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.

  • Article
  • Published:

Intestinal inhibition of Atg7 prevents tumour initiation through a microbiome-influenced immune response and suppresses tumour growth

Subjects

Abstract

Here, we show that autophagy is activated in the intestinal epithelium in murine and human colorectal cancer and that the conditional inactivation of Atg7 in intestinal epithelial cells inhibits the formation of pre-cancerous lesions in Apc+/− mice by enhancing anti-tumour responses. The antibody-mediated depletion of CD8+ T cells showed that these cells are essential for the anti-tumoral responses mediated by the inhibition of autophagy. We show that Atg7 deficiency leads to intestinal dysbiosis and that the microbiota is required for anticancer responses. In addition, Atg7 deficiency resulted in a stress response accompanied by metabolic defects, AMPK activation and p53-mediated cell-cycle arrest in tumour cells but not in normal tissue. This study reveals that the inhibition of autophagy within the epithelium may prevent the development and progression of colorectal cancer in genetically predisposed patients.

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

Access options

Buy this article

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

Figure 1: Autophagy is active in vivo during murine and human intestinal tumorigenesis.
Figure 2: Atg7 deficiency strongly attenuates the initiation and progression of intestinal tumours resulting from APC loss.
Figure 3: Expression of genes related to immunity in non-tumoral intestinal mucosa of Apc+/−Atg7−/− mice.
Figure 4: T-cell infiltration in Apc+/−Atg7−/− mice is associated with an anti-tumour immune response.
Figure 5: Gut mucosal integrity is disrupted in Apc+/−Atg7−/− mice.
Figure 6: The microbiota is essential for immune anti-tumoral response in Apc+/−Atg7−/− mice.
Figure 7: Atg7 deficiency strongly attenuates adenoma cell proliferation.
Figure 8: AMPK signalling is activated in adenomas from Apc+/−Atg7−/− mice.

Similar content being viewed by others

References

  1. Liang, X. H. et al. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 402, 672–676 (1999).

    Article  CAS  PubMed  Google Scholar 

  2. Degenhardt, K. et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 10, 51–64 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Mathew, R., Karantza-Wadsworth, V. & White, E. Role of autophagy in cancer. Nat. Rev. Cancer 7, 961–967 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Mathew, R. et al. Autophagy suppresses tumorigenesis through elimination of p62. Cell 137, 1062–1075 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wei, H. et al. Suppression of autophagy by FIP200 deletion inhibits mammary tumorigenesis. Genes Dev. 25, 1510–1527 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Yang, S. et al. Pancreatic cancers require autophagy for tumor growth. Genes Dev. 25, 717–729 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Guo, J. Y. et al. Autophagy suppresses progression of K-ras-induced lung tumors to oncocytomas and maintains lipid homeostasis. Genes Dev. 27, 1447–1461 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tjalsma, H., Boleij, A., Marchesi, J. R. & Dutilh, B. E. A bacterial driver–passenger model for colorectal cancer: beyond the usual suspects. Nat. Rev. Microbiol. 10, 575–582 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Korinek, V. et al. Constitutive transcriptional activation by a β-catenin-Tcf complex in APC-/- colon carcinoma. Science 275, 1784–1787 (1997).

    Article  CAS  PubMed  Google Scholar 

  10. Colnot, S. et al. Colorectal cancers in a new mouse model of familial adenomatous polyposis: influence of genetic and environmental modifiers. Lab. Invest. 84, 1619–1630 (2004).

    Article  CAS  PubMed  Google Scholar 

  11. Sansom, O. J. et al. Loss of Apc in vivo immediately perturbs Wnt signaling, differentiation, and migration. Genes Dev. 18, 1385–1390 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Andreu, P. et al. Crypt-restricted proliferation and commitment to the Paneth cell lineage following Apc loss in the mouse intestine. Development 132, 1443–1451 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Zeineldin, M. & Neufeld, K. L. More than two decades of Apc modeling in rodents. Biochim. Biophys. Acta 1836, 80–89 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Peignon, G. et al. Complex interplay between β-catenin signalling and Notch effectors in intestinal tumorigenesis. Gut 60, 166–176 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Komatsu, M. et al. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J. Cell Biol. 169, 425–434 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Petherick, K. J. et al. Autolysosomal β-catenin degradation regulates Wnt-autophagy-p62 crosstalk. EMBO J. 32, 1903–1916 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Dunn, G. P., Koebel, C. M. & Schreiber, R. D. Interferons, immunity and cancer immunoediting. Nat. Rev. Immunol. 6, 836–848 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Cerovic, V., Bain, C. C., Mowat, A. M. & Milling, S. W. Intestinal macrophages and dendritic cells: what’s the difference? Trends Immunol. 35, 270–277 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Tosolini, M. et al. Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, th2, treg, th17) in patients with colorectal cancer. Cancer Res. 71, 1263–1271 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Berg, D. J. et al. Enterocolitis and colon cancer in interleukin-10-deficient mice are associated with aberrant cytokine production and CD4+ TH1-like responses. J. Clin. Invest. 98, 1010–1020 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Mlecnik, B. et al. Biomolecular network reconstruction identifies T-cell homing factors associated with survival in colorectal cancer. Gastroenterology 138, 1429–1440 (2010).

    Article  CAS  PubMed  Google Scholar 

  22. Murai, M. et al. Interleukin 10 acts on regulatory T cells to maintain expression of the transcription factor FOXP3 and suppressive function in mice with colitis. Nat. Immunol. 10, 1178–1184 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Xiao, H. et al. Loss of single immunoglobulin interlukin-1 receptor-related molecule leads to enhanced colonic polyposis in Apc(min) mice. Gastroenterology 139, 574–585 (2010).

    Article  CAS  PubMed  Google Scholar 

  24. Cadwell, K., Stappenbeck, T. S. & Virgin, H. W. Role of autophagy and autophagy genes in inflammatory bowel disease. Curr. Top. Microbiol. Immunol. 335, 141–167 (2009).

    CAS  PubMed  Google Scholar 

  25. Patel, K. K. et al. Autophagy proteins control goblet cell function by potentiating reactive oxygen species production. EMBO J. 32, 3130–3144 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Serebrennikova, O. B. et al. Tpl2 ablation promotes intestinal inflammation and tumorigenesis in Apcmin mice by inhibiting IL-10 secretion and regulatory T-cell generation. Proc. Natl Acad. Sci. USA 109, E1082–E1091 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Berkers, C. R., Maddocks, O. D., Cheung, E. C., Mor, I. & Vousden, K. H. Metabolic regulation by p53 family members. Cell Metab. 18, 617–633 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Fujishita, T., Aoki, K., Lane, H. A., Aoki, M. & Taketo, M. M. Inhibition of the mTORC1 pathway suppresses intestinal polyp formation and reduces mortality in ApcΔ716 mice. Proc. Natl Acad. Sci. USA 105, 13544–13549 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Zhou, G. et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Invest. 108, 1167–1174 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Rao, S. et al. A dual role for autophagy in a murine model of lung cancer. Nat. Commun. 5, 3056 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Rosenfeldt, M. T. et al. p53 status determines the role of autophagy in pancreatic tumour development. Nature 504, 296–300 (2013).

    Article  CAS  PubMed  Google Scholar 

  32. Yang, A. et al. Autophagy is critical for pancreatic tumor growth and progression in tumors with p53 alterations. Cancer Discov. 4, 905–913 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Strohecker, A. M. et al. Autophagy sustains mitochondrial glutamine metabolism and growth of BrafV600E-driven lung tumors. Cancer Discov. 3, 1272–1285 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Michaud, M. et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 334, 1573–1577 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Loddenkemper, C. et al. In situ analysis of FOXP3+ regulatory T cells in human colorectal cancer. J. Transl. Med. 4, 52 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Pages, F. et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N. Engl. J. Med. 353, 2654–2666 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Sato, E. et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc. Natl Acad. Sci. USA 102, 18538–18543 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gao, Q. et al. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J. Clin. Oncol. 25, 2586–2593 (2007).

    Article  PubMed  Google Scholar 

  39. Castellino, F. & Germain, R. N. Cooperation between CD4+ and CD8+ T cells: when, where, and how. Annu. Rev. Immunol. 24, 519–540 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Toes, R. E., Ossendorp, F., Offringa, R. & Melief, C. J. CD4 T cells and their role in antitumor immune responses. J. Exp. Med. 189, 753–756 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Grimm, W. A. et al. The Thr300Ala variant in ATG16L1 is associated with improved survival in human colorectal cancer and enhanced production of type I interferon. Gut 10.1136/gutjnl-2014-308735 (2015).

  42. Cadwell, K. et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature 456, 259–263 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Marchiando, A. M. et al. A deficiency in the autophagy gene Atg16L1 enhances resistance to enteric bacterial infection. Cell Host Microbe 14, 216–224 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. Benjamin, J. L., Sumpter, R. Jr, Levine, B. & Hooper, L. V. Intestinal epithelial autophagy is essential for host defense against invasive bacteria. Cell Host Microbe 13, 723–734 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Conway, K. L. et al. Atg16l1 is required for autophagy in intestinal epithelial cells and protection of mice from Salmonella infection. Gastroenterology 145, 1347–1357 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Hu, B. et al. Microbiota-induced activation of epithelial IL-6 signaling links inflammasome-driven inflammation with transmissible cancer. Proc. Natl Acad. Sci. USA 110, 9862–9867 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Zhan, Y. et al. Gut microbiota protects against gastrointestinal tumorigenesis caused by epithelial injury. Cancer Res. 73, 7199–7210 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Paulos, C. M. et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J. Clin. Invest. 117, 2197–2204 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Viaud, S. et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 342, 971–976 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Iida, N. et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 342, 967–970 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ruffell, B. et al. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell 26, 623–637 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ghiringhelli, F. et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1β-dependent adaptive immunity against tumors. Nat. Med. 15, 1170–1178 (2009).

    Article  CAS  PubMed  Google Scholar 

  53. Weigmann, B. et al. Isolation and subsequent analysis of murine lamina propria mononuclear cells from colonic tissue. Nat. Protoc. 2, 2307–2311 (2007).

    Article  CAS  PubMed  Google Scholar 

  54. Lepage, P. et al. Biodiversity of the mucosa-associated microbiota is stable along the distal digestive tract in healthy individuals and patients with IBD. Inflamm. Bowel Dis. 11, 473–480 (2005).

    Article  PubMed  Google Scholar 

  55. Seksik, P. et al. Alterations of the dominant faecal bacterial groups in patients with Crohn’s disease of the colon. Gut 52, 237–242 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Caporaso, J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 7, 335–336 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Cole, J. R. et al. The ribosomal database project: improved alignments and new tools for rRNA analysis. Nucleic Acids Res. 37, D141–D145 (2009).

    Article  CAS  PubMed  Google Scholar 

  58. Segata, N. et al. Metagenomic biomarker discovery and explanation. Genome Biol. 12, R60 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Komatsu (Niigata University, Japan) and S. Robine (Institut Curie, France) for a generous supply of mutant mice, S. Pham, P. Mariani, Y. Bieche, S. Vacher, B. Violet, M. Foretz, B. Radenen-Bussiere and V. Maillet for technical help and for helpful discussions. We are grateful to the staff of Cochin’s animal housing facility and in particular to F. Lager and I. Lagoutte. This work was supported by Institut National du Cancer, the Comité de Paris de la Ligue Contre le Cancer, la Fondation Arc, and by the Cancer Research Personalized Medicine (CARPEM). W.C. was supported by Poste d’acceuil INSERM, M.F. and F.D. by CARPEM, and J.L. held a fellowship from the Ministère de la Recherche et de la Technologie and was also financially supported by la Fondation Arc. This work was also supported by funds from Inserm and by grants from the Fondation pour la Recherche Médicale (‘Equipe FRM 2013’ to M.C.).

Author information

Authors and Affiliations

Authors

Contributions

B.R. conceived and supervised the study, analysed the data and wrote the manuscript with the contribution of J.-P.C., P.L. and M.C. J.-P.C. supervised and analysed the immunological study and gave helpful insights and discussion. A.L. and A.-M.C. contributed to the immunological experiments M.C. and P.L. supervised and analysed the microbiota studies. M.D. contributed to the microbiota studies. J.L. designed, carried out and analysed most of the experiments with crucial help from W.C., M.A.B. and M.F. C.T. assisted with immunohistochemistry experiments and provided helpful comments of the manuscript. J.L. and A.D. performed genotyping and immunohistochemistry experiments. F.D. and F.L. performed the microarray and bioinformatic analysis. C.M. and G.R. acquired the ultrasound images. A.S. assisted with electron microscopy analysis. B.T. and W.C. provided and analysed the surgical specimens. C.P. provided helpful discussions.

Corresponding author

Correspondence to Béatrice Romagnolo.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 5 Autophagy-related gene expression in murine and human intestinal tumors.

(a) Representative LC3 immunostaining in colonic adenoma and adjacent non tumoral tissue from Apc+/− mice. (b) Transcript levels of Atg7, Atg5, Beclin1 and Lamp2 mRNA were analysed by qPCR in samples of small intestinal dysplasia (VilCreERT2Apcflox/flox mice 5 days post tamoxifen induction, Apc−/− mice) and adenomas (VilCreERT2Apcflox/+ 135 days post induction, Apc+/−). The abundance was assessed relative to control tissue (tamoxifen injected Apcflox/flox mice). Data were normalized to the abundance of 18s rRNA (mean ± SD, n: Controls = 3 extracts from 3 control mice, n: Dysplasias = 3 extracts from 3 Apc−/− mice, n: Adenomas = 3 adenomas from 3 Apc+/− mice, significant differences, 1P = 0.0074, 2P = 0.0108, 3P = 0.0009, 4P = 0.0023, 5P = 0.0028, 6P = 0.0039, 7P = 0.0089, 8P = 0.0436, two-tailed unpaired t-test). (c) The expression of Atg7, Atg5, Beclin1 and Lamp2 mRNA were analysed by qPCR in human untreated intestinal tumors and in normal matched tissue. Twenty-six colonic adenomas, 31 non-metastatic colon carcinomas, 39 metastatic colon carcinomas, and 24 liver metastasis were analysed. The percentage indicate the tumors that showed a higher level of Atg gene expression than normal matched-tissue (>1.5 fold) for at least 3 out of 4 tested genes. Gene expression levels were normalized to the abundance of 18s rRNA for each sample.

Supplementary Figure 6 Immune cell infiltration following IEC-Atg7 deficiency.

(a) Transcriptomic analysis of normal duodenum of Apc+/−Atg7−/− mice compared to Apc+/− mice revealed that among 129 genes that were significantly deregulated following Atg7 deletion, 32 are related to type I IFN signaling response (one way ANOVA, P < 0.05). (b) qPCR analyses confirmed that the IFN-mediated response was deregulated in the duodenum and colon from Apc+/− and Apc+/−Atg7−/− mice. (mean ± SD, n: Apc+/− = 9 extracts from 9 mice, Apc+/−Atg7−/− = 10 extracts from 10 mice; mice were pooled from 2 independent experiments, significant differences, 1P = 1.97 × 10−05, 2P = 0.0002, 3P = 0.0007, 4P = 0.0005, 5P = 0.0005, 6P = 0.0376, 7P = 0.0003, 8P = 0.0005, 9P = 0.0008, 10P = 0.0091, two-tailed unpaired t-test). (c) Representative immunostainings for CD45 and CD3 showing immune cell infiltration in non tumoral colonic mucosa of Apc+/−Atg7−/− and Apc+/− mice. (d) Representative CD11c immunostaining showing an increase in CD11c+ cells in the lamina propria of the small intestine of Apc+/−Atg7−/− compared to Apc+/− mice. (e) Percentages of DC subsets gated on CD11chigh MHCIIhigh by flow cytometry analyses in the MLN from Apc+/−Atg7−/− and Apc+/− mice (mean ± SD, n: Apc+/− = 3 and Apc+/−Atg7−/− = 4 mice, significant differences, 1P = 0.0155, 2P = 0.0483, 3P = 0.0422, two-tailed unpaired t-test). Representative dot plots of DC subsets based on CD103, CD11b expression in MLN from Apc+/−Atg7−/− and Apc+/− mice. (f) Secretion of IL-12p70 produced following PMA/Ionomycin stimulation of immune cells extracted from the small intestine lamina propria of Apc+/−Atg7−/− and Apc+/ mice (mean ± SD, n: Apc+/− = 3 and Apc+/−Atg7−/− = 3 mice, significant differences, 1P = 0.0335, two-tailed unpaired t-test).

Supplementary Figure 7 Immune gene expression following IEC-Atg7 deletion.

(a,b) The expression of genes associated with cytotoxic CD8+ T cells (CD2, CD3γ, CD8α, CD8β, IL15, ZAP70, GZMA, GZMβ, GZMκ, PRF1), TH1 cells (Stat1, IRF1, IFNγ, TBX21, IL12Rb1), Treg cells (FOXP3, CTLA4, TGFβ1), and TH17 cells (Il17Rβ, IL23α, RORC, IRF4, CCL20, CCR6, STAT3) assessed by qPCR in normal colon (a) and duodenum (b) from Apc+/−Atg7−/− compared to Apc+/− mice. (c) Transcript levels of of CX3CL1, CXCL9, CXCL10 mRNA was analysed by qPCR in the colon and duodenum from Apc+/− mice or Apc+/−Atg7−/− mice. Gene expression levels were normalized to the abundance of 18s rRNA for each sample (mean ± SD, (ac) n: Apc+/− = 9 extracts from 9 mice and Apc+/−Atg7−/− = 10 extracts from 10 mice, mice were pooled from three independent experiments, significant differences, (a) 1P = 0.0108, 2P = 0.0257, 3P = 0.0185, 4P = 0.0150, 5P = 0.0180, 6P = 0.0123, 7P = 0.0109, 8 = 0.0200, 9P = 0.0006, 10P = 5.25 × 10−08, 11P = 2.51 × 10−07, 12P = 0.0006, 13P = 0.0134, 14P = 0.0009, 15P = 0.0154, 16P = 0.0195; (b) 1P = 0.0366, 2P = 0.0472, 3P = 0.0413, 4P = 0.0313, 5P = 0.0006, 6P = 0.0156, 7P = 0.0055, 8P = 0.0473, 9P = 0.0187, 10P = 0.0294, 11P = 0.0142, 12P = 0.0356, 13P = 0.0002, 14P = 0.0070, 15P = 0.0038, 16P = 0.0006, 17P = 0.0305, 18P = 0.0109 (c)1P = 0.0005, 2P = 0.0278, 3P = 0.0136, 4P = 0.0281, 5P = 0.0338, 6P = 0.0021, two-tailed unpaired t-test).

Supplementary Figure 8 Anti-CD8 and anti-CD25 antibody treatments on Apc+/−Atg7−/− and Apc+/− mice.

(a) Protocol of CD8 and CD25 depletion. Apc+/−Atg7−/− and Apc+/− mice were injected with tamoxifen and treated with anti-CD8, anti-CD25 antibodies or IgG isotype and their diet was supplemented with tamoxifen for 5 days per month. Mice were injected with antibodies or with control IgG twice a week for 90 days. (b) Successful CD8+ T and Treg-cell depletion in Apc+/−Atg7−/− and Apc+/− mice was monitored by flow cytometry of splenocytes. Percentage of CD8+ IFNγ T cells or FOXP3 CD4+T cells within the CD45+ cell population from mice of the indicated genotype and treatment. (mean ± SD, for CD8+ T cells: n: Apc+/− Isotype IgG = 3, Apc+/− dCD8 = 3,Apc+/−Atg7−/− Isotype IgG = 3,Apc+/−Atg7−/− dCD8 = 4 mice; for CD4+ T cells: n: Apc+/− Isotype IgG = 3,Apc+/−dCD8 = 3,Apc+/−Atg7−/− Isotype IgG = 5,Apc+/−Atg7−/− dCD8 = 5 mice, significant differences, 1P = 0.0144, 2P = 0.0066, 3P = 0.0261, 4P = 0.0076, two-tailed unpaired t-test).

Supplementary Figure 9 IEC-autophagy deficiency induces microbial dysbiosis.

(a) Transcriptomic analysis of normal duodenum of Apc+/−Atg7−/− mice compared to Apc+/− mice revealed that among the 54 genes that were significantly downregulated following Atg7 deletion, 20 genes encode Paneth cell markers (one way ANOVA, P < 0.05). qPCR analyses confirmed that Paneth cell markers are less abundant in the distal small intestine of Apc+/−Atg7−/−. Gene expression levels were normalized to the abundance of 18s rRNA for each sample (mean ± SD, n: Apc+/− = 9,Apc+/−Atg7−/− = 10 mice, significant differences, 1P = 4.15 × 10−05, 2P = 7.61 × 10−04, 3P = 1.39 × 10−05, two-tailed unpaired t-test). (b) Heatmap of differentially represented bacterial species in feces between Apc+/− and Apc+/−Atg7−/− mice. Log10-transformation was applied on the relative abundance data matrix. Phyla assignment of the different bacterial species is indicated by a cap letter at the beginning of the species name. F: Firmicutes, B: Bacteroidetes, P: Proteobacteria, A: Actinobacteria. (n = 8 extracts of feces from 8 mice per condition, mice were pooled from 2 independent experiments, P values are listed in Supplementary Table 2 (two-tailed unpaired t-test). (c) Diversity of the gut microbiota in Apc+/− and Apc+/−Atg7−/− mice. Simpson diversity index was calculated to estimate bacterial diversity in both fecal and ileal mucosa samples of Apc+/− and Apc+/−Atg7−/− mice (mean ± SD, for feces, n: Apc+/− = 8 extracts from 8 mice, n: Apc+/−Atg7−/− = 8 extracts from 8 mice, for ileum n: Apc+/− = 7, n: Apc+/−Atg7−/− = 7, mice were pooled from 2 independent experiments, P = 0.015, two-tailed unpaired t-test). (d) Main bacterial genera repartition in both ileal and feces mucosa of Apc+/− and Apc+/−Atg7−/− mice (n: Apc+/− = 8 extracts from 8 mice, n: Apc+/−Atg7−/− = 8 extracts from 8 mice, for ileum n: Apc+/− = 7, n: Apc+/−Atg7−/− = 7, mice were pooled from 2 independent experiments, P values are listed in Supplementary Table 3 (two-tailed unpaired t-test). (e) Aerobic culture of fecal and colonic microbiota (mean ± SD, n: (Gram)Apc+/− = 3,Apc+/−Atg7−/− = 4 mice; (Gram+)Apc+/− = 4,Apc+/−Atg7−/− = 4 mice, significant differences, 1P = 0.0428, 2P = 0.0029, 3P = 0.0276, 4P = 0.0008, two-tailed unpaired t-test). (f) Protocol of short and long term antibiotic treatments (ATB).

Supplementary Figure 10 β-catenin signaling in adenoma following IEC-Atg7 deletion.

Representative hematoxylin/eosin staining of colonic adenomas from Apc+/− and Apc+/−Atg7−/− mice. Polyps from Apc+/−Atg7−/− mice were smaller than those from Apc+/− mice. (b,c) Gene expression levels of several β-catenin target genes (Axin2, c-Myc, c-Jun, Sox9) assessed by qPCR of adenomas (Ade) from the indicated genotype relative to non-tumoral colon of Apc+/− mice (NT). Gene expression levels were normalized to the abundance of 18s rRNA for each sample (mean ± SD., n: NT Apc+/− = 8 extracts from 8 mice; Ade Apc+/− = 8 tumors from 8 mice and Ade Apc+/−Atg7−/− = 8 tumors from 8 mice, mice were pooled from two independent, significant differences, (b) colon, 1P = 0.0038, 2P = 0.0042, 3P = 0.0032, 4P = 0.0025, 5P = 0.0047, 6P = 0.0485, 7P = 0.0466, 8P = 0.0472, (c) duodenum 1P = 0.0091, 2P = 0.0192, 3P = 0.0086, 4P = 0.0082, 5P = 0.0037, 6P = 0.0008, 7P = 0.0384, 8P = 0.0478 two-tailed unpaired t-test).

Supplementary Figure 11 Effect of metformin treatment on intestinal adenomas from Apc+/− mice.

(a) Collected ultrasound measurements of colonic tumor volumes from Apc+/− mice and those from metformin-treated-Apc+/− mice. Box plots show the 5-95 percentiles which are delineated by the upper and the lower limits of the box and the median is shown by the horizontal line inside the box. n: Apc+/− = 10,Apc+/−Met = 32 tumors pooled from 3 mice. P(genotype) < 0,0001 and P(Time) < 0,0001 (two-way ANOVA test). (b) Western blotting for phosphorylated-AMPK (pAMPK), AMPK, phosphorylated-Raptor (pRaptor), Raptor, phosphorylated-p70S6 kinase (pp70S6K), p70S6 kinase (p70S6K), phosphorylated-S6 ribosomal protein (pS6) and S6 ribosomal protein (S6) in adenomas from Apc+/− mice and those from metformin-treated-Apc+/− mice.γ-tubulin served as a loading control. Each lane represents a sample from a different animal. Unprocessed original scans of blots are shown in Supplementary Fig. 9.

Supplementary Figure 12 Model of the differential effects of IEC-Atg7 deletion on the initiation and progression of intestinal tumorigenesis driven by Apc-loss.

IEC-autophagy blockade by Atg7 deletion alters Paneth cell numbers and leads to abnormal mucin accumulation in goblet cells. This host defense alteration is accompanied by an increase in intestinal permeability and contributes to a shift in the composition of the gut microbiota characterized by an outgrowth of Firmicutes and a decrease in Proteobacteria. Remodeling of the microbiota architecture and composition is associated with an increased abundance of CD103+CD11b within the mesenteric lymph node reported to prime Th1 and CD8 T cell through IL-1β and IL-12 secretion. Infiltration of cytotoxic CD8+ T cells in the lamina propria has a major impact on the elimination of transformed cells induced by Apc loss. However, this antitumoral response is incomplete as few cancer cells escape and persist in Apc+/−Atg7−/− mice. At this stage, cell cycle arrest associated with p53 accumulation, AMPK signaling activation and low abundance of glycolytic enzymes lead to a drastic decrease in Atg7-deficient tumor cell growth.

Supplementary Figure 13 Unprocessed original scans of Western blots presented in the indicated Figures and Supplementary Figures.

Supplementary Table 1 Transcriptomic analysis of normal duodenum of Apc+/−Atg7−/− mice compared to Apc+/− mice.
Supplementary Table 2 Differentially represented species in feces of Apc+/−Atg7−/ mice compared to Apc+/− mice.
Supplementary Table 3 Main bacterial genera repartition in both ileal and feces mucosa of Apc+/− and Apc+/−Atg7−/− mice revealed by linear discriminant analysis effect size (LEfSe) analysis (P values are indicated, two-tailed unpaired t-test).
Supplementary Table 4 Primers used for qPCR assays in this study.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2829 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lévy, J., Cacheux, W., Bara, M. et al. Intestinal inhibition of Atg7 prevents tumour initiation through a microbiome-influenced immune response and suppresses tumour growth. Nat Cell Biol 17, 1062–1073 (2015). https://doi.org/10.1038/ncb3206

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3206

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