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:

ELOF1 is a transcription-coupled DNA repair factor that directs RNA polymerase II ubiquitylation

Abstract

Cells employ transcription-coupled repair (TCR) to eliminate transcription-blocking DNA lesions. DNA damage-induced binding of the TCR-specific repair factor CSB to RNA polymerase II (RNAPII) triggers RNAPII ubiquitylation of a single lysine (K1268) by the CRL4CSA ubiquitin ligase. How CRL4CSA is specifically directed towards K1268 is unknown. Here, we identify ELOF1 as the missing link that facilitates RNAPII ubiquitylation, a key signal for the assembly of downstream repair factors. This function requires its constitutive interaction with RNAPII close to K1268, revealing ELOF1 as a specificity factor that binds and positions CRL4CSA for optimal RNAPII ubiquitylation. Drug–genetic interaction screening also revealed a CSB-independent pathway in which ELOF1 prevents R-loops in active genes and protects cells against DNA replication stress. Our study offers key insights into the molecular mechanisms of TCR and provides a genetic framework of the interplay between transcriptional stress responses and DNA replication.

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: RNAPII-associated ELOF1 is a putative TCR gene.
Fig. 2: ELOF1 is required for efficient transcription elongation.
Fig. 3: ELOF1 is essential for transcription recovery after UV irradiation.
Fig. 4: ELOF1 is essential for repair of transcription-blocking DNA damage.
Fig. 5: ELOF1–RNAPII interaction is required for RNAPII ubiquitylation.
Fig. 6: ELOF1 interacts with the CRL4CSA complex.
Fig. 7: CRISPR screens identify determinants of illudin S sensitivity in the absence of ELOF1 or CSB.
Fig. 8: ELOF1 protects cells against DNA damage during replication.

Similar content being viewed by others

Data availability

Both raw and processed ChIP-seq, Bru-seq, ATAC-seq and RNA-seq data shown in main Figs. 24 and Extended Data Figs. 39 have been deposited into the Gene Expression Omnibus (GEO) under GSE149760. The mass spectrometry proteomics data shown in main Fig. 1 and Extended Data Fig. 2 have been deposited into the ProteomeXchange Consortium via the PRIDE partner repository54 (https://www.ebi.ac.uk/pride/) with the dataset identifier PXD024051. Additionally, publicly available reference datasets of the Hg38 genome and hg19 genome and the known Canonical gene table from the UCSC genome database (https://genome.ucsc.edu/cgi-bin/hgTables; hg38 genome, lifted-over to hg19 when needed) and gene interactions from the GeneMANIA database (https://genemania.org/) have been obtained and used in this manuscript. Published structural information has been obtained for Saccharomyces cerevisiae RAD26 bound to RNAPII https://www.rcsb.org/ PBD: 5VVS) and Komagataella pastoris ELF1 bound to RNAPII (PDB: 5XOG). Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding authors upon reasonable request.

Code availability

Custom code used for the analysis of NGS data was written in R and is available from GitHub (https://git.lumc.nl/dvandenheuvel/van-der-weegen-et-al_elof_ncb2021.git).

References

  1. Brueckner, F., Hennecke, U., Carell, T. & Cramer, P. CPD damage recognition by transcribing RNA polymerase II. Science 315, 859–862 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Nakazawa, Y. et al. Ubiquitination of DNA damage-stalled RNAPII promotes transcription-coupled repair. Cell 180, 1228–1244.e24 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Tufegdžić Vidaković, A. et al. Regulation of the RNAPII pool is integral to the DNA damage response. Cell 180, 1245–1261.e21 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Nakazawa, Y. et al. Mutations in UVSSA cause UV-sensitive syndrome and impair RNA polymerase IIo processing in transcription-coupled nucleotide-excision repair. Nat. Genet. 44, 586–592 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Schwertman, P. et al. UV-sensitive syndrome protein UVSSA recruits USP7 to regulate transcription-coupled repair. Nat. Genet. 44, 598–602 (2012).

    Article  CAS  PubMed  Google Scholar 

  6. van der Weegen, Y. et al. The cooperative action of CSB, CSA, and UVSSA target TFIIH to DNA damage-stalled RNA polymerase II. Nat. Commun. 11, 2104 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Laugel, V. et al. Mutation update for the CSB/ERCC6 and CSA/ERCC8 genes involved in Cockayne syndrome. Hum. Mutat. 31, 113–126 (2010).

    Article  CAS  PubMed  Google Scholar 

  8. Xu, J. et al. Structural basis for the initiation of eukaryotic transcription-coupled DNA repair. Nature 551, 653–657 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Jaspers, N. G. et al. Anti-tumour compounds illudin S and irofulven induce DNA lesions ignored by global repair and exclusively processed by transcription- and replication-coupled repair pathways. DNA Repair (Amst.) 1, 1027–1038 (2002).

    Article  CAS  Google Scholar 

  10. Mair, B. et al. Essential gene profiles for human pluripotent stem cells identify uncharacterized genes and substrate dependencies. Cell Rep. 27, 599–615.e12 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. Yu, X. et al. Up-regulation of human prostaglandin reductase 1 improves the efficacy of hydroxymethylacylfulvene, an antitumor chemotherapeutic agent. J. Pharmacol. Exp. Ther. 343, 426–433 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Adam, S., Polo, S. E. & Almouzni, G. Transcription recovery after DNA damage requires chromatin priming by the H3.3 histone chaperone HIRA. Cell 155, 94–106 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Oksenych, V. et al. Histone methyltransferase DOT1L drives recovery of gene expression after a genotoxic attack. PLoS Genet. 9, e1003611 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Boeing, S. et al. Multiomic analysis of the UV-induced DNA damage response. Cell Rep. 15, 1597–1610 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Daniels, J. P., Kelly, S., Wickstead, B. & Gull, K. Identification of a crenarchaeal orthologue of Elf1: implications for chromatin and transcription in Archaea. Biol. Direct 4, 24 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Ehara, H. et al. Structural insight into nucleosome transcription by RNA polymerase II with elongation factors. Science 363, 744–747 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Ehara, H. et al. Structure of the complete elongation complex of RNA polymerase II with basal factors. Science 357, 921–924 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Prather, D., Krogan, N. J., Emili, A., Greenblatt, J. F. & Winston, F. Identification and characterization of Elf1, a conserved transcription elongation factor in Saccharomyces cerevisiae. Mol. Cell. Biol. 25, 10122–10135 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Veloso, A. et al. Rate of elongation by RNA polymerase II is associated with specific gene features and epigenetic modifications. Genome Res. 24, 896–905 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mayne, L. V. & Lehmann, A. R. Failure of RNA synthesis to recover after UV irradiation: an early defect in cells from individuals with Cockayne’s syndrome and xeroderma pigmentosum. Cancer Res. 42, 1473–1478 (1982).

    CAS  PubMed  Google Scholar 

  21. Perdiz, D. et al. Distribution and repair of bipyrimidine photoproducts in solar UV-irradiated mammalian cells. Possible role of Dewar photoproducts in solar mutagenesis. J. Biol. Chem. 275, 26732–26742 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Epanchintsev, A. et al. Cockayne’s syndrome A and B proteins regulate transcription arrest after genotoxic stress by promoting ATF3 degradation. Mol. Cell 68, 1054–1066.e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Bugai, A. et al. P-TEFb activation by RBM7 shapes a pro-survival transcriptional response to genotoxic stress. Mol. Cell 74, 254–267 e210 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Geijer, I. et al. NCB-M43309B. Nat. Cell Biol. doi:Placeholder (2021).

  25. Zhang, X. et al. Mutations in UVSSA cause UV-sensitive syndrome and destabilize ERCC6 in transcription-coupled DNA repair. Nat. Genet. 44, 593–597 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Fei, J. & Chen, J. KIAA1530 protein is recruited by Cockayne syndrome complementation group protein A (CSA) to participate in transcription-coupled repair (TCR). J. Biol. Chem. 287, 35118–35126 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Olivieri, M. et al. A genetic map of the response to DNA damage in human cells. Cell 182, 481–496 e421 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. de Vivo, A. et al. The OTUD5–UBR5 complex regulates FACT-mediated transcription at damaged chromatin. Nucleic Acids Res. 47, 729–746 (2019).

    Article  PubMed  CAS  Google Scholar 

  29. Panier, S. et al. Tandem protein interaction modules organize the ubiquitin-dependent response to DNA double-strand breaks. Mol. Cell 47, 383–395 (2012).

    Article  CAS  PubMed  Google Scholar 

  30. Benedict, B. et al. WAPL-dependent repair of damaged DNA replication forks underlies oncogene-induced loss of sister chromatid cohesion. Dev. Cell 52, 683–698.e7 (2020).

    Article  CAS  PubMed  Google Scholar 

  31. Doench, J. G. et al. Rational design of highly active sgRNAs for CRISPR–Cas9-mediated gene inactivation. Nat. Biotechnol. 32, 1262–1267 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hart, T. et al. High-Resolution CRISPR screens reveal fitness genes and genotype-specific cancer liabilities. Cell 163, 1515–1526 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Colic, M. et al. Identifying chemogenetic interactions from CRISPR screens with drugZ. Genome Med. 11, 52 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Typas, D. et al. The de-ubiquitylating enzymes USP26 and USP37 regulate homologous recombination by counteracting RAP80. Nucleic Acid Res. 43, 6919–6933 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Rappsilber, J., Mann, M. & Ishihama, Y. Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips. Nat. Protoc. 2, 1896–1906 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Tyanova, S., Temu, T. & Cox, J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat. Protoc. 11, 2301–2319 (2016).

    Article  CAS  PubMed  Google Scholar 

  37. Tyanova, S. et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731–740 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Parra, I. & Windle, B. High resolution visual mapping of stretched DNA by fluorescent hybridization. Nat. Genet. 5, 17–21 (1993).

    Article  CAS  PubMed  Google Scholar 

  39. Garcia-Rubio, M., Barroso, S. I. & Aguilera, A. Detection of DNA–RNA hybrids in vivo. Methods Mol. Biol. 1672, 347–361 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Sanz, L. A. & Chedin, F. High-resolution, strand-specific R-loop mapping via S9.6-based DNA–RNA immunoprecipitation and high-throughput sequencing. Nat. Protoc. 14, 1734–1755 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Andrade-Lima, L. C., Veloso, A., Paulsen, M. T., Menck, C. F. & Ljungman, M. DNA repair and recovery of RNA synthesis following exposure to ultraviolet light are delayed in long genes. Nucleic Acids Res. 43, 2744–2756 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. Preprint at https://arxiv.org/abs/1303.3997 (2013).

  48. Tischler, G. & Leonard, S. biobambam: tools for read pair collation based algorithms on BAM files. Source Code Biol. Med. 9, 13 (2014).

    Article  PubMed Central  Google Scholar 

  49. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. R Development Core Team. R: a Language and Environment for Statistical Computing. (R Foundation for Statistical Computing, 2019).

  52. Benaglia, T., Hunter, C. D. & Young, D. R. D mixtools: an R Package for analyzing finite mixture models. J. Stat. Softw. 32, 1–29 (2009).

    Article  Google Scholar 

  53. Danko, C. G. et al. Signaling pathways differentially affect RNA polymerase II initiation, pausing, and elongation rate in cells. Mol. Cell 50, 212–222 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge A. Kragten, J. Balk, J. Poell, K. Kato, M. Shimada, S. Kloet, M. Paulsen, M. Vukic, D. Warmerdam, A. Ramadhin and L. Daxinger for help during this project. We also thank J. Moffat, K. Chan and A. Tong for sharing the pLCKO-TKOv3 library before publication. We thank the Amsterdam UMC NGS sequencing facilities for support. We thank P. van Veelen and A. de Ru for MS equipment maintenance. This work was funded by a LUMC Research Fellowship, a NWO-ENW-M grant (OCENW.KLEIN.090) and a NWO-VIDI grant (ALW.016.161.320) to M.S.L., a Leiden University Fund (LUF) grant to D.v.d.H. (W18355-2-EM), a KWF/Alpe Young Investigator 10701 grant to J.d.L., a CCA proof-of-concept grant to K.d.L. and R.W., an Amsterdam UMC Innovation Grant (CRISPR Expertise Center, 2019) to R.W., UM1 HG009382 and R01 CA213214 NCI grants to M.L., a KWF Young Investigator grant 11367 to R.G.-P., and an ERC starting grant 310913 to A.C.O.V. J.C.W. was supported by NIH grant HL098316 and is a Howard Hughes Medical Institute (HHMI) Investigator and an American Cancer Society Research Professor. T.E.T.M. was supported by an EMBO Long-term fellowship (ALTF 1316-2016) and a HHMI fellowship of The Jane Coffin Childs Memorial Fund for Medical Research.

Author information

Authors and Affiliations

Authors

Contributions

Y.v.d.W. generated plasmids, U2OS and RPE1-iCas9 single KO and dKO cells, U2OS Flp-In cell-lines, performed clonogenic survivals, co-IP experiments, RRS and DRB-RRS experiments, western blot analyses, in situ PLA experiments, generated samples for pan-RNAPII ChIP-seq, ser2-RNAPII ChIP-seq, ATAC-seq, BruDRB-seq and Bru-seq, generated the figures and wrote the paper. K.d.L. optimized, performed and analysed all CRISPR screens and RNA-seq experiments, performed and analysed drug-sensitivity assays, generated gene-gene interaction network and Venn diagrams, performed γH2AX foci and DNA fibre experiments and helped write the paper. D.v.d.H. performed co-IP experiments, developed tools and analysed pan-RNAPII ChIP-seq, ser2-RNAPII ChIP-seq, ATAC-seq, BruDRB-seq, Bru-seq and helped write the paper. Y.N. performed co-IP experiments and generated TCR-seq libraries. T.E.T.M. generated recombinant xlELOF1, xlCSB and xlCRL4CSA, purified xlRNAPII and performed pull-down and in vitro ubiquitylation assays. J.J.M.v.S. created the RPE1-iCas9 cell line, performed CRISPR screens, performed γH2AX foci and DNA fibre analyses. M.S.M.A. performed and analysed DRIP–qPCR. I.V.N. analysed BruDRB-seq and Bru-seq. D.E.C.B. generated stable cell lines and performed clonogenic survival assays. R.G.-P. analysed the MS with support from A.C.O.V. N.H.M.K. generated U2OS single KO clones, Flp-In cell-lines, performed clonogenic survivals and co-IP experiments. A.P.W. performed the unscheduled DNA synthesis experiments and generated plasmids. K.R. processed and analysed the RNA-seq data, performed gene–gene interaction network analysis, and performed and analysed CRISPR screens. Y.H. analysed ser2-RNAPII ChIP-seq. J.d.L. supervised J.J.M.v.S.; J.C.D. supervised K.R.; J.C.W. supervised T.E.T.M.; S.M.N. supervised M.S.M.A.; M.L. supervised I.V.N. and analysed BruDRB-seq and Bru-seq. T.O. supervised Y.N. and Y.H. and analysed Ser2-RNAPII ChIP-seq. R.M.F.W. supervised K.d.L., co-supervised K.R. and J.J.M.v.S., conceived, coordinated and supervised the project and helped write the paper. M.S.L. supervised Y.v.d.W., D.v.d.H., D.E.C.B., N.H.M.K. and A.P.W., conceived, coordinated and supervised the project, generated all cryo-electron microscopy images and wrote the paper.

Corresponding authors

Correspondence to Rob M. F. Wolthuis or Martijn S. Luijsterburg.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Cell Biology thanks Dong Wang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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

Extended data

Extended Data Fig. 1 Human ELOF1 protects cells against transcription stress.

a, Front-view and side-view of the yeast orthologue of CSB, Saccharomyces cerevisiae RAD26 (purple), bound to RNAPII (grey) (PDB: 5VVS). b, Schematic representation of the CRISPR/Cas9 screen in RPE1-iCas9 cells in the presence of Illudin S (IC60; 25 nM). c, Network analysis of highly significant hits representing genes that promote Illudin S toxicity. Grey lines reflect known protein-protein interactions (Cytoscape, BioGRID). d, Sanger sequencing of the indicated RPE1-iCas9 single ELOF1-KO clones. e, 72 h drug sensitivity assay of indicated RPE1-iCas9 KO clones. The experiment has been performed twice and each symbol represents the median of 6 technical replicates of an independent experiment. UT, untreated f, Clonogenic Illudin S survival of the indicated RPE1-iCas9 cells. The experiment has been performed three times (except for ELOF1-KO 2-12 where the experiment has been done twice) and each symbol represents the mean of 2 technical replicates of an independent experiment. Numerical data are provided in Source data extended data Fig. 1.

Source data

Extended Data Fig. 2 Human ELOF1 and yeast ELF1 show similar RNAPII-binding modes.

a, Sanger sequencing of the indicated U2OS (FRT) ELOF1-KO clone. b, Clonogenic Illudin S survival of U2OS (FRT) WT, CSB-KO, ELOF1-KO, and ELOF1-KO complemented with ELOF1WT-GFP. The experiment has been performed twice and each symbol represents the mean of 2 technical replicates of an independent experiment. c, Alignment of human ELOF1, Xenopus leavis ELOF1, and Saccharomyces cerevisiae ELF1. Conserved residues are indicated in yellow, zinc-finger cysteines in magenta, residues involved in the RPB1 or RPB2 interaction in green. Note that the C-terminus of Saccharomyces cerevisiae ELF1 (83–145) is absent in human ELOF1 and Xenopus leavis ELOF1. (d) Co-immunoprecipitation (IP) of ELOF1WT-GFP and ELOF1S72K/D73K-GFP on the combined soluble and chromatin fraction. Data shown represent 4 independent experiments. (e) Volcano plot depicting the statistical differences between 4 replicates of the MS analysis on ELOF1WT-GFP pull-down in mock treated and UV-irradiated samples. The fold change (log2) is plotted on the x-axis and the significance (t-test -log10(P value)) is plotted on the y-axis. RNAPII subunits are indicated in red, elongation factors are indicated in blue, GFP is indicated in green, and PAF1 subunits are indicated in purple. (f) Clonogenic Illudin S survival of U2OS (FRT) WT, ELOF1-KO, and TY1-tagged ELOF1 rescue cell lines. The experiment has been performed twice and each symbol represents the mean of 2 technical replicates of an independent experiment. Uncropped blots and numerical data are provided in Source data extended data Fig. 2.

Source data

Extended Data Fig. 3 ELOF1 promotes transcription elongation.

a, Calculated speed of RNAPII, quantified by wave-front analyses, in WT (red) or ELOF1-KO (blue) cells 30 min (n=3 experiments) or 60 min (n=2 experiments) after DRB release. Data represents the median per condition (black line) and median within individual replicates (black circles). b, Metaplots of BrU signal (nascent transcription) from 2 kb before the TSS to 2 kb after the TTS in 767 genes between 25–50 kb (upper), 562 genes between 50–100 kb (middle), or 400 genes of >100 kb in WT (red) or ELOF1-KO (blue) cells. Profiles are normalized to 100% at promoter-proximal BrU peaks instead of area under the curve for better comparison of transcription profiles. Profiles are averages of 2 independent replicates. c, Heatmaps of ATAC-seq data around the TSS (−5 kb until +5 kb) of 3,000 genes of 3–100 kb in unirradiated RPE1-iCas9 cells (WT or ELOF1-KO). Numerical data are provided in Source data extended data Fig. 3.

Source data

Extended Data Fig. 4 ELOF1 promotes genome-wide transcription recovery.

a, Quantification of 5-EU levels of the indicated RPE1-iCas9 cells normalized to mock-treated levels for each cell line. Cells were either mock-treated or UV-irradiated (3 h or 24 h; 12 J m-2). The experiment has been performed twice and each black circle represents the median of 2 technical replicates of an independent experiment, >80 cells collected per technical replicate. The black line represents the median of all the cells collected. b, Quantification of 5-EU levels normalized to the baseline level before DRB treatment for each condition. The experiment has been performed twice and each black circle represents the median of 2 technical replicates of an independent experiment, >80 cells collected per technical replicate. The black line represents the median of all the cells collected. c, Western blot analysis of RPE1-iCas9 ELOF1-KO cells complemented with GFP-tagged versions of ELOF1. Data shown represent 3 independent experiments. d, Metaplots of BrU signal (nascent transcription) in 767 genes between 25–50 kb, or in 561 genes between 50–100 kb in WT (upper) or ELOF1-KO (lower) cells after mock treatment (red), or 3 h (blue), 8 h (black), and 24 h (green) after UV irradiation (9 J m-2). Profiles are averages of 2 independent replicates. Uncropped blots and numerical data are provided in Source data extended data Fig. 4.

Source data

Extended Data Fig. 5 Global gene-expression changes in response to UV irradiation.

Volcano plots of RNA-seq in the indicated RPE1-iCas9 cell lines depicting the downregulation (blue) or upregulation (red) of gene expression in response to UV irradiation (24 h; 9 J m-2). The fold change (log2) is plotted on the x-axis and the significance (-log10 P value) is plotted on the y-axis. a, WT, (b) CSB-KO, (c) ELOF1-KO. Only genes indicating at least 2 counts per million (CPM) in at least 33% of samples were included in the analysis. FDR-adjusted P values < 0.05 were considered significant. Two short UV-response genes (ATF3, CDKN1A) are highlighted. d, Box plot depicting the gene length of the 650 most significantly downregulated genes (in blue) or the 650 most significantly upregulated genes (in red). The horizontal line represents the median (center); Upper Bound: gene length scores larger than 75% of all data points; Lower Bound: gene lengths scores shorter than 75% of all data points. Points above and below the box represent the outliers.

Extended Data Fig. 6 Genome-wide redistribution of RNAPII in response to UV irradiation.

a, Heatmaps of pan-RNAPII ChIP-seq data around the TSS of 3,000 genes of 3–100 kb, ranked according to RNAPII signal in mock-treated WT cells. Heatmaps of the same genes are shown after UV irradiation (9 J m-2) in WT or ELOF1-KO cells. b, Averaged metaplots of pan-RNAPII ChIP-seq of 3,000 genes of 3–100 kb from the TSS until the TTS in the indicated RPE1-iCas9 cells after mock-treatment (red) or at 8 h (blue) after UV irradiation (9 J m-2). (c) As in b showing the area around the TTS (−2 kb until +2 kb).

Extended Data Fig. 7 Metaplots of TCR-seq using a Ser2-RNAPII antibody.

a, Individual metaplots (two replicates for each condition) of ser2-RNAPII TCR-seq of 3,000 genes for 3–100 kb from the TSS until the TTS (−5 kb and +5 kb, respectively) in the indicated RPE1-iCas9 cells after mock-treatment or at 1 h, 4 h, or 8 h after UV irradiation (9 J m-2). The coding (non-transcribed) strand in shown in red, while the template (transcribed) strand is shown in blue.

Extended Data Fig. 8 Histogram plots of TCR-seq using a Ser2-RNAPII antibody.

a, Frequency distribution plots of the gene-by-gene ser2-RNAPII strand-specificity index (SSI). SSIs below −0.1 or above 0.1 are presented in red. A unimodal distribution indicates no strand-bias (and thus no DNA damage in the template strand), while a trimodal distribution reflects a strand-bias caused by DNA damage in the template strand.

Extended Data Fig. 9 Validation of TCR-seq with a pan-RNAPII antibody.

a, Individual metaplots (two replicates for each condition) of pan-RNAPII TCR-seq of 3,000 genes of 3–100 kb from the TSS until the TTS (−5 kb and +5 kb, respectively) in the indicated RPE1-iCas9 cells after mock-treatment or at 1 h, 4 h, or 8 h after UV irradiation (9 J m-2). The coding (non-transcribed) strand in shown in red, while the template (transcribed) strand is shown in blue. b, Frequency distribution plots of the gene-by-gene pan-RNAPII strand-specificity index (SSI). SSIs below −0.1 or above 0.1 are presented in red. A unimodal distribution indicates no strand-bias (and thus no DNA damage in the template strand), while a trimodal distribution reflects a strand-bias caused by DNA damage in the template strand.

Extended Data Fig. 10 ELOF1 is not involved in global genome repair.

a-b, Unscheduled DNA synthesis (UDS) in the indicated RPE1-iCas9 cells following local UV irradiation (30 J m-2; 1 h). DNA damage was identified by CPD staining. a, Representative images (Scale bar, 10 µm) and (b) quantification of EdU levels normalized to WT cells. The experiment has been performed twice and each black circle represents the median of 2 technical replicates of an independent experiment, >80 cells collected per technical replicate. The black line represents the median of all the cells collected. c, Endogenous RNAPIIo Co-IP on U2OS (FRT) ELOF1-KO cells complemented with ELOF1WT-GFP after knockdown of CSB (siCSB) or as a control luciferase (siLUC). Data shown represent 2 independent experiments. d, Western blot analysis of CSA protein levels in the indicated RPE1-iCas9 cells after mock treatment, or 7 h, 24 h, and 48 h after UV irradiation (9 J m-2; n=2). e, Quantification of 5-EU levels of the indicated RPE1-iCas9 cells normalized to mock-treated levels for each cell line. Cells were either mock-treated or UV-irradiated (3 h or 24 h; 9 J m-2). 10 µM FT671 (USP7 inhibitor) was added to the indicated cells 24 h prior to UV irradiation. The experiment has been performed three times and each black circle represents the median of 2 technical replicates of an independent experiment, >50 cells collected per technical replicate. The black line represents the median of all the cells collected. f, Western blot analysis of CSA protein levels from e. g, GST pull-down of immobilized recombinant Xenopus laevis (xl) ELOF1 incubated with recombinant xlRAD23B. Data shown represent 2 independent experiments. h, In vitro ubiquitylation of recombinant xlELOF1 and xlCSB with recombinant xlCRL4CSA, E1, E2, ubiquitin, and ATP. In vitro ubiquitylation reactions were stopped at the indicated times. Data shown represent 3 independent experiments. (i) Representative images of staining with TY1 antibodies in U2OS (FRT) WT cells and ELOF1-KO cells complemented with TY1-tagged ELOF1. Data shown represent 2 independent experiments. j, Representative image of staining with TY1 and RBX1 antibodies at 1 h after local UV irradiation (50 J m-2) in U2OS (FRT) ELOF1-KO cells complemented with TY1-tagged ELOF1. Data shown represent 2 independent experiments. k, Results of mining a recent CRISPR screen repository27. Shown are the Z-scores for the indicated sgRNAs (targeting ELOF1 (blue), CSA (orange), CSB (green), UVSSA (black) after exposure to the indicated genotoxic agents. Uncropped blots and numerical data are provided in Source data extended data fig. 10.

Source data

Supplementary information

Reporting Summary

Peer Review Information

Supplementary Tables

Supplementary Table 1: Cell lines. Supplementary. Table 2: sgRNAs. Supplementary Table 3: Plasmids. Supplementary Table 4: Primers. Supplementary Table 5: Antibodies. Supplementary Table 6: Sequence depth. Supplementary Table 7: Read counts (no mismatches allowed) per pLCKO-TKOv3 guide for the four CRISPR screens presented. (A) General information for the screens. (B) Raw read counts of the CRISPR screens.

Source data

Source Data Fig. 1

Unprocessed images of western blots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Unprocessed images of western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 5

Unprocessed images of western blots.

Source Data Fig. 6

Unprocessed images of western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed images of western blots..

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed images of western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed images of western blots.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

van der Weegen, Y., de Lint, K., van den Heuvel, D. et al. ELOF1 is a transcription-coupled DNA repair factor that directs RNA polymerase II ubiquitylation. Nat Cell Biol 23, 595–607 (2021). https://doi.org/10.1038/s41556-021-00688-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-021-00688-9

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing