Purpose:

One of the main limitations to anticancer radiotherapy lies in irreversible damage to healthy tissues located within the radiation field. “FLASH” irradiation at very high dose-rate is a new treatment modality that has been reported to specifically spare normal tissue from late radiation-induced toxicity in animal models and therefore could be a promising strategy to reduce treatment toxicity.

Experimental Design:

Lung responses to FLASH irradiation were investigated by qPCR, single-cell RNA sequencing (sc-RNA-Seq), and histologic methods during the acute wound healing phase as well as at late stages using C57BL/6J wild-type and Terc−/− mice exposed to bilateral thorax irradiation as well as human lung cells grown in vitro.

Results:

In vitro studies gave evidence of a reduced level of DNA damage and induced lethality at the advantage of FLASH. In mouse lung, sc-RNA-seq and the monitoring of proliferating cells revealed that FLASH minimized the induction of proinflammatory genes and reduced the proliferation of progenitor cells after injury. At late stages, FLASH-irradiated lungs presented less persistent DNA damage and senescent cells than after CONV exposure, suggesting a higher potential for lung regeneration with FLASH. Consistent with this hypothesis, the beneficial effect of FLASH was lost in Terc−/− mice harboring critically short telomeres and lack of telomerase activity.

Conclusions:

The results suggest that, compared with conventional radiotherapy, FLASH minimizes DNA damage in normal cells, spares lung progenitor cells from excessive damage, and reduces the risk of replicative senescence.

Translational Relevance

“FLASH” radiotherapy involves delivering large doses of radiation in a single fraction in less than 0.1 second. FLASH has been reported to spare normal tissues from dose-limiting toxicity while keeping the antitumor efficiency unchanged. One pressing issue to advance FLASH to early-phase clinical trials is to uncover the molecular and physiologic mechanisms that underlie this differential response. Using a combination of transcriptomic, biochemical, immunochemical, and histologic in vivo and in vitro approaches, we identified early and late markers of murine lung response that are differentially expressed following FLASH relative to conventional radiotherapy. Our data show that FLASH minimizes the induction of proinflammatory genes and persistent DNA damage and facilitates radiation recovery by preserving lung progenitor cells.

For decades, radiation oncologists have been delivering 2 Gy daily fractions at dose rates around 0.01 Gy/second for up to a total dose reaching the limit of tolerance of healthy tissues. We recently developed another methodology named “FLASH” that consists of delivering ≥ 10 Gy in a single microsecond pulse or in a limited number of pulses of 1–2 Gy each given in ≤ 100 ms temporal sequence (1). In mice, FLASH was found to elicit a dramatic decrease of damage to normal tissues, with protection against lung fibrosis as well as memory loss after brain irradiation while keeping the antitumoral effect unchanged (1, 2). Such protection against normal tissue injury has been confirmed in large mammals (3), thus evoking a strong interest for the FLASH methodology in anticancer radiotherapy.

Radiation-induced damage to the lung leads to an acute pneumonitis followed by abortive regenerative processes that, together with chronic inflammation, foster fibrosis development in the months following radiation (4). Pathophysiology of radiation-induced pulmonary fibrosis is complex and lung regeneration after radiation injury is poorly understood. However, authors (5, 6) have reported the presence of persistent DNA damage foci as well as senescent cells in the months following thoracic irradiation, two factors that may impair lung regenerative processes. Using a variety of analytic approaches, we identified some early and late molecular indicators of the differential response of mouse lung to FLASH versus conventional dose-rate irradiation (CONV). Data show that FLASH minimizes the induction of proinflammatory genes, facilitates radiation recovery and reduces the number of persistent DNA damage foci as well as radiation-induced senescence and fibrogenesis.

Mice and ethics statement

Studies were performed in accordance with the recommendations of the European Community (2010/63/UE) for the care and use of laboratory animals. Experimental procedures were specifically approved by the ethics committee of the Institut Curie CEEA-IC #118 (Authorization number APAFIS#5479-201605271 0291841 given by National Authority) in compliance with the international guidelines. Females C57BL/6J mice purchased from Charles River Laboratories at the age of 6 weeks were housed in Institut Curie animal facilities. Terc−/− mice described previously (7, 8) were purchased from Jackson Laboratories and intercrossed until the third generation, at which time mice became sterile or pups died at the next generation.

Irradiation

Mice were exposed to bilateral thorax irradiation at the age of 10–12 weeks using the 4.5-MeV linear electron accelerator facility (Kinetron; ref. 9). Collimation, time-resolved fluence measurement, chemical dosimetry, depth-dose distribution, mouse immobilization, and irradiation of mouse thorax were described previously (1) except for anesthesia that was carried out with a nose cone using 2.5% isoflurane in air, without adjunction of oxygen. Other details are given under Supplementary Data S1.

In vitro studies with human cells

For the immunofluorescent determination of radio-induced foci of the TRP53BP1 (53BP1) and γH2AX proteins at sites of DNA damage, we used two nontransformed human lung fibroblast cell lines, namely, MRC5 and IMR-90 and the human lung epithelial carcinoma cell line A-549. Details of cell culture conditions and immunofluorescence assays are given in Supplementary Data S2. For studies of differentiation and survival, we used Human Pulmonary Basal Epithelial Cells (PBEC). The preparation, irradiation, and analysis of PBECs are described under Supplementary Data S3.

Scoring persistent foci of Trp53bp1 (53bp1) in irradiated mouse lung

The methods used for determination of the number of late, persistent foci of 53bp1 in cells isolated from mouse lungs at one week and three months postirradiation, are described under Supplementary Data S4.

RNA isolation and qPCR analysis

The lower lung right lobe was disrupted in cold lysis buffer with electrical homogenizer and total RNA extracted using the RNEasy Plus Mini Kit (Qiagen #74134). Two micrograms of RNA was then reverse transcribed using High Capacity cDNA Reverse Transcription Kit (Applied Biosystems #4368814). Quantitative gene expression was measured by real-time qPCR using Fast Sybr Green Master Mix (Bio-Rad #4385612). Values are presented as the ratio of target mRNA to β-actin mRNA obtained using the 2–ΔΔCt method and normalized to nonirradiated control (fold change).

Histology

For histologic analysis, lungs were excised, washed with ice-cold PBS, gently inflated in 4% PFA under mild vacuum (25 Torr, 1 hour at room temperature) followed by 3 hours in the cold room, then embedded in paraffin and cut into 7-μm–thick slices. The preparations were stained by hematoxylin–eosin or Masson trichrome (R.A.L Diagnostics, #361350).

Monitoring cell proliferation

The proliferation rate of lung cells during the week postirradiation was assessed by 5-ethenyl-2′-deoxyuridine (EdU) incorporation. Irradiated mice and controls received daily intraperitoneal injection of EdU (4 mg/kg) and were euthanized 7 days postirradiation. The left lung was fixed in 4% PFA, embedded in paraffin, and EdU was revealed using Click-IT chemistry (Baseclick GmBH, BCK488-IV-IM-S) on 7-μm–thick sections. For EdU quantification, lung cells were dissociated from lung lobes, cytospinned, and stained for EdU incorporation as well as CD45 antibody (eBioscience #11-0451, 1/200 dilution). More than 1,000 cells were analyzed per condition.

Single-cell RNA sequencing

Lungs were resected and single-cell suspensions were prepared by enzymatic dissociation and sequential filtering (100 and 40 μm) before red blood cell lysis and cell counting. Single-cell 3′-RNAseq samples were prepared using single cell V2 reagent kit and Chromium controller (10× Genomics, PN-120237). cDNA quality control was assessed by capillary electrophoresis (Bioanalyzer, Agilent) before libraries' preparation and sequencing on HiSeq 2500 (Illumina). Raw and processed data were deposited in GEO (GSE133992, for review use token sfaresuwplurtcb). Data processing and analysis was performed as described under Supplementary Data S5.

SA-β-gal analysis in mouse lung

Senescence-associated β-galactosidase (SA-β-gal) activity was assessed with a commercially available assay (Cell Signaling Technology #9860) as per manufacturer's instructions. Briefly, the left lobes were fixed and incubated overnight with staining solution at 37°C. Before paraffin embedding, macroscopic images representing 80% of the lobe was taken with a stereomicroscope and, once embedded, sections were cut and counterstained with hematoxylin to visualize tissue architecture. β-gal+ cell clusters were counted over more than 15 lung sections per mouse.

CT imaging and analysis

Three-dimensional X-ray imaging of lung was performed on the cone beam computed tomography (CBCT) module of the Small Animal Radiation Research Platform (SARRP, Xstrahl), operated at 60-kVp X-ray tube voltage, 0.8-mA tube current and 6 frames/second. Animals were anesthetized with 1.5%–2% isoflurane and maintained on a PMMA vertical stand in the upright position. Using the integrated software Murislice (XStrahl), the 3D reconstruction was calculated from 1,440 projections, with isotropic voxels of size 117 μm. The reconstructed slices were analyzed with ImageJ/FIJI (ImageJ, NIH, Bethesda, MD). First, we normalized the voxel intensity of all the acquisitions on a common scale of density (NDU, Normalized Density Units) in applying a linear contrast stretching based on two ROI, in the air and in the PMMA stand, respectively. The lung was then segmented from the rest of the cavity chest with the FIJI k-means Clustering plugin. We calculated the mean NDU of the voxel intensity distribution within the lung, and the 75th percentile. The latter was used to draw the highest 25% of NDU on the whole 3D image. We determined the fibrosis status of the mouse based on the following criteria: either the total mean NDU of the lung was higher than the upper bound of the 95% confidence interval measured on the nonirradiated mice or the highest 25% of NDU were observed notably at the base of the right superior lobe (Supplementary Fig. S6). Previous experiments showed that all the mice presenting a densification of the lung at the base of the right superior lobe will inevitably develop complete lung fibrosis in the following weeks (Supplementary Fig. S6).

Statistical analysis

Data analysis was performed using StatEL (AD Software) or Prism software (GraphPad software) and, unless otherwise stated, expressed as mean ± SEM. Two-group comparisons were analyzed either by two-tailed t test for independent samples, Mann–Whitney U test or Fisher exact test (*, P < 0.1; **, P < 0.01; ***, P < 0.001).

FLASH minimizes DNA damage and lethality in normal human lung cells in vitro

The Histone H2AX and the TRP53-binding protein 1 (53BP1) are rapidly phosphorylated in response to DNA double-strand breaks (DSB) and aggregate into discrete foci that can be analyzed by immunofluorescence microscopy. γH2AX recruitment extends over several megabases from both sides of the DSB. 53BP1 is at the crossroad of recombination and endjoining and promotes microhomology end-joining in G0–G1 phase cells (10). We thus performed combined analysis of γH2AX and 53BP1 foci by immunofluorescence microscopy for a comprehensive analysis of DSBs in the early times after FLASH versus CONV irradiation in MRC5 and IMR-90 normal human fibroblasts as well as in A549 human lung adenocarcinoma cells grown on coverslips in vitro. Cells at midconfluence were given 5.2 ± 0.2 Gy delivered in the FLASH or CONV mode, fixed and examined at 5, 30, and 180 minutes postirradiation. In the three cell lines, radiation produced a sharp increase in the number of 53BP1 and γH2AX foci per nucleus culminating at 30 minutes followed by a ≥ 60% decrease at 180 minutes postirradiation (Fig. 1A) in close agreement with published data (11, 12). We then chose the 30-minute time point for systematic investigation of foci formation. The results are shown in Fig. 1B. FLASH elicited significantly less 53BP1 foci than CONV in the two fibroblast cell lines. In contrast, the response of A549 tumor cells in terms of 53BP1 foci was indifferent to the modality of radiation. The number of γH2AX foci was slightly reduced in FLASH relative to CONV conditions in the two fibroblast cell lines, but this tiny difference was not statistically significant. These results suggest that FLASH induces less initial, 53BP1-relevant DNA damage than CONV and that this difference is specific to normal cells.

Figure 1.

FLASH minimizes DNA damage and survival responses of normal human lung cells in vitro. A, Representative images of the time-dependent formation and decay of 53BP1 and γH2AX foci in nuclei of IMR-90 fibroblasts exposed to 5.2 ± 0.2 Gy in both CONV and FLASH modes. B, Box and whisker chart of the number of foci per nucleus of 53BP1 and γH2AX at 30 minutes postirradiation in human lung fibroblasts (MRC5, IMR-90) and tumor cells (A-549) grown on coverslips and exposed to radiation. A total of 148 (MRC5, CONV), 114 (MRC5, FLASH), 235 (IMR-90, CONV), 229 (IMR-90, FLASH), 223 (A-549, CONV), and 218 nuclei (A-549, FLASH) were analyzed. NI, nonirradiated. The statistical analysis of data pooled by cell line and radiation modality was performed using the nonparametric Mann–Whitney U test. Bold horizontal bars, median; T-bars, SD; NS, nonsignificant. C, PEBCs from three different donors were exposed to 2 or 4 Gy in the CONV or FLASH mode (two separate experiments) and allowed to grow in triplicate at an Air–Liquid interface for 21 days, at which time cells were fixed and prepared for P63 immunostaining. Left, the bulk P63pos cell count. The statistical analysis was made using a Mann–Whitney U test based on the data pooled from the three donors. Bold horizontal bars, median; T-bars, SD; NS, nonsignificant.

Figure 1.

FLASH minimizes DNA damage and survival responses of normal human lung cells in vitro. A, Representative images of the time-dependent formation and decay of 53BP1 and γH2AX foci in nuclei of IMR-90 fibroblasts exposed to 5.2 ± 0.2 Gy in both CONV and FLASH modes. B, Box and whisker chart of the number of foci per nucleus of 53BP1 and γH2AX at 30 minutes postirradiation in human lung fibroblasts (MRC5, IMR-90) and tumor cells (A-549) grown on coverslips and exposed to radiation. A total of 148 (MRC5, CONV), 114 (MRC5, FLASH), 235 (IMR-90, CONV), 229 (IMR-90, FLASH), 223 (A-549, CONV), and 218 nuclei (A-549, FLASH) were analyzed. NI, nonirradiated. The statistical analysis of data pooled by cell line and radiation modality was performed using the nonparametric Mann–Whitney U test. Bold horizontal bars, median; T-bars, SD; NS, nonsignificant. C, PEBCs from three different donors were exposed to 2 or 4 Gy in the CONV or FLASH mode (two separate experiments) and allowed to grow in triplicate at an Air–Liquid interface for 21 days, at which time cells were fixed and prepared for P63 immunostaining. Left, the bulk P63pos cell count. The statistical analysis was made using a Mann–Whitney U test based on the data pooled from the three donors. Bold horizontal bars, median; T-bars, SD; NS, nonsignificant.

Close modal

Next, we used primary human pulmonary bronchial epithelial cells (PBEC) expressing TRP63 (P63), a known marker for upper airway basal stem cells (13), to determine whether cells with essential characteristics of stemness elicited a differential response to CONV versus FLASH irradiation. We used an in vitro air–liquid interface (ALI) system to establish a three-dimensional stratified and polarized bronchial epithelium to compare the effects of the two modalities of irradiation at 2 and 4 Gy on the ability of PBECs to proliferate and differentiate. Upon injury, P63+ basal stem cells expand and spread over the denuded basal lamina and proliferate and differentiate into ciliated and secretory cells (ref. 14; Supplementary Data and Supplementary Fig. S3 for full details). We observed that the decrease of P63+ PBECs relative to the total number of cells at 21 days postirradiation was less pronounced when irradiated with FLASH (Fig. 1C). The difference was significant at the 4 Gy dose point. Therefore, FLASH spares PBECs grown in vitro from radiation-induced differentiation and cell death at moderate doses of radiation.

FLASH reduces the pressure to repopulate after radiation injury in mouse lung

We reasoned that induction of proliferation required to replace irreversibly damaged cells might start in the lungs in the days following radiation injury. To test this hypothesis, mice received daily injection of EdU all over the first week postirradiation (Fig. 2A). With this technique, the count of EdU+ cells includes all the cells that crossed S-phase, at least once during the week postirradiation. Lungs exposed to CONV irradiation showed an increase in the number of proliferating cells highlighted by the presence of clusters of 2–3 EdU+ cells in the lung parenchyma (Fig. 2B and C), from 4% in the nonirradiated controls to 8% after CONV irradiation. Remarkably, the number of EdU+ cells one week after FLASH irradiation was not significantly higher than in nonirradiated controls.

Figure 2.

FLASH reduces the need for cell proliferation following radiation injury. A, Ten-week-old C57BL/6J female mice exposed to CONV or FLASH thoracic irradiation were injected daily with 4 mg/kg EdU for 7 days. At the end of the week, mice were sacrificed and the lungs excised and sectioned. B, Representative images of EdU incorporation revealed by click-IT chemistry on 7-μm–thick lung sections. C, Representative images of costaining EdU (green) and CD45 (red). Arrows point to major colocalizations. D, Quantification of EdU+ (left), EdU+/CD45 (middle), and EdU+/CD45+ (right) cells obtained from lungs that were enzymically dissociated, cytospinned, and stained for EdU incorporation and CD45 expression. At least 3 mice and >2,500 cells were analyzed per condition.

Figure 2.

FLASH reduces the need for cell proliferation following radiation injury. A, Ten-week-old C57BL/6J female mice exposed to CONV or FLASH thoracic irradiation were injected daily with 4 mg/kg EdU for 7 days. At the end of the week, mice were sacrificed and the lungs excised and sectioned. B, Representative images of EdU incorporation revealed by click-IT chemistry on 7-μm–thick lung sections. C, Representative images of costaining EdU (green) and CD45 (red). Arrows point to major colocalizations. D, Quantification of EdU+ (left), EdU+/CD45 (middle), and EdU+/CD45+ (right) cells obtained from lungs that were enzymically dissociated, cytospinned, and stained for EdU incorporation and CD45 expression. At least 3 mice and >2,500 cells were analyzed per condition.

Close modal

Because radiation injury elicits an inflammatory response that may cause an infiltration of proliferating immune cells, we quantified the proportion of EdU+ cells that were either CD45 (nonimmune cells) or CD45+ 1 week postirradiation. While neither CONV nor FLASH irradiation induced any significant increase in the proportion of CD45+ EdU+ cells, the CD45 population showed a higher proportion of EdU+ cells after CONV irradiation only (Fig. 2D).

Single-cell RNA sequencing analysis identifies cell type–specific transcriptional changes of FLASH irradiation

To assess the molecular changes induced specifically after FLASH irradiation, we next performed single-cell RNA sequencing (sc-RNAseq) analyses 4 days postirradiation (Fig. 3A). This time point was chosen to avoid acute transcriptional changes induced by DNA damage and apoptosis and, second, because it showed the highest proportion of EdU+ proliferating cells over a 24-hour pulse period (data not shown). Using a droplet-based single-cell technology (Chromium, 10X Genomics), we analyzed 1,555 cells from nonirradiated mouse lung, then 2,283 and 2,189 cells after CONV and FLASH irradiation, respectively. Integrated sc-RNAseq analysis with Seurat package (15) allowed us to identify 16 distinct lung cell populations present in the three different conditions (Fig. 3B). A major decrease in the percentage of B and T cells (naïve and activated subpopulations) was observed following both CONV and FLASH irradiation (Fig. 3C).

Figure 3.

sc-RNAseq analysis identifies specific molecular alterations after FLASH versus CONV irradiation. Mice were exposed to 17 Gy by bilateral thorax irradiation and euthanized at 4 days postirradiation. A, Mouse lungs (n = 3) were enzymatically dissociated, cell suspensions loaded in the Chromium controller (10x Genomics) following manufacturer's protocol before libraries' preparation and sequencing on HiSeq 2500 (Illumina). B, 6,027 cells passed the quality control and were clustered using a graph-based approach, visualized with Uniform Manifold Approximation and Projection (UMAP) plot, and cell populations identified based on published markers expression. C, Cell types distribution across NI, CONV, and FLASH conditions. D, Heatmap of genes differentially expressed between the different conditions. E, Tgfb1 and Cebpb expression in interstitial macrophages and monocytes. F, Egr1 expression in AT2 epithelial cells. E and F, FLASH is less efficient than CONV in upregulating (Tgfb1, Cebpb) or downregulating (Egr1) the expression of specific response genes at the time point considered.

Figure 3.

sc-RNAseq analysis identifies specific molecular alterations after FLASH versus CONV irradiation. Mice were exposed to 17 Gy by bilateral thorax irradiation and euthanized at 4 days postirradiation. A, Mouse lungs (n = 3) were enzymatically dissociated, cell suspensions loaded in the Chromium controller (10x Genomics) following manufacturer's protocol before libraries' preparation and sequencing on HiSeq 2500 (Illumina). B, 6,027 cells passed the quality control and were clustered using a graph-based approach, visualized with Uniform Manifold Approximation and Projection (UMAP) plot, and cell populations identified based on published markers expression. C, Cell types distribution across NI, CONV, and FLASH conditions. D, Heatmap of genes differentially expressed between the different conditions. E, Tgfb1 and Cebpb expression in interstitial macrophages and monocytes. F, Egr1 expression in AT2 epithelial cells. E and F, FLASH is less efficient than CONV in upregulating (Tgfb1, Cebpb) or downregulating (Egr1) the expression of specific response genes at the time point considered.

Close modal

Single-cell analysis in bulk (i.e., aggregating all cells to simulate bulk RNAseq data) failed to uncover major radiation-induced transcriptional alterations between CONV and FLASH conditions at the organ level (Fig. 3D). However, zooming-in at the cell populations level revealed molecular changes specific of FLASH-irradiated lung. FLASH was less efficient than CONV irradiation in eliciting upregulation of proinflammatory factors like Tgfb1 in the interstitial macrophage population and Cebpb, the Nf-κB canonical transcription factor in monocytes (Fig. 3E). Differential analysis of the alveolar epithelial AT2 cells, which contain stem/progenitor pools in the adult lung (16–18), showed a less pronounced decrease in Egr1 expression after FLASH than after CONV irradiation (Fig. 3F). Consistent with the known role of Egr1 in the maintenance of hematopoietic stem cell quiescence (19), calculation of the cell-cycle score of AT2 cells based on the expression of cell-cycle–specific genes with the CycleMix package, showed that FLASH is less efficient than CONV in pushing AT2 cells to exit quiescence and progress through S- and G2-phase (Supplementary Fig. S5).

In conclusion, FLASH proved to be less efficient than CONV irradiation in inducing expression of inflammatory genes and exit from quiescence in lung AT2 cells.

FLASH elicits less persistent nuclear 53bp1 foci than CONV irradiation in lung

Persisting focal accumulation of the adaptor protein 53bp1 at sites of chromosomal damage has been observed in liver, brain, and lung tissues long after exposure to a sublethal dose of radiation (20). These foci have been associated with the DNA segments with chromatin alterations reinforcing senescence (DNA-SCARS; ref. 5).

We sought to determine by immunofluorescence the fate of such persistent 53bp1 foci in cells isolated from irradiated lung at 1 week and 3 months postirradiation versus nonirradiated lungs collected from mice of the same age (Fig. 4A). With regard to the nonirradiated condition, after one week postirradiation, cells exposed to CONV and FLASH showed a higher damage signal in both the number of cells with 53bp1 foci and the number of foci per cell, which were significantly higher after CONV (Fig. 4B). After 3 months, the number of foci per cell decreased in FLASH-irradiated cells while it increased in the CONV condition. Moreover, while the number of cells bearing DNA damage foci remained stable after 3 months following FLASH, this number significantly increased in the CONV condition, suggesting that DNA damage continues to accumulate in the absence of exogenous injury.

Figure 4.

FLASH induces less persistent nuclear 53bp1 foci than CONV in irradiated lung. A, Representative images of 53bp1-associated immunofluorescence in cytospinned mouse lung cells (Supplementary Data S4) showing typical 53bp1 foci at 1 week postirradiation. B, Score of foci in individual nuclei (n > 850 per condition) at 1 week and 3 months postirradiation. C, Analysis of the multiplicity of foci at the same time points. The numbers in abscissa indicate the number of nuclei scored manually.

Figure 4.

FLASH induces less persistent nuclear 53bp1 foci than CONV in irradiated lung. A, Representative images of 53bp1-associated immunofluorescence in cytospinned mouse lung cells (Supplementary Data S4) showing typical 53bp1 foci at 1 week postirradiation. B, Score of foci in individual nuclei (n > 850 per condition) at 1 week and 3 months postirradiation. C, Analysis of the multiplicity of foci at the same time points. The numbers in abscissa indicate the number of nuclei scored manually.

Close modal

With rare exceptions, 53bp1 foci were arranged in multiplets consisting of individual foci paired in close proximity to each other and bound to the interior face of the nuclear membrane. The multiplicity changed with time (Fig. 4C). At 1-week postirradiation, the percentage of nuclei bearing singlets was not different from that seen in nonirradiated controls. In contrast, nearly half of the nuclei presented with foci assembled into doublets, quadruplets, or sextuplets. The ratio of quadruplets to doublets was slightly higher after CONV than FLASH irradiation and, as said above, the difference between the two modalities in terms of the number of foci per nucleus, was significant. The scene was profoundly altered at 3 months postirradiation. In FLASH-irradiated lungs, the count of nuclei presenting 53bp1 foci was not very different from that measured at 1-week postirradiation, but the total number of foci was significantly smaller due to a large decrease of higher-multiplicity foci, namely, sextuplets, and quadruplets. While the proportion of doublets did not change very much with time, the proportion of singlets was 10-fold higher than at 1-week postirradiation. A similar interconversion from high to low multiplicity was observed in nuclei from CONV-irradiated lungs (Fig. 4C). In the CONV group, however, the number of nuclei-bearing foci as well as the total foci count was significantly higher than at 1-week postirradiation, again suggesting that CONV-irradiated cells accumulate spontaneous DNA damage at distance from irradiation.

FLASH preserves the lung from radio-induced senescence

Replicative (stem) cell senescence is known to play a leading role in the induction of lung fibrosis (6). To test the hypothesis that FLASH may prevent, at least in part, the induction of senescent cells in the lung after irradiation and therefore limit the development of pulmonary fibrosis, we analyzed the presence of senescent clusters by detecting SA-β-gal activity in lung tissue 4 months postirradiation, that is, at the onset of fibrosis. Whole-mount SA-β-gal staining of the superior lobe revealed the presence of multiple senescent subpleural clusters after CONV irradiation, whereas only one or two clusters were visible after FLASH by whole mount as well as on histologic sections (Fig. 5A and B; Supplementary Fig. S7). Quantitative analysis of the number of SA-β-gal clusters per section indicated that there were half as many SA-β-gal+ senescent clusters 4 months after FLASH than after CONV irradiation (Fig. 5C).

Figure 5.

FLASH preserves the lung from radio-induced senescence. A, Macroscopic view of SA-β-gal staining of the right superior lobe from mice sacrificed 4 months after CONV or FLASH irradiation versus nonirradiated controls (NI) of the same age. B, Representative images of SA-β-gal staining of lung tissue at 16 weeks postirradiation. C, Diagram of the mean number of SA-β-gal–positive clusters per lung section (n ≥ 3 per condition). D, mRNA expression levels of the senescence markers genes Cdkn2a (p16INK4A), Serpine1 (PAI-1), and Mmp-2 by qRT-PCR from lungs at 3 (3M), 4 (4M), and 5 months (5M) postirradiation. Values are normalized to β-actin mRNA and expressed as the fold increase compared with the nonirradiated controls.

Figure 5.

FLASH preserves the lung from radio-induced senescence. A, Macroscopic view of SA-β-gal staining of the right superior lobe from mice sacrificed 4 months after CONV or FLASH irradiation versus nonirradiated controls (NI) of the same age. B, Representative images of SA-β-gal staining of lung tissue at 16 weeks postirradiation. C, Diagram of the mean number of SA-β-gal–positive clusters per lung section (n ≥ 3 per condition). D, mRNA expression levels of the senescence markers genes Cdkn2a (p16INK4A), Serpine1 (PAI-1), and Mmp-2 by qRT-PCR from lungs at 3 (3M), 4 (4M), and 5 months (5M) postirradiation. Values are normalized to β-actin mRNA and expressed as the fold increase compared with the nonirradiated controls.

Close modal

To confirm this observation, we quantified by qRT-PCR the mRNA expression of Cdkn2a (p16INK4A), Serpine1 (PAI-1) and Mmp-2, classical markers of senescence, and proinflammatory secreted associated senescence proteins (SASP) at the time of fibrogenesis, that is, 3 to 5 months postirradiation. In agreement with the histologic results, we observed that the expression of these three senescence markers was comparatively lower after FLASH than after CONV irradiation. This difference was particularly significant for p16INK4A mRNA, which underwent strong upregulation 4 months after CONV radiation exposure (Fig. 5D).

The FLASH sparing effect is lost in Terc−/− mice

Telomere length and proper maintenance are critical to preserve the replication potential of stem/progenitor cells (21). To determine whether the status of telomeres plays a role in normal tissue sparing in the FLASH mode, wild-type and Terc−/− mice harboring critically short telomeres were exposed to CONV or FLASH irradiation and followed for the development of lung fibrosis by periodical CT scan in the months postirradiation. Mice developed a densification of the lung parenchyma evocative of fibrosis (Fig. 6A and B; Supplementary Fig. S6) and were euthanized at 4 and 5 months postirradiation. Masson trichrome staining of lung sections showed subpleural areas with thickening of alveolar septa and collagen invasion characteristic of lung fibrosis (Fig. 6C). One wild-type FLASH-irradiated mouse out of 8 presented signs of fibrosis while 10 of 11 CONV-irradiated wild-type mice were committed to fibrosis (Fig. 6B), as expected (1). However, 12 of 12 CONV-irradiated and 7 of 8 FLASH-irradiated Terc−/− mice developed pulmonary fibrosis 4 and 5 months postirradiation. These results indicated that Terc−/− mice are not spared from fibrosis by FLASH irradiation.

Figure 6.

Terc−/− mice develop pulmonary fibrosis after FLASH irradiation. A, Representative high-resolution CT scan images from WT and third-generation Terc−/− mice at 16–20 weeks after thoracic irradiation. B, Quantification of the WT and Terc−/− mice harboring pulmonary fibrosis following CONV (n = 11 WT and 12 Terc−/− mice) versus FLASH (n = 8 WT and 9 Terc−/− mice) irradiation based on CT scan image analysis. Fisher exact test was performed to calculate the P value for each comparison. C, Representative images of Masson trichrome staining of the WT and Terc−/− lungs presented in A. Whole lung images are shown in Supplementary Fig. S8. D, SA-β-gal staining of Terc−/− lungs showing the presence of SA-β-gal+ clusters after CONV and FLASH irradiation. Nonirradiated Terc−/− mice did not present any SA-β-gal+ cells. Mosaic images of whole SA-β-gal–stained lung sections at 16 weeks postirradiation are shown in Supplementary Fig. S7.

Figure 6.

Terc−/− mice develop pulmonary fibrosis after FLASH irradiation. A, Representative high-resolution CT scan images from WT and third-generation Terc−/− mice at 16–20 weeks after thoracic irradiation. B, Quantification of the WT and Terc−/− mice harboring pulmonary fibrosis following CONV (n = 11 WT and 12 Terc−/− mice) versus FLASH (n = 8 WT and 9 Terc−/− mice) irradiation based on CT scan image analysis. Fisher exact test was performed to calculate the P value for each comparison. C, Representative images of Masson trichrome staining of the WT and Terc−/− lungs presented in A. Whole lung images are shown in Supplementary Fig. S8. D, SA-β-gal staining of Terc−/− lungs showing the presence of SA-β-gal+ clusters after CONV and FLASH irradiation. Nonirradiated Terc−/− mice did not present any SA-β-gal+ cells. Mosaic images of whole SA-β-gal–stained lung sections at 16 weeks postirradiation are shown in Supplementary Fig. S7.

Close modal

As lack of telomerase induces complex mouse phenotypes (22), we determined whether Terc−/− mice may be primed toward senescence by evaluating SA-β-gal staining on nonirradiated Terc−/− lung sections as well as after CONV and FLASH irradiation. In age-matched nonirradiated Terc−/− lung sections, we did not find any SA-β-gal cluster, whereas, as observed in CONV-irradiated wild-type mice, several SA-β-gal clusters were found in CONV- and FLASH-irradiated Terc−/− mice (Fig. 6D).

The goal of this study was to pave the way for identification of the mechanistic basis for the protective effect of FLASH to radiation-induced toxicity. Early DNA damage response was investigated in vitro using immunofluorescence characterization of radio-induced γH2AX and 53BP1 foci in normal human lung fibroblasts. The difference between FLASH and CONV with γH2AX as an endpoint was not significant. In contrast, a significant difference was observed at the level of 53BP1 foci. This suggests that the bulk of DNA double-strand breaks does not depend very much on the radiation mode while 53BP1 recognizes specifically a subset of DNA damage that is less abundant in the FLASH mode. FLASH also elicited a higher survival of P63+ human PBECs. In mouse lung, we observed a dramatic downregulation of all major indicators of tissue damage including (i) reduced proliferation after radiation injury; (ii) attenuated downregulation of Egr1 in AT2 cells revealed by sc-RNAseq, suggesting a reduced mobilization of the AT2 stem/progenitor pool after radiation injury; (iii) attenuated proinflammatory responses; (iv) a reduction of 53bp1 persistent nuclear foci in lung parenchymal cells; and finally (v) a reduced number of subpleural clusters of senescent cells. Furthermore, the protective effect of FLASH against radiation-induced fibrosis was lost in Terc−/− mice that present critically short telomeres, thus suggesting that a full potential of replication, most likely of the progenitor cell population, is required for the FLASH effect in vivo. However, even though lungs from nonirradiated Terc−/− mice did not present any sign of spontaneous senescence, more lung phenotypic analysis is required to confirm this conclusion.

It was recently shown that whole-brain FLASH irradiation preserves neurogenesis in the hippocampus and spares mice from the memory loss subsequent to CONV radiotherapy (2, 23). Actually, radiation injury is known to impact stem cell functions and their regenerative potential in different organs such as the brain and the intestine (24–26). In the lung, recent work from Stripp and colleagues showed that radiation reduces the in vitro colony-forming ability of epithelial progenitor cells from distal airways (27, 28). We recently demonstrated that NOTCH signaling promotes the survival of irradiated P63+ basal stem cells through activation of the 53BP1/DNA damage response pathway (29). Whether this also impacts on the protective response of FLASH is not known. Our results indicate that CONV thoracic irradiation leads to an acute proliferation phase in the days following radiation injury and that this pressure to repopulate the irradiated organ is reduced after FLASH irradiation. In line with this result, our sc-RNAseq analysis reveals that after CONV irradiation, the expression of Egr1, a transcription factor whose downregulation is necessary to trigger proliferation of hematopoietic stem cells (19), is reduced specifically in the AT2 compartment which is known to contain a stem/progenitor pool (16, 17, 19). This supports the idea that, owing to an intrinsically lower level of damage, FLASH does not push lung progenitor cells to replicate to the same extent as CONV irradiation, thus preserving the progenitor pool of cells from exhaustion.

The long-term consequences of radiation injury have been associated with persistent foci of the DNA damage protein 53bp1 concomitantly with senescence of the stem/progenitor cells (6, 20, 30) including secretion of IL6, an important SASP component (5). FLASH-irradiated cells isolated from whole lung also presented persistent 53bp1 foci and some occasional clusters of senescent cells. However, their number was significantly lower than in CONV-irradiated lungs and declined with time, while additional 53bp1 foci accumulated with aging in CONV-irradiated lungs. In line with our results, a recent study showed that senolytic treatment kills senescent AT2 cells and reverses radiation-induced pulmonary fibrosis, suggesting a causal link between the presence of senescent cells at late stages after irradiation and fibrogenesis (30).

In conclusion, our findings uncover a first mechanistic clue for the beneficial FLASH effect through normal tissue sparing. These results lend support to the design of preclinical studies investigating the therapeutic benefit of FLASH in cancer treatment.

No potential conflicts of interest were disclosed.

Conception and design: C. Fouillade, A. Londoño-Vallejo, P. Verrelle, V. Favaudon

Development of methodology: C. Fouillade, S. Curras-Alonso, L. Giuranno, S. Heinrich, A. Beddok, M. Vooijs, A. Londoño-Vallejo, V. Favaudon

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): C. Fouillade, S. Curras-Alonso, E. Quelennec, S. Heinrich, S. Bonnet-Boissinot, A. Beddok, S. Leboucher, S. Baulande, M. Vooijs, A. Londoño-Vallejo, V. Favaudon

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): C. Fouillade, S. Curras-Alonso, L. Giuranno, E. Quelennec, S. Heinrich, S. Bonnet-Boissinot, A. Beddok, H.U. Karakurt, M. Vooijs, V. Favaudon

Writing, review, and/or revision of the manuscript: C. Fouillade, S. Curras-Alonso, S. Heinrich, M. Vooijs, P. Verrelle, M. Dutreix, A. Londoño-Vallejo, V. Favaudon

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M. Bohec, P. Verrelle, V. Favaudon

Study supervision: C. Fouillade, A. Londoño-Vallejo, V. Favaudon

The authors wish to thank Marie-Noëlle Soler and Claire Lovo from the PICT-IBiSA Orsay Imaging facility of Institut Curie and Christophe Alberti, Frédéric Bertrand, Elodie Belloir, Virginie Dangles-Marie, Casper Caspersen and Isabelle Grandjean from the animal core facility of Institut Curie. The contribution of the bioinformatics core facility at U900 Inserm-Institut Curie is gratefully acknowledged. Thanks are due to Dr. Marie-Catherine Vozenin (Radio-oncology laboratory, Department of radio-oncology, Lausanne University Hospital, Switzerland) and Paul-Henri Roméo (IRCM-SCSR, CEA-DSV, Fontenay-aux-Roses, France) for kind support and helpful discussion. We also thank the dedicated work of Ikbal Rhimi on CT analysis. This work was funded by the Agence Nationale de la Recherche (ANR-14-CE36-0008-02 program), the Comprehensive Cancer Center “SIRIC” program of Institut Curie (Grant INCa-DGOS-4654) and the Nanotherad IDEX (Paris-Saclay University). High-throughput sequencing was performed by the ICGex NGS platform of Institut Curie supported by the grants ANR-10-EQPX-03 (Equipex) and ANR-10-INBS-09-08 (France Génomique Consortium) from the Agence Nationale de la Recherche (Investissements d'Avenir program) and by the Cancéropole Ile-de-France. This project has received funding from the European Union's Horizon 2020 Research and Innovation Program under the Marie Skłodowska-Curie grant agreements No. 666003 (S. Curras-Alonso and H.U. Karakurt PhDs, Institut Curie CO-FUND/IC3i program) and 642623 (L. Giuranno PhD, ITN RADIATE Grant). We are also grateful to the Prolific association for their financial support through a Graine de Chercheur grant to C. Fouillade. Generous financial aid from Mrs. Marie Mahler (Gisikon, Switzerland) is gratefully acknowledged.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Favaudon
V
,
Caplier
L
,
Monceau
V
,
Pouzoulet
F
,
Sayarath
M
,
Fouillade
C
, et al
Ultrahigh dose-rate FLASH irradiation increases the differential response between normal and tumor tissue in mice
.
Sci Transl Med
2014
;
6
:
245ra93
.
2.
Montay-Gruel
P
,
Petersson
K
,
Jaccard
M
,
Boivin
G
,
Germond
JF
,
Petit
B
, et al
Irradiation in a flash: unique sparing of memory in mice after whole brain irradiation with dose rates above 100Gy/s
.
Radiother Oncol
2017
;
124
:
365
9
.
3.
Vozenin
MC
,
De Fornel
P
,
Petersson
K
,
Favaudon
V
,
Jaccard
M
,
Germond
JF
, et al
The advantage of FLASH radiotherapy confirmed in mini-pig and cat-cancer patients
.
Clin Cancer Res
2019
;
25
:
35
42
.
4.
Yarnold
J
,
Brotons
MC
. 
Pathogenetic mechanisms in radiation fibrosis
.
Radiother Oncol
2010
;
97
:
149
61
.
5.
Rodier
F
,
Munoz
DP
,
Teachenor
R
,
Chu
V
,
Le
O
,
Bhaumik
D
, et al
DNA-SCARS: distinct nuclear structures that sustain damage-induced senescence growth arrest and inflammatory cytokine secretion
.
J Cell Sci
2011
;
124
:
68
81
.
6.
Citrin
DE
,
Shankavaram
U
,
Horton
JA
,
Shield
W
 3rd
,
Zhao
S
,
Asano
H
, et al
Role of type II pneumocyte senescence in radiation-induced lung fibrosis
.
J Natl Cancer Inst
2013
;
105
:
1474
84
.
7.
Blasco
MA
,
Lee
HW
,
Hande
MP
,
Samper
E
,
Lansdorp
PM
,
DePinho
RA
, et al
Telomere shortening and tumor formation by mouse cells lacking telomerase RNA
.
Cell
1997
;
91
:
25
34
.
8.
Wong
KK
,
Chang
S
,
Weiler
SR
,
Ganesan
S
,
Chaudhuri
J
,
Zhu
C
, et al
Telomere dysfunction impairs DNA repair and enhances sensitivity to ionizing radiation
.
Nat Genet
2000
;
26
:
85
8
.
9.
Favaudon
V
,
Lentz
JM
,
Heinrich
S
,
Patriarca
A
,
de Marzi
L
,
Fouillade
C
, et al
Time-resolved dosimetry of pulsed electron beams in very high dose-rate, FLASH irradiation for radiotherapy preclinical studies.
Nucl Inst Meth Phys Res A
2019
;
944
:
162537
.
10.
Xiong
X
,
Du
Z
,
Wang
Y
,
Feng
Z
,
Fan
P
,
Yan
C
, et al
53BP1 promotes microhomology-mediated end-joining in G1-phase cells
.
Nucleic Acids Res
2015
;
43
:
1659
70
.
11.
Schultz
LB
,
Chehab
NH
,
Malikzay
A
,
Halazonetis
TD
. 
p53 binding protein 1 (53BP1) is an early participant in the cellular response to DNA double-strand breaks
.
J Cell Biol
2000
;
151
:
1381
90
.
12.
Suzuki
K
,
Nakashima
M
,
Yamashita
S
. 
Dynamics of ionizing radiation-induced DNA damage response in reconstituted three-dimensional human skin tissue
.
Radiat Res
2010
;
174
:
415
23
.
13.
Smirnova
NF
,
Schamberger
AC
,
Nayakanti
S
,
Hatz
R
,
Behr
J
,
Eickelberg
O
. 
Detection and quantification of epithelial progenitor cell populations in human healthy and IPF lungs
.
Respir Res
2016
;
17
:
83
.
14.
Tadokoro
T
,
Wang
Y
,
Barak
LS
,
Bai
Y
,
Randell
SH
,
Hogan
BL
. 
IL-6/STAT3 promotes regeneration of airway ciliated cells from basal stem cells
.
Proc Natl Acad Sci U S A
2014
;
111
:
E3641
9
.
15.
Butler
A
,
Hoffman
P
,
Smibert
P
,
Papalexi
E
,
Satija
R
. 
Integrating single-cell transcriptomic data across different conditions, technologies, and species
.
Nat Biotechnol
2018
;
36
:
411
20
.
16.
Kim
CF
,
Jackson
EL
,
Woolfenden
AE
,
Lawrence
S
,
Babar
I
,
Vogel
S
, et al
Identification of bronchioalveolar stem cells in normal lung and lung cancer
.
Cell
2005
;
121
:
823
35
.
17.
Barkauskas
CE
,
Cronce
MJ
,
Rackley
CR
,
Bowie
EJ
,
Keene
DR
,
Stripp
BR
, et al
Type 2 alveolar cells are stem cells in adult lung
.
J Clin Invest
2013
;
123
:
3025
36
.
18.
Nabhan
AN
,
Brownfield
DG
,
Harbury
PB
,
Krasnow
MA
,
Desai
TJ
. 
Single-cell Wnt signaling niches maintain stemness of alveolar type 2 cells
.
Science
2018
;
359
:
1118
23
.
19.
Min
IM
,
Pietramaggiori
G
,
Kim
FS
,
Passegue
E
,
Stevenson
KE
,
Wagers
AJ
. 
The transcription factor EGR1 controls both the proliferation and localization of hematopoietic stem cells
.
Cell Stem Cell
2008
;
2
:
380
91
.
20.
Le
ONL
,
Rodier
F
,
Fontaine
F
,
Coppe
JP
,
Campisi
J
,
DeGregori
J
, et al
Ionizing radiation-induced long-term expression of senescence markers in mice is independent of p53 and immune status
.
Aging Cell
2010
;
9
:
398
409
.
21.
Hiyama
E
,
Hiyama
K
. 
Telomere and telomerase in stem cells
.
Br J Cancer
2007
;
96
:
1020
4
.
22.
Sahin
E
,
Colla
S
,
Liesa
M
,
Moslehi
J
,
Müller
FL
,
Guo
M
, et al
Telomere dysfunction induces metabolic and mitochondrial compromise
.
Nature
2011
;
470
:
359
65
.
23.
Montay-Gruel
P
,
Acharya
MM
,
Petersson
K
,
Alikhani
L
,
Yakkala
C
,
Allen
BD
, et al
Long-term neurocognitive benefits of FLASH radiotherapy driven by reduced reactive oxygen species
.
Proc Natl Acad Sci U S A
2019
;
116
:
10943
51
.
24.
Potten
CS
,
Hendry
JH
. 
Differential regeneration of intestinal proliferative cells and cryptogenic cells after irradiation
.
Int J Radiat Biol
1975
;
27
:
413
24
.
25.
Monje
ML
,
Mizumatsu
S
,
Fike
JR
,
Palmer
TD
. 
Irradiation induces neural precursor-cell dysfunction
.
Nat Med
2002
;
8
:
955
62
.
26.
Metcalfe
C
,
Kljavin
NM
,
Ybarra
R
,
de Sauvage
FJ
. 
Lgr5+ stem cells are indispensable for radiation-induced intestinal regeneration
.
Cell Stem Cell
2014
;
14
:
149
59
.
27.
Farin
AM
,
Manzo
ND
,
Kirsch
DG
,
Stripp
BR
. 
Low- and high-LET radiation drives clonal expansion of lung progenitor cells in vivo
.
Radiat Res
2015
;
183
:
124
32
.
28.
McConnell
AM
,
Konda
B
,
Kirsch
DG
,
Stripp
BR
. 
Distal airway epithelial progenitor cells are radiosensitive to high-LET radiation
.
Sci Rep
2016
;
6
:
33455
.
29.
Giuranno
L
,
Wansleeben
C
,
Iannone
R
,
Arathoon
L
,
Hounjet
J
,
Groot
AJ
, et al
NOTCH signaling promotes survival of irradiated basal airway stem cells
.
Am J Physiol Lung Cell Mol Physiol
2019
;
317
:
L414
23
.
30.
Shao
L
,
Feng
W
,
Li
H
,
Gardner
D
,
Luo
Y
,
Wang
Y
, et al
Total body irradiation causes long-term mouse BM injury via induction of HSC premature senescence in an Ink4a- and Arf-independent manner
.
Blood
2014
;
123
:
3105
15
.
31.
Niroomand-Rad
A
,
Blackwell
CR
,
Coursey
BM
,
Gall
KP
,
Galvin
JM
,
McLaughlin
WL
, et al
Radiochromic film dosimetry: recommendations of AAPM Radiation Therapy Committee Task Group 55. American Association of Physicists in Medicine
.
Med Phys
1998
;
25
:
2093
115
.
32.
Amatngalim
GD
,
van Wijck
Y
,
de Mooij-Eijk
Y
,
Verhoosel
RM
,
Harder
J
,
Lekkerkerker
AN
, et al
Basal cells contribute to innate immunity of the airway epithelium through production of the antimicrobial protein RNase 7
.
J Immunol
2015
;
194
:
3340
50
.
33.
van Wetering
S
,
Zuyderduyn
S
,
Ninaber
DK
,
van Sterkenburg
MA
,
Rabe
KF
,
Hiemstra
PS
. 
Epithelial differentiation is a determinant in the production of eotaxin-2 and -3 by bronchial epithelial cells in response to IL-4 and IL-13
.
Mol Immunol
2007
;
44
:
803
11
.
34.
McInnes
L
,
Healy
J
,
Melville
J
. 
UMAP: uniform manifold approximation and projection for dimension reduction
.
ArXiv
2018
:
1803
.
02528
.
35.
Reyfman
PA
,
Walter
JM
,
Joshi
N
,
Anekalla
KR
,
McQuattie-Pimentel
AC
,
Chiu
S
, et al
Single-cell transcriptomic analysis of human lung provides insights into the pathobiology of pulmonary fibrosis
.
Am J Respir Crit Care Med
2019
;
199
:
1517
36
.
36.
Xie
T
,
Wang
Y
,
Deng
N
,
Huang
G
,
Taghavifar
F
,
Geng
Y
, et al
Single-cell deconvolution of fibroblast heterogeneity in mouse pulmonary fibrosis
.
Cell Rep
2018
;
22
:
3625
40
.
37.
Finak
G
,
McDavid
A
,
Yajima
M
,
Deng
J
,
Gersuk
V
,
Shalek
AK
, et al
MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data
.
Genome Biol
2015
;
16
:
278
.
38.
Gauthier
NP
,
Jensen
LJ
,
Wernersson
R
,
Brunak
S
,
Jensen
TS
. 
Cyclebase.org: version 2.0, an updated comprehensive, multi-species repository of cell cycle experiments and derived analysis results
.
Nucleic Acids Res
2010
;
38
:
D699
702
.
39.
Scialdone
A
,
Natarajan
KN
,
Saraiva
LR
,
Proserpio
V
,
Teichmann
SA
,
Stegle
O
, et al
Computational assignment of cell-cycle stage from single-cell transcriptome data
.
Methods
2015
;
85
:
54
61
.