Elsevier

Placenta

Volume 35, Issue 2, February 2014, Pages 85-91
Placenta

The impact of ionizing radiation on placental trophoblasts

https://doi.org/10.1016/j.placenta.2013.12.011Get rights and content

Abstract

Introduction

Exposure to low-dose radiation is widespread and attributable to natural sources. However, occupational, medical, accidental, and terrorist-related exposures remain a significant threat. Information on radiation injury to the feto-placental unit is scant and largely observational. We hypothesized that radiation causes trophoblast injury, and alters the expression of injury-related transcripts in vitro or in vivo, thus affecting fetal growth.

Methods

Primary human trophoblasts (PHTs), BeWo or NCCIT cells were irradiated in vitro, and cell number and viability were determined. Pregnant C57Bl/6HNsd mice were externally irradiated on E13.5, and placentas examined on E17.5. RNA expression was analyzed using microarrays and RT-qPCR. The experiments were repeated in the presence of the gramicidin S (GS)-derived nitroxide JP4-039, used to mitigate radiation-induced cell injury.

Results

We found that survival of in vitro–irradiated PHT cell was better than that of irradiated BeWo trophoblast cell line or the radiosensitive NCCIT mixed germ cell tumor line. Radiation altered the expression of several trophoblast genes, with a most dramatic effect on CDKN1A (p21, CIP1). Mice exposed to radiation at E13.5 exhibited a 25% reduction in mean weight by E17.5, and a 9% reduction in placental weight, which was associated with relatively small changes in placental gene expression. JP4-039 had a minimal effect on feto-placental growth or on gene expression in irradiated PHT cells or mouse placenta.

Discussion and conclusion

While radiation affects placental trophoblasts, the established placenta is fairly resistant to radiation, and changes in this tissue may not fully account for fetal growth restriction induced by ionizing radiation.

Introduction

Exposure to ionizing radiation remains a reality in today's world. Worldwide, the average annual exposure to natural radiation is about 2.4 milli Sievert (mSv) [1]. Occupational exposures are most relevant to people working with nuclear fuel and medical devices, in defense-related functions, and in occupations associated with enhanced exposure to natural sources of radiation. For example, aircrew members are exposed to 5–8 mSv per hour while flying [1]. Medical sources of radiation include diagnostic procedures that expose individuals to low doses (commonly 0.1–10 mSv) and therapeutic exposures, typically 20–60 Gray (Gy), to a targeted tissue [1]. Accidental exposures in nuclear fuel processing plants typically expose workers to 1–20 Gy [1]. These risks may be greatly amplified if “dirty bombs” are deployed by terrorists against civilians [2].

Research into diagnosis, treatment, and prevention of radiation injury in pregnancy is limited by appropriate ethical concerns and by the scarcity of information on mechanisms underlying the effect of ionizing radiation on the developing feto-placental unit. Anecdotal reports or observational studies have generated some information pertaining to gestational age and radiation dose. During the pre-implantation period, as little as 0.3 Gy is lethal to the mouse embryo [3]. In the post-implantation period, the main risks from radiation include embryonic death, congenital anomalies, growth restriction, and neurologic maldevelopment [4]. Exposing mice at E14 to 0.3–1.5 Gy of whole body irradiation caused decreased neonatal body length and body weight [5]. Minimal effects on litter size or fetal growth were observed when mice at E7-16 were exposed to low dose radiation, 10–13 mSv per day over 10 days [6]. In humans, data from children exposed to in utero radiation after catastrophic events in Hiroshima, Nagasaki, and Chernobyl revealed lower height and weight in adolescence [4], [7].

Ionizing radiation damages tissues through diverse mechanisms [8]. A major consequence of radiation is direct and indirect DNA damage. Direct effects include the transfer of kinetic energy from radioactive particles to the DNA backbone, which breaks phosphodiester bonds. Indirect effects include the generation of reactive oxygen species, which cause DNA double-strand breaks and cell-cycle arrest. Other types of injury include p53-dependent and -independent apoptosis [9], mitochondrial damage, loss of regenerative capacity, and premature senescence [8]. NFκB mediates several radiation-stimulated signal transduction pathways, which may explain the degree of radiation-sensitivity of differing cells types [10]. These pathways implicate CDKN1A (also known as p21, CIP1), epidermal growth factor receptors, and the apoptosis-related proteins BAX and BCL2 in radiation injury [11]. Whereas radiation-induced pathways have been interrogated in non-placental cell types, there are no studies of radiation injury to cultured primary human trophoblast (PHT) cells; there has been a single study that included the choriocarcinoma line JEG3 and showed no effect on gene expression of gap junction protein alpha 1 [12].

Methods to scavenge reactive oxygen species have been proposed to mitigate radiation damage. This effect has been attributed, at least in part, to the action of manganese superoxide dismutase (MnSOD, [13]). The nitroxides, which have superoxide dismutase–mimetic activity and inhibit lipid peroxidation [14], constitute one such class of radioprotectors. JP4-039 is a nitroxide linked to a short alkene isostere analog of hemigramicidin S, which allows concentration at the mitochondrial membrane, the site of radiation-induced lipid peroxidation [15]. It has been shown to protect against radiation damage in vivo [16], [17]. In this study, we tested the hypothesis that ionizing radiation causes injury to PHT cells in vitro and to the mouse placenta in vivo. We also assessed whether the nitroxide JP4-039 mitigates that damage.

Section snippets

Cell culture and irradiation in vitro

All studies involving human placental cells were approved by the Institutional Review Board at the University of Pittsburgh. For control, we used the immortalized choriocarcinoma line BeWo (ATCC, Manassas, VA), which captures aspects of trophoblast biology but maintains its undifferentiated state and proliferative capacity [18], and NCCIT cells (ATCC, Manassas, VA), a mixed germ cell tumor line that is particularly radiosensitive [19]. Term PHT cells were isolated and cultured using a modified

The effect of cell irradiation on PHT cells in vitro

We first examined the effect of in vitro irradiation on PHT cell number. We measured total protein as a surrogate for cell number, because PHT cells do not divide in vitro, but differentiate into syncytia. As shown in Fig. 1A, radiation had no effect on protein content in PHT cells, determined 24 h after exposure. In contrast, radiation markedly reduced protein content in the BeWo placental line, as well as in the radiosensitive NCCIT cells, both capable of proliferation in vitro. We obtained

Discussion

Our data indicate that, while ionizing radiation affects human trophoblasts in vitro and mouse fetal growth in vivo, the impact of radiation on placental gene expression is relatively modest and might not account for the full effect of radiation on fetal growth.

In vitro, irradiation of PHT cells led to reduced medium β-hCG levels and gene expression, and enhanced apoptosis. The extent of global gene expression was overall small. Among gene products that exhibited a significant change in vitro,

Acknowledgments

This work was supported in part by an NIH grant, U19 AI068021, to JSG; NIH grants R01HD045675 and R01ES011597 to YS; and Pennsylvania Department of Health Research Formula Funds to YS. The authors thank Judy Ziegler and Elena Sadovsky for technical assistance and Lori Rideout and Bruce Campbell for assistance in manuscript preparation.

References (49)

  • F.A. Mettler et al.

    Major radiation exposure–what to expect and how to respond

    N Engl J Med

    (2002)
  • Y. Gu et al.

    The embryonic and fetal effects in ICR mice irradiated in the various stages of the preimplantation period

    Radiat Res

    (1997)
  • M. De Santis et al.

    Radiation effects on development

    Birth Defects Res C Embryo Today

    (2007)
  • P.U. Devi et al.

    Effect of early fetal irradiation on the postnatal development of mouse

    Teratology

    (2001)
  • E.K. Howell et al.

    Radioadaptive response following in utero low-dose irradiation

    Radiat Res

    (2013)
  • A. Petrova et al.

    Morbidity in a large cohort study of children born to mothers exposed to radiation from chernobyl

    Stem Cells

    (1997)
  • P. Jeggo et al.

    Cellular radiosensitivity: how much better do we understand it?

    Int J Radiat Biol

    (2009)
  • A.R. Snyder et al.

    Differential induction and activation of NF-kappaB transcription complexes in radiation-induced chromosomally unstable cell lines

    Environ Mol Mutagen

    (2005)
  • F. Banaz-Yasar et al.

    Cell line specific modulation of connexin43 expression after exposure to ionizing radiation

    Cell Commun Adhes

    (2005)
  • L.A. Macmillan-Crow et al.

    Invited review: manganese superoxide dismutase in disease

    Free Radic Res

    (2001)
  • J. Ji et al.

    Lipidomics identifies cardiolipin oxidation as a mitochondrial target for redox therapy of brain injury

    Nat Neurosci

    (2012)
  • H. Kim et al.

    Amelioration of radiation esophagitis by orally administered p53/Mdm2/Mdm4 inhibitor (BEB55) or GS-nitroxide

    In Vivo

    (2011)
  • J.S. Greenberger et al.

    Strategies for discovery of small molecule radiation protectors and radiation mitigators

    Front Oncol

    (2011)
  • B. Huppertz et al.
  • Cited by (7)

    • Placental pathology in cancer during pregnancy and after cancer treatment exposure

      2021, Placenta
      Citation Excerpt :

      In vitro studies and animal studies are the only option to study direct irradiation effects on the placenta. Primary (term) human trophoblast cells were exposed to a single dose of 10 Gy radiation [40]. Cyclin-dependent kinase inhibitor 1A (CDKN1A), a gene known to be involved in the p53-dependent cell cycle arrest, was highly upregulated compared to the control group.

    • Effects of ionizing radiation and HLY78 on the zebrafish embryonic developmental toxicity

      2019, Toxicology
      Citation Excerpt :

      Previous studies or reports have shown that the main adverse effects of IR during embryonic development include embryonic death, teratogenesis, growth restriction, decreased body length and weight, and neurodevelopmental deficits (De Santis et al., 2007; Devi and Hossain, 2001; Gu et al., 1997). IR induces tissue damage through various mechanisms, including DNA damage, p53-dependent and -independent apoptosis, and mitochondrial damage (Kanter et al., 2014). Significant developmental effects, such as an increase in the rates of malformations and apoptosis, have also been observed after IR in our previous studies (Si et al., 2013, 2017; Zhou et al., 2014, 2015).

    • Other Maternal Disorders Complicating Pregnancy

      2021, Benirschke’s Pathology of the Human Placenta: Seventh Edition
    • Pregnancy and Cancer: the INCIP Project

      2020, Current Oncology Reports
    View all citing articles on Scopus
    View full text