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:

Non-canonical HIF-1 stabilization contributes to intestinal tumorigenesis

Abstract

The hypoxia-inducible transcription factor HIF-1 is appreciated as a promising target for cancer therapy. However, conditional deletion of HIF-1 and HIF-1 target genes in cells of the tumor microenvironment can result in accelerated tumor growth, calling for a detailed characterization of the cellular context to fully comprehend HIF-1’s role in tumorigenesis. We dissected cell type-specific functions of HIF-1 for intestinal tumorigenesis by lineage-restricted deletion of the Hif1a locus. Intestinal epithelial cell-specific Hif1a loss reduced activation of Wnt/β-catenin, tumor-specific metabolism and inflammation, significantly inhibiting tumor growth. Deletion of Hif1a in myeloid cells reduced the expression of fibroblast-activating factors in tumor-associated macrophages resulting in decreased abundance of tumor-associated fibroblasts (TAF) and robustly reduced tumor formation. Interestingly, hypoxia was detectable only sparsely and without spatial association with HIF-1α, arguing for an importance of hypoxia-independent, i.e., non-canonical, HIF-1 stabilization for intestinal tumorigenesis that has not been previously appreciated. This adds a further layer of complexity to the regulation of HIF-1 and suggests that hypoxia and HIF-1α stabilization can be uncoupled in cancer. Collectively, our data show that HIF-1 is a pivotal pro-tumorigenic factor for intestinal tumor formation, controlling key oncogenic programs in both the epithelial tumor compartment and the tumor microenvironment.

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

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A, et al. Colorectal cancer statistics, 2017. CA Cancer J Clin. 2017;67:177–93.

    Article  Google Scholar 

  2. Kohne CH. Successes and limitations of targeted cancer therapy in colon cancer. Prog tumor Res. 2014;41:36–50.

    Article  Google Scholar 

  3. Huang M, Shen A, Ding J, Geng M. Molecularly targeted cancer therapy: some lessons from the past decade. Trends Pharm Sci. 2014;35:41–50.

    Article  Google Scholar 

  4. Semenza GL. Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy. Trends Pharm Sci. 2012;33:207–14.

    Article  CAS  Google Scholar 

  5. Baba Y, Nosho K, Shima K, Irahara N, Chan AT, Meyerhardt JA, et al. HIF1A overexpression is associated with poor prognosis in a cohort of 731 colorectal cancers. AmJPathol. 2010;176:2292–301.

    CAS  Google Scholar 

  6. Yoshimura H, Dhar DK, Kohno H, Kubota H, Fujii T, Ueda S, et al. Prognostic impact of hypoxia-inducible factors 1alpha and 2alpha in colorectal cancer patients: correlation with tumor angiogenesis and cyclooxygenase-2 expression. ClinCancer Res. 2004;10:8554–60.

    CAS  Google Scholar 

  7. Zhong H, De Marzo AM, Laughner E, Lim M, Hilton DA, Zagzag D, et al. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 1999;59:5830–5.

    CAS  PubMed  Google Scholar 

  8. Imamura T, Kikuchi H, Herraiz MT, Park DY, Mizukami Y, Mino-Kenduson M, et al. HIF-1alpha and HIF-2alpha have divergent roles in colon cancer. Int J Cancer. 2009;124:763–71.

    Article  CAS  Google Scholar 

  9. Mizukami Y, Jo WS, Duerr EM, Gala M, Li J, Zhang X, et al. Induction of interleukin-8 preserves the angiogenic response in HIF-1alpha-deficient colon cancer cells. Nat Med. 2005;11:992–7.

    Article  CAS  Google Scholar 

  10. Shay JE, Imtiyaz HZ, Sivanand S, Durham AC, Skuli N, Hsu S, et al. Inhibition of hypoxia-inducible factors limits tumor progression in a mouse model of colorectal cancer. Carcinogenesis. 2014;35:1067–77.

    Article  CAS  Google Scholar 

  11. Xue X, Ramakrishnan SK, Shah YM. Activation of HIF-1alpha does not increase intestinal tumorigenesis. Am J Physiol Gastrointest Liver Physiol. 2014;307:G187–195.

    Article  CAS  Google Scholar 

  12. Xue X, Ramakrishnan S, Anderson E, Taylor M, Zimmermann EM, Spence JR, et al. Endothelial PAS domain protein 1 activates the inflammatory response in the intestinal epithelium to promote colitis in mice. Gastroenterology. 2013;145:831–41.

    Article  CAS  Google Scholar 

  13. Mladenova DN, Dahlstrom JE, Tran PN, Benthani F, Bean EG, Ng I, et al. HIF1alpha deficiency reduces inflammation in a mouse model of proximal colon cancer. Dis Model Mech. 2015;8:1093–103.

    Article  CAS  Google Scholar 

  14. Stockmann C, Doedens A, Weidemann A, Zhang N, Takeda N, Greenberg JI, et al. Deletion of vascular endothelial growth factor in myeloid cells accelerates tumorigenesis. Nature. 2008;456:814–8.

    Article  CAS  Google Scholar 

  15. Kim JW, Evans C, Weidemann A, Takeda N, Lee YS, Stockmann C, et al. Loss of fibroblast HIF-1alpha accelerates tumorigenesis. Cancer Res. 2012;72:3187–95.

    Article  CAS  Google Scholar 

  16. Palazon A, Tyrakis PA, Macias D, Velica P, Rundqvist H, Fitzpatrick S, et al. An HIF-1alpha/VEGF-A axis in cytotoxic T cells regulates tumor progression. Cancer Cell. 2017;32:669. e665

    Article  CAS  Google Scholar 

  17. Cramer T, Yamanishi Y, Clausen BE, Forster I, Pawlinski R, Mackman N, et al. HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell. 2003;112:645–57.

    Article  CAS  Google Scholar 

  18. Ryan HE, Poloni M, McNulty W, Elson D, Gassmann M, Arbeit JM, et al. Hypoxia-inducible factor-1alpha is a positive factor in solid tumor growth. Cancer Res. 2000;60:4010–5.

    CAS  PubMed  Google Scholar 

  19. Okayasu I, Ohkusa T, Kajiura K, Kanno J, Sakamoto S. Promotion of colorectal neoplasia in experimental murine ulcerative colitis. Gut. 1996;39:87–92.

    Article  CAS  Google Scholar 

  20. Su LK, Kinzler KW, Vogelstein B, Preisinger AC, Moser AR, Luongo C, et al. Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. Science. 1992;256:668–70.

    Article  CAS  Google Scholar 

  21. LaGory EL, Giaccia AJ. The ever-expanding role of HIF in tumour and stromal biology. Nat Cell Biol. 2016;18:356–65.

    Article  CAS  Google Scholar 

  22. Madison BB, Dunbar L, Qiao XT, Braunstein K, Braunstein E, Gumucio DL. Cis elements of the villin gene control expression in restricted domains of the vertical (crypt) and horizontal (duodenum, cecum) axes of the intestine. J Biol Chem. 2002;277:33275–83.

    Article  CAS  Google Scholar 

  23. Mazumdar J, O’Brien WT, Johnson RS, LaManna JC, Chavez JC, Klein PS, et al. O2 regulates stem cells through Wnt/beta-catenin signalling. Nat. Cell Biol. 2010;12:1007–13.

    Article  CAS  Google Scholar 

  24. Semenza GL. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J Clin Invest. 2013;123:3664–71.

    Article  CAS  Google Scholar 

  25. Janakiram NB, Rao CV. The role of inflammation in colon cancer. Adv Exp Med Biol. 2014;816:25–52.

    Article  CAS  Google Scholar 

  26. Eichele DD, Kharbanda KK. Dextran sodium sulfate colitis murine model: an indispensable tool for advancing our understanding of inflammatory bowel diseases pathogenesis. World J Gastroenterol. 2017;23:6016–29.

    Article  CAS  Google Scholar 

  27. Louis NA, Hamilton KE, Canny G, Shekels LL, Ho SB, Colgan SP. Selective induction of mucin-3 by hypoxia in intestinal epithelia. J Cell Biochem. 2006;99:1616–27.

    Article  CAS  Google Scholar 

  28. Dilly AK, Lee YJ, Zeh HJ, Guo ZS, Bartlett DL, Choudry HA. Targeting hypoxia-mediated mucin 2 production as a therapeutic strategy for mucinous tumors. Transl Res. 2016;169:19–30.e1.

    Article  CAS  Google Scholar 

  29. Mikami Y, Hisatsune A, Tashiro T, Isohama Y, Katsuki H. Hypoxia enhances MUC1 expression in a lung adenocarcinoma cell line. Biochem Biophys Res Commun. 2009;379:1060–5.

    Article  CAS  Google Scholar 

  30. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118:229–41.

    Article  CAS  Google Scholar 

  31. Biswas SK, Allavena P, Mantovani A. Tumor-associated macrophages: functional diversity, clinical significance, and open questions. Semin Immunopathol. 2013;35:585–600.

    Article  CAS  Google Scholar 

  32. Palazon A, Goldrath AW, Nizet V, Johnson RS. HIF transcription factors, inflammation, and immunity. Immunity. 2014;41:518–28.

    Article  CAS  Google Scholar 

  33. Trottier MD, Irwin R, Li Y, McCabe LR, Fraker PJ. Enhanced production of early lineages of monocytic and granulocytic cells in mice with colitis. Proc Natl Acad Sci USA. 2012;109:16594–9.

    Article  CAS  Google Scholar 

  34. Mantovani A, Sica A. Macrophages, innate immunity and cancer: balance, tolerance, and diversity. Curr Opin Immunol. 2010;22:231–7.

    Article  CAS  Google Scholar 

  35. Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013;73:1128–41.

    Article  CAS  Google Scholar 

  36. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, et al. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007;445:111–5.

    Article  CAS  Google Scholar 

  37. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449:1003–7.

    Article  CAS  Google Scholar 

  38. Ragusa S, Cheng J, Ivanov KI, Zangger N, Ceteci F, Bernier-Latmani J, et al. PROX1 promotes metabolic adaptation and fuels outgrowth of Wnt(high) metastatic colon cancer cells. Cell Rep. 2014;8:1957–73.

    Article  CAS  Google Scholar 

  39. Heijmans J, Buller NV, Muncan V, van den Brink GR. Role of mast cells in colorectal cancer development, the jury is still out. Biochim Biophys Acta. 2012;1822:9–13.

    Article  CAS  Google Scholar 

  40. Isella C, Terrasi A, Bellomo SE, Petti C, Galatola G, Muratore A. et al. Stromal contribution to the colorectal cancer transcriptome. Nat Genet. 2015;47:312–9.

    Article  CAS  Google Scholar 

  41. Calon A, Lonardo E, Berenguer-Llergo A, Espinet E, Hernando-Momblona X, Iglesias M. et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat Genet. 2015;47:320–9.

    Article  CAS  Google Scholar 

  42. Wynn TA, Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity. 2016;44:450–62.

    Article  CAS  Google Scholar 

  43. Ross R, Benditt EP. Wound healing and collagen formation. I. Sequential changes in components of guinea pig skin wounds observed in the electron microscope. J Biophys Biochem Cytol. 1961;11:677–700.

    Article  CAS  Google Scholar 

  44. Travis MA, Sheppard D. TGF-beta activation and function in immunity. Annu Rev Immunol. 2014;32:51–82.

    Article  CAS  Google Scholar 

  45. Pradere JP, Kluwe J, De Minicis S, Jiao JJ, Gwak GY, Dapito DH, et al. Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology. 2013;58:1461–73.

    Article  CAS  Google Scholar 

  46. Gascard P, Tlsty TD. Carcinoma-associated fibroblasts: orchestrating the composition of malignancy. Genes Dev. 2016;30:1002–19.

    Article  CAS  Google Scholar 

  47. Waldner MJ, Foersch S, Neurath MF. Interleukin-6–a key regulator of colorectal cancer development. Int J Biol Sci. 2012;8:1248–53.

    Article  CAS  Google Scholar 

  48. Neufert C, Becker C, Tureci O, Waldner MJ, Backert I, Floh K, et al. Tumor fibroblast-derived epiregulin promotes growth of colitis-associated neoplasms through ERK. J Clin Invest. 2013;123:1428–43.

    Article  CAS  Google Scholar 

  49. Kramann R, Schneider RK, DiRocco DP, Machado F, Fleig S, Bondzie PA, et al. Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem Cell. 2015;16:51–66.

    Article  CAS  Google Scholar 

  50. Bucala R, Spiegel LA, Chesney J, Hogan M, Cerami A. Circulating fibrocytes define a new leukocyte subpopulation that mediates tissue repair. Mol Med. 1994;1:71–81.

    Article  CAS  Google Scholar 

  51. Raffaghello L, Dazzi F. Classification and biology of tumour associated stromal cells. Immunol Lett. 2015;168:175–82.

    Article  CAS  Google Scholar 

  52. Crawford JR, Pilling D, Gomer RH. Improved serum-free culture conditions for spleen-derived murine fibrocytes. J Immunol Methods. 2010;363:9–20.

    Article  CAS  Google Scholar 

  53. Karhausen J, Furuta GT, Tomaszewski JE, Johnson RS, Colgan SP, Haase VH. Epithelial hypoxia-inducible factor-1 is protective in murine experimental colitis. J Clin Invest. 2004;114:1098–106.

    Article  CAS  Google Scholar 

  54. Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med. 2004;10:789–99.

    Article  CAS  Google Scholar 

  55. Sansom OJ, Reed KR, Hayes AJ, Ireland H, Brinkmann H, Newton IP, et al. Loss of Apc in vivo immediately perturbs Wnt signaling, differentiation, and migration. Genes Dev. 2004;18:1385–90.

    Article  CAS  Google Scholar 

  56. Riemer P, Rydenfelt M, Marks M, van Eunen K, Thedieck K, Herrmann BG, et al. Oncogenic β-catenin and PIK3CA instruct network states and cancer phenotypes in intestinal organoids. J Cell Biol. 2017;216:1567–77.

    Article  CAS  Google Scholar 

  57. Farrall AL, Riemer P, Leushacke M, Sreekumar A, Grimm C, Herrmann BG, et al. Wnt and BMP signals control intestinal adenoma cell fates. Int J Cancer. 2012;131:2242–52.

    Article  CAS  Google Scholar 

  58. Shah YM, Ito S, Morimura K, Chen C, Yim SH, Haase VH, et al. Hypoxia-inducible factor augments experimental colitis through an MIF-dependent inflammatory signaling cascade. Gastroenterology. 2008;134:2036–48. 2048.e2031-2033

    Article  Google Scholar 

  59. Mahler M, Bristol IJ, Leiter EH, Workman AE, Birkenmeier EH, Elson CO, et al. Differential susceptibility of inbred mouse strains to dextran sulfate sodium-induced colitis. Am J Physiol. 1998;274:G544–551.

    CAS  PubMed  Google Scholar 

  60. Cummins EP, Seeballuck F, Keely SJ, Mangan NE, Callanan JJ, Fallon PG, et al. The hydroxylase inhibitor dimethyloxalylglycine is protective in a murine model of colitis. Gastroenterology. 2008;134:156–65.

    Article  CAS  Google Scholar 

  61. Robinson A, Keely S, Karhausen J, Gerich ME, Furuta GT, Colgan SP. Mucosal protection by hypoxia-inducible factor prolyl hydroxylase inhibition. Gastroenterology. 2008;134:145–55.

    Article  CAS  Google Scholar 

  62. Kim YE, Lee M, Gu H, Kim J, Jeong S, Yeo S et al. HIF-1alpha activation in myeloid cells accelerates dextran sodium sulfate-induced colitis progression in mice. Dis Model Mech 2018;11. https://doi.org/10.1242/dmm.033241.

    Article  Google Scholar 

  63. Bäcker V, Cheung FY, Siveke JT, Fandrey J, Winning S. Knockdown of myeloid cell hypoxia-inducible factor-1α ameliorates the acute pathology in DSS-induced colitis. PLoS ONE. 2017;12:e0190074

    Article  Google Scholar 

  64. Fluck K, Breves G, Fandrey J, Winning S. Hypoxia-inducible factor 1 in dendritic cells is crucial for the activation of protective regulatory T cells in murine colitis. Mucosal Immunol. 2016;9:379–90.

    Article  CAS  Google Scholar 

  65. Hoebler C, Gaudier E, De Coppet P, Rival M, Cherbut C. MUC genes are differently expressed during onset and maintenance of inflammation in dextran sodium sulfate-treated mice. Dig Dis Sci. 2006;51:381–9.

    Article  CAS  Google Scholar 

  66. Peterson LW, Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol. 2014;14:141–53.

    Article  CAS  Google Scholar 

  67. Doedens AL, Stockmann C, Rubinstein MP, Liao D, Zhang N, DeNardo DG, et al. Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor progression. Cancer Res. 2010;70:7465–75.

    Article  CAS  Google Scholar 

  68. Tymoszuk P, Evens H, Marzola V, Wachowicz K, Wasmer MH, Datta S, et al. In situ proliferation contributes to accumulation of tumor-associated macrophages in spontaneous mammary tumors. Eur J Immunol. 2014;44:2247–62.

    Article  CAS  Google Scholar 

  69. Wynn TA, Barron L. Macrophages: master regulators of inflammation and fibrosis. Semin Liver Dis. 2010;30:245–57.

    Article  CAS  Google Scholar 

  70. Xue J, Sharma V, Hsieh MH, Chawla A, Murali R, Pandol SJ, et al. Alternatively activated macrophages promote pancreatic fibrosis in chronic pancreatitis. Nat Commun. 2015;6:7158.

    Article  CAS  Google Scholar 

  71. Krishnamachary B, Berg-Dixon S, Kelly B, Agani F, Feldser D, Ferreira G, et al. Regulation of colon carcinoma cell invasion by hypoxia-inducible factor 1. Cancer Res. 2003;63:1138–43.

    CAS  PubMed  Google Scholar 

  72. Kaidi A, Qualtrough D, Williams AC, Paraskeva C. Direct transcriptional up-regulation of cyclooxygenase-2 by hypoxia-inducible factor (HIF)-1 promotes colorectal tumor cell survival and enhances HIF-1 transcriptional activity during hypoxia. Cancer Res. 2006;66:6683–91.

    Article  CAS  Google Scholar 

  73. Hung SP, Yang MH, Tseng KF, Lee OK. Hypoxia-induced secretion of TGF-beta1 in mesenchymal stem cell promotes breast cancer cell progression. Cell Transpl. 2013;22:1869–82.

    Article  Google Scholar 

  74. Hu J, Discher DJ, Bishopric NH, Webster KA. Hypoxia regulates expression of the endothelin-1 gene through a proximal hypoxia-inducible factor-1 binding site on the antisense strand. Biochem Biophys Res Commun. 1998;245:894–9.

    Article  CAS  Google Scholar 

  75. Ozdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25:719–34.

    Article  CAS  Google Scholar 

  76. Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–47.

    Article  CAS  Google Scholar 

  77. Cooper HS, Murthy SN, Shah RS, Sedergran DJ. Clinicopathologic study of dextran sulfate sodium experimental murine colitis. Lab Investig. 1993;69:238–49.

    CAS  PubMed  Google Scholar 

  78. Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141:1762–72.

    Article  CAS  Google Scholar 

  79. Hammerich L, Warzecha KT, Stefkova M, Bartneck M, Ohl K, Gassler N, et al. Cyclic adenosine monophosphate-responsive element modulator alpha overexpression impairs function of hepatic myeloid-derived suppressor cells and aggravates immune-mediated hepatitis in mice. Hepatology. 2015;61:990–1002.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Research in the Cramer lab was supported by grants from Deutsche Krebshilfe (109160) and Deutsche Forschungsgemeinschaft (CR 133/2-1 until 2–4). NR was supported by a grant from the BMBF (MAPTor-NET (031A426A)). We are indebted to Ralf Weiskirchen (University Hospital Aachen) for help regarding the MLEC assay. We are grateful to Christine Sers (Charité, Berlin) and Florian R. Greten (Georg-Speyer-Haus, Frankfurt) for helpful discussions and to Ilia N. Buhtoiarov (Children’s Hospital of New Jersey, USA), Glenn S. Belinsky, Daniel W. Rosenberg (University of Connecticut Health Center, Farmington, USA), and Takuji Tanaka (Gifu Municipal Hospital, Japan) for providing control reagents. We are grateful to Deborah Gumucio (University of Michigan, USA) for providing Villin-Cre mice. The excellent technical assistance of Birgit Bogdanoff and Simone Spiekermann is highly appreciated. Parts of this work were granted the “Best Poster Award” at the 2015 CELL symposium “Cancer, Inflammation and Immunity” in Sitges, Spain.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, NR, MM, and TC; methodology, NR, ME, SJ, AE, KTW, AAK, AF, SN, MG, IR, T.E., CZ, SK, RH, MBM, WF, and MM; formal analysis, investigation, and visualization, NR, ME, SJ, AE, AAK, RK, SN, IR, MG, CZ, SK, MBM, MM, and TC; writing, NR, FT, MM, and TC; supervision, resources, and funding acquisition, RK, SK, CEL, WB, MS, LS, OS, FT, MM, and TC; project administration, TC.

Corresponding author

Correspondence to Thorsten Cramer.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rohwer, N., Jumpertz, S., Erdem, M. et al. Non-canonical HIF-1 stabilization contributes to intestinal tumorigenesis. Oncogene 38, 5670–5685 (2019). https://doi.org/10.1038/s41388-019-0816-4

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-019-0816-4

This article is cited by

Search

Quick links