You are on page 1of 8

Life Sciences in Space Research 31 (2021) 43–50

Contents lists available at ScienceDirect

Life Sciences in Space Research


journal homepage: www.elsevier.com/locate/lssr

Mitigation of late cardiovascular effects of oxygen ion radiation by


γ-tocotrienol in a mouse model
Ashley S. Nemec-Bakk a, *, Vijayalakshmi Sridharan a, Reid D. Landes b, Preeti Singh a,
Maohua Cao c, John W. Seawright d, Xingui Liu e, Guangrong Zheng f, Paari Dominic g,
Rupak Pathak a, Marjan Boerma a
a
Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
b
Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
c
College of Dentistry, Texas A&M University, Dallas TX, USA
d
McLennan Community College, Waco, TX, USA
e
Department of Pharmacodynamics, University of Florida, Gainesville, FL, USA
f
Department of Medicinal Chemistry, University of Florida, Gainesville, FL, USA
g
Department of Medicine and Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA

A R T I C L E I N F O A B S T R A C T

Key words: Purpose: While there is concern about degenerative tissue effects of exposure to space radiation during deep-
High-LET radiation space missions, there are no pharmacological countermeasures against these adverse effects. γ-Tocotrienol
Cardiovascular system (GT3) is a natural form of vitamin E that has anti-oxidant properties, modifies cholesterol metabolism, and has
Degenerative tissue effects
anti-inflammatory and endothelial cell protective properties. The purpose of this study was to test whether GT3
γ-Tocotrienol
could mitigate cardiovascular effects of oxygen ion (16O) irradiation in a mouse model. Materials and methods:
Radiation countermeasure
Male C57BL/6 J mice were exposed to whole-body 16O (600 MeV/n) irradiation (0.26–0.33 Gy/min) at doses of
0 or 0.25 Gy at 6 months of age and were followed up to 9 months after irradiation. Animals were administered
GT3 (50 mg/kg/day s.c.) or vehicle, on Monday – Friday starting on day 3 after irradiation for a total of 16
administrations. Ultrasonography was used to measure in vivo cardiac function and blood flow parameters.
Cardiac tissue remodeling and inflammatory infiltration were assessed with histology and immunoblot analysis
at 2 weeks, 3 and 9 months after radiation. Results: GT3 mitigated the effects of 16O radiation on cardiac
function, the expression of a collagen type III peptide, and markers of mast cells, T-cells and monocytes/mac­
rophages in the left ventricle. Conclusions: GT3 may be a potential countermeasure against late degenerative
tissue effects of high-linear energy transfer radiation in the heart.

1. Introduction (LET) radiation exposures of human populations described above,


research is needed to identify the risk of cardiovascular disease from
Studies of Japanese atomic bomb survivors (Preston et al., 2003; exposure to space radiation.
Ozasa et al., 2016) and other human populations exposed to low doses of Currently, there is minimal opportunity to obtain data on biological
ionizing radiation to a large part of the body (Little et al., 2012; Simo­ effects of HZE particles from human subjects, which makes the deter­
netto et al., 2014; Carr et al., 2005) have shown an increased rate in mination of the biological response almost exclusively dependent on
cardiovascular disease. This has raised the concern about potential animal models (Sridharan et al., 2020; Garikipati et al., 2021; Sasi et al.,
degenerative tissue effects in the cardiovascular system due to low-dose 2017; S.S. Ramadan et al., 2016; Koturbash et al., 2016; Miousse et al.,
exposure to ionizing radiation during deep space travel (Chancellor 2019). Studies in rodent models of HZE radiation exposure have shown
et al., 2014; Hellweg and Baumstark-Khan, 2007; NCRP 2006). Since the alterations in cardiac function and cardiac tissue remodeling (X. Yan
biological properties of the high-Z high-energy (HZE) particle radiation et al., 2014; X. Yan et al., 2014), indicators of inflammation in the heart
in deep space are different from those of the low-linear energy transfer (Tungjai et al., 2013) and vascular stiffness and endothelial dysfunction

* Corresponding author at: 4301 West Markham Slot 522-10, Little Rock, AR 72205, USA.
E-mail address: anemecbakk@uams.edu (A.S. Nemec-Bakk).

https://doi.org/10.1016/j.lssr.2021.07.006
Received 13 May 2021; Received in revised form 2 July 2021; Accepted 29 July 2021
Available online 3 August 2021
2214-5524/© 2021 The Committee on Space Research (COSPAR). Published by Elsevier B.V. All rights reserved.
A.S. Nemec-Bakk et al. Life Sciences in Space Research 31 (2021) 43–50

(Soucy et al., 2011). radiation, 10 mice at the 3 months-time point, and 15 mice at 9 months.
Since astronauts in deep space will spend most of their time inside a
space craft, it is important that research models for the assessment of 2.2. GT3 administration and plasma measurement
health effects of deep space travel mimic the radiation environment that
may be found inside the space craft as opposed to free space. Frag­ GT3 was separated from DeltaGold® using flash column with silica
mentation of heavy ions in the wall of a space craft will lead to a radi­ gel (230 – 400 mesh) as the stationary phase. Gradient elution was
ation environment inside the craft that, for a large part, consists of ions performed with ethyl acetate and hexanes (20/1 to 10/1). Purity of GT3
of Z ≤ 10 (Walker et al., 2013). Therefore, we previously performed a (> 95%) was determined with gas chromatography- and liquid
study in which adult male C57BL/6J mice were exposed to protons (150 chromatography-mass spectrometry.
MeV) or oxygen ions (16O, 600 MeV/n) (Seawright et al., 2019). We Subcutaneous administration of GT3 (50 mg/kg bodyweight once a
found that 16O at doses from 0.05 to 1 Gy caused small changes in day) or vehicle (saline, with 5% Tween 80) started on day 3 after irra­
cardiac function and induced the expression of left ventricular collagen diation (a Friday) and continued on Monday – Friday for a total of 16
type III and left ventricular protein levels of mast cell tryptase, CD2 administrations. Plasma samples of mice sacrificed 2 weeks after irra­
(marker of T-cells) and CD68 (monocytes/macrophages). In the current diation, as described under the heading Blood and tissue collection, were
project, we used the same mouse model to test whether a radiation used to assess plasma concentration of GT3 using high-pressure liquid
countermeasure may mitigate these effects of 16O exposure. chromatography (HPLC).
γ-Tocotrienol (GT3) is a natural form of vitamin E that has long been
known for its properties as protector against the acute radiation syn­ 2.3. High-resolution ultrasonography
drome from exposure to low-LET radiation (Berbee et al., 2009; Singh
et al., 2011; Ghosh et al., 2009). While GT3 has anti-oxidant properties Ultrasonography was performed at 3, 5, 7 and 9 months after irra­
similar to those of the most common form of vitamin E, α-tocopherol diation (n = 15 mice per group) by one investigator who was blinded to
(Yoshida et al., 2003; Suarna et al., 1993), GT3 has additional benefits. It the treatment groups. The day before ultrasonography, mice were
is a potent inhibitor of 3‑hydroxy-3-methyl-glutaryl-coenzyme A anesthetized with 2% isoflurane, and hair was removed from the thorax
(HMG-CoA) reductase, the rate-limiting enzyme in cholesterol biosyn­ and abdomen using a depilatory cream. On the day of ultrasonography,
thesis (Parker et al., 1993; Song and Bose-Boyd, 2006). Moreover, GT3 mice were anesthetized with 1.5 – 2% isoflurane and placed supine on a
accumulates in endothelial cells to at least 30-fold higher levels than heated platform that monitors respiration rate and ECG. The mice were
α-tocopherol and modulates larger numbers of genes in these cells scanned with a Vevo® 2100 imaging system (VisualSonics, Inc.) with a
(Berbee et al., 2012; Naito et al., 2005). We have previously shown that MS400 (18–38 MHz) transducer. Echocardiographs were obtained in the
GT3 exerts its radiation protection at least in part by inhibiting the short axis M-mode at the mid-left ventricular level.
cholesterol synthesis pathway (Berbee et al., 2009) and by reducing Pulsed-wave Doppler was used to determine abdominal aorta blood
endothelial dysfunction (Pathak et al., 2015). Altogether, we hypothe­ flow as an indicator of both cardiac and vascular function. For this
size that GT3 has beneficial effects in the cardiovascular response to purpose, the probe was placed in the transverse axis, immediately
ionizing radiation and tested whether administration of GT3 would alter anterior of the renal artery branch point.
the effects of a single dose of 16O (600 MeV/n, 0.25 Gy) on cardiac The Vevo® LAB cardiac software package was used to obtain ultra­
function and structure in male C57BL/6J mice. sonography parameters from three M-mode scans per animal, and the
vascular package was used to assess blood flow parameters from three
2. Materials and methods consecutive beats per abdominal aorta Pulsed-wave Doppler scan, in up
to three scans per animal. A description of all parameters obtained from
2.1. Animal housing and radiation ultrasonography is provided in Supplemental Materials, Table S1.

All animal procedures within this study were approved by the 2.4. Blood and tissue collection
Institutional Animal Care and Use Committees of the University of
Arkansas for Medical Sciences (UAMS) and Brookhaven National Lab­ Two weeks, 3 months, and 9 months after irradiation, mice were
oratory (BNL). Male C57BL/6J mice (n = 140 total; Jackson Laboratory; weighed in preparation for blood and tissue collection. Calipers were
Bar Harbor, ME) were housed at UAMS, 5 per cage on a 12:12 hour light used to measure tibia length as an additional indicator of body size. Mice
: dark cycle and were administered standard rodent chow low in soy were anesthetized with 3% isoflurane, and a modified infusion set (27 G
(2020X, Harlan Laboratories) and water ad libitum. The cages were with shortened tubing) was used to inject a single dose of heparin
randomly divided into 4 experimental groups: 16O or sham-irradiation, (30− 40 U/kg) into the abdominal vena cava. Without changing the
each with vehicle or GT3 administration after irradiation (n = 35 mice position of the infusion set, a blood sample was drawn and transferred
per group). into an ethylenediaminetetraacetic acid (EDTA) coated tube. Immedi­
At 6 months of age, all mice were transported to Brookhaven Na­ ately after blood collection, the heart was collected, rinsed, weighed and
tional Laboratory (BNL; Upton, NY) by overnight air and housed on a cut longitudinally. One half of the heart was fixed in 5% formalin for 24
12:12 hour light:dark cycle, 2020X diet and water ad libitum. After 1 h and embedded in paraffin. For the other half, a specimen of left
week acclimatization to BNL, mice were exposed to whole-body 16O ventricle was processed as described under the heading Electron micro­
irradiation at the NASA Space Radiation Laboratory (NSRL). Since in a scopy. The remainder of the heart was divided into atria, right ventricle,
previous study we had tested the effects of 16O in a dose range of 0.05 – and 4 samples of left ventricle and snap-frozen in liquid nitrogen.
1 Gy (Seawright et al., 2019), we selected 0.25 Gy as an intermediate
dose for the current study. For this purpose, mice were individually 2.5. Blood chemistry values
positioned in a clear Lucite cube, placed within the NSRL beam line and
were exposed to 16O (600 MeV/n) at a dose rate of 0.26 − 0.33 Gy/min. Blood samples collected at 9 months were used to measure blood
Radiation dosimetry was performed by the NSRL physics team. chemistry values with an i-STAT blood analyzer and CHEM8 + car­
Sham-irradiated mice were also transported to NSRL and placed in the tridges (Abbott Labs, Lake Bluff, IL), and blood glucose levels were
clear Lucite cubes for about 1 min (the same time as the irradiated mice), determined by using FreeStyle Precision Neo blood glucose monitoring
but were not exposed to 16O. Two days after irradiation or system and test strips (Abbott Laboratories, Lake Bluff, IL). Values were
sham-treatment, mice were returned to UAMS. Within each experi­ determined for blood anion gap (ANGAP), blood concentration of urea
mental group, 10 mice were scheduled for sacrifice at 2 weeks after nitrogen (BUN), Cl, blood concentration of creatine (CREA), glucose,

44
A.S. Nemec-Bakk et al. Life Sciences in Space Research 31 (2021) 43–50

hemoglobin (HB), hematocrit (HCT), K, Na, concentration of free 2.10. Statistical analyses
ionized calcium (ICA), and blood concentration of carbon dioxide
(TCO2). Ultrasonography measured 18 heart function parameters from M-
mode and Pulsed-wave Doppler recordings of the heart and 4 parameters
2.6. Histology from pulsed-wave Doppler of the abdominal aorta, as listed in Supple­
mentary Materials, Table S1. The measures of each these parameters
For the assessment of cardiac collagen deposition, 5 µm longitudinal at a given time point were fitted with an analysis of covariance
sections of heart were rehydrated and incubated with Sirius Red sup­ comprised of radiation (0 or 0.25 Gy), GT3 (GT3 or vehicle), and their
plemented with Fast Green. Sections were scanned with a ScanScope interaction as factors. Bodyweight at euthanasia and heart rate during
CS2 slide scanner and analyzed with ImageScope 12 software (Leica ultrasonography recording were included as covariates. Others have
Biosystems, Wetzlar, Germany). The relative tissue area of collagens was previously seen that measures of ultrasonography parameters may
calculated as the red-stained area expressed as a percentage of the total depend on heart rate (Wu et al., 2010) and bodyweight (Pelosi et al.,
tissue area of each section. 2013). For each parameter, we evaluated whether the assumptions of
normal errors and equal variances among independent groups were
reasonable. Normal assumptions were reasonable for all parameters.
2.7. Immunohistochemistry
However, Levene’s test for homogeneous variance indicated that for
several parameters, variances were not equal among the groups. We thus
Immunohistochemistry was used to identify cluster of differentiation
allowed the variances among independent groups to differ for those
(CD) 45-positive cells. Left ventricle sections were rehydrated and an­
parameters having heterogeneous variances. Error degrees of freedom
tigen retrieval was performed by incubating the sections in Target
were estimated with Kenward-Roger’s method (Kenward and Roger,
Retrieval buffer (DAKO, Glostrup, Denmark) at 120 ◦ C in a decloaking
2009). Comparisons of groups were conducted with t-tests within the
chamber (Biocare Medical, Pacheco, CA) for 20 min, cooling for 45 min
analysis of covariance framework; the group means were computed at
in the retrieval buffer, and then were rinsed in distilled water. Imme­
the mean values of heart rate and bodyweight. Analyses of all other
diately after, sections were then incubated in 1% H2O2 in methanol to
parameters were the same as for the analysis of covariance described
block endogenous peroxidase, followed by 10% normal serum in 3% dry
above, except the covariates of heart rate and bodyweight were not
milk powder and 0.2% bovine serum albumin. Sections were incubated
included. All tests were conducted at a 0.05 significance level.
overnight with rat anti-CD45 (Santa Cruz Biotechnology, Santa Crus,
Regarding multiple comparisons, rather than adjusting the significance
CA), followed by biotinylated goat anti-rat IgG (Bio-Rad, Hercules, CA)
level, the positive False Discovery Rate (pFDR) was computed for all
and an avidin-biotin-peroxidase complex (Vector Laboratories, Burlin­
significant results found (Storey, 2002). The ANCOVAs and ANOVAs
game, CA) for 45 min. Antibodies were visualized with 0.5 mg/mL 3, 3′ -
were conducted with the MIXED procedure in SAS/STAT software,
diaminobenzidine (Sigma-Aldrich) and counterstained with hematoxy­
version 9.4 (SAS System for Windows, SAS Institute, Inc.). The pFDR was
lin. Stained sections were examined with an Axioskop transmitted-light
computed in R version 3.6.2 with custom code. Statistical code and data
microscope (Carl Zeiss), and CD45-positive cells were counted in 10
for analyses reported herein is available on request.
optical areas per section. Cells were counted by an individual who was
When reporting summary statistics (i.e., means ± standard deviation
aware of the animal identification numbers but not the treatment
(SD)), the reported means and SDs were estimated from the statistical
groups. The number of CD-positive cells were divided by the total area of
models since these are the values on which inferences are based. Pa­
the 10 views.
rameters for which the assumption of homogeneous variance across
groups was reasonable, the pooled SD is reported; otherwise the SDs
2.8. Electron microscopy were estimated (within the model) for each group.
Sample size considerations. In general, the planned sample sizes for
Tissue specimens of left ventricle were fixed and processed for each of the four groups defined by the combinations of radiation dose
electron microscopy using a method described by Cocchiaro et al. and GT3 administration at a specific post-irradiation time point were n
(Cocchiaro et al., 2008). Sections were analyzed with a Tecnai F20 200 = 6, 10, and 15, depending on the parameter under investigation. With
keV electron microscope (FEI Company, Hillsboro OR). these sample sizes, main effects of size 1.20 SD (n = 6), 0.92 SD (n = 10),
and 0.74 SD (n = 15) were detectable with 0.80 power on a 0.05 sig­
2.9. Immunoblot analysis nificance level test. With the same assumptions, simple effects of size
1.71 SD (n = 6), 1.29 SD (n = 10), and 1.05 SD (n = 15) were detectable,
To determine protein expression indicative of cardiac remodeling and interaction effects of size 2.41 SD (n = 6), 1.84 SD (n = 10), and 1.48
and inflammatory infiltration, immunoblot analysis was performed. A SD (n = 15) were detectable.
Potter-Elvehjem mechanical compact stirrer (BDC2002, Caframo Lab
Solutions, Georgian Bluffs, Ontario) was used to homogenize left ven­ 3. Results
tricular samples in a 1% Triton-X100 RIPA buffer containing protease
(1:100, Sigma-Aldrich, St Louis, MO) and phosphatase inhibitors (1:100, 3.1. Animal characteristics
Sigma-Aldrich). Protein concentration was determined using a BCA
protein assay (Bio-Rad Laboratories, Hercules, CA) and 30 µg protein Plasma GT3 concentrations were measured in mice sacrificed at 2
was added to a 2x Laemmli buffer containing β-mercaptoethanol (5%). weeks after irradiation, when GT3 administrations were still ongoing.
Gel electrophoresis was performed, and proteins were transferred to a Plasma GT3 concentrations were 0.47 ± 0.15 µM (n = 5 mice) in the
PVDF membrane. sham-irradiated animals and 0.69 ± 0.16 µM (n = 5 mice) in the irra­
The following antibodies were obtained from Santa Cruz Biotech­ diated animals. These plasma levels were not significantly different.
nology (Santa Cruz, CA) and used to determine protein content: Across all time points examined, there was no evidence of differences
Collagen type III (1:1000), mast cell tryptase (1:20,000), CD2 (T-lym­ in bodyweight among the four groups (Supplemental Materials,
phocytes; 1:3000), and CD-68 (monocytes/macrophages; 1:1000). Im­ Table S2).
munoblots were imaged with an AlphaImager® gel documentation In blood chemistry values measured 9 months after total body 16O
system (ProteinSimple, San Jose, CA), protein bands quantified by exposure increased Cl and decreased ICA, Na, TCO2, Glucose, and BUN
densitometry through use of publically available ImageJ software and compared to sham animals. GT3 mitigated these 16O induced changes in
expressed relative to GAPDH (Santa Cruz Biotechnology, 1:20,000). Cl, ICA, Na, and TCO2. For BUN, levels in GT3-treated mice exposed to

45
A.S. Nemec-Bakk et al. Life Sciences in Space Research 31 (2021) 43–50

16
O radiation were also decreased from their sham irradiated peers. GT3 3.4. Cardiac immune cell infiltration
also demonstrated a significant increase in HB and HCT in irradiated
mice (Supplemental Materials, Table S3). In previous studies with high-dose local X-ray exposure of the heart
in rodent models, we found a correlation between cardiac mast cell
3.2. In vivo cardiac function numbers and collagen deposition (Boerma et al., 2004). Therefore, we
assessed cardiac mast cell numbers as potential indicators of
Ultrasonography provided 18 measures related to cardiac size and radiation-induced tissue remodeling at 9 months after irradiation. There
function (described in Supplementary Materials, Table S4). At the 3 was a significant decrease in mast cell numbers in the radiation group
months’ time point (9 weeks after the completion of GT3 treatment), compared to sham (Fig. 3). On the other hand, left ventricular mast cell
GT3 treated animals exposed to 16O showed an increase in ejection tryptase content was significantly elevated in irradiated vehicle-treated
fraction and fractional shortening, but these increases seemed to animals over sham at 3 and 9 months. A significant interaction was seen
normalize over time (Fig. 1). Exposure to 16O caused small but signifi­ between GT3 and radiation at all 3 time points, indicating that GT3
cant increases in ejection fraction and fractional shortening as measured mitigated the effects of radiation on left ventricular mast cell tryptase
9 months in vehicle-treated animals. These increases may be an indi­ levels (Fig. 3).
cation that the irradiated heart needs to compensate to maintain cardiac We examined CD45 positive cells as a general marker of leukocytes
output or may be an indication of left ventricular tissue remodeling. (Supplementary Materials, Figure S1). In our prior study of male
Treatment with GT3 normalized the effects of radiation on ejection C57BL/6J mice exposed to 0.25 Gy 16O (600 MeV/n), we found
fraction and fractional shortening in irradiated animals. No major consistent increases in markers of immune cells at 3 months after irra­
changes were seen in any of the other cardiac ultrasonography param­ diation (Seawright et al., 2019), therefore, in the current study we
eters (Supplementary Materials, Table S4). immunostained tissue sections for CD45, as a general marker of leuko­
Exposure to 16O and administration of GT3 did not change blood cytes, and counted the number of CD45 positive cells per mm2 tissue at
flow parameters in the abdominal aorta (Supplementary Materials, the 3 months’ time point. While t-tests within the analysis of covariance
Table S5), suggesting that larger vessel function was not altered. showed no significant differences between the four experimental groups,
there was a significant interaction between radiation and GT3 on the
number of CD45 positive cells that pointed to a potential mitigative
3.3. Cardiac remodeling
effect of GT3 (Fig. 4A).
Even though the total number of leukocytes did not differ signifi­
No differences were found in heart weight relative to tibia length to
cantly between groups, we examined individual immune cell types by
correct for body size at any of the post-radiation time points (Supple­
examining left ventricular CD2 and CD68 protein levels (Fig. 4B).
mentary Materials, Table S2). If radiation fibrosis develops in the
Consistent with our prior study (Seawright et al., 2019), CD2 and CD68
heart, it is expected to appear late after radiation exposure. Therefore,
were significantly increased in the 16O group compared to sham at 2
assessment of collagen levels in the heart was performed at 9 months
weeks and 3 months after irradiation. GT3 alone also significantly
after irradiation. There was no significant difference in percentage area
increased CD2 and CD68 at 2 weeks (when GT3 administration was
of cardiac tissue occupied by collagens 9 months after radiation expo­
ongoing) compared to sham-irradiated animals. On the other hand, GT3
sure (Fig. 2).
treatment significantly reduced CD2 and CD68 content compared to
Immunoblot analysis of collagen type III revealed a band of
radiation alone at 3 months and reduced CD68 at 9 months (Fig. 4C–D).
approximately 75 kDa, as we previously observed in the mouse heart
(Seawright et al., 2019; S.S. Ramadan et al., 2016) that appears to be a
truncated peptide of collagen type III. As seen in our prior study (Sea­ 3.5. Cardiac subcellular structure
wright et al., 2019), even though histological staining did not show an
increase in total collagen deposition in the heart, the left ventricular Electron microscopy of hearts at 9 months after irradiation showed
content of the 75 kDa collagen type III peptide in vehicle-treated animals damaged mitochondria with disrupted cristae. This damage was not
was significantly increased in the 16O exposed mice over sham at 2 observed in mice treated with radiation and GT3 (Fig. 5).
weeks and 3 months. The change in left ventricular content of this
peptide may be an indication of ongoing remodeling of collagen type III. 3.6. False discovery rate
A significant interaction between radiation and GT3 at 2 weeks and 3
months indicates that GT3 mitigated the effects of radiation on the 75 For each time point at which an outcome was measured, we made
kDa collagen type III peptide content (Fig. 2). three comparisons: (i) 0 Gy vs 0.25 Gy, (ii) 0 Gy vs 0 Gy+GT3, (iii) (0 Gy

Fig. 1. Ejection fraction and fractional shortening after 16O and GT3. Cardiac ejection fraction (A) and fractional shortening (B) were determined by ultra­
sonography at 3, 5, 7, and 9 months after 16O radiation exposure. n = 11–14/group. a indicates p < 0.05 compared to time matched 0 Gy, b indicates p < 0.05
compared to time matched 0 Gy+GT3, and c indicates p < 0.05 compared to time matched 0.25 Gy. Error bars are model-estimated SDs. Horizontal lines indicate
there was a significant interaction between GT3 and radiation.

46
A.S. Nemec-Bakk et al. Life Sciences in Space Research 31 (2021) 43–50

Fig. 2. Cardiac collagen levels after 16O and


GT3. Histological analysis of cardiac collagen
deposition at 9 months after irradiation (A),
representative immunoblot of left ventricular
contents of collagen type III and GAPDH at 3
months (B), and collagen type III normalized to
GAPDH at 2 weeks, 3 months, and 9 months after
radiation (C). Error bars are model-estimated
SDs. Total n = 131 animals; time points 2
weeks and 3 months had n = 10/group; and
month 9 had n = 11–14/group. a indicates p <
0.05 compared to time matched 0 Gy, b indicates
p < 0.05 compared to time matched 0 Gy+GT3,
and c indicates p < 0.05 compared to time
matched 0.25 Gy. Horizontal lines indicate there
was a significant interaction between GT3 and
radiation.

Fig. 3. Cardiac mast cells after 16O


and GT3. Cardiac mast cell numbers per
tissue area at 9 months after radiation
(A), representative immunoblot of mast
cell tryptase (MCT) and GAPDH at 3
months after radiation (B), and MCT
density normalized to GAPDH at 2
weeks, 3 months, and 9 months (C).
Error bars are model-estimated SDs.
Total n = 71 animals, with n = 5–6/
group at each time point. a indicates p <
0.05 compared to time matched 0 Gy, b
indicates p < 0.05 compared to time
matched 0 Gy+GT3, and c indicates p <
0.05 compared to time matched 0.25
Gy. Horizontal lines indicate there was a
significant interaction between GT3 and
radiation.

– 0.25 Gy) vs (0 Gy+GT3 - 0.25 Gy+GT3). Altogether, we made 336 While some studies have shown a relation between vitamin E intake and
tests; 52 were significant at the 0.05 level. The positive False Discovery increased cancer rate (Klein et al., 2011), the majority of studies in
Rate was 0.285, meaning that about 15 of these 52 significant results humans and animal subjects show that several forms of vitamin E,
may be false discoveries. including GT3 actually have cancer preventative properties (Jiang,
2017; Yang et al., 2020). On the basis of these combined properties of
4. Discussion GT3 and its apparent safety, we decided to test its effects on cardio­
vascular function and structure when administered after exposure to 16O
This study aimed to provide the first evidence to indicate whether as a model of space radiation.
GT3 may be considered as a safe countermeasure against the degener­ Ultrasonography demonstrated small but significant increases in
ative tissue effects of space radiation in the heart. GT3 has long been ejection fraction and fractional shortening in vehicle-treated animals at
known as a potent protector against the acute radiation syndrome when 9 months after 16O exposure. Yan et al. have also reported a small in­
administered before exposure to low-LET radiation (Berbee et al., 2009; crease in ejection fraction in male C57BL/6NT mice from 1 to 10 months
Singh et al., 2011; Ghosh et al., 2009). However, GT3 also has several after a single dose of 0.15 Gy 56Fe (1 GeV/n) (X. Yan et al., 2014). We
properties that are beneficial to the cardiovascular system. GT3 is a hypothesize that this may be an indication that the irradiated heart
potent inhibitor of HMG-CoA reductase, the rate-limiting enzyme in needs to compensate to maintain cardiac output or a consequence of
cholesterol biosynthesis. Interestingly, in contrast to statins, which are cardiac remodeling that is not yet fully understood. Treatment with GT3
direct inhibitors of HMG-CoA reductase, GT3 reduces the levels of the mitigated the effects of radiation on ejection fraction and fractional
enzyme by stimulating its intracellular degradation (Parker et al., 1993; shortening at 9 months.
Song and Bose-Boyd, 2006). Intermediates of the cholesterol biosyn­ The normal heart contains low numbers of mast cells (Hellstrom and
thesis pathway are used for post-translational modification of Rho pro­ Holmgren, 1950; Constantinides and Rutherdale, 1957). Mast cells
teins that are involved in endothelial cell functions such as stress fiber produce a large variety of pro- and anti-fibrogenic mediators that
formation, expression of nitric oxide synthase, and production of cyto­ regulate tissue remodeling. In our previous studies of localized cardiac
kines and growth factors (Jaffe and Hall, 2005). Therefore, inhibition of x-ray exposure, radiation-induced pathological changes such as degen­
cholesterol biosynthesis has indirect benefits to endothelial function. eration and fibrosis are closely correlated with increased mast cell

47
A.S. Nemec-Bakk et al. Life Sciences in Space Research 31 (2021) 43–50

Fig. 4. Left ventricular protein levels of immune cell markers after 16O and GT3. Immunohistochemistry results for CD45 positive cells at 3 months after
radiation exposure (A), representative immunoblots for left ventricular CD2 and CD68 at 3 months after radiation (B), Immunoblot analysis of CD2 (C) and CD68 (D)
normalized to GAPDH at 2 weeks, 3 months, and 9 months after radiation. Error bars are model-estimated SDs. Total n = 71 animals, with n = 5–6/group at each time
point. a indicates p < 0.05 compared to time matched 0 Gy, b indicates p < 0.05 compared to time matched 0 Gy+GT3, and c indicates p < 0.05 compared to time
matched 0.25 Gy. Horizontal lines indicate there was a significant interaction between GT3 and radiation.

numbers (Boerma et al., 2004). In the current study, mast cell numbers
as determined from histological staining were significantly decreased
after radiation exposure in vehicle-treated animals. It is possible that
whole body irradiation has an effect on overall mast cell numbers or
their ability to migrate into peripheral tissues. Nonetheless, an increase
in left ventricular protein content of mast cell tryptase was seen in
irradiated animals at the 3 and 9 month time points. These results may
suggest that existing mast cells in the heart show increased activity in
response to 16O irradiation as part of a subclinical event, rather than an
actual accumulation of mast cells in the heart. In addition to normalizing
the radiation-induced increases in ejection fraction and fractional
shortening, GT3 administration mitigated the radiation-induced in­
creases in left ventricular mast cell tryptase, suggesting its potential to
alter adverse tissue remodeling.
To further assess tissue remodeling, we examined collagens in the
heart. In vehicle-treated animals exposed to 0.25 Gy 16O, at 2 weeks and
3 months we found an increase in left ventricular content of a 75 kDa
cleavage product of collagen type III, one of the most abundant collagens
in the heart. On the other hand, there was no change in total collagen
deposition as measured with histology. These results are in accordance
with our prior findings of collagen type III at 2 weeks and 3 months and
total collagens at 9 months in male C57BL/6J mice exposed to 0.25 Gy of
16
O (600 MeV/n) (Seawright et al., 2019). Together, these results may
Fig. 5. Electron microscopy analysis of cardiac tissue after 16O and GT3.
Tissue specimens were examined at 9 months after radiation. Mitochondria in indicate that while radiation fibrosis does not occur, there is active
irradiated vehicle treated mice showed disrupted cristae. Magnification: 9600 remodeling of extracellular matrix in the heart after exposure to 16O
×; scale bar = 1000 nm. (600 MeV/n). In contrast, in a prior study, Yan et al. reported an increase
in cardiac collagen deposition at 10 months after exposure to protons (1
GeV, 0.5 Gy) or iron ions (1 GeV/n, 0.15 Gy) in adult male C57BL/6N

48
A.S. Nemec-Bakk et al. Life Sciences in Space Research 31 (2021) 43–50

mice (X. Yan et al., 2014). Differences may be due to the use of a HZE particles during space travel are many fold lower (Cucinotta et al.,
different mouse strain or different ions. Nonetheless, in the current 2003). Therefore, if GT3 is further developed as radiation countermea­
study, the effects of 16O on left ventricular contents of 75 kDa collagen sure for space travel, it should be tested in animal models of chronic or
type III in GT3-treated animals was significantly lower than those in protracted exposures to HZE radiation. Also, while the current study
vehicle-treated animals at 2 weeks and 3 months, again suggesting the focused on male C57BL/6J mice, a widely used animal model for radi­
potential of GT3 to mitigate cardiac tissue remodeling from space ation studies in the cardiovascular system (S.S. Ramadan et al., 2016;
radiation. Patties et al., 2014; Ghosh et al., 2016), there may be sex differences in
In addition to mast cell tryptase, we examined left ventricular pro­ the risk for cardiovascular disease, and therefore future studies will be
tein content of CD2, and CD68, markers of T-lymphocytes and mono­ required in female animals.
cytes/macrophages, respectively. CD45 (leukocytes) cell numbers were In conclusion, results of this study provide some evidence of mild
also assessed but there was no change seen in this marker. CD2 and changes in cardiac function and remodeling after whole body exposure
CD68 immune cell markers were increased at 2 weeks and 3 months to a low dose of 16O (600 MeV/n) in male C57BL/6J mice, and some
after radiation in vehicle-treated animals, but were normalized by GT3 indication that administration of GT3 for 3 weeks after radiation miti­
treatment. Previous studies have shown an increase in the number of gates these effects. If GT3 is considered as one of the radiation coun­
CD68 positive cells at 2 and 4 weeks after 56Fe (0.15 Gy, 1 GeV/n) in termeasures to be developed further for space travel, its effects should be
mouse models (X. Yan et al., 2014; Coleman et al., 2015). Moreover, tested in female animals and in animals chronically exposed to simu­
Tungjai et al. (Tungjai et al., 2013) showed an increase in inflammatory lated GCR.
cytokines in the mouse heart in response to 28Si (300 MeV/n). These
results suggest HZE exposure may only affect specific immune cells and Declaration of Competing Interest
not the immune system as a whole. Within irradiated animals, GT3
lowered CD2 levels at 2 weeks and 3 months and CD68 levels at 3 All authors declare no conflict of interest.
months and 9 months. Studies have previously shown tocopherols and
tocotrienols can directly regulate signaling pathways in macrophages Acknowledgements
(Shen et al., 2018) and gene expression in T-lymphocytes (Zingg et al.,
2013). However, the effects seen in the current study may also be due to This work supported by the National Space Biomedical Research
an indirect regulation of immune cell function by GT3. Institute under grant RE03701 through NCC 9–58, NASA grants
Since GT3 is known to protect mitochondria from radiation expo­ 80NSSC17K0425 and 80NSSC19K0437, and by the National Institute of
sure, cardiac mitochondria were imaged using electron microscopy. General Medical Sciences under grant P20 GM109005. The authors wish
Previous studies have demonstrated beneficial effects of tocotrienols on to thank the BNL support group, the NSRL physicists, the UAMS
the mitochondria where a single dose of tocotrienols prevented mPTP Experimental Pathology Core, and the animal care staff at UAMS and
opening alterations in mitochondrial respiration, and mitochondrial BNL for their excellent technical support.
membrane potential (Nowak et al., 2012; Sridharan et al., 2015). GT3
has also been shown to inhibit apoptosis by preventing cytochrome c Supplementary materials
release from the mitochondria resulting in suppressed caspase -3 and -9
activation (Makpol et al., 2012). In the current study, 16O irradiation Supplementary material associated with this article can be found, in
disrupted mitochondrial cristae but this damage was not seen in the the online version, at doi:10.1016/j.lssr.2021.07.006.
groups treated with GT3. This result suggests GT3 can also prevent
cardiac mitochondrial damage when exposed to HZE radiation which
References
has not been shown before.
Blood chemistry values were obtained at 9 months after 16O exposure Berbee, M., et al., 2009. Gamma-Tocotrienol ameliorates intestinal radiation injury and
to put in context with cardiovascular function and structure. A small reduces vascular oxidative stress after total-body irradiation by an HMG-CoA
reductase-dependent mechanism. Radiat. Res. 171 (5), 596–605.
increase in blood concentrations of Cl and decreases in ICA, Na and
Berbee, M., et al., 2012. Mechanisms underlying the radioprotective properties of
TCO2 may be an indication of reduced kidney function in 16O exposed gamma-tocotrienol: comparative gene expression profiling in tocol-treated
mice. However, kidney dysfunction was not severe enough to elevate endothelial cells. Genes Nutr. 7, 75–81. PMCID: PMC3250525.
BUN levels. In a prior study, exposure of male Long Evans rats to 0.5 Gy Boerma, M., et al., 2004. Histopathology of ventricles, coronary arteries and mast cell
16 accumulation in transverse and longitudinal sections of the rat heart after
O (600 MeV/n) did not alter any of these blood chemistry values at 12 irradiation. Oncol. Rep. 12, 213–219.
months after irradiation (Sridharan et al., 2020), suggesting a species or Carr, Z.A., et al., 2005. Coronary heart disease after radiotherapy for peptic ulcer disease.
strain dependent effect. Tocotrienol administration cause long-term Int. J. Radiat. Oncol. Biol. Phys. 61 (3), 842–850.
Chancellor, J.C., Scott, G.B.I., Sutton, J.P., 2014. Space radiation: the number one risk to
improvements in kidney function in patients with diabetes (Koay astronaut health beyond low earth orbit. Life 4, 491–510.
et al., 2021). In the current study, GT3 mitigated the effects of 16O on Cocchiaro, J.L., et al., 2008. Cytoplasmic lipid droplets are translocated into the lumen of
blood concentrations of Cl, ICA, Na and TCO2, suggesting that GT3 may the Chlamydia trachomatis parasitophorous vacuole. Proc. Natl. Acad. Sci. U S A,
105 (27), 9379–9384.
indirectly affect the cardiovascular system by preserving kidney func­ Coleman, M., et al. Delayed cardiomyocyte response to total body heavy ion particle
tion. In addition, GT3 caused a small increase in HB and HCT only in radiation exposure - identification of regulatory gene networks. 2015.
irradiated animals. In prior animal studies, a single administration of Constantinides, P., Rutherdale, J., 1957. Effects of age and endocrines on the mast cell
counts of the rat myocardium. J. Gerontol. 12 (3), 264–269.
tocotrienols caused an early decrease in HB and HCT that stabilized at
Cucinotta, F.A., et al., 2003. Radiation dosimetry and biophysical models of space
day 7 (Satyamitra et al., 2012) or had no effects on HB or HCT levels radiation effects. Gravit. Space Biol. Bull. 16 (2), 11–18.
(Qureshi et al., 2011). The biological mechanisms behind the apparent Garikipati, V.N.S., et al., 2021. Long-term effects of very low dose particle radiation on
gene expression in the heart: degenerative disease risks. LID. Cells 10 (2), 387.
interaction between GT3 and radiation on HB and HCT in the current
Ghosh, P., et al., 2016. Effects of high-LET radiation exposure and hindlimb unloading on
study are unknown. skeletal muscle resistance artery vasomotor properties and cancellous bone
The high charge and ionization density of 16O are representative of microarchitecture in Mice. Radiat. Res. 185 (3), 257–266.
the relative biological effectiveness of the radiation environment inside Ghosh, S.P., et al., 2009. Gamma-tocotrienol, a tocol antioxidant as a potent
radioprotector. Int. J. Radiat. Biol. 85 (7), 598–606.
a space craft, therefore we selected 16O as the model charged particle in Hellstrom, B., Holmgren, H., 1950. Numerical distribution of mast cells in the human
this and our prior study (Seawright et al., 2019). Although the radiation skin and heart. Acta. Anat. (Basel) 10 (1–2), 81–107.
dose of 0.25 Gy in this study is within the range of total doses experi­ Hellweg, C.E., Baumstark-Khan, C., 2007. Getting ready for the manned mission to Mars:
the astronauts’ risk from space radiation. Naturwissenschaften 94 (7), 517–526.
enced in deep space missions, practical considerations have limited this Jaffe, A.B., Hall, A., 2005. RHO GTPASES: biochemistry and biology. Annu. Rev. Cell
study to administering this dose within minutes, while the dose rates of Dev. Biol. 21, 247–269.

49
A.S. Nemec-Bakk et al. Life Sciences in Space Research 31 (2021) 43–50

Jiang, Q., 2017. Natural forms of vitamin E as effective agents for cancer prevention and Sasi, S.P., et al., 2017. Different sequences of fractionated low-dose proton and single
therapy. Adv. Nutr. 8 (6), 850–867. iron-radiation-induced divergent biological responses in the heart. Radiat. Res. 188
Kenward, M.G., Roger, J.H., 2009. An improved approximation to the precision of fixed (2), 191–203.
effects from restricted maximum likelihood. Comput. Stat. Data Anal. 53 (7), Satyamitra, M., et al., 2012. Mechanism of radioprotection by δ-tocotrienol:
2583–2595. pharmacokinetics, pharmacodynamics and modulation of signalling pathways. Br. J.
Klein, E.A., et al., 2011. Vitamin E and the risk of prostate cancer: the Selenium and Radiol. 85 (1019), e1093–e1103.
vitamin E Cancer Prevention Trial (SELECT). JAMA 306 (14), 1549–1556. Seawright, J.W., et al., 2019. Effects of low-dose oxygen ions and protons on cardiac
Koay, Y.Y., et al., 2021. A phase IIb randomized controlled trial investigating the effects function and structure in male C57BL/6 J mice. Life Sci. Space Res. (Amst) 20
of tocotrienol-rich vitamin E on diabetic kidney disease. Nutrients 13 (1), 258. (2214–5532 (Electronic)), 72–84.
Koturbash, I., et al., 2016. Radiation-induced changes in DNA methylation of repetitive Shen, J., et al., 2018. δ-Tocotrienol, isolated from rice bran, exerts an anti-inflammatory
elements in the mouse heart. Mutat. Res. 787, 43–53. effect via MAPKs and PPARs signaling pathways in lipopolysaccharide-stimulated
Little, M.P., et al., 2012. Systematic review and meta-analysis of circulatory disease from macrophages. Int. J. Mol. Sci. 19 (10).
exposure to low-level ionizing radiation and estimates of potential population Simonetto, C., et al., 2014. Ischemic heart disease in workers at Mayak PA: latency of
mortality risks. Environ. Health Perspect. 120 (11), 1503–1511. incidence risk after radiation exposure. PLoS ONE 9 (5), e96309.
Makpol, S., et al., 2012. Inhibition of mitochondrial cytochrome c release and Singh, P.K., et al., 2011. Radioprotective efficacy of tocopherol succinate is mediated
suppression of caspases by gamma-tocotrienol prevent apoptosis and delay aging in through granulocyte-colony stimulating factor. Cytokine 56 (2), 411–421.
stress-induced premature senescence of skin fibroblasts. Oxid. Med. Cell Longev. Song, B.L., Bose-Boyd, R.A., 2006. Insig-dependent ubiquitination and degradation of 3-
2012, 785743. hydroxy-3-methylglutaryl coenzyme a reductase stimulated by delta- and gamma-
Miousse, I.R., et al., 2019. Changes in one-carbon metabolism and DNA methylation in tocotrienols. J. Biol. Chem. 281 (35), 25054–25061.
the hearts of mice exposed to space environment-relevant doses of oxygen ions Soucy, K.G., et al., 2011. HZE (5)(6)Fe-ion irradiation induces endothelial dysfunction in
(16O). Life Sci. Space Res. (Amst) 22, 8–15. rat aorta: role of xanthine oxidase. Radiat. Res. 176 (4), 474–485.
Naito, Y., et al., 2005. Tocotrienols reduce 25-hydroxycholesterol-induced monocyte- Sridharan, V., et al., 2015. A tocotrienol-enriched formulation protects against radiation-
endothelial cell interaction by inhibiting the surface expression of adhesion induced changes in cardiac mitochondria without modifying late cardiac function or
molecules. Atherosclerosis 180 (1), 19–25. structure. Radiat. Res. 183 (3), 357–366.
NCRP, 2006. Report No. 153, Information Needed to Make Radiation Protection Sridharan, V., et al., 2020. Effects of single-dose protons or oxygen ions on function and
Recommendations for Space Missions Beyond Low-Earth Orbit. National Council on structure of the cardiovascular system in male Long Evans rats. Life. Sci. Space. Res.
Radiation Protection & Measurements. 26, 62–68.
Nowak, G., et al., 2012. γ-Tocotrienol protects against mitochondrial dysfunction and Storey, J.D., 2002. A direct approach to false discovery rates. J. Roy. Stat. Soc. 64 (3),
renal cell death. J. Pharmacol. Exp. Ther. 340 (2), 330–338. 479–498.
Ozasa, K., I. Takahashi, and E.J. Grant, Radiation-related risks of non-cancer outcomes in Suarna, C., et al., 1993. Comparative antioxidant activity of tocotrienols and other
the atomic bomb survivors. Ann. ICRP, 2016. natural lipid-soluble antioxidants in a homogeneous system, and in rat and human
Parker, R.A., et al., 1993. Tocotrienols regulate cholesterol production in mammalian lipoproteins. Biochim. Biophys. Acta. 1166 (2–3), 163–170.
cells by post-transcriptional suppression of 3-hydroxy-3-methylglutaryl-coenzyme A Tungjai, M., Whorton, E.B., Rithidech, K.N., 2013. Persistence of apoptosis and
reductase. J. Biol. Chem. 268 (15), 11230–11238. inflammatory responses in the heart and bone marrow of mice following whole-body
Pathak, R., et al., 2015. Thrombomodulin contributes to gamma tocotrienol-mediated exposure to (2)(8)Silicon ((2)(8)Si) ions. Radiat. Environ. Biophys. 52 (3), 339–350.
lethality protection and hematopoietic cell recovery in irradiated mice. PLoS ONE 10 Walker, S.A., Townsend, L.W., Norbury, J.W., 2013. Heavy ion contributions to organ
(4), e0122511. dose equivalent for the 1977 galactic cosmic ray spectrum. Adv. Space Res. 51,
Patties, I., et al., Late inflammatory and thrombotic changes in irradiated hearts of 1792–1799.
C57BL/6 wild-type and atherosclerosis-prone ApoE-deficient mice. Strahlenther. Wu, J., et al., 2010. Effects of heart rate and anesthetic timing on high-resolution
Onkol., 2014. echocardiographic assessment under isoflurane anesthesia in mice. J. Ultrasound
Pelosi, A., et al., 2013. Cardiac effect of short-term experimental weight gain and loss in Med. 29 (12), 1771–1778.
dogs. Vet. Rec. 172 (6), 153. Yan, X., et al., 2014b. Radiation-associated cardiovascular risks for future deep-space
Preston, D.L., et al., 2003. Studies of mortality of atomic bomb survivors. Report 13: solid missions. J. Radiat. Res. 55 (1), i37–i39. Suppl.
cancer and noncancer disease mortality: 1950-1997. Radiat. Res. 160 (4), 381–407. Yan, X., et al., 2014a. Cardiovascular risks associated with low dose ionizing particle
Qureshi, A.A., et al., 2011. Tocotrienols-induced inhibition of platelet thrombus radiation. PLoS ONE 9 (10), e110269.
formation and platelet aggregation in stenosed canine coronary arteries. Lipids Yang, C.S., et al., 2020. Vitamin E and cancer prevention: studies with different forms of
Health Dis. 10, 58. tocopherols and tocotrienols. Mol. Carcinog. 59 (4), 365–389.
Ramadan, S.S., et al., 2016a. A priming dose of protons alters the early cardiac cellular Yoshida, Y., Niki, E., Noguchi, N., 2003. Comparative study on the action of tocopherols
and molecular response to (56)Fe irradiation. Life Sci. Space Res. (Amst) 8, 8–13. and tocotrienols as antioxidant: chemical and physical effects. Chem. Phys. Lipids
Ramadan, S.S., et al., 2016b. A priming dose of protons alters the early cardiac cellular 123 (1), 63–75.
and molecular response to (56)Fe irradiation. Life. Sci. Space Res. 8, 8–13. Zingg, J.M., et al., 2013. In vivo regulation of gene transcription by alpha- and gamma-
tocopherol in murine T lymphocytes. Arch. Biochem. Biophys. 538 (2), 111–119.

50

You might also like