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.

  • Technical Report
  • Published:

Targeting specific DNA G-quadruplexes with CRISPR-guided G-quadruplex-binding proteins and ligands

Abstract

Despite the demonstrated importance of DNA G-quadruplexes (G4s) in health and disease, technologies to readily manipulate specific G4 folding for functional analysis and therapeutic purposes are lacking. Here we employ G4-stabilizing protein/ligand in conjunction with CRISPR to selectively facilitate single or multiple targeted G4 folding within specific genomic loci. We demonstrate that fusion of nucleolin with a catalytically inactive Cas9 can specifically stabilize G4s in the promoter of oncogene MYC and muscle-associated gene Itga7 as well as telomere G4s, leading to cell proliferation arrest, inhibition of myoblast differentiation and cell senescence, respectively. Furthermore, CRISPR can confer intra-G4 selectivity to G4-binding compounds pyridodicarboxamide and pyridostatin. Compared with traditional G4 ligands, CRISPR-guided biotin-conjugated pyridodicarboxamide enables a more precise investigation into the biological functionality of de novo G4s. Our study provides insights that will enhance understanding of G4 functions and therapeutic interventions.

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: Targeted G4s in specific genome loci using dCas9–nucleolin.
Fig. 2: MycG4 is selectively stabilized by dCas9–nucleolin.
Fig. 3: Targeting stabilization of telomere G4s disrupts telomere homeostasis via dCas9–nucleolin.
Fig. 4: Itga7 promoter G4 stabilized by dCas9–nucleolin inhibits muscle cell differentiation.
Fig. 5: dCas9–mSA/Bio–PDC stabilizes G4 in targeted genome loci.
Fig. 6: Selectively stabilizing de novo G4 by dCas9–SunTag–mSA-guided Bio–PDC.
Fig. 7: dCas9–SunTag–mSA/Bio–PDC selectively stabilizes NEAT1 and MALAT1 G4s.

Similar content being viewed by others

Data availability

The ATAC-seq and RNA-seq data generated from this study have been deposited in the Gene Expression Omnibus (GSE255625). The following previously published datasets were used: hg38 (https://www.ensembl.org/Homo_sapiens/Info/Index), PDS and PhenDC3 Chem-map (GSE209713)23, BG4 CUT&Tag (GSE181373)22, ATAC-seq (GSE162299)24. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Davis, J. T. G-Quartets 40 years later: from 5′-GMP to molecular biology and supramolecular chemistry. Angew. Chem. Int. Ed. Engl. 43, 668–698 (2004).

    Article  CAS  PubMed  Google Scholar 

  2. Varshney, D., Spiegel, J., Zyner, K., Tannahill, D. & Balasubramanian, S. The regulation and functions of DNA and RNA G-quadruplexes. Nat. Rev. Mol. Cell Biol. 21, 459–474 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kosiol, N., Juranek, S., Brossart, P., Heine, A. & Paeschke, K. G-Quadruplexes: a promising target for cancer therapy. Mol. Cancer 20, 40 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Qin, H., Zhao, C., Sun, Y., Ren, J. & Qu, X. Metallo-supramolecular complexes enantioselectively eradicate cancer stem cells in vivo. J. Am. Chem. Soc. 139, 16201–16209 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Meier-Stephenson, V. G4-Quadruplex-binding proteins: review and insights into selectivity. Biophys. Rev. 14, 635–654 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lejault, P., Mitteaux, J., Sperti, F. R. & Monchaud, D. How to untie G-quadruplex knots and why? Cell Chem. Biol. 28, 436–455 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. Santos, T., Salgado, G. F., Cabrita, E. J. & Cruz, C. Nucleolin: a binding partner of G-quadruplex structures. Trends Cell Biol. 32, 561–564 (2022).

    Article  CAS  PubMed  Google Scholar 

  8. Roychoudhury, S. et al. Endogenous oxidized DNA bases and APE1 regulate the formation of G-quadruplex structures in the genome. Proc. Natl Acad. Sci. USA 117, 11409–11420 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mendoza, O., Bourdoncle, A., Boule, J. B., Brosh, R. M. Jr. & Mergny, J. L. G-Quadruplexes and helicases. Nucleic Acids Res. 44, 1989–2006 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chen, M. C. et al. Structural basis of G-quadruplex unfolding by the DEAH/RHA helicase DHX36. Nature 558, 465–469 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Rodriguez, R. et al. A novel small molecule that alters shelterin integrity and triggers a DNA-damage response at telomeres. J. Am. Chem. Soc. 130, 15758–15759 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Liu, L. Y., Ma, T. Z., Zeng, Y. L., Liu, W. & Mao, Z. W. Structural basis of pyridostatin and its derivatives specifically binding to G-quadruplexes. J. Am. Chem. Soc. 144, 11878–11887 (2022).

    Article  CAS  PubMed  Google Scholar 

  13. Verga, D., Hamon, F., Poyer, F., Bombard, S. & Teulade-Fichou, M. P. Photo-cross-linking probes for trapping G-quadruplex DNA. Angew. Chem. Int. Ed. Engl. 53, 994–998 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Masson, T. et al. BrdU immuno-tagged G-quadruplex ligands: a new ligand-guided immunofluorescence approach for tracking G-quadruplexes in cells. Nucleic Acids Res. 49, 12644–12660 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. De Cian, A., Delemos, E., Mergny, J. L., Teulade-Fichou, M. P. & Monchaud, D. Highly efficient G-quadruplex recognition by bisquinolinium compounds. J. Am. Chem. Soc. 129, 1856–1857 (2007).

    Article  PubMed  Google Scholar 

  16. Lemarteleur, T. et al. Stabilization of the c-myc gene promoter quadruplex by specific ligands’ inhibitors of telomerase. Biochem. Biophys. Res. Commun. 323, 802–808 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517, 583–588 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Chiarella, A. M. et al. Dose-dependent activation of gene expression is achieved using CRISPR and small molecules that recruit endogenous chromatin machinery. Nat. Biotechnol. 38, 50–55 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Qin, G., Yang, J., Zhao, C., Ren, J. & Qu, X. Manipulating complex chromatin folding via CRISPR-guided bioorthogonal chemistry. Proc. Natl Acad. Sci. USA 119, e2204725119 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wang, C. et al. dCas9-based gene editing for cleavage-free genomic knock-in of long sequences. Nat. Cell Biol. 24, 268–278 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Siddiqui-Jain, A., Grand, C. L., Bearss, D. J. & Hurley, L. H. Direct evidence for a G-quadruplex in a promoter region and its targeting with a small molecule to repress c-MYC transcription. Proc. Natl Acad. Sci. USA 99, 11593–11598 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hui, W. W. I., Simeone, A., Zyner, K. G., Tannahill, D. & Balasubramanian, S. Single-cell mapping of DNA G-quadruplex structures in human cancer cells. Sci. Rep. 11, 23641 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yu, Z. et al. Chem-map profiles drug binding to chromatin in cells. Nat. Biotechnol. 41, 1265–1271 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Shen, J. et al. Promoter G-quadruplex folding precedes transcription and is controlled by chromatin. Genome Biol. 22, 143 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Hansel-Hertsch, R. et al. G-Quadruplex structures mark human regulatory chromatin. Nat. Genet. 48, 1267–1272 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Bian, W. X. et al. Binding of cellular nucleolin with the viral core RNA G-quadruplex structure suppresses HCV replication. Nucleic Acids Res. 47, 56–68 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Li, C. et al. Ligand-induced native G-quadruplex stabilization impairs transcription initiation. Genome Res. 31, 1546–1560 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Bochman, M. L., Paeschke, K. & Zakian, V. A. DNA secondary structures: stability and function of G-quadruplex structures. Nat. Rev. Genet. 13, 770–780 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lago, S., Tosoni, E., Nadai, M., Palumbo, M. & Richter, S. N. The cellular protein nucleolin preferentially binds long-looped G-quadruplex nucleic acids. Biochim. Biophys. Acta Gen. Subj. 1861, 1371–1381 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Weintraub, H. et al. The myoD gene family: nodal point during specification of the muscle cell lineage. Science 251, 761–766 (1991).

    Article  CAS  PubMed  Google Scholar 

  31. Shklover, J., Weisman-Shomer, P., Yafe, A. & Fry, M. Quadruplex structures of muscle gene promoter sequences enhance in vivo MyoD-dependent gene expression. Nucleic Acids Res. 38, 2369–2377 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yafe, A., Etzioni, S., Weisman-Shomer, P. & Fry, M. Formation and properties of hairpin and tetraplex structures of guanine-rich regulatory sequences of muscle-specific genes. Nucleic Acids Res. 33, 2887–2900 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. McClure, M. J. et al. Role of integrin α7β1 signaling in myoblast differentiation on aligned polydioxanone scaffolds. Acta Biomater. 39, 44–54 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Qin, G. et al. Unlocking G-quadruplexes as targets and tools against COVID-19. Chin. J. Chem. 41, 560–568 (2023).

  35. Abiri, A. et al. Unlocking G-quadruplexes as antiviral targets. Pharmacol. Rev. 73, 897–923 (2021).

    Article  CAS  PubMed  Google Scholar 

  36. Neidle, S. Quadruplex nucleic acids as novel therapeutic targets. J. Med. Chem. 59, 5987–6011 (2016).

    Article  CAS  PubMed  Google Scholar 

  37. Renaud de la Faverie, A. et al. Nucleic acids targeted to drugs: SELEX against a quadruplex ligand. Biochimie 93, 1357–1367 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Gu, B., Posfai, E. & Rossant, J. Efficient generation of targeted large insertions by microinjection into two-cell-stage mouse embryos. Nat. Biotechnol. 36, 632–637 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Di Antonio, M. et al. Single-molecule visualization of DNA G-quadruplex formation in live cells. Nat. Chem. 12, 832–837 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Tanenbaum, M. E., Gilbert, L. A., Qi, L. S., Weissman, J. S. & Vale, R. D. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159, 635–646 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Morita, S. et al. Targeted DNA demethylation in vivo using dCas9-peptide repeat and scFv-TET1 catalytic domain fusions. Nat. Biotechnol. 34, 1060–1065 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Burger, A. M. et al. The G-quadruplex-interactive molecule BRACO-19 inhibits tumor growth, consistent with telomere targeting and interference with telomerase function. Cancer Res. 65, 1489–1496 (2005).

    Article  CAS  PubMed  Google Scholar 

  43. Kim, M. Y., Gleason-Guzman, M., Izbicka, E., Nishioka, D. & Hurley, L. H. The different biological effects of telomestatin and TMPyP4 can be attributed to their selectivity for interaction with intramolecular or intermolecular G-quadruplex structures. Cancer Res. 63, 3247–3256 (2003).

    CAS  PubMed  Google Scholar 

  44. Hu, M. H., Lin, J. H. & Huang, Q. Discovery of a fluorescent, long chain-bridged bispurine that selectively targets the c-MYC G-quadruplex. Bioorg. Chem. 122, 105750 (2022).

    Article  CAS  PubMed  Google Scholar 

  45. Chambers, V. S. et al. High-throughput sequencing of DNA G-quadruplex structures in the human genome. Nat. Biotechnol. 33, 877–881 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Yang, W. S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Venugopal, R. & Jaiswal, A. K. Nrf2 and Nrf1 in association with Jun proteins regulate antioxidant response element-mediated expression and coordinated induction of genes encoding detoxifying enzymes. Oncogene 17, 3145–3156 (1998).

    Article  CAS  PubMed  Google Scholar 

  48. Maxwell, P. H. et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399, 271–275 (1999).

    Article  CAS  PubMed  Google Scholar 

  49. Chen, J. et al. Expression of Q227L-Gαs in MCF-7 human breast cancer cells inhibits tumorigenesis. Proc. Natl Acad. Sci. USA 95, 2648–2652 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. West, J. A. et al. The long noncoding RNAs NEAT1 and MALAT1 bind active chromatin sites. Mol. cell 55, 791–802 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Robinson, J., Raguseo, F., Nuccio, S. P., Liano, D. & Di Antonio, M. DNA G-quadruplex structures: more than simple roadblocks to transcription? Nucleic Acids Res. 49, 8419–8431 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Rodriguez, R. et al. Small-molecule-induced DNA damage identifies alternative DNA structures in human genes. Nat. Chem. Biol. 8, 301–310 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Filippakopoulos, P. et al. Selective inhibition of BET bromodomains. Nature 468, 1067–1073 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Felsenstein, K. M. et al. Small molecule microarrays enable the identification of a selective, quadruplex-binding inhibitor of MYC expression. ACS Chem. Biol. 11, 139–148 (2016).

    Article  CAS  PubMed  Google Scholar 

  55. Calabrese, D. R. et al. Chemical and structural studies provide a mechanistic basis for recognition of the MYC G-quadruplex. Nat. Commun. 9, 4229 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Esain-Garcia, I. et al. G-Quadruplex DNA structure is a positive regulator of MYC transcription. Proc. Natl Acad. Sci. USA 121, e2320240121 (2024).

  57. Umar, M. I., Chan, C. Y. & Kwok, C. K. Development of RNA G-quadruplex (rG4)-targeting l-RNA aptamers by rG4-SELEX. Nat. Protoc. 17, 1385–1414 (2022).

    Article  CAS  PubMed  Google Scholar 

  58. Ellenbroek, B. D., Kahler, J. P., Evers, S. R. & Pomplun, S. J. Synthetic peptides: promising modalities for the targeting of disease-related nucleic acids. Angew. Chem. Int Ed. 63, e202401704 (2024).

    Article  CAS  Google Scholar 

  59. Galli, S. et al. DNA G-quadruplex recognition in vitro and in live cells by a structure-specific nanobody. J. Am. Chem. Soc. 144, 23096–23103 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Yang, L. Z. et al. Dynamic imaging of RNA in living cells by CRISPR-Cas13 systems. Mol. Cell 76, 981–997.e987 (2019).

    Article  CAS  PubMed  Google Scholar 

  61. Qin, G. et al. A step by step protocol for stabilizing specific DNA G-quadruplexes by CRISPR guided G-quadruplex binding proteins and ligands. Protoc. Exch. https://doi.org/10.21203/rs.3.pex-2648/v1 (2024).

    Article  Google Scholar 

Download references

Acknowledgements

We thank S. Balasubramanian and M. Farrow for their help on the synthesis. Financial support was provided by the National Key R&D Program of China (2019YFA0709202 to X.Q.; 2021YFF1200700 to J.R.; 2022YFA1205804 to C.Z.), the National Natural Science Foundation of China (91856205 and 21820102009 to X.Q.; 22237006 to J.R.; 22107098 to G.Q.; 22122704 to C.Z.) and Jilin Innovation Project (2023DJ02 to X.Q.).

Author information

Authors and Affiliations

Authors

Contributions

X.Q. was the principal investigator who conceived and designed the study, obtained financial supports and approved the final version of the paper. J.R. was the other principal investigator who conceived the study. G.Q. and Z.L. performed the experiments; G.Q., Z.L., J.Y., X.L. and C.Z. conducted data management; G.Q. performed the statistical analyses, interpreted the results and drafted the paper. All the authors read and approved the final version of the paper.

Corresponding author

Correspondence to Xiaogang Qu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks David Monchaud and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Targeted G4s in specific oncogene promoter region using dCas9–nucleolin.

a, The proposed model of MYC transcriptional regulation by MycG4 and nucleolin. b, Binding curves of nucleolin with MycG4 and a hairpin as the example of non-G4 structure determined by an adapted enzyme linked immunosorbent assay (ELISA). c, The human MYC locus. MycG4 site is indicated by a yellow box. The locations of the targets (3–6) for sgRNAs (sgR-3-6) are indicated by red bars. d, Binding curves of nucleolin with KRAS G4 and VEGFA G4 determined by ELISA. e, The human KRAS locus. KRAS G4 site is indicated by a yellow box. The locations of the targets (1–4) for sgRNAs (sgR-KRAS-1-4) are indicated by red bars. f, Inhibition of EGFP protein expression in HEK293T cells by sgR-KRAS-1-4, each expressed with or without dCas9–nucleolin. EGFP reporter gene was downstream of the KRAS promoter with or without KRAS G4. g, The human VEGFA locus. VEGFA G4 site is indicated by a yellow box. The locations of the targets (1–3) for sgRNAs (sgR-VEGFA-1-4) are indicated by red bars. h, Inhibition of EGFP protein expression in HEK293T cells by sgR-VEGFA-1-4, each expressed with or without dCas9–nucleolin. EGFP reporter gene was downstream of the VEGFA promoter with or without VEGFA G4. i, The mRNA levels of EGFP with the MycG4-WT or MycG4-Mut MYC promoter in the HEK293T cells transfected with dCas9–nucleolin and the indicated sgRNAs. j, The protein levels of dCas9 and dCas9–nucleolin in the HEK293T cells transfected with dCas9 or dCas9–nucleolin, detected by western blotting assays. Data are shown as mean ± SEM of three independent experiments, and P values were determined using two-tailed Student’s t-test. Source numerical data and unprocessed blots are available in Source data.

Source data

Extended Data Fig. 2 dCas9–nucleolin targeting MycG4 inhibits MYC transcription and induces cell growth arrest.

a, The dCas9–nucleolin decreases the levels of both unprocessed and mature MYC RNA transcripts. Unprocessed MYC RNA transcript was measured using the intron specific primers. Mature MYC mRNA levels were measured using a pair of primer flanking intron 1. All RNA transcript levels were measured at desired time points after transfecting dCas9–nucleolin and sgR-5. b, The nucleolin/G4 complexes were captured by BG4 antibody through Anti-Flag Magnetic Beads, detected by western blotting assays. c, The proposed model of MYC transcriptional regulation by G4s, NM23H2, SP1, CNBP, hnRNP K and nucleolin. d-g, The occupancy of SP1, CNBP, hnRNP K and nucleolin in the promoter region of MYC was measured by ChIP in HeLa cells transfected with dCas9–nucleolin and sgR-5. h-k, The occupancy of SP1, CNBP, hnRNP K and nucleolin in the promoter region of MYC was measured by ChIP in HEK293T cells transfected with dCas9–nucleolin and sgR-5. l, Cells growth assays on HEK293T, HeLa, MCF-7 and K562 cells expressing dCas9–nucleolin and sgR-5. m, Genome browser tracks overlaid at HIF1A and NRAS genes. The dark green box highlights the G4 sites. Data are shown as mean ± SEM of three independent experiments, and P values were determined using two-tailed Student’s t-test. Source numerical data and unprocessed blots are available in Source data.

Source data

Extended Data Fig. 3 Targeting telomere and Itga7 G4s by using dCas9–nucleolin.

a, Binding curves of nucleolin with telomere G4 determined by ELISA. b, Left: Immunofluorescence (IF) showing BG4 foci (red) in the nuclei of HeLa cells transfected with dCas9–nucleolin, sgR-Telo or sgR-Ctrl (n = 24). Scale bars, 5 μm. Right: The number of BG4 foci per nucleus. c, Left: IF showing BG4 foci (red) in the nuclei of HeLa cells transfected with dCas9 or dCas9–nucleolin (n = 30). Scale bars, 5 μm. Right: The number of BG4 foci per nucleus. d, The telomere length of the HeLa cells transfected with dCas9–nucleolin/sgR-Telo was detected by qRT-PCR. e, Binding curves of nucleolin with Itga7 G4 determined by ELISA. Data are shown as mean ± SEM of three independent experiments, and P values were determined using two-tailed Student’s t-test. Source numerical data are available in Source data.

Source data

Extended Data Fig. 4 Targeting G4 in specific genome loci by dCas9-mSA/Bio-PDC complexes.

a, The cytotoxicity of Bio-PDC in HEK293T, MCF-7, A375 and K562 cells was detected by CCK8 assays. b, Binding curves of Bio-PDC with a hairpin as the example of non-G4 structure, MycG4, KRAS G4, c-kit1 G4 and VEGFA G4 determined by ELISA. c,d, The FAM labelled G4s was captured by Bio-PDC through Magnetic streptavidin-coated beads (c), and the FAM signals were detected (d). e, The mRNA expression of MYC, KARS, KIT and VEGFA in HeLa cell treated with the indicated concentration Bio-PDC for 48 hours. Data are shown as mean ± SEM of three independent experiments, and P values were determined using two-tailed Student’s t-test. Source numerical data are available in Source data.

Source data

Extended Data Fig. 5 dCas9-mSA/Bio-PDC complexes guided by sgRNAs targeting MycG4 selectively inhibit MYC expression.

a, ChIP-qRT-PCR assays of dCas9-mSA constructs demonstrates correct localization to their intended genomic loci. b, The effects of Bio-PDC on EGFP expression in EGFP reporter expressed HEK293T cells. EGFP reporter gene was downstream of the MYC promoter with or without MycG4. c-e, The effects of Bio-PDC on EGFP expression in EGFP reporter expressed HEK293T cells transfecting with dCas9-mSA and sgR-3 (c), sg-4 (d) or sgR-6 (e). EGFP reporter gene was downstream of the MYC promoter with or without MycG4. f, The EGFP reporter expressed HEK293T cells pre-transfected with dCas9 or 4 MYC-targeting sgRNAs were treated with 500 nM Bio-PDC for 48 hours, and then the EGFP expression was detected. g, The occupancy of BG4 in the promoter region of MYC was measured by ChIP in HeLa cells transfected without or with dCas9-mSA and 4 MYC-targeting sgRNAs in response to 500 nM Bio-PDC for 48 hours. h, The mRNA expression of MYC, KARS, KIT, VEGFA and HRAS in HeLa cell transfected without or with dCas9-mSA and 4 MYC-targeting sgRNAs in response to 500 nM Bio-PDC for 48 hours. Data are shown as mean ± SEM of three independent experiments, and P values were determined using two-tailed Student’s t-test. Source numerical data are available in Source data.

Source data

Extended Data Fig. 6 Targeting G4 in specific genome loci by dCas9-SunTag-mSA/Bio-PDC complexes.

a, ChIP-qRT-PCR assays of dCas9-SunTag-mSA constructs demonstrates correct localization to their intended genomic loci. b, The mRNA expression of MYC in HeLa cells expressed with dCas9-mSA/4 sgRNAs or dCas9-SunTag-mSA/sgR-5 in response to 500 nM Bio-PDC for 48 hours. c, The occupancy of BG4 in the promoter region of MYC was measured by ChIP in HeLa cells expressed with dCas9-mSA/4 sgRNAs or dCas9-SunTag-mSA/sgR-5 in response to 500 nM Bio-PDC for 48 hours. d, The mRNA levels of VEGFA and HRAS in the HeLa cells expressed dCas9-SunTag-mSA/sgR-5 with the indicated treatment of PDC or Bio-PDC. e, The fold change of G4-driven oncogenes expression in the HeLa cells expressed dCas9-SunTag-mSA/sgR-5 in response to 500 nM Bio-PDC, 5 μM BRACO-19, 10 μM TMPyP4 and 10 μM DA3 (a G4-stabilized ligand with preference for MycG4) for 48 hours. Data are shown as mean ± SEM of three independent experiments, and P values were determined using two-tailed Student’s t-test. Source numerical data are available in Source data.

Source data

Extended Data Fig. 7 The formation of de novo G4s in vitro.

a, Genome browser tracks overlaid at GPX4, NFE2L2, VHL, MAPK1, NEAT1, MALAT1 genes. The orange box highlights the G4 sites, and the putative G4-forming sequences (PQSs) were shown. b, Circular dichroism spectroscopy of GPX4, NFE2L2, VHL, MAPK1, NEAT1, MALAT1 G4s and their mutants. c, Fluorescence turn-on assays of N-methyl mesophorphyrin IX (NMM) in the absence or presence of GPX4, NFE2L2, VHL, MAPK1, NEAT1, MALAT1 G4s and their mutants under indicated conditions. NMM, a well-known fluorescent G4-specific targeting compound. Source numerical data are available in Source data.

Source data

Extended Data Fig. 8 dCas9-SunTag-mSA guided Bio-PDC selectively stabilizes de novo G4.

a, Binding curves of Bio-PDC with GPX4, NFE2L2, VHL, MAPK1, NEAT1 and MALAT1 G4s determined by ELISA. b, The occupancy of BG4 in the promoter region of GPX4, NFE2L2, VHL, MAPK1, NEAT1 and MALAT1 was measured by ChIP in HeLa cells expressed with dCas9-SunTag-mS and sgRNAs targeting GPX4, NFE2L2, VHL, MAPK1, NEAT1 and MALAT1 G4s in response to 500 nM Bio-PDC for 48 hours. c, The Venn diagram shows the overlap of genes containing G4 motifs at the promoter region and differentially expressed genes. d, The Venn diagram shows the overlap of differentially expressed genes and the genes with differential ATAC signal in promoter. e, Genome browser tracks overlaid at KRAS and VEGFA genes. The brown box highlights the G4 sites. f, The occupancy of BG4 in the promoter region of PLEC and SEMA4B was measured by ChIP in HeLa cells expressed with dCas9-SunTag-mSA and two sgRNAs targeting NEAT1 and MALAT1 promoter G4s (sgR-lncRNAs) in response to 500 nM Bio-PDC for 48 hours. Data are shown as mean ± SEM of three independent experiments, and P values were determined using two-tailed Student’s t-test. Source numerical data are available in Source data.

Source data

Extended Data Fig. 9 dCas9-SunTag-mSA/Bio-PDS complexes can stabilizes G4 in targeted genome loci.

a, Chemical structures of Bio-PDS. b,c The FAM labelled G4s was captured by Bio-PDS through Anti-Flag Magnetic Beads (b), and the FAM signals were detected (c). d, Inhibition of EGFP protein expression in the HEK293T cells expressed with dCas9-SunTag-mSA and sgR-5 by Bio-PDS concentration ranging from 50 to 1,000 nM treatment for 48 hours. e, The mRNA expression of MYC in HeLa cells expressed with dCas9-SunTag-mSA/sgR-5 in response to 200 nM Bio-PDS for 48 hours. f, The occupancy of BG4 in the promoter region of MYC was measured by ChIP in HeLa cells expressed with dCas9-SunTag-mSA/sgR-5 in response to 200 nM Bio-PDS for 48 hours. g, The mRNA levels of MYC and SRC in the HeLa cells expressed dCas9-SunTag-mSA/sgR-5 with the indicated treatment of PDS or Bio-PDS. h, Volcano plot (left) and heatmap (right) showing gene expression changes in HeLa cells transfected with dCas9-SunTag-mSA and sgR-lncRNAs in response to 200 nM Bio-PDS treatment for 48 hours. i, MA plots showing fold change of the expression of the genes with G4s in promoter. Data are shown as mean ± SEM of three independent experiments, and P values were determined using two-tailed Student’s t-test. Source numerical data are available in Source data.

Source data

Extended Data Fig. 10 dCas9-SunTag-mSA/Bio-JQ1 complexes can activate gene transcription in targeted genome loci.

a, Chemical structures of Bio-JQ1. b, The mRNA expression of MyoD and CXCR4 in HEK293T cells transfected with dCas9-SunTag-mSA and sgR-MyoD or sgR-CXCR4 in response to 200 nM Bio-JQ1 treatment for 48 hours. Data are shown as mean ± SEM of three independent experiments, and P values were determined using two-tailed Student’s t-test. Source numerical data are available in Source data.

Source data

Supplementary information

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qin, G., Liu, Z., Yang, J. et al. Targeting specific DNA G-quadruplexes with CRISPR-guided G-quadruplex-binding proteins and ligands. Nat Cell Biol 26, 1212–1224 (2024). https://doi.org/10.1038/s41556-024-01448-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-024-01448-1

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