Autophagy inhibition radiosensitizes in vitro, yet reduces radioresponses in vivo due to deficient immunogenic signalling

A Ko, A Kanehisa, I Martins, L Senovilla, C Chargari, D Dugue, G Mariño, O Kepp, M Michaud, J-L Perfettini, G Kroemer, E Deutsch, A Ko, A Kanehisa, I Martins, L Senovilla, C Chargari, D Dugue, G Mariño, O Kepp, M Michaud, J-L Perfettini, G Kroemer, E Deutsch

Abstract

Clinical oncology heavily relies on the use of radiotherapy, which often leads to merely transient responses that are followed by local or distant relapse. The molecular mechanisms explaining radioresistance are largely elusive. Here, we identified a dual role of autophagy in the response of cancer cells to ionizing radiation. On one hand, we observed that the depletion of essential autophagy-relevant gene products, such as ATG5 and Beclin 1, increased the sensitivity of human or mouse cancer cell lines to irradiation, both in vitro (where autophagy inhibition increased radiation-induced cell death and decreased clonogenic survival) and in vivo, after transplantation of the cell lines into immunodeficient mice (where autophagy inhibition potentiated the tumour growth-inhibitory effect of radiotherapy). On the other hand, when tumour proficient or deficient for autophagy were implanted in immunocompetent mice, it turned out that defective autophagy reduced the efficacy of radiotherapy. Indeed, radiotherapy elicited an anti-cancer immune response that was dependent on autophagy-induced ATP release from stressed or dying tumour cells and was characterized by dense lymphocyte infiltration of the tumour bed. Intratumoural injection of an ecto-ATPase inhibitor restored the immune infiltration of autophagy-deficient tumours post radiotherapy and improved the growth-inhibitory effect of ionizing irradiation. Altogether, our results reveal that beyond its cytoprotective function, autophagy confers immunogenic properties to tumours, hence amplifying the efficacy of radiotherapy in an immunocompetent context. This has far-reaching implications for the development of pharmacological radiosensitizers.

Figures

Figure 1
Figure 1
Knockdown of ATG5 or BECN 1 alters autophagy after irradiation and enhances radiation sensitivity of A549, H460 and CT26 cells. (a) Decreased ATG5 and BECN1 expression in A549 and H460 cells by RNA interference was observed by immunoblot. A representative immunoblot of three independent experiments is shown. (bd) Cell lines as indicated. Radiation sensitivity of ATG5- and BECN1-depleted cells was assessed by clonogenic assay. Cells were transfected with a control shRNA or with an shRNA-targeting ATG5 or BECN1, then cells were left untreated or irradiated by 2 or 4 Gy. Cellular colonies were coloured and counted after 12–14 days and survival fraction (SF) was calculated and represented as described before. Quantitative data are reported as means±S.E.M. (n=3 triplicate samples, Student's t-test, ***P<0.001, as compared with untreated cells)
Figure 2
Figure 2
IR-induced cell death is increased in ATG5-depleted cells. (a) ATG5 depletion triggers mitochondrial membrane depolarization of A549 cells after ionizing radiation. A549 cells transfected by scrambled siRNA (SCR) or by specific shRNA for ATG5 (ATG5KD) were irradiated by 4 Gy and then subjected one day later to dual staining with the vital propidium iodide (PI) dye and with the mitochondrial transmembrane potential-sensitive dye 3,3 dihexyloxacarbocyanine iodide DiOC6(3) dye. Cell death was quantified by cytofluorometric analysis. (b) Histogram reporting the experiment shown in (a). Quantitative data are shown as means±S.E.M. (n=3 triplicate samples, Student's t-test, ***P<0.001 comparing ATG5KD (4 Gy) with ATG5KD (0 Gy) and SCR (4 Gy) with SCR (0 Gy); ###P<0.001 comparing ATG5KD (4 Gy) with SCR (4 Gy))
Figure 3
Figure 3
Autophagy delays radiation-induced growth in vivo. (ac) Effects of ATG5 or BECN1 knockdowns on the growth of irradiated A549 or H460 tumours. Five nude BALB/c mice were subcutaneously injected with ATG5- or BECN1-depleted A549 cells (a and b) or H460 cells (c). Once xenografted tumours became palpable, mice were randomly assigned to a control group or to a group that received localized tumour irradiation with a single dose 8 Gy. Then, tumour volumes were monitored every 2–4 days. Quantitative data are reported as means±S.E.M. (Student's t-test, **P<0.01, ***P<0.001, as compared with the control group)
Figure 4
Figure 4
Autophagy inhibition decreases irradiation-induced ATP release and impairs anti-tumoural immune response. (ac) Role of ATG5 on the growth of irradiated murine CT26 tumours implanted on immunodeficient mice. Nude BALB/c mice were subcutaneously injected with SCR or ATG5-depleted CT26 cells, then irradiated or left untreated with (c) or without an inhibitor of ecto-ATPase ARL67156 (ARL) (a). Tumour volumes were monitored every 3 or 4 days by direct measurement, as previously described. (b) Same experiment as in (a) in an immunocompetent host. Wild-type BALB/c mice were subcutaneously injected with ATG5-depleted CT26 cells, left untreated or irradiated, then tumour volume was monitored. Quantitative data presented are reported as means±S.E.M. (Student's t-test, *P<0.05, **P<0.01). (d and e) Tumour growth of irradiated tumours is inhibited by ATP release via autophagy. To determine the impact of irradiation and autophagy on ATP release, ATG5depleted CT26 (ATG5KD) or control (SCR) cells were irradiated with 4 Gy and incubated with or without an inhibitor of ecto-ATPase ARL67156 (ARL). ATP release was determined in vitro by using enzymatic methods at indicated times (e). Quantitative data are reported as means±S.E.M. (n=3 triplicate samples, Student's t-test, *P<0.05 comparing ATG5KD (4 Gy+ARL) with ATG5KD (Co) at 24 h; #P<0.05 comparing ATG5KD (4 Gy+ARL) with ATG5KD (4 Gy) at 24 h). To assess the impact of extracellular ATP release by irradiation on tumour growth, Balb/c mice were injected with scramble or Atg5-depleted CT26 tumour cells, then xenografted tumours were randomly assigned to treatment groups that received localized tumour irradiation with one single 8 Gy irradiation, with ARL (d). Tumour volumes were determined as indicated above and quantitative data are reported as means±S.E.M. (Student's t-test, ***P<0.001)
Figure 5
Figure 5
ARL67156 restores the lymphocyte infiltration in Atg5-depleted CT26 tumour after irradiation. (a) Representative pictures of CT26-recovered tumours. Scale bar, 10 μm. Arrows point lymphocytes. Quantitative data are reported in (b). Samples were compared using one-tailed Student's t-test. Error bars indicate S.E.M. **P<0.01, comparing SCR (IR) with SCR (Co) and SCR (IR+ARL) with SCR (Co); ##P<0.01, comparing SCR (IR+ARL) with SCR (IR); **P<0.01 comparing ATG5KD (IR) with ATG5KD (Co) and ATG5KD (IR+ARL) with ATG5KD (Co); ##P<0.01 comparing ATG5KD (IR+ARL) with ATG5KD (IR) in (b)

References

    1. Gudkov AV, Komarova EA. The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer. 2003;3:117–129.
    1. Eriksson D, Stigbrand T. Radiation-induced cell death mechanisms. Tumour Biol. 2010;31:363–372.
    1. Paglin S, Hollister T, Delohery T, Hackett N, McMahill M, Sphicas E, et al. A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Res. 2001;61:439–444.
    1. Stewart RD, Yu VK, Georgakilas AG, Koumenis C, Park JH, Carlson DJ. Effects of radiation quality and oxygen on clustered DNA lesions and cell death. Radiat Res. 2011;176:587–602.
    1. Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell. 2011;147:728–741.
    1. Marino G, Martins I, Kroemer G. Autophagy in Ras-induced malignant transformation: fatal or vital. Mol Cell. 2011;42:1–3.
    1. Mizushima N. The pleiotropic role of autophagy: from protein metabolism to bactericide. Cell Death Differ. 2005;12 (Suppl 2:1535–1541.
    1. Klionsky DJ, Cregg JM, Dunn WA, Jr., Emr SD, Sakai Y, Sandoval IV, et al. A unified nomenclature for yeast autophagy-related genes. Dev Cell. 2003;5:539–545.
    1. Klionsky DJ, Emr SD. Autophagy as a regulated pathway of cellular degradation. Science. 2000;290:1717–1721.
    1. Ohsumi Y. Molecular dissection of autophagy: two ubiquitin-like systems. Nat Rev Mol Cell Biol. 2001;2:211–216.
    1. Petiot A, Ogier-Denis E, Blommaart EF, Meijer AJ, Codogno P. Distinct classes of phosphatidylinositol 3'-kinases are involved in signaling pathways that control macroautophagy in HT-29 cells. J Biol Chem. 2000;275:992–998.
    1. Yang Z, Klionsky DJ. Eaten alive: a history of macroautophagy. Nat Cell Biol. 2010;12:814–822.
    1. Rabinowitz JD, White E. Autophagy and metabolism. Science. 2010;330:1344–1348.
    1. Ito H, Daido S, Kanzawa T, Kondo S, Kondo Y. Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int J Oncol. 2005;26:1401–1410.
    1. Apel A, Herr I, Schwarz H, Rodemann HP, Mayer A. Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res. 2008;68:1485–1494.
    1. Chen YS, Song HX, Lu Y, Li X, Chen T, Zhang Y, et al. Autophagy inhibition contributes to radiation sensitization of esophageal squamous carcinoma cells. Dis Esophagus. 2011;24:437–443.
    1. Kim KW, Moretti L, Mitchell LR, Jung DK, Lu B. Combined Bcl-2/mammalian target of rapamycin inhibition leads to enhanced radiosensitization via induction of apoptosis and autophagy in non-small cell lung tumor xenograft model. Clin Cancer Res. 2009;15:6096–6105.
    1. Lin CI, Whang EE, Abramson MA, Jiang X, Price BD, Donner DB, et al. Autophagy: a new target for advanced papillary thyroid cancer therapy. Surgery. 2009;146:1208–1214.
    1. Lomonaco SL, Finniss S, Xiang C, Decarvalho A, Umansky F, Kalkanis SN, et al. The induction of autophagy by gamma-radiation contributes to the radioresistance of glioma stem cells. Int J Cancer. 2009;125:717–722.
    1. Stone HB, Peters LJ, Milas L. Effect of host immune capability on radiocurability and subsequent transplantability of a murine fibrosarcoma. J Natl Cancer Inst. 1979;63:1229–1235.
    1. Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–1059.
    1. Ladoire S, Hannani D, Vetizou M, Locher C, Aymeric L, Apetoh L, et al. Cell death associated molecular patterns (CDAMP) as determinants of cancer immunogenicity Antiox Redox Signal 2013. e-pub ahead of print 8 February 2013; PubMed PMID: 23394620.
    1. Obeid M, Panaretakis T, Joza N, Tufi R, Tesniere A, van Endert P, et al. Calreticulin exposure is required for the immunogenicity of gamma-irradiation and UVC light-induced apoptosis. Cell Death Differ. 2007;14:1848–1850.
    1. Obeid M, Tesniere A, Panaretakis T, Tufi R, Joza N, van Endert P, et al. Ecto-calreticulin in immunogenic chemotherapy. Immunol Rev. 2007;220:22–34.
    1. Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Walk SF, et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature. 2009;461:282–286.
    1. Tsukimoto M, Homma T, Ohshima Y, Kojima S. Involvement of purinergic signaling in cellular response to gamma radiation. Radiat Res. 2010;173:298–309.
    1. Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 2011;334:1573–1577.
    1. Haas-Kogan DA, Dazin P, Hu L, Deen DF, Israel A. P53-independent apoptosis: a mechanism of radiation-induced cell death of glioblastoma cells. Cancer J Sci Am. 1996;2:114–121.
    1. Chautan M, Chazal G, Cecconi F, Gruss P, Golstein P. Interdigital cell death can occur through a necrotic and caspase-independent pathway. Curr Biol. 1999;9:967–970.
    1. Yuan J, Kroemer G. Alternative cell death mechanisms in development and beyond. Genes Dev. 2010;24:2592–2602.
    1. Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012;19:107–120.
    1. Liang B, Kong D, Liu Y, Liang N, He M, Ma S, et al. Autophagy inhibition plays the synergetic killing roles with radiation in the multi-drug resistant SKVCR ovarian cancer cells. Radiat Oncol. 2012;7:213.
    1. Wang K, Klionsky DJ. Mitochondria removal by autophagy. Autophagy. 2011;7:297–300.
    1. White E, DiPaola RS. The double-edged sword of autophagy modulation in cancer. Clin Cancer Res. 2009;15:5308–5316.
    1. Mancias JD, Kimmelman AC. Targeting autophagy addiction in cancer. Oncotarget. 2011;2:1302–1306.
    1. Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen HY, et al. Autophagy suppresses tumorigenesis through elimination of p62. Cell. 2009;137:1062–1075.
    1. Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest. 2003;112:1809–1820.
    1. Marino G, Salvador-Montoliu N, Fueyo A, Knecht E, Mizushima N, Lopez-Otin C. Tissue-specific autophagy alterations and increased tumorigenesis in mice deficient in Atg4C/autophagin-3. J Biol Chem. 2007;282:18573–18583.
    1. Raman D, Baugher PJ, Thu YM, Richmond A. Role of chemokines in tumor growth. Cancer Lett. 2007;256:137–165.
    1. Mantovani A, Romero P, Palucka AK, Marincola FM. Tumour immunity: effector response to tumour and role of the microenvironment. Lancet. 2008;371:771–783.
    1. Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G. Immunological aspects of cancer chemotherapy. Nat Rev Immunol. 2008;8:59–73.
    1. Saitoh T, Akira S. Regulation of innate immune responses by autophagy-related proteins. J Cell Biol. 2010;189:925–935.
    1. Mandapathil M, Hilldorfer B, Szczepanski MJ, Czystowska M, Szajnik M, Ren J, et al. Generation and accumulation of immunosuppressive adenosine by human CD4+CD25highFOXP3+ regulatory T cells. J Biol Chem. 2010;285:7176–7186.

Source: PubMed

3
Subskrybuj