Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis

Subjects

An Author Correction to this article was published on 08 October 2020

This article has been updated

Abstract

Although pyroptosis is critical for macrophages against pathogen infection, its role and mechanism in cancer cells remains unclear. PD-L1 has been detected in the nucleus, with unknown function. Here we show that PD-L1 switches TNFα-induced apoptosis to pyroptosis in cancer cells, resulting in tumour necrosis. Under hypoxia, p-Stat3 physically interacts with PD-L1 and facilitates its nuclear translocation, enhancing the transcription of the gasdermin C (GSDMC) gene. GSDMC is specifically cleaved by caspase-8 with TNFα treatment, generating a GSDMC N-terminal domain that forms pores on the cell membrane and induces pyroptosis. Nuclear PD-L1, caspase-8 and GSDMC are required for macrophage-derived TNFα-induced tumour necrosis in vivo. Moreover, high expression of GSDMC correlates with poor survival. Antibiotic chemotherapy drugs induce pyroptosis in breast cancer. These findings identify a non-immune checkpoint function of PD-L1 and provide an unexpected concept that GSDMC/caspase-8 mediates a non-canonical pyroptosis pathway in cancer cells, causing tumour necrosis.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: PD-L1 translocates into the nucleus in response to hypoxia.
Fig. 2: p-Y705-Stat3 binds to PD-L1 and facilitates its nuclear translocation under hypoxia.
Fig. 3: nPD-L1 switches TNFα-induced apoptosis to pyroptosis under hypoxia.
Fig. 4: nPD-L1 and p-Y705-Stat3 act as co-activators in the transcriptional activation of GSDMC expression in response to hypoxia.
Fig. 5: GSDMC cleavage by caspase-8 determines the hypoxia-induced apoptosis-to-pyroptosis switch with TNFα treatment.
Fig. 6: The N-terminal domain of GSDMC (amino acids 1–365) binds to the cell membrane and induces pyroptosis.
Fig. 7: GSDMC, nPD-L1 and caspase-8 are required for TNFα-induced tumour necrosis in hypoxic regions.
Fig. 8: Chemotherapeutic drugs induce pyroptotic cell death via the nPD-L1/GSDMC-mediated non-canonical pathway in breast tumours.

Similar content being viewed by others

Data availability

The consensus caspase-8 recognition motifs were analysed using the CaspDB database (http://caspdb.sanfordburnham.org/). The p-Y705-Stat3-binding site in the GSDMC promoter was analysed using the GPMiner program (http://gpminer.mbc.nctu.edu.tw/). All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Change history

References

  1. Shi, J. et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526, 660–665 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Kayagaki, N. et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526, 666–671 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Liu, X. et al. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature 535, 153–158 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Shi, J. et al. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature 514, 187–192 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Ding, J. et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature 535, 111–116 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Wang, Y. et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 547, 99–103 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Rogers, C. et al. Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death. Nat. Commun. 8, 14128 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wang, Q. et al. A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature 579, 421–426 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Zhang, Z. et al. Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature 579, 415–420 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhou, Z. et al. Granzyme A from cytotoxic lymphocytes cleaves GSDMB to trigger pyroptosis in target cells. Science 368, eaaz7548 (2020).

  11. Kovacs, S. B. & Miao, E. A. Gasdermins: effectors of pyroptosis. Trends Cell Biol. 27, 673–684 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Powles, T. et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature 515, 558–562 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Schmid, P. et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N. Engl. J. Med. 379, 2108–2121 (2018).

    Article  CAS  PubMed  Google Scholar 

  15. Socinski, M. A. et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N. Engl. J. Med. 378, 2288–2301 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Satelli, A. et al. Potential role of nuclear PD-L1 expression in cell-surface vimentin positive circulating tumor cells as a prognostic marker in cancer patients. Sci. Rep. 6, 28910 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ghebeh, H. et al. Doxorubicin downregulates cell surface B7-H1 expression and upregulates its nuclear expression in breast cancer cells: role of B7-H1 as an anti-apoptotic molecule. Breast Cancer Res. 12, R48 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Shimizu, S. et al. Induction of apoptosis as well as necrosis by hypoxia and predominant prevention of apoptosis by Bcl-2 and Bcl-XL. Cancer Res. 56, 2161–2166 (1996).

    CAS  PubMed  Google Scholar 

  19. Grivennikov, S. I., Greten, F. R. & Karin, M. Immunity, inflammation, and cancer. Cell 140, 883–899 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Scaffidi, P., Misteli, T. & Bianchi, M. E. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 418, 191–195 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Vakkila, J. & Lotze, M. T. Inflammation and necrosis promote tumour growth. Nat. Rev. Immunol. 4, 641–648 (2004).

    Article  CAS  PubMed  Google Scholar 

  22. Carswell, E. et al. An endotoxin-induced serum factor that causes necrosis of tumors. Proc. Natl Acad. Sci. USA 72, 3666–3670 (1975).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Noman, M. Z. et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 211, 781–790 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Pawlus, M. R., Wang, L. & Hu, C. J. STAT3 and HIF1α cooperatively activate HIF1 target genes in MDA-MB-231 and RCC4 cells. Oncogene 33, 1670–1679 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Yang, Y. et al. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 28, 862–864 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Li, C.-W. et al. Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat. Commun. 7, 12632 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Timmer, J. & Salvesen, G. Caspase substrates. Cell Death Differ. 14, 66–72 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Poręba, M., Stróżyk, A., Salvesen, G. S. & Drąg, M. Caspase substrates and inhibitors. Cold Spring Harb. Perspect. Biol. 5, a008680 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Salvesen, G. S. & Ashkenazi, A. Snapshot: caspases. Cell 147, 476–476.e1 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Brown, J. M. & Wilson, W. R. Exploiting tumour hypoxia in cancer treatment. Nat. Rev. Cancer 4, 437–447 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Murdoch, C., Muthana, M., Coffelt, S. B. & Lewis, C. E. The role of myeloid cells in the promotion of tumour angiogenesis. Nat. Rev. Cancer 8, 618–631 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Barkal, A. A. et al. Engagement of MHC class I by the inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy. Nat. Immunol. 19, 76–84 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Karagiannis, G. S. et al. Neoadjuvant chemotherapy induces breast cancer metastasis through a TMEM-mediated mechanism. Sci. Transl. Med. 9, eaan0026 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Aglietti, R. A. & Dueber, E. C. Recent insights into the molecular mechanisms underlying pyroptosis and gasdermin family functions. Trends Immunol. 38, 261–271 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Zhang, M. et al. Programmed death-ligand 1 triggers PASMCs pyroptosis and pulmonary vascular fibrosis in pulmonary hypertension. J. Mol. Cell. Cardiol. 138, 23–33 (2020).

    Article  CAS  PubMed  Google Scholar 

  36. Ehrnhoefer, D. E. et al. p53 increases caspase-6 expression and activation in muscle tissue expressing mutant huntingtin. Hum. Mol. Genet. 23, 717–729 (2013).

    Article  PubMed  Google Scholar 

  37. Curiel, T. J. et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 10, 942–949 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Lin, H. et al. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression. J. Clin. Investig. 128, 805–815 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Tang, H. et al. PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression. J. Clin. Investig. 128, 580–588 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Kumar, V. et al. CD45 phosphatase inhibits STAT3 transcription factor activity in myeloid cells and promotes tumor-associated macrophage differentiation. Immunity 44, 303–315 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Edwards, J. G. et al. Tumor necrosis correlates with angiogenesis and is a predictor of poor prognosis in malignant mesothelioma. Chest 124, 1916–1923 (2003).

    Article  PubMed  Google Scholar 

  42. Koukourakis, M. I. et al. Lactate dehydrogenase-5 (LDH-5) overexpression in non-small-cell lung cancer tissues is linked to tumour hypoxia, angiogenic factor production and poor prognosis. Br. J. Cancer 89, 877–885 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Obeid, M. et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat. Med. 13, 54–61 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Krysko, D. V. et al. Immunogenic cell death and DAMPs in cancer therapy. Nat. Rev. Cancer 12, 860–875 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Galluzzi, L., Buque, A., Kepp, O., Zitvogel, L. & Kroemer, G. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell 28, 690–714 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Erkes, D. A. et al. Mutant BRAF and MEK inhibitors regulate the tumor immune microenvironment via pyroptosis. Cancer Discov. 10, 254–269 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. Kosugi, S. et al. Six classes of nuclear localization signals specific to different binding grooves of importin α. J. Biol. Chem. 284, 478–485 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. Bassi, C. et al. Nuclear PTEN controls DNA repair and sensitivity to genotoxic stress. Science 341, 395–399 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kosugi, S., Hasebe, M., Tomita, M. & Yanagawa, H. Systematic identification of cell cycle-dependent yeast nucleocytoplasmic shuttling proteins by prediction of composite motifs. Proc. Natl Acad. Sci. USA 106, 10171–10176 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Pichler, A., Gast, A., Seeler, J. S., Dejean, A. & Melchior, F. The nucleoporin RanBP2 has SUMO1 E3 ligase activity. Cell 108, 109–120 (2002).

    Article  CAS  PubMed  Google Scholar 

  51. Lo, H. W. et al. Nuclear interaction of EGFR and STAT3 in the activation of the iNOS/NO pathway. Cancer Cell 7, 575–589 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Lin, S.-Y. et al. Nuclear localization of EGF receptor and its potential new role as a transcription factor. Nat. Cell Biol. 3, 802–808 (2001).

    Article  CAS  PubMed  Google Scholar 

  53. Shen, J. et al. EGFR modulates microRNA maturation in response to hypoxia through phosphorylation of AGO2. Nature 497, 383–387 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Chang, C. J. et al. EZH2 promotes expansion of breast tumor initiating cells through activation of RAF1-β-catenin signaling. Cancer Cell 19, 86–100 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Cha, T.-L. et al. Akt-mediated phosphorylation of EZH2 suppresses methylation of lysine 27 in histone H3. Science 310, 306–310 (2005).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Dunner Jr. and acknowledge the National Institutes of Health Cancer Center (Support grant no. P30CA016672) for supporting the High Resolution Electron Microscopy Facility at The University of Texas MD Anderson Cancer Center for the electron microscopy analysis. This work was funded in part by the National Institutes of Health (grant nos P30CA016672 and R01 AI116722 (to M.-C.H.)); a National Breast Cancer Foundation Breast Cancer Research Foundation grant to M.-C.H.; the Patel Memorial Breast Cancer Endowment Fund; the Sister Institution Network Fund for collaboration between The University of Texas MD Anderson Cancer Center and China Medical University (to M.-C.H.); the YingTsai Young Scholar Award (grant no. CMU108-YTY02 to J.-M.H.); China Medical Univesity Start Up Fund (grant no. CMU108-N-02 to W.-J.W.); the NIH (grant nos R35 CA220430 and PO1 CA092584); the Cancer Prevention and Research Institute of Texas (grant no. RP180813); Robert A. Welch Chemistry Chair (to J.A.T.); Cancer Prevention and Research Institute of Texas (grant no. RP160710 to M.-C.H.); and Drug Development Center, Ministry of Education, Taiwan (grant no. 108-2311-B-241-001 to M.-C.H. and Y.C.).

Author information

Authors and Affiliations

Authors

Contributions

J.H. and M.-C.H. designed and conceived the study. J.H. and M.-C.H. wrote the manuscript. J.L.H. and K.H. contributed to the preparation of the manuscript. J.H., R.Z., W.X., Y. You, J.-M.H., Y.C., Y.-C.W., C.L., W.-J.W., B.K., Z.Y., Y. Yang, X.X., Y.L., C.-W.L., B.S. and J.A.T. performed experiments and analysed data. L.N. and C.-W.C. analysed the PD-L1 and GSDMC sequences. Y.W. provided patient tissue samples. M.-C.H. supervised the entire project.

Corresponding author

Correspondence to Mien-Chie Hung.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Establishment of stable cells in the MDA-MB-231 cell line.

(a) Deletion of endogenous Stat3 by CRISPR–Cas9. KO, knockout. Blots are representative of three independent experiments. (b) Stable re-expression of Stat3-WT or Stat3-Y705F in Stat3-knockout MDA-MB-231 cells. Validation of p-Y705-Stat3 expression level by immunoblotting. Blots are representative of three independent experiments. (c) Analysis of the nuclear localization signal (NLS) and nuclear export signal (NES) in the human PD-L1 amino acid sequence compared with that of other species. Sequence alignment of PD-L1 from human, mouse, cow, rat, and dog. The alignment was generated and presented by the ClustalW2 algorithm and ESPript 3.0 (http://espript.ibcp.fr/ESPript/cgi-bin/ESPript.cgi). Identical residues are indicated by the dark red background and conserved residues are in red. The NLS and NES sequences of PD-L1 are conserved across species. (d-f) Stable re-expression of PD-L1-WT (wild-type PD-L1), PD-L1-NLS (NLS-mutated PD-L1) or PD-L1-NES (NES-mutated PD-L1) in the PD-L1–knockout MDA-MB-231 cell line. Immunoblotting of total PD-L1 expression level in stable transfectants (d). Blots are representative of three independent experiments. Representative images of PD-L1 distribution of three independent experiments was determined by confocal assay under hypoxia (e-f). Scale bar, 20 μm. Unprocessed blots are provided in Source Data Extended Data Fig. 1.

Source data

Extended Data Fig. 2 nPD-L1-mediated pyroptotic cell death in cell lines of multiple cancer types.

(a) Immunoblotting analysis of PD-L1 expression levels in liver cancer and lung cancer cell lines. Blots are representative of three independent experiments. (b) Hypoxia-induced pyroptotic cell death in PD-L1-positive cells of multiple cancer types treated with p-Stat3 inhibitor, HO-3867. Red arrows indicate the pyroptotic cells with big bubbles. The experiment was repeated three times with similar results. Scale bar, 20 μm. Unprocessed blots are provided in Source Data Extended Data Fig. 2.

Source data

Extended Data Fig. 3 nPD-L1 and p-Y705-Stat3 are required for hypoxia-induced GSDMC expression.

(a) Real-time quantitative polymerase chain reaction (RT–qPCR) analysis of mRNA levels of gasdermin family members in the indicated MDA-MB-231 stable transfectants. Data are shown as mean ± SD of n = 3 independent experiments. (b and c) GSDMC mRNA (b) and protein (c) levels in MDA-MB-231 cells under hypoxia. Data of (b) are shown as mean ± SD of n = 3 independent experiments. Blots are representative of three independent experiments. N, normoxia; H, hypoxia. (d) RT–qPCR analysis of mRNA levels of GSDMC in the indicated MDA-MB-231 stable transfectants. Data are shown as mean ± SD of n = 3 independent experiments. (e-g) MDA-MB-231 cells were cultured under hypoxia and treated with HO-3867 (20 μM) or ivermectin (25 μM). GSDMC mRNA (e) and protein (f) levels were analysed by RT–qPCR and immunoblotting, respectively. GSDMC promoter luciferase reporter plasmids were transfected to cells, and luciferase activity was measured in (g). Data of (e) and (g) are shown as mean ± SD of n = 3 independent experiments. Blots are representative of three independent experiments. P values of all statistical analysis were determined by two-sided Student’s t-test. Statistical source data and unprocessed blots are provided in Source Data Extended Data Fig. 3.

Source data

Extended Data Fig. 4 The p-Y705-Stat3 binding site (in red) in the nucleic acid sequence of GSDMC promoter and the expression level of GSDME in MDA-MB-231 cells.

(a) GSDMC promoter sequence was obtained from GeneCopoeia and analysed using the GPMiner program (http://gpminer.mbc.nctu.edu.tw/). Two putative p-Y705-Stat3-binding sites in the GSDMC promoter were predicted. p-Y705-Stat3-binding site was validated by screening with luciferase reporter assay. (b-d) Immunoblotting of GSDME in MDA-MB-231 cells. H226 cells as a positive control. Immunoblotting of the endogenous GSDME (b). Immunoblotting of GSDME in MDA-MB-231 cells under hypoxia (c). Immunoblotting of GSDME in MDA-MB-231 cells treated with daunorubicin, doxorubicin, epirubicin, and actinomycin D (d). The experiments of (b-d) were repeated three times with similar results respectively. Unprocessed blots are provided in Source Data Extended Data Fig. 4.

Source data

Extended Data Fig. 5 Caspase-8, but not caspase-6, is required for TNFα-induced pyroptosis under hypoxia.

(a and b) MDA-MB-231 cells were cultured under normoxia (N) or hypoxia (H) and treated as indicated. LDH-released cell death was measured as shown in (a). Data are shown as mean ± SD of n = 3 independent experiments. P values were determined by two-sided Student’s t-test. Representative images of dying cell morphology of three independent experiments (b). Red arrows indicate cell swelling with big bubbles. Scale bar, 20 μm. Caspase-3i, caspase-3 inhibitor Z-DEVD-FMK; Caspase-6i, caspase-6 inhibitor Z-VEID-FMK; Caspase-8i, caspase-8 inhibitor Z-IETD-FMK (each at 10 μM). Statistical source data are provided in Source Data Extended Data Fig. 5.

Source data

Extended Data Fig. 6 The pyroptotic cell death in cell lines with different GSDMC and PD-L1 expression status.

(a) Immunoblotting of PD-L1 and GSDMC in Hs578T, BT549, MDA-MB-231 and MCF-7 cells. Blots are representative of three independent experiments. (b) LDH-released cell death in Hs578T, BT549, MDA-MB-231 and MCF-7 cells treated with TNFα under normoxia or hypoxia. Data are shown as mean ± SD of n = 3 independent experiments. P values were determined by two-sided Student’s t-test. Statistical source data and unprocessed blots are provided in Source Data Extended Data Fig. 6.

Source data

Extended Data Fig. 7 The N-terminal domain (1–365) of GSDMC is sufficient to induce pyroptosis.

(a) 3D structural modelling of GSDMC. The modelling was performed using SWISS-MODEL. The structural fold of GSDMC N- and C-terminal domains is in blue cartoon presentation. The linker region and the solvent-exposed loop region of the C-terminal domain of GSDMC are coloured in green with numbered Asp/Glu residues coloured in magenta. The structural figure was prepared using PyMOL Molecular Graphics System, Version 2.0 Schrödinger, LLC. (b) Confocal microscopy analysis of the distribution of GSDMC-FL, GSDMC (1¬–365), and GSDMD-N expressions in HeLa cells. GSDMD-N, N-terminal domain of GSDMD. Images are representative of three independent experiments. Scale bar, 20 μm. (c–d) HeLa cells were transfected with GSDMC-FL or GSDMC (1–365). Representative images of dying cell morphology of three independent experiments (c). Scale bar, 20 μm. Dying cells were stained with nucleic acid dye SYTOX green. Red arrows indicate cell swelling with large bubbles. LDH-released cell death is shown as mean ± SD of n = 3 independent experiments (d). P values were determined by two-sided Student’s t-test. (e) HEK293T cells were transfected with Flag-GSDMC-FL or Flag-GSDMC (1–365) for 16 h. Supernatants and cell pellets were collected and subjected to immunoblotting. The blot is representative of three independent experiments. (f) Cartoon diagram of GSDMC structure and the cleavage by caspase-8 across species. Statistical source data and unprocessed blots are provided in Source Data Extended Data Fig. 7.

Source data

Extended Data Fig. 8 Nuclear PD-L1 translocation in vivo causes poor survival in both immunocompetent and nude mice.

(a) IHC staining of HIF1α, GSDMC, PD-L1, and p-Y705-Stat3 in MDA-MB-231 xenografts in nude mice (n = 8). MDA-MB-231 cells were injected subcutaneously into nude mice. Four weeks after tumour cell injection, all mice were sacrificed and tumours excised for IHC staining. The expression levels and patterns of HIF1α, GSDMC, PD-L1, and p-Y705-Stat3 in normoxic and hypoxic area in tumours were analysed and indicated by green arrows. Scale bar, 100 μm. The experiment was repeated three times with similar results. (b) Survival analysis of 4T1 cells expressing PD-L1-WT or PD-L1-NLS in BALB/c nude mice. Data are shown of n = 10 mice. The experiment was repeated three times with similar results. Statistical significance for survival analysis was determined using a log-rank (Mantel–Cox) test. (c) Survival analysis of 4T1 cells expressing PD-L1-WT or PD-L1-NLS in immunocompetent BALB/c mice. Data are shown of n = 10 mice. The experiment was repeated three times with similar results. Statistical significance for survival analysis was determined using a log-rank (Mantel–Cox) test. Statistical source data are provided in Source Data Extended Data Fig. 8.

Source data

Extended Data Fig. 9 Chemotherapeutic drugs induce pyroptotic cell death by nPD-L1/GSDMC-mediated non-canonical pathway in breast tumour.

(a) Representative images of dying cell morphology by phase-contrast microscopy in MDA-MB-231 stable transfectants as indicated treated with daunorubicin, doxorubicin, epirubicin, and actinomycin D. The experiment was repeated three times with similar results. Scale bar, 20 μm. (b) LDH-released cell death in MDA-MB-231 stable transfectants as indicated treated with daunorubicin, doxorubicin, epirubicin, and actinomycin D. Data are shown as mean ± SD of n = 3 independent experiments. P values were determined by two-sided Student’s t-test. Statistical source data are provided in Source Data Extended Data Fig. 9.

Source data

Extended Data Fig. 10 A proposed model of nuclear PD-L1-mediated apoptosis-to-pyroptosis switch under hypoxia.

Under normoxia, caspase-3 can be activated by TNFα-induced caspase-8 to cause apoptotic cell death. However, hypoxia-activated p-Stat3 facilitates nPD-L1 translocation. nPD-L1 cooperates with p-Stat3 to transcriptionally activate GSDMC expression. GSDMC is specifically cleaved by macrophage-derived TNFα-activated caspase-8, which generates an N-terminal pore-forming fragment that causes pyroptotic cell death to induce tumour necrosis in breast cancer.

Supplementary information

Reporting Summary

Supplementary Video 1

MDA-MB-231–GFP cells were treated with TNFα plus CHX under normoxia. MDA-MB-231–GFP, MDA-MB-231 cells with stable expression of GFP protein.

Supplementary Video 2

MDA-MB-231–GFP cells were treated with TNFα plus CHX under hypoxia.

Supplementary Video 3

MDA-MB-231–GFP cells were co-treated with TNFα plus CHX and the Stat3 inhibitor HO-3867 (20 μM) under hypoxia.

Supplementary Video 4

MDA-MB-231–GFP cells were co-treated with TNFα plus CHX and caspase-8 inhibitor Z-IETD-FMK (10 μM) under hypoxia.

Supplementary Table 1

The final concentrations for TNFα, CHX and various stimuli.

Source data

Source Data Fig. 1

Statistical source data

Source Data Fig. 1

Unprocessed western blots

Source Data Fig. 2

Unprocessed western blots

Source Data Fig. 3

Statistical source data

Source Data Fig. 4

Statistical source data

Source Data Fig. 4

Unprocessed western blots

Source Data Fig. 5

Statistical source data

Source Data Fig. 5

Unprocessed western blots

Source Data Fig. 6

Statistical source data

Source Data Fig. 6

Unprocessed western blots and gels

Source Data Fig. 7

Statistical source data

Source Data Fig. 8

Statistical source data

Source Data Fig. 8

Unprocessed western blots

Source Data Extended Data Fig. 1

Unprocessed western blots

Source Data Extended Data Fig. 2

Unprocessed western blots

Source Data Extended Data Fig. 3

Statistical source data

Source Data Extended Data Fig. 3

Unprocessed western blots

Source Data Extended Data Fig. 4

Unprocessed western blots

Source Data Extended Data Fig. 5

Statistical source data

Source Data Extended Data Fig. 6

Statistical source data

Source Data Extended Data Fig. 6

Unprocessed western blots

Source Data Extended Data Fig. 7

Statistical source data

Source Data Extended Data Fig. 7

Unprocessed western blots

Source Data Extended Data Fig. 8

Statistical source data

Source Data Extended Data Fig. 9

Statistical source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hou, J., Zhao, R., Xia, W. et al. PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis. Nat Cell Biol 22, 1264–1275 (2020). https://doi.org/10.1038/s41556-020-0575-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-020-0575-z

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer