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:

NuRD–ZNF827 recruitment to telomeres creates a molecular scaffold for homologous recombination

Abstract

Alternative lengthening of telomeres (ALT) is a homologous recombination (HR)-dependent mechanism for de novo synthesis of telomeric DNA in mammalian cells. Nuclear receptors are bound to the telomeres of cells that use ALT. Here we demonstrate that nuclear receptors recruit ZNF827, a zinc-finger protein of unknown function, which recruits the nucleosome remodeling and histone deacetylation (NuRD) complex via binding to an N-terminal RRK motif within ZNF827. This results in decreased shelterin binding, hypoacetylation of telomeric chromatin, enhanced telomere-telomere interactions and recruitment of HR proteins, and it is critically important for cell viability and proliferation. We propose that NuRD–ZNF827 recruitment to human telomeres causes remodeling of telomeric chromatin and creates an environment that promotes telomere-telomere recombination and integrates and controls multiple mechanistic elements of ALT activity.

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: Nuclear-receptor recruitment of NuRD to ALT telomeres is cell cycle regulated.
Figure 2: The N-terminal RRK motif of ZNF827 is required for nuclear receptor–dependent NuRD recruitment to ALT telomeres.
Figure 3: NuRD–ZNF827 deacetylates histones and prevents shelterin binding at ALT telomeres.
Figure 4: NuRD–ZNF827 recruitment to telomeres promotes ALT activity.
Figure 5: ALT-specific telomere-telomere interactions are caused by NuRD–ZNF827.
Figure 6: ZNF827 knockdown increases the telomeric DDR and causes senescence and apoptosis in ALT cells.
Figure 7: NuRD–ZNF827 recruitment to telomeres drives HR and protects against the DDR in ALT cells.

Similar content being viewed by others

References

  1. Moyzis, R.K. et al. A highly conserved repetitive DNA sequence, (TTAGGG)n, present at the telomeres of human chromosomes. Proc. Natl. Acad. Sci. USA 85, 6622–6626 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. van Steensel, B., Smogorzewska, A. & de Lange, T. TRF2 protects human telomeres from end-to-end fusions. Cell 92, 401–413 (1998).

    Article  CAS  PubMed  Google Scholar 

  3. d'Adda di Fagagna, F. et al. A DNA damage checkpoint response in telomere-initiated senescence. Nature 426, 194–198 (2003).

    Article  CAS  PubMed  Google Scholar 

  4. Wang, R.C., Smogorzewska, A. & de Lange, T. Homologous recombination generates T-loop-sized deletions at human telomeres. Cell 119, 355–368 (2004).

    Article  CAS  PubMed  Google Scholar 

  5. Hockemeyer, D., Sfeir, A.J., Shay, J.W., Wright, W.E. & de Lange, T. POT1 protects telomeres from a transient DNA damage response and determines how human chromosomes end. EMBO J. 24, 2667–2678 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Celli, G.B., Denchi, E.L. & de Lange, T. Ku70 stimulates fusion of dysfunctional telomeres yet protects chromosome ends from homologous recombination. Nat. Cell Biol. 8, 885–890 (2006).

    Article  PubMed  Google Scholar 

  7. Sfeir, A., Kabir, S., van Overbeek, M., Celli, G.B. & de Lange, T. Loss of Rap1 induces telomere recombination in the absence of NHEJ or a DNA damage signal. Science 327, 1657–1661 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Shay, J.W. & Bacchetti, S. A survey of telomerase activity in human cancer. Eur. J. Cancer 33, 787–791 (1997).

    Article  CAS  PubMed  Google Scholar 

  9. Dunham, M.A., Neumann, A.A., Fasching, C.L. & Reddel, R.R. Telomere maintenance by recombination in human cells. Nat. Genet. 26, 447–450 (2000).

    Article  CAS  PubMed  Google Scholar 

  10. Déjardin, J. & Kingston, R.E. Purification of proteins associated with specific genomic loci. Cell 136, 175–186 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Conomos, D. et al. Variant repeats are interspersed throughout the telomeres and recruit nuclear receptors in ALT cells. J. Cell Biol. 199, 893–906 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lee, M. et al. Telomere extension by telomerase and ALT generates variant repeats by mechanistically distinct processes. Nucleic Acids Res. 42, 1733–1746 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Heaphy, C.M. et al. Altered telomeres in tumors with ATRX and DAXX mutations. Science 333, 425 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Schwartzentruber, J. et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482, 226–231 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Lovejoy, C.A. et al. Loss of ATRX, genome instability, and an altered DNA damage response are hallmarks of the alternative lengthening of telomeres pathway. PLoS Genet. 8, e1002772 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bower, K. et al. Loss of wild-type ATRX expression in somatic cell hybrids segregates with activation of alternative lengthening of telomeres. PLoS ONE 7, e50062 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. O'Sullivan, R.J. et al. Rapid induction of alternative lengthening of telomeres by depletion of the histone chaperone ASF1. Nat. Struct. Mol. Biol. 21, 167–174 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lai, A.Y. & Wade, P.A. Cancer biology and NuRD: a multifaceted chromatin remodelling complex. Nat. Rev. Cancer 11, 588–596 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Conomos, D., Pickett, H.A. & Reddel, R.R. Alternative lengthening of telomeres: remodeling the telomere architecture. Front. Oncol. 3, 27 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Cui, S. et al. Nuclear receptors TR2 and TR4 recruit multiple epigenetic transcriptional corepressors that associate specifically with the embryonic β-type globin promoters in differentiated adult erythroid cells. Mol. Cell. Biol. 31, 3298–3311 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Pickett, H.A., Cesare, A.J., Johnstone, R.L., Neumann, A.A. & Reddel, R.R. Control of telomere length by a trimming mechanism that involves generation of t-circles. EMBO J. 28, 799–809 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hong, W. et al. FOG-1 recruits the NuRD repressor complex to mediate transcriptional repression by GATA-1. EMBO J. 24, 2367–2378 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lauberth, S.M. & Rauchman, M. A conserved 12-amino acid motif in Sall1 recruits the nucleosome remodeling and deacetylase corepressor complex. J. Biol. Chem. 281, 23922–23931 (2006).

    Article  CAS  PubMed  Google Scholar 

  24. Cismasiu, V.B. et al. BCL11B functionally associates with the NuRD complex in T lymphocytes to repress targeted promoter. Oncogene 24, 6753–6764 (2005).

    Article  CAS  PubMed  Google Scholar 

  25. Topark-Ngarm, A. et al. CTIP2 associates with the NuRD complex on the promoter of p57KIP2, a newly identified CTIP2 target gene. J. Biol. Chem. 281, 32272–32283 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Wang, Y. et al. LSD1 is a subunit of the NuRD complex and targets the metastasis programs in breast cancer. Cell 138, 660–672 (2009).

    Article  CAS  PubMed  Google Scholar 

  27. Porro, A., Feuerhahn, S. & Lingner, J. TERRA-reinforced association of LSD1 with MRE11 promotes processing of uncapped telomeres. Cell Reports 6, 765–776 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Yeager, T.R. et al. Telomerase-negative immortalized human cells contain a novel type of promyelocytic leukemia (PML) body. Cancer Res. 59, 4175–4179 (1999).

    CAS  PubMed  Google Scholar 

  29. Henson, J.D. et al. DNA C-circles are specific and quantifiable markers of alternative-lengthening-of-telomeres activity. Nat. Biotechnol. 27, 1181–1185 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Londoño-Vallejo, J.A., Der-Sarkissian, H., Cazes, L., Bacchetti, S. & Reddel, R.R. Alternative lengthening of telomeres is characterized by high rates of telomeric exchange. Cancer Res. 64, 2324–2327 (2004).

    Article  PubMed  Google Scholar 

  31. Bechter, O.E., Zou, Y., Walker, W., Wright, W.E. & Shay, J.W. Telomeric recombination in mismatch repair deficient human colon cancer cells after telomerase inhibition. Cancer Res. 64, 3444–3451 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Cesare, A.J. et al. Spontaneous occurrence of telomeric DNA damage response in the absence of chromosome fusions. Nat. Struct. Mol. Biol. 16, 1244–1251 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Cesare, A.J. & Reddel, R.R. Alternative lengthening of telomeres: models, mechanisms and implications. Nat. Rev. Genet. 11, 319–330 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Pan, M.R. et al. Chromodomain helicase DNA-binding protein 4 (CHD4) regulates homologous recombination DNA repair, and its deficiency sensitizes cells to poly(ADP-ribose) polymerase (PARP) inhibitor treatment. J. Biol. Chem. 287, 6764–6772 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lin, S.Y., Liang, Y. & Li, K. Multiple roles of BRIT1/MCPH1 in DNA damage response, DNA repair, and cancer suppression. Yonsei Med. J. 51, 295–301 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Wu, G., Jiang, X., Lee, W.H. & Chen, P.L. Assembly of functional ALT-associated promyelocytic leukemia bodies requires Nijmegen breakage syndrome 1. Cancer Res. 63, 2589–2595 (2003).

    CAS  PubMed  Google Scholar 

  37. Draskovic, I. et al. Probing PML body function in ALT cells reveals spatiotemporal requirements for telomere recombination. Proc. Natl. Acad. Sci. USA 106, 15726–15731 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Smeenk, G. et al. The NuRD chromatin-remodeling complex regulates signaling and repair of DNA damage. J. Cell Biol. 190, 741–749 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lejon, S. et al. Insights into association of the NuRD complex with FOG-1 from the crystal structure of an RbAp48.FOG-1 complex. J. Biol. Chem. 286, 1196–1203 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Muntoni, A., Neumann, A.A., Hills, M. & Reddel, R.R. Telomere elongation involves intra-molecular DNA replication in cells utilizing alternative lengthening of telomeres. Hum. Mol. Genet. 18, 1017–1027 (2009).

    Article  CAS  PubMed  Google Scholar 

  41. Chan, K.L., North, P.S. & Hickson, I.D. BLM is required for faithful chromosome segregation and its localization defines a class of ultrafine anaphase bridges. EMBO J. 26, 3397–3409 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Barefield, C. & Karlseder, J. The BLM helicase contributes to telomere maintenance through processing of late-replicating intermediate structures. Nucleic Acids Res. 40, 7358–7367 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Park, S.W. et al. SUMOylation of Tr2 orphan receptor involves Pml and fine-tunes Oct4 expression in stem cells. Nat. Struct. Mol. Biol. 14, 68–75 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Polo, S.E., Kaidi, A., Baskcomb, L., Galanty, Y. & Jackson, S.P. Regulation of DNA-damage responses and cell-cycle progression by the chromatin remodelling factor CHD4. EMBO J. 29, 3130–3139 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Sims, J.K. & Wade, P.A. Mi-2/NuRD complex function is required for normal S phase progression and assembly of pericentric heterochromatin. Mol. Biol. Cell 22, 3094–3102 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Chou, D.M. et al. A chromatin localization screen reveals poly (ADP ribose)-regulated recruitment of the repressive polycomb and NuRD complexes to sites of DNA damage. Proc. Natl. Acad. Sci. USA 107, 18475–18480 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Doksani, Y., Wu, J.Y., de Lange, T. & Zhuang, X. Super-resolution fluorescence imaging of telomeres reveals TRF2-dependent T-loop formation. Cell 155, 345–356 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sfeir, A. et al. Mammalian telomeres resemble fragile sites and require TRF1 for efficient replication. Cell 138, 90–103 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Martínez, P. et al. Increased telomere fragility and fusions resulting from TRF1 deficiency lead to degenerative pathologies and increased cancer in mice. Genes Dev. 23, 2060–2075 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Episkopou, H. et al. Alternative lengthening of telomeres is characterized by reduced compaction of telomeric chromatin. Nucleic Acids Res. 42, 4391–4405 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Benetti, R. et al. Suv4–20h deficiency results in telomere elongation and derepression of telomere recombination. J. Cell Biol. 178, 925–936 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Gonzalo, S. et al. DNA methyltransferases control telomere length and telomere recombination in mammalian cells. Nat. Cell Biol. 8, 416–424 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Benetti, R., Garcia-Cao, M. & Blasco, M.A. Telomere length regulates the epigenetic status of mammalian telomeres and subtelomeres. Nat. Genet. 39, 243–250 (2007).

    Article  CAS  PubMed  Google Scholar 

  54. Stern, J.L., Zyner, K.G., Pickett, H.A., Cohen, S.B. & Bryan, T.M. Telomerase recruitment requires both TCAB1 and Cajal bodies independently. Mol. Cell. Biol. 32, 2384–2395 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Cohen, S.B. & Reddel, R.R. A sensitive direct human telomerase activity assay. Nat. Methods 5, 355–360 (2008).

    Article  CAS  PubMed  Google Scholar 

  56. Conomos, D. et al. Variant repeats are interspersed throughout the telomeres and recruit nuclear receptors in ALT cells. J. Cell Biol. 199, 893–906 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Pickett, H.A., Cesare, A.J., Johnstone, R.L., Neumann, A.A. & Reddel, R.R. Control of telomere length by a trimming mechanism that involves generation of t-circles. EMBO J. 28, 799–809 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Smyth, C.M., Helmer, M.A., Dalla-Pozza, L. & Rowe, P.B. Flow cytometric DNA analyses of frozen samples from children's solid tumors. Pathology 25, 388–393 (1993).

    Article  CAS  PubMed  Google Scholar 

  59. Henson, J.D. et al. DNA C-circles are specific and quantifiable markers of alternative-lengthening-of-telomeres activity. Nat. Biotechnol. 27, 1181–1185 (2009).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by an Australian Postgraduate Award (to D.C.), a Cancer Institute of New South Wales (NSW) Research Scholar Award (to D.C.), National Health and Medical Research Council of Australia project grant 1009231 (to R.R.R. and H.A.P.), Cancer Council NSW project grant 1069550 (to H.A.P.), Cancer Council NSW program grant PG11-08 (to R.R.R.) and a Cancer Institute NSW Fellowship (to H.A.P.).

Author information

Authors and Affiliations

Authors

Contributions

All authors conceived and designed the project. Experiments were conducted by D.C. and H.A.P. All authors analyzed data and authored the manuscript. We thank J. Mackay (University of Sydney) for advice and helpful discussion.

Corresponding authors

Correspondence to Roger R Reddel or Hilda A Pickett.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Cell cycle regulation of NuRD recruitment by nuclear receptors.

(a) Western blot analysis of panel of mortal, telomerase-positive and ALT cell lines showing global expression levels of nuclear receptors and NuRD components.

(b) Telomere-ChIP against nuclear receptors in synchronized WI38-VA13/2RA cells. Quantitation of the percentage of telomeric DNA pulled down is presented in the accompanying graph (mean ± range; n = 2 biological replicates).

(c) Western blot analysis of synchronized WI38-VA13/2RA cells showing global expression levels of nuclear receptors and NuRD components.

(d) Telomere-ChIP against nuclear receptors in HT1080 hTR and HT1080 hTR TCA cells. Quantitation is presented in the accompanying graph (mean ± range; n = 2 biological replicates; *P<0.05 by two tailed t test).

(e) Western blot analysis of HT1080 hTR and HT1080 hTR TCA cells showing global expression levels of nuclear receptors as well as NuRD components.

Supplementary Figure 2 ZNF827 binding to telomeres is cell cycle regulated and ALT specific.

(a) Telomere-ChIP against ZNF827 in the HT1080 hTR and HT1080 hTR TCA cell lines. Quantitation of the percentage of telomeric DNA pulled down is presented in the accompanying graph (mean ± range; n = 2 biological replicates; *P<0.05 by two tailed t test).

(b) Reverse Co-IP of NuRD and shelterin components from WI38-VA13/2RA cells overexpressing ZNF827 followed by western blot analysis to detect ZNF827.

(c) Indirect immunofluorescence against the NuRD component RBAP46 (red) and Myc-tagged ZNF827 (purple) coupled with telomere-FISH (green) and DAPI (blue) in WI38-VA13/2RA nuclei 72 h following knockdown of both TR4 and COUP-TF2. Bar represents 10 μm.

(d) Real-time RT-PCR showing ZNF827 transcript levels in WI38-VA13/2RA cells 72 h post-transfection with siRNA against different nuclear receptors (mean ± 1 s.d.; n = 3 technical replicates; **P<0.005 by two tailed t test).

(e) Real-time RT-PCR showing ZNF827 transcript levels in WI38-VA13/2RA cells 48 h after overexpression of different nuclear receptors (mean ± 1 s.d.; n = 3 technical replicates; **P<0.005 by two tailed t test).

(f) Real-time RT-PCR showing ZNF827 transcript levels in a panel of mortal, telomerase-positive and ALT cell lines (mean ± 1 s.d.; n = 3 technical replicates).

(g) Telomere-ChIP against ZNF827 in synchronized WI38-VA13/2RA cells. Quantitation is presented in the accompanying graph (mean ± range; n = 2 biological replicates).

(h) Real-time RT-PCR showing ZNF827 transcript levels in synchronized WI38-VA13/2RA cells (mean ± 1 s.d.; n = 3 technical replicates).

(i) Real-time RT-PCR showing ZNF827 transcript levels in HT1080 hTR and HT1080 hTR TCA cells (mean ± 1 s.d.; n = 3 technical replicates; ***P<0.0001 by two tailed t test).

Supplementary Figure 3 NuRD–ZNF827 remodels ALT telomeric chromatin.

(a) Telomere-ChIP against histones and histone marks in WI38-VA13/2RA cells 72 h after simultaneous siRNA-mediated knockdown of TR4 and COUP-TF2. Quantitation of the percentage of telomeric DNA pulled down normalized to the appropriate histone (either H3 or H4) is presented in the two accompanying graphs (mean ± range; n = 2 biological replicates; *P<0.05 by two tailed t test).

(b) Telomere-ChIP against histones and histone marks in a panel of mortal, telomerase-positive and ALT cell lines. Quantitation normalized to the appropriate histone is presented in the two accompanying graphs (mean ± range; n = 2 biological replicates).

Supplementary Figure 4 ALT phenotypic characteristics are affected by ZNF827 expression.

(a) Real-time RT-PCR confirming knockdown of ZNF827 transcript levels 72 h post-transfection with siRNA in WI38-VA13/2RA and IIICF/c cells (mean ± 1 s.d.; n = 3 technical replicates; **P<0.005 and ***P<0.0001 by two tailed t test).

(b) Western blot analysis of WI38-VA13/2RA and IIICF/c cells 48 h after overexpression of ZNF827.

(c) Western blot analysis of HT1080 hTR cells 48 h after overexpression of ZNF827.

(d) CO-FISH of metaphases from HT1080 hTR cells 48 h post-ZNF827 overexpression. Leading (red; Texas Red-conjugated (TTAGGG)3) and lagging (green; Alexa Fluor 488-conjugated (CCCTAA)3) telomere-FISH coupled with DAPI (blue) counterstaining. Bar represents 10 μm.

(e) Quantitation of the percentage of chromosome ends with two colors on only one sister chromatid observed 48 h after ZNF827 overexpression in WI38-VA13/2RA cells via CO-FISH (mean ± 1 s.d.; n = 3 biological replicates, quantifying >2,000 chromosome ends per replicate; *P<0.05 by two tailed t test).

(f) Quantitation of the percentage of fragile telomeres observed 48 h after ZNF827 overexpression in WI38-VA13/2RA cells via CO-FISH (mean ± 1 s.d.; n = 3 biological replicates, quantifying >2,000 chromosome ends per replicate; *P<0.05 by two tailed t test).

(g) Quantitation of the percentage of chromosome ends associating with ECTR DNA observed 48 h after ZNF827 overexpression in WI38-VA13/2RA cells via CO-FISH (mean ± 1 s.d.; n = 3 biological replicates, quantifying >2,000 chromosome ends per replicate; *P<0.05 by two tailed t test).

Supplementary Figure 5 ALT-specific telomere bridges caused by the NuRD–ZNF827 complex lead to prolonged prometaphase.

(a) Quantitation of the percentage of (pro)metaphases with telomere bridges in the HT1080 hTR and HT1080 hTR TCA cell lines (mean ± 1 s.d.; n = 3 biological replicates, quantifying ~50 (pro)metaphases per replicate; ***P<0.0001 by two tailed t test).

(b) Quantitation of the percentage of WI38-VA13/2RA cells with telomere bridges 48 h after ZNF827 overexpression and 24 h after treatment with various HDAC inhibitors (mean ± 1 s.d.; n = 3 biological replicates, quantifying ~50 cells per replicate; *P<0.05 by two tailed t test).

(c) Telomere-ChIP against NuRD components in WI38-VA13/2RA cells 24 h after TSA treatment. Quantitation of the percentage of telomeric DNA pulled down is presented in the accompanying graph (mean ± range; n = 2 biological replicates; *P<0.05 by two tailed t test).

(d) Quantitation of the percentage of WI38-VA13/2RA cells with telomere bridges with or without the use of standard cytogenetic methods (mean ± 1 s.d.; n = 3 biological replicates, quantifying ~50 cells per replicate; ***P<0.0001 by two tailed t test).

(e) Quantitation of the percentage of cells with telomere bridges 48 h after ZNF827 overexpression in synchronized WI38-VA13/2RA cells (mean ± 1 s.d.; n = 3 biological replicates, quantifying ~50 cells per replicate; *P<0.05 and **P<0.005 by two tailed t test).

(f) Western blot analysis of WI38-VA13/2RA cells following siRNA-mediated knockdown (72 h) and overexpression (48 h) of ZNF827 showing the global expression of cell cycle markers.

(g) Quantitation of the percentage of HT1080 hTR, WI38-VA13/2RA and IIICF/c cells in each stage of the cell cycle following siRNA-mediated knockdown (72 h) and overexpression (48 h) of ZNF827 (mean ± 1 s.d.; n = 3 biological replicates).

Supplementary Figure 6 Validation of ZNF827 knockdown in cell-line panel.

Real-time RT-PCR confirming the knockdown of ZNF827 transcript levels 72 h post-transfection with siRNA in the entire panel of cell lines shown in Figure 6a (mean ± 1 s.d.; n = 3 technical replicates; *P<0.05 by two tailed t test).

Supplementary Figure 7 Original western blots for main figures.

(a) Complete western blots of nuclear receptors and NuRD components in WI38-VA13/2RA cells after nuclear receptor knockdown shown in Figure 1d. The molecular weight standards for all blots are shown on the left.

(b) Complete western blot of HT1080 hTR and WI38-VA13/2RA cells after ZNF827 overexpression shown in Figure 2b. The molecular weight standards for all blots are shown on the left.

(c) Complete western blots of NuRD and shelterin components for Co-IP of ZNF827 from WI38-VA13/2RA cells overexpressing ZNF827 shown in Figure 2g. The molecular weight standards for all blots are shown on the left.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 5532 kb)

Supplementary Table 1

List of primary antibodies used in this study (XLSX 15 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Conomos, D., Reddel, R. & Pickett, H. NuRD–ZNF827 recruitment to telomeres creates a molecular scaffold for homologous recombination. Nat Struct Mol Biol 21, 760–770 (2014). https://doi.org/10.1038/nsmb.2877

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2877

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