, and 2–7 d later received the first of four consecutive doses of 168 cGy. Several hours after the last irradiation, a single thymus from a non-irradiated mouse was transplanted subcutaneously under the right chest or upper abdomen of each of the previously thymectomized, irradiated hosts. Surprisingly, thymic lymphoma incidence and latency arising from the grafts matched that observed in irradiated, intact mice. Furthermore, the tumors were histologically identical to those found in the intact mice, and exhibited a similar pattern of metastasis [29]. This study showed that radiation-induced thymic lymphomas can occur even when the grafted thymus was never exposed to radiation, suggesting a systemic effect of radiation in the host.

This systemic mechanism of tumor induction was elucidated in their second study, which showed that shielding a thigh of the host during irradiation or promptly injecting fresh bone marrow into the host shortly after the last irradiation could neutralize the tumor-inducing effect of radiation [30]. The next study showed that prior radiation exposure impaired regeneration, which was mediated by bone marrow derived cells as thigh-shielded mice exhibited an identical degree of graft regeneration as observed in unirradiated mice [31]. The final publication provided conclusive evidence that the tumors that arose in the unirradiated thymic grafts were indeed composed of donor cells and not invading host cells that had received radiation by using FI hybrid mice [29]. The susceptible C57BL strain of mice was crossed with the C3H strain, which is resistant to radiation-induced lymphomas, to generate an F1 hybrid. These studies showed that the genetic background of the graft donor determined tumor incidence and thus, susceptibility was a property of the thymus, even though the mechanism of induction occurred through the host. This series of papers highlight the host as an effective target of radiation in the induction of thymic lymphomas in grafts that were never irradiated.

In a similar series of studies, Billingham and colleagues used the carcinogen methylcholanthrene to determine which compartment was the site of carcinogenic action in mouse skin. Skin grafts of various thicknesses (including or excluding hair follicles) from carcinogen-treated sites were transplanted to untreated sites in the same animal. Such an approach revealed that the underlying dermis layer conferred equivalent tumorigenic potential, even if the overlying epidermis was untreated. Tumors occurred when untreated grafts were transplanted into treated dermis, but not when treated grafts were placed into untreated dermis [32].

Morgan and colleagues showed that an immortal myogenic cell line formed tumors far more rapidly in irradiated compared to non-irradiated host muscle. The accelerated tumor phenotype was a direct effect of irradiation on the stroma, rather than due to systemic effects, because tumors did not form in distant muscle sites [33]. Interestingly, when transplanted to normal mice, these tumors formed large amounts of muscle. Likewise, irradiated pancreatic fibroblasts mixed with pancreatic carcinoma cells formed more aggressive and invasive cancer than when the pancreatic cancer cells were mixed with nonirradiated pancreatic fibroblasts [34]. These authors further demonstrated that an antagonist of hepatocyte growth factor completely blocked the increased invasiveness of pancreatic cancer cells that was induced by co-culture with irradiated fibroblasts.

Even cell transformation, often ascribed to misrepaired DNA damage, is susceptible to microenvironment. The frequency of neoplastic transformation in cultured irradiated tracheal epithelial cells [35, 36] or C3H 10 T1/2 cells [37] is inversely correlated to the number of cells seeded, i.e. the fewer cells seeded the more transformed colonies were evident, suggesting that cell density/interactions suppressed this supposedly mutagenic consequence. Bauer and colleagues also showed that the frequency of radiation, chemical and virally mediated transformation of cultured human and rodent fibroblasts is actively suppressed by non-transformed cells (reviewed in [38]). In a process called intercellular induction of apoptosis, non-transformed cells induce selective ablation of transformed cells via apoptosis [39]. If this control system acts in vivo as efficiently as it does in vitro, tumor formation should require the establishment of resistance mechanisms directed against intercellular induction of apoptosis. Indeed, transformed foci from cells cultured from established tumors are not influenced by the presence of normal cells [39].

Terzaghi-Howe showed that transforming growth factor β (TGFβ) produced by the differentiated normal epithelial cells inhibited the growth and phenotype of radiation-transformed cells [40]. Bauer described three distinct, but competing, roles for TGFβ during transformation (reviewed in [41]: TGFβ actually helps maintain the transformed state of mesenchymal cells, enables non-transformed neighbors to recognize transformed cells, and triggers an apoptosis-inducing signal. Bauer and colleagues showed that the latter two processes are enhanced following very low radiation doses [42].

The Role of Stroma in Radiogenic Mammary Cancer

The mammary glands of mice irradiated with 0.5–5 Gy show rapid changes in the stromal matrix [4345]. Collagen type III is induced in the adipose stroma and peri-epithelial stroma, while collagen type I undergoes remodeling in the peri-epithelial stroma. Consistent with the immunoreactivity, production of new collagen fibrils is increased in the irradiated mouse mammary gland. Surprisingly, tenascin, which is down-regulated in adult mammary gland, is rapidly induced in the peri-epithelial stroma by radiation. Consistent with this wound-like stroma, TGFβ is activated and mediates much of the stromal remodeling [46]. In some ways, the irradiated microenvironment exhibits features of an ‘activated’ stroma, capable of further evolution that could modify the behavior and function of resident epithelial cells.

This rapid remodeling of the mammary microenvironment led us to hypothesize that the irradiated stroma augments breast cancer potential [2, 43, 47]. To test this, we created a radiation chimera by transplanting unirradiated, preneoplastic mammary cells to the mammary glands of irradiated hosts [10]. The mammary gland is uniquely suited to studies of stromal-epithelial interactions because the epithelium develops postnatally from a rudiment that is readily removed from the inguinal glands of female mice at 3 weeks of age. The so-called clearing of the fat pad results in a stable gland-free fat pad. Transplantation of normal mammary epithelial cells at the time of clearing produces a normal ductal outgrowth that fills the fat pad in 10 weeks and is indistinguishable in from intact gland by whole mounts or histological analysis (92).

In the radiation-chimera model, the mammary epithelium is surgically removed at puberty, the adult animal is irradiated some time later, and then non-irradiated mammary epithelial cells are transplanted into the irradiated host [10]. Initial studies used COMMA-1D mammary epithelial cells, which undergo mammary morphogenesis when transplanted into a 3-wk old mammary gland. They are non-tumorigenic if injected into the cleared fat pads of 3-wk old mice, subcutaneously in immature and adult mice, or into nude mice. Although clonal in origin, COMMA-1D cells harbor two mutant Trp53 alleles that may confer neoplastic potential [48]. When transplanted into mice irradiated 1–14 d earlier with 4 Gy, outgrowths rapidly developed tumors, ranging from a peak of 100% at day 3 and twice that of sham-irradiated mice at 14 d post-irradiation. Furthermore, tumors from irradiated animals were nearly five times larger than the few tumors that arose in sham-irradiated hosts, indicating that tumor biology, as well as frequency, was affected. These data support the idea that high dose radiation promotes carcinogenesis by inducing a hospitable tissue environment. The effect of the irradiated microenvironment on neoplastic progression persisted for several weeks and appears to be independent of systemic radiation effects (as tested by hemi-body irradiation), which support the hypothesis that non-mutagenic effects of radiation can contribute significantly to radiation carcinogenesis in vivo.

Similar observations have also been made for chemical carcinogenesis. Soto and colleagues showed that the stroma is a target of the n-methylnitrosurea (NMU) in the rat mammary gland [8]. NMU treatment of the rat mammary stroma promoted tumorigenesis of mammary epithelial cells that were not treated with the chemical carcinogen. In contrast, Medina and colleagues performed a similar experiment using 7,12-dimethylbenzanthrcene (DMBA)-treated mice and found that the treated mouse mammary stroma did not alter tumorigenesis by untreated preneoplastic mouse mammary outgrowth lines [49].

In preliminary studies (Nguyen et al., submitted), we used the radiation chimera transplanted with Trp53 null mammary epithelium to assess radiation dose dependence. The Trp53 null mammary epithelium progress from ductal outgrowths to ductal carcinoma in situ to invasive breast carcinomas over the course of a year and exhibit many features similar to those of human tumors, including genomic instability and heterogeneous tumor histology that can be either estrogen receptor (ER) positive or negative [50, 51]. The experiments demonstrate that low dose (<1 Gy) host irradiation reduces tumor latency, but also affects the tumor type in a manner similar as to that found in breast tumors of women treated with radiation [25, 27].

Mammary Composition and Radiation

Girls exposed to ionizing radiation at Nagasaki-Hiroshima who were peri-pubertal (approximately aged 10–14 years) were much more likely to develop breast cancer than older girls or adult women who were exposed to comparable radiation doses [52]. Similar results were found for high dose radiation exposures to the breast from fluoroscopy for radiation therapy for Hodgkin’s disease [5355]. A major question that arises from these epidemiological data is: What is the biological basis for the window of susceptibility during adolescence? Ongoing studies in our laboratory suggest that radiation exposure can affect stem cell self-renewal and that it may be mediated through the microenvironment.

It is becoming clearer that “stemness” as measured ex vivo is not a single property, but several properties that can be manifest under different conditions [56]. A stem cell is the mechanism for regenerating tissue after injury [57]. Hence, it must not only be capable of producing many differentiated progeny, but able to switch between these options when appropriate. Thus, the properties, and probably the number, of stem cells can change in response to circumstances, including experimental manipulations. A variety of recent studies in tissues ranging from brain to liver have brought to light the ability of partially-differentiated cells to dedifferentiate and replenish the supply of true stem cells. This degree of plasticity is unexpected based on the traditional understanding of stem-cell function [56], but may be relevant to the concern that these cells are particularly susceptible to changes in the stroma.

Recent studies in mouse mammary gland have provided new insight into how signaling between cells determines the activity of the stem cell pool and cell-fate decisions that determine mammary lineages. Studies from Visvader show that macrophages influence mammary stem cell activity and support stem/progenitor cell functions [58]. Some research suggests that bone marrow derived cells might contribute to the niche, at least in cancer [59]. It is clear that stromal signals are important; the best studied is Wnt/β-catenin. Wnt proteins are secreted morphogens that are required for cell-fate specification, progenitor-cell proliferation and the control of asymmetric cell division. Wnt binds to Frizzled receptors to induce β-catenin stabilization and translocation to the nucleus where it interacts with transcriptional factor TCF/Lef-1 [60]. MMTV-Wnt1 expands cell populations expressing CK4, Sca1, CK6, CD24med/49fhigh, and other profiles that have been associated with stem and early progenitor populations. Increased β-catenin transcriptional activity reduced latency in mammary tumor models, with tumors displaying a higher proportion of progenitor cell markers [61, 62]. This experimental model supports the concept that increased stem cell number may itself be a major risk factor for cancer development [63].

A remarkable series of experiments by Smith and colleagues demonstrate that mammary epithelial cells can reprogram diverse cell types [64, 65]. Transplantation into the mammary fat pad of normal mammary epithelial cells mixed with marked cells isolated from mature testis induce conversion or re-programming of the beta-galactosidase marked testis cells with mammary repopulating capacity. The idea that the niche can supersede established phenotype demonstrates that this population has a high degree of phenotypic plasticity [66]. We showed that a single radiation exposure predisposes human mammary epithelial cells to undergo TGFβ-mediated epithelial-to-mesenchymal transition (EMT) [67]. Recent studies from Weinberg indicate that transition through an EMT can endow otherwise differentiated cells with a stem-like phenotype [68]. Our in vitro model suggests that irradiation alters the response to TGFβ, which when taken together with our prior observations that radiation induces TGFβ activation, suggest that radiation can operate at multiple levels to deregulate stem cell pool size.

Concluding Remarks

Stroma is a critical player in carcinogenesis; indeed the normal stroma is a significant barrier to malignancy. An important question arises: how does stroma convert from a defensive to offensive player in tumorigenesis? Disruption of stromal-epithelial interactions is an understudied activity of carcinogens and a novel avenue through which to explore new strategies for intervening in the neoplastic process. Our studies using radiation show that that the mammary stroma can be activated by even a single radiation exposure, putting the stroma into play much earlier than generally thought in the carcinogenic process.

Since radiation is a well-documented human breast carcinogen, understanding its actions is essential for predicting risk medical exposures. A deeper understanding of radiation’s action as a carcinogen might also reveal previously unsuspected routes to spontaneous cancer, just as the knowledge of its effect on DNA damage and responses has focused attention on DNA damage pathway gene polymorphisms in general populations. Moreover, the basic biology of radiation overlaps that of oxidative stress and can be used to probe tissue response to reactive oxygen species generated during other tissue processes like inflammation. Finally, understanding how radiation causes cancer may provide the means to protect children from the risk associated with exposures to necessary diagnostic procedures like computerized tomography scans, which can result in doses comparable to those in which risk has been measured, or from radiation treatment for other cancers.

Acknowledgements

The author wishes to acknowledge funding from NASA Specialized Center for Research in Radiation Health Effects, the Low Dose Radiation Program of the Office of Biological and Environmental Research, United States Department of Energy DE AC03 76SF00098, and the Bay Area Breast Cancer and the Environment Research Center grant number U01 ES012801 from the National Institute of Environmental Health Sciences, NIH and the National Cancer Institute, NIH.

Abbreviations

TGFβ

Transforming growth factor β

ER

estrogen receptor

PR

progesterone receptor

EMT

epithelial-mesenchymal transition

References

  • 1.Rubin H. Cancer as a dynamic developmental disorder. Cancer Res. 1985;45:2935–2942. [PubMed] [Google Scholar]
  • 2.Barcellos-Hoff MH. The potential influence of radiation-induced microenvironments in neoplastic progression. J Mammary Gland Biol Neoplasia. 1998;3:165–175. doi: 10.1023/a:1018794806635. [DOI] [PubMed] [Google Scholar]
  • 3.Sonnenschein C, Soto AM. Somatic mutation theory of carcinogenesis: why it should be dropped and replaced. Mol Carcinog. 2000;29(4):205–211. doi: 10.1002/1098-2744(200012)29:4<205::aid-mc1002>3.0.co;2-w. [DOI] [PubMed] [Google Scholar]
  • 4.Bissell MJ, Radisky D. Putting tumours in context. Nat Rev Cancer. 2001;1(1):1–11. doi: 10.1038/35094059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wiseman BS, Werb Z. Stromal effects on mammary gland development and breast cancer. Science. 2002;296(5570):1046–1049. doi: 10.1126/science.1067431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kuperwasser C, Chavarria T, Wu M, Magrane G, Gray JW, Carey L, et al. From the cover: reconstruction of functionally normal and malignant human breast tissues in mice. PNAS. 2004;101(14):4966–4971. doi: 10.1073/pnas.0401064101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S, et al. TGF-{beta} signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science. 2004;303(5659):848–851. doi: 10.1126/science.1090922. [DOI] [PubMed] [Google Scholar]
  • 8.Maffini MV, Soto AM, Calabro JM, Ucci AA, Sonnenschein C. The stroma as a crucial target in rat mammary gland carcinogenesis. J Cell Sci. 2004;117(8):1495–1502. doi: 10.1242/jcs.01000. [DOI] [PubMed] [Google Scholar]
  • 9.de Visser KE, Eichten A, Coussens LM. Paradoxical roles of the immune system during cancer development. Nat Rev Cancer. 2006;6(1):24–37. doi: 10.1038/nrc1782. [DOI] [PubMed] [Google Scholar]
  • 10.Barcellos-Hoff MH, Ravani SA. Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Res. 2000;60:1254–1260. [PubMed] [Google Scholar]
  • 11.NAS/NRC. Health risks from exposure to low levels of ionizing radiation: phase 2. Washington: National Academy Press; 2006. [PubMed] [Google Scholar]
  • 12.Barcellos-Hoff MH. Cancer as an emergent phenomenon in systems radiation biology. Radiat Env Biophys. 2007;47(1):33–38. doi: 10.1007/s00411-007-0141-0. [DOI] [PubMed] [Google Scholar]
  • 13.Herskind C, Rodemann HP. Spontaneous and radiation-induced differentiationof fibroblasts. Exp Gerontol. 2000;35(6–7):747–755. doi: 10.1016/s0531-5565(00)00168-6. [DOI] [PubMed] [Google Scholar]
  • 14.Rave-Frank M, Virsik-Kopp P, Pradier O, Nitsche M, Grunefeld SHS. In vitro response of human dermal fibroblasts to X-irradiation: relationship between radiation-induced clonogenic cell death, chromosome aberrations and markers of proliferative senescence or differentiation. Int J Radiat Biol. 2001;77:1163–1174. doi: 10.1080/09553000110086372. [DOI] [PubMed] [Google Scholar]
  • 15.Park CC, Henshall-Powell RL, Erickson AC, Talhouk R, Parvin B, Bissell MJ, et al. Ionizing radiation induces heritable disruption of epithelial cell interactions. Proc Natl Acad Sci USA. 2003;100(19):10728–10733. doi: 10.1073/pnas.1832185100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tsai KK, Chuang EY, Little JB, Yuan ZM. Cellular mechanisms for low-dose ionizing radiation-induced perturbation of the breast tissue microenvironment. Cancer Res. 2005;65(15):6734–6744. doi: 10.1158/0008-5472.CAN-05-0703. [DOI] [PubMed] [Google Scholar]
  • 17.Kadhim MA, Lorimore SA, Hepburn MD, Goodhead DT, Buckle VJ, Wright EG. Alpha-particle-induced chromosomal instability in human bone marrow cells. Lancet. 1994;344(8928):987–988. doi: 10.1016/s0140-6736(94)91643-8. [DOI] [PubMed] [Google Scholar]
  • 18.Kadhim MA, Lorimore SA, Townsend KM, Goodhead DT, Buckle VJ, Wright EG. Radiation-induced genomic instability: delayed cytogenetic aberrations and apoptosis in primary human bone marrow cells. Int J Radiat Biol. 1995;67(3):287–293. doi: 10.1080/09553009514550341. [DOI] [PubMed] [Google Scholar]
  • 19.Kadhim MA, Macdonald DA, Goodhead DT, Lorimore SA, Marsden SJ, Wright EG. Transmission of chromosomal instability after plutonium alpha-particle irradiation [see comments] Nature. 1992;355(6362):738–740. doi: 10.1038/355738a0. [DOI] [PubMed] [Google Scholar]
  • 20.Clutton SM, Townsend KM, Goodhead DT, Ansell JD, Wright EG. Differentiation and delayed cell death in embryonal stem cells exposed to low doses of ionising radiation. Cell Death Differ. 1996;3(1):141–148. [PubMed] [Google Scholar]
  • 21.Limoli CL, Kaplan MI, Corcoran J, Meyers M, Boothman DA, Morgan WF. Chromosomal instability and its relationship to other end points of genomic instability. Cancer Res. 1997;57(24):5557–5563. [PubMed] [Google Scholar]
  • 22.Preston DL, Pierce DA, Shimizu Y, Ron E, Mabuchi K. Dose response and temporal patterns of radiation-associated solid cancer risks. Health Phys. 2003;85(1):43–46. doi: 10.1097/00004032-200307000-00010. [DOI] [PubMed] [Google Scholar]
  • 23.Preston DL, Ron E, Tokuoka S, Funamoto S, Nishi N, Soda M, et al. Solid cancer incidence in atomic bomb survivors: 1958–1998. Radiat Res. 2007;168(1):1–64. doi: 10.1667/RR0763.1. [DOI] [PubMed] [Google Scholar]
  • 24.Preston DL, Mattsson A, Holmberg E, Shore R, Hildreth NG, Boice JD., Jr Radiation effects on breast cancer risk: a pooled analysis of eight cohorts. Radiat Res. 2002;158(2):220–235. doi: 10.1667/0033-7587(2002)158[0220:reobcr]2.0.co;2. [DOI] [PubMed] [Google Scholar]
  • 25.Castiglioni F, Terenziani M, Carcangiu ML, Miliano R, Aiello P, Bertola L, et al. Radiation effects on development of HER2-positive breast carcinomas. Clin Cancer Res. 2007;13(1):46–51. doi: 10.1158/1078-0432.CCR-06-1490. [DOI] [PubMed] [Google Scholar]
  • 26.Van Leeuwen FE, Klokman WJ, Stovall M, Dahler EC, van’t Veer MB, Noordijk EM, et al. Roles of radiation dose, chemotherapy, and hormonal factors in breast cancer following Hodgkin’s disease. J Natl Cancer Inst. 2003;95(13):971–980. doi: 10.1093/jnci/95.13.971. [DOI] [PubMed] [Google Scholar]
  • 27.Broeks A, Braaf LM, Wessels LF, Van de Vjver M, De Bruin ML, Stovall M, et al. Radiation-associated breast tumors display a distinct gene expression profile. Int J Radiat Oncol Biol Phys. 2010;76(2):540–547. doi: 10.1016/j.ijrobp.2009.09.004. [DOI] [PubMed] [Google Scholar]
  • 28.Barcellos-Hoff MH. Integrative radiation carcinogenesis: interactions between cell and tissue responses to DNA damage. Semin Cancer Biol. 2005;15(2):138–148. doi: 10.1016/j.semcancer.2004.08.010. [DOI] [PubMed] [Google Scholar]
  • 29.Kaplan HS, Carnes WH, Brown MB, Hirsch BB. Indirect induction of lymphomas in irradiated mice: I. Tumor incidence and morphology in mice bearing nonirradiated thymic grafts. Cancer Res. 1956;16(5):422–425. [PubMed] [Google Scholar]
  • 30.Kaplan HS, Brown MB, Hirsch BB, Carnes WH. Indirect induction of lymphomas in irradiated mice: II. Factor of irradiation of the host. Cancer Res. 1956;16(5):426–428. [PubMed] [Google Scholar]
  • 31.Carnes WH, Kaplan HS, Brown MB, Hirsch BB. Indirect induction of lymphomas in irradiated mice: III. Role of the thymic graft. Cancer Res. 1956;16(5):429–433. [PubMed] [Google Scholar]
  • 32.Billingham RE, Orr JW, Woodhouse DL. Transplantation of skin components during chemical carcinogenesis with 20-methylcholanthrene. Br J Cancer. 1951;5:417–432. doi: 10.1038/bjc.1951.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Morgan JE, Gross JG, Pagel CN, Beauchamp JR, Fassati A, Thrasher AJ, et al. Myogenic cell proliferation and generation of a reversible tumorigenic phenotype are triggered by preirradiation of the recipient site. J Cell Biol. 2002;157(4):693–702. doi: 10.1083/jcb.200108047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ohuchida K, Mizumoto K, Murakami M, Qian L-W, Sato N, Nagai E, et al. Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res. 2004;64(9):3215–3222. doi: 10.1158/0008-5472.can-03-2464. [DOI] [PubMed] [Google Scholar]
  • 35.Terzaghi M, Little JB. X-radiation-induced transformation in C3H mouse embryo-derived cell line. Cancer Res. 1976;36:1367–1374. [PubMed] [Google Scholar]
  • 36.Terzaghi M, Nettesheim P. Dynamics of neoplastic development in carcinogen-exposed tracheal mucosa. Cancer Res. 1979;39:3004–3010. [PubMed] [Google Scholar]
  • 37.Kennedy AR, Fox M, Murphy G, Little JB. Relationship between x-ray exposure and malignant transformation in C3H 10 T1/2 cells. Proc Natl Acad Sci USA. 1980;77(12):7262–7266. doi: 10.1073/pnas.77.12.7262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bauer G. Elimination of transformed cells by normal cells: a novel concept for the control of carcinogenesis. Histol Histopathol. 1996;11(1):237–255. [PubMed] [Google Scholar]
  • 39.Engelmann I, Eichholtz-Wirth H, Bauer G. Ex vivo tumor cell lines are resistant to intercellular induction of apoptosis and independent of exogenous survival factors. Anticancer Res. 2000;20(4):2361–2370. [PubMed] [Google Scholar]
  • 40.Terzaghi-Howe M. Inhibition of carcinogen-altered rat tracheal epithelial cell proliferation by normal epithelial cells in vivo. Carcinogenesis. 1986;8:145–150. doi: 10.1093/carcin/8.1.145. [DOI] [PubMed] [Google Scholar]
  • 41.Häufel T, Dormann S, Hanusch J, Schwieger A, Bauer G. Three distinct roles for TGF-beta during intercellular induction of apoptosis: a review. Anticancer Res. 1999;19(1A):105–111. [PubMed] [Google Scholar]
  • 42.Portess DI, Bauer G, Hill MA, O’Neill P. Low dose irradiation of non-transformed cells stimulates the selective removal of precancerous cells via intercellular induction of apoptosis. Cancer Res. 2007;67(3):1246–1253. doi: 10.1158/0008-5472.CAN-06-2985. [DOI] [PubMed] [Google Scholar]
  • 43.Barcellos-Hoff MH. Radiation-induced transforming growth factor β and subsequent extracellular matrix reorganization in murine mammary gland. Cancer Res. 1993;53:3880–3886. [PubMed] [Google Scholar]
  • 44.Ehrhart EJ, Gillette EL, Barcellos-Hoff MH. Immunohistochemical evidence of rapid extracellular matrix remodeling after iron-particle irradiation of mouse mammary gland. Rad Res. 1996;145:157–162. [PubMed] [Google Scholar]
  • 45.Ehrhart EJ, Carroll A, Segarini P, Tsang ML-S, Barcellos-Hoff MH. Latent transforming growth factor-β activation in situ: quantitative and functional evidence following low dose irradiation. FASEB J. 1997;11:991–1002. doi: 10.1096/fasebj.11.12.9337152. [DOI] [PubMed] [Google Scholar]
  • 46.Barcellos-Hoff MH, Derynck R, Tsang ML-S, Weatherbee JA. Transforming growth factor-β activation in irradiated murine mammary gland. J Clin Invest. 1994;93:892–899. doi: 10.1172/JCI117045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Barcellos-Hoff MH. How do tissues respond to damage at the cellular level? The role of cytokines in irradiated tissues. Radiat Res. 1998;150(5):S109–S120. [PubMed] [Google Scholar]
  • 48.Jerry DJ, Medina D, Butel JS. p53 mutations in COMMA-D cells. In Vitro Cell. Dev Biol. 1994;30A:87–89. doi: 10.1007/BF02631398. [DOI] [PubMed] [Google Scholar]
  • 49.Medina D, Kittrell FS. Stroma is not a major target in 7, 12-dimethlybenzanthracene mediated tumorigenesis of mouse mammary preneoplasia. J Cell Sci. 2005;118:123–127. doi: 10.1242/jcs.01597. [DOI] [PubMed] [Google Scholar]
  • 50.Jerry DJ, Kittrell FS, Kuperwasser C, Laucirica R, Dickinson ES, Bonilla PJ, et al. A mammary-specific model demonstrates the role of the p53 tumor suppressor gene in tumor development. Oncogene. 2000;19(8):1052–1058. doi: 10.1038/sj.onc.1203270. [DOI] [PubMed] [Google Scholar]
  • 51.Medina D, Kittrell FS, Shepard A, Stephens LC, Jiang C, Lu J, et al. Biological and genetic properties of the p53 null preneoplastic mammary epithelium. FASEB J. 2002;16(8):881–883. doi: 10.1096/fj.01-0885fje. [DOI] [PubMed] [Google Scholar]
  • 52.Tokunaga M, Land CE, Aoki Y, Yamamoto T, Asano M, Sato E, et al. Proliferative and nonproliferative breast disease in atomic bomb survivors. Results of a histopathologic review of autopsy breast tissue. Cancer. 1993;72(5):1657–1665. doi: 10.1002/1097-0142(19930901)72:5<1657::aid-cncr2820720527>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
  • 53.Boice JD, Jr, Preston D, Davis FG, Monson RR. Frequent chest x-ray fluoroscopy and breast cancer incidence among tuberculosis patients in Massachusetts. Radiat Res. 1991;125:214–222. [PubMed] [Google Scholar]
  • 54.Hancock SL, Tucker MA, Hoppe RT. Breast cancer after treatment of Hodgkin's disease. J Natl Cancer Inst. 1993;85(1):25–31. doi: 10.1093/jnci/85.1.25. [DOI] [PubMed] [Google Scholar]
  • 55.Howe GR, McLaughlin J. Breast cancer mortality between 1950 and 1987 after exposure to fractionated moderate-dose-rate ionizing radiation in the Canadian fluoroscopy cohort study and a comparison with breast cancer mortality in the atomic bomb survivor study. Radiat Res. 1996;145:694–707. [PubMed] [Google Scholar]
  • 56.Booth C, Potten CS. Gut instincts: thoughts on intestinal epithelial stem cells. J Clin Invest. 2000;105(11):1493–1499. doi: 10.1172/JCI10229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Potten CS, Loeffler M. Stem cells: attributes, cycles, spirals, pitfalls and uncertainties. Lessons for and from the crypt. Development. 1990;110(4):1001–1020. doi: 10.1242/dev.110.4.1001. [DOI] [PubMed] [Google Scholar]
  • 58.Gyorki D, Asselin-Labat M-L, van Rooijen N, Lindeman G, Visvader J. Resident macrophages influence stem cell activity in the mammary gland. Breast Cancer Res. 2009;11(4):R62. doi: 10.1186/bcr2353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Psaila B, Lyden D. The metastatic niche: adapting the foreign soil. Nat Rev Cancer. 2009;9(4):285–293. doi: 10.1038/nrc2621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hatsell S, Rowlands T, Hiremath M, Cowin P. beta-Catenin and Tcfs in mammary development and cancer. J Mammary Gland Biol Neoplasia. 2003;8:145–158. doi: 10.1023/a:1025944723047. [DOI] [PubMed] [Google Scholar]
  • 61.Liu BY, McDermott SP, Khwaja SS, Alexander CM. The transforming activity of Wnt effectors correlates with their ability to induce the accumulation of mammary progenitor cells. PNAS. 2004;101(12):4158–4163. doi: 10.1073/pnas.0400699101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Teissedre B, Pinderhughes A, Incassati A, Hatsell S, Hiremath M, Cowin P. MMTV-Wnt1 and -DeltaN89beta-catenin induce canonical signaling in distinct progenitors and differentially activate Hedgehog signaling within mammary tumors. PLoS ONE. 2009;4:e4537. doi: 10.1371/journal.pone.0004537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Incassati A, Pinderhughes A, Eelkema R, Cowin P. Links between transforming growth factor-beta and canonical Wnt signaling yield new insights into breast cancer susceptibility, suppression and tumor heterogeneity. Breast Cancer Res. 2009;11(3):103. doi: 10.1186/bcr2253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Booth BW, Mack DL, Androutsellis-Theotokis A, McKay RDG, Boulanger CA, Smith GH. The mammary microenvironment alters the differentiation repertoire of neural stem cells. Proc Natl Acad Sci. 2008;105(39):14891–14896. doi: 10.1073/pnas.0803214105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Boulanger CA, Mack DL, Booth BW, Smith GH. Interaction with the mammary microenvironment redirects spermatogenic cell fate in vivo 10.1073/pnas.0611637104. PNAS. 2007;104(10):3871–3876. doi: 10.1073/pnas.0611637104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Smith G, Medina D. Re-evaluation of mammary stem cell biology based on in vivo transplantation. Breast Cancer Res. 2008;10(1):203. doi: 10.1186/bcr1856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Andarawewa KL, Erickson AC, Chou WS, Costes SV, Gascard P, Mott JD, et al. Ionizing radiation predisposes nonmalignant human mammary epithelial cells to undergo transforming growth factor beta Induced epithelial to mesenchymal transition. Cancer Res. 2007;67:8662–8670. doi: 10.1158/0008-5472.CAN-07-1294. [DOI] [PubMed] [Google Scholar]
  • 68.Mani SA, Guo W, Liao M-J, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–715. doi: 10.1016/j.cell.2008.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES