Recovery from radiation-induced (RI) myelosuppression depends on hematopoietic stem and progenitor cell survival and the active proliferation/differentiation process, which requires early cytokine support. Single cytokine or late-acting growth factor therapy has proved to be inefficient in ensuring reconstitution after severe RI damage. This work was aimed at evaluating the in vivo survival effect of combinations of early-acting cytokines whose antiapoptotic activity has been demonstrated in vitro: stem cell factor (SCF [S]), FMS-like tyrosine kinase 3 ligand (FLT-3 ligand [F]), thrombopoietin (TPO [T]), interleukin-3 (IL-3 [3]), and stromal derived factor-1 (SDF-1). B6D2F1 mice underwent total body irradiation at 8 Gy cesium Cs 137 γ radiation (ie, lethal dose 90% at 30 days) and were treated soon after irradiation, at 2 hours and at 24 hours, with recombinant murine cytokines, each given intraperitoneally at 50 μg/kg per injection. All treatments induced 30-day survival rates significantly higher than control (survival rate, 8.3%). 4F (SFT3) and 5F (4F + SDF-1) were the most efficient combinations (81.2% and 87.5%, respectively), which was better than 3F (SFT, 50%), TPO alone (58.3%), and SDF-1 alone (29.2%) and also better than 4F given at 10 μg/kg per injection (4F10, 45.8%) or as a 50 μg/kg single injection at 2 hours (4Fs, 62.5%). Despite delayed death occurring mainly from day 150 on and possible long-term hematopoiesis impairment, half the 30-day protective effects of 4F and 5F were preserved at 300 days. Our results show that short- and long-term survival after irradiation depends on appropriate multiple cytokine combinations and at optimal concentrations. The proposal is made that an emergency cytokine regimen could be applied to nuclear accident victims as part of longer cytokine treatment, cell therapy, or both.

The pancytopenia that occurs after irradiation is mainly the consequence of early death of hematopoietic stem and progenitor cells (HSPCs), and its severity and duration depend on the level of bone marrow (BM) cell loss.1 Numerous studies have shown the efficacy of hematopoietic growth factors (HGFs) in stimulating neutrophil and platelet recovery in animals and humans after myeloablation.2 Granulocyte-colony-stimulating factor (G-CSF), granulocyte macrophage-colony-stimulating factor (GM-CSF), interleukin-3 (IL-3), IL-11, and thrombopoietin (TPO) are among the most efficient cytokines.3 These molecules have primarily been evaluated as monotherapy, but the simultaneous or sequential administration of 2 complementary cytokines has resulted in better patterns of hematologic recovery.4-7 As a clinical application, victims of the criticality nuclear accident in Tokai-mura, Japan, recently were given G-CSF, megakaryocyte growth and development factor (MGDF), and erythropoietin in combination.8 In most preclinical trials, HGF administration starts the day after myeloablative injury. The duration of treatment covers at least 15 days until neutrophil or platelet recovery. Prolonged administration stimulates HSPCs to cycle, proliferate, and differentiate, but it may also induce adverse effects such as proinflammatory and immunogenic activity.9 This led us to consider short-term treatment schedules. Thus far, few cytokines have been tested as early and acute therapy. IL-1 seemed to be a pivotal hematopoietic cytokine in the therapy for radiation injury before adverse effects were established.10 G-CSF injected into mice 2 hours after irradiation as a single injection has been shown to induce a good level of protection against 30-day mortality.11 Unfortunately, this factor was unsuccessful in restoring thrombopoiesis in myeloablated nonhuman primates. Despite a notable level of platelet recovery observed in sublethally or lethally irradiated mice, TPO did not prevent a profound neutropenic nadir after lethal irradiation, which may be involved in the observed lethality.12 

Apoptosis has been shown to play a major role in HSPC death soon after irradiation in vitro13 and in vivo.14,15 If a subset of highly radioresistant stem cells recapitulates long-term hematopoiesis16,17 (a notion supported by the eventual autologous reconstitution in accidentally irradiated victims despite bone marrow allotransplantation), the preservation of residual cytokine-sensitive HSPCs from radiation-induced (RI) apoptosis would be critical for myeloablated host survival.15 In particular, the mitigation of morbidity and mortality caused by the initial neutropenia and thrombocytopenia requires the preservation and stimulation of less immature HSPCs. In vitro studies dealing with RI apoptosis of CD34+ cells have shown that early management is necessary to maintain cell clonogenicity. In fact, cytokines active in the early stages of hematopoiesis preserve CD34+ cells, provided they are added in combination soon after irradiation.13 Thus far, we found the best antiapoptotic combination to be stem cell factor (SCF) + FMS-like tyrosine kinase 3 ligand (FLT-3 ligand) + TPO + IL-3 (4F). In addition, stromal derived factor-1 (SDF-1) chemokine has been reported to be an antiapoptotic factor toward HSPC in vitro18,19and to participate in host defenses against DNA damage.20All these data prompted us to use cytokine combinations as an early treatment of RI injury.

The purpose of our study was to assess the capacity of antiapoptotic cytokines to rescue mice from radiation when cytokines are given as an early treatment to counteract cell death, especially potentially lethal lesions, as a combination to ensure multilineage protection, and as a short-term treatment to avert adverse effects. In this study, we report the therapeutic benefit for lethally irradiated mice of the administration at 2 hours and 24 hours after irradiation of multiple cytokine combinations involving SCF, FLT-3 ligand, TPO, IL-3, and SDF-1 in comparison with TPO or SDF-1 alone. The highest 30-day survival rates were obtained with 4F and 4F + SDF-1 (5F). Long-term follow-up revealed delayed death, and treated irradiated mice displayed a reduced long-term hematopoietic potential compared with normal mice. However, approximately half the protection provided by the cytokines at 30 days was still evident 300 days after total body irradiation (TBI). To dissect the mechanism of action of 4F, we used a nonhuman primate model and showed that 4F administration 2 hours after TBI prevented BM CD34+ cells from apoptosis and that 4F did not stimulate CD34+ cells to cycle within the first 24 hours after in vitro irradiation.

Animals

Housing and experiments were approved by the French Army Ethics Committee in accordance with European rules and regulations.

Mouse model

Six-week-old male B6D2F1 mice (Charles River, Les Oncins, France) were quarantined for 7 days before irradiation at the Centre de Transfusion Sanguine des Armées (Clamart, France). Mice were housed in groups of 6 per cage in rooms equipped with a reverse-filtered air barrier and received commercial food and acidified water ad libitum.

Nonhuman primate model

Adult male cynomolgus monkeys (Macaca fascicularis) weighing 8 kg each were housed at the Centre de Recherches du Service de Santé des Armées (CRSSA)–accredited animal facility in individual cages in holding rooms equipped with a reverse-filtered air barrier (10 air changes per hour, 100% fresh air) and full-spectrum light (8:00 am to 8:00 pm) and conditioned to 23°C with a relative humidity of 60%. Animals were fed commercial primate chow supplemented with fresh fruit and were provided tap water ad libitum.

Irradiation

In vitro irradiation.

Cells were irradiated at 2.5 Gy using a cobalt Co 60 γ source (dose rate, 100 cGy/min). This dose appears relevant because it is greater than the D0 (dose required to reduce cell survival to 37% of its initial value) value, estimated to be approximately 1 Gy for HSPC.21 

Total body irradiation.

Mice placed in ventilated polycarbonate canisters underwent TBI with a single 8 Gy dose using a cesium Cs 137 gamma source (dose rate, 10 Gy/min). Dosimetry was performed using an ionization chamber and thermoluminescent aluminum oxide dosimeters. Survival curves previously performed established that 8 Gy constitutes the 90% ± 5% lethal dose at 30 days.

Anesthetized monkeys (n = 6) placed in a restraining chair were exposed to global unilateral front irradiation with a 60Co γ source at a total midline tissue dose of 5 Gy (measured in air at the anterior iliac crest level) with a dose rate of 20 cGy/min. Dosimetry was performed using ionizing chambers and thermoluminescent dosimeters.

Cytokines

Recombinant murine SCF (S), FLT-3 ligand (F), TPO (T), IL-3 (3), and SDF-1α were used (R&D Systems, Abingdon, United Kingdom) for the mouse study. Recombinant human (rh) TPO, rhSCF, rhFLT-3 ligand, and rhIL-3 (R&D Systems) were used for the monkey study.

CD34+ cells

Cell collection.

For the in vitro study, baboon-mobilized peripheral blood (PB) CD34+ cells were collected using leukapheresis as previously described.22 

Cell culture.

Within 30 minutes of irradiation or sham irradiation, cells were washed and resuspended (1 × 105 cells/mL) in serum-free medium H00 (RTM-Mabio, Tourcoing, France). Unirradiated and irradiated cells were then incubated with the 4F cytokine combination (FLT-3 ligand + TPO + SCF + IL-3; 50 ng/mL each) in 6-well plates at 37°C in a humidified incubator with 5% CO2 in air. Cells were analyzed after 24 hours and 3 days of incubation relative to fresh control cells.

Cell cycle analysis.

DNA and nuclear protein Ki67 (only present during the G1and S/G2/M phases of the cell cycle) cell content were evaluated using flow cytometry.23 Briefly, after CD34+ cell labeling with antihuman CD34, clone 566, monoclonal antibody (mAb) conjugated to phycoerythrin (PE) (BD Biosciences, Le Pont de Claix, France), and cell fixation in phosphate-buffered saline (PBS)–formaldehyde 1%, cells were permeabilized in PBS-Triton 0.1% (30 minutes on ice). After two washes, cells were incubated with anti-Ki-67 MIB-1 mAb conjugated to fluorescein isothiocyanate (FITC; Beckman Coulter, Villepinte, France) for 1 hour at 4°C. Finally cells were washed and resuspended in PBS 7-aminoactinomycin (7 AAD, 0.5μg/mL; Sigma, St Louis, MO). Flow cytometric analysis was performed within 1hour, and G0, G1, S/G2/M cell contents were determined (FCS Express; De novo Software, Toronto, ON, Canada).

In vivo apoptosis evaluation.

Current methodologies used for evaluating in vitro cell death are inefficient for in vivo evaluation because apoptotic cells are progressively eliminated by phagocytes. It is, therefore, necessary to identify a pertinent early marker. We chose to measure the mitochondrial transmembrane potential (MTP) at the CD34+cell level by flow cytometry using 3, 3′ dihexyloxacarbocianine iodide (DiOC6; Interchim, Montluçon, France).24MTP is known to be increased during the early phase of apoptosis,25 before the final step of depolarization. Humeral BM was harvested after TBI, and mononuclear cells were isolated by cell density separation and were DiOC6/CD34 double-labeled in 2 steps. First, cells were incubated with 25 nM/L DiOC6 for 15 minutes at 37°C in PBS. Cells were then put on ice to prevent the leak of DiOC6 and were then stained with anti-CD34 mAb for 1 hour. Nucleated cells were selected for analysis using 0.5 μg/mL LDS751 (Interchim) addition. DiOC6 uptake was expressed in terms of mean fluorescence intensity (MFI). For each sample, DiOC6 uptake in the granulocyte population was used as an internal standard. Thus, the ratio of DiOC6 uptake in CD34+cells to DiOC6 uptake in granulocytes was evaluated before and 6 hours after TBI. The increase in the ratio accounts for a status of hyperpolarization of the mitochondrial membrane, which is considered to be an early marker of apoptosis.

Study groups and treatments

Mouse study.

Twenty-four mice were randomly assigned to each group. Seven treatments were evaluated using S, F, T, 3, and SDF-1 alone or in combination: TPO, SDF-1, 3F (S + F + T), 4F (S + F + T + 3), and 5F (4F + SDF-1) were given 2 hours and 24 hours after TBI. Each cytokine was administered at 50 μg/kg for each administration. The last 2 treatments were 4Fsingle (4F given only at 2 hours after TBI, each cytokine given at 50 μg/kg) and 4F10 (4F given at 2 hours and 24 hours after TBI, each cytokine given at 10 μg/kg). Preliminary experiments showed that 4F induced a similarly protective effect by either the intravenous or the intraperitoneal route. Cytokine solutions were prepared extemporaneously with pyrogen-free sterile Dulbecco phosphate-buffered solution containing 0.5% human serum albumin and injected intraperitoneally in a volume of 200 ± 20 μL (according to the animal's weight) at 2 hours and 24 hours after irradiation. Four control groups that received cytokine diluent were evaluated throughout the study; 4F and 5F evaluation was carried out twice.

Monkey study.

Six monkeys underwent 5 Gy γ irradiation. Three of them were treated with 4F at 2 hours after TBI (each cytokine was administered at 50 μg/kg intravenously), and 3 control animals received the cytokine diluent. Humeral BM of control and treated animals was harvested before irradiation and 6 hours after TBI for the evaluation of apoptosis at the CD34+ cell level.

Survival follow-up

During the first 30 days, mice were observed daily to record survival. Weight and behavior were monitored. After 30 days, animals were observed 3 times a week. Autopsies were performed in long-term deceased animals. After 360 days, surviving mice were killed by cervical dislocation and were autopsied. Spleens and femurs were collected. Spleens were stained with formalin and weighed. BM was harvested, and total nucleated cell (TNC) counts were assessed, as were differential cell counts using cytospins (Shandon, Pittsburgh, PA) stained with May-Grünwald-Giemsa. BM clonogenicity (granulocyte macrophage-colony-forming unit [GM-CFU] and erythroid burst-forming unit [BFU-e]) was evaluated using a short-term methylcellulose assay (RTM Mabio). Briefly, 15 × 103/mL femoral BM TNCs were plated in triplicate in 6-well plates with recombinant murine SCF, FLT-3 ligand, IL-3, GM-CSF, and EPO (R&D Systems), and CFU scoring was performed at 10 days of culture.

Statistics

The mouse study was conducted over 4 experiments, and no significant difference in survival among the 4 control groups was observed (χ2 evaluation), allowing for a global statistical comparison. Survival curves were compared using Kaplan-Meier analysis, and statistical significance was determined between groups using χ2 analysis with Yates correction. Spleen weights, BM TNC, and clonogenicity were compared using the Mann-Whitney U test. The latter test was also used to analyze the effect of 4F on the cell cycle distribution of baboon BM-irradiated CD34+ cells versus unirradiated cells.

Short- term survival

The onset of lethality was delayed by 3 days in mice treated with 5F in comparison with the other 2 treatments and with the control group (11.5 days instead of 8 days). Thirty-day survival rates are presented in Figure 1A and Table1. The average survival rate of controls was 8.3%. All treated groups displayed 30-day survival rates significantly higher than those of the control group. Multiple cytokine treatments proved to be more efficient than single therapy. In fact, 5F and 4F afforded the best protection (survival rates, 87.5% and 81.25%, respectively) when given at 50 μg/kg 2 hours and 24 hours after TBI. Neither was statistically different from the other (P = .36). Both treatments were more efficient than TPO (58.3%), 3F (50%), 4F10 (45.8%), and SDF-1 (29.2%), as shown in Table 2, and 5F was better than 4Fsingle (62.5%). The benefit conferred by 2 injections of 4F rather than a single injection was not significant. Thus, the combination of cytokines administered appears crucial for efficacy.

Fig. 1.

Effect of 7 treatments with SCF (S), FLT-3 ligand (F), TPO (T), IL-3 (3), and SDF-1 after 8 Gy 127Cs γ irradiation on survival of B6D2F1 mice.

Cytokines TPO alone, SDF-1α alone, SFT (3F), SFT3 (4F), and SFT3 + SDF-1 (5F) were given at 50 μg/kg intraperitoneally 2 hours and 24 hours after TBI. 4F was also evaluated 2 hours after TBI at 50 μg/kg (4Fsingle also indicated as 4Fs) or 2 hours and 24 hours after TBI at 10 μg/kg (4F10). The data presented summarize the results of 4 experiments. (A) 30-day survival: any therapy was effective in prolonging short-term survival. (B) 300-day survival: Kaplan-Meier analysis was performed to analyze survival curves and showed a global statistical significance (P < .0001). Statistical significance was determined between groups using χ2analysis with Yates correction (see Table 2).

Fig. 1.

Effect of 7 treatments with SCF (S), FLT-3 ligand (F), TPO (T), IL-3 (3), and SDF-1 after 8 Gy 127Cs γ irradiation on survival of B6D2F1 mice.

Cytokines TPO alone, SDF-1α alone, SFT (3F), SFT3 (4F), and SFT3 + SDF-1 (5F) were given at 50 μg/kg intraperitoneally 2 hours and 24 hours after TBI. 4F was also evaluated 2 hours after TBI at 50 μg/kg (4Fsingle also indicated as 4Fs) or 2 hours and 24 hours after TBI at 10 μg/kg (4F10). The data presented summarize the results of 4 experiments. (A) 30-day survival: any therapy was effective in prolonging short-term survival. (B) 300-day survival: Kaplan-Meier analysis was performed to analyze survival curves and showed a global statistical significance (P < .0001). Statistical significance was determined between groups using χ2analysis with Yates correction (see Table 2).

Close modal
Table 1.

Survival percentages at 30 and 300 days after TBI for the different treatments

Survival, d Control
n = 120
Treatment
TPO
n = 24
4Fsingle
n = 24
4F
n = 48
5F
n = 48
4F 10
n = 24
3F
n = 24
SDF-1
n = 24
30  8.3  58.3  62.50  81.25  87.5  45.8  50.0  29.2 
300  3.3  29.2  41.7  50.0  43.8  8.3  20.8  12.5 
Survival, d Control
n = 120
Treatment
TPO
n = 24
4Fsingle
n = 24
4F
n = 48
5F
n = 48
4F 10
n = 24
3F
n = 24
SDF-1
n = 24
30  8.3  58.3  62.50  81.25  87.5  45.8  50.0  29.2 
300  3.3  29.2  41.7  50.0  43.8  8.3  20.8  12.5 

TPO or SDF-1 indicates each cytokine alone; 3F, SCF + FLT-3 ligand + TPO; 4F, 3F + IL-3; 5F, 4F + SDF-1. TPO, SDF-1, 3F, 4F, and 5F were given 2 hours and 24 hours after TBI. Each cytokine was administered at 50 μg/kg. 4Fsingle is 4F given only at 2 hours after TBI. Each cytokine was given at 50 μg/kg. 4F10 is 4F given at 2 hours and 24 hours after TBI. Each cytokine was given at 10 μg/kg. Control, irradiated controls treated with cytokine diluent. The number (n) of mice for each group is indicated. See Table 2 for statistical significance.

Table 2.

Levels of statistical significance between treatments regarding survival at 30 days, 300 days, or both

P Days 30 and 300 Day 30 only Day 300 only
< .0001  5F, 4F, 4Fs, and TPO > C  4F10 > C, 3F > C  —  
    5F and 4F > SDF-1   
< .001  —  5F > 3F, 5F > 4F10  4F10 > C  
      4F > 4F10 
< .01  —  5F > TPO  3F > C  
    4F > 3F  5F and 4F > SDF-1  
    4F > 4F10   
< .05  4Fs > SDF-1  5F > 4Fs, TPO > SDF-1  5F and 4F > 3F 
    SDF-1 > C  5F and 4Fs > 4F10  
    4F > TPO   
NS  5F and 4Fs = 4F  4Fs = 4F10  5F = 4Fs,  
  4Fs and 4F10 = 3F    4F10 and SDF-1 = C  
  4Fs, 4F10, and 3F = TPO    TPO = SDF-1  
  4F10 and 3F = SDF-1    5F and 4F = TPO 
P Days 30 and 300 Day 30 only Day 300 only
< .0001  5F, 4F, 4Fs, and TPO > C  4F10 > C, 3F > C  —  
    5F and 4F > SDF-1   
< .001  —  5F > 3F, 5F > 4F10  4F10 > C  
      4F > 4F10 
< .01  —  5F > TPO  3F > C  
    4F > 3F  5F and 4F > SDF-1  
    4F > 4F10   
< .05  4Fs > SDF-1  5F > 4Fs, TPO > SDF-1  5F and 4F > 3F 
    SDF-1 > C  5F and 4Fs > 4F10  
    4F > TPO   
NS  5F and 4Fs = 4F  4Fs = 4F10  5F = 4Fs,  
  4Fs and 4F10 = 3F    4F10 and SDF-1 = C  
  4Fs, 4F10, and 3F = TPO    TPO = SDF-1  
  4F10 and 3F = SDF-1    5F and 4F = TPO 

Statistical significance was determined between groups using χ2 analysis with Yates correction. 4Fs means 4Fsingle, which is 4F given only at 2 hours after TBI. Each cytokine was given at 50 μg/kg. 4F10 is 4F given at 2 hours and 24 hours after TBI. Each cytokine was given at 10 μg/kg. NS indicates not significant; —, no results.

In vitro effect of 4F on irradiated HSPC cell cycle distribution

Freshly isolated baboon-mobilized PB CD34+ cells are mainly in the G0/G1 phases of the cell cycle, with only a reduced subset in the S and G2M phases (67.8% ± 5.8%, 29.9% ± 4.8%, and 2.4% ± 1.2%, respectively), as shown in Table 3 and Figure 2. Unirradiated (unir) CD34+ cells incubated for 24 hours in serum-free medium in the presence of 4F exhibited an important reduction of G0cell subset and an increase of G1 and S/G2/M cell subsets (40.2% ± 12.6%, 39.5% ± 6%, 19.8% ± 7.6%, respectively; P < .05 for G0, G1, and S/G2/M) (day 1 unir vs day 0). In contrast to unirradiated cells, in vitro–irradiated cells incubated with 4F for 24 hours showed the maintenance of their initial cell cycle distribution (P > .05 for G0 and S/G2/M, day 0 vs day 1, 2.5 Gy) (P < .05 for G0, G1, and S/G2/M, day 1 unir vs day1, 2.5 Gy). From day 3 of culture with 4F, irradiated CD34+ cells were stimulated to cycle as did unirradiated cells. At 3 days, the relative cell cycle distributions were similar even though, in terms of absolute cell number, unirradiated cells exhibited a greater expansion rate (K4F = 2.6 ± 0.7) than irradiated cells (K4Firrad = 0.55 ± 0.34). At 7 days, the respective cell amplification rates were K4F = 12.5 ± 0.7 and K4Firrad = 2.9 ± 0.6.

Table 3.

In vitro effect of 4F on CD34+ cell cycle distribution

% CD34+ cells Day 0 Day 1 Day 3
Unirradiated 2.5 Gy Unirradiated 2.5 Gy
G0  67.8 ± 5.8  40.2 ± 12.6  78.4 ± 7.9  4.6 ± 4.3  7.8 ± 3.7 
G1  29.9 ± 4.8  39.5 ± 6.0  14.3 ± 6.8  55.3 ± 14.7  53.1 ± 9.5 
S/G2/M  2.4 ± 1.2  19.8 ± 7.6  6.5 ± 3.2  40.0 ± 12.2  37.3 ± 7.0 
% CD34+ cells Day 0 Day 1 Day 3
Unirradiated 2.5 Gy Unirradiated 2.5 Gy
G0  67.8 ± 5.8  40.2 ± 12.6  78.4 ± 7.9  4.6 ± 4.3  7.8 ± 3.7 
G1  29.9 ± 4.8  39.5 ± 6.0  14.3 ± 6.8  55.3 ± 14.7  53.1 ± 9.5 
S/G2/M  2.4 ± 1.2  19.8 ± 7.6  6.5 ± 3.2  40.0 ± 12.2  37.3 ± 7.0 

Unirradiated (unir) and in vitro–irradiated baboon-mobilized PB CD34+ cells were incubated for 7 days in serum-free medium in the presence of 4F, and cell cycle analysis was performed by flow cytometry after 1 or 3 days of culture. Results from 4 different baboons are expressed as mean ± SEM. All intergroup comparisons, made using the Mann-Whitney U test, are significant (P < .05) except the following: day 0 versus day 1, 2.5 Gy; day 3 unir versus day 3, 2.5 Gy regarding both G0 and S/G2/M; day 3 unir versus day 3, 2.5 Gy regarding G1.

Fig. 2.

In vitro effect of 4F on CD34+ cell cycle distribution.

Unirradiated and in vitro–irradiated (2.5 Gy) baboon-mobilized PB CD34+ cells were incubated for 7 days in serum-free medium in the presence of 4F. Cell cycle analysis was performed by flow cytometry before and after 1 and 3 days of culture. G0, G1, and S/G2/M cell content was determined using triple labeling with anti-CD34-PE mAb, anti-Ki67-FITC mAb, and 7 AAD. Cell cycle phenotype of gated CD34+ cells is shown as a biparametric dot plot. x-axis indicates DNA/7 AAD cell content; y-axis, nuclear protein Ki67 cell content. Lower left quadrant, G0CD34+ cells; upper left quadrant, G1CD34+ cells; upper right quadrant, S/G2/M CD34+ cells. Results are taken from tests on one representative baboon.

Fig. 2.

In vitro effect of 4F on CD34+ cell cycle distribution.

Unirradiated and in vitro–irradiated (2.5 Gy) baboon-mobilized PB CD34+ cells were incubated for 7 days in serum-free medium in the presence of 4F. Cell cycle analysis was performed by flow cytometry before and after 1 and 3 days of culture. G0, G1, and S/G2/M cell content was determined using triple labeling with anti-CD34-PE mAb, anti-Ki67-FITC mAb, and 7 AAD. Cell cycle phenotype of gated CD34+ cells is shown as a biparametric dot plot. x-axis indicates DNA/7 AAD cell content; y-axis, nuclear protein Ki67 cell content. Lower left quadrant, G0CD34+ cells; upper left quadrant, G1CD34+ cells; upper right quadrant, S/G2/M CD34+ cells. Results are taken from tests on one representative baboon.

Close modal

4F administration leads to a reduction of in vivo RI apoptosis at the HSPC level

Monkey BM CD34+ cells harvested 6 hours after 5 Gy TBI exhibited a notable increase in mitochondrial transmembrane potential compared with their own initial value (Figure3). In contrast, this hyperpolarization did not occur in animals given 4F 2 hours after TBI.

Fig. 3.

Early single administration of 4F to irradiated monkeys reduced RI apoptosis at the BM CD34+ cell level.

The mitochondrial transmembrane potential (MTP) was evaluated ex vivo in BM CD34+ cells 6 hours (H6) after TBI (4 hours after 4F injection) using DiOC6 uptake expressed as mean fluorescence intensity (MFI). Results are expressed as mean indices (± SEM) using an internal standard. Each index is the ratio of DiOC6 uptake MFI in CD34+ cells to DiOC6 uptake MFI in granulocytes from a given BM sample. Unlike 4F-treated animals (n = 3), untreated animals (n = 3) exhibited a pattern of MTP hyperpolarization at the BM CD34+ cell level, which is an early marker of apoptosis. H0 indicates before irradiation.

Fig. 3.

Early single administration of 4F to irradiated monkeys reduced RI apoptosis at the BM CD34+ cell level.

The mitochondrial transmembrane potential (MTP) was evaluated ex vivo in BM CD34+ cells 6 hours (H6) after TBI (4 hours after 4F injection) using DiOC6 uptake expressed as mean fluorescence intensity (MFI). Results are expressed as mean indices (± SEM) using an internal standard. Each index is the ratio of DiOC6 uptake MFI in CD34+ cells to DiOC6 uptake MFI in granulocytes from a given BM sample. Unlike 4F-treated animals (n = 3), untreated animals (n = 3) exhibited a pattern of MTP hyperpolarization at the BM CD34+ cell level, which is an early marker of apoptosis. H0 indicates before irradiation.

Close modal

Long-term survival

Data regarding 300-day survival are presented in Figure 1B and Table 1. Delayed death occurred in all treated groups and in the few surviving controls, mainly from day 150 after TBI, well beyond the classical period of RI hematologic death. In contrast, age-matched unirradiated control (UC) animals experienced no deaths in 360 days of follow-up. Despite delayed death in irradiated animals, all treatments except 4F10 and SDF-1 induced significantly improved long-term survival rates. Multiple cytokine combinations remained the most effective treatments, resulting in survival rates of 43.8% (5F), 50% (4F), and 41.7% (4Fsingle) versus 29.2% (TPO), 12.5% (SDF-1), and 3.3% (control). Moreover, 4F injected only 2 hours after TBI at 50 μg/kg (4Fsingle) resulted in greater long-term survival (41.7%) than when injected 2 hours and 24 hours after TBI at 10 μg/kg (4F10, 8.3%), in accordance with short-term results. The benefit conferred by 5F and 4F versus TPO was not significant.

Autopsies carried out on animals that died between 30 days and 300 days after TBI showed a pattern of cachexia and multiorgan failure. Autopsies performed at 360 days in long-term survivors revealed signs of macroscopic anemia associated with liver disease in most animals and 4 cases of cancer (1 myosarcoma, 2 thymomas, 1 leukemia) in different groups. Spleens and femurs from mice of groups 5F, 4F, TPO, SDF-1, and UC were collected at that time. Spleen weights are reported in Table4. In contrast to the spleens of groups 4F and TPO, those of group 5F were significantly smaller than UC spleens. BM TNC counts (Tables 4 and 5) decreased in all treated groups to values equal to 64.5% (5F), 77.4% (4F), 76.3% (TPO), and 61.5% (SDF-1) of the UC level. However, this reduction in BM TNC was not significant for 5F (P = .06 vs UC). The most striking unexpected observation was that all treated animals proved to be significantly depleted of BM colony-forming units (CFUs) in comparison with UC. BM CFU of treated groups were 27.8% (5F), 56.1% (4F), 59.1% (TPO), and 44.1% (SDF-1) of UC CFU. In fact, though group 5F appeared significantly more depleted of CFU than other treated groups (Table 5), BM megakaryocytic progenitors were twice as numerous in group 5F as in other groups, UC included (4.1% of TNC vs 2% on average; P < .01). At 360 days, there was only one surviving irradiated control mouse whose BM displayed a marked reduction in TNC and clonogenicity (Table 4).

Table 4.

Spleen weight, total femoral BM nucleated cell count, and clonogenicity for different treatments evaluated 360 days after irradiation

Treatment group UC (n = 4) 5F (n = 17) 4F (n = 8) TPO (n = 5) SDF-1 (n = 4)
Spleen weight  62.8 ± 5.1  49.4 ± 15.1  54.9 ± 21.6  56 ± 22.6  48.5 ± 15.7 
 Range  56-68  36-101  28-96  31-96  31-69 
 Median  63.5  46  51  46  47  
BM TNC  43.4 ± 6.4  28 ± 13.6  33.6 ± 5.8  33.1 ± 3.4  26.7 ± 6.5 
 Range  34.4-48.4  14.4-47.6  25-42.6  31-38.8  18-32.8 
 Median  45.4  24  33.8  31.2  28 
 (%)  (100)  (64.5)  (77.4)  (76.3)  (61.5)  
Total BM CFU  143.7 ± 24.3  40 ± 24.7  80.6 ± 19.1  84.9 ± 8.6  63.4 ± 13.4 
 Range  121.5-173.6  19.7-54.7  59.7-113  79.6-99.6  44.4-74.5 
 Median  139.8  41  75.3  80.1  67.3 
 (%)  (100)  (27.8)  (56.1)  (59.1)  (44.1) 
Treatment group UC (n = 4) 5F (n = 17) 4F (n = 8) TPO (n = 5) SDF-1 (n = 4)
Spleen weight  62.8 ± 5.1  49.4 ± 15.1  54.9 ± 21.6  56 ± 22.6  48.5 ± 15.7 
 Range  56-68  36-101  28-96  31-96  31-69 
 Median  63.5  46  51  46  47  
BM TNC  43.4 ± 6.4  28 ± 13.6  33.6 ± 5.8  33.1 ± 3.4  26.7 ± 6.5 
 Range  34.4-48.4  14.4-47.6  25-42.6  31-38.8  18-32.8 
 Median  45.4  24  33.8  31.2  28 
 (%)  (100)  (64.5)  (77.4)  (76.3)  (61.5)  
Total BM CFU  143.7 ± 24.3  40 ± 24.7  80.6 ± 19.1  84.9 ± 8.6  63.4 ± 13.4 
 Range  121.5-173.6  19.7-54.7  59.7-113  79.6-99.6  44.4-74.5 
 Median  139.8  41  75.3  80.1  67.3 
 (%)  (100)  (27.8)  (56.1)  (59.1)  (44.1) 

Parameters were evaluated in long-term survivors versus age-matched unirradiated controls (UC). Results are expressed as follows: spleen weight in mg; BM total nucleated cell count in million cells from 2 femurs following erythrocyte lysis; total BM CFU × 10−3 per mouse (GM-CFU + BFU-e) enumerated after 10 days of culture in semisolid medium. The only irradiated animal surviving at 360 days exhibited the following values: spleen weight, 48 mg; femoral BM TNC, 20.6 × 106cells; BM CFU, 48,753.

Table 5.

Levuls of statistical significance between treatments versus age-matched unirradiated controls regarding spleen weight, femoral BM TNC, and clonogenics

P Spleen weight BM TNC BM CFU
< .01  5F < UC  —  5F and 4F < UC
5F < 4F and TPO 
      TPO and SDF-1 < UC 
< .05  —  4F, TPO, and SDF-1 < UC
5F < UC (P = .06) 
5F < SDF-1
SDF-1 < TPO 
P Spleen weight BM TNC BM CFU
< .01  5F < UC  —  5F and 4F < UC
5F < 4F and TPO 
      TPO and SDF-1 < UC 
< .05  —  4F, TPO, and SDF-1 < UC
5F < UC (P = .06) 
5F < SDF-1
SDF-1 < TPO 

Comparisons were made using the Mann-Whitney U test. All other comparisons were not significant. — indicates no results.

The results of this study support the concept that cytokine cooperation is important for hematopoietic recovery from radiation injury. They also suggest that short-term treatment is efficient. This work not only accounts for the short-term hematopoietic activity of cytokines on the irradiated organism, it also accounts for the long-term response to cytokine treatment. To date the prolonged administration of GM-CSF or G-CSF remains the criterion standard of cytokine therapy for irradiated victims in spite of their granulocyte-restricted activity.7,26 In most preclinical trials, HGF administration starts from the day after myeloablative injury, and the duration of treatment covers 15 to 21 days until neutrophil or platelet recovery.2-6,27 Few combinations have been used despite promising studies in myeloablated nonhuman primates that entailed 2 cytokines4-6 or synthetic dual cytokine receptor agonists.3,27 Thus far, nuclear accident victims have been treated with GM-CSF and IL-328 or G-CSF + GM-CSF.7 More recently, the victims of the criticality accident in Tokai-mura were given G-CSF, MGDF, and erythropoietin in combination.8 However, in these dramatic situations, cytokine therapy was initiated after a variable delay following radiation exposure, which might have reduced its efficacy. Furthermore, the prolonged administration of HGF may induce notable proinflammatory29 or immunogenic activity.3,9 

The potential therapeutic benefit of early and short-term administration of antiapoptotic cytokines has been suggested by some authors whose studies have been primarily focused on TPO.12,15 For some years the latter has emerged as a key factor, not only for its thrombopoietic activity but also as a survival factor on the HSPC.30 TPO/MGDF counteracts lethal myelosuppression when given early after radiobiologic or radiocombined damage.12,15,31-33 Moreover, this beneficial effect can be obtained with a single administration,12,15,31-34 which is as effective as 7 repeated injections for hematopoietic recovery and prevention of death.32 The single administration of TPO to humans has been reported to maintain hematopoiesis without inducing adverse effects.35 As demonstrated by Pestina et al,15 the survival effect involves the mitigation of the p53 apoptotic pathway, but TPO/Mpl-ligand appears to act downstream of p53.

IL-1 and tumor necrosis factor (TNF) also exhibit radioprotective effects, but these cytokines have been discarded because of high toxicity.36 In fact, given that hematopoiesis is regulated by a subtle balance between numerous positive and negative regulators whose cooperation is required to optimize cell survival, proliferation, and differentiation, we chose to evaluate in vivo the therapeutic efficacy of combinations of 5 different cytokines—SCF, FLT-3 ligand, TPO, IL-3, and SDF-1—whose antiapoptotic activity has been screened in vitro or in vivo.13,18,20 The SFT3 combination exhibited the highest antiapoptotic efficacy toward in vitro–irradiated CD34+ cells (dose range, 2.5-4 Gy) in comparison with the same cytokines used as single factors or other combinations.13 For instance, IL-3 has been demonstrated to reduce p53-dependent apoptosis in response to DNA damage by inducing the expression of Bcl-2 and Bcl-Xl.15 The SFT combination (3F) has recently been described to increase antiapoptotic survivin protein levels in quiescent CD34+cells.37 Moreover, the same authors have reported an inverse correlation between survivin and active caspase 3.38 In our in vitro study,13 the SFT3 combination reduced RI Fas/Fas ligand up-regulation and maintained the Bcl-2 expression level. Cell cycle analysis of irradiated cells treated with SFT3 showed almost no activation after 24 hours of incubation, unlike treated unirradiated cells. Altogether 3 days of incubation were necessary for irradiated cells to display a similar activation pattern of cell cycle as unirradiated cells, which resulted in subsequent CD34+ cell expansion within 7 days of liquid culture. Regarding SDF-1α, this is a promising stromal factor locally induced at the BM level, namely after irradiation, which keeps in vitro CD34+ cells from apoptosis after serum deprivation.20 Recently, SDF-1 has been described as a survival factor for CD34+ cells because it counteracts spontaneous apoptosis through an autocrine/paracrine mechanism.18,19 Preliminary studies suggested a multilineage activity in vivo because SDF-1 administration to nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice grafted with human cord blood mononuclear cells resulted in the mobilization to peripheral blood and spleen of CD41+ cells and myeloid/monocytic cells.39 

In the present study, we first focused on RI short-term hematopoietic cell death evaluated by the classical 30-day survival end-point. We have based our 2 injection treatments on the in vitro capacity of SFT3 to exert a strong antiapoptotic effect when added early to HSPC and to initiate the production of radioprotective progenitors. The cytokine dose (50 μg/kg for each cytokine of the combination) was chosen in accordance with our in vitro study, which demonstrated the necessity for using high concentrations to counteract RI apoptosis (ie, 50 ng/mL is better than 10 ng/mL for each cytokine of the combination),1 and the Pestina et al15 study, in which a dose range of 50 to 65 μg/kg was selected to reduce in vivo RI apoptosis. Here we show that administering the SFT3 combination at 50 μg/kg (each cytokine), supplemented or not with SDF-1α, 2 and 24 hours after acute, lethal TBI results in significantly improved survival.

To confirm that 4F efficacy was related at least partially to apoptosis reduction at the HSPC level, we used a monkey model of 5 Gy TBI. BM CD34+ cells were harvested 6 hours after irradiation, and we looked for mitochondrial membrane hyperpolarization as an early marker of active cell death.25 We brought evidence that 4F preserved CD34+ cells in vivo from RI apoptosis because no hyperpolarization occurred. On the contrary, BM CD34+ cells from untreated irradiated animals exhibited significant hyperpolarization in comparison with the preirradiation value, accounting for the initial phase of apoptosis. These data and those on the cell cycle sustain the hypothesis that the predominant effect of 4F treatment during the first 24 hours after irradiation consists of apoptosis reduction rather than cell proliferation.

The benefit provided to mice by SFT3 was also demonstrated in comparison with TPO-treated mice, which argues in favor of an additive or synergistic effect of the combination. In fact, TPO-treated mice in our model exhibited a survival rate consistent with that shown by Mouthon et al,12,32 who administered TPO 2 hours after TBI. By contrast, Pestina et al15 and Abushullaih et al33 obtained a higher survival rate by giving TPO immediately after lethal myelosuppression, which is unfortunately incompatible with the medical management of accidentally irradiated victims.15,33 Yet, their result is in agreement with the benefit that should be expected from the early counteraction of apoptosis. Preliminary data with the 4F combination in irradiated monkeys suggest that SFT3 can sustain multilineage recovery, including lymphopoiesis (F.H., M.D., manuscript in preparation). In the study by Neelis et al,31 single administration 2 hours after TBI proved to be significantly more efficient than treatment at 24 hours; therefore, we evaluated the efficacy of SFT3 given only at 2 hours after TBI. The benefit conferred by 2 injections of 4F versus a single injection was not actually significant. Interestingly, the dose administered appeared crucial, as did the multiple cytokine composition of the combination. In our model, dose reduction of SFT3 to 10 μg/kg or IL-3 withdrawal (2-hour and 24-hour schedules) only allowed for reduced survival rates. Finally, SDF-1 alone was less efficient than TPO, but recent studies suggest that SDF-1 bioavailability has to be improved before development for clinical use.39 

In this study we then focused on long-term follow-up to assess whether the cytokines selected and their combinations could behave as long-term protectors. In fact, if numerous studies conducted in mice have accounted for long-term follow-up,40-43 data on the effects in aging irradiated animals of early cytokine administration are lacking. In our study, delayed death mainly observed from day 150 after TBI was likely to occur from hematologic failure (hematopoietic and thrombotic disorders) and nonhematopoietic defects (fibrosis-associated multiorgan failure). In the Barnes et al42 study, most late deaths of untreated irradiated mice occurred between 9 and 32 weeks after mid-lethal X-irradiation (earlier than in our treated irradiated mice), and the most frequent syndromes observed were liver necrosis, diarrhea, pleural effusion, and aplastic anemia. In our study, no predominant etiology for death was observed. In particular, oncogenesis, which could be expected to be highly increased after antiapoptotic treatment, affected only a minority of long-term survivors, at least from a macroscopic analysis. From a clinical point of view, these survivors were comparable to healthy animals regarding body weight, food intake, and locomotion. However, when compared with UC, hematopoiesis was impaired in most of the treated animals, as shown by the reduction in BM TNC and clonogenic cell counts. This impairment was observed for all treated groups but appeared greater for 5F than for TPO, 4F, and SDF-1. These results are consistent with lasting impairment of marrow stem and progenitor cell compartments previously reported in various species following irradiation.40,41,43,44 

BM CFU depletion could be explained by 3 mechanisms: (1) RI apoptosis, because our in vitro results showing that SFT3 protects approximately 12.3% of CD34+ cells irradiated at 4 Gy from apoptosis13 support the hypothesis that such cytokine combinations prevent apoptosis only partially in vivo; (2) partial stem cell exhaustion following cytokine-induced overstimulation.31 In fact, whole-body survival provided by multiple cytokines involves the production of short-term radioprotective progenitors that occurs, in the range of lethal radiation doses, at the expense of long-term stem and progenitor cells. As proof of the contribution of radioprotective progenitors, we clearly observed 2 striking waves of platelet production 8 and 23 days later, issued respectively from megakaryocytes and megakaryoblasts in 5 Gy γ-irradiated cynomolgus monkeys receiving a single 4F injection 2 hours after TBI. In contrast, irradiated control animals displayed no response, but a transient profound phase of thrombocytopenia (F.H., M.D., manuscript in preparation) (3) reduced effectiveness of residual stem cells.41,45 One can wonder whether the level of HSPC depletion could be related to the multiple cytokine composition of the combination (4F and 5F) or short-term treatment and whether in this context high survival rates paradoxically are obtained at the expense of the HSPC compartment. We found that 5F-treated animals exhibited increased BM megakaryocytes. This result is consistent with the megakaryopoietic activity of SDF-1 reported by some authors.39 Finally, we showed that despite delayed death occurring from day 150 on, half the protective effect of selected cytokines was preserved at 300 days.

A crucial issue is the selection of the ideal treatment for patients who have sustained severe radiobiologic injury. In our study, the short-term injection of multiple cytokine combinations was well tolerated and proved to be more efficient than single factors in promoting short-term survival in mice. It is possible that the administration of factors such as protease inhibitors, p53 chemical inhibitors,46 and negative regulators of hematopoiesis, with or without sequential cytokines such as antiapoptotic factors followed by colony-stimulating factors, may result in improved long-term hematopoiesis.

We thank S. Doulay, C. Dupuy, N. Grenier, V. Leroux, and L. Crépin for their technical assistance.

Prepublished online as Blood First Edition Paper, December 5, 2002; DOI 10.1182/blood-2002-06-1634.

Supported by a grant from Electricité de France.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Monroy
RL
Radiation effects on the lymphohematopoietic system: a compromise in immune competency.
Military Radiobiology.
Conklin
JJ
Walker
RI
1987
111
134
Academic Press
Orlando, FL
2
Thierry
D
Gourmelon
P
Parmentier
C
Nenot
JC
Hematopoietic growth factors in the treatment of therapeutic and accidental irradiation induced bone marrow aplasia.
Int J Radiat Biol.
67
1995
103
117
3
MacVittie
TJ
Farese
AM
Cytokine-based treatment for acute radiation-induced myelosuppression: preclinical and clinical perspective.
The Medical Basis for Radiation Accident Preparedness.
Ricks
RC
Berger
ME
O'Hara
FM
Jr
2002
53
72
Parthenon Publishing Group
New York, NY
4
Farese
AM
Williams
DE
Seiler
FR
MacVittie
TJ
Combination protocols of cytokine therapy with interleukin-3 and granulocyte-macrophage-colony-stimulating factor in a primate model of radiation-induced marrow aplasia.
Blood.
82
1993
3012
3018
5
Farese
AM
Hunt
P
Grab
LB
MacVittie
TJ
Combined administration of recombinant human megakaryocyte growth and development factor and granulocyte colony-stimulating factor enhances multilineage hematopoietic reconstitution in nonhuman primates after radiation-induced marrow aplasia.
J Clin Invest.
97
1996
2145
2151
6
Neelis
KJ
Dubbelman
YD
Qingliang
L
Thomas
R
Eaton
DL
Wagemaker
G
Simultaneous administration of TPO and G-CSF after cytoreductive treatment of rhesus monkeys prevents thrombocytopenia, accelerates platelet and red cell reconstitution, alleviates neutropenia, and promotes the recovery of immature bone marrow cells.
Exp Hematol.
25
1997
1084
1093
7
Jinaratana
V
The radiological accident in Thailand.
The Medical Basis for Radiation Accident Preparedness.
Ricks
RC
Berger
ME
O'Hara
FM
Jr
2002
283
301
Parthenon Publishing Group
New York, NY
8
Maekawa
K
Overview of medical care for highly exposed victims in the Tokai Mura accident.
The Medical Basis for Radiation Accident Preparedness.
Ricks
RC
Berger
ME
O'Hara
FM
Jr
2002
313
318
Parthenon Publishing Group
New York, NY
9
Li
J
Xia
Y
Bertino
A
Coburn
J
Kuter
DJ
Characterization of an anti-thrombopoietin antibody that developing a cancer patient following the injection of Peg-rhuMGDF [abstract].
Blood.
94
1999
51
10
Neta
R
Oppenheim
JJ
Cytokines in therapy of radiation injury.
Blood.
72
1988
1093
1095
11
Sureda
A
Valls
A
Kadar
E
et al
A single dose of G-CSF modifies radiation-induced death in B6D2F1 mice.
Exp Hematol.
21
1993
1605
1607
12
Mouthon
MA
Gaugler
MH
Van der Meeren
A
Vandamme
M
Gourmelon
P
Wagemaker
G
Single administration of thrombopoietin to lethally irradiated mice prevents infectious and thrombotic events leading to mortality.
Exp Hematol.
29
2001
30
40
13
Drouet
M
Mathieu
J
Grenier
N
Multon
E
Sotto
JJ
Hérodin
F
The reduction of in vitro radiation-induced Fas-related apoptosis in CD34+ progenitor cells by SCF, Flt-3 ligand, TPO and IL-3 in combination resulted in CD34+ cell proliferation and differentiation.
Stem Cells.
17
1999
273
285
14
Domen
J
Gandy
KL
Weissman
IL
Systemic overexpression of Bcl-2 in the hematopoietic system protects transgenic mice from the consequences of lethal irradiation.
Blood.
91
1998
2272
2282
15
Pestina
TI
Cleveland
JL
Yang
C
Zambetti
GP
Jackson
CW
MPL-ligand prevents lethal myelosuppression by inhibiting p53-dependent apoptosis.
Blood.
98
2001
2084
2090
16
Dainiak
N
Hematologic consequences of exposure to ionizing radiation.
Exp Hematol.
30
2002
513
528
17
Van Bekkum
DW
Radiation sensitivity of the hematopoietic stem cell.
Radiat Res.
128
1991
S4
S8
18
Lataillade
JJ
Clay
D
Dupuy
C
et al
Chemokine SDF-1 enhances circulating CD34+ cell proliferation in synergy with cytokines: possible role in progenitor survival.
Blood.
95
2000
756
768
19
Lataillade
JJ
Clay
D
Bourin
P
et al
SDF-1 regulates primitive hematopoiesis by suppressing apoptosis and by promoting G0/G1 transition in cells: evidence for an autocrine/paracrine mechanism.
Blood.
99
2002
1117
1129
20
Ponomaryov
T
Peled
A
Petit
I
et al
Induction of the chemokine stromal-derived factor-1 following DNA damage improves human stem cell function.
J Clin Invest.
106
2000
1331
1339
21
FitzGerald
TJ
McKenna
M
Rothstein
L
Daugherty
C
Kase
K
Greenberger
JS
Radiosensitivity of human bone marrow granulocyte–macrophage progenitor cells and stromal colony–forming cells: effect of dose rate.
Radiat Res.
107
1986
205
215
22
Norol
F
Drouet
M
Pflumio
F
et al
Ex vivo expansion marginally amplifies repopulating cells from baboon peripheral blood mobilized CD34+ cells.
Br J Haematol.
117
2002
924
934
23
Jordan
CT
Yamasaki
G
Minamoto
D
High-resolution cell cycle analysis of defined phenotypic subsets within primitive human hematopoietic cell populations.
Exp Hematol.
24
1996
1347
1355
24
Hertveldt
K
Philippé
J
Thierens
J
et al
Flow cytometry as a quantitative and sensitive method to evaluate low dose radiation induced apoptosis in vitro in human peripheral blood lymphocytes.
Int J Radiat Biol.
71
1997
429
433
25
Scarlett
JL
Sheard
PW
Hughes
G
et al
Changes in mitochondrial membrane potential during stauroporine induced apoptosis in Jurkat cells.
FEBS Lett.
475
2000
267
272
26
Gale
RP
Butturini
A
Use of hematopoietic growth factors in radiation accidents.
Int J Radiat Oncol Biol Phys.
19
1990
1291
1295
27
Farese
AM
Smith
WG
Giri
JG
Siegel
N
McKearn
JP
MacVittie
TJ
Promegapoeitin 1A, an engineered chimeric IL-3 and MPL receptor agonist, stimulates hematopoietic recovery in conventional and abbreviated schedule following radiation-induced myelosuppression in nonhuman primates.
Stem Cells.
19
2001
329
338
28
Baranov
AE
Selidovkin
GD
Butturini
A
Gale
RP
Hematopoietic recovery after 10 Gy acute total body irradiation.
Blood.
83
1994
596
599
29
Negrin
RS
Clinical applications of hematopoietic growth factors.
The Cytokine Handbook.
Thomson
AW
1998
803
822
Academic Press
San Diego, CA
30
Borge
OJ
Ramsfjell
V
Cui
L
et al
Ability of early acting cytokines to directly promote survival and suppress apoptosis of human primitive CD34+CD38− bone marrow cells with multilineage potential at the single-cell level: key role of thrombopoietin.
Blood.
90
1997
2282
2292
31
Neelis
KJ
Visser
TP
Dimjati
W
et al
A single dose of thrombopoietin shortly after myelosuppressive total body irradiation prevents pancytopenia in mice by promoting short term multilineage spleen repopulating cells at the transient expense of bone marrow repopulating cells.
Blood.
92
1998
1586
1597
32
Mouthon
MA
Van der Meeren
A
Gaugler
MH
et al
Thrombopoietin promotes hematopoietic recovery and survival after high-dose whole body irradiation.
Int J Radiat Oncol Biol Phys.
43
1999
867
875
33
Abushullaih
BA
Pestina
TI
Srivastava
DK
Jackson
CW
Daw
NC
A schedule of recombinant MPL ligand highly effective at preventing lethal myelosuppresion in mice given carboplatin and radiation.
Exp Hematol.
29
2001
1425
1431
34
Ulich
TR
del Castillo
J
Senaldi
G
et al
The prolonged hematologic effects of a single injection of PEG-rhuMGDF in normal and thrombocytopenic mice.
Exp Hematol.
27
1999
117
130
35
Kuter
DJ
Goodnough
LT
Romo
J
et al
Thrombopoietin therapy increases platelet yields in healthy platelet donors.
Blood.
98
2001
1339
1345
36
Dinarello
CA
Interleukin-1.
The Cytokine Handbook.
Thomson
AW
1998
35
72
Academic Press
San Diego, CA
37
Fukuda
S
Foster
RG
Porter
SB
Pelus
LM
The antiapoptotic protein survivin is associated with cell cycle entry of normal cord blood CD34+ cells and modulates cell cycle and proliferation of mouse hematopoietic progenitor cells.
Blood.
100
2002
2463
2471
38
Fukuda
S
Pelus
LM
Regulation of the inhibitor-of-apoptosis family member survivin in normal cord blood and bone marrow CD34+ cells by hematopoietic growth factors: implication of survivin expression in normal hematopoiesis.
Blood.
98
2001
2091
2100
39
Perez
L
Alpdogan
O
Shieh
J
et al
In vivo effects of stromal derived factor 1 (SDF-1/CXCL12) on human megakaryocytopoiesis and mobilization of human hematopoietic progenitors [abstract].
Blood.
98
2001
799
40
Croizat
H
Frindel
E
Tubiana
M
Long-term radiation effects on the bone marrow stem cells of C3H mice.
Int J Radiat Biol.
36
1979
91
99
41
Wangenheim
KH
Peterson
HP
Hubner
GE
Feinendegen
LE
Long-term residual radiation effect in the murine hematopoietic stem cell compartment.
Ann N Y Acad Sci.
459
1985
221
231
42
Barnes
DWH
Bungay
GT
Mole
RH
Delayed mortality after mid-lethal exposure to whole body irradiation and its modification by treatment with syngenic lymph node or bone marrow cells.
Int J Rad Biol.
11
1966
409
427
43
Knospe
WH
Long-term bone marrow damage after irradiation.
Hematopoiesis: long-term effects of chemotherapy and radiation.
Testa
NG
Gale
RP
1988
93
130
Marcel Dekker
New York, NY
44
Nothdurft
W
Steinbach
KH
Fliedner
TN
Dose- and time-related quantitative and qualitative alterations in the granulocyte/macrophage progenitor cell (GM-CFC) compartment of dogs after total body irradiation.
Radiat Res.
98
1984
332
344
45
Cronkite
EP
Inoue
T
Hirabayashi
Y
Are stem cells exposed to ionizing radiation in vivo as effective as nonirradiated transfused stem cells in restoring hematopoiesis?
Exp Hematol.
21
1993
823
825
46
Komarov
PG
Komarova
EA
Kondratov
RV
et al
A chemical inhibitor of p53 that protects mice from side effects of cancer therapy.
Science.
285
1999
1733
1737

Author notes

Francis Hérodin, Centre de Recherches du Service de Santé des Armées, Departement of Radiobiology, Radiohematology Unit 24, avenue des Maquis du Grésivaudan 38702, La Tronche Cedex France; e-mail:francis.herodin@wanadoo.fr.

Sign in via your Institution