Biology Contribution
Differential Effects of X-Rays and High-Energy 56Fe Ions on Human Mesenchymal Stem Cells

https://doi.org/10.1016/j.ijrobp.2008.10.002Get rights and content

Purpose

Stem cells hold great potential for regenerative medicine, but they have also been implicated in cancer and aging. How different kinds of ionizing radiation affect stem cell biology remains unexplored. This study was designed to compare the biological effects of X-rays and of high–linear energy transfer (LET) 56Fe ions on human mesenchymal stem cells (hMSC).

Methods and Materials

A multi-functional comparison was carried out to investigate the differential effects of X-rays and 56Fe ions on hMSC. The end points included modulation of key markers such as p53, cell cycle progression, osteogenic differentiation, and pathway and networks through transcriptomic profiling and bioinformatics analysis.

Results

X-rays and 56Fe ions differentially inhibited the cell cycle progression of hMSC in a p53-dependent manner without impairing their in vitro osteogenic differentiation process. Pathway and network analyses revealed that cytoskeleton and receptor signaling were uniquely enriched for low-dose (0.1 Gy) X-rays. In contrast, DNA/RNA metabolism and cell cycle regulation were enriched for high-dose (1 Gy) X-rays and 56Fe ions, with more significant effects from 56Fe ions. Specifically, DNA replication, DNA strand elongation, and DNA binding/transferase activity were perturbed more severely by 1 Gy 56Fe ions than by 1 Gy X-rays, consistent with the significant G2/M arrest for the former while not for the latter.

Conclusions

56Fe ions exert more significant effects on hMSC than X-rays. Since hMSC are the progenitors of osteoblasts in vivo, this study provides new mechanistic understandings of the relative health risks associated with low- and high-dose X-rays and high-LET space radiation.

Introduction

Stem cells hold great potential for regenerative medicine because they can self-renew and differentiate along tissue-specific lineages. Mesenchymal stem cells (MSCs), the multipotent stromal cells from bone marrow, play important roles in the maintenance and repair of various tissues, including bone, cartilage, and muscle (1). The differentiation of MSCs into osteoblasts is controlled by the transcription factor Runx2 (2). Stem cells also respond to genotoxic stresses, for example, ionizing radiation (IR), which may lead to the induction of mutations and chromosome aberrations, transient cell-cycle arrest, apoptosis, mitotic catastrophe, cellular senescence, or malignant transformation. Abnormalities in stem cell biology have been implicated in cancer and aging, but the mechanisms are poorly understood (3). For example, MSCs have been shown as a target for neoplastic transformation following manipulations 4, 5. However, major challenges still remain to define the normal molecular signaling and tumorigenic pathways in stem cells.

Because of their high tissue-penetrating property, IR such as X-rays/gamma rays is used in CT scans, cancer radiotherapy, and nuclear medicine. However, IR is also a well-known DNA-damaging agent, clastogen, and carcinogen 6, 7. During deep space missions, space crews encounter an elevated radiation environment that includes high-energy–charged (HZE) particles, such as 56Fe ions from the galactic cosmic rays (8). Such densely ionizing radiation induces more severe clustering of damage on DNA compared to X-rays, and gives rise to complex DNA double-strand breaks (DSBs) that are more prone to be misrepaired and cause permanent genomic changes (9). Another complicating factor during space travel is the possibility of synergistic relationships with other risk factors, such as microgravity. The possibility that HZE radiation affects the differentiation process of human mesenchymal stem cells (hMSC) into osteoblasts, important for bone regeneration and loss in space because of microgravity, has not been investigated.

Advances in systems biology have opened up new possibilities for elucidating the molecular mechanisms underlying cellular responses to IR. Global gene expression changes in response to IR have been reported for Saccharomyces cerevisiae(10), murine brain (11), normal human skin fibroblasts (12), and human lung fibroblasts (13). These studies suggested that IR regulates gene expression at the transcriptional level in a dose-dependent manner and depending on radiation quality, for example, X-rays vs. HZE particles. On the other hand, proteomic and transcriptomic studies of hMSC have provided important insights into their proliferation and differentiation 14, 15. However, no direct correlations between phenotypic radioresponses and pathway regulations have been reported for hMSC, particularly in the context of osteogenic differentiation.

Here we describe the first comprehensive study of the radioresponses of hMSC to X-rays and high energy 56Fe ions. We show that neither X-rays nor 56Fe ions impaired the in vitro osteogenic differentiation process of hMSC. We further show that they differentially regulated the cell cycle progression of hMSC in a dose-dependent and p53-dependent manner. Pathway and network analyses of transcriptomic profiles revealed the underlying mechanisms.

Section snippets

Cell culture

Human mesenchymal stem cells were obtained from Lonza Group (Walkersville, MD). The log number was 4F1560, which was derived from the bone marrow of a healthy 23-year-old woman. We characterized expanded hMSC (up to passage 10) again by their surface markers and differentiation potential as we described previously 14, 15. For cell proliferation without differentiation, hMSC (typical passage number, five to eight) were cultured in MSC growth medium, supplemented with prescreened fetal bovine

X-rays and 56Fe ions inhibited proliferation without impairing in vitro osteogenic differentiation process of hMSC

Figure 1a demonstrates clear morphologic changes in hMSC over a 7-day period when stimulated with osteogenic differentiation media. These changes became noticeable starting on Day 3 with an increasingly three-dimensional, cobblestone-like morphology potentially indicative of osteogenic differentiation. However, no obvious morphologic differences were detected in irradiated cultures when comparing between radiation doses or types at any given time point. Quantitative ALP activity data in Fig. 1b

Discussion

Ionizing radiation induces a variety of lesions to DNA, including single-strand breaks, double-strand breaks, and base damages, depending on radiation quality and dose. 56Fe ions induce denser clustering of DNA lesions than X-rays, increasing the likelihood of failed repairs. In addition, DSBs in close proximity can misrejoin to give rise to chromosome aberrations and permanent genetic damage (22). Our results indicated that these biophysical events were associated with perturbed DNA synthesis,

Conclusion

In conclusion, we showed both phenotypically and mechanistically the differential effects of X-rays and high-energy 56Fe ions on hMSC. Our results will assist the Department of Energy with risk estimations for low-dose radiation, and the National Aeronautics and Space Admininstration with HZE risk estimations and countermeasures for space exploration.

Acknowledgments

The authors thank Dr. M. Bissell for advice and Drs. B. Sutherland and P. Guida for help with the logistics of National Aeronautics and Space Administration (NASA) Space Radiation Laboratory (NSRL) experiments. This work was supported by the Low Dose Radiation Research Program jointly funded by the U.S. Department of Energy (DOE) and NASA (to D.W.). A.W. acknowledges the funding support from the DOE Low Dose Radiation Research Program. S.L. acknowledges the funding support from the National

References (39)

  • C.M. Beausejour et al.

    Ageing: Balancing regeneration and cancer

    Nature

    (2006)
  • D. Rubio et al.

    Spontaneous human adult stem cell transformation

    Cancer Res

    (2005)
  • N. Serakinci et al.

    Adult human mesenchymal stem cell as a target for neoplastic transformation

    Oncogene

    (2004)
  • E.J. Hall et al.

    Genomic instability and bystander effects induced by high-LET radiation

    Oncogene

    (2003)
  • W.F. Morgan

    Is there a common mechanism underlying genomic instability, bystander effects and other nontargeted effects of exposure to ionizing radiation?

    Oncogene

    (2003)
  • G.A. Nelson

    Fundamental space radiobiology

    Gravit Space Biol Bull

    (2003)
  • G.W. Birrell et al.

    Transcriptional response of Saccharomyces cerevisiae to DNA-damaging agents does not identify the genes that protect against these agents

    Proc Natl Acad Sci U S A

    (2002)
  • E. Yin et al.

    Gene expression changes in mouse brain after exposure to low-dose ionizing radiation

    Int J Radiat Biol

    (2003)
  • L.H. Ding et al.

    Gene expression profiles of normal human fibroblasts after exposure to ionizing radiation: A comparative study of low and high doses

    Radiat Res

    (2005)
  • Cited by (43)

    • Human mesenchymal stromal cells maintain their stem cell traits after high-LET particle irradiation – Potential implications for particle radiotherapy and manned space missions

      2022, Cancer Letters
      Citation Excerpt :

      It should be noted that the previously reported strong G2/M arrest in MSCs was mainly observed after considerably higher radiation doses [13,14,38]. Another publication has previously demonstrated a stronger G2/M block after 1 Gy 56Fe ions compared to 1 Gy photons; however, in this study, RBE values were not taken into account for the experimental design, making a comparison between these two radiation types difficult [32]. Particle radiation-induced cell cycle effects have been linked to the downregulation of cyclins B1 and E2 even after low doses of 56Fe [32].

    • Total body irradiation-induced colon damage is prevented by nitrate-mediated suppression of oxidative stress and homeostasis of the gut microbiome

      2020, Nitric Oxide - Biology and Chemistry
      Citation Excerpt :

      The opposite changes were observed for Bcl-2 expression (Fig. 3D). IR inhibits cell cycle progression in human mesenchymal stem cells and induces senescence by activating the p53 pathway [22]. Moreover, IR-induced senescence caused long-term damage by preventing the renewal of tissues progenitor cells [23].

    • Chronological age affects msc senescence in vitro—a systematic review

      2021, International Journal of Molecular Sciences
    View all citing articles on Scopus

    Conflict of interest: none.

    Supplementary material for this article can be found at www.redjournal.org.

    View full text