Open Access
How to translate text using browser tools
26 September 2019 Mitigating Effects of 1-Palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) on Hematopoietic Acute Radiation Syndrome after Total-Body Ionizing Irradiation in Mice
Yong-Jae Kim, Jinseon Jeong, Su-Hyun Shin, Do Young Lee, Ki-Young Sohn, Sun Young Yoon, Jae Wha Kim
Author Affiliations +
Abstract

Acute radiation syndrome (ARS) occurs as a result of partial- or whole-body, high-dose exposure to radiation in a very short period of time. Survival is dependent on the severity of the hematopoietic sub-syndrome of ARS. In this study, we investigated the mitigating effects of a lipid molecule, 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG), on the kinetics of hematopoietic cells, including absolute neutrophil count (ANC), red blood cells (RBCs) and platelet counts, in mice after gamma-ray total-body irradiation (TBI). Male and female BALB/c mice (11 weeks old) received a LD70/30 dose of TBI. PLAG significantly and dose-dependently attenuated radiation-induced mortality (P = 0.0041 for PLAG 50 mg/kg; P < 0.0001 for PLAG 250 mg/kg) and body weight loss (P < 0.0001 for PLAG 50 and 250 mg/kg) in mice. Single-fraction TBI sharply reduced ANC within 3 days postirradiation and maintained the neutropenic state (ANC < 500 cells/µl) by approximately 26.8 ± 0.8 days. However, administration of PLAG attenuated radiation-induced severe neutropenia (ANC < 100 cells/µl) by effectively delaying the mean day of its onset and decreasing its duration. PLAG also significantly mitigated radiation-induced thrombocytopenia (P < 0.0001 for PLAG 250 mg/kg) and anemia (P = 0.0023 for PLAG 250 mg/kg) by increasing mean platelet and RBC counts, as well as hemoglobin levels, in peripheral blood. Moreover, delayed administration of PLAG, even at 48 and 72 h after gamma-ray irradiation, significantly attenuated radiation-induced mortality in a time-dependent manner. When compared to olive oil and palmitic linoleic hydroxyl (PLH), only PLAG effectively attenuated radiation-induced mortality, indicating that it has a distinctive mechanism of action. Based on these preclinical observations, we concluded that PLAG has high potential as a radiation countermeasure for the improvement of survivability and the treatment of hematopoietic injury in gamma-ray-induced ARS.

INTRODUCTION

Acute radiation syndrome (ARS) is a broad term used to describe a range of signs and symptoms that occur after an entire or large portion of the body is exposed to a high dose of ionizing radiation (1). ARS has been traditionally divided into three sub-syndromes, each with a specific dose threshold for the appearance of clinical symptoms: hematopoietic sub-syndrome (H-ARS, 1–6 Gy), gastrointestinal sub-syndrome (GI-ARS, 6–8 Gy), and neurovascular sub-syndrome (>10 Gy) (24). However, because the damage caused by radiation is not confined to one isolated system, irradiated patients require immediate intensive care to minimize damage to other systems (4).

Over several decades, the U.S. Department of Health and Human Services (specifically, National Institutes of Health and the Biomedical Advanced Research and Development Authority) has sought to investigate potential radiation countermeasures that are safe, easily administered and effective at reducing adverse health effects that occur after radiation exposure (57). Despite these ongoing efforts, as of 2015, only Neupogen® (granulocyte colony-stimulating factor; G-CSF) and Neulasta® (pegylated G-CSF) have been approved by the U.S. Food and Drug Administration (FDA) as radiation countermeasures for the treatment of H-ARS (811). Recently, Leukine® (granulocyte-macrophage colony-stimulating factor; GM-CSF) was approved by the FDA as a radiomitigator to increase survival and to facilitate the recovery of white blood cells in adults and pediatric patients acutely exposed to a sub-lethal dose of radiation (12).

Hematopoietic organs are highly susceptible to the cytotoxic effects of radiation, resulting in immune suppression including neutropenia, thrombocytopenia, and/or anemia (1315). Patients with neutropenia often experience reduced defense against infection and inflammation, consequently followed by mortality due to sepsis (16). Low platelet counts in peripheral blood confer a greater risk of bleeding and delayed wound healing (17). The contributory mechanisms of radiation-induced immune suppression are complex. These involve the death of immune cells, blocking of cell maturation and differentiation and failure of bone marrow to produce immune cells (18). A deeper understanding of the mechanisms by which radiation damages the immune system is necessary for the development of radiation countermeasures that boost immunity after exposure.

The chemically synthesized lipid molecule, 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG), also known as EC-18, is identical to a major constituent found in the antlers of sika deer (19, 20). Previously, the therapeutic efficacy of PLAG has been demonstrated in chemotherapy-associated hematopoietic dysfunction (21). In a published clinical study, PLAG reduced the incidence of gemcitabine-induced neutropenia in patients with unresectable pancreatic cancer (22). In addition, we have previously demonstrated that PLAG exerts a synergistic effect with PEGylated G-CSF (pegfilgrastim) in the treatment of chemotherapy-induced neutropenia by regulating neutrophil extravasation (23). Based on the effect of PLAG on neutrophil modulation, we sought to determine whether it also had therapeutic efficacy as a radiation countermeasure for H-ARS, after exposure to a sub-lethal dose of gamma radiation in BALB/c mice. In this study, survival rate, body weight reduction, peripheral counts of neutrophils, platelets and RBCs, and hemoglobin levels were examined over a 30-day period.

MATERIALS AND METHODS

Animals

Specific-pathogen-free male and female BALB/c mice (10 weeks old) were obtained from Koatech Co. (Pyongtaek, Republic of Korea). Upon receipt, five mice per cage were housed in a specific-pathogen-free facility and acclimatized for one week under consistent temperature and 12:12 h light-dark schedule. All animals were fed a standard mouse diet with water was available ad libitum. All experimental procedures were approved by the Institutional Animal Care and Use Committee of the Korea Research Institute of Bioscience and Biotechnology and were performed in compliance with the “Guide for the Care and Use of Laboratory Animals” by the National Research Council and Korean national laws for animal welfare.

Gamma-ray Irradiation

Male and female BALB/c mice (11 weeks old) were placed in single chambers of a lead-shielding irradiation apparatus and received a single uniform total-body dose of gamma-ray irradiation from a 60Co source (JL Shepherd & Associates, San Fernando, CA) at an exposure rate of 0.833 Gy/min. Dose rates were measured using the EPD Mk2+ electronic dosimeter (Thermo Scientific, Waltham, MA).

Administration of PLAG, Olive Oil and Palmitic Linoleic Hydroxyl Glycerol (PLH)

PLAG was obtained from Enzychem Lifesciences Corp. (Jecheon, South Korea) and re-suspended in sterile phosphate buffered saline (PBS). For the dose experiment, mice received PLAG [50 or 250 mg/ kg, oral (p.o.) administration] or vehicle (sterile PBS; 0.1 ml/mouse, p.o.) beginning 24 h postirradiation and continuing daily to day 30. For the time experiment, mice received PLAG (250 mg/kg, p.o.) either immediately postirradiation (+0 day) or at 24 h (+1 day), 48 h (+2 days) or 72 h (+3 days) postirradiation. and continuing daily to day 30. For the comparative experiment, olive oil (Sigma-Aldrich® LLC, St. Louis, MO) and PLH (Enzychem Lifesciences Corp.) were re-suspended in sterile PBS. Mice were orally administered 250 mg/kg PLAG, 250 mg/kg olive oil, 250 mg/kg PLH or vehicle beginning 24 h postirradiation and continuing daily to day 30.

Assessment of Body Weight

The body weights of mice (10/10 male/female; 20 mice per group) were measured daily for 30 days. The results are expressed as body weights normalized to initial state. Since the mice died from radiation injury, the number of mice that were weighed was different as the study progressed.

Assessment of Survival and the Kinetics of Blood Cells in Peripheral Blood

Mice (20/20 male/female; 40 mice per group) were monitored at least twice daily for survival for 30 days. For assessment of blood cell kinetics, the mice were divided into two cohorts of 20 mice/cohort based on blood collection time. Blood collection schedules were as follows. Cohort 1 collection was performed on days 1, 5, 10, 15, 20 and 27; cohort 2 collection was performed on days 3, 7, 12, 17, 22 and 30. Approximately 30–40 µl of whole blood was collected from the orbital sinuses using EDTA-free capillary tubes (Kimble Chase, Rockwood, TN) and collection tubes containing K3EDTA (Greiner Bio-One International, Kremsmünster, Austria). Since the mice died from radiation injury, the number of blood samples taken from the mice was different as the study progressed. The blood cells were counted and classified by complete blood count (CBC) analysis using a Mindray BC-5000 auto-hematology analyzer (Shenzhen Mindray Biomedical Electronics, Guangdong Sheng, China). The numbers of blood cells were recorded at the appointed dates for 30 days. The mice surviving after the end of experiments underwent euthanasia by CO2 inhalation followed by cervical dislocation.

Statistical Analyses

Dose-dependent mortality was examined over 30 days using logistic regression. For statistical analysis of hematologic and body weight data, one-way ANOVA followed by Tukey-Kramer post hoc test was performed using GraphPad Prism version 8.0 (LaJolla, CA). P values <0.05 were considered statistically significant, and the results were expressed as the mean ± SD. Paired log-rank (Mantel-Cox) test was used to analyze the survivability and the duration of neutropenia, thrombocytopenia and anemia between control and PLAG-treated groups.

RESULTS

Radiation Dose-Response Relationship (DRR) and Determination of LDXX/30

The mortality rate of irradiated mice positively correlates with radiation dose (24). Prior to the PLAG efficacy test, we first investigated the relationship between gamma-ray dose and lethality of mice to determine the lethality dose (LD) of gamma-ray irradiation during the 30-day survival observation. Figure 1A shows Kaplan-Meier survival curves of BALB/c mice irradiated at various doses of 60Co gamma rays; increasing radiation dose significantly decreased the overall survival time. The mean survival time (MST) of decedents for each radiation dose cohort ranged from 13.69 to 15.30 days, with the overall MST of decedents across all dose cohorts being 14.38 days (Table 1). Figure 1B shows the radiation dose-response relationship (DRR) using a probit model. Thirty-day survival was calculated at each radiation dose and is shown as percentage mortality on the y-axis. Based on the probit model in Fig. 1B, we determined LDXX/30 with 95% confidence intervals around each dose. The LD30/30, LD50/30, LD70/30 and LD95/30 values were 5.45, 5.85, 6.11 and 6.35 Gy, respectively (Table 2). The established LD70/30 in this experiment was applied in subsequent experiments to determine PLAG efficacy.

FIG. 1

Survival rates and logistic regression probability of 30-day mortality in BALB/c mice after TBI. Mice (n = 20 per group, 10 males and 10 females) received gamma-ray doses ranging from 6.0 to 6.4 Gy gamma-rays. Panel A: Kaplan-Meier survival curves show the proportion of mice surviving at each time point for each radiation dose. Panel B: Radiation dose-response relationship (DRR) using a probit model. Survival at day 30 was analyzed for each radiation dose and is shown as percentage mortality on the y-axis.

img-z3-1_602.jpg

TABLE 1

Thirty-day Mortality of BALB/c Mice after Gamma-Ray Irradiation

img-AgeL_602.gif

TABLE 2

Estimated Radiation Dose in BALB/c Mice after Gamma-Ray Irradiation

img-ASRY_602.gif

Administration of PLAG Attenuates Mortality and Body Weight Loss in Irradiated Mice

We investigated whether administration of PLAG increases the survivability of mice after receiving LD70/30 (6.11 Gy) TBI. Exposure alone resulted in the death of 68.5% of the animals in the vehicle control group over the 30-day observation period, with an average survival time of 21.2 days among the decedents (Fig. 2A, Table 3). Therefore, the survival rate of the vehicle control group was 32.5%. Conversely, administration of PLAG (50 and 250 mg/kg) significantly enhanced 30-day survival to 60% (P=0.0041) and 85% (P < 0.0001), respectively (Fig. 2A). Moreover, the average survival time of the decedents with PLAG 50 and 250 mg/kg treatment increased to 24.3 and 27.8 days, respectively (Table 3). Based on these observations, PLAG has therapeutic potential for improving survivability and increasing the average duration of life in gamma-ray-induced ARS.

FIG. 2

PLAG increased survival rates and mitigated body weight loss in mice receiving an LD70/30 dose of gamma rays. Mice (n = 40 per group, 20 males and 20 females) received the LD70/30 dose (6.11 Gy) of gamma rays, and were administered 50 or 250 mg/kg of PLAG once a day starting the day after irradiation and continuing until the final day of observation. Panel A: Survival was monitored for 30 days. P = 0.0041, 6.11 Gy + PLAG 50 mg/kg vs. 6.11 Gy; P < 0.0001, 6.11 Gy + PLAG 250 mg/kg vs. 6.11 Gy (log-rank test). Panel B: Body weights were measured daily for 30 days. Data are presented as mean. #6.11 Gy vs. 6.11 Gy + PLAG 50 mg/kg; *6.11 Gy vs. 6.11 Gy + PLAG 250 mg/kg. #/*P < 0.05, ##/**P < 0.01, ###/***P < 0.005.

img-z4-1_602.jpg

TABLE 3

Dose Effect of PLAG (Daily Administration) on Survivability and Average Life Duration of Irradiated Mice

img-A0Ke_602.gif

Figure 2B shows the effect of PLAG on changes in the body weights of irradiated mice over a 30-day observation period. Weight loss was examined in a different batch from that of survival and hematological analyses. The LD70/30 gamma-ray dose resulted in substantial decreases in body weight in the mice. Eighty percent of mice in the vehicle control group receiving irradiation alone had more than 10% body weight loss, and 40% had extreme weight loss, defined as body weight reduction of 20% or more. Only 35% and 15% of mice in the groups receiving PLAG 50 and 250 mg/kg experienced severe radiation-induced weight loss, respectively (Table 4). This observation indicates that PLAG is very effective in mitigating body weight loss in gamma-ray-induced ARS.

TABLE 4

Effect of PLAG (Daily Administration) on Occurrence and Severity of Body Weight Loss of Irradiated Mice

img-ASiV_602.gif

Administration of PLAG Mitigates ANC Loss in Peripheral Blood of Irradiated Mice

Hematological nadirs are known to be closely associated with the decreased survival from ARS (25). Using CBC analysis, we investigated whether enhanced survivability by PLAG results from increases in nadir values. A single-dose of TBI (LD70/30, 6.11 Gy) rapidly diminished the absolute neutrophil counts (ANC) within 3 days after gamma-ray irradiation (Fig. 3A). In particular, the administration of PLAG 50 and 250 mg/kg significantly attenuated radiation-induced depletion of ANC in mice in a dose-dependent manner (Fig. 3A and B). The mean first day of severe neutropenia (ANC < 100 cells/µl) in control and PLAG 50 and 250 mg/kg-treated groups was 3.8 ± 0.3, 5.7 ± 0.6 and 8.5 ± 1.0 days, respectively (Table 5). Although PLAG did not protect irradiated mice from experiencing severe neutropenia, it significantly reduced the duration of severe neutropenia (Table 5). In addition, the group treated with PLAG 250 mg/kg exhibited a significant increase in the mean nadir of ANC, from 20.5 ± 2.2 cells/µl to 49.0 ± 4.9 cells/µl after irradiation (Table 6). These observations show that administration of PLAG has a remarkable effect in preventing gamma-ray-induced ANC depletion.

FIG. 3

PLAG mitigated ANC depletion in mice receiving an LD70/30 dose of gamma rays. Mice (n = 20 per group, 10 males and 10 females) received the LD70/30 dose (6.11 Gy) of gamma rays and were administered 50 or 250 mg/kg of PLAG once a day starting the day after irradiation. Panel A: Effect of PLAG administration on the kinetics of ANC after 6.11 Gy irradiation for 30 days. Panel B: Dots indicate individual ANC data for days 3, 7, 17 and 22. #No radiation vs. 6.11 Gy; *6.11 Gy vs. 6.11 Gy + PLAG 250 mg/kg. #/*P < 0.05, ##/**P < 0.01, ###/***P < 0.005.

img-z5-6_602.jpg

TABLE 5

Mean First Day and Mean Duration of Severe Neutropenia (ANC < 100 cells/µl), Thrombocytopenia (PLT <100 × 103 cells/µl) and Anemia (HGB <12 g/dL) in Control and PLAG-Treated Mice Exposed to 6.11 Gy

img-Aj4_602.gif

TABLE 6

Mean Nadir and Mean Number of Days to Recovery of ANC, Platelets and RBC in Control and PLAG-Treated Mice After 6.11 Gy Irradiation

img-Aa7_602.gif

Administration of PLAG Mitigates Platelet Loss in Peripheral Blood in Irradiated Mice

Low platelet counts increase bleeding risk (26). Thrombocytopenia is a condition in which peripheral blood platelet counts are below 100,000 platelets/µl (17, 27, 28). Single-dose gamma-ray TBI (6.11 Gy) rapidly reduced mean platelet counts to below 50% of the baseline within 5 days, and below 100 × 103 cells/µl within approximately 9 days postirradiation (Fig. 4A). Administration of PLAG did not significantly prevent radiation-induced peripheral platelet depletion. However, administration of PLAG 250 mg/kg significantly reduced the duration of thrombocytopenia from 13.1 ± 1.1 to 6.7 ± 0.5 days (P < 0.0001) (Table 5). In addition, mice treated with PLAG 250 mg/kg exhibited a remarkable increase in the mean nadir of platelet counts, from 27.6 ± 1.9 × 103 cells/µl to 50.1 ± 3.2 × 103 cells/µl, after gamma-ray irradiation, and significantly reduced the mean number of days to platelet recovery (≥100,000 cells/ µl), from 22.8 ± 1.1 to 16.8 ± 0.4 cells/µl (Table 6). Based on this observation, PLAG administration is very effective for recovering gamma-ray-induced depletion of platelet counts.

FIG. 4

PLAG mitigated the depletion of platelet counts in mice receiving an LD70/30 dose of gamma rays. Mice (n = 20 per group, 10 males and 10 females) received the LD70/30 dose (6.11 Gy) of gamma rays and were administered 50 or 250 mg/kg of PLAG once a day starting the day after irradiation. Panel A: The effect of PLAG administration on the kinetics of platelet counts after 6.11 Gy irradiation for 30 days. Panel B: Dots indicate individual platelet counts for days 3, 7, 17 and 22. #No radiation vs. 6.11 Gy; *6.11 Gy vs. 6.11 Gy + PLAG 250 mg/kg. #/*P < 0.05, ##/**P < 0.01, ###/***P < 0.005.

img-z6-6_602.jpg

Administration of PLAG Mitigates the Reduction of RBC Counts and Hemoglobin Levels in Peripheral Blood in Irradiated Mice

In addition to reduced ANC and platelet counts, single-dose gamma-ray TBI (LD70/30, 6.11 Gy) resulted in reduction of RBC counts and hemoglobin over the 30-day observation period (Fig. 5A and C). Anemia is a condition in which RBC count and hemoglobin level are decreased in peripheral blood, defined as a hemoglobin level of less than 13 g/dl in men and of less than 12 g/dl in women (29, 30). Administration of PLAG 250 mg/kg significantly prevented radiation-induced reduction of RBC counts and hemoglobin on days 17 and 22 (Fig. 5B and D). Moreover, administration of PLAG 250 mg/kg significantly reduced the duration of anemia from 15.5 ± 0.8 to 9.7 ± 0.7 days (P < 0.0001) (Table 5). Mice treated with PLAG 250 mg/kg exhibited a remarkable increase in the mean nadir of RBC counts, from 3.0 ± 0.3 × 106 cells/µl to 4.0 ± 0.2 × 106 cells/µl after gamma-ray irradiation, and significantly advanced the mean number of days to recovery of RBCs (≥6.3 × 106 cells/µl) from 26.9 ± 0.8 to 24.1 ± 0.7 days (Table 6). These results indicate that PLAG has a remarkable effect in attenuating gamma-ray-induced anemia.

FIG. 5

PLAG mitigated the depletion of RBC counts and hemoglobin levels in mice receiving an LD70/30 dose of gamma rays. Mice (n = 20 per group, 10 males and 10 females) received the LD70/30 dose (6.11 Gy) of gamma rays and were administered 50 or 250 mg/kg of PLAG once a day starting the day after irradiation. Panels A and C: Effect of PLAG administration on the kinetics of RBC counts and hemoglobin levels after 6.11 Gy irradiation for 30 days. Panels B and D: Dots indicate individual platelet counts or hemoglobin levels for days 3, 7, 17 and 22. #No radiation vs. 6.11 Gy; *6.11 Gy vs. 6.11 Gy + PLAG 250 mg/kg. #/*P < 0.05, ##/**P < 0.01, ###/***P < 0.005.

img-z7-1_602.jpg

Time Effect of PLAG Administration on Survivability of Irradiated Mice

Currently, there are no radiation countermeasures approved by the FDA that are effective for alleviating radiation-induced damage when administered later than 48 h postirradiation (31). Therefore, we investigated how 24-h, 48-h and 72-h delayed administration of PLAG after irradiation influences survival rate for 30 days. In this experiment, using single-stage, TBI of BALB/c mice at a dose of 6.11 Gy gamma rays (LD70/30) caused the death of 65% of the animals in the vehicle control group within 23 days, with the average life span of decedents being 19.2 days, as shown in Figure 6A and Table 6. The survival rate in the vehicle control group was 35%. Survival percentages among irradiated mice receiving PLAG at +0, +1, +2 and +3 days were 80%, 70%, 55% and 55%, respectively. Moreover, the average life spans of decedents that received PLAG at +0, +1, +2 and +3 days were 25.0, 23.7, 18.1, and 22.3 days, respectively (Table 7). This observation indicates that PLAG has therapeutic potential for improving survivability dependent on the starting time of administration in gamma-ray-induced ARS conditions, even if it is administered late in time postirradiation.

FIG. 6

Time effect of PLAG administration on survivability and body weight loss in mice receiving an LD70/30 dose of gamma rays. Mice (n = 20 per group, 10 males and 10 females) received an LD70/30 dose (6.11 Gy) of gamma rays and were administered 250 mg/kg of PLAG once a day starting the same day (+0), as well as 24 h (+1 day), 48 h (+2 days) or 72 h (+3 days) postirradiation. Survival was monitored for 30 days. *P < 0.001, 6.11 Gy + PLAG (+0 day) vs. 6.11 Gy; *P < 0.001, 6.11 Gy + PLAG (+1 day) vs. 6.11 Gy; *P = 0.008, 6.11 Gy + PLAG (+2 days) vs. 6.11 Gy; *P= 0.0012, 6.11 Gy + PLAG (+3 days) vs. 6.11 Gy (log-rank test).

img-z7-3_602.jpg

TABLE 7

Time Effect of Administration of PLAG 250mg/kg on Survivability and Average Life Duration of Irradiated Mice

img-AvIE_602.gif

Effect of Administration of PLAG, Olive Oil and PLH on Survivability and Body Weight Loss in Irradiated Mice

PLAG is a lipid molecule, containing 883 kcal/100 g, with palmitic and linoleic acid esterified at the first and second position of the glycerol backbone and acetyl acid at the third position. We investigated whether the therapeutic efficacy of PLAG results from the uptake of additional calories, compared to olive oil, which contains 884.1 kcal/ 100 g, and whether the PLAG acetyl moiety contributes to improved survivability, compared to its unacetylated form, PLH. The chemical structures of PLAG and PLH are shown in Fig. 7A. A similar set of experiments was simultaneously performed to analyze the time effect of PLAG administration. The survival rate of the vehicle control group was 35%. The survival percentages of irradiated mice receiving PLAG, olive oil or PLH 250 mg/kg were 70%, 25% and 40%, respectively (Fig. 7B). Moreover, the average life spans of the decedents that received PLAG, olive oil or PLH 250 mg/kg were 23.7, 17.9 and 18.8 days, respectively (Table 8). This observation indicates that the acetyl moiety of PLAG might be attributed to improved survivability in gamma-ray-induced ARS conditions, and the mitigating effect of PLAG is not result of the uptake of additional calories.

TABLE 8

Effect of administration of PLAG, Olive Oil, PLH 250 mg/kg on Survivability and Average Life Duration of Irradiated Mice

img-AfvaT_602.gif

DISCUSSION

Since the Fukushima nuclear power plant accident in 2011, public awareness of the threat of radiation has greatly increased, and has prompted the formation of contingency plans for the prophylaxis and treatment of ARS (1, 32, 33). Many government agencies involved in national security and public health, including the U.S. FDA, have been searching for suitable radiation countermeasures for the treatment of H-ARS and GI-ARS for several decades (5–7). However, to date, only a limited number of medical options are available, including G-CSF and PEGylated G-CSF, as mitigators of H-ARS (811). Therefore, the development of safe and effective radiation countermeasures is a priority project for the government.

Here we report that gamma-ray TBI at a lethal dose (LD70/30, 6.11 Gy) has a mortality rate of approximately 70%, and results in substantial reduction of body weight and H-ARS symptoms, including neutropenia, thrombocytopenia and anemia in BALB/c mice. Administration of PLAG 50 mg/ kg and 250 mg/kg resulted in 60% and 85% survival, respectively, after irradiation, and significantly mitigated radiation-induced severe weight loss. We extended the 15-day observation beyond the end of PLAG administration to ensure the durability of the mitigating effect on survival enhancement. During 15 days of the extended period, the survivability of all groups did not change (data not shown). Moreover, PLAG 250 mg/kg significantly alleviated radiation-induced neutropenia, thrombocytopenia and anemia by increasing the counts of neutrophils, platelets and RBCs, respectively, in peripheral blood.

FIG. 7

Effects of administration of PLAG, olive oil and PLH on survivability in mice receiving an LD70/30 dose of gamma rays. Mice (n = 20 per group, 10 males and 10 females) received the LD70/30 dose (6.11 Gy) of gamma rays and were administered 250 mg/kg of PLAG, olive oil and PLH once a day starting the day after irradiation and continuing until the final day of observation. Panel A: Chemical structures of PLAG and PLH. Panel B: Survival was monitored for 30 days. *P < 0.001, 6.11 Gy + PLAG 250 mg/kg vs. 6.11 Gy; *P = 0.432, 6.11 Gy + olive oil 250 mg/kg vs. 6.11 Gy; *P = 0.069, 6.11 Gy + PLH 250 mg/kg vs. 6.11 Gy (log-rank test).

img-z7-5_602.jpg

As reported elsewhere, it has been shown that PLAG is an effective therapeutic candidate in different inflammatory-associated diseases, such as rheumatoid arthritis, atopic dermatitis, asthma and hepatitis (20, 3436). In addition, the therapeutic efficacy of PLAG has been demonstrated in chemotherapy and radiation-induced oral mucositis in hamster and mouse models, through the effective attenuation of neutrophil infiltration into cheek pouches and tongues and a decrease in inflammatory cytokines (37). PLAG is currently undergoing a phase II clinical trial for chemotherapy and radiation-induced oral mucositis. We believe that the efficacy of PLAG in modulating an excessive inflammatory response would also contribute to the improved biological indices in ARS.

Ionizing radiation releases very high energy, which damages molecular components in cells of various tissues (4). DNA is the most susceptible target of radiation, and its damage results in chromosomal aberrations, mutations and eventually cellular death (38, 39). Radiation-induced cellular death by mitotic death and necrosis is associated with the inflammatory response, resulting in hyperinflammatory and hypercytokinemia-like syndromes (40, 41). We observed that PLAG effectively decreased the radiation-induced release of damage-associated molecular patterns (DAMPs) in the bloodstream that induce systemic inflammation, such as high motility group box 1 and s100A9 (data not shown). We believe that the putative mechanism of PLAG in ARS is the accelerated resolution of inflammation by effectively eliminating radiation-induced inflammatory factors.

For the study of the hematopoietic syndrome, most researchers have used, in addition to canine models and nonhuman primates, inbred mouse strains, including BALB/ c, C3H/HeN, B6D2F1/J and C57BL/6. Of these strains, BALB/c is the most sensitive to radiation, and C57BL/6 is the most resistant (18). Because radiation directly affects single- and double-stranded DNA breaks and/or impairment of DNA-damage repair machinery (42), the relatively high sensitivity of BALB/c mice to radiation can be attributed to defects in double-stranded DNA repair (43). In this study, BALB/c mice were used to evaluate the effect of PLAG, since it was believed that this strain, due to its inherent genetic defects, would more apparently exhibit a radiation-induced systemic inflammatory response by releasing DAMPs. However, it is necessary to assess the efficacy of this drug in more than one mouse strain because of strain variables with respect to radiation response.

The cells in the bone marrow and gastrointestinal (GI) tract are the most sensitive to radiation damage, because these cells are highly proliferative and have a low sensitivity threshold to radiation, caused by fast cellular turnover (1, 44, 45). Along with hematopoietic injury, GI-ARS is a serious injury that causes various clinical manifestations, from those occurring in the mouth to those in the colon and rectum. Patients with GI-ARS suffer not only from malnutrition due to reduced digestive capacity, but also from excessive inflammation and bacterial overgrowth after radiation exposure (46, 47). The co-appearance of GI damage with hematopoietic injury increases the mortality rate even at low radiation doses (48, 49). Therefore, it is necessary to investigate whether PLAG also attenuates the signs and symptoms of GI injury after radiation exposure.

In conclusion, the administration of PLAG significantly improved gamma-ray-induced mortality and body weight loss in BALB/c mice. Moreover, it effectively attenuated radiation-associated hematopoietic injuries, including neutropenia, thrombocytopenia and anemia. Together, these findings demonstrate that PLAG has therapeutic potential as a radiation countermeasure for the improvement of survivability and treatment of the hematopoietic sub-syndrome of ARS.

ACKNOWLEDGMENTS

PLAG has been granted Orphan Drug Designation by the U.S. Food and Drug Administration (FDA) for the treatment of acute radiation syndrome under the name of EC-18. Corresponding author, JWH, declares that the research was conducted in the absence of any commercial or financial relationships that could be constructed as a potential conflict of interest. Author, K-YS, is a president of Enzychem Lifesciences. The remaining authors are employees of Enzychem Lifesciences.

REFERENCES

1.

Heslet L, Bay C, Nepper-Christensen S. Acute radiation syndrome (ARS) - treatment of the reduced host defense. Int J Gen Med 2012; 5:105–15. Google Scholar

2.

Anno GH, Baum SJ, Withers HR, Young RW. Symptomatology of acute radiation effects in humans after exposure to doses of 0.5–30 Gy. Health Phys 1989; 56:821–38. Google Scholar

3.

Mettler FA, Jr., Gus'kova AK, Gusev I. Health effects in those with acute radiation sickness from the Chernobyl accident. Health Phys 2007; 93:462–9. Google Scholar

4.

Christensen DM, Iddins CJ, Parrillo SJ, Glassman ES, Goans RE. Management of ionizing radiation injuries and illnesses, part 4: acute radiation syndrome. J Am Osteopath Assoc 2014; 114:702–11. Google Scholar

5.

Singh VK, Seed TM. A review of radiation countermeasures focusing on injury-specific medicinals and regulatory approval status: part I. Radiation sub-syndromes, animal models and FDA-approved countermeasures. Int J Radiat Biol 2017; 93:851–69. Google Scholar

6.

Singh VK, Newman VL, Berg AN, MacVittie TJ. Animal models for acute radiation syndrome drug discovery. Expert Opin Drug Discov 2015; 10:497–517. Google Scholar

7.

Singh VK, Romaine PL, Newman VL. Biologics as countermeasures for acute radiation syndrome: where are we now? Expert Opin Biol Ther 2015; 15:465–71. Google Scholar

8.

Farese AM, MacVittie TJ. Filgrastim for the treatment of hematopoietic acute radiation syndrome. Drugs Today (Barc) 2015; 51:537–48. Google Scholar

9.

Kiang JG, Zhai M, Bolduc DL, Smith, JT, Anderson MN, Ho C, et al. Combined therapy of pegylated G-CSF and Alxn4100TPO improves survival and mitigates acute radiation syndrome after whole-body ionizing irradiation alone and followed by wound trauma. Radiat Res 2017; 188:476–90. Google Scholar

10.

Singh VK, Newman VL, Seed TM. Colony-stimulating factors for the treatment of the hematopoietic component of the acute radiation syndrome (H-ARS): a review. Cytokine 2015; 71:22–37. Google Scholar

11.

Farese AM, Bennett AW, Gibbs AM, Hankey KG, Prado K, Jackson W 3rd , et al. Efficacy of Neulasta or Neupogen on H-ARS and GI-ARS mortality and hematopoietic recovery in nonhuman primates after 10 Gy irradiation with 2.5% bone-marrow sparing. Health Phys 2019; 116:339–53. Google Scholar

12.

Singh VK, Seed TM. An update on sargramostim for treatment of acute radiation syndrome. Drugs Today (Barc) 2018; 54:679–93. Google Scholar

13.

Adams TG, Sumner LE, Casagrande R. Estimating risk of hematopoietic acute radiation syndrome in children. Health Phys 2017; 113:452–57. Google Scholar

14.

Hu S. Linking doses with clinical scores of hematopoietic acute radiation syndrome. Health Phys 2016; 111:337–47. Google Scholar

15.

Singh VK, Hauer-Jensen M. Gamma-Tocotrienol as a promising countermeasure for acute radiation syndrome: current status. Int J Mol Sci 2016; 17. Google Scholar

16.

Boada Burutaran M, Guadagna R, Grille S, Stevenazzi M, Guillermo C, Diaz L. Results of high-risk neutropenia therapy of hematology-oncology patients in a university hospital in Uruguay. Rev Bras Hematol Hemoter 2015; 37:28–33. Google Scholar

17.

Swinkels M, Rijkers M, Voorberg J, Vidarsson G, Leebeek FWG, Jansen AJG. Emerging concepts in immune thrombocytopenia. Front Immunol 2018; 9:880. Google Scholar

18.

Williams JP, Brown SL, Georges GE, Hauer-Jensen M, Hill RP, Huser AK, et al. Animal models for medical countermeasures to radiation exposure. Radiat Res 2010; 173:557–78. Google Scholar

19.

Yang HO, Park JS, Cho SH, Yoon JY, Kim MG, Jhon GJ, et al. Stimulatory effects of monoacetyldiglycerides on hematopoiesis. Biol Pharm Bull 2004; 27:1121–5. Google Scholar

20.

Jeong J, Kim YJ, Yoon SY, Yong-Jae Kim, Joo Heon Kim, Ki-Young Sohn, et al. PLAG (1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol) modulates eosinophil chemotaxis by regulating CCL26 expression from epithelial cells. PLoS One 2016; 11:e0151758. Google Scholar

21.

Jeong J, Kim Y-J, Lee DY, Moon BG, Sohn KY, Yoon SY, et al. 1-Palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) attenuates gemcitabine-induced neutrophil extravasation. Cell Biosci 2019; 9:4. Google Scholar

22.

Oh D, Kim MH, Song TJ, Cho CJ, Nam K, Cho MK, et al. 1-Pamitoyl-2-linoleoyl-3-acetyl-rac-glycerol may reduce incidence of gemcitabine-induced neutropenia: a pilot case-controlled study. World J Oncol 2015; 6:410–15. Google Scholar

23.

Yoo N, Lee HR, Shin SH, Sohn KY, Kim HJ, Han YH, et al. PLAG (1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol) augments the therapeutic effect of pegfilgrastim on gemcitabine-induced neutropenia. Cancer Lett 2016; 377:25–31. Google Scholar

24.

Kendall GM, Muirhead CR, MacGibbon BH, O'Hagan JA, Conquest AJ, Goodill AA, et al. Mortality and occupational exposure to radiation: first analysis of the National Registry for Radiation Workers. BMJ 1992; 304:220–5. Google Scholar

25.

Gluzman-Poltorak Z, Vainstein V, Basile LA. Association of hematological nadirs and survival in a nonhuman primate model of hematopoietic syndrome of acute radiation syndrome. Radiat Res 2015; 184:226–30. Google Scholar

26.

Slichter SJ. Relationship between platelet count and bleeding risk in thrombocytopenic patients. Transfus Med Rev 2004; 18:153–67. Google Scholar

27.

Stasi R. How to approach thrombocytopenia. Hematology Am Soc Hematol Educ Program 2012; 2012:191–7. Google Scholar

28.

Chow L, Aslam R, Speck ER, Kim M, Cridland N, Webster ML, et al. A murine model of severe immune thrombocytopenia is induced by antibody- and CD8+ T cell-mediated responses that are differentially sensitive to therapy. Blood 2010; 115:1247–53. Google Scholar

29.

Groopman JE, Itri LM. Chemotherapy-induced anemia in adults: incidence and treatment. J Natl Cancer Inst 1999; 91:1616–34. Google Scholar

30.

Izaks GJ, Westendorp RG, Knook DL. The definition of anemia in older persons. JAMA 1999; 281:1714–7. Google Scholar

31.

Singh VK, Romaine PL, Seed TM. Medical countermeasures for radiation exposure and related injuries: characterization of medicines, FDA-approval status and inclusion into the Strategic National Stockpile. Health Phys 2015; 108:607–30. Google Scholar

32.

Cerezo L, Macia I Garau M. Acute radiation syndrome and Fukushima: a watershed moment? Rep Pract Oncol Radiother 2012; 17:1–3. Google Scholar

33.

Waterman G, Kase K, Orion I, Broisman A, Milstein O. Selective shielding of bone marrow: an approach to protecting humans from external gamma radiation. Health Phys 2017; 113:195–208. Google Scholar

34.

Kim YJ, Shin JM, Shin SH, Kim JH, Sohn KY, Kim HJ, et al. 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol ameliorates arthritic joints through reducing neutrophil infiltration mediated by IL-6/ STAT3 and MIP-2 activation. Oncotarget 2017; 8:96636–48. Google Scholar

35.

Ko YE, Yoon SY, Ly SY, Kim JH, Sohn KY, Kim JW. 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) reduces hepatic injury in concanavalin A-treated mice. J Cell Biochem 2018; 119:1392–405. Google Scholar

36.

Yoon SY, Kang HB, Ko YE, Shin SH, Kim YJ, Sohn KY, et al. 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (EC-18) modulates Th2 immunity through attenuation of IL-4 expression. Immune Netw 2015; 15:100–9. Google Scholar

37.

Lee HR, Yoo N, Kim JH, Sohn KY, Kim HJ, Kim MH, et al. The therapeutic effect of PLAG against oral mucositis in hamster and mouse model. Front Oncol 2016; 6:209. Google Scholar

38.

Ward JF. DNA damage produced by ionizing radiation in mammalian cells: identities, mechanisms of formation, and reparability. Prog Nucleic Acid Res Mol Biol 1988; 35:95–125. Google Scholar

39.

Lomax ME, Folkes LK, O'Neill P. Biological consequences of radiation-induced DNA damage: relevance to radiotherapy. Clin Oncol (R Coll Radiol) 2013; 25:578–85. Google Scholar

40.

Schaue D, Micewicz ED, Ratikan JA, Xie MW, Cheng G, McBride WH. Radiation and inflammation. Semin Radiat Oncol 2015; 25:4–10. Google Scholar

41.

Yoritsune E, Furuse M, Kuwabara H, Miyata T, Nonoguchi N, Kawabata S, et al. Inflammation as well as angiogenesis may participate in the pathophysiology of brain radiation necrosis. J Radiat Res 2014; 55:803–11. Google Scholar

42.

Lai H, Singh NP. Single- and double-strand DNA breaks in rat brain cells after acute exposure to radiofrequency electromagnetic radiation. Int J Radiat Biol 1996; 69:513–21. Google Scholar

43.

Okayasu R, Suetomi K, Yu Y, Silver A, Bedford JS, Cox R, et al. A deficiency in DNA repair and DNA-PKcs expression in the radiosensitive BALB/c mouse. Cancer Res 2000; 60:4342–5. Google Scholar

44.

Gourmelon P, Benderitter M, Bertho JM, Huet C, Gorin NC, De Revel P. European consensus on the medical management of acute radiation syndrome and analysis of the radiation accidents in Belgium and Senegal. Health Phys 2010; 98:825–32. Google Scholar

45.

Teo MT, Sebag-Montefiore D, Donnellan CF. Prevention and management of radiation-induced late gastrointestinal toxicity. Clin Oncol (R Coll Radiol) 2015; 27:656–67. Google Scholar

46.

Shadad AK, Sullivan FJ, Martin JD, Egan LJ. Gastrointestinal radiation injury: prevention and treatment. World J Gastroenterol 2013; 19:199–208. Google Scholar

47.

Shadad AK, Sullivan FJ, Martin JD, Egan LJ. Gastrointestinal radiation injury: symptoms, risk factors and mechanisms. World J Gastroenterol 2013; 19:185–98. Google Scholar

48.

Deng W, Kimura Y, Gududuru V, Wu W, Balogh A, Szabo E, et al. Mitigation of the hematopoietic and gastrointestinal acute radiation syndrome by octadecenyl thiophosphate, a small molecule mimic of lysophosphatidic acid. Radiat Res 2015; 183:465–75. Google Scholar

49.

Patil R, Szabo E, Fells JI, Balogh A, Lim KG, Fujiwara Y, et al. Combined mitigation of the gastrointestinal and hematopoietic acute radiation syndromes by an LPA2 receptor-specific nonlipid agonist. Chem Biol 2015; 22:206–16. Google Scholar
©2019 by Radiation Research Society. All rights of reproduction in any form reserved.
Yong-Jae Kim, Jinseon Jeong, Su-Hyun Shin, Do Young Lee, Ki-Young Sohn, Sun Young Yoon, and Jae Wha Kim "Mitigating Effects of 1-Palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) on Hematopoietic Acute Radiation Syndrome after Total-Body Ionizing Irradiation in Mice," Radiation Research 192(6), 602-611, (26 September 2019). https://doi.org/10.1667/RR15440.1
Received: 22 May 2019; Accepted: 14 August 2019; Published: 26 September 2019
Back to Top