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:

An ID2-dependent mechanism for VHL inactivation in cancer

Subjects

Abstract

Mechanisms that maintain cancer stem cells are crucial to tumour progression. The ID2 protein supports cancer hallmarks including the cancer stem cell state. HIFα transcription factors, most notably HIF2α (also known as EPAS1), are expressed in and required for maintenance of cancer stem cells (CSCs). However, the pathways that are engaged by ID2 or drive HIF2α accumulation in CSCs have remained unclear. Here we report that DYRK1A and DYRK1B kinases phosphorylate ID2 on threonine 27 (Thr27). Hypoxia downregulates this phosphorylation via inactivation of DYRK1A and DYRK1B. The activity of these kinases is stimulated in normoxia by the oxygen-sensing prolyl hydroxylase PHD1 (also known as EGLN2). ID2 binds to the VHL ubiquitin ligase complex, displaces VHL-associated Cullin 2, and impairs HIF2α ubiquitylation and degradation. Phosphorylation of Thr27 of ID2 by DYRK1 blocks ID2–VHL interaction and preserves HIF2α ubiquitylation. In glioblastoma, ID2 positively modulates HIF2α activity. Conversely, elevated expression of DYRK1 phosphorylates Thr27 of ID2, leading to HIF2α destabilization, loss of glioma stemness, inhibition of tumour growth, and a more favourable outcome for patients with glioblastoma.

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

Figure 1: Inhibition of DYRK1-mediated phosphorylation of ID2 at Thr27 promotes NSC properties.
Figure 2: DYRK1 kinases and ID2 Thr27 phosphorylation are inhibited by hypoxia and enhanced by PHD1.
Figure 3: The DYRK1–ID2–Thr27 pathway controls GSCs and HIF2α.
Figure 4: Unphosphorylated ID2 binds to and disrupts the VCB–Cul2 complex.
Figure 5: DYRK1 kinases inhibit human glioma growth by repressing an ID2–HIF2α network.

Similar content being viewed by others

References

  1. Keith, B., Johnson, R. S. & Simon, M. C. HIF1α and HIF2 α: sibling rivalry in hypoxic tumour growth and progression. Nature Rev. Cancer 12, 9–22 (2012)

    CAS  Google Scholar 

  2. Gordan, J. D., Bertout, J. A., Hu, C. J., Diehl, J. A. & Simon, M. C. HIF-2α promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity. Cancer Cell 11, 335–347 (2007)

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Kondo, K., Kim, W. Y., Lechpammer, M. & Kaelin, W. G., Jr. Inhibition of HIF2α is sufficient to suppress pVHL-defective tumor growth. PLoS Biol. 1, E83 (2003)

    PubMed  PubMed Central  Google Scholar 

  4. Li, Z. et al. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell 15, 501–513 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Pietras, A. et al. HIF-2α maintains an undifferentiated state in neural crest-like human neuroblastoma tumor-initiating cells. Proc. Natl Acad. Sci. USA 106, 16805–16810 (2009)

    ADS  CAS  PubMed  Google Scholar 

  6. Lasorella, A., Benezra, R. & Iavarone, A. The ID proteins: master regulators of cancer stem cells and tumour aggressiveness. Nature Rev. Cancer 14, 77–91 (2014)

    CAS  Google Scholar 

  7. Perk, J., Iavarone, A. & Benezra, R. Id family of helix-loop-helix proteins in cancer. Nature Rev. Cancer 5, 603–614 (2005)

    CAS  Google Scholar 

  8. Lasorella, A., Rothschild, G., Yokota, Y., Russell, R. G. & Iavarone, A. Id2 mediates tumor initiation, proliferation, and angiogenesis in Rb mutant mice. Mol. Cell. Biol. 25, 3563–3574 (2005)

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Niola, F. et al. Mesenchymal high-grade glioma is maintained by the ID–RAP1 axis. J. Clin. Invest. 123, 405–417 (2013)

    CAS  PubMed  Google Scholar 

  10. Vandeputte, D. A. et al. Expression and distribution of id helix-loop-helix proteins in human astrocytic tumors. Glia 38, 329–338 (2002)

    PubMed  Google Scholar 

  11. Hara, E., Hall, M. & Peters, G. Cdk2-dependent phosphorylation of Id2 modulates activity of E2A-related transcription factors. EMBO J. 16, 332–342 (1997)

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Nam, H. S. & Benezra, R. High levels of Id1 expression define B1 type adult neural stem cells. Cell Stem Cell 5, 515–526 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Niola, F. et al. Id proteins synchronize stemness and anchorage to the niche of neural stem cells. Nature Cell Biol. 14, 477–487 (2012)

    CAS  PubMed  Google Scholar 

  14. Himpel, S. et al. Specificity determinants of substrate recognition by the protein kinase DYRK1A. J. Biol. Chem. 275, 2431–2438 (2000)

    CAS  PubMed  Google Scholar 

  15. Lee, K., Deng, X. & Friedman, E. Mirk protein kinase is a mitogen-activated protein kinase substrate that mediates survival of colon cancer cells. Cancer Res. 60, 3631–3637 (2000)

    CAS  PubMed  Google Scholar 

  16. Göckler, N. et al. Harmine specifically inhibits protein kinase DYRK1A and interferes with neurite formation. FEBS J. 276, 6324–6337 (2009)

    PubMed  Google Scholar 

  17. Himpel, S. et al. Identification of the autophosphorylation sites and characterization of their effects in the protein kinase DYRK1A. Biochem. J. 359, 497–505 (2001)

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Kaelin, W. G. Jr & Ratcliffe, P. J. Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway. Mol. Cell 30, 393–402 (2008)

    CAS  PubMed  Google Scholar 

  19. Semenza, G. L. HIF-1, O(2), and the 3 PHDs: how animal cells signal hypoxia to the nucleus. Cell 107, 1–3 (2001)

    CAS  PubMed  Google Scholar 

  20. Warnecke, C. et al. Differentiating the functional role of hypoxia-inducible factor (HIF)-1α and HIF-2α (EPAS-1) by the use of RNA interference: erythropoietin is a HIF-2α target gene in Hep3B and Kelly cells. FASEB J. 18, 1462–1464 (2004)

    CAS  PubMed  Google Scholar 

  21. Kershaw, N. J. & Babon, J. J. VHL: cullin-g the hypoxic response. Structure 23, 435–436 (2015)

    CAS  PubMed  Google Scholar 

  22. Nguyen, H. C., Yang, H., Fribourgh, J. L., Wolfe, L. S. & Xiong, Y. Insights into Cullin-RING E3 ubiquitin ligase recruitment: structure of the VHL–EloBC–Cul2 complex. Structure 23, 441–449 (2015)

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Kamura, T. et al. Rbx1, a component of the VHL tumor suppressor complex and SCF ubiquitin ligase. Science 284, 657–661 (1999)

    ADS  CAS  PubMed  Google Scholar 

  24. Ohta, T., Michel, J. J., Schottelius, A. J. & Xiong, Y. ROC1, a homolog of APC11, represents a family of cullin partners with an associated ubiquitin ligase activity. Mol. Cell 3, 535–541 (1999)

    CAS  PubMed  Google Scholar 

  25. Giorgi, F. M. et al. Inferring protein modulation from gene expression data using conditional mutual information. PLoS ONE 9, e109569 (2014)

    ADS  PubMed  PubMed Central  Google Scholar 

  26. Alvarez, M. J., Giorgi, F. M. & Califano, A. Using viper, a package for virtual inference of protein-activity by enriched regulon analysis. Bioconductor, 1–14 (2014)

  27. Löfstedt, T. et al. Induction of ID2 expression by hypoxia-inducible factor-1: a role in dedifferentiation of hypoxic neuroblastoma cells. J. Biol. Chem. 279, 39223–39231 (2004)

    PubMed  Google Scholar 

  28. Chakrabarti, L., Galdzicki, Z. & Haydar, T. F. Defects in embryonic neurogenesis and initial synapse formation in the forebrain of the Ts65Dn mouse model of Down syndrome. J. Neurosci. 27, 11483–11495 (2007)

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Contestabile, A. et al. Cell cycle alteration and decreased cell proliferation in the hippocampal dentate gyrus and in the neocortical germinal matrix of fetuses with Down syndrome and in Ts65Dn mice. Hippocampus 17, 665–678 (2007)

    PubMed  Google Scholar 

  30. Guimeá, J. et al. A human homologue of Drosophila minibrain (MNB) is expressed in the neuronal regions affected in Down syndrome and maps to the critical region. Hum. Mol. Genet. 5, 1305–1310 (1996)

    Google Scholar 

  31. Hibaoui, Y. et al. Modelling and rescuing neurodevelopmental defect of Down syndrome using induced pluripotent stem cells from monozygotic twins discordant for trisomy 21. EMBO Mol. Med. 6, 259–277 (2014)

    CAS  PubMed  Google Scholar 

  32. Aranda, S., Laguna, A. & de la Luna, S. DYRK family of protein kinases: evolutionary relationships, biochemical properties, and functional roles. FASEB J. 25, 449–462 (2011)

    CAS  PubMed  Google Scholar 

  33. Becker, W. Emerging role of DYRK family protein kinases as regulators of protein stability in cell cycle control. Cell Cycle 11, 3389–3394 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Hämmerle, B. et al. Transient expression of Mnb/Dyrk1a couples cell cycle exit and differentiation of neuronal precursors by inducing p27KIP1 expression and suppressing NOTCH signaling. Development 138, 2543–2554 (2011)

    PubMed  PubMed Central  Google Scholar 

  35. Litovchick, L., Florens, L. A., Swanson, S. K., Washburn, M. P. & DeCaprio, J. A. DYRK1A protein kinase promotes quiescence and senescence through DREAM complex assembly. Genes Dev. 25, 801–813 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Park, J. et al. Dyrk1A phosphorylates p53 and inhibits proliferation of embryonic neuronal cells. J. Biol. Chem. 285, 31895–31906 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Yabut, O., Domogauer, J. & D'Arcangelo, G. Dyrk1A overexpression inhibits proliferation and induces premature neuronal differentiation of neural progenitor cells. J. Neurosci. 30, 4004–4014 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Lauth, M. et al. DYRK1B-dependent autocrine-to-paracrine shift of Hedgehog signaling by mutant RAS. Nature Struct. Mol. Biol. 17, 718–725 (2010)

    CAS  Google Scholar 

  39. Mao, J. et al. Regulation of Gli1 transcriptional activity in the nucleus by Dyrk1. J. Biol. Chem. 277, 35156–35161 (2002)

    CAS  PubMed  Google Scholar 

  40. Yang, E. J., Ahn, Y. S. & Chung, K. C. Protein kinase Dyrk1 activates cAMP response element-binding protein during neuronal differentiation in hippocampal progenitor cells. J. Biol. Chem. 276, 39819–39824 (2001)

    CAS  PubMed  Google Scholar 

  41. Pozo, N. et al. Inhibition of DYRK1A destabilizes EGFR and reduces EGFR-dependent glioblastoma growth. J. Clin. Invest. 123, 2475–2487 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Carro, M. S. et al. The transcriptional network for mesenchymal transformation of brain tumours. Nature 463, 318–325 (2010)

    ADS  CAS  PubMed  Google Scholar 

  43. Reynolds, B. A. & Weiss, S. Clonal and population analyses demonstrate that an EGF-responsive mammalian embryonic CNS precursor is a stem cell. Dev. Biol. 175, 1–13 (1996)

    CAS  PubMed  Google Scholar 

  44. Deleyrolle, L. P. et al. Determination of somatic and cancer stem cell self-renewing symmetric division rate using sphere assays. PLoS One. 6, e15844 (2011)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  45. Tropepe, V. et al. Distinct neural stem cells proliferate in response to EGF and FGF in the developing mouse telencephalon. Dev. Biol. 208, 166–188 (1999)

    CAS  PubMed  Google Scholar 

  46. Mîinea, C. P. & Lienhard, G. E. Stoichiometry of site-specific protein phosphorylation estimated with phosphopeptide-specific antibodies. Biotechniques 34, 828–831 (2003)

    PubMed  Google Scholar 

  47. Bordner, A. J. & Abagyan, R. Ab initio prediction of peptide-MHC binding geometry for diverse class I MHC allotypes. Proteins 63, 512–526 (2006)

    CAS  PubMed  Google Scholar 

  48. Abagyan, R. & Totrov, M. Biased probability Monte Carlo conformational searches and electrostatic calculations for peptides and proteins. J. Mol. Biol. 235, 983–1002 (1994)

    CAS  PubMed  Google Scholar 

  49. Almond, D. & Cardozo, T. Assessment of immunologically relevant dynamic tertiary structural features of the HIV-1 V3 loop crown R2 sequence by ab initio folding. J. Vis. Exp. 43, 2118 (2010)

    Google Scholar 

  50. Zhao, X. et al. The HECT-domain ubiquitin ligase Huwe1 controls neural differentiation and proliferation by destabilizing the N-Myc oncoprotein. Nature Cell Biol. 10, 643–653 (2008)

    CAS  PubMed  Google Scholar 

  51. Cawthorne, C., Swindell, R., Stratford, I. J., Dive, C. & Welman, A. Comparison of doxycycline delivery methods for Tet-inducible gene expression in a subcutaneous xenograft model. J. Biomol. Tech. 18, 120–123 (2007)

    PubMed  PubMed Central  Google Scholar 

  52. Zhao, X. et al. The N-Myc-DLL3 cascade is suppressed by the ubiquitin ligase Huwe1 to inhibit proliferation and promote neurogenesis in the developing brain. Dev. Cell 17, 210–221 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Wang, K. et al. Genome-wide identification of post-translational modulators of transcription factor activity in human B cells. Nature Biotechnol. 27, 829–837 (2009)

    CAS  Google Scholar 

Download references

Acknowledgements

We thank N. Sherman for phospho-ID2 and ID2-associated proteins analysis by mass spectrometry, C. Warnecke for the plasmid expressing HIF2α-TM, Z. Ronai for Flag–PHD1, Flag–PHD2, and Flag–PHD3, plasmids, A. Flores-Morales for the plasmid expressing Flag–SOCS2, M. Pagano for cDNAs for RBX1, Elongin B, Elongin C, and K. H. Kim for pcDNA-VHL. We thank D. D’Arca for preparation of VHL and HIFα constructs. This work was supported by National Institute of Health grants to A. L. (R01CA101644 and R01CA131126), and A. I. (R01CA178546 and R01NS061776 and a grant from The Chemotherapy Foundation). V. F. is supported by a fellowship from the American Brain Tumor Association (ABTA). S. B. L. was supported by NRF-2013R1A6A3A03063888 fellowship.

Author information

Authors and Affiliations

Authors

Contributions

A.I. and A.L. conceived the project, coordinated the study, oversaw the results and wrote the manuscript. S.B.L. designed and performed most biochemical and cell biology experiments and helped with writing the manuscript; V.F. generated lentiviral vectors, performed in vitro GSC infections, and RT–PCR and binding studies; A.M.C. generated and analysed glioma xenografts and assisted in GSC experiments; G.L. conducted the in vitro screening assay that identified DYRK1 as the ID2 Thr27 kinase. M.B. and A.C. conducted gene expression and bioinformatics analyses. J.N.B. provided excess tissue from human GBM for GSC isolation. D.S., K.H. and T.C. generated the computational molecular docking model. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Antonio Iavarone or Anna Lasorella.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 ID2 is phosphorylated on Ser5, Ser14 and Thr27.

Chromatographic results of mass spectrometry analysis of ID2 protein immunoprecipitated from IMR32 human neuroblastoma cells. a, The peptide identified as ID2 A3–R8 shows phosphorylation of Ser5. b, The peptide identified as ID2 K12–R24 shows phosphorylation of Ser14. c, The peptide identified as ID2 S25–L36 shows phosphorylation of Thr27.

Extended Data Figure 2 T27A missense mutation in ID2 in human cancer cells and Thr27 phosphorylation of ID2 by DYRK1 kinases.

a, Sequence analysis of genomic DNA from the neuroblastoma cell line IMR32 shows the wild-type sequence (left). Sequencing of DNA from the colon cancer cell line HRT-18 shows a heterozygous mutation resulting in the change of codon-27 from ACC (Thr) to GCC (Ala) (right). b, Both wild-type and mutant ID2(T27A) are expressed in HRT-18 colon cancer cells. Sequence analysis of representative clones (out of 20 clones) derived from HRT-18 cDNA demonstrates expression of wild-type (left panel) and mutant (right panel) alleles. c, Amino acid sequence flanking Thr27 of ID2 (marked in red), including the DYRK1 consensus motif (bold), is evolutionarily conserved. d, In vitro kinase assay using bacterially expressed GST–ID proteins and recombinant DYRK1A. e, U87 cells transfected with Flag–ID2, Flag–ID2(T27A) or the empty vector were immunoprecipitated with Flag antibody. Co-precipitated proteins were analysed by western blot using DYRK1A, DYRK1B and Flag antibodies. β-actin was used as control for loading. WCL, whole cellular lysate. f, U87 stably transfected with Flag–ID2 were treated with harmine (10 μM) or vehicle for 24 h and analysed by western blot using the indicated antibodies.

Extended Data Figure 3 DYRK1 kinase activity and Thr27 phosphorylation of ID2 are inhibited by hypoxia.

a, U87 glioma cells were treated with 100 μM CoCl2 for the indicated times. Cellular lysates were analysed by western blot using the indicated antibodies. b, SK-N-SH cells were treated with 300 μM CoCl2 for the indicated times and assayed by western blot using the indicated antibodies. c, Stoichiometric evaluation of pThr-27-ID2 in SK-N-SH cells untreated or treated with CoCl2 for 24 h. Cellular lysates prepared in denaturing buffer were immunoprecipitated using pT27-ID2 antibody or normal rabbit IgG. Aliquots of whole cellular lysates (WCL, μg) and immunoprecipitates were assayed by western blot using pT27-ID2 and non-phosphorylated ID2 antibodies (upper panels). The efficiency of immunoprecipitation with anti-pT27-ID2 antibody from untreated cells was determined to calculate the percent of the pT27-ID2 in the absence and in the presence of CoCl2 (lower panel). d, 293T cells expressing GFP–DYRK1 proteins untreated or treated with 100 μM CoCl2 for 12 h were used as a source of active kinase. The kinase activity of the anti-GFP–DYRK1 immunoprecipitates was tested in vitro using bacterially expressed and purified Flag–ID2 as substrate. Kinase reactions were evaluated by western blot using p-T27-ID2 antibodies (top). Analysis of kinase reactions by Flag immunoblot shows similar amount of ID2 protein in each kinase reaction (middle). Immunocomplexes were analysed by western blot using GFP antibody (bottom). e, Lysates from U251 cells expressing GFP–DYRK1 proteins untreated or treated with 100 μM CoCl2 for 6 h were immunoprecipitated using GFP antibodies. Western blot was performed using anti-p-Tyrosine (p-Tyr) or GFP antibodies. Analysis of WCL shows similar expression levels of DYRK1 proteins. α-tubulin was used as control for loading. f, Lysates from 293T cells expressing GFP–DYRK1A untreated or treated with 100 μM CoCl2 for 12 h were immunoprecipitated with anti-p-Tyr antibodies and analysed by western blot using antibodies against GFP. α-tubulin was used as control for loading. g, Lysates from 293T cells expressing GFP–DYRK1B untreated or treated with 100 μM CoCl2 for 12 h were immunoprecipitated with anti-p-Tyr antibodies and analysed by western blot using antibodies against GFP. α-tubulin was used as control for loading. h, U87 transfected with GFP–DYRK1A, GFP–DYRK1B or GFP and Flag–PHD1, Flag–PHD2, or Flag–PHD3 were immunoprecipitated using anti-hydroxyproline antibody. Western blot was performed using GFP antibody (upper panels). HC, IgG heavy chain. Lower panels, WCL.

Extended Data Figure 4 The DYRK1–ID2 Thr27 pathway controls GSCs and HIF2α.

a, GSC line 48 cells were transduced with lentiviruses expressing ID2(WT), ID2(T27A), or the empty vector. b, Cells were analysed by in vitro LDA. Representative regression plot used to calculate gliomasphere frequency in panel c. c, The frequency of cells capable of forming gliomaspheres by in vitro LDA. Data in the histograms represent means of 3 biological replicates ± s.d.; **P = 0.00163. d, The microphotographs show representative gliomasphere cultures of cells treated as in a. e, HIF2α mRNAs from cells treated as in a were analysed by semi-quantitative RT–PCR. f, U87 cells stably expressing Flag–ID2 or Flag–ID2(T27A) were analysed by western blot using the indicated antibodies. Arrow points to specific band. Arrowhead indicates Flag–ID2. Asterisk indicates endogenous ID2. g, GSC line 34 cells were transduced with lentiviruses expressing DYRK1B-V5 or empty vector. Cells were analysed by western blot using the indicated antibodies. Arrow points to specific band. Asterisk indicates a non-specific band. h, qRT–PCR from cells treated as in g. Data in the histograms represent means ± s.d. (n = 9, triplicate experiments each performed in triplicate; ***P = 8.44524 × 10−7 for TGFA). i, GSC line 31 was transduced with lentiviruses expressing DYRK1B-V5 or empty vector. Expression of HIF2α, DYRK1B-V5 and α-tubulin was analysed by western blot. j, mRNAs from experiment shown in Fig. 3a–c were analysed by semiquantitative RT–PCR for HIF2α. k, GSC line 31 cells were transduced with lentiviruses expressing DYRK1B and ID2, ID2(T27A), or the empty vector. Cells were analysed by LDA. Representative regression plot used to calculate gliomasphere frequency in Fig. 3b. l, GSC cell line 31 cells were transduced with lentiviruses expressing DYRK1B or the empty vector in the absence or in the presence of undegradable HIF2α (HIF2α-TM). Cells were analysed by in vitro LDA. Representative regression plot used to calculate the frequency of gliomaspheres in cultures from three independent infections (Vect plus Vect = 13.55%; DYRK1B-Vect = 4.36%; DYRK1B-HIF2α-TM = 9.73%).

Extended Data Figure 5 The DYRK1–ID2-Thr27 pathway modulates HIFα stability by regulating the interaction between ID2 and VHL.

a, In vivo ubiquitylation of HIF1α protein. U87 cells transfected with the expression plasmids HIF1α and MYC-ubiquitin were co-transfected with Flag–ID2, Flag–ID2(T27A), or the empty vector in the presence or in the absence of GFP–DYRK1B. After treatment with MG132 (20 μM) for 6 h, lysates were prepared in denaturing buffer and identical aliquots were immunoprecipitated with antibodies directed against MYC. An anti-HA antibody was used to detect HIF1α ubiquitin conjugates (left); Cellular lysates, WCL, were analysed by western blot using the indicated antibodies (right). b, U87 cells were co-transfected with plasmids expressing HA–HIF2α and GFP–DYRK1B or GFP-vector. Cells were treated with 50 μg ml−1 of CHX for the indicated times and analysed by western blot. c, Quantification of HIF2α protein from the experiment in panel b as the log2 of the percent of HIF2α relative to untreated cells. d, IMR32 cells were co-transfected with ID2 and Flag–VHL or Flag–HIF1α expression vectors. Immunoprecipitation was performed using Flag antibody and immunocomplexes and whole cellular lysates (WCL) were analysed by western blot using the indicated antibodies. e, IMR32 cells transfected with Flag–VHL expression vector were used for IgG or ID2 antibody immunoprecipitation. Immunocomplexes and WCL were analysed by western blot. Arrow points to the specific Flag–VHL band; asterisk indicates IgG light chain. f, Flag immunoprecipitation of binding reactions of in vitro translated Flag–ID and HA–Elongin C proteins. Immunocomplexes were analysed by western blot for HA and Flag. g, Flag-ID proteins and HA–VHL were translated and incubated in vitro. Flag immunocomplexes were analysed by western blot for HA and Flag. h, In vitro streptavidin pulldown assay of biotinylated ID2 peptides (amino acid 14–34 (WT), pT27, and T27W) and in vitro translated HA–VHL. Bound polypeptides were detected by western blot.

Extended Data Figure 6 Molecular docking of an ID2 (15–31) peptide on the VHL–Elongin C complex.

a, Ribbon representation of the backbone of the VHL-Elongin C complex and the predicted binding conformation of the ID2 peptide. VHL (red ribbon), Elongin C (blue ribbon) and the docked ID2 peptide (purple ribbon). Cul2 contact residues are colored yellow ribbon in both VHL and Elongin C. Arrow indicates the ID2 peptide. b, Docking result for the phospho-Thr-27-ID2 peptide shown from the same perspective as in panel a. c, The view and complex in a is rotated 90 degrees around an axis parallel to the page so that the perspective is from the arrow shown in panel a. d, Electrostatic molecular surface representation of the VHL–Elongin C complex with the docked ID2 peptide. The perspective is the same as in panel c. The T27 side chain is shown as space-filling spheres and is indicates by the red arrow. The N-terminus and C-terminus of the ID2 peptide are indicated by purple arrows.

Extended Data Figure 7 DYRK1-mediated phosphorylation of ID2 prevents dissociation of the VCB–Cul2 complex.

a, In vivo binding assay using lysates from U87 cells co-transfected with HA–VHL and Flag–ID2 or Flag–ID2(T27E) expression vectors. Flag immunocomplexes were analysed by western blot using HA and Flag antibodies. Whole cell lysates, WCL, were analysed by western blot using the indicated antibodies. Binding of Flag–ID2 and Flag–ID2(T27E) to the bHLH protein E47 is shown as control for ID2 binding. b, U87 cells were transfected with Flag–ID2, Flag–ID2(T27A) or Flag–ID2(T27E) plasmids. Cellular lysates were analysed by western blot using the indicated antibodies. c, In vitro binding between purified Flag-ID2 and His-VHL following in vitro kinase reaction using recombinant DYRK1B and Flag–ID2. d, Analysis of the HA–Elongin C immunocomplexes in U87 cells transfected with HA–Elongin C in the absence or presence of Flag–ID2(T27A). Anti-HA immunoprecipitation reactions and WCL were analysed by western blot using antibodies against Cul2, HA (Elongin C), and Flag (ID2). e, Analysis of the Flag–SOCS2 immunocomplexes in U87 cells transfected with ID2, ID2(T27A) or the empty vector. Flag immunoprecipitation reactions and WCL were analysed by western blot using antibodies against Cul5, ID2, and Flag (SOCS2). f, Stoichiometric analysis of ID2 and VHL in cellular lysates. Decreasing amount of WCL from 1 × 106 U87 cells and purified proteins were assayed by western blot (left). Regression plots of densitometry analysis were used to determine ID2 and VHL protein concentration and the ID2:VHL ratio (right). g, Immunoprecipitation of endogenous VHL in U87 cells in the presence and in the absence of CoCl2. Western blot for Cul2 and VHL are analysed by western blot. Vinculin is shown as loading control.

Extended Data Figure 8 DYRK1 kinase inhibits proliferation of human glioma.

a, Malignant glioma were induced in Id1Flox/Flox-Id2Flox/Flox-Id3-/- mice via injection of lentivirus expressing H-RAS-V12-IRES-CRE-ER linked to U6-shp53 cassette into the dentate gyrus as described9. Mice were treated for 5 days with tamoxifen or vehicle and euthanized 2 days later. Tumours were analysed by immunohistochemistry using HIF2α and OLIG2 antibodies. Nuclei were counterstained with haematoxylin. b, Western blot analysis of DYRK1B in U87 cells stably expressing a doxycycline inducible DYRK1B or the empty vector. Cells were treated with 0.75 μg ml−1 doxycycline or vehicle for 36 h. Lysates of adult mouse cortex (CX) and cerebellum (CB) were used to compare exogenous DYRK1B with endogenous levels of the protein. c, Tissue sections from experiment in Fig. 5e, f were analysed by immunostaining using BrdU antibodies. d, Quantification of BrdU positive cells from the experiment in c. Data in the histograms represent means ± s.d. (n = 5; ***P = 3.065 × 10−7, DYRK1B – Dox versus DYRK1B + Dox). Asterisks indicate statistical significance by two-tailed t-test. e, Western blot analysis of ectopically expressed V5-DYRK1B, V5-DYRK1B-K140R in U87 cells. f, Brain cross-sections of mice intracranially injected with U87 cells in e were analysed by immunofluorescence using V5 antibody (red, upper panels) to identify exogenous DYRK1B and human vimentin antibody (red, lower panels) to identify human glioma cells. Nuclei were counterstained with DAPI (blue). T, tumour; B, brain.

Extended Data Figure 9 Analysis of DYRK1A, DYRK1B and ID2 expression in human GBM.

a, Scatter plot showing the expression of DYRK1A and DYRK1B in GBM. Blue and red dots indicate GBM samples with high or low expression of both DYRK1A and DYRK1B, respectively. GBM samples were used for Kaplan–Meier survival analysis to evaluate the prognostic power of the expression of DYRK1A and DYRK1B shown in Fig. 5i. b, Distribution of ∆ESrand, representing the null model, for ID2 activity (left) and ID2 expression (right). This distribution is used to calculate the P value for enrichment of ∆ES. Red dot (or vertical black bar) represents the ∆ES using HIF2α targets. The P value is calculated as ratio of number of times ∆ESrand is greater than ∆ES (falls in green regions) over the total trials (= 1000).

Extended Data Figure 10 Model for the regulation of HIFα stability by the DYRK1 kinase and ID2 pathway.

In cellular contexts that favour HIFα protein instability (normal oxygen levels, but also low ID2 expression and high DYRK1 expression) prolyl hydroxylases (PHD1) is active and positively regulates DYRK1 kinases. Active, tyrosine phosphorylated DYRK1 kinases keep ID2 under functional constraint by phosphorylation of Thr27. The VCB–Cul2 ubiquitin ligase complex efficiently ubiquitylates HIFα (left). With decreasing oxygenation and PHD1 inactivation but also in the presence of downregulation of DYRK1, elevated expression of ID2, or ID2(T27A) mutation, the un-phosphorylated/un-phosphorylatable pool of ID2 exerts an inhibitory function towards the VCB–Cul2 complex by binding directly VHL and Elongin C proteins and displacing Cul2. This results in HIFα accumulation (right). The transcriptional activation of the ID2 gene, a HIFα target, by HIF2α generates a feed-forward ID2–HIF2α loop that amplifies the effects.

Supplementary information

Supplementary Figure 1

This file contains the uncropped gels for Figures 1, 2, 3, 4, 5 and Extended Data Figures 2, 3, 4, 5, 7, 8. (PDF 5363 kb)

Supplementary Tables

This file contains Supplementary Tables 1-4. (XLSX 12537 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, S., Frattini, V., Bansal, M. et al. An ID2-dependent mechanism for VHL inactivation in cancer. Nature 529, 172–177 (2016). https://doi.org/10.1038/nature16475

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature16475

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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