Open Access
How to translate text using browser tools
29 July 2022 Mitigation of Multi-Organ Radiation Injury with ACE2 Agonist Diminazene Aceturate
Tracy Gasperetti, Guru Prasad Sharma, Anne C. Frei, Lauren Pierce, Dana Veley, Nathan Szalewski, Jayashree Narayanan, Brian L. Fish, Heather A. Himburg
Author Affiliations +
Abstract

The renin-angiotensin system (RAS) is known to regulate the pathogenesis of radiation-induced injury as inhibitors of the RAS enzyme angiotensin converting enzyme (ACE) have established function as mitigators of multi-organ radiation injury. To further elucidate the role of RAS signaling during both the acute and delayed syndromes of radiation exposure, we have evaluated whether pharmacologic modulation of alternate RAS enzyme angiotensin converting enzyme 2 (ACE2) reduces the pathogenesis of multi-organ radiation-induced injuries. Here, we demonstrate pharmacologic ACE2 activation with the small molecule ACE2 agonist diminazene aceturate (DIZE) improves survival in rat models of both hematologic acute radiation syndrome (H-ARS) and multi-organ delayed effects of acute radiation exposure (DEARE). In the H-ARS model, DIZE treatment increased 30-day survival by 30% compared to vehicle control rats after a LD50/30 total-body irradiation (TBI) dose of 7.75 Gy. In the mitigation of DEARE, ACE2 agonism with DIZE increased median survival by 30 days, reduced breathing rate, and reduced blood urea nitrogen (BUN) levels compared to control rats after partial-body irradiation (PBI) of 13.5 Gy. DIZE treatment was observed to have systemic effects which may explain the multi-organ benefits observed including mobilization of hematopoietic progenitors to the circulation and a reduction in plasma TGF-beta levels. These data suggest the ACE2 enzyme plays a critical role in the RAS-mediated pathogenesis of radiation injury and may be a potential therapeutic target for the development of medical countermeasures for acute radiation exposure.

INTRODUCTION

In the event of a radiological or nuclear disaster, victims of radiation exposure will experience dose-dependent toxicity to multiple organ systems. In the early days to weeks after acute radiation exposure, medical treatment must focus on the critical need to mitigate life-threatening injury to the hematopoietic and gastrointestinal systems. In accordance with this need, the FDA has approved the use of filgrastim (Neupogen®), pegfilgrastim (Neulasta®), sargramostim (Leukine®), and romiplostim (Nplate®) for treatment of the hematopoietic acute radiation syndrome (H-ARS). However, survivors of ARS are likely to experience the delayed effects of acute radiation exposure (DEARE) which encompass late morbidities and progressive functional decline of multiple organ systems. While there are no FDA-approved treatments to date for mitigation of DEARE, pharmacologic modulation of the renin-angiotensin system (RAS) has therapeutic potential.

RAS is known to regulate the response of several organ systems to radiation injury as pharmacologic inhibition of the key RAS enzyme angiotensin converting enzyme (ACE) is known to reduce hematopoietic, cardiac, respiratory, and renal damage after radiation exposure (19). In the regulation of vascular homeostasis, RAS signaling enzymes ACE and ACE2 are thought to act in opposition with ACE promoting vasoconstriction and ACE2 mediating vasorelaxation. ACE catalyzes the conversion of hormone peptide angiotensin I (Ang I) to the vasoconstrictor product angiotensin II (Ang II). Conversely, ACE2 counterbalances ACE-mediated vasoconstriction via reduction of Ang II to Ang (1–7) (10). The local RAS system and specifically the balance of ACE/ACE2 activity is thought to play a key role in the pathogenesis of many disease processes and has most recently been implicated in the pathogenesis of respiratory distress syndrome in COVID-19 patients (11, 12).

Here we have evaluated whether pharmacologic agonism of ACE2 mitigates early and late radiation injury in a similar manner to ACE inhibition. To pharmacologically promote ACE2 signaling in vivo, we have administered the small molecule ACE2 agonist diminazene aceturate (DIZE). DIZE is a well-tolerated anti-parasitic medication commonly used for the treatment of trypanosomiasis in livestock (1316). Previously, DIZE treatment prior to radiation exposure has been shown to reduce acute radiation-induced renal injury in rats by increasing ACE2/Mas signaling (17). In this study, we have evaluated the efficacy of DIZE as a mitigator of radiation injury using two adult rat models: total-body irradiation (TBI) for H-ARS and partial-body irradiation (PBI) for lung and renal DEARE.

METHODS

Animals

All studies described were performed in accordance with an approved Institutional Animal Care and Use Committee protocol. WAG/RijCmcr rats were bred and maintained in a barrier facility at our institution. Two weeks prior to irradiation, rats were switched to a moderate antioxidant diet (Teklad Global 2018 diet) which is more representative of antioxidant levels in a human diet (18). All rats were provided reverse osmosis hyper-chlorinated water ad libitum.

Diminazene Aceturate (DIZE) Administration

Diminazene aceturate (DIZE) was purchased from Sigma-Aldrich (St. Louis, MO, D7770) and MedChemExpress (HY-12404, Monmouth Junction, NJ) and reconstituted in sterile water at a stock concentration of 10 mg/ml. Rats were weighed immediately prior to drug administration and administered 15 mg/kg of the 10 mg/ml DIZE solution via subcutaneous injection (19). Vehicle control rats received subcutaneous injections of an equivalent volume of sterile water.

Total-Body Irradiation Model

Adult female WAG/RijCmcr rats (11–12 weeks old) were exposed to 7.25–7.75 Gy TBI. All rats were irradiated without the use of anesthetics by being placed in a plastic jig and the entire body was exposed using a XRAD 320 kV orthovoltage x-ray system (Precision X-Ray, Madison, CT). The X-ray system was operated at 320 kVp and 13 mAs with a half-value layer of 1.4 mm copper and a dose rate of 169 cGy/min. For survival studies to day 30 postirradiation, rats were exposed to a total dose of 7.75 Gy TBI which is the LD50/30 in this model. Rats were dosed daily subcutaneously starting at 24 h postirradiation with either 15 mg/kg DIZE or sterile water until the end of the study. To examine the recovery of peripheral blood and bone marrow compartment, a subset of rats was exposed to a myelosuppressive dose of 7.25 Gy TBI which is approximately the LD10/30 in this model. At 24 h postirradiation, 15 mg/kg DIZE or sterile water were dosed daily subcutaneously until harvest on day 10 postirradiation.

Partial-Body Irradiation Model

Adult female WAG/RijCmcr rats were exposed to 13.5 Gy PBI with bone marrow shielding to one hind-limb (X-RAD 320 Precision, 320 kVp; 169 cGy/min). This dose is based on the established LD50/120 radiation dose for female all-cause mortality through lung-DEARE in this strain (20). All rats received supportive care after irradiation consisting of antibiotics (enrofloxacin ∼10 mg/kg/day) in the drinking water from days 2–14, subcutaneous saline (40 ml/kg) days 2–10, and powdered diet days 35–70. Rats were dosed subcutaneously starting at 24 h after PBI with either 15 mg/kg DIZE or sterile water (3×/week [Monday-Wednesday-Friday (MWF)] for the duration of the study. Daily health checks were performed by trained staff. Additionally, body weight, breathing rate and renal function were monitored to assess the health of the rats. Rats were followed for survival through gastrointestinal, hematopoietic, lung and renal injury up to 190 days.

Mobilization Assay

To assess for the mobilization of bone marrow progenitor cells to the peripheral blood, normal non-irradiated adult male rats were treated with a single subcutaneous dose of either 15 mg/kg DIZE, 0.55 mg/kg Neulasta® (Amgen, Thousand Oaks, California), or vehicle control. The dose for Neulasta® was selected based on prior studies in our lab using PEGylated G-CSF (21).

At 6 h after injection, the peripheral blood was collected, a complete blood cell count (CBC) was performed, and the mononuclear fraction was isolated by Ficoll-Paque separation. Mononuclear cells were then analyzed by flow cytometry and CFU assay as described below.

Blood Collection

Blood was collected from anesthetized rats via the jugular vein. Briefly, rats were restrained with one hand by positioning the forelimbs in the caudodorsal direction with the thumb and middle finger. The head of the rat was secured with the index finger, creating a straight line along the ventral side of the rat. A 23-gauge needle attached with a syringe was carefully inserted into either the right or left external jugular vein and blood was collected. Syringes were coated with EDTA to prevent clotting for all CBC blood draws.

Peripheral Blood Analysis

Complete blood counts were determined using a Heska Element 5 Veterinary Hematology Analyzer.

Blood Urea Nitrogen (BUN) Determination

BUN levels were monitored starting at day 90 postirradiation and every 30 days thereafter as using a urease-nitroprusside colorimetric assay as described previously (22, 23). Irradiated rats with BUN>120 mg/dl were euthanized and given a value of 120 mg/dl to account for attrition, since such rats were previously confirmed to have severe and irreversible renal damage (9, 24).

Breathing Rate Measurement

Breathing rates were measured using a MouseOx Plus Pulse Oximeter (Starr Life Sciences Corp) in accordance with manufacturer's instructions. Rats were acclimated to the recording processing on the day prior to recording. After calibration for movement, the breathing rate was measured over a five-minute period and averaged to obtain the reported rate for each rat.

Bronchoalveolar Lavage Fluid (BALF) Collection

BALF was collected immediately after euthanasia as previously described (25). Lavage fluid was centrifuged at 300g for 5 min at 4°C to remove cellular components and the resulting BALF supernatant was 10× concentrated (Millipore Amicon Ultra-4 Centrifugal Filter Unit) prior to ELISA analysis.

ACE and ACE2 Activity Assays

ACE and ACE2 activity were determined in whole lung digests with commercially purchased kits: ACE2 (MAK377, Sigma Aldrich) and ACE (CS0002, Sigma-Aldrich).

Bone Marrow Isolation

Bone marrow was harvested from the femurs of euthanized rats. Each femur was carefully excised from the animal and flushed with PBS containing 10% FBS and 1% penicillin-streptomycin using a 1 ml syringe with a 25G needle, making sure to flush from both ends to ensure all cells were removed. Cells were pelleted by centrifugation and the red blood cell fraction was removed with ACK lysis buffer. Total viable cells were determined using a Countess Automated Cell Counter Hemocytometer C10227 (Invitrogen) with trypan blue exclusion.

RT-PCR Assay

Lung total RNA was isolated using RNeasy Micro Kit (74004, Qiagen) and quantified on Nanodrop 2000 Spectrophotometer (Model: 840-274200, Thermo Fisher Scientific). C- DNA was prepared using the high-capacity RNA-to-cDNA Kit (4387406, Thermo Fisher Scientific) and used to examine respective transcript expression using TaqMan Gene Expression Assay primer probes (4331182, Thermo Fisher Scientific). The rat primer probes used were ACE (Rn00561094_m1), ACE2 (Rn01416293_m1), AT2R (Rn00560677_s1), MASR (Rn00562673_s1) and the reference GAPDH (Rn01775763_m1). Expression of target genes was normalized to GAPDH. The relative expression of the gene was calculated with respect to control (0 Gy) and presented as fold change using the 2-ΔΔCT method.

Flow Cytometry Analysis

Bone marrow or peripheral blood mononuclear cells were stained with the following fluorescence-conjugated antibodies used to differentiate rat hematopoietic stem and progenitor cell populations (26): APC-CD45 (17-0461-82, Thermo Fisher Scientific), PE/Cy7-CD71 (204424, Biolegend), BV510-CD90 (202535, Biolegend), FITC-Ox82 (205103, Biolegend).

Samples were acquired live on MACSQuant 10 Analyzer Flow Cytometer (Miltenyi) with 7-AAD used to exclude dead cells. Data was analyzed using FlowJo software version 10.0 (BD Life Sciences).

Colony Forming Unit (CFU) Assay

CFU assays were conducted using MethoCult (R3774, Stem Cell Technologies, Vancouver, BC, Canada) media in six-well Smartdish (Stem Cell Technologies) plates at a seeding density of 10,000 bone marrow mononuclear cells per well or the mononuclear cell fraction isolated by Ficoll-Paque (Stem Cell Technologies) gradient separation from 0.5 ml of whole blood.

Cytokine Estimation by ELISA

Rat TGF-beta (BMS623-3, Invitrogen) and IL-10 (ab100764, Abcam) cytokine concentrations were determined per the manufacturer's instruction.

Statistical Analyses

Statistical analysis was performed using the GraphPad Prism version 9 (GraphPad Software, Inc.). P values for comparison of survival curves were determined using Log-rank test.

Statistical analysis of multiple groups and time points was conducted using 2-way ANOVA with Tukey's multiple comparison test. A one-way ANOVA with Tukey's multiple comparison was used to analyze multiple groups at a single time point. P values of <0.05 were considered statistically significant.

RESULTS

H-ARS

We first evaluated survival during H-ARS in adult female rats exposed to 7.75 Gy TBI and treated with daily subcutaneous injection of water vehicle or 15 mg/kg DIZE starting at 24 h after TBI. DIZE treated rats had significantly improved survival to day 30 after TBI vs. water treated controls (Fig. 1A, P = 0.046). We then assessed recovery of peripheral blood complete blood counts (CBCs) and the bone marrow compartment in irradiated and DIZE treated rats at day 10 after a myelosuppressive dose of 7.25 Gy. DIZE treatment increased total white blood cell counts (WBCs) compared to vehicle treated controls (Fig. 1B). In the bone marrow, DIZE improved recovery of total cellularity (Fig. 1C). In rats, the CD71-CD45RA-OX82+CD90+ population is enriched for hematopoietic stem and progenitor cells (26). At day 10 after 7.25 Gy, this population is significantly decreased compared to non-irradiated controls at day 10 (Fig. 1D). There was a non-significant trend towards an increase in the percentage of CD71-CD45RA-OX82+CD90+ cells in DIZE treated bone marrow vs. vehicle control (Fig. 1D); however, the absolute numbers of CD71-CD45RA-OX82+CD90+ cells were significantly increased in DIZE-treated rats (Fig. 1E). Consistent with improved bone marrow cellularity, myeloid colony forming potential (CFU-GM) was also significantly increased in DIZE versus vehicle-treated rats (Fig. 1F).

FIG. 1

ACE2 agonism with DIZE mitigates H-ARS. Panel A. Survival after 7.75 Gy total-body irradiation (TBI) in adult female WAG/RijCmcr rats treated with daily subcutaneous injection of DIZE (N = 27) or water vehicle (N = 29). P value for log-rank analysis: P = 0.046. Panels B–F: Day 10 assessment of a cohort of female rats after 7.25 Gy TBI and treatment with DIZE (N = 9) or water vehicle (N = 9). Five age-matched non-irradiated animals were included as controls. Panel B: Scatter plot of complete blood cell counts. Panel C: Scatter plot of total cells per femur. Panel D: Left, representative FACS plots depicting the percentage of bone marrow cells within the hematopoietic stem cell-enriched population of CD71-OX82+CD90+ cells. Right, scatter plots of percent CD71-OX82+CD90+. Panel E: Calculated total CD71-OX82+CD90+ cells per femur. Panel F: Colony forming units per 10,000 whole bone marrow cells. Solid bar indicates median, significance values determined by ANOVA with multiple comparison tests.

img-z4-1_325.jpg

Multi-Organ Late Effects

Next, to see if ACE2 agonism with DIZE mitigates radiation pneumonitis, we employed a rat partial body irradiation model with shielding to one hind limb. This model reproduces the major sequelae of radiation injury to the bone marrow, gastrointestinal tract, lungs, and kidney (8, 27, 28). Pneumonitis manifests at days 50–120 postirradiation as evidenced by morbidity due to increased respiration, pleural effusion, and weight loss. Rats which survive past 120 days eventually succumb to renal failure evidenced by elevated blood urea nitrogen (BUN) levels. In this model, we have previously demonstrated lisinopril mediated ACE inhibition supports multi-organ recovery after radiation injury (8, 28). Here, we evaluated whether long-term administration of DIZE could similarly mitigate radiation injury. DIZE or water vehicle were administered subcutaneously starting at 24 h after PBI and was administered three times weekly (MWF) until endpoint. DIZE treatment after 13.5 Gy PBI in adult female rats significantly improved survival through 190 days (Fig. 2A). DIZE treatment increased median survival from 147 days in control-irradiated rats to 177 days. Long-term DIZE treatment had no effect on body weight compared to irradiated controls (Fig. 2B). Consistent with the observed improvement in survival during pneumonitis, DIZE treatment significantly reduced the radiation-induced elevation in breathing rate at day 70 (Fig. 2C). As part of our IACUC protocol, BUN levels are assessed monthly in all irradiated rats starting at day 90 and rats in renal failure (BUN > 120 mg/dL) are humanely euthanized. DIZE treatment significantly reduced BUN levels at days 90, 120 and 150 relative to 13.5 Gy PBI alone rats (Fig. 2D).

FIG. 2

ACE2 agonism with DIZE mitigates multi-organ DEARE. Panel A: Survival after 13.5 Gy partial-body irradiation (PBI) in adult female WAG/RijCmcr rats treated with subcutaneous injection of DIZE (N = 15) or water vehicle (N = 16) 3×/week (MWF). Gray region from days 50–120 indicates period of risk for morbidity due to lung-DEARE, while beige region from days 120–200 indicates period of risk for morbidity due to kidney-DEARE. Median survival for each group is indicated with a dashed line. P value for log-rank analysis: P = 0.0005. Panel B: Corresponding mean percentage body weight change for each treatment group. Panel C: Breathing rates at day 70 after 13.5 Gy TBI and treatment with DIZE (N = 4) or water vehicle (N = 4). Five age-matched non-irradiated animals were included as controls. Panel D: Blood urea nitrogen (BUN) levels at days 90-180 after 13.5 Gy TBI and treatment with DIZE (N = 9) or water vehicle (N = 7). Solid bar indicates median, significance values determined by ANOVA with multiple comparison tests.

img-z5-1_325.jpg

Modulation of ACE/ACE2 Activity

We assessed the effects of DIZE treatment on modulation of ACE/ACE2 transcription and protein activity in the whole lung after 13.5 Gy PBI. At day 42 after 13.5 Gy PBI, ACE2 transcript levels in the lung are decreased relative to non-irradiated rats (Fig. 3A). Although DIZE treatment did not significantly alter ACE2 expression compared to vehicle treatment, DIZE treatment did decrease transcription of ACE relative to vehicle treatment. Additionally, DIZE increased mRNA expression of two receptors known to act as downstream targets of ACE2 enzymatic products: AT2R and MasR (Fig. 3A). At day 70 after 13.5 Gy PBI, we then assessed enzymatic activity levels of ACE and ACE2 in whole lungs (Fig. 3B). Although DIZE treatment did not alter ACE activity, DIZE increased ACE2 activity by more than twofold and decreased the ratio of ACE/ACE2 activity (Fig. 3B).

FIG. 3

DIZE increases ACE2 Signaling. Panel A: RT-PCR analysis performed on RNA isolated from whole lung lysates at day 42 after 13.5 Gy PBI in female rats treated with DIZE (N = 5) or water vehicle (N = 4). Data represent means ± SEM. P values determined by two-way ANOVA analysis with multiple comparison test. Panel B: Enzymatic ACE activity (left), ACE2 activity (middle), and the calculated ratio of ACE:ACE2 activity (right) in whole lung lysates at day 70 after 13.5 Gy PBI in female rats treated with DIZE (N = 5) or water vehicle (N = 4). Solid bar indicates median, significance values determined by ANOVA with multiple comparison tests.

img-z6-1_325.jpg

Modulation of TGF-Beta and IL-10 Levels

At days 42 and 70 after 13.5 Gy PBI, we measured cytokines levels of two proteins known to affect radiation-induced lung injury: TGF-beta and IL-10. Both factors were measured in the plasma and bronchoalveolar lavage fluid (BALF). Levels of the pro-fibrotic factor TGF-beta were elevated in the plasma and BALF at both time points after 13.5 Gy PBI compared to non-irradiated control levels (Fig. 4A). However, treatment with DIZE reduced both plasma and BALF TGF-b levels vs. 13.5 Gy PBI alone (Fig. 4A). Conversely, anti-inflammatory cytokine IL-10 was reduced after 13.5 Gy PBI in the BALF at day 42 compared to non-irradiated levels (Fig. 4B). Treatment with DIZE increased levels of IL-10 in both the BALF and plasma at the day 70 timepoint (Fig. 4B).

FIG. 4

DIZE regulates systemic TGF-beta and IL-10 levels. ELISA analysis was used to determine protein concentrations at days 42 and 70 after 13.5 Gy PBI in female rats treated with subcutaneous injection of DIZE (N = 5) or water vehicle (N = 4). A. TGF-beta levels in the bronchoalveolar fluid (BALF, left) and plasma (right). B. IL-10 levels in the BALF (left) and plasma (right). Four age-matched non-irradiated animals were included as controls. Significance values determined by two-way ANOVA with multiple comparison tests.

img-z7-1_325.jpg

Bone Marrow Mobilization

Prior studies have suggested agonism of ACE2/Ang (1-7) signaling promotes mobilization of bone marrow progenitors with vasoreparative function (19, 29). Here, we evaluated whether a single subcutaneous injection of 15 mg/kg DIZE in non-irradiated rats induces bone marrow mobilization at 6 h. Data were compared to mobilization with a single dose of 0.55 mg/kg pegylated-G-CSF (Neulasta). Treatment with either DIZE or Neulasta increased total white blood cell and neutrophil counts at 6 h (Fig. 5A) and the percentage of hematopoietic progenitors present in the circulation (Fig. 5B). Both DIZE treatment and Neulasta also increased the numbers of circulating cells capable of forming hematopoietic colonies in a colony forming unit assay (Fig. 5C).

FIG. 5

DIZE treatment induces mobilization of bone marrow progenitor cells. At 6 h after a single injection of either vehicle (n = 5), Neulasta (N = 5; 0.55 mg/kg), DIZE (n = 5, 15 mg/kg) in non-irradiated adult rats, peripheral blood was assessed for the following: Panel A: Complete blood cell counts including WBC differential. Panel B: Circulating progenitor cell populations as identified by FACS analysis (representative plots left) of CD71-OX82+ myeloid progenitors and CD71-OX82+CD90+ HSCs. C. Colony forming units per 1 ml whole blood. Solid bar indicates median, significance values determined by ANOVA with multiple comparison tests.

img-z8-1_325.jpg

DISCUSSION

Diminazene aceturate (DIZE) is an aromatic diamidine compound developed in 1955 as anti-Trypanosoma medication for domestic livestock under the trade name Berenil (13). In addition to its anti-protozoal activity, DIZE has been observed to reduce immune activation and systemic inflammatory cytokine production (30). More recently, molecular docking and enzymatic activity assays have shown DIZE strongly enhances ACE2 catalytic activity (31). Based on this finding, DIZE has been employed as a pharmacologic ACE2 agonist in animal models of hypertension (19, 3234) and cardiac dysfunction (3537). Here, we have observed DIZE administration after radiation injury promotes multi-organ recovery without any overt tissue toxicity. To our knowledge, this is the first study to demonstrate DIZE mitigates lethal radiation injury in either an H-ARS or DEARE model. Moreover, the observation that DIZE can improve multi-organ survival in acute and delayed settings is a key translational finding as this may be a potential therapeutic mechanism to promote multi-organ recovery after radiation injury.

The ACE2/Ang (1-7) signaling pathway is known to play an important role in bone marrow regeneration after radiation or chemotherapy injury as administration of Ang (1-7) accelerates bone marrow recovery (3841). Recently, ACE2 has been shown to be increased in highly purified human hematopoietic stem cell populations (42). Direct agonism of Ang (1-7) signaling promotes HSC expansion in vitro (39). Consistent with these findings, we observed DIZE-mediated ACE2 agonism significantly increased recovery of total bone marrow cellularity and functional colony forming capacity at both early and late time points after radiation injury. Additionally, we observed a survival benefit in DIZE-mediated ACE2 agonism in rats after 7.75 Gy TBI.

In addition to mediating an improved survival after hematopoietic injury, we observed that DIZE treatment also mitigated late morbidities due to pulmonary and renal failure. After 13.5 Gy PBI, rats in this model manifest morbidity from radiation pneumonitis characterized by rapid respiration (25). Here, we observed DIZE treatment reduces pneumonitis related morbidities and mitigates the increase in respiration rate at day 70 after PBI seen in irradiated control rats. DIZE treatment also significantly delays morbidity from renal failure as evidenced by significantly reduced BUN levels and increased overall median survival. These data are consistent with a prior report which demonstrated prophylactic DIZE treatment reduced acute renal damage after radiation injury (17).

Our observations with ACE2 agonism after radiation injury bear many similarities to ACE inhibitor treatment in radiation injury models. For example, ACE inhibitors have been shown to increase survival during H-ARS in rodent and pig models (1, 43, 44). Additionally, extensive studies by our group in the rat PBI model have demonstrated ACE inhibitors mitigate lung and kidney DEARE (8, 25, 28). It is possible ACE2 agonism may be acting to negate downstream ACE signaling by reducing the available amount of Angiotensin II (Ang II) to bind and activate Angiotensin II type 1 receptor (AT1R). Indeed, like ACE inhibitor treatment, DIZE treatment is known to lower blood pressure in animal models (45).

However, several findings from this study suggest ACE2 agonism with DIZE elicits a unique downstream signaling mechanism summarized in Fig. 6. As anticipated, DIZE induces high levels of ACE2 activity in the lung, but we also observed increased transcription of RAS receptors AT2R and MasR. Both AT2R and MasR are thought to directly counterbalance AT1R receptor function by promoting vasodilation, reducing inflammation, and reducing fibrosis (46). The necessity for AT2R and MasR in mediating the observed effects will be the focus of future studies. In keeping with a systemic suppression of pro-fibrotic signaling, we observed a dramatic reduction in both circulating and BALF TGF-beta levels in DIZE-treated rats. This is a key finding as TGF-beta is a master regulator in the initiation of radiation fibrosis. It is also consistent with prior experimental lung and cardiac fibrosis models where DIZE treatment was observed to decrease fibrotic progression and lower TGF-beta expression (47, 48). Increased levels of IL-10 in DIZE treated rats suggests DIZE treatment reduces inflammation as IL-10 is an anti-inflammatory mediator and is considered a candidate therapy for treatment of idiopathic pulmonary fibrosis (49).

FIG. 6

ACE2 signaling pathway. ACE2 metabolizes the conversion of angiotensin peptide Ang II to Ang (1-7). This both reduces the amount of Ang II available to bind the AT1R and increases signaling of Ang (1-7) through AT2R and MasR. Diminazene aceturate (DIZE) treatment increased ACE2 activity and increases AT2R and MasR transcription. We hypothesize this mechanism promoted the observed increase in survival by suppressing pro-inflammatory and pro-fibrotic signaling cascades.

img-z9-1_325.jpg

One of the most interesting DIZE-mediated effects observed was the mobilization of hematopoietic progenitors into the circulation. At 6 h after DIZE administration, we saw a marked increase in circulating hematopoietic progenitors which was similar to Neulasta-mediated mobilization. Our observation is consistent with DIZE treatment in a murine model of myocardial infarction, in which DIZE was observed to increase circulating CD90+ cells thought to act as reparative endothelial progenitor cells (19). Additionally, agonism of Ang (1-7) signaling has also been observed to increase circulating hematopoietic progenitor cell populations (39, 50). Moreover, in diabetic patients the loss of vasoreparative function has been linked to decreased ACE2 expression in CD34+ hematopoietic progenitors as lentiviral-mediated ACE2 gene transfer in CD34+ cells derived from diabetic patients rescued vasoreparative function in hind-limb ischemia models (29). Together these studies suggest DIZE may promote multi-organ recovery via activation of hematopoietic progenitors with vasoreparative function.

In conclusion, we have observed DIZE treatment promotes multi-organ and long-term recovery after radiation injury. Since ACE2 is likely to promote recovery via mechanisms distinct from ACE pathway inhibition, it is likely the combination of ACE inhibitors and ACE2 agonists may have additive effects as radiation mitigators. In the mitigation of multi-organ injury after 13.5 Gy PBI, we observe that median survival in DIZE-treated female rats is 177 days. While this survival benefit is less than previously observed in female rats treated with ACE inhibitor lisinopril (213 days, unpublished data), we have not fully optimized the dose or schedule of either DIZE or lisinopril. Mitigation will also need to be validated in male rats as there are established sex differences in radiation sensitivity to DEARE (20). For this reason, future studies are warranted to optimize the potential combination of ACE inhibitors with ACE2 agonists for mitigation of multi-organ injury.

ACKNOWLEDGMENTS

This work was supported by funding from NIH/NIAID U01AI133594, U01AI138331, the MCW Department of Radiation Oncology, and the MCW Cancer Center.

REFERENCES

1.

McCart EA, Lee YH, Jha J, Mungunsukh O, Rittase WB, Summers TA, Jr., et al. Delayed captopril administration mitigates hematopoietic injury in a murine model of total body irradiation. Sci Rep 2019; 9, 2198. Google Scholar

2.

Medhora M, Gao F, Jacobs ER, Moulder JE. Radiation damage to the lung: mitigation by angiotensin-converting enzyme (ACE) inhibitors. Respirology 2012; 17, 66–71. Google Scholar

3.

Mungunsukh O, George J, McCart EA, Snow AL, Mattapallil JJ, Mog SR, et al. Captopril reduces lung inflammation and accelerated senescence in response to thoracic radiation in mice. J Radiat Res 2021; 62, 236–48. Google Scholar

4.

van der Veen SJ, Ghobadi G, de Boer RA, Faber H, Cannon MV, Nagle PW, et al. ACE inhibition attenuates radiation-induced cardiopulmonary damage. Radiother Oncol 2015; 114, 96–103. Google Scholar

5.

Ryu S, Kolozsvary A, Jenrow KA, Brown SL, Kim JH. Mitigation of radiation-induced optic neuropathy in rats by ACE inhibitor ramipril: importance of ramipril dose and treatment time. J Neurooncol 2007; 82, 119–24. Google Scholar

6.

Kim JH, Brown SL, Kolozsvary A, Jenrow KA, Ryu S, Rosenblum ML, et al. Modification of radiation injury by ramipril, inhibitor of angiotensin-converting enzyme, on optic neuropathy in the rat. Radiat Res 2004; 161, 137–42. Google Scholar

7.

Moulder JE, Fish BL, Cohen EP. Treatment of radiation nephropathy with ACE inhibitors and AII type-1 and type-2 receptor antagonists. Curr Pharm Des 2007; 13, 1317–25. Google Scholar

8.

Fish BL, Gao F, Narayanan J, Bergom C, Jacobs ER, Cohen EP, et al. Combined hydration and antibiotics with lisinopril to mitigate acute and delayed high-dose radiation injuries to multiple organs. Health Phys 2016; 111, 410–9. Google Scholar

9.

Moulder JE, Fish BL, Cohen EP. Treatment of radiation nephropathy with ACE inhibitors. Int J Radiat Oncol Biol Phys 1993; 27, 93–9. Google Scholar

10.

Santos RAS, Sampaio WO, Alzamora AC, Motta-Santos D, Alenina N, Bader M, et al. The ACE2/Angiotensin-(1-7)/MAS axis of the renin-angiotensin system: Focus on angiotensin-(1-7). Physiol Rev 2018; 98, 505–53. Google Scholar

11.

Pagliaro P, Penna C. ACE/ACE2 Ratio: A key also in 2019 Coronavirus Disease (Covid-19)? Front Med (Lausanne) 2020; 7, 335. Google Scholar

12.

Wosten-van Asperen RM, Lutter R, Specht PA, Moll GN, van Woensel JB, van der Loos CM, et al. Acute respiratory distress syndrome leads to reduced ratio of ACE/ACE2 activities and is prevented by angiotensin-(1-7) or an angiotensin II receptor antagonist. J Pathol 2011; 225, 618–27. Google Scholar

13.

Peregrine AS, Mamman M. Pharmacology of diminazene: a review. Acta Trop 1993; 54, 185–203. Google Scholar

14.

Abaru DE, Liwo DA, Isakina D, Okori EE. Retrospective long-term study of effects of berenil by follow-up of patients treated since 1965. Tropenmed Parasitol 1984; 35, 148–50. Google Scholar

15.

Rajapaksha IG, Mak KY, Huang P, Burrell LM, Angus PW, Herath CB. The small molecule drug diminazene aceturate inhibits liver injury and biliary fibrosis in mice. Sci Rep 2018; 8, 10175. Google Scholar

16.

Kuriakose S, Uzonna JE. Diminazene aceturate (Berenil), a new use for an old compound? Int Immunopharmacol 2014; 21, 342–5. Google Scholar

17.

Hasan HF, Elgazzar EM, Mostafa DM. Diminazene aceturate extenuate the renal deleterious consequences of angiotensin-II induced by gamma-irradiation through boosting ACE2 signaling cascade. Life Sci 2020; 253, 117749. Google Scholar

18.

Moulder JE, Fish BL, Cohen EP, Flowers JB. Medhora M, Effects of Diet on Late Radiation Injuries in Rats. Health Phys 2019; 116, 566–70. Google Scholar

19.

Qi Y, Zhang J, Cole-Jeffrey CT, Shenoy V, Espejo A, Hanna M, et al. Diminazene aceturate enhances angiotensin-converting enzyme 2 activity and attenuates ischemia-induced cardiac pathophysiology. Hypertension 2013; 62, 746–52. Google Scholar

20.

Fish BL, MacVittie TJ, Gao F, Narayanan J, Gasperetti T, Scholler D, et al. Rat Models of Partial- body Irradiation with Bone Marrow-sparing (Leg-out PBI) Designed for FDA Approval of Countermeasures for Mitigation of Acute and Delayed Injuries by Radiation. Health Phys 2021; 121, 419– 33. Google Scholar

21.

Gasperetti T, Miller T, Gao F, Narayanan J, Jacobs ER, Szabo A, et al. Polypharmacy to Mitigate Acute and Delayed Radiation Syndromes. Front Pharmacol 2021; 12, 634477. Google Scholar

22.

Cohen EP. Predictors of mortality in patients undergoing hemodialysis. N Engl J Med 1994; 330, 573; author reply 73-4. Google Scholar

23.

Medhora M, Gao F, Wu Q, Molthen RC, Jacobs ER, Moulder JE, et al. Model development and use of ACE inhibitors for preclinical mitigation of radiation-induced injury to multiple organs. Radiat Res 2014; 182, 545–55. Google Scholar

24.

Moulder JE, Cohen EP, Fish BL, Hill P. Prophylaxis of bone marrow transplant nephropathy with captopril, an inhibitor of angiotensin-converting enzyme. Radiat Res 1993; 136, 404–7. Google Scholar

25.

Sharma GP, Fish BL, Frei AC, Narayanan J, Gasperetti T, Scholler D, et al. Pharmacological ACE- inhibition mitigates radiation-induced pneumonitis by suppressing ACE-expressing lung myeloid cells. Int J Radiat Oncol Biol Phys 2022. Google Scholar

26.

Crook K, Hunt SV. Enrichment of early fetal-liver hemopoietic stem cells of the rat using monoclonal antibodies against the transferrin receptor, Thy-1, and MRC-OX82. Dev Immunol 1996; 4, 235–46. Google Scholar

27.

Fish BL, MacVittie TJ, Szabo A, Moulder JE, Medhora M. WAG/ RijCmcr rat models for injuries to multiple organs by single high dose ionizing radiation: similarities to nonhuman primates (NHP). Int J Radiat Biol 2020; 96, 81–92. Google Scholar

28.

Medhora M, Gao F, Gasperetti T, Narayanan J, Khan AH, Jacobs ER, et al. Delayed effects of acute radiation exposure (Deare) in juvenile and old rats: mitigation by Lisinopril. Health Phys 2019; 116, 529–45. Google Scholar

29.

Joshi S, Montes de Oca I, Maghrabi A, Lopez-Yang C, Quiroz-Olvera J, Garcia CA, et al. ACE2 gene transfer ameliorates vasoreparative dysfunction in CD34+ cells derived from diabetic older adults. Clin Sci (Lond) 2021; 135, 367–85. Google Scholar

30.

Kuriakose S, Muleme HM, Onyilagha C, Singh R, Jia P, Uzonna JE. Diminazene aceturate (Berenil) modulates the host cellular and inflammatory responses to Trypanosoma congolense infection. PLoS One 2012; 7, e48696. Google Scholar

31.

Kulemina LV, Ostrov DA. Prediction of off-target effects on angiotensin-converting enzyme 2. J Biomol Screen 2011; 16, 878–85. Google Scholar

32.

Bennion DM, Haltigan EA, Irwin AJ, Donnangelo LL, Regenhardt RW, Pioquinto DJ, et al. Activation of the neuroprotective angiotensin-converting enzyme 2 in rat ischemic stroke. Hypertension 2015; 66, 141–8. Google Scholar

33.

Veit F, Weissmann N. Angiotensin-converting enzyme 2 activation for treatment of pulmonary hypertension. Am J Respir Crit Care Med 2013; 187, 569–71. Google Scholar

34.

Chen IC, Lin JY, Liu YC, Chai CY, Yeh JL, Hsu JH, et al. Angiotensin-converting enzyme 2 Activator ameliorates severe pulmonary hypertension in a rat model of left pneumonectomy combined with VEGF inhibition. Front Med (Lausanne) 2021; 8, 619133. Google Scholar

35.

De Maria ML, Araújo LD, Fraga-Silva RA, Pereira LA, Ribeiro HJ, Menezes GB, et al. Anti-hypertensive effects of diminazene aceturate: An angiotensin-converting enzyme 2 activator in rats. Protein Pept Lett 2016; 23, 9–16. Google Scholar

36.

Shenoy V, Gjymishka A, Jarajapu YP, Qi Y, Afzal A, Rigatto K, et al. Diminazene attenuates pulmonary hypertension and improves angiogenic progenitor cell functions in experimental models. Am J Respir Crit Care Med 2013; 187, 648–57. Google Scholar

37.

Velkoska E, Patel SK, Griggs K, Pickering RJ, Tikellis C, Burrell LM, Short-term treatment with diminazene aceturate ameliorates the reduction in kidney ACE2 activity in rats with subtotal nephrectomy. PLoS One 2015; 10, e0118758. Google Scholar

38.

Rodgers KE, Espinoza T, Roda N, Meeks CJ, Hill C, Louie SG, et al. Accelerated hematopoietic recovery with angiotensin-(1-7) after total body radiation. Int J Radiat Biol 2012; 88, 466–76. Google Scholar

39.

Heringer-Walther S, Eckert K, Schumacher SM, Uharek L, Wulf-Goldenberg A, Gembardt F, et al. Angiotensin-(1-7) stimulates hematopoietic progenitor cells in vitro and in vivo. Haematologica 2009; 94, 857–60. Google Scholar

40.

Ellefson DD, diZerega GS, Espinoza T, Roda N, Maldonado S, Rodgers KE. Synergistic effects of co-administration of angiotensin 1-7 and Neupogen on hematopoietic recovery in mice. Cancer Chemother Pharmacol 2004; 53, 15–24. Google Scholar

41.

Rodgers K, Xiong S, DiZerega GS. Effect of angiotensin II and angiotensin(1-7) on hematopoietic recovery after intravenous chemotherapy. Cancer Chemother Pharmacol 2003; 51, 97–106. Google Scholar

42.

Ropa J, Cooper S, Capitano ML, Van't Hof W, Broxmeyer HE, Human hematopoietic stem, progenitor, and immune cells respond ex vivo to SARS-CoV-2 spike protein. Stem Cell Rev Rep 2021; 17, 253–65. Google Scholar

43.

Rittase WB, McCart EA, Muir JM, Bouten RM, Slaven JE, Mungunsukh O, et al. Effects of captopril against radiation injuries in the Gottingen minipig model of hematopoietic-acute radiation syndrome. PLoS One 2021; 16, e0256208. Google Scholar

44.

Saunders J, Niswander LM, McGrath KE, Koniski A, Catherman SC, Ture SK, et al. Long-acting PGE2 and Lisinopril Mitigate H-ARS. Radiat Res 2021; 196, 284–96. Google Scholar

45.

Sartorio CL, Pimentel EB, Dos Santos RL, Rouver WN, Mill JG, Acute hypotensive effect of diminazene aceturate in spontaneously hypertensive rats: role of NO and Mas receptor. Clin Exp Pharmacol Physiol 2020; 47, 1723–30. Google Scholar

46.

Matavelli LC, Siragy HM, AT2 receptor activities and pathophysiological implications. J Cardiovasc Pharmacol 2015; 65, 226–32. Google Scholar

47.

Macedo LM, Souza AP, De Maria ML, Borges CL, Soares CM, Pedrino GR, et al. Cardioprotective effects of diminazene aceturate in pressure-overloaded rat hearts. Life Sci 2016; 155, 63–9. Google Scholar

48.

Prata LO, Rodrigues CR, Martins JM, Vasconcelos PC, Oliveira FM, Ferreira AJ, et al. Original research: ACE2 activator associated with physical exercise potentiates the reduction of pulmonary fibrosis. Exp Biol Med (Maywood) 2017; 242, 8–21. Google Scholar

49.

Shamskhou EA, Kratochvil MJ, Orcholski ME, Nagy N, Kaber G, Steen E, et al. Hydrogel-based delivery of Il-10 improves treatment of bleomycin-induced lung fibrosis in mice. Biomaterials 2019; 203, 52–62. Google Scholar

50.

Cole-Jeffrey CT, Pepine CJ, Katovich MJ, Grant MB, Raizada MK, Hazra S, Beneficial effects of Angiotensin-(1-7) on CD34+ cells from patients with heart failure. J Cardiovasc Pharmacol 2018; 71, 155–59. Google Scholar
©2022 by Radiation Research Society. All rights of reproduction in any form reserved.
Tracy Gasperetti, Guru Prasad Sharma, Anne C. Frei, Lauren Pierce, Dana Veley, Nathan Szalewski, Jayashree Narayanan, Brian L. Fish, and Heather A. Himburg "Mitigation of Multi-Organ Radiation Injury with ACE2 Agonist Diminazene Aceturate," Radiation Research 198(4), 325-335, (29 July 2022). https://doi.org/10.1667/RADE-22-00055.1
Received: 8 March 2022; Accepted: 17 June 2022; Published: 29 July 2022
Back to Top