Open Access

X‑irradiation induces acute and early term inflammatory responses in atherosclerosis‑prone ApoE‑/‑ mice and in endothelial cells

  • Authors:
    • Raghda Ramadan
    • Michaël Claessens
    • Ellen Cocquyt
    • Mohamed Mysara
    • Elke Decrock
    • Sarah Baatout
    • An Aerts
    • Luc Leybaert
  • View Affiliations

  • Published online on: March 25, 2021     https://doi.org/10.3892/mmr.2021.12038
  • Article Number: 399
  • Copyright: © Ramadan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Thoracic radiotherapy is an effective treatment for many types of cancer; however it is also associated with an increased risk of developing cardiovascular disease (CVD), appearing mainly ≥10 years after radiation exposure. The present study investigated acute and early term physiological and molecular changes in the cardiovascular system after ionizing radiation exposure. Female and male ApoE‑/‑ mice received a single exposure of low or high dose X‑ray thoracic irradiation (0.1 and 10 Gy). The level of cholesterol and triglycerides, as well as a large panel of inflammatory markers, were analyzed in serum samples obtained at 24 h and 1 month after irradiation. The secretion of inflammatory markers was further verified in vitro in coronary artery and microvascular endothelial cell lines after exposure to low and high dose of ionizing radiation (0.1 and 5 Gy). Local thoracic irradiation of ApoE‑/‑ mice increased serum growth differentiation factor‑15 (GDF‑15) and C‑X‑C motif chemokine ligand 10 (CXCL10) levels in both female and male mice 24 h after high dose irradiation, which were also secreted from coronary artery and microvascular endothelial cells in vitro. Sex‑specific responses were observed for triglyceride and cholesterol levels, and some of the assessed inflammatory markers as detailed below. Male ApoE‑/‑ mice demonstrated elevated intercellular adhesion molecule‑1 and P‑selectin at 24 h, and adiponectin and plasminogen activator inhibitor‑1 at 1 month after irradiation, while female ApoE‑/‑ mice exhibited decreased monocyte chemoattractant protein‑1 and urokinase‑type plasminogen activator receptor at 24 h, and basic fibroblast growth factor 1 month after irradiation. The inflammatory responses were mainly significant following high dose irradiation, but certain markers showed significant changes after low dose exposure. The present study revealed that acute/early inflammatory responses occurred after low and high dose thoracic irradiation. However, further research is required to elucidate early asymptomatic changes in the cardiovascular system post thoracic X‑irradiation and to investigate whether GDF‑15 and CXCL10 could be considered as potential biomarkers for the early detection of CVD risk in thoracic radiotherapy‑treated patients.

Introduction

Adjuvant radiotherapy is an effective treatment for thoracic malignancies and has resulted in a significant improvement in the chances of cancer patient's survival over the past decades (14). However, while high-energy radiotherapy treatment successfully kills cancer cells, unavoidable radiation exposure to the heart and large arteries occurs during treatment, resulting in radiation-induced secondary cardiovascular disease (CVD) in cancer survivors (516). Indeed, epidemiological, clinical and experimental data have established the link between radiation exposure at high and medium doses that are received during radiotherapy (>0.5 Gy) and the risk for CVD (6,9,10,1720). In addition, recent data suggest that radiation-induced CVD occurs at much lower doses than previously thought (<0.5 Gy) (7,10,19,2124). However, the underlying cellular and molecular mechanisms of radiation-induced CVD are not fully understood, possibly resulting in improper radiation protection. Most of the clinical and experimental data focus on identifying the late cardiovascular response after radiation exposure, although physiological changes and cellular and molecular damage in the cardiovascular system may happen directly after radiation exposure (7,18,2532). Moreover, there are currently no validated biomarkers that predict the risk to develop CVD after radiotherapy. Therefore, experimental animal studies of acute and early cardiovascular response after thoracic irradiation may improve our knowledge on early asymptomatic changes in the cardiovascular system after ionizing exposure. This may help in identifying biomarkers indicating developing cardiovascular complications after radiotherapy, which may help in screening patients at risk for developing CVD, thus countermeasures and early medical intervention might be applied to prevent further cardiac toxicity.

Atherosclerosis, a progressive inflammatory disease of the arterial wall, is the main underlying cause of major CVD. Multiple inflammatory markers have been correlated with the pathogenesis of atherosclerosis (3336). Radiation exposure is also known to trigger the release of several inflammatory and adhesion molecules, involved in the pathogenesis of atherosclerosis (18,26,3742). Here, we explored a large panel of inflammatory markers in the serum of atherosclerosis-prone ApoE/ mice at 24 h (acute response) and 1 month (early term response) of thoracic X-ray irradiation. The inflammatory markers were further verified in vitro in coronary artery and microvascular endothelial cell lines exposed to low and high dose X-rays. Serum triglyceride and cholesterol levels, which are known to be strongly associated with atherogenesis (43,44), were also evaluated at 24 h and 1 month post-irradiation. We identified two inflammatory markers, growth differentiation factor-15 (GDF-15) and C-X-C motif chemokine ligand 10 (CXCL10), which were significantly elevated 24 h after irradiation in both female and male mice, which were found to be also increased in irradiated coronary artery and microvascular endothelial cells. Interestingly, we observed gender-specific responses in triglyceride and cholesterol levels 1 month post-irradiation, and in the assessed inflammatory markers at 24 h and 1 month post-irradiation.

Materials and methods

Animals

Animal experiments were approved by the Ethical Committee Animal Studies of the Faculty of Medicine and Health Sciences, Ghent University (ECD 17/60), and were performed in compliance with the Belgian laboratory animal legislation and the European Communities Council Directive of 22 September 2010 (2010/63/EU). Apolipoprotein E-deficient ApoE/) mice were purchased from Charles River Laboratories, and animals were housed at the animal facility of UGent. Pups were weaned at the age of three weeks and housed in temperature-controlled, individually ventilated cages, with a 12 h light-dark cycle. They received a standardized mouse chow diet (3.7% fat) and water ad libitum. At the age of 10–12 weeks, ApoE/ mice were randomly allocated to receive irradiation or sham treatment. In total 72 female and male ApoE/ mice were used (6 mice per group, except for the control group at 1 month, which were 3 for male and 9 for female mice).

Mouse irradiation

The Small Animal Radiation Research Platform (SARRP; Xstrahl®; in collaboration with INFINITY lab, UGent) was used to irradiate ApoE/ mice. Mice were anesthetized with isoflurane (5% induction and 2% maintenance) and subjected to a full body CT-scan (50 kV, 1.5 mA, 360 projections >360°, 1 mm aluminium filter) prior to irradiation. CT images were analyzed by using Muriplan software (Xstrahl®) to determine the coordinates of the isocenter for subsequent irradiation. Thoracic irradiation was performed in a ventro-dorsal direction, with 220-kV X-rays, operating at 13 mA and filtered with 0.15 mm of copper, operating with a dose rate of 3.4 Gy/min. The field size, created by the collimator of 10×10 mm, was encompassing both carotid arteries, the aortic arch and apical portion of the heart. The doses that were delivered to the heart region are 0 Gy (sham treatment; also referred to as ‘controls’), 0.1 and 10 Gy.

Cell culture

Telomerase Immortalized human Microvascular Endothelial cells (TIME) from the American Type Cell Culture (ATCC, France; ATCC® CRL-4025™) (https://www.lgcstandards-atcc.org/Products/All/CRL-4025) was used. In addition, TICAE cells, which are primary human coronary artery endothelial cells from the European Collection of Authenticated Cell Cultures (ECACC; HCAECs cat. no. 300-05a) that were transduced with retroviruses bearing the est2 gene, a yeast homologue of the human TERT protein (45,46), were used. TIME and TICAE cells are from a male human donor. MesoEndo Cell Growth Medium (Sigma-Aldrich; Merck KGaA) was used for TIME and TICAE cells. The passage number that was used in the experiments is 38 for TIME cells and 33 for TICAE cells. Cells were grown at 37°C in a humidified incubator supplemented with 5% CO2 and were split with a 0.05% trypsin supplemented with 0.02% ethylenediaminetetraacetic acid (EDTA) every 3–4 days. Moxi Z Mini Automated Cell Counter (Orflo Technologies) was used to count the cells. For the cytokine detection experiment, TIME and TICAE cells were seeded in 6-well plate at a density of 2.5×105 per well in 6 biological replicates. Three days later, cells reached 100% confluence. The medium was changed before irradiation, and at 24 and 72 h after irradiation the supernatant was collected.

Cell irradiation

Both TIME and TICAE cells were irradiated at 100% confluence. Single X-rays doses [0 Gy (also referred to as ‘control’), 0.1 and 5 Gy] were applied to the cells using a vertical X-ray beam using a Xstrahl RX generator (320 kV, Filtration: 3.8 mm Al and 1 mm Cu, tube current: 12 mA) (Camberley), at a dose rate of 0.5 Gy/min. X-irradiation was performed in accordance to ISO 4037 and under ISO 17025 accreditation of the Laboratory for Nuclear Calibrations (LNK) of the Belgian Nuclear Research Centre (SCK•CEN). Cells were moved to the irradiation facility using a mobile incubator.

Blood sampling

The blood collection from each mouse was done at two different time points: 1) One week before irradiation and 2) a specific time period after radiation exposure (24 h or 1 month). The mice were anesthetized intraperitoneally with xylazine (10 mg/kg) and ketamine (80 mg/kg) using a 30G needle, and anesthetic depth was assessed using the pedal withdrawal reflex. Retro-orbital puncture was performed to take a 100 µl blood sample with a Brand micropipette one week before irradiation, and at the specific time points after irradiation, where the eye was pulled out to collect at least 500 µl blood. Immediately after the second blood sampling, anesthetized mice were euthanized due to exsanguination via transcardial perfusion. To obtain serum, the collected blood was allowed to clot in eppendorf tubes for 1.5 h at room temperature and was subsequently centrifuged at 1,500 g for 15 min at 4°C. The supernatant was removed and centrifuged again at the same conditions to obtain pure serum. The latter was collected in eppendorf tubes and stored at −80°C.

Triglyceride quantification

The Triglyceride Quantification assay kit (Abcam, ab65336) was used according to the manufacturer's instructions. Briefly, a 50 µl triglyceride standard curve was prepared and 1 µl serum was added to 49 µl assay buffer and all samples were performed in duplicate. Subsequently, 2 µl of lipase, which converts the triglycerides to free fatty acids and glycerol, was added to each well and incubated for 20 min at room temperature. Then, 50 µl reaction mix consisting of 46 µl triglyceride assay buffer, 2 µl triglyceride probe and 2 µl triglyceride enzyme mix, was added to each well and incubated at room temperature for 60 min protected from light. The glycerol molecules are oxidized to generate hydrogen peroxide (H2O2), which reacts with the probe to generate fluorescence that was measured at Ex/Em 535/587 nm with a microplate reader (Victor3, 1420 multilabel counter, PerkinElmer).

Cholesterol quantification

The Cholesterol Assay kit (Abcam, ab65390) was used according to the manufacturer's instructions. A 50 µl cholesterol standard curve was prepared. The serum samples were diluted 1/400 in assay buffer and 50 µl of this diluted serum was used. Then, 50 µl of a cholesterol reaction mix, consisting of 44 µl assay buffer, 2 µl cholesterol probe, 2 µl enzyme mix, and 2 µl cholesterol esterase was added to each well for an incubation period of 60 min at 37°C. The cholesterol esterase hydrolyzes cholesteryl esters to free cholesterol. Subsequently, cholesterol oxidase specifically recognizes free cholesterol and produce H2O2 that reacts with the probe to generate fluorescence (Ex/Em=538/587 nm) that was measured with a microplate reader (Victor3, 1420 multilabel counter, PerkinElmer).

Inflammatory cytokine detection

Simultaneous detection of multiple inflammatory markers in mice serum samples (Adiponectin, CRP, CXCL10, Endoglin, FGF-basic, GDF-15, ICAM-1, IL-6, IL-1β, MCP-1, P-selectin, PAI-1, PCSK9, and uPAR) or in cell supernatant samples (GDF-15, CXCL10, ICAM-1, MCP-1, uPAR, PAI-1, P-selectin, FGF-basic and IL-6) was performed using multiplex bead assay (Luminex® MAGPIX Assay, R&D Systems) following manufacturer's instructions. Luminex® MAGPIX technology is based on the use of a mixture of magnetic microspheres that is added to the sample in a pre-coated plate with analyte-specific capture antibodies, followed by adding specific biotinylated detection antibodies and phycoerythrin that binds to the biotinylated antibodies. Magnetic beads are captured and held in a monolayer by a magnet, while the beads are illuminated by two spectrally distinct light-emitting diodes (LEDs), one to identify the analyte and the other to determine amount of analyte based on the magnitude of the phycoerythrin-derived signal. Signals are captured and imaged with a charge-coupled device (CDD) camera (4750). Briefly, standards, serum or supernatant samples, and magnetic microparticles cocktail were incubated into cytokine-specific antibodies in a pre-coated 96-well plate. After 2-h incubation in the dark, the plate was washed, and a biotinylated detection antibody cocktail was added. After applying a second wash to remove the unbound biotinylated antibodies, a streptavidin-phycoerythrin conjugate was added to each well to bind to the biotinylated antibodies. Further, a final wash was performed and the microparticles were detected using the Luminex® MAGPIX Analyzer. Inflammatory markers concentration in the supernatant of TIME and TICAE cells was normalized to cell count performed using IncuCyte ZOOM™ phase contrast imaging with ×10 magnification.

Statistical analysis

Analysis of cholesterol, triglyceride and supernatant inflammatory cytokine data were performed using Kruskal-Wallis test followed by Dunn's test, and the P-value was adjusted using Benjamini-Hochberg procedure to control the False Discovery Rate. Data are presented as mean ± standard error of the mean. GraphPad Prism 5.01 (GraphPad Software Inc.) was used for these statistical analysis. For in vivo serum inflammatory cytokines, the fold change in inflammatory marker levels before and after irradiation was first calculated, followed by statistical analysis. Statistical analysis was done with a Kruskal-Wallis test followed by Dunn's test and the P-value was adjusted using Benjamini-Hochberg procedure, using ggsignif R package. The results are considered significant if the P-value is <0.05. Data are presented as boxplot, ggplot2 R package, showing the median with the lower and upper hinges that correspond to the first and third quartiles (the 25 and 75th percentiles) and upper/lower whisker extends from the hinges to the largest/smallest value no further than 1.5 * IQR from the corresponding upper and lower hinge, respectively. Data beyond the end of the whiskers are called ‘outlying’ points and are plotted individually. In vivo data are represented separately in female ApoE/ mice and male ApoE/ mice, or as the sum of female and male data, which are referred to as combined data.

Results

Thoracic irradiation induces early term alterations in triglyceride and cholesterol levels in ApoE−/− mice
At 24 h post-thoracic irradiation

Thoracic irradiation of female ApoE/ mice with 10 Gy induced a significant decrease in triglyceride and cholesterol levels at 24 h after irradiation, compared to age-matched controls (Fig. 1A and B), (summarized in Table SI). In male ApoE/ mice, thoracic irradiation induced a significant increase in triglyceride level only at 0.1 Gy, but no significant changes in cholesterol level was observed at 24 h after irradiation (Fig. 1A and B). Female and male ApoE/ mice combined data showed no significant changes in triglyceride and cholesterol levels at 24 h after irradiation (Fig. 1A and B).

At 1 month post thoracic irradiation

At 1 month after irradiation, a significant increase in triglyceride levels in female ApoE/ mice was observed at 10 Gy, whereas total cholesterol levels were significantly decreased at 0.1 and 10 Gy, compared to age-matched controls (Fig. 1C and D). In male ApoE/ mice, a significant decrease in triglyceride level was observed in a dose-dependent manner at 1 month after irradiation, but no significant changes in cholesterol level was observed (Fig. 1C and D). Female and male ApoE/ mice combined data showed no significant changes in triglyceride levels, while a significant dose-dependent decrease in cholesterol level at 1 month after irradiation was observed (Fig. 1C and D).

Thoracic irradiation induces acute and early term systemic inflammatory response in ApoE−/− mice

To assess the inflammatory response, fourteen different cytokines, which are involved in the pathogenesis of atherosclerosis (Adiponectin, CRP, CXCL10, Endoglin, FGF-basic, GDF-15, IGMA-1, IL-6, IL-1β, MCP-1, P-selectin, PAI-1, PCSK9, and uPAR), have been assessed in serum samples of female and male ApoE/ mice at 24 h and 1 month after local thoracic X-ray irradiation. Fold changes between the baseline cytokines levels and at 24 h or at 1 month post irradiation cytokine levels were analyzed (summarized in Table SII).

At 24 h post thoracic irradiation

At 24 h post thoracic irradiation, a significant increase in GDF-15 at 0.1 and 10 Gy in female ApoE/ mice and a significant increase at 10 Gy in male ApoE/ mice were observed (Fig. 2A). Female and male ApoE/ mice combined data showed a significant increase in GDF-15 at 10 Gy. A significant increase in CXCL10 was also observed at 10 Gy in female and male ApoE/ mice, as well as in the combined female and male data (Fig. 2B). In addition, a significant increase in ICAM-1 was observed at 10 Gy only in male ApoE/ mice (Fig. 2C). Whereas, thoracic irradiation induced a significant decrease in MCP-1 at 0.1 and 10 Gy only in female ApoE/ mice (Fig. 2D). Moreover, a significant decrease in female uPAR at 10 Gy was observed, and female and male uPAR combined data showed a significant decrease at 0.1 and 10 Gy (Fig. 2E). Next to that, in male ApoE/ mice, there was a significant increase in PCSK9 at 0.1 Gy (Fig. 2F), and an increase in P-selectin at 10 Gy (Fig. 2G). Nonetheless, no significant changes were observed in adiponectin, PAI-1, CRP, endoglin, FGF-basic, IL-1β and IL-6 at 24 h post thoracic irradiation (Fig. S1).

At 1 month post thoracic irradiation

The systemic inflammatory response at 1 month was different from the acute response at 24 h post thoracic irradiation. Adiponectin was significantly increased at 10 Gy post-irradiation only in male ApoE/ mice (Fig. 3A). CXCL10 and IL-6 were significantly decreased at 0.1 Gy in female ApoE/ mice, and in combined female and male data (Fig. 3B and E). FGF-basic level was significantly decreased at 10 Gy in female mice, and in the female and male mice combined data (Fig. 3C). Furthermore, PAI-1 showed a significant increase at 10 Gy in male ApoE/ mice (Fig. 3D). No significant changes were observed in Endoglin, P-selectin, GDF-15, IL-1β, ICAM-1, MCP-1, PCSK-9, and uPAR at 1 month post thoracic irradiation (Fig. S2).

X-irradiation induces inflammatory response in coronary artery and microvascular endothelial cells

To verify whether the observed inflammatory responses in serum cytokines at 24 h and 1 month after irradiation is linked to endothelial cell responses, two endothelial cell lines (TICAE and TIME cells, originating from a male donor) were irradiated at low dose (0.1 Gy) or high dose (5 Gy), with supernatant being collected at 24 h or 72 h after irradiation. The 72 h time point was chosen since it is not possible to keep cells in culture for 1 month post-irradiation without passaging them. In addition, the high dose was chosen to be 5 Gy in vitro, since we previously reported that 5 Gy induced significant apoptotic cell death from 4 h after X-irradiation in TICAE and TIME cells, which increased with time (51), and also considering that we are irradiating a monolayer of endothelial cells (not tissue like the in vivo situation). The cytokines tested were GDF-15, CXCL10, ICAM-1, MCP-1, uPAR, IL-6, PAI-1, P-selectin, and FGF-basic (summarized in Table SIII).

At 24 h post endothelial irradiation

At 24 h after irradiation, GDF-15 and CXCL10 increased significantly at 5 Gy in TICAE and TIME cells (Fig. 4A, B, J and K) which correspond to the increase in these markers in female and male mice at 24 h post irradiation (Fig. 2A and B). A significant increase in ICAM-1 was observed at 5 Gy in TIME cells (Fig. 4L), which may correlate with the increase in serum ICAM-1 in male mice at 10 Gy for the 24 h time point (Fig. 2C). Additionally, MCP-1 level was not changed in response to radiation in TICAE and TIME cells at 24 h post irradiation (Fig. 4D and M), and this was the case in the irradiated male mice at the same time point (Fig. 2D). Moreover, a significant increase in P-selectin was observed at 5 Gy in TICAE and TIME cells at 24 h post irradiation (Fig. 4H and Q), which corresponds to the observed trend of increase in male mice data (Fig. 2G). The levels of uPAR, IL-6 and PAI-1 were significantly increased in TICAE and TIME cells, and FGF-basic increased in TICAE cells at 5 Gy after 24 h of irradiation (Fig. 4E-G, I, N, O and P), while this was not the case in the irradiated female and male mice at the same time point.

At 72 h post endothelial irradiation

At 72 h after irradiation, GDF-15 was still significantly elevated in TICAE and TIME cells at 5 Gy (Fig. 5A and J). CXCL10, ICAM-1, MCP-1, uPAR, IL-6 and FGF-basic were also significantly elevated at 5 Gy in the irradiated TICAE cells (Fig. 5B-E, F and I), while ICAM-1 and MCP-1 significantly decreased in a dose-dependent manner in the irradiated TIME cells at 72 h post irradiation (Fig. 5L and M). Moreover, there was a significant increase in P-selectin at 5 Gy in TICAE cells, and significant increase in PAI-1 at 5 Gy in TICAE and TIME cells (Fig. 5G, H and P).

Discussion

Thoracic cancer radiotherapy significantly increases the risk for developing CVD (6,9,10,52,53). The underlying pathophysiology is complex and the mechanisms are not fully understood (6,7,9,1721). Previous in vivo studies have identified late cardiovascular responses to ionizing radiation exposure, which was mainly manifested by pro-inflammatory responses and accelerated formation of atherosclerotic lesions/plaques 2–15 months after irradiation (17,18,2629,54). Most of these pre-clinical studies were performed in a single gender, with limited data available related to possible gender differences. Here, we investigated acute (24 h) and early term (1 month) cardiovascular responses to ionizing radiation exposure, by assessing triglyceride and total cholesterol levels, and exploring a large panel of inflammatory markers in serum of female and male ApoE/ mice that received low or high doses of local thoracic X-ray irradiation. We report, for the first time, that local thoracic irradiation of ApoE/ mice increases serum GDF-15 and CXCL10 in both female and male mice 24 h after high dose irradiation (10 Gy). GDF-15 and CXCL10 levels were also significantly elevated at 24 h in irradiated coronary artery and microvascular endothelial (TICAE and TIME) cells in vitro after high dose of X-ray irradiation. In addition, we report gender-specific responses in the tested triglyceride and cholesterol levels at 1 month after irradiation, and in the assessed inflammatory markers at 24 h and 1 month post-irradiation. Below we discuss these findings in more detail.

Increased cholesterol and triglyceride levels in atherosclerosis have been shown in several clinical studies (55), and it was suggested that triglyceride level can be used as independent risk factor biomarker for coronary artery disease (56). Increased cholesterol and triglyceride levels may contribute to a large number of biological actions and consequences, including inflicting endothelial cell injury, increasing adhesion molecule expression, increasing leukocytes recruitment, as well as the formation of foam cells, therefore contributing to the atherosclerotic process (43,44). We observed different responses in cholesterol and triglyceride levels in the irradiated female and male ApoE/ mice, especially at 1 month after exposure. Although triglyceride levels were increased in 10 Gy-irradiated female mice, levels dose-dependently decreased in male mice, demonstrating that triglyceride levels display gender specific responses. In contrast, cholesterol levels dose-dependently decreased in female mice, and showed a decreasing trend in male mice, resulting in a significant dose-dependent decrease when combining female and male responses 1 month post-irradiation. A similar observation for the decrease in total cholesterol and the increase in triglyceride levels in female mice was reported in female breast cancer patients after 50–60 Gy of radiotherapy which were given over 5 weeks (57). Previous studies performed 2–5 months after irradiation of ApoE/ mice, revealed variable responses in cholesterol and triglyceride levels depending mainly on the time point after exposure, gender used in the experiment, and on radiation doses/quality used (17,18,27,29). Cholesterol and triglyceride changes can perhaps be secondary to the inflammatory or oxidative stress responses after radiation exposure, though further investigations are needed to unveil the involved mechanisms.

Our results further demonstrate that local thoracic irradiation induced an increase in serum GDF-15, which was significant at low and high doses (0.1 and 10 Gy) in female mice and at high dose in male mice 24 h after irradiation. In vitro, GDF-15 was secreted from irradiated TICAE and TIME cells at 24 h after high dose (5 Gy) irradiation. An increased GDF-15 gene expression was previously observed in human aortic endothelial cells at 4–24 h after 4 Gy (58), and in carotid arteries of male ApoE/ mice at 1 week after 14 Gy of local neck X-ray irradiation (59). GDF-15 is a member of the transforming growth factor β superfamily that increases its expression under inflammatory conditions (60,61). Elevated GDF-15 serum levels have been associated with an increased risk for a range of CVD, including atherosclerosis, and currently being evaluated as a biomarker in CVD (6265). Previous studies revealed that GDF-15 may contribute to the initiation and the progression of atherosclerotic lesions by regulating apoptosis and IL-6-dependent inflammatory responses (66), promoting migration of macrophage, and by contributing to plaque instability (67). In line with this, our in vitro data showed an increased IL-6 level in TICAE and TIME cells 24 h after 5 Gy irradiation. GDF-15 may play an important role in ionizing radiation-induced endothelial cell senescence through an oxidative stress-mediated p16 pathway (58). Another inflammatory marker of potential interest is CXCL10, also known as Interferon-γ-inducible protein 10 (IP-10), which acts as a chemoattractant cytokine. CXCL10 was shown to promote atherosclerosis by recruitment and retention of activated T lymphocytes to vascular wall lesions during the atherogenesis process (68,69), and also is being investigated as a potential biomarker for CVD (7073). In our study, the serum level of CXCL10 was significantly elevated in female and male ApoE/ mice 24 h after high dose thoracic irradiation, and was found to be secreted from both TICAE and TIME cells 24 h post-irradiation. In line with our in vitro data, CXCL10 gene expression was previously reported to be upregulated in human coronary artery endothelial cells after 10 Gy of fractionated X-irradiation (74), and in human umbilical vascular endothelial cells following exposure to 20 Gy of acute gamma irradiation (75).

One of the earliest responses to endothelial cell injury, which is considered one potential initiating event of the atherosclerotic process, is upregulation of adhesion molecules including ICAM-1 and P-selectin, leading to leukocyte adherence to the endothelium (30,76,77). Multiple in vitro and in vivo studies have reported post-irradiation increases in ICAM-1 and P-selectin levels, thereby promoting leucocyte adhesion to the endothelium (26,7883). However, most of the in vivo studies were performed in endothelial cells of irradiated arteries weeks to months after radiation exposure. Here, we report an acute dose-dependent increase in serum ICAM-1 level 24 h after thoracic irradiation in male ApoE/ mice, and this increase was stabilized 1 month later. Previous observations have shown that local irradiation of carotid arteries in female and male ApoE/ mice did not induce ICAM-1 changes at 1 and 22 weeks after 14 Gy X-ray exposure (17,18), indicating that the early 24 h response reported here may be transient in nature. We further found a corresponding ICAM-1 increase in the irradiated TIME cells at 24 h for the 5 Gy dose, which is in line with previous in vitro studies performed at the same time window (39,84). Moreover, we showed that the serum level of P-selectin was increased 24 h after irradiation, again only in male ApoE/ mice. This elevated P-selectin level was also observed in irradiated TICAE and TIME cells, which confirms previous in vitro findings (79). The inflammatory responses observed at high irradiation dose (5 Gy) in TICAE and TIME cells could be an injury response, since an increased DNA damage and persistent cell death were previously observed in these cells after 5 Gy X-irradiation (51).

Our data further demonstrate a significant decrease in atheroprotective basic fibroblast growth factor (FGF-basic), in female ApoE/ mice at 10 Gy, and a decreasing trend in male ApoE/ mice 1 month after irradiation, resulting is a significant dose-dependent decrease in the combined female and male data. A previous study performed in non-Hodgkin lymphoma cancer patients showed a significant decrease in FGF-2 serum level after doses ranging between 6 and 52 Gy of radiotherapy (85), and another study performed in patients with different tumour histotypes, also reported that FGF-2 serum level decreased after radiotherapy (86). FGF-basic was observed to have a protective effect against irradiation (87), since it inhibited radiation-induced apoptosis of endothelial cells under both in vitro and in vivo conditions (88,89), and was also reported to decrease VCAM-1 expression and macrophage presence in atherosclerotic plaques from rabbits fed with a high cholesterol diet (90).

In contrast to the proatherogenic inflammatory response after thoracic irradiation, our results showed an acute decrease in the proatherogenic cytokine MCP-1, only observed in female mice 24 h after irradiation. MCP-1 is involved in atherosclerosis initiation and progression by recruiting monocytes and contributing to macrophage infiltration into the subendothelial cell layer (91,92). Interestingly, it has been shown that post-irradiation MCP-1-mediated chemoattraction is regulated by the proatherogenic uPAR expression (93,94). In line with this observation, our results showed a decrease in uPAR serum level only in female ApoE/ mice 24 h after 10 Gy exposure. It is worth mentioning that the observed alterations in inflammatory markers, as discussed, are limited to multiplex bead assay assessment, and further validation using other approaches, such as western blotting or RT-qPCR, are required.

The gender difference in response to radiation exposure could be explained by hormone differences between male and female. Several groups have reported that estrogen is protective against vascular dysfunction and atherosclerotic lesions in mice (9598). Estrogen is known to exert protective and beneficial effects in the cardiovascular system by improving vascular function, increasing NO production and by inhibiting proliferation and migration of vascular smooth muscle cells (99101). It was also shown that atherosclerotic lesions were significantly less extensive in female ApoE/ mice than in male mice (101). Indeed, in our experiment, we observed an increase in the pro-inflammatory markers ICAM-1, P-selectin in male mice while a decrease in the pro-inflammatory MCP-1 and uPAR levels in female mice. Though, further investigations are required to unveil mechanisms behind the observed gender specific response after irradiation in ApoE/ mice, by scrutinizing the effect of sex steroid hormones on cytokine response in vitro, or by using knock-down/out techniques in vivo.

Taken together, our results reveal acute and early term inflammatory responses after X-ray exposure of ApoE/ mice and of coronary artery and microvascular endothelial cells. Future research is needed to fully grasp the scope of early changes in the cardiovascular system after thoracic irradiation, and to determine whether GDF-15 and CXCL10 could be used as potential biomarkers for early detection of cardiovascular risks in thoracic radiotherapy-treated patients, thus identifying patients who may benefit from early medical intervention.

Supplementary Material

Supporting Data

Acknowledgements

The authors would like to thank Dr Kenneth Raj (Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Didcot, UK) who donated the TICAE cell line.

Funding

This work was supported by the Fund for Scientific Research Flanders, Belgium (grant no. G040720N). RR was supported by a doctoral grant obtained from Belgian Nuclear Research Centre/Ghent University.

Availability of data and materials

The datasets used and/or analysed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

RR conducted ex-vivo and in vitro experiments, guided and supervised the in vivo irradiation and blood collection experiments, and wrote the manuscript. MC and EC conducted in vivo irradiation and blood sampling, as well as the cholesterol and triglyceride experiments. MM analysed the in vivo experiments. RR and AA confirm the authenticity of all the raw data. ED, SB, AA and LL designed the experiments and supervised the study. All authors, except MC, contributed equally in reviewing the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Mice were treated according to the European Ethics Committee guidelines and the study protocol was approved by the Animal Experiment Ethical Committee of the Faculty of Medicine and Health Sciences, Ghent University, Belgium (approval no. ECD 17/60).

Patient consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

References

1 

Kong FM, Zhao L and Hayman JA: The role of radiation therapy in thoracic tumors. Hematol Oncol Clin North Am. 20:363–400. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Baskar R, Lee KA, Yeo R and Yeoh KW: Cancer and radiation therapy: Current advances and future directions. Int J Med Sci. 9:193–199. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Delaney G, Jacob S, Featherstone C and Barton M: The role of radiotherapy in cancer treatment: Estimating optimal utilization from a review of evidence-based clinical guidelines. Cancer. 104:1129–1137. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein KD, Alteri R and Jemal A: Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 66:271–289. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Darby S, McGale P, Peto R, Granath F, Hall P and Ekbom A: Mortality from cardiovascular disease more than 10 years after radiotherapy for breast cancer: Nationwide cohort study of 90 000 Swedish women. BMJ. 326:256–257. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Darby SC, Ewertz M, McGale P, Bennet AM, Blom-Goldman U, Brønnum D, Correa C, Cutter D, Gagliardi G, Gigante B, et al: Risk of ischemic heart disease in women after radiotherapy for breast cancer. N Engl J Med. 368:987–998. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Baselet B, Rombouts C, Benotmane AM, Baatout S and Aerts A: Cardiovascular diseases related to ionizing radiation: The risk of low-dose exposure (review). Int J Mol Med. 38:1623–1641. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Yusuf SW, Sami S and Daher IN: Radiation-induced heart disease: A clinical update. Cardiol Res Pract. 2011:3176592011. View Article : Google Scholar : PubMed/NCBI

9 

Authors on behalf of ICRP, ; Stewart FA, Akleyev AV, Hauer-Jensen M, Hendry JH, Kleiman NJ, Macvittie TJ, Aleman BM, Edgar AB, Mabuchi K, et al: ICRP publication 118: ICRP statement on tissue reactions and early and late effects of radiation in normal tissues and organs-threshold doses for tissue reactions in a radiation protection context. Ann ICRP. 41:1–322. 2012. View Article : Google Scholar

10 

Little MP: Radiation and circulatory disease. Mutat Res. 770:299–318. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Eftekhari M, Anbiaei R, Zamani H, Fallahi B, Beiki D, Ameri A, Emami-Ardekani A, Fard-Esfahani A, Gholamrezanezhad A, Seid Ratki KR and Roknabadi AM: Radiation-induced myocardial perfusion abnormalities in breast cancer patients following external beam radiation therapy. Asia Ocean J Nucl Med Biol. 3:3–9. 2015.PubMed/NCBI

12 

Kole TP, Aghayere O, Kwah J, Yorke ED and Goodman KA: Comparison of heart and coronary artery doses associated with intensity-modulated radiotherapy versus three-dimensional conformal radiotherapy for distal esophageal cancer. Int J Radiat Oncol Biol Phys. 83:1580–1586. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Tillman GF, Pawlicki T, Koong AC and Goodman KA: Preoperative versus postoperative radiotherapy for locally advanced gastroesophageal junction and proximal gastric cancers: A comparison of normal tissue radiation doses. Dis Esophagus. 21:437–444. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Hong JC, Rahimy E, Gross CP, Shafman T, Hu X, Yu JB, Ross R, Finkelstein SE, Dosoretz A, Park HS, et al: Radiation dose and cardiac risk in breast cancer treatment: An analysis of modern radiation therapy including community settings. Pract Radiat Oncol. 8:e79–e86. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Al-Kindi SG and Oliveira GH: Incidence and trends of cardiovascular mortality after common cancers in young adults: Analysis of surveillance, epidemiology and end-results program. World J Cardiol. 8:368–374. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Drost L, Yee C, Lam H, Zhang L, Wronski M, McCann C, Lee J, Vesprini D, Leung E and Chow E: A systematic review of heart dose in breast radiotherapy. Clin Breast Cancer. 18:e819–e824. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Hoving S, Heeneman S, Gijbels MJ, te Poele JA, Russell NS, Daemen MJ and Stewart FA: Single-dose and fractionated irradiation promote initiation and progression of atherosclerosis and induce an inflammatory plaque phenotype in ApoE(−/−) mice. Int J Radiat Oncol Biol Phys. 71:848–857. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Stewart FA, Heeneman S, Te Poele J, Kruse J, Russell NS, Gijbels M and Daemen M: Ionizing radiation accelerates the development of atherosclerotic lesions in ApoE−/− mice and predisposes to an inflammatory plaque phenotype prone to hemorrhage. Am J Pathol. 168:649–658. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Kreuzer M, Auvinen A, Cardis E, Hall J, Jourdain JR, Laurier D, Little MP, Peters A, Raj K, Russell NS, et al: Low-dose ionising radiation and cardiovascular diseases-strategies for molecular epidemiological studies in Europe. Mutat Res Rev Mutat Res. 764:90–100. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Shimizu Y, Kodama K, Nishi N, Kasagi F, Suyama A, Soda M, Grant EJ, Sugiyama H, Sakata R, Moriwaki H, et al: Radiation exposure and circulatory disease risk: Hiroshima and Nagasaki atomic bomb survivor data, 1950–2003. BMJ. 340:b53492010. View Article : Google Scholar : PubMed/NCBI

21 

Yamada M, Naito K, Kasagi F, Masunari N and Suzuki G: Prevalence of atherosclerosis in relation to atomic bomb radiation exposure: An RERF adult health study. Int J Radiat Biol. 81:821–826. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Azizova TV, Grigoryeva ES, Haylock RG, Pikulina MV and Moseeva MB: Ischaemic heart disease incidence and mortality in an extended cohort of Mayak workers first employed in 1948–1982. Br J Radiol. 88:201501692015. View Article : Google Scholar : PubMed/NCBI

23 

Kashcheev VV, Chekin SY, Karpenko SV, Maksioutov MA, Menyaylo AN, Tumanov KA, Kochergina EV, Kashcheeva PV, Gorsky AI, Shchukina NV, et al: Radiation risk of cardiovascular diseases in the cohort of Russian emergency workers of the chernobyl accident. Health Phys. 113:23–29. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Ramadan R, Vromans E, Anang DC, Decrock E, Mysara M, Monsieurs P, Baatout S, Leybaert L and Aerts A: Single and fractionated ionizing radiation induce alterations in endothelial connexin expression and channel function. Sci Rep. 9:46432019. View Article : Google Scholar : PubMed/NCBI

25 

Mathias D, Mitchel RE, Barclay M, Wyatt H, Bugden M, Priest ND, Whitman SC, Scholz M, Hildebrandt G, Kamprad M and Glasow A: Low-dose irradiation affects expression of inflammatory markers in the heart of ApoE−/− mice. PLoS One. 10:e01196612015. View Article : Google Scholar : PubMed/NCBI

26 

Sievert W, Trott KR, Azimzadeh O, Tapio S, Zitzelsberger H and Multhoff G: Late proliferating and inflammatory effects on murine microvascular heart and lung endothelial cells after irradiation. Radiother Oncol. 117:376–381. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Mitchel RE, Hasu M, Bugden M, Wyatt H, Little MP, Gola A, Hildebrandt G, Priest ND and Whitman SC: Low-dose radiation exposure and atherosclerosis in ApoE(−)/(−) mice. Radiat Res. 175:665–676. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Mancuso M, Pasquali E, Braga-Tanaka I III, Tanaka S, Pannicelli A, Giardullo P, Pazzaglia S, Tapio S, Atkinson MJ and Saran A: Acceleration of atherogenesis in ApoE−/− mice exposed to acute or low-dose-rate ionizing radiation. Oncotarget. 6:31263–31271. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Kumarathasan P, Vincent R, Blais E, Saravanamuthu A, Gupta P, Wyatt H, Mitchel R, Hannan M, Trivedi A and Whitman S: Cardiovascular changes in atherosclerotic ApoE-deficient mice exposed to Co60 (ү) radiation. PLoS One. 8:e654862013. View Article : Google Scholar : PubMed/NCBI

30 

Massberg S, Brand K, Gruner S, Page S, Müller E, Müller I, Bergmeier W, Richter T, Lorenz M, Konrad I, et al: A critical role of platelet adhesion in the initiation of atherosclerotic lesion formation. J Exp Med. 196:887–896. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Heidenreich PA, Hancock SL, Lee BK, Mariscal CS and Schnittger I: Asymptomatic cardiac disease following mediastinal irradiation. J Am Coll Cardiol. 42:743–749. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Yusuf SW, Venkatesulu BP, Mahadevan LS and Krishnan S: Radiation-induced cardiovascular disease: A clinical perspective. Front Cardiovasc Med. 4:662017. View Article : Google Scholar : PubMed/NCBI

33 

Corrado E, Rizzo M, Coppola G, Fattouch K, Novo G, Marturana I, Ferrara F and Novo S: An update on the role of markers of inflammation in atherosclerosis. J Atheroscler Thromb. 17:1–11. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Soeki T and Sata M: Inflammatory biomarkers and atherosclerosis. Int Heart J. 57:134–139. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Libby P: Inflammation in atherosclerosis. Arterioscler Thromb Vasc Biol. 32:2045–2051. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Murabito JM, Keyes MJ, Guo CY, Keaney JF Jr, Vasan RS, D'Agostino RB Sr and Benjamin EJ: Cross-sectional relations of multiple inflammatory biomarkers to peripheral arterial disease: The Framingham offspring study. Atherosclerosis. 203:509–514. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Halle M, Gabrielsen A, Paulsson-Berne G, Gahm C, Agardh HE, Farnebo F and Tornvall P: Sustained inflammation due to nuclear factor-kappa B activation in irradiated human arteries. J Am Coll Cardiol. 55:1227–1236. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Kiyohara H, Ishizaki Y, Suzuki Y, Katoh H, Hamada N, Ohno T, Takahashi T, Kobayashi Y and Nakano T: Radiation-induced ICAM-1 expression via TGF-β1 pathway on human umbilical vein endothelial cells; comparison between X-ray and carbon-ion beam irradiation. J Radiat Res. 52:287–292. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Hallahan D, Kuchibhotla J and Wyble C: Cell adhesion molecules mediate radiation-induced leukocyte adhesion to the vascular endothelium. Cancer Res. 56:5150–5155. 1996.PubMed/NCBI

40 

Di Maggio FM, Minafra L, Forte GI, Cammarata FP, Lio D, Messa C, Gilardi MC and Bravatà V: Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond). 12:142015. View Article : Google Scholar : PubMed/NCBI

41 

Baluna RG, Eng TY and Thomas CR: Adhesion molecules in radiotherapy. Radiat Res. 166:819–831. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Min X, Lu M, Tu S, Wang X, Zhou C, Wang S, Pang S, Qian J, Ge Y, Guo Y, et al: Serum cytokine profile in relation to the severity of coronary artery disease. Biomed Res Int. 2017:40136852017. View Article : Google Scholar : PubMed/NCBI

43 

Peng J, Luo F, Ruan G, Peng R and Li X: Hypertriglyceridemia and atherosclerosis. Lipids Health Dis. 16:2332017. View Article : Google Scholar : PubMed/NCBI

44 

Sloop GD: A critical analysis of the role of cholesterol in atherogenesis. Atherosclerosis. 142:265–268. 1999. View Article : Google Scholar : PubMed/NCBI

45 

Lowe D and Raj K: Premature aging induced by radiation exhibits pro-atherosclerotic effects mediated by epigenetic activation of CD44 expression. Aging Cell. 13:900–910. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Lowe D, Horvath S and Raj K: Epigenetic clock analyses of cellular senescence and ageing. Oncotarget. 7:8524–8531. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Leligdowicz A, Conroy AL, Hawkes M, Zhong K, Lebovic G, Matthay MA and Kain KC: Validation of two multiplex platforms to quantify circulating markers of inflammation and endothelial injury in severe infection. PLoS One. 12:e01751302017. View Article : Google Scholar : PubMed/NCBI

48 

Vitkova V, Panek M, Janec P, Šibíková M, Vobruba V, Haluzík M, Živný J and Janota J: Endothelial microvesicles and soluble markers of endothelial injury in critically Ill newborns. Mediators Inflamm. 2018:19750562018. View Article : Google Scholar : PubMed/NCBI

49 

Bahlas S, Damiati L, Dandachi N, Sait H, Alsefri M and Pushparaj PN: Rapid immunoprofiling of cytokines, chemokines and growth factors in patients with active rheumatoid arthritis using luminex multiple analyte profiling technology for precision medicine. Clin Exp Rheumatol. 37:112–119. 2019.PubMed/NCBI

50 

Reslova N, Michna V, Kasny M, Mikel P and Kralik P: xMAP technology: Applications in detection of pathogens. Front Microbiol. 8:552017. View Article : Google Scholar : PubMed/NCBI

51 

Ramadan R, Vromans E, Anang DC, Goetschalckx I, Hoorelbeke D, Decrock E, Baatout S, Leybaert L and Aerts A: Connexin43 hemichannel targeting with TAT-Gap19 alleviates radiation-induced endothelial cell damage. Front Pharmacol. 11:2122020. View Article : Google Scholar : PubMed/NCBI

52 

Darby SC, Cutter DJ, Boerma M, Constine LS, Fajardo LF, Kodama K, Mabuchi K, Marks LB, Mettler FA, Pierce LJ, et al: Radiation-related heart disease: Current knowledge and future prospects. Int J Radiat Oncol Biol Phys. 76:656–665. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Aleman BM, Moser EC, Nuver J, Suter TM, Maraldo MV, Specht L, Vrieling C and Darby SC: Cardiovascular disease after cancer therapy. EJC Suppl. 12:18–28. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Monceau V, Meziani L, Strup-Perrot C, Morel E, Schmidt M, Haagen J, Escoubet B, Dörr W and Vozenin MC: Enhanced sensitivity to low dose irradiation of ApoE−/− mice mediated by early pro-inflammatory profile and delayed activation of the TGFβ1 cascade involved in fibrogenesis. PLoS One. 8:e570522013. View Article : Google Scholar : PubMed/NCBI

55 

Brunner D, Altman S, Loebl K, Schwartz S and Levin S: Serum cholesterol and triglycerides in patients suffering from ischemic heart disease and in healthy subjects. Atherosclerosis. 28:197–204. 1977. View Article : Google Scholar : PubMed/NCBI

56 

Sarwar N, Danesh J, Eiriksdottir G, Sigurdsson G, Wareham N, Bingham S, Boekholdt SM, Khaw KT and Gudnason V: Triglycerides and the risk of coronary heart disease: 10,158 incident cases among 262,525 participants in 29 Western prospective studies. Circulation. 115:450–458. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Ozmen HK, Erdemci B, Askin S and Sezen O: Carnitine and adiponectin levels in breast cancer after radiotherapy. Open Med (Wars). 12:189–194. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Park H, Kim CH, Jeong JH, Park M and Kim KS: GDF15 contributes to radiation-induced senescence through the ROS-mediated p16 pathway in human endothelial cells. Oncotarget. 7:9634–9644. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Hoving S, Heeneman S, Gijbels MJ, Te Poele JA, Visser N, Cleutjens J, Russell NS, Daemen MJ and Stewart FA: Irradiation induces different inflammatory and thrombotic responses in carotid arteries of wildtype C57BL/6J and atherosclerosis-prone ApoE(−/−) mice. Radiother Oncol. 105:365–370. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Bootcov MR, Bauskin AR, Valenzuela SM, Moore AG, Bansal M, He XY, Zhang HP, Donnellan M, Mahler S, Pryor K, et al: MIC-1, a novel macrophage inhibitory cytokine, is a divergent member of the TGF-beta superfamily. Proc Natl Acad Sci USA. 94:11514–11519. 1997. View Article : Google Scholar : PubMed/NCBI

61 

Hsiao EC, Koniaris LG, Zimmers-Koniaris T, Sebald SM, Huynh TV and Lee SJ: Characterization of growth-differentiation factor 15, a transforming growth factor beta superfamily member induced following liver injury. Mol Cell Biol. 20:3742–3751. 2000. View Article : Google Scholar : PubMed/NCBI

62 

Wollert KC and Kempf T: Growth differentiation factor 15 in heart failure: An update. Curr Heart Fail Rep. 9:337–345. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Chen J, Luo F, Fang Z and Zhang W: GDF-15 levels and atherosclerosis. Int J Cardiol. 257:362018. View Article : Google Scholar : PubMed/NCBI

64 

Kempf T and Wollert KC: Growth differentiation factor-15: A new biomarker in cardiovascular disease. Herz. 34:594–599. 2009. View Article : Google Scholar : PubMed/NCBI

65 

Xu X, Li Z and Gao W: Growth differentiation factor 15 in cardiovascular diseases: From bench to bedside. Biomarkers. 16:466–475. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Bonaterra GA, Zugel S, Thogersen J, Walter SA, Haberkorn U, Strelau J and Kinscherf R: Growth differentiation factor-15 deficiency inhibits atherosclerosis progression by regulating interleukin-6-dependent inflammatory response to vascular injury. J Am Heart Assoc. 1:e0025502012. View Article : Google Scholar : PubMed/NCBI

67 

de Jager SC, Bermudez B, Bot I, Koenen RR, Bot M, Kavelaars A, de Waard V, Heijnen CJ, Muriana FJ, Weber C, et al: Growth differentiation factor 15 deficiency protects against atherosclerosis by attenuating CCR2-mediated macrophage chemotaxis. J Exp Med. 208:217–225. 2011. View Article : Google Scholar : PubMed/NCBI

68 

Heller EA, Liu E, Tager AM, Yuan Q, Lin AY, Ahluwalia N, Jones K, Koehn SL, Lok VM, Aikawa E, et al: Chemokine CXCL10 promotes atherogenesis by modulating the local balance of effector and regulatory T cells. Circulation. 113:2301–2312. 2006. View Article : Google Scholar : PubMed/NCBI

69 

Mach F, Sauty A, Iarossi AS, Sukhova GK, Neote K, Libby P and Luster AD: Differential expression of three T lymphocyte-activating CXC chemokines by human atheroma-associated cells. J Clin Invest. 104:1041–1050. 1999. View Article : Google Scholar : PubMed/NCBI

70 

van den Borne P, Quax PH, Hoefer IE and Pasterkamp G: The multifaceted functions of CXCL10 in cardiovascular disease. Biomed Res Int. 2014:8931062014. View Article : Google Scholar : PubMed/NCBI

71 

Ardigo D, Assimes TL, Fortmann SP, Go AS, Hlatky M, Hytopoulos E, Iribarren C, Tsao PS, Tabibiazar R and Quertermous T; ADVANCE Investigators, : Circulating chemokines accurately identify individuals with clinically significant atherosclerotic heart disease. Physiol Genomics. 31:402–409. 2007. View Article : Google Scholar : PubMed/NCBI

72 

Orn S, Breland UM, Mollnes TE, Manhenke C, Dickstein K, Aukrust P and Ueland T: The chemokine network in relation to infarct size and left ventricular remodeling following acute myocardial infarction. Am J Cardiol. 104:1179–1183. 2009. View Article : Google Scholar : PubMed/NCBI

73 

Tavakolian Ferdousie V, Mohammadi M, Hassanshahi G, Khorramdelazad H, Khanamani Falahati-Pour S, Mirzaei M, Allah Tavakoli M, Kamiab Z, Ahmadi Z, Vazirinejad R, et al: Serum CXCL10 and CXCL12 chemokine levels are associated with the severity of coronary artery disease and coronary artery occlusion. Int J Cardiol. 233:23–28. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Palayoor ST, John-Aryankalayil M, Makinde AY, Falduto MT, Magnuson SR and Coleman CN: Differential expression of stress and immune response pathway transcripts and miRNAs in normal human endothelial cells subjected to fractionated or single-dose radiation. Mol Cancer Res. 12:1002–1015. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Heinonen M, Milliat F, Benadjaoud MA, François A, Buard V, Tarlet G, d'Alché-Buc F and Guipaud O: Temporal clustering analysis of endothelial cell gene expression following exposure to a conventional radiotherapy dose fraction using Gaussian process clustering. PLoS One. 13:e02049602018. View Article : Google Scholar : PubMed/NCBI

76 

Libby P, Ridker PM and Maseri A: Inflammation and atherosclerosis. Circulation. 105:1135–1143. 2002. View Article : Google Scholar : PubMed/NCBI

77 

Lusis AJ: Atherosclerosis. Nature. 407:233–241. 2000. View Article : Google Scholar : PubMed/NCBI

78 

Hallahan DE and Virudachalam S: Accumulation of P-selectin in the lumen of irradiated blood vessels. Radiat Res. 152:6–13. 1999. View Article : Google Scholar : PubMed/NCBI

79 

Tu J, Hu Z and Chen Z: Endothelial gene expression and molecular changes in response to radiosurgery in in vitro and in vivo models of cerebral arteriovenous malformations. Biomed Res Int. 2013:4082532013. View Article : Google Scholar : PubMed/NCBI

80 

Hallahan DE and Virudachalam S: Ionizing radiation mediates expression of cell adhesion molecules in distinct histological patterns within the lung. Cancer Res. 57:2096–2099. 1997.PubMed/NCBI

81 

Gaugler MH, Squiban C, van der Meeren A, Bertho JM, Vandamme M and Mouthon MA: Late and persistent up-regulation of intercellular adhesion molecule-1 (ICAM-1) expression by ionizing radiation in human endothelial cells in vitro. Int J Radiat Biol. 72:201–209. 1997. View Article : Google Scholar : PubMed/NCBI

82 

Haubner F, Leyh M, Ohmann E, Pohl F, Prantl L and Gassner HG: Effects of external radiation in a co-culture model of endothelial cells and adipose-derived stem cells. Radiat Oncol. 8:662013. View Article : Google Scholar : PubMed/NCBI

83 

Azimzadeh O, Sievert W, Sarioglu H, Merl-Pham J, Yentrapalli R, Bakshi MV, Janik D, Ueffing M, Atkinson MJ, Multhoff G and Tapio S: Integrative proteomics and targeted transcriptomics analyses in cardiac endothelial cells unravel mechanisms of long-term radiation-induced vascular dysfunction. J Proteome Res. 14:1203–1219. 2015. View Article : Google Scholar : PubMed/NCBI

84 

Cervelli T, Panetta D, Navarra T, Andreassi MG, Basta G, Galli A, Salvadori PA, Picano E and Del Turco S: Effects of single and fractionated low-dose irradiation on vascular endothelial cells. Atherosclerosis. 235:510–518. 2014. View Article : Google Scholar : PubMed/NCBI

85 

Ria R, Cirulli T, Giannini T, Bambace S, Serio G, Portaluri M, Ribatti D, Vacca A and Dammacco F: Serum levels of angiogenic cytokines decrease after radiotherapy in non-Hodgkin lymphomas. Clin Exp Med. 8:141–145. 2008. View Article : Google Scholar : PubMed/NCBI

86 

Ria R, Portaluri M, Russo F, Cirulli T, Di Pietro G, Bambace S, Cucci F, Romano T, Vacca A and Dammacco F: Serum levels of angiogenic cytokines decrease after antineoplastic radiotherapy. Cancer Lett. 216:103–107. 2004. View Article : Google Scholar : PubMed/NCBI

87 

Yang X, Liaw L, Prudovsky I, Brooks PC, Vary C, Oxburgh L and Friesel R: Fibroblast growth factor signaling in the vasculature. Curr Atheroscler Rep. 17:5092015. View Article : Google Scholar : PubMed/NCBI

88 

Fuks Z, Persaud RS, Alfieri A, McLoughlin M, Ehleiter D, Schwartz JL, Seddon AP, Cordon-Cardo C and Haimovitz-Friedman A: Basic fibroblast growth factor protects endothelial cells against radiation-induced programmed cell death in vitro and in vivo. Cancer Res. 54:2582–2590. 1994.PubMed/NCBI

89 

Zhang S, Qiu X, Zhang Y, Fu K, Zhao X, Wu J, Hu Y, Zhu W and Guo H: Basic fibroblast growth factor ameliorates endothelial dysfunction in radiation-induced bladder injury. Biomed Res Int. 2015:9676802015.PubMed/NCBI

90 

Six I, Mouquet F, Corseaux D, Bordet R, Letourneau T, Vallet B, Dosquet CC, Dupuis B, Jude B, Bertrand ME, et al: Protective effects of basic fibroblast growth factor in early atherosclerosis. Growth Factors. 22:157–167. 2004. View Article : Google Scholar : PubMed/NCBI

91 

Aiello RJ, Bourassa PA, Lindsey S, Weng W, Natoli E, Rollins BJ and Milos PM: Monocyte chemoattractant protein-1 accelerates atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 19:1518–1525. 1999. View Article : Google Scholar : PubMed/NCBI

92 

Harrington JR: The role of MCP-1 in atherosclerosis. Stem Cells. 18:65–66. 2000. View Article : Google Scholar : PubMed/NCBI

93 

Nalla AK, Gogineni VR, Gupta R, Dinh DH and Rao JS: Suppression of uPA and uPAR blocks radiation-induced MCP-1 mediated recruitment of endothelial cells in meningioma. Cell Signal. 23:1299–1310. 2011. View Article : Google Scholar : PubMed/NCBI

94 

Farris SD, Hu JH, Krishnan R, Emery I, Chu T, Du L, Kremen M, Dichek HL, Gold E, Ramsey SA and Dichek DA: Mechanisms of urokinase plasminogen activator (uPA)-mediated atherosclerosis: Role of the uPA receptor and S100A8/A9 proteins. J Biol Chem. 286:22665–22677. 2011. View Article : Google Scholar : PubMed/NCBI

95 

Kimura M, Sudhir K, Jones M, Simpson E, Jefferis AM and Chin-Dusting JP: Impaired acetylcholine-induced release of nitric oxide in the aorta of male aromatase-knockout mice: Regulation of nitric oxide production by endogenous sex hormones in males. Circ Res. 93:1267–1271. 2003. View Article : Google Scholar : PubMed/NCBI

96 

Adams MR, Golden DL, Register TC, Anthony MS, Hodgin JB, Maeda N and Williams JK: The atheroprotective effect of dietary soy isoflavones in apolipoprotein E−/− mice requires the presence of estrogen receptor-alpha. Arterioscler Thromb Vasc Biol. 22:1859–1864. 2002. View Article : Google Scholar : PubMed/NCBI

97 

Zhu Y, Bian Z, Lu P, Karas RH, Bao L, Cox D, Hodgin J, Shaul PW, Thoren P, Smithies O, et al: Abnormal vascular function and hypertension in mice deficient in estrogen receptor beta. Science. 295:505–508. 2002. View Article : Google Scholar : PubMed/NCBI

98 

Hodgin JB, Krege JH, Reddick RL, Korach KS, Smithies O and Maeda N: Estrogen receptor alpha is a major mediator of 17beta-estradiol's atheroprotective effects on lesion size in Apoe−/− mice. J Clin Invest. 107:333–340. 2001. View Article : Google Scholar : PubMed/NCBI

99 

Hurtado R, Celani M and Geber S: Effect of short-term estrogen therapy on endothelial function: A double-blinded, randomized, controlled trial. Climacteric. 19:448–451. 2016. View Article : Google Scholar : PubMed/NCBI

100 

Zheng S, Chen X, Hong S, Long L, Xu Y, Simoncini T and Fu X: 17β-Estradiol inhibits vascular smooth muscle cell migration via up-regulation of striatin protein. Gynecol Endocrinol. 31:618–624. 2015. View Article : Google Scholar : PubMed/NCBI

101 

Zhang G, Li C, Zhu N, Chen Y, Yu Q, Liu E and Wang R: Sex differences in the formation of atherosclerosis lesion in apoE−/−mice and the effect of 17β-estrodiol on protein S-nitrosylation. Biomed Pharmacother. 99:1014–1021. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2021
Volume 23 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ramadan R, Claessens M, Cocquyt E, Mysara M, Decrock E, Baatout S, Aerts A and Leybaert L: X‑irradiation induces acute and early term inflammatory responses in atherosclerosis‑prone ApoE‑/‑ mice and in endothelial cells. Mol Med Rep 23: 399, 2021
APA
Ramadan, R., Claessens, M., Cocquyt, E., Mysara, M., Decrock, E., Baatout, S. ... Leybaert, L. (2021). X‑irradiation induces acute and early term inflammatory responses in atherosclerosis‑prone ApoE‑/‑ mice and in endothelial cells. Molecular Medicine Reports, 23, 399. https://doi.org/10.3892/mmr.2021.12038
MLA
Ramadan, R., Claessens, M., Cocquyt, E., Mysara, M., Decrock, E., Baatout, S., Aerts, A., Leybaert, L."X‑irradiation induces acute and early term inflammatory responses in atherosclerosis‑prone ApoE‑/‑ mice and in endothelial cells". Molecular Medicine Reports 23.6 (2021): 399.
Chicago
Ramadan, R., Claessens, M., Cocquyt, E., Mysara, M., Decrock, E., Baatout, S., Aerts, A., Leybaert, L."X‑irradiation induces acute and early term inflammatory responses in atherosclerosis‑prone ApoE‑/‑ mice and in endothelial cells". Molecular Medicine Reports 23, no. 6 (2021): 399. https://doi.org/10.3892/mmr.2021.12038