Changes in one-carbon metabolism and DNA methylation in the hearts of mice exposed to space environment-relevant doses of oxygen ions (16O)

https://doi.org/10.1016/j.lssr.2019.05.003Get rights and content

Highlights

  • Space-relevant doses of radiation lead to long-lasting changes in DNA methylation.

  • Major satellites are particularly affected, at the methylation and expression levels.

  • Transsulfuration of methionine is impacted at low dose.

Abstract

Cardiovascular disease constitutes an important threat to humans after space missions beyond the Earth's magnetosphere. Epigenetic alterations have an important role in the etiology and pathogenesis of cardiovascular disease. Previous research in animal models has shown that protons and 56Fe ions cause long-term changes in DNA methylation and expression of repetitive elements in the heart. However, astronauts will be exposed to a variety of ions, including the smaller fragmented products of heavy ions after they interact with the walls of the space craft. Here, we investigated the effects of 16O on the cardiac methylome and one-carbon metabolism in male C57BL/6 J mice. Left ventricles were examined 14 and 90 days after exposure to space-relevant doses of 0.1, 0.25, or 1 Gy of 16O (600 MeV/n). At 14 days, the two higher radiation doses elicited global DNA hypomethylation in the 5′-UTR of Long Interspersed Nuclear Elements 1 (LINE-1) compared to unirradiated, sham-treated mice, whereas specific LINE-1 elements exhibited hypermethylation at day 90. The pericentromeric major satellites were affected both at the DNA methylation and expression levels at the lowest radiation dose. DNA methylation was elevated, particularly after 90 days, while expression showed first a decrease followed by an increase in transcript abundance. Metabolomics analysis revealed that metabolites involved in homocysteine remethylation, central to DNA methylation, were unaffected by radiation, but the transsulfuration pathway was impacted after 90 days, with a large increase in cystathione levels at the lowest dose. In summary, we observed dynamic changes in the cardiac epigenome and metabolome three months after exposure to a single low dose of oxygen ions.

Introduction

The potential health hazards associated with space radiation are a major obstacle preventing deep space exploration (Chancellor et al., 2014). Beyond the protective magnetosphere of the Earth, astronauts are exposed to radiation from galactic cosmic rays and solar flares. For even the shortest round-trip to Mars, exposure for the crew is estimated to reach 0.66 Sv (or Gy) (Zeitlin et al., 2013). Furthermore, contrary to terrestrial radiation, current technology does not provide effective shielding against the type of high energy particles encountered in space. It is therefore crucial to learn more about the types of long-term biological changes associated with exploration beyond the orbit of the Earth.

Experimental evidence suggests cardiac remodeling, as well as apoptotic and inflammatory responses in the mouse heart as a result of exposure to high-LET radiation (Boerma et al., 2016, Ramadan et al., 2016, Tungjai et al., 2013, Boerma et al., 2015, Hughson et al., 2018). Studies of cancer survivors indicate that cardiac injury due to exposure to ionizing radiation increases linearly with mean dose of radiation to the heart, and progresses very slowly, with onset observed decades after exposure (Darby et al., 2013). Recent studies indicate that epigenetic and metabolic alterations may substantially contribute to pathogenesis of cardiovascular disease (Stenvinkel et al., 2007, Sharma et al., 2008). DNA methylation is one of the key epigenetic mechanisms for the regulation of genetic information. It is responsive to environmental exposures, providing a possible mechanism linking radiation exposure and late-onset injury to the heart.

Methyl groups for DNA methylation are supplied through the methyl donor methionine, with homocysteine as a byproduct. The methionine cycle is critical for normal heart development, and folic acid supplementation during pregnancy is associated with a decreased incidence of congenital heart defects (Feng et al., 2015, Liu et al., 2016). Beyond the development period, an elevated level of homocysteine is an independent risk factor for cardiovascular disease (Perry et al., 1995, Wald et al., 2011), highlighting the importance of the methionine cycle in cardiac health throughout life.

Studies report that radiation-induced changes in DNA methylation stem primarily from the repetitive elements rather than individual genes (Nzabarushimana et al., 2014, Miousse et al., 2017b, Koturbash et al., 2016). Of particular interest are two subtypes of repetitive elements; LINE-1 retrotransposons and satellite DNA. LINE-1 repeats cover about 20% of the murine genome and are heavily methylated within their 5′-untranslated regions (UTRs) (Miousse and Koturbash, 2015). Satellite DNA repeats are characteristic of the pericentromeric regions. It is a less abundant, but still heavily methylated type of repetitive element. DNA methylation is a critical regulator of aberrant transcription for both elements, and DNA hypomethylation is associated with their reactivation and insertional mutagenesis (for LINE-1 elements) and chromosomal aberrations (for satellite DNA) (Miousse and Koturbash, 2015, Miousse et al., 2015b, Koturbash et al., 2007, Miousse et al., 2015a, Qu et al., 1999, Ji et al., 1997). Recent studies demonstrated alterations in DNA methylation during the pathogenesis of a number of heart diseases (Baccarelli et al., 2010, Kim et al., 2010, Gilsbach et al., 2014, Fiorito et al., 2014). DNA hypomethylation and accumulation of satellite DNA transcripts has also been observed in humans diagnosed with heart failure (Haider et al., 2012). Effects of high-LET irradiation on DNA methylation of those two types of repetitive elements in the mouse heart, as well as alterations to one-carbon metabolism, were reported (Koturbash et al., 2016).

In this study, we investigate the short- and long-term (14 and 90 days post exposure, respectively) effects of exposure to one of the under-investigated heavy ions yet relevant for the radiation environment inside a space craft, oxygen (16O), on the mouse cardiac methionine cycle using epigenetic and metabolomic approaches. The most biologically relevant radiation source in free space is 56Fe (Cucinotta et al., 2003), and we previously reported epigenetic changes in the heart associated with this type of radiation (Koturbash et al., 2016). However, interaction with the hull of the spacecraft and space suit materials also creates ions of smaller masses, such as 16O (Walker et al., 2013). Previous results from our group and others indicate a distinctly large cardiovascular toxicity associated with exposure to 16O, which prompted us to take a closer look at the epigenetic signature of 16O in the heart (Ramadan et al., 2016, Yan et al., 2014, Seawright et al., 2019). Levels of radiation exposure have been estimated for different types of charged particles in outer space and on the surface of Mars (Nelson, 2016, Cucinotta et al., 2003, Zeitlin et al., 2013). Several Russian, European and Canadian space agencies have proposed 1 Sv (or 1 Gy) as the astronaut career exposure limit, a dose only slightly above the high estimates for the cumulative exposure associated with a round trip mission to Mars. Based on these estimates, we analyzed doses of 0.1, 0.25, and 1 Gy of 16O radiation (600 MeV/n) in an effort to replicate the most current estimation of exposures associated with a round trip to Mars, but with all of the dose coming from a single particle species.

Section snippets

Animals and whole body irradiation

Four to 8 week-old male C57BL/6J mice purchased from the Jackson Laboratory (Bar Harbor, ME) were housed 5 per cage at the University of Arkansas for Medical Sciences (UAMS). At 6 months of age, mice were shipped to the NASA Space Radiation Laboratory (NSRL) at Brookhaven National Laboratories (BNL) in Upton, NY. Two cages, each with 5 mice, were assigned to one of the 8 groups (n = 10/group) for a total of 80 mice in this study. After a one-week acclimation period, the mice received a single

Repetitive element DNA methylation

To investigate the effect of 16O radiation on the cardiac epigenome, DNA methylation at LINE-1 elements and satellite DNA was first measured. DNA methylation status of 18 most represented LINE-1 elements that evolved in the murine genome during the last 13 million years (Myr) was measured. At the 14 day time point, exposure to two doses of 16O (0.25 and 1 Gy) led to significant global DNA hypomethylation of LINE-1 elements within their 5′-UTRs, independently of evolutionary age (Fig. 1A).

Discussion

The present study investigated the short and long term effects of exposure to space-relevant doses of high energy 16O ions on DNA methylation and methyl group metabolism in the heart. Previous studies indicated changes in DNA methylation, repetitive element repression, and expression of methylation-related genes in this organ caused by exposure to two other types of charged particles, protons and 56Fe (Koturbash et al., 2016). This new study confirms and expands these findings. In addition, the

Authors' contribution

MB, MHJ, IK, and AKC conceptualized the experiment. IRM, CMS, VS, JWS, PS, RDL, and AKC performed investigations and data analysis. IRM, IK, MB, and RDL wrote the original draft and did review and editing.

Declaration of Competing Interest

We wish to confirm that there are no known conflicts of interest associated with this publication and there has been no significant financial support for this work that could have influenced its outcome.

Acknowledgments

We would like to thank Dr. Gregory Nelson for his assistance with the revision of this paper. The study was supported in part by the National Space Biomedical Research Institute through NCC 9–58 (MB), and Centers of Biomedical Research Excellence (NIH) (P20, GM109005, MHJ and IK).

References (51)

  • J.W. Seawright et al.

    Effects of low-dose oxygen ions and protons on cardiac function and structure in male C57BL/6J mice

    Life Sci. Space Res.

    (2019)
  • Y. Tian et al.

    UHRF1 contributes to DNA damage repair as a lesion recognition factor and nuclease scaffold

    Cell Rep.

    (2015)
  • S.A. Walker et al.

    Heavy ion contributions to organ dose equivalent for the 1977 galactic cosmic ray spectrum

    Adv. Space Res.

    (2013)
  • A. Baccarelli et al.

    Ischemic heart disease and stroke in relation to blood DNA methylation

    Epidemiol. Camb. Mass

    (2010)
  • V. Batra et al.

    Modulation of enzymes involved in folate dependent one-carbon metabolism by gamma-radiation stress in mice

    J. Radiat. Res.

    (2004)
  • M. Boerma et al.

    Space radiation and cardiovascular disease risk

    World J. Cardiol.

    (2015)
  • J.C. Chancellor et al.

    Space radiation: the number one risk to astronaut health beyond low earth orbit

    Life

    (2014)
  • J. Chang et al.

    Low doses of oxygen ion irradiation cause acute damage to hematopoietic cells in mice

    PloS One

    (2016)
  • S. Cronin et al.

    Dose-related association of MTHFR 677T allele with risk of ischemic stroke

    Stroke

    (2005)
  • F.A. Cucinotta et al.

    Nuclear interactions in heavy ion transport and event-based risk models

    Radiat. Prot. Dosimetry

    (2011)
  • F.A. Cucinotta et al.

    Radiation dosimetry and biophysical models of space radiation effects

    Gravit. Space Biol. Bull.

    (2003)
  • S.C. Darby et al.

    Risk of ischemic heart disease in women after radiotherapy for breast cancer

    N. Engl. J. Med.

    (2013)
  • Y. Feng et al.

    Maternal folic acid supplementation and the risk of congenital heart defects in offspring: a meta-analysis of epidemiological observational studies

    Sci. Rep.

    (2015)
  • R. Gilsbach et al.

    Dynamic DNA methylation orchestrates cardiomyocyte development, maturation and disease

    Nat. Commun.

    (2014)
  • M.G. Guren et al.

    Biochemical signs of impaired cobalamin status during and after radiotherapy for rectal cancer

    Int. J. Radiat. Oncol.

    (2004)
  • Cited by (13)

    • The glycolytic pathway to heart failure

      2023, Glycolysis: Tissue-Specific Metabolic Regulation in Physio-pathological Conditions
    • Impact of spaceflight stressors on behavior and cognition: A molecular, neurochemical, and neurobiological perspective

      2022, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      While a direct link between space radiation-induced changes in cognition and the microbiome remain difficult to establish, this does represent one potential area for future investigation. Although heavy ion exposure of cardiac tissue has been reported to alter one-carbon metabolism and epigenetics (DNA methylation) with long-term-elevations in cystathione (Miousse et al., 2019), it remains uncertain how such changes might interact with CNS function. Other metabolomic studies analyzing liver following higher doses of low LET radiation (gamma and proton) find changes that implicate multiple biological pathways, but again are difficult to pinpoint to the CNS (Xiao et al., 2017).

    • Mitigation of late cardiovascular effects of oxygen ion radiation by γ-tocotrienol in a mouse model

      2021, Life Sciences in Space Research
      Citation Excerpt :

      Since the biological properties of the high-Z high-energy (HZE) particle radiation in deep space are different from those of the low-linear energy transfer (LET) radiation exposures of human populations described above, research is needed to identify the risk of cardiovascular disease from exposure to space radiation. Currently, there is minimal opportunity to obtain data on biological effects of HZE particles from human subjects, which makes the determination of the biological response almost exclusively dependent on animal models (Sridharan et al., 2020; Garikipati et al., 2021; Sasi et al., 2017; S.S. Ramadan et al., 2016; Koturbash et al., 2016; Miousse et al., 2019). Studies in rodent models of HZE radiation exposure have shown alterations in cardiac function and cardiac tissue remodeling (X. Yan et al., 2014; X. Yan et al., 2014), indicators of inflammation in the heart (Tungjai et al., 2013) and vascular stiffness and endothelial dysfunction (Soucy et al., 2011).

    • 5’UTR methylation in different genes from workers exposed to volatile organic compounds: A new insight for considering an epigenetic mark as a functional correlate

      2020, Toxicology Letters
      Citation Excerpt :

      b) It is highly complex to give an explanation about our results regarding methylation looking for a link with gene expression, since we did not measured protein expression levels, and also, beacuse there are reports claiming that 5′UTR methylation is closely related with gene silencing (Liu et al., 2018), but also there are others linking this methylation status only to translation initiation (Zhou et al., 2015). This becomes more challenging if we analyze recent findings on 5′UTR methylation in animals chronically exposed to Oxygen-16 (Miousse et al., 2019), where it has been observed a global hypomethylation in this region at day 14 of exposure, while they observed hypermethylation at day 90 of exposure. This latter observation rises more questions about the meaning of our findings regarding if they are transitory or even concomitant (higher methylation in promoter and 5′UTR for TNF-α in LS workers), as a result of VOC exposure.

    • Leveraging advances in modern science to revitalize low-dose radiation research in the United States

      2022, Leveraging Advances in Modern Science to Revitalize Low-Dose Radiation Research in the United States
    View all citing articles on Scopus
    View full text