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:

Stabilization of AFF1 by PARylation ensures transcriptional restart after DNA damage

Abstract

Precise control of transcription is essential for cell survival under stress conditions, including DNA damage. While mechanisms of DNA damage-induced transcriptional silencing are well characterized, how transcription resumes remains less understood. Here we identify a new role for poly(ADP-ribose) polymerase 1 (PARP1) in transcriptional restart during the DNA damage response (DDR) through a mechanism termed poly(ADP-ribose)-mediated stabilization (PARSTA) of AFF1. Upon DNA damage, PARP1 binds to and PARylates AFF1 in a region targeted by the E3 ligase Siah1, preventing AFF1 ubiquitination and promoting its stability. This stabilization supports efficient transcriptional recovery after DNA damage. Notably, cells resistant to genotoxic stress exhibit elevated PARP1 activity and AFF1 levels, while AFF1 depletion impairs DNA repair and survival. Together, these findings expand PARP1’s role to the transcriptional recovery phase in DDR and suggest that targeting the PARSTA pathway may offer therapeutic potential in diseases characterized by hyperactive PARP1 and elevated levels of AFF1.

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: AFF1 responds to DNA damage in a PARP1-dependent manner.
Fig. 2: DNA damage induces PARP1–AFF1 interaction.
Fig. 3: AFF1 is PARylated by PARP1 following DNA damage.
Fig. 4: PARylation of AFF1 enhances protein stability by disrupting its interaction with Siah1.
Fig. 5: AFF1 is required for transcriptional recovery after DNA damage.
Fig. 6: The PARP1–AFF1 axis is essential for cell survival in response to DNA damage.

Similar content being viewed by others

Data availability

All data that were generated and/or analyzed in this study are included in the article and the Supplementary Information. The raw sequencing data generated in this study were deposited to the Genome Sequence Archive of the National Genomics Data Center (HRA010143). MS data were uploaded to ProteomeXchange (PXD049977). Source data are provided with this paper.

References

  1. Chen, F. X., Smith, E. R. & Shilatifard, A. Born to run: control of transcription elongation by RNA polymerase II. Nat. Rev. Mol. Cell Biol. 19, 464–478 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Adelman, K. & Lis, J. T. Promoter-proximal pausing of RNA polymerase II: emerging roles in metazoans. Nat. Rev. Genet 13, 720–731 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Zhou, Q., Li, T. & Price, D. H. RNA polymerase II elongation control. Annu Rev. Biochem 81, 119–143 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Vos, S. M., Farnung, L., Urlaub, H. & Cramer, P. Structure of paused transcription complex Pol II–DSIF–NELF. Nature 560, 601–606 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. He, N. & Zhou, Q. New insights into the control of HIV-1 transcription: when Tat meets the 7SK snRNP and super elongation complex (SEC). J. Neuroimmune Pharm. 6, 260–268 (2011).

    Article  Google Scholar 

  6. Schulze-Gahmen, U., Lu, H., Zhou, Q. & Alber, T. AFF4 binding to Tat–P-TEFb indirectly stimulates TAR recognition of super elongation complexes at the HIV promoter. eLife 3, e02375 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Ott, M., Geyer, M. & Zhou, Q. The control of HIV transcription: keeping RNA polymerase II on track. Cell Host Microbe 10, 426–435 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Luo, Z., Lin, C. & Shilatifard, A. The super elongation complex (SEC) family in transcriptional control. Nat. Rev. Mol. Cell Biol. 13, 543–547 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Aoi, Y. & Shilatifard, A. Transcriptional elongation control in developmental gene expression, aging, and disease. Mol. Cell 83, 3972–3999 (2023).

    Article  CAS  PubMed  Google Scholar 

  10. Lu, H. et al. AFF1 is a ubiquitous P-TEFb partner to enable Tat extraction of P-TEFb from 7SK snRNP and formation of SECs for HIV transactivation. Proc. Natl Acad. Sci. USA 111, E15–E24 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Chou, S. et al. HIV-1 Tat recruits transcription elongation factors dispersed along a flexible AFF4 scaffold. Proc. Natl Acad. Sci. USA 110, E123–E131 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Liu, K. et al. The super elongation complex drives neural stem cell fate commitment. Dev. Cell 40, 537–551 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Lin, C. et al. Dynamic transcriptional events in embryonic stem cells mediated by the super elongation complex (SEC). Genes Dev. 25, 1486–1498 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wang, D. et al. The super elongation complex drives transcriptional addiction in MYCN-amplified neuroblastoma. Sci. Adv. 9, eadf0005 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Jin, J. et al. PRMT2 promotes HIV-1 latency by preventing nucleolar exit and phase separation of Tat into the super elongation complex. Nat. Commun. 14, 7274 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ui, A., Nagaura, Y. & Yasui, A. Transcriptional elongation factor ENL phosphorylated by ATM recruits Polycomb and switches off transcription for DSB repair. Mol. Cell 58, 468–482 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Mourgues, S. et al. ELL, a novel TFIIH partner, is involved in transcription restart after DNA repair. Proc. Natl Acad. Sci. USA 110, 17927–17932 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kumari, N., Hassan, M. A., Lu, X., Roeder, R. G. & Biswas, D. AFF1 acetylation by p300 temporally inhibits transcription during genotoxic stress response. Proc. Natl Acad. Sci. USA 116, 22140–22151 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Tubbs, A. & Nussenzweig, A. Endogenous DNA damage as a source of genomic instability in cancer. Cell 168, 644–656 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lu, H., Yang, M. & Zhou, Q. Reprogramming transcription after DNA damage: recognition, response, repair, and restart. Trends Cell Biol. 33, 682–694 (2023).

    Article  CAS  PubMed  Google Scholar 

  22. Milano, L., Gautam, A. & Caldecott, K. W. DNA damage and transcription stress. Mol. Cell 84, 70–79 (2024).

    Article  CAS  PubMed  Google Scholar 

  23. Sang, C. C. et al. PARP1 condensates differentially partition DNA repair proteins and enhance DNA ligation. EMBO Rep. https://doi.org/10.1038/s44319-024-00285-5 (2024).

  24. Gupte, R., Liu, Z. & Kraus, W. L. PARPs and ADP-ribosylation: recent advances linking molecular functions to biological outcomes. Genes Dev. 31, 101–126 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chappidi, N., et al. PARP1–DNA co-condensation drives DNA repair site assembly to prevent disjunction of broken DNA ends. Cell 187, 945–961 (2024).

    Article  CAS  PubMed  Google Scholar 

  26. Fu, H. et al. Poly(ADP-ribosylation) of P-TEFb by PARP1 disrupts phase separation to inhibit global transcription after DNA damage. Nat. Cell Biol. 24, 513–525 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Adamowicz, M. et al. XRCC1 protects transcription from toxic PARP1 activity during DNA base excision repair. Nat. Cell Biol. 23, 1287–1298 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Khanam, T. et al. CDKL5 kinase controls transcription-coupled responses to DNA damage. EMBO J. 40, e108271 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Awwad, S. W., Abu-Zhayia, E. R., Guttmann-Raviv, N. & Ayoub, N. NELF-E is recruited to DNA double-strand break sites to promote transcriptional repression and repair. EMBO Rep. 18, 745–764 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Li, D. F. et al. Tandem mass tag (TMT)-based proteomic analysis of Cryptosporidium andersoni oocysts before and after excystation. Parasit. Vectors 14, 608 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Cho, K. F. et al. Proximity labeling in mammalian cells with TurboID and split-TurboID. Nat. Protoc. 15, 3971–3999 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Chai, Y. N. et al. TMT proteomics analysis of intestinal tissue from patients of irritable bowel syndrome with diarrhea: implications for multiple nutrient ingestion abnormality. J. Proteom. 231, 103995 (2021).

    Article  CAS  Google Scholar 

  33. Blessing, C. et al. XPC–PARP complexes engage the chromatin remodeler ALC1 to catalyze global genome DNA damage repair. Nat. Commun. 13, 4762 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dong, C. et al. Screen identifies DYRK1B network as mediator of transcription repression on damaged chromatin. Proc. Natl Acad. Sci. USA 117, 17019–17030 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hoch, N. C. et al. XRCC1 mutation is associated with PARP1 hyperactivation and cerebellar ataxia. Nature 541, 87–91 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Hu, K., et al. Poly(ADP-ribosyl)ation of BRD7 by PARP1 confers resistance to DNA-damaging chemotherapeutic agents. EMBO Rep. 20, e46166 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Hu, K. et al. ATM-dependent recruitment of BRD7 is required for transcriptional repression and DNA repair at DNA breaks flanking transcriptional active regions. Adv. Sci. (Weinh.) 7, 2000157 (2020).

    CAS  PubMed  Google Scholar 

  38. Bursen, A. et al. Interaction of AF4 wild-type and AF4·MLL fusion protein with SIAH proteins: indication for t(4;11) pathobiology? Oncogene 23, 6237–6249 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Larsen, S. C. et al. Proteome-wide identification of in vivo ADP-ribose acceptor sites by liquid chromatography–tandem mass spectrometry. Methods Mol. Biol. 1608, 149–162 (2017).

    Article  CAS  PubMed  Google Scholar 

  40. Huang, D. & Kraus, W. L. The expanding universe of PARP1-mediated molecular and therapeutic mechanisms. Mol. Cell 82, 2315–2334 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Gagne, J. P. et al. Quantitative site-specific ADP-ribosylation profiling of DNA-dependent PARPs. DNA Repair (Amst.) 30, 68–79 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Moss, J., Yost, D. A. & Stanley, S. J. Amino acid-specific ADP-ribosylation. J. Biol. Chem. 258, 6466–6470 (1983).

    Article  CAS  PubMed  Google Scholar 

  43. Liu, M., Hsu, J., Chan, C., Li, Z. & Zhou, Q. The ubiquitin ligase Siah1 controls ELL2 stability and formation of super elongation complexes to modulate gene transcription. Mol. Cell 46, 325–334 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. van der Weegen, Y. et al. ELOF1 is a transcription-coupled DNA repair factor that directs RNA polymerase II ubiquitylation. Nat. Cell Biol. 23, 595–607 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Liang, K. et al. Targeting processive transcription elongation via SEC disruption for MYC-induced cancer therapy. Cell 175, 766–779 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Francette, A. M. & Arndt, K. M. Multiple direct and indirect roles of the Paf1 complex in transcription elongation, splicing, and histone modifications. Cell Rep. 43, 114730 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lavigne, M. D., Konstantopoulos, D., Ntakou-Zamplara, K. Z., Liakos, A. & Fousteri, M. Global unleashing of transcription elongation waves in response to genotoxic stress restricts somatic mutation rate. Nat. Commun. 8, 2076 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Allen, C., Her, S. & Jaffray, D. A. Radiotherapy for cancer: present and future. Adv. Drug Deliv. Rev. 109, 1–2 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Liu, H., et al. Transcriptional pausing induced by ionizing radiation enables the acquisition of radioresistance in nasopharyngeal carcinoma. J. Mol. Cell Biol. 15, mjad044 (2023).

    Article  CAS  PubMed Central  Google Scholar 

  50. Li, Z., Lu, H. & Zhou, Q. A minor subset of super elongation complexes plays a predominant role in reversing HIV-1 latency. Mol. Cell Biol. 36, 1194–1205 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hou, S., et al. PARP5A and RNF146 phase separation restrains RIPK1-dependent necroptosis. Mol. Cell 84, 938–954 (2024).

    Article  CAS  PubMed  Google Scholar 

  52. DaRosa, P. A. et al. Allosteric activation of the RNF146 ubiquitin ligase by a poly(ADP-ribosyl)ation signal. Nature 517, 223–226 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Liu, L. et al. Tankyrase-mediated ADP-ribosylation is a regulator of TNF-induced death. Sci. Adv. 8, eabh2332 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Zhang, Y. et al. RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation and Wnt signalling. Nat. Cell Biol. 13, 623–629 (2011).

    Article  CAS  PubMed  Google Scholar 

  55. Callow, M. G., et al. Ubiquitin ligase RNF146 regulates tankyrase and axin to promote Wnt signaling. PLoS ONE 6, e22595 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Chen, A., Kleiman, F. E., Manley, J. L., Ouchi, T. & Pan, Z. Q. Autoubiquitination of the BRCA1·BARD1 RING ubiquitin ligase. J. Biol. Chem. 277, 22085–22092 (2002).

    Article  CAS  PubMed  Google Scholar 

  57. Fang, S., Jensen, J. P., Ludwig, R. L., Vousden, K. H. & Weissman, A. M. Mdm2 is a RING finger-dependent ubiquitin protein ligase for itself and p53. J. Biol. Chem. 275, 8945–8951 (2000).

    Article  CAS  PubMed  Google Scholar 

  58. Hu, G. & Fearon, E. R. Siah-1 N-terminal RING domain is required for proteolysis function, and C-terminal sequences regulate oligomerization and binding to target proteins. Mol. Cell Biol. 19, 724–732 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Depaux, A., Regnier-Ricard, F., Germani, A. & Varin-Blank, N. Dimerization of hSiah proteins regulates their stability. Biochem Biophys. Res Commun. 348, 857–863 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. Yu, D., Liu, R., Yang, G. & Zhou, Q. The PARP1–Siah1 axis controls HIV-1 transcription and expression of Siah1 substrates. Cell Rep. 23, 3741–3749 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Gibbs-Seymour, I., Fontana, P., Rack, J. G. M. & Ahel, I. HPF1/C4orf27 is a PARP-1-interacting protein that regulates PARP-1 ADP-ribosylation activity. Mol. Cell 62, 432–442 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Bonfiglio, J. J. et al. Serine ADP-ribosylation depends on HPF1. Mol. Cell 65, 932–940 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Smith, R. et al. HPF1-dependent histone ADP-ribosylation triggers chromatin relaxation to promote the recruitment of repair factors at sites of DNA damage. Nat. Struct. Mol. Biol. 30, 678–691 (2023).

    Article  CAS  PubMed  Google Scholar 

  64. Suskiewicz, M. J. et al. HPF1 completes the PARP active site for DNA damage-induced ADP-ribosylation. Nature 579, 598–602 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Pellegrino, S. & Altmeyer, M. Interplay between ubiquitin, SUMO, and poly(ADP-ribose) in the cellular response to genotoxic stress. Front. Genet. 7, 63 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Wei, H. & Yu, X. Functions of PARylation in DNA damage repair pathways. Genomics Proteomics Bioinformatics 14, 131–139 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Chen, Q. et al. ADP-ribosylation of histone variant H2AX promotes base excision repair. EMBO J. 40, e104542 (2021).

    Article  CAS  PubMed  Google Scholar 

  68. Chen, Y. P. et al. Nasopharyngeal carcinoma. Lancet 394, 64–80 (2019).

    Article  PubMed  Google Scholar 

  69. Longarini, E. J. et al. Modular antibodies reveal DNA damage-induced mono-ADP-ribosylation as a second wave of PARP1 signaling. Mol. Cell 83, 1743–1760 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Li, P. et al. Nimbolide targets RNF114 to induce the trapping of PARP1 and synthetic lethality in BRCA-mutated cancer. Sci. Adv. 9, eadg7752 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Hendriks, I. A., Larsen, S. C. & Nielsen, M. L. An advanced strategy for comprehensive profiling of ADP-ribosylation sites using mass spectrometry-based proteomics. Mol. Cell Proteom. 18, 1010–1026 (2019).

    Article  CAS  Google Scholar 

  72. Shao, Y. et al. A chaperone-like function of FUS ensures TAZ condensate dynamics and transcriptional activation. Nat. Cell Biol. 26, 86–99 (2024).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the core facility of the Life Sciences Institute for technical assistance. This work was supported in part by the National Key R&D Program of China (2020YFA0908100), the National Natural Science Foundation of China (32570646, 32370591 and 32470588), the Zhejiang Provincial Natural Science Foundation of China (LRG25C060001), the Natural Science Foundation of Xiamen (3502Z202473020), the Scientific Research Foundation of State Key Laboratory of Vaccines for Infectious Diseases, the Xiang An Biomedicine Laboratory (2023XAKJ0102052) and the Fundamental Research Funds for the Central Universities (226-2025-00028).

Author information

Authors and Affiliations

Authors

Contributions

F.Z., H.F., W.Z. and C.L. performed the majority of the molecular biology and biochemistry experiments. M.Y., Z.F., Y.S., Z.L., Y.G., M.H., Y.Z. and Y.H. helped with the imaging and MS experiments. F.Z., H.F. and W.Z. analyzed the data. Y.L., Z.W., Q.Z., H.J., J.H. and L.Z. provided valuable discussion. F.Z., Y.X. and H.L. designed and supervised the project and wrote the manuscript. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Yuhua Xue or Huasong Lu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Xiaohua Shen 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 Proteomic analysis of chromatin-bound factions in response to DNA damage.

a, Correlation analysis depicting the TMT labeling quantification of chromatin-bound fractions derived from cells untreated or treated with H2O2. Quantitative mass spectrometry (MS) analysis for each group was performed in triplicate as indicated by 1-3. b, c, Gene Ontology analysis of up-regulated proteins (b) or down-regulated proteins (c) bound to chromatin after H2O2 treatment. GO enrichment analysis was conducted using Metascape, with statistical significance calculated by cumulative hypergeometric test (default setting). Only terms with Min Enrichment ≥ 1.5, Min Overlap ≥ 3 and P < 0.01 were retained. Bar plots display –log₁₀ (P value) of enriched terms for up-regulated (b) and down-regulated (c) proteins after H2O2 treatment. d, Functional interaction network of proteins showed increased binding to PAR-PARP1 and chromatin retention after H2O2. e, f, Lysates from HeLa control, or PARP1 KD (e) or PARP2 KD (f) cells were analyzed by WB. g, h, Control or PARP1 KD (g) or PARP2 KD (h) cells were untreated or treated with 2 mM H2O2 for 30 min. After fractionation, soluble and chromatin-bound fractions were analyzed by WB for the indicated proteins. All Western blots are representative of three independent experiments. Gel source data are available online.

Source Data

Extended Data Fig. 2 AFF1 binds to PARP1.

a, Confocal microscopy images of cells expressing different GFP-AFF1 variants. b, Lysates from HeLa cells expressing different GFP-AFF1 variants were analyzed by WB. c, HeLa cells expressing different GFP-AFF1 variants were subjected to co-immunoprecipitation assays. Input and anti-Flag IPs were analyzed by WB. d, Immobilized PARP1-Strep or PAR-PARP1-Strep was incubated with purified F-AFF1 for pull-down (PD) assays. Input and PD samples were analyzed by WB. e, g, Quantification of PLA assay shown in Fig. 2g (e) and Fig. 2h (g). Red lines indicate the mean intensity in each group. n = 20. f, HeLa cells expressing GFP or GFP-AFF1 were subjected to laser microirradiation and then analyzed by immunofluorescence staining with anti-PARP1. Bottom: line plot of fluorescence intensity along the dashed line. Scale bars, 20 μM in a and 10 μM in f. Experiment in a was representative of three independent experiments with similar results. Experiment in f was repeated twice, yielding similar results. All Western blots are representative of three independent experiments. Gel source data are available online.

Source Data

Extended Data Fig. 3 AFF1 is PARylated by PARP1.

a, f, g, h, WB of affinity-purified AFF1-F from cells exposed to increasing IR doses (a), or H2O2 for indicated times (f), or H2O2 ± SK575 (g), or H2O2 ± DNA-PK/ATM inhibitors (h). b, WB of Input and affinity-purified AFF1-F from cells ± H2O2 using pan-ADPr, poly-ADPr, mono-ADPr antibodies as indicated. c, Purified and immobilized af1521-Strep was incubated with lysates from AFF1-F-expressing cells ± H2O2. Input and PD samples were analyzed by WB. d, WB of affinity-purified AFF1-F from soluble / chromatin fractions of cells ± H2O2. e, WB of affinity-purified various SEC subunits from cells ± H2O2. i, j, WB of affinity-purified AFF1-F from control or PARP2 KD (i) or PARG KD (j) cells ± H2O2. k, m, Lysates from HeLa control or PARG KD (k) or HPF1 KD (m) cells were analyzed by WB. l, Affinity-purified PARylated AFF1-F were treated with PARG or NH2OH, followed by WB analysis. n, Chromatin retention of GFP-AFF1 in H2O2-exposed cells pretreated ± ActD / DRB following pre-extraction. Bottom: percentage of cells showing nuclear signal (n = 3 biologically independent replicates). Error bars indicate mean ± s.d. o, WB of soluble / chromatin fractions from cells pretreated with DRB ± H2O2. p, Quantification of GFP-AFF1 enrichment at damage sites following treatment with DMSO, ActD or AZD2281 (n = 19). Box plots show minimum, quartiles, and maximum that are representative of three independent experiments. q, WB of affinity-purified AFF1-F from cells pretreated with ActD or DRB ± H2O2, and then detected via Odyssey imaging. Scale bars, 10 μM. All Western blots are representative of three independent experiments. Gel source data are available online.

Source Data

Extended Data Fig. 4 Identification of AFF1 PARylation sites by MS.

a, WB of affinity-purified AFF1-601-900-F from control or HPF1 KD cells ± H2O2. b, Confocal microscopy images of cells expressing different AFF1 variants. Scale bar, 20 μM. c, WB of affinity-purified AFF1-F variants as indicated from cells ± H2O2. d, WB of in vitro PARylation of AFF1-601-900-F using recombinant PARP1 and indicated components. e, f, g, Fully annotated ETD tandem mass spectra of representative peptides containing an ADP-ribosylated residue, with both c-ions (red) and z-ions (blue) pinpointing the modification site. h, The sequence of the AFF1 aa 601-900 region with the 16 identified PARylation sites highlighted in red and the remaining serine residues in blue. i, HeLa cells expressing Flag-tagged AFF1-WT, -M1 or -M2 were subjected to co-immunoprecipitation assays. Input and anti-Flag IPs were analyzed by WB. j, Purified and immobilized Flag-tagged AFF1-WT, -M1 or -M2 proteins were incubated with PAR for in vitro binding analysis. Input and PD samples were analyzed by dot blotting or WB. All Western blots are representative of three independent experiments. Gel source data are available online.

Source Data

Extended Data Fig. 5 AFF1 becomes stabilized after PARylation.

a, b, HeLa cells expressing Flag-tagged AFF1-WT (a) or AFF1-M2 (b) or not (-) were untreated or treated with 2 mM H2O2 for 30 min. Input and anti-Flag IPs were analyzed by WB. c, HeLa cells expressing AFF1-M2 and/or CycT1-Mut2 as indicated were subjected to co-IP assays. Input and anti-Flag IPs were analyzed by WB. d, e, HeLa cells expressing AFF1-F were pretreated with DMSO or AZD2281 (e), or left untreated (d), and then exposed to 0.25 mM H2O2 or 1 μM Doxorubicin for the indicated times. Affinity-purified AFF1-F proteins were analyzed by WB. f, HeLa cells expressing Flag-tagged AFF1-WT or AFF1-M2 were exposed to 0.25 mM H2O2 or 1 μM Doxorubicin for the indicated times and then analyzed as in e. g, h, WB of affinity-purified AFF1-WT or -2R from cells untreated or treated with 1 μM Doxorubicin (g) or 2 mM H2O2 (h). i, j, k, l, b, HeLa cells were treated with CHX to block protein synthesis and harvested at indicated times after no treatment or treatment with MG132 (i) or MNNG (j) or MMS (k) or UV (l). m, Control or PARP1 KO cells expressing PARP1-E988A were treated ana analyzed as in j. n, HeLa cells expressing Flag-tagged AFF1-WT or AFF1-M2 were treated ana analyzed as in j. o, The mRNA levels of Siah1 in control or Siah1 KD cells were measured by qRT-PCR. Error bars indicate mean ± s.d., n = 3 biologically independent samples. p, HeLa cells expressing AFF1-F and/or His-Ub were treated with 2 mM H2O2 for 30 min and then followed by MG132 incubation for 8 hrs. Cells were harvested for Ni-NTA pull-down assays and analyzed by WB. All Western blots are representative of three independent experiments. Gel source data are available online.

Source Data

Extended Data Fig. 6 Depletion of AFF1 attenuates transcriptional recovery after DNA damage.

a, b, Metaplots of average TT-seq signals for genes (10-25 kb, 25-50 kb, 50-100 kb; a) or AFF1 targets (b) in control/AFF1 KD cells at different times after H2O2 treatment. c, Box plot of log2 fold change in TT-seq signals of AFF1 targets at 2.5 h post-H2O2 vs untreated. Box plots show minimum, quartiles, and maximum, n = 10229. d, GO analysis of 2311 AFF1 targets with impaired recovery in KD cells. e, Metaplots of spike-in normalized TT-seq signals in control or AFF1 KD cells at different time points post-H2O2. f, h, Heatmaps of pSer2 occupancy across gene bodies in control (f) or AFF1 KD (h) cells post-H2O2. g, i, Metaplots of pSer2 ChIP-seq signals in control (g) or AFF1 KD (i) cells at different time points post-H2O2. j, k, Metaplots showing the average level of nascent transcription of genes in control (j) or AFF1 KD (k) cells. Wavefront positions are defined as the distance from the TSS where the reads density drops below an arbitrary threshold (dashed line). l, WB of lysates from AFF1 KD cells reconstituted with AFF1-WT or M2 ± 0.25 mM H2O2 for 5 min, followed by recovery for the indicated times. m, HeLa cells pre-treated with DMSO or AZD2281 were treated ± H2O2 for 5 min and allowed to recover for the indicated times. Nascent RNA was labeled with 5-EU for 20 min and quantified by fluorescence. Quantification: relative fluorescence intensity per cell normalized to mock (set to 100). Red lines indicate mean intensity. n = 97,100,109,74,95,108, left to right. Scale bars = 20 μM. Statistical analysis was performed using two-tailed unpaired t-tests. ****P < 0.0001. All Western blots are representative of three independent experiments. Gel source data are available online.

Source Data

Extended Data Fig. 7 Depletion of AFF1 enhances the sensitivity of cancer cells to genotoxic stress.

a, Representative colony formation assay images (as Fig.7a). b, k, WB of lysates from control or AFF1 KD HeLa (b) or CR (k) cells. c, Colony formation of control or AFF1 KD cells with increasing IR doses (n = 3). d, o, Control or AFF1 KD HeLa (d) or CR (o) cells were exposed to 2 Gy IR and then harvested at the indicated time points for WB analysis. e, γH2AX immunofluorescence in control or AFF1 KD cells after 5 Gy IR. Right: quantification of γH2AX foci per cell. Red lines indicate mean, n = 100. f, Alkaline comet assays of control, PARP1 KD, or AFF1 KD cells at the indicated time points after treatment with 0.25 mM H2O2 for 5 min. Quantification showing the tail moment per cell. Red lines indicate the mean in each group. n = 98. Statistical analysis was performed using two-tailed unpaired t-tests. ****P < 0.0001, ***P < 0.001. g, m, Colony formation of CNE2 or CR cells (g, n = 3) or CR control or AFF1 KD cells (m, n = 3) with increasing IR doses. h, Crystal violet staining of CNE2 or CR cells ± AZD2281 1 week after 10 Gy IR. i, LFQ intensities of indicated proteins in CNE2 or CR cells. j, qRT-PCR analysis of AFF1 mRNA levels. l, Proliferation curves of control, AFF1 KD, and AFF4 KD CR cells (n = 3). n, SA-β-gal staining of control or AFF1 KD CR cells 3 days after 6 Gy IR. Right: quantification of the number of SA-β-gal positive cells per field. p, q, Graphs of isolated tumors in Fig.7k (p) or Fig.7 m (q). All error bars indicate mean ± s.d. with n = 3 biologically independent samples. All Western blots are representative of three independent experiments. Gel source data are available online.

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

Zhu, F., Fu, H., Zhu, W. et al. Stabilization of AFF1 by PARylation ensures transcriptional restart after DNA damage. Nat Chem Biol (2025). https://doi.org/10.1038/s41589-025-02045-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41589-025-02045-5

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