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:

GUARDIN is a p53-responsive long non-coding RNA that is essential for genomic stability

Abstract

The list of long non-coding RNAs (lncRNAs) involved in the p53 pathway of the DNA damage response is rapidly expanding, but whether lncRNAs have a role in maintaining the de novo structure of DNA is unknown. Here, we demonstrate that the p53-responsive lncRNA GUARDIN is important for maintaining genomic integrity under steady-state conditions and after exposure to exogenous genotoxic stress. GUARDIN is necessary for preventing chromosome end-to-end fusion through maintaining the expression of telomeric repeat-binding factor 2 (TRF2) by sequestering microRNA-23a. Moreover, GUARDIN also sustains breast cancer 1 (BRCA1) stability by acting as an RNA scaffold to facilitate the heterodimerization of BRCA1 and BRCA1-associated RING domain protein 1 (BARD1). As such, GUARDIN silencing triggered apoptosis and senescence, enhanced cytotoxicity of additional genotoxic stress and inhibited cancer xenograft growth. Thus, GUARDIN may constitute a target for cancer treatment.

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: Identification of GUARDIN as a p53-inducible lncRNA.
Fig. 2: GUARDIN is expressed at higher levels in colon cancers with wild-type p53 than in those carrying mutant p53.
Fig. 3: GUARDIN is important for cell survival and proliferation.
Fig. 4: GUARDIN sequesters miR-23a to stabilize TRF2.
Fig. 5: GUARDIN binds to and stabilizes BRCA1.
Fig. 6: GUARDIN protects genomic integrity through TRF2 and BRCA1.
Fig. 7: GUARDIN protects cells from apoptosis induced by genotoxic insults through TRF2 and BRCA1.

Similar content being viewed by others

References

  1. Hoeijmakers, J. H. J. DNA damage, aging, and cancer. N. Engl. J. Med. 361, 1475–1485 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Hoeijmakers, J. H. J. The key role of DNA damage on cancer, aging and longevity. Environ. Mol. Mutagen. 53, S13 (2012).

    Google Scholar 

  3. Braig, M. & Schmitt, C. A. Oncogene-induced senescence: putting the brakes on tumor development. Cancer Res. 66, 2881–2884 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Best, B. P. Nuclear DNA damage as a direct cause of aging. Rejuvenation Res. 12, 199–208 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Lahtz, C. & Pfeifer, G. P. Epigenetic changes of DNA repair genes in cancer. J. Mol. Cell Biol. 3, 51–58 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Xin, H. W., Liu, D. & Zhou, S. Y. The telosome/shelterin complex and its functions. Genome Biol. 9, 232 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Palm, W. & de Lange, T. How shelterin protects mammalian telomeres. Annu. Rev. Genet. 42, 301–334 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Liu, D., O’Connor, M. S., Qin, J. & Songyang, Z. Telosome, a mammalian telomere-associated complex formed by multiple telomeric proteins. J. Biol. Chem. 279, 51338–51342 (2004).

    Article  CAS  PubMed  Google Scholar 

  9. Fumagalli, M. et al. Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nat. Cell Biol. 14, 355–365 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. van Tuyn, J. & Adams, P. D. Signalling the end of the line. Nat. Cell Biol. 14, 339–341 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Martinez, P. & Blasco, M. A. Role of shelterin in cancer and aging. Aging Cell 9, 653–666 (2010).

    Article  CAS  PubMed  Google Scholar 

  12. Majidinia, M. & Yousefi, B. DNA damage response regulation by microRNAs as a therapeutic target in cancer. DNA Repair 47, 1–11 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Idogawa, M. et al. Identification and analysis of large intergenic non-coding RNAs regulated by p53 family members through a genome-wide analysis of p53-binding sites. Hum. Mol. Genet. 23, 2847–2857 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Huarte, M. et al. A large intergenic noncoding RNA Induced by p53 mediates global gene repression in the p53 response. Cell 142, 409–419 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Schmitt, A. M. et al. An inducible long noncoding RNA amplifies DNA damage signaling. Nat. Genet. 48, 1370–1376 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Di Micco, R. et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 444, 638–642 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Léveillé, N. et al. Genome-wide profiling of p53-regulated enhancer RNAs uncovers a subset of enhancers controlled by a lncRNA. Nat. Commun. 27, 6520 (2015).

    Article  Google Scholar 

  18. Wang, W., Cheng, B., Miao, L., Mei, Y. & Wu, M. Mutant p53-R273H gains new function in sustained activation of EGFR signaling via suppressing miR-27a expression. Cell Death Dis. 4, e574 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Tsang, W. P., Ho, F. Y., Fung, K. P., Kong, S. K. & Kwok, T. T. p53-R175H mutant gains new function in regulation of doxorubicin-induced apoptosis. Int. J. Cancer 114, 331–336 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Liu, D. P., Song, H. & Xu, Y. A common gain of function of p53 cancer mutants in inducing genetic instability. Oncogene 29, 949–956 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Jiang, P. et al. p53 regulates biosynthesis through direct inactivation of glucose-6-phosphate dehydrogenase. Nat. Cell Biol. 13, 310–316 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Georgakilas, A. G. et al. Are common fragile sites merely structural domains or highly organized “functional” units susceptible to oncogenic stress? Cell. Mol. Life Sci. 71, 4519–4544 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Henrich, K. O., Schwab, M. & Westermann, F. 1p36 tumor suppression—a matter of dosage? Cancer Res. 72, 6079–6088 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Hunten, S. et al. p53-regulated networks of protein, mRNA, miRNA, and lncRNA expression revealed by integrated pulsed stable isotope labeling with amino acids in cell culture (pSILAC) and next generation sequencing (NGS) analyses. Mol. Cell. Proteom. 14, 2609–2629 (2015).

    Article  Google Scholar 

  25. Ashouri, A. et al. Pan-cancer transcriptomic analysis associates long non-coding RNAs with key mutational driver events. Nat. Commun. 7, 13197 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Sarkar, S. et al. Different combinations of genetic/epigenetic alterations inactivate the p53 and pRb pathways in invasive human bladder cancers. Cancer Res. 60, 3862–3871 (2000).

    CAS  PubMed  Google Scholar 

  27. Vikhanskaya, F., Lee, M. K., Mazzoletti, M., Broggini, M. & Sabapathy, K. Cancer-derived p53 mutants suppress p53-target gene expression—potential mechanism for gain of function of mutant p53. Nucleic Acids Res. 35, 2093–2104 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Li, Q. et al. C23 promotes tumorigenesis via suppressing p53 activity. Oncotarget. 7, 58274–58285 (2016).

    PubMed  PubMed Central  Google Scholar 

  29. Debacq-Chainiaux, F., Erusalimsky, J. D., Campisi, J. & Toussaint, O. Protocols to detect senescence-associated β-galactosidase (SA-β-gal) activity, a biomarker of senescent cells in culture and in vivo. Nat. Protoc. 4, 1798–1806 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Dimri, G. P. et al. A biomarker that identifies senescent human-cells in culture and in aging skin in vivo. Proc. Natl Acad. Sci. USA 92, 9363–9367 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sharma, V. et al. A BRCA1-interacting lncRNA regulates homologous recombination. EMBO Rep. 16, 1520–1534 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kartha, R. V & Subramanian, S. Competing endogenous RNAs (ceRNAs): new entrants to the intricacies of gene regulation. Front. Genet. 5, 8 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Tay, Y., Rinn, J. & Pandolfi, P. P. The multilayered complexity of ceRNA crosstalk and competition. Nature 505, 344–352 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dodd, D. W., Gagnon, K. T. & Corey, D. R. Digital quantitation of potential therapeutic target RNAs. Nucleic Acid. Ther. 23, 188–194 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Luo, Z. H. et al. mir-23a induces telomere dysfunction and cellular senescence by inhibiting TRF2 expression. Aging Cell 14, 391–399 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Choi, K. H., Farrell, A. S., Lakamp, A. S. & Ouellette, M. M. Characterization of the DNA binding specificity of shelterin complexes. Nucleic Acids Res. 39, 9206–9223 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yan, Y., Liang, Z., Du, Q., Yang, M. & Geller, D. A. MicroRNA-23a downregulates the expression of interferon regulatory factor-1 in hepatocellular carcinoma cells. Oncol. Rep. 36, 633–640 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Baer, R. & Ludwig, T. The BRCA1/BARD1 heterodimer, a tumor suppressor complex with ubiquitin E3 ligase activity. Curr. Opin. Genet. Dev. 12, 86–91 (2002).

    Article  CAS  PubMed  Google Scholar 

  39. Hashizume, R. et al. The RING heterodimer BRCA1–BARD1 is a ubiquitin ligase inactivated by a breast cancer-derived mutation. J. Biol. Chem. 276, 14537–14540 (2001).

    Article  CAS  PubMed  Google Scholar 

  40. Brzovic, P. S., Rajagopal, P., Hoyt, D. W., King, M. C. & Klevit, R. E. Structure of a BRCA1–BARD1 heterodimeric RING–RING complex. Nat. Struct. Biol. 8, 833–837 (2001).

    Article  CAS  PubMed  Google Scholar 

  41. Shrivastav, M., De Haro, L. P. & Nickoloff, J. A. Regulation of DNA double-strand break repair pathway choice. Cell Res. 18, 134–147 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Vogelstein, B., Lane, D. & Levine, A. J. Surfing the p53 network. Nature 408, 307–310 (2000).

    Article  CAS  PubMed  Google Scholar 

  43. Galanos, P. et al. Chronic p53-independent p21 expression causes genomic instability by deregulating replication licensing. Nat. Cell Biol. 18, 777–789 (2016).

    Article  CAS  PubMed  Google Scholar 

  44. Yin, D. et al. miR-34a functions as a tumor suppressor modulating EGFR in glioblastoma multiforme. Oncogene 32, 1155–1163 (2013).

    Article  CAS  PubMed  Google Scholar 

  45. Okada, N. et al. A positive feedback between p53 and miR-34 miRNAs mediates tumor suppression. Genes Dev. 28, 438–450 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wynford-Thomas, D. p53: guardian of cellular senescence. J. Pathol. 180, 118–121 (1996).

    Article  CAS  PubMed  Google Scholar 

  47. Stiewe, T. The p53 family in differentiation and tumorigenesis. Nat. Rev. Cancer 7, 165–168 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Karlseder, J., Broccoli, D., Dai, Y. M., Hardy, S. & de Lange, T. p53- and ATM-dependent apoptosis induced by telomeres lacking TRF2. Science 283, 1321–1325 (1999).

    Article  CAS  PubMed  Google Scholar 

  49. Takai, H., Smogorzewska, A. & de Lange, T. DNA damage foci at dysfunctional telomeres. Curr. Biol. 13, 1549–1556 (2003).

    Article  CAS  PubMed  Google Scholar 

  50. Wang, Y. et al. BASC, a super complex of BRCA1-associated proteins involved in the recognition and repair of aberrant DNA structures. Genes Dev. 14, 927–939 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Friedenson, B. The BRCA1/2 pathway prevents hematologic cancers in addition to breast and ovarian cancers. BMC Cancer 7, 152 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Tapia, T. et al. Promoter hypermethylation of BRCA1 correlates with absence of expression in hereditary breast cancer tumors. Epigenetics 3, 157–163 (2008).

    Article  PubMed  Google Scholar 

  53. Shen, J., Ambrosone, C. B. & Zhao, H. Novel genetic variants in microRNA genes and familial breast cancer. Int. J. Cancer 124, 1178–1182 (2009).

    Article  CAS  PubMed  Google Scholar 

  54. Beck, M. et al. The quantitative proteome of a human cell line. Mol. Syst. Biol. 7, 549 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Badie, S. et al. BRCA1 and CtIP promote alternative non-homologous end-joining at uncapped telomeres. EMBO J. 34, 410–424 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Isono, M. et al. BRCA1 directs the repair pathway to homologous recombination by promoting 53BP1 dephosphorylation. Cell Rep. 18, 520–532 (2017).

    Article  CAS  PubMed  Google Scholar 

  57. Powell, S. N. & Kachnic, L. A. Roles of BRCA1 and BRCA2 in homologous recombination, DNA replication fidelity and the cellular response to ionizing radiation. Oncogene 22, 5784–5791 (2003).

    Article  CAS  PubMed  Google Scholar 

  58. Feng, Z. & Zhang, J. A dual role of BRCA1 in two distinct homologous recombination mediated repair in response to replication arrest. Nucleic Acids Res. 40, 726–738 (2012).

    Article  CAS  PubMed  Google Scholar 

  59. Bau, D. T., Mau, Y. C. & Shen, C. Y. The role of BRCA1 in non-homologous end-joining. Cancer Lett. 240, 1–8 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. Jiang, G. et al. BRCA1–Ku80 protein interaction enhances end-joining fidelity of chromosomal double-strand breaks in the G1 phase of the cell cycle. J. Biol. Chem. 288, 8966–8976 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Brown, J. S., O’Carrigan, B., Jackson, S. P. & Yap, T. A. Targeting DNA repair in cancer: beyond PARP inhibitors. Cancer Discov. 7, 20–37 (2017).

    Article  CAS  PubMed  Google Scholar 

  62. Audeh, M. W. et al. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial. Lancet 376, 245–251 (2010).

    Article  CAS  PubMed  Google Scholar 

  63. Tutt, A. et al. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet 376, 235–244 (2010).

    Article  CAS  PubMed  Google Scholar 

  64. Dong, L. et al. Ets-1 mediates upregulation of Mcl-1 downstream of XBP-1 in human melanoma cells upon ER stress. Oncogene 30, 3716–3726 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Dodd, D. W., Gagnon, K. T. & Corey, D. R. Digital quantitation of potential therapeutic target RNAs. Nucleic Acid. Ther. 23, 188–194 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Karbaschi, M. & Cooke, M. S. Novel method for the high-throughput processing of slides for the comet assay. Sci. Rep. 4, 7200 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Liu, X. Y. et al. RIP1 kinase is an oncogenic driver in melanoma. Cancer Res. 75, 1736–1748 (2015).

    Article  CAS  PubMed  Google Scholar 

  68. Jin, L. et al. MicroRNA-149*, a p53-responsive microRNA, functions as an oncogenic regulator in human melanoma. Proc. Natl Acad. Sci. USA 108, 15840–15845 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Guzman, C., Bagga, M., Kaur, A., Westermarck, J. & Abankwa, D. ColonyArea: an ImageJ plugin to automatically quantify colony formation in clonogenic assays. PLoS ONE 9, e92444 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank X. Xu of Shenzhen University for providing U2OS-HR and U2OS-NHEJ cells and T. Ohta of St. Marianna University for providing the HA-BARD1 plasmid. We thank L. Kong and Q. Cheng of Henan Provincial People’s Hospital for their assistance with preparation of tissue sections and immunohistochemistry experiments. This work was supported by grants from the National Key R&D Program of China (2016YFC1302302) and the National Natural Science Foundation of China (81430065, 31371388, 31601117 and 81471551).

Author information

Authors and Affiliations

Authors

Contributions

W.L.H., L.J., A.X., X.D.Z. and M.W. designed the research. W.L.H., L.J. and A.X. performed most of the experiments and data analysis. Y.F.W. participated in the experiments and data analysis. R.F.T. participated in the data analysis and manuscript preparation. M.W., R.F.T. and X.D.Z. wrote the manuscript.

Corresponding authors

Correspondence to Xu Dong Zhang or Mian Wu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 lncRNA#6, GUARDIN, is regulated by wildtype p53 and its depletion induces apoptosis and reduced cell viability.

(a) p53-upregulated lncRNAs, 1, 2, 6, 7 and 8 identified by lncRNA profiling along with lincRNA-p21 and DINO were validated after induction of p53 in H1299 cells carrying an inducible wild-type p53 expression system using qPCR. Induced expression of p53 is shown as inset. Mean ± s.e.m.; n=3 independent experiments, two-tailed Student’s t-test. (b) Silencing of lncRNA#6 but not 1, 2, 7 or 8 triggered activation of caspase-3 and cleavage of PARP in HCT116 cells. Data shown represent three independent experiments. (c) Silencing of lncRNA#6 but not 1, 2, 7 or 8 caused reduction in cell viability in HCT116 cells. Mean ± s.e.m.; n=3 independent experiments, two-tailed Student’s t-test. (d) shRNA knockdown efficacy of lncRNA#1, 2, 6, 7 and 8 in HCT116 cells. Mean ± s.e.m.; n=3 independent experiments, two-tailed Student’s t-test. (e) The longest isoform of GUARDIN, RP3-510D11.2-1, was markedly more abundant than the others (RP3-510D11.2-2 and RP3-510D11.2-3) in H1299 cells with or without induction of p53. Mean ± s.e.m.; n=3 independent experiments, two-tailed Student’s t-test. (f) Ectopic expression wild-type p53 but not p53 mutants, p53R175H and p53R273H caused upregulation of GUARDIN in H1299 cells. Mean ± s.e.m.; n=3 independent experiments, two-tailed Student’s t-test. Statistics source data for a and c-f are provided in Supplementary Table 7. Uncropped images of blots for a, b and f in Supplementary Fig. 7.

Supplementary Figure 2 GUARDIN expression in colon cancer cells and tissues and the relation between miR-34a and MIR34AHG expression, copy number loss and p53 status.

(a) Schematic illustration of the genomic location of GUARDIN at chromosome 1p36.22 and its relation to the genes encoding miR-34a, MIR34AHG, and H6PD. (b) GUARDIN expression was positively correlated with the expression of miR-34a and MIR34AHG in laser capture micro-dissected (LCM) cancer cells from fresh surgical samples of 40 colon cancers. Linear regression analysis. (c) GUARDIN expression along with the expression of miR-34a and MIR34AHG were reduced in LCM-dissected colon cancer cells with copy number loss of their genes. n=40 biologically independent samples, two-tailed Student’s t-test. (d) Silencing or overexpression of GUARDIN did not alter the expression of miR-34a and its target Snail as well as MIR34AHG. n=3 independent experiments, two-tailed Student’s t-test. (e) Introduction of anti-miR-23a or knockdown of MIR34AHG did not affect expression of GUARDIN. n=3 independent experiments, two-tailed Student’s t-test. (f) Schematic illustrations of the putative p53-binding region (p53-BR) located 305-329 bp upstream of the GUARDIN transcriptional start site and the pGL3-basic based GUARDIN promoter reporter constructs. (g) In situ hybridization analysis of GUARDIN expression in HCT116 cells and HCT116 cells with GUARDIN knocked down by shRNA that were used as positive and negative biological controls, respectively. Data shown represent three independent experiments. (h) GUARDIN expression in LCM-dissected colon cancer cells from tumours without copy number loss of its gene, similar to the expression of p21 mRNA, was lower in mutant TP53 cases compared with paired LCM pre-neoplastic epithelial cells (hyperplastic polyps (n=5 biologically independent samples) and adenomas (n=7 biologically independent samples)), whereas there was no significant difference in GUARDIN and p21 mRNA expression between LCM-dissected pre-neoplastic and paired normal colon epithelial cells. n=12 biologically independent samples. two-tailed Student’s t-test. Statistics source data for b-e and h are provided in Supplementary Table 7.

Supplementary Figure 3 Rescue of the inhibitory effects of GUARDIN depletion by pan-caspase inhibitor z-VAD-fmk and the identification of complementarity between GUARDIN and miR-23a.

(a) Knockdown of GUARDIN in HCT116 cells using independent shRNAs. Mean ± s.e.m.; n=3 independent experiments, two-tailed Student’s t-test. (b) Treatment with z-VAD-fmk abolished activation of caspase-3 and cleavage of PARP caused by silencing of GUARDIN in HCT116 cells. Data shown represent three independent experiments. (c) Treatment with z-VAD-fmk partially reversed inhibition of cell number expansion caused by silencing of GUARDIN in HCT116, U2OS, and A549 cells. Mean ± s.e.m.; n=3 independent experiments, two-tailed Student’s t-test. (d) Schematic Illustration of base pairing between miR-23a and GUARDIN. (e) Schematic illustration of psiCHECK2-based luciferase reporter constructs containing wild-type GUARDIN (psiCHECK2-GUARDIN-WT) and a mutant reporter construct in which three putative miR-23a binding sites were mutated (psiCHECK2-GUARDIN-mt). Mutated bases are depicted in red. Statistics source data for a and c are provided in Supplementary Table 7. Uncropped images of blots for b are shown in Supplementary Fig. 7.

Supplementary Figure 4 EMSA assays confirm the GUARDIN-BRCA1-BARD1 interaction with domain mapping experiments identifying distinct regions of GUARDIN which bind to BRCA1 and BARD1.

(a) GUARDIN physically associates with BRCA1 and BARD1 in electrophoretic mobility-shift assays. Data shown represent three independent experiments. (b) Schematic illustration of division of GUARDIN into three fragments corresponding to individual exons of the GUARDIN gene (E1, E2 and E3) along with corresponding deletion mutants used. (c) The fragment of GUARDIN corresponding to exon 3 of its gene was required for its association with BARD1. Data shown represent three independent experiments. (d) The fragments of GUARDIN corresponding to exon 1 and 2 of its gene were necessary for its association with BRCA1. Data shown represent three independent experiments. Uncropped images of blots for a, c and d are shown in Supplementary Fig. 7.

Supplementary Figure 5 GUARDIN binding to BRCA1 and BARD1 occurs through their respective amino-terminal domains but independently of embodied RING domains.

(a) Schematic illustration of BRCA1 and the corresponding mutants with deletion of individual fragments. FL: Full length; NT: N Terminus; SCD: serine cluster domain; CT: C Terminus; ΔRING: RING domain deletion. (b) Schematic illustration of BARD1 and the corresponding mutants with deletion of individual fragments. FL: Full length; NT: N Terminus; FAR: Four ankyrin repeats; CT: C Terminus; ΔRING: RING domain deletion. (c) RNA immunoprecipitation assays were performed in HCT116 cells transfected with indicated BRCA1 segment cDNA. The NT of BRCA1 was required for its association with endogenous GUARDIN. Data shown represent three independent experiments. (d) RNA immunoprecipitation assays were performed in HCT116 cells transfected with the indicated BARD1 construct. The NT of BARD1 was required for its association with endogenous GUARDIN. Data shown represent three independent experiments. (e) RNA immunoprecipitation assays were performed in HCT116 cells transfected with indicated BRCA1 segment cDNA. The RING domain encompassed within the NT of BRCA1 was not required for its association with endogenous GUARDIN. Data shown represent three independent experiments. (f) RNA immunoprecipitation assays were performed in HCT116 cells transfected with indicated BARD1 segment cDNA. The RING domain encompassed within the NT of BARD1 was not required for its association with endogenous GUARDIN. Data shown represent three independent experiments. Uncropped images of blots for c-f are shown in Supplementary Fig. 7.

Supplementary Figure 6 GUARDIN depletion induces cell cycle arrest and defective DNA repair through effects on BRCA1, TRF2 and/or miR-23a.

(a) Silencing of GUARDIN caused cell cycle arrest in the G0/G1 phase in HCT116 cells. Mean ± s.e.m.; n=3 independent experiments, two-tailed Student’s t-test. (b) Treatment with z-VAD-fmk did not affect reduction in activation of the HR and NHEJ repair pathways caused by silencing of GUARDIN. Mean ± s.e.m.; n=3 independent experiments, two-tailed Student’s t-test. (c) Neither overexpression of TRF2, introduction of anti-miR-23a, nor overexpression of BRCA1 alone significantly affected inhibition of DDR activation caused by GUARDIN knockdown, whereas co-overexpression of TRF2 and BRCA1 or co-introduction of anti-miR-23a and BRCA1-expressing constructs abolished reduction in DDR activation in cells with GUARDIN knocked down. Mean ± s.e.m.; n=3 independent experiments, two-tailed Student’s t-test. (d) Overexpression of TRF2, introduction of anti-miR-23a, or overexpression of BRCA1 alone did not rescue HCT116 cells from GUARDIN knockdown, whereas the combination of TRF2 overexpression or anti-miR-23a and BRCA1 overexpression abolished inhibition of cell viability caused by GUARDIN knockdown. Mean ± s.e.m.; n=3 independent experiments, two-tailed Student’s t-test. (e) Model depicting the GUARDIN-mediated pathway required to maintain genomic stability. Statistics source data for a-d are provided in Supplementary Table 7.

Supplementary Figure 7 Uncropped images from Western blots.

Uncropped images for Fig.1a-j. Uncropped images for Fig. 3f and Fig. 4g, i-k. Uncropped images for Fig. 5b-j. Uncropped images for Fig. 7b, c, Supplementary Fig. 1a, b, f, and Supplementary Fig. 3b. Uncropped images for Supplementary Fig. 4a, c, d and Supplementary Fig. 5c-f.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hu, W.L., Jin, L., Xu, A. et al. GUARDIN is a p53-responsive long non-coding RNA that is essential for genomic stability. Nat Cell Biol 20, 492–502 (2018). https://doi.org/10.1038/s41556-018-0066-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0066-7

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