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:

CUL-2LRR-1 and UBXN-3 drive replisome disassembly during DNA replication termination and mitosis

Abstract

Replisome disassembly is the final step of DNA replication in eukaryotes, involving the ubiquitylation and CDC48-dependent dissolution of the CMG helicase (CDC45–MCM–GINS). Using Caenorhabditis elegans early embryos and Xenopus laevis egg extracts, we show that the E3 ligase CUL-2LRR-1 associates with the replisome and drives ubiquitylation and disassembly of CMG, together with the CDC-48 cofactors UFD-1 and NPL-4. Removal of CMG from chromatin in frog egg extracts requires CUL2 neddylation, and our data identify chromatin recruitment of CUL2LRR1 as a key regulated step during DNA replication termination. Interestingly, however, CMG persists on chromatin until prophase in worms that lack CUL-2LRR-1, but is then removed by a mitotic pathway that requires the CDC-48 cofactor UBXN-3, orthologous to the human tumour suppressor FAF1. Partial inactivation of lrr-1 and ubxn-3 leads to synthetic lethality, suggesting future approaches by which a deeper understanding of CMG disassembly in metazoa could be exploited therapeutically.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The CDC-48 cofactor NPL-4 is required for CMG helicase disassembly during S phase in the C. elegans early embryo.
Figure 2: CUL-2LRR-1 is required for CMG helicase disassembly during S phase in C. elegans.
Figure 3: A mitotic pathway for CMG helicase disassembly is revealed in the absence of CUL-2LRR-1.
Figure 4: The mitotic CMG helicase disassembly pathway requires UBXN-3 and is modulated by the SUMO protease ULP-4, both of which become essential when LRR-1 is depleted.
Figure 5: Isolation of the post-termination worm replisome.
Figure 6: CUL2LRR1 associates with the post-termination vertebrate replisome and is recruited to chromatin during DNA replication termination in Xenopus egg extracts.
Figure 7: Active CUL2LRR1 is required for extraction of CMG components from chromatin during DNA replication termination in Xenopus egg extracts.

Similar content being viewed by others

References

  1. Gambus, A. et al. GINS maintains association of Cdc45 with MCM in replisome progression complexes at eukaryotic DNA replication forks. Nat. Cell Biol. 8, 358–366 (2006).

    CAS  PubMed  Google Scholar 

  2. Moyer, S. E., Lewis, P. W. & Botchan, M. R. Isolation of the Cdc45/Mcm2-7/GINS (CMG) complex, a candidate for the eukaryotic DNA replication fork helicase. Proc. Natl Acad. Sci. USA 103, 10236–10241 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Bell, S. P. & Labib, K. Chromosome duplication in Saccharomyces cerevisiae. Genetics 203, 1027–1067 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Deegan, T. D. & Diffley, J. F. MCM: one ring to rule them all. Curr. Opin. Struct. Biol. 37, 145–151 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. O’Donnell, M. & Li, H. The eukaryotic replisome goes under the microscope. Curr. Biol. 26, R247–R256 (2016).

    PubMed  PubMed Central  Google Scholar 

  6. Labib, K., Tercero, J. A. & Diffley, J. F. X. Uninterrupted MCM2-7 function required for DNA replication fork progression. Science 288, 1643–1647 (2000).

    CAS  PubMed  Google Scholar 

  7. Ilves, I., Petojevic, T., Pesavento, J. J. & Botchan, M. R. Activation of the MCM2-7 helicase by association with Cdc45 and GINS proteins. Mol. Cell 37, 247–258 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Bell, S. P. DNA replication. Terminating the replisome. Science 346, 418–419 (2014).

    CAS  PubMed  Google Scholar 

  9. Dewar, J. M., Budzowska, M. & Walter, J. C. The mechanism of DNA replication termination in vertebrates. Nature 525, 345–350 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Maric, M., Maculins, T., De Piccoli, G. & Labib, K. Cdc48 and a ubiquitin ligase drive disassembly of the CMG helicase at the end of DNA replication. Science 346, 1253596 (2014).

    PubMed  PubMed Central  Google Scholar 

  11. Moreno, S. P., Bailey, R., Campion, N., Herron, S. & Gambus, A. Polyubiquitylation drives replisome disassembly at the termination of DNA replication. Science 346, 477–481 (2014).

    PubMed  Google Scholar 

  12. Maculins, T., Nkosi, P. J., Nishikawa, H. & Labib, K. Tethering of SCF(Dia2) to the replisome promotes efficient ubiquitylation and disassembly of the CMG helicase. Curr. Biol. 25, 2254–2259 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Morohashi, H., Maculins, T. & Labib, K. The amino-terminal TPR domain of Dia2 tethers SCF(Dia2) to the replisome progression complex. Curr. Biol. 19, 1943–1949 (2009).

    CAS  PubMed  Google Scholar 

  14. Soucy, T. A. et al. An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer. Nature 458, 732–736 (2009).

    CAS  PubMed  Google Scholar 

  15. Duda, D. M. et al. Structural insights into NEDD8 activation of cullin-RING ligases: conformational control of conjugation. Cell 134, 995–1006 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Saha, A. & Deshaies, R. J. Multimodal activation of the ubiquitin ligase SCF by Nedd8 conjugation. Mol. Cell 32, 21–31 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Sonneville, R., Querenet, M., Craig, A., Gartner, A. & Blow, J. J. The dynamics of replication licensing in live Caenorhabditis elegans embryos. J. Cell Biol. 196, 233–246 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Sonneville, R., Craig, G., Labib, K., Gartner, A. & Blow, J. J. Both chromosome decondensation and condensation are dependent on DNA replication in C. elegans embryos. Cell Rep. 12, 405–417 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Avci, D. & Lemberg, M. K. Clipping or extracting: two ways to membrane protein degradation. Trends Cell Biol. 25, 611–622 (2015).

    CAS  PubMed  Google Scholar 

  20. Franz, A., Ackermann, L. & Hoppe, T. Ring of change: CDC48/p97 drives protein dynamics at chromatin. Front. Genet. 7, 73 (2016).

    PubMed  PubMed Central  Google Scholar 

  21. Ramadan, K., Halder, S., Wiseman, K. & Vaz, B. Strategic role of the ubiquitin-dependent segregase p97 (VCP or Cdc48) in DNA replication. Chromosoma http://dx.doi.org/10.1007/s00412-016-0587-4 (2016).

  22. Meyer, H. H., Shorter, J. G., Seemann, J., Pappin, D. & Warren, G. A complex of mammalian ufd1 and npl4 links the AAA-ATPase, p97, to ubiquitin and nuclear transport pathways. EMBO J. 19, 2181–2192 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Mouysset, J., Kahler, C. & Hoppe, T. A conserved role of Caenorhabditis elegans CDC-48 in ER-associated protein degradation. J. Struct. Biol. 156, 41–49 (2006).

    CAS  PubMed  Google Scholar 

  24. Sarikas, A., Hartmann, T. & Pan, Z. Q. The cullin protein family. Genome Biol. 12, 220 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Kloppsteck, P., Ewens, C. A., Forster, A., Zhang, X. & Freemont, P. S. Regulation of p97 in the ubiquitin-proteasome system by the UBX protein-family. Biochim. Biophys. Acta 1823, 125–129 (2012).

    CAS  PubMed  Google Scholar 

  26. Meyer, H., Bug, M. & Bremer, S. Emerging functions of the VCP/p97 AAA-ATPase in the ubiquitin system. Nat. Cell Biol. 14, 117–123 (2012).

    CAS  PubMed  Google Scholar 

  27. Vaz, B., Halder, S. & Ramadan, K. Role of p97/VCP (Cdc48) in genome stability. Front. Genet. 4, 60 (2013).

    PubMed  PubMed Central  Google Scholar 

  28. Pelisch, F. et al. Dynamic SUMO modification regulates mitotic chromosome assembly and cell cycle progression in Caenorhabditis elegans. Nat. Commun. 5, 5485 (2014).

    CAS  PubMed  Google Scholar 

  29. Merlet, J. et al. The CRL2LRR-1 ubiquitin ligase regulates cell cycle progression during C. elegans development. Development 137, 3857–3866 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Starostina, N. G., Simpliciano, J. M., McGuirk, M. A. & Kipreos, E. T. CRL2(LRR-1) targets a CDK inhibitor for cell cycle control in C. elegans and actin-based motility regulation in human cells. Dev. Cell 19, 753–764 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Fullbright, G., Rycenga, H. B., Gruber, J. D. & Long, D. T. p97 promotes a conserved mechanism of helicase unloading during DNA cross-link repair. Mol. Cell. Biol. 36, 2983–2994 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Semlow, D. R., Zhang, J., Budzowska, M., Drohat, A. C. & Walter, J. C. Replication-dependent unhooking of DNA interstrand cross-links by the NEIL3 glycosylase. Cell 167, 498–511 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Cuvier, O., Stanojcic, S., Lemaitre, J. M. & Mechali, M. A topoisomerase II-dependent mechanism for resetting replicons at the S-M-phase transition. Genes Dev. 22, 860–865 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Bandau, S., Knebel, A., Gage, Z. O., Wood, N. T. & Alexandru, G. UBXN7 docks on neddylated cullin complexes using its UIM motif and causes HIF1α accumulation. BMC Biol. 10, 36 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Ossareh-Nazari, B., Katsiarimpa, A., Merlet, J. & Pintard, L. RNAi-based suppressor screens reveal genetic interactions between the CRL2LRR-1 E3-ligase and the DNA replication machinery in Caenorhabditis elegans. G3 (Bethesda) 6, 3431–3442 (2016).

    CAS  Google Scholar 

  36. Dewar, J. M., Low, E., Mann, M., Raschle, M. & Walter, J. C. CRL2Lrr1 promotes unloading of the vertebrate replisome from chromatin during replication termination. Genes Dev. http://dx.doi.org/10.1101/gad.291799.116 (2017).

  37. Franz, A. et al. CDC-48/p97 coordinates CDT-1 degradation with GINS chromatin dissociation to ensure faithful DNA replication. Mol. Cell 44, 85–96 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Franz, A. et al. Chromatin-associated degradation is defined by UBXN-3/FAF1 to safeguard DNA replication fork progression. Nat. Commun. 7, 10612 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Lee, J. J. et al. Complex of Fas-associated factor 1 (FAF1) with valosin-containing protein (VCP)-Npl4-Ufd1 and polyubiquitinated proteins promotes endoplasmic reticulum-associated degradation (ERAD). J. Biol. Chem. 288, 6998–7011 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Menges, C. W., Altomare, D. A. & Testa, J. R. FAS-associated factor 1 (FAF1): diverse functions and implications for oncogenesis. Cell Cycle 8, 2528–2534 (2009).

    CAS  PubMed  Google Scholar 

  41. Sharrock, W. J., Sutherlin, M. E., Leske, K., Cheng, T. K. & Kim, T. Y. Two distinct yolk lipoprotein complexes from Caenorhabditis elegans. J. Biol. Chem. 265, 14422–14431 (1990).

    CAS  PubMed  Google Scholar 

  42. Gambus, A. et al. A key role for Ctf4 in coupling the MCM2-7 helicase to DNA polymerase alpha within the eukaryotic replisome. EMBO J. 28, 2992–3004 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Sengupta, S., van Deursen, F., de Piccoli, G. & Labib, K. Dpb2 integrates the leading-strand DNA polymerase into the eukaryotic replisome. Curr. Biol. 23, 543–552 (2013).

    CAS  PubMed  Google Scholar 

  44. Brenner, S. The genetics of Caenorhabditis elegans. Genetics 77, 71–94 (1974).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Edgley, M. L., Baillie, D. L., Riddle, D. L. & Rose, A. M. Genetic balancers. WormBook 6, 1–32 (2006).

    Google Scholar 

  46. Timmons, L. & Fire, A. Specific interference by ingested dsRNA. Nature 395, 854 (1998).

    CAS  PubMed  Google Scholar 

  47. Gillespie, P. J., Gambus, A. & Blow, J. J. Preparation and use of Xenopus egg extracts to study DNA replication and chromatin associated proteins. Methods 57, 203–213 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Gambus, A., Khoudoli, G. A., Jones, R. C. & Blow, J. J. MCM2-7 form double hexamers at licensed origins in Xenopus egg extract. J. Biol. Chem. 286, 11855–11864 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Khoudoli, G. A. et al. Temporal profiling of the chromatin proteome reveals system-wide responses to replication inhibition. Curr. Biol. 18, 838–843 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Prokhorova, T. A. & Blow, J. J. Sequential MCM/P1 subcomplex assembly is required to form a heterohexamer with replication licensing activity. J. Biol. Chem. 275, 2491–2498 (2000).

    CAS  PubMed  Google Scholar 

  51. Heubes, S. & Stemmann, O. The AAA-ATPase p97-Ufd1-Npl4 is required for ERAD but not for spindle disassembly in Xenopus egg extracts. J. Cell Sci. 120, 1325–1329 (2007).

    CAS  PubMed  Google Scholar 

  52. Walter, J. C. Evidence for sequential action of cdc7 and cdk2 protein kinases during initiation of DNA replication in Xenopus egg extracts. J. Biol. Chem. 275, 39773–39778 (2000).

    CAS  PubMed  Google Scholar 

  53. Hodgson, B., Li, A., Tada, S. & Blow, J. J. Geminin becomes activated as an inhibitor of Cdt1/RLF-B following nuclear import. Curr. Biol. 12, 678–683 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Lee, D. W. et al. The Dac-tag, an affinity tag based on penicillin-binding protein 5. Anal. Biochem. 428, 64–72 (2012).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge the support of the Medical Research Council (core grant MC_UU_12016/13 for K.L.; award MR/K007106/1 to A.Gambus) the Wellcome Trust (reference 102943/Z/13/Z for award to K.L.; reference 0909444/Z/09/Z for award to A.Gartner) and the Lister Institute (award to A.Gambus) for funding our work. We thank J. Blow for geminin protein, MRC PPU reagents (https://mrcppureagents.dundee.ac.uk) for recombinant frog LRR1 and for producing antibodies, and T. Deegan for helpful comments on the manuscript. We also thank L. Pintard (Institut Jacques Monod, France) for providing the worm line heterozygous for lrr-1Δ, C. Ponting for advice regarding orthologues of the budding yeast Dia2 protein, and J. Walter and E. Low for discussing unpublished findings.

Author information

Authors and Affiliations

Authors

Contributions

R.S. performed the experiments in Figs 15 and Supplementary Figs 14. S.P.M. performed the experiments in Figs 6 and 7 and Supplementary Fig. 5. K.L. and A.Gambus conceived the project and designed experiments in collaboration with R.S. and S.P.M. A.K. and C.J. produced recombinant CUL2–RBX1 and C.J.H. provided recombinant LRR1. A.Gartner provided invaluable support in the early stages of the project. K.L. wrote the manuscript, with contributions and critical comments from the other authors.

Corresponding authors

Correspondence to Agnieszka Gambus or Karim Labib.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 The CDC-48_UFD-1_NPL-4 complex is required for CMG helicase disassembly in C. elegans.

(a) cdc-48 RNAi leads to persistence of GINS and CDC-45 on chromatin during prophase and throughout mitosis (examples indicated by arrows). (b) Adaptors of CDC-48 in C. elegans. (c) ufd-1 RNAi leads to persistence of GINS and CDC-45 on chromatin during prophase and throughout mitosis (examples indicated by arrows). (d) Equivalent experiment to that in Figure 1d, illustrating the effect of npl-4 RNAi on embryos expressing GFP-MCM-3. To help visualise the small proportion of GFP-MCM-3 on chromatin in early metaphase (marked by an arrow), the experiment also included RNAi to the 3’UTR of endogenous MCM3 (this 3’ UTR is not present in the GFP-MCM-3 transgene), to increase the incorporation of GFP-MCM3 into replication complexes. (e) cdc-48 RNAi experiment, analogous to that in Figure 1d. (f) Homozygous GFP-psf-1 worms were exposed to the indicated RNAi. Embryos were then isolated and processed as in Figure 1e-f. The middle panels show that the amount of CMG isolated from RNR-1 depleted extract was reduced compared to control (compare levels of MCM-7, MCM-2 and CDC-45), due to the inhibition of DNA replication in each embryonic cell cycle. In the right panels, loading of the GFP-PSF-1 IP samples was adjusted to obtain a similar level of CMG (compare MCM-2 and CDC-45). (g,h) Photobleaching experiments for GFP-SLD5 and GFP-MCM3, equivalent to the experiment in Figure 1h. The scale bars correspond to 5 μm. Unprocessed scans of key immunoblots are shown in Supplementary Figure 8.

Supplementary Figure 2 CUL-2LRR-1 is required for removal of GINS from chromatin during S-phase in C. elegans.

(a) C. elegans contain six families of cullin complexes, each with a specific cullin and a unique set of substrate adaptors. (b) Embryos from GFP-sld-5 mCherry-H2B worms were exposed to RNAi against the indicated cullins and processed as in Figure 2. Timelapse images are shown from S-phase to mid-prophase. (c) Six forms of the CUL-2 ligase in C. elegans, each with a unique substrate adaptor. (d) Embryos from GFP-sld-5 mCherry-H2B worms were exposed to RNAi against the indicated substrate adaptors of CUL-2 and processed as in Figure 2. Timelapse images are shown from S-phase to mid-prophase. RNAi for zyg-11 produces meiotic defects and leads to abnormal nuclear morphology in the first embryonic cell cycle. Arrows in this figure indicate the persistent association of GFP-SLD-5 with mitotic chromatin in embryos treated with npl-4 RNAi. Scale bars correspond to 5 μm.

Supplementary Figure 3 A new pathway for CMG helicase disassembly acts during mitosis.

(a) Embryos from GFP-psf-1 mCherry-H2B worms were exposed to the indicated RNAi and processed as in Figure 3. Timelapse images of the first embryonic cell cycle are shown from S-phase to metaphase. GFP-PSF1 initially persists on prophase chromatin following RNAi to components of CUL-2LRR-1 (the arrows denote examples), before being released in late prophase (indicated by asterisks). (b) Extended timecourses for the GFP-SLD-5 data presented in Figure 2a, b. (c) Data from the first cell cycle, for the experiment in Figure 3b. (d) Embryos from GFP-cdc-45 mCherry-H2B worms were exposed to the indicated RNAi and processed as above. (e) Illustration of CMG disassembly defects produced either by depletion of CDC-48/UFD-1/NPL-4, or by depletion of components of CUL-2LRR-1. Scale bars correspond to 5 μm.

Supplementary Figure 4 The mitotic disassembly pathway for the CMG helicase requires UBXN-3 and is modulated by ULP-4.

(a) Embryos from GFP-sld-5 mCherry-H2B worms were exposed to the indicated RNAi and processed as in Figure 4a. The arrows indicate persistent association of GFP-PSF1 with mitotic chromatin (throughout mitosis in the case of RNAi to npl-4, or after simultaneous RNAi to lrr-1 + ubxn-3), whereas the asterisk denotes release of GFP-PSF-1 from chromatin in late prophase in embryos treated only with lrr-1 RNAi. Scale bars correspond to 5 μm. (b) Embryos from GFP-cdc-45 mCherry-H2B worms were processed as for Figure 4b. (c) Embryos from GFP-psf-1 mCherry-H2B worms were exposed to the indicated RNAi and processed as above. The data correspond to the AB cell in the second cell cycle and CMG components remained on chromatin until at or after nuclear envelope breakdown in 3/5 embryos treated with lrr-1 ulp-4 double RNAi. The panel shows an example of an embryo where CMG persists on chromatin until nuclear envelope breakdown upon co-depletion of LRR-1 and ULP-4. (d) Data from a similar experiment, corresponding to the EMS cell in the third cell cycle. Note that in this case we also depleted the ATL-1 checkpoint kinase, to shorten the otherwise long cell cycle delay that is induced by the combination of ulp-4 lrr-1 double RNAi. CMG components remained on chromatin until late metaphase in 5/5 embryos treated with lrr-1 ulp-4 atl-1 triple RNAi. CMG was extracted normally from chromatin during S-phase in embryos subjected to ulp-4 atl-1 double RNAi (5/5 embryos tested), whereas lrr-1 atl-1 double RNAi resembled lrr-1 single RNAi treatment (CMG extracted before the end of prophase in 5/5 embryos).

Supplementary Figure 5 Additional supplementary material for experiments with Xenopus egg extracts.

(a) In a similar experiment to that in Figure 6c, replisome disassembly was blocked during chromosome replication by addition of MLN4924 to Xenopus egg extracts. After isolation of chromatin and digestion of DNA, immunoprecipitation of LRR1 led to co-depletion of CUL2. (b) Analysis of ongoing DNA synthesis at the indicated timepoints for the experiment in Figure 6e, f, by addition of short pulses of α-dATP (see Methods). Data for repeats of this experiment are included in Supplementary Table 6. (c) Replication kinetics for the experiment in Figure 6h, measured by monitoring total incorporation of α-dATP into nascent DNA by the indicated timepoints (see Methods). Data for repeats of this experiment are included in Supplementary Table 6. Unprocessed scans of key immunoblots from this Figure are shown in Supplementary Figure 8.

Supplementary Figure 6 CUL2 is very highly conserved in vertebrates.

Alignment of Xenopus CUL2 with the human and mouse orthologues, showing that the mammalian and frog proteins are almost identical. Moreover, previous work indicated that all key residues in CUL2 that contact EloB-C and substrate adaptors are 100% conserved between the human and frog orthologues1.

Supplementary Figure 7 Validation of new antibodies generated in this study for C. elegans proteins.

(ad) In each case, RNAi was used to deplete the corresponding protein, before immunoblotting of embryonic extracts (upper panels). Ponceau S staining of the nitrocellulose membare (lower panels) provides a loading control in each case.

Supplementary information

Supplementary Information

Supplementary Information (PDF 5929 kb)

Supplementary Table 1

Supplementary Information (XLSX 34 kb)

Supplementary Table 2

Supplementary Information (XLSX 33 kb)

Supplementary Table 3

Supplementary Information (XLSX 34 kb)

Supplementary Table 4

Supplementary Information (XLSX 33 kb)

Supplementary Table 5

Supplementary Information (XLSX 33 kb)

Supplementary Table 6

Supplementary Information (XLSX 13 kb)

Supplementary Table 7

Supplementary Information (XLSX 33 kb)

Supplementary Table 8

Supplementary Information (XLSX 43 kb)

The CMG helicase component PSF-1 does not associate with condensing chromatin during mitotic prophase or throughout mitosis.

Video of a single optical section through an embryo expressing GFP-PSF-1 (left panel) and mCherry-Histone H2B (right panel) progressing throughout the first and second embryonic cell cycles. Images were acquired every 10 sec with a spinning disk confocal microscope and processed with ImageJ software. The female and male pronuclei are orientated respectively towards the left and right of the video. (MOV 2692 kb)

GFP-PSF-1 associates with condensing chromatin during prophase in embryos depleted for NPL-4 and remains on chromatin throughout mitosis.

Images were acquired and analysed as for Supplementary Movie 1. (MOV 5257 kb)

FRAP analysis of GFP-CDC-45 after depletion of NPL-4.

The movie was generated as above and shows an embryo expressing GFP-CDC-45 (left panel) and mCherry-Histone H2B (right panel). The female pronucleus (left side of the embryo) was photobleached during early S-phase (shown as a white disk in the video at 1’50”) and the chromosomes from the female and male pronuclei were then analysed during the following mitosis (see 19’30” to 24’50”). No recovery of the GFP-CDC-45 signal was observed on the female chromatin, indicating that depletion of NPL-4 causes CDC-45 to persist on chromatin from S-phase until the end of mitosis. (MOV 1176 kb)

GFP-PSF-1 associates with condensing chromatin during prophase in embryos depleted for CUL-2, but is then released from chromatin during late prophase.

Images were acquired and analysed as for Supplementary Movie 1. Note that depletion of CUL-2 leads to meiotic defects in the embryo and thus to abnormal nuclear morphology, reflecting the important role of CUL-2ZYG-11 during the second meiotic cell division2,3. In addition, mitotic entry is delayed in the first embryonic cell cycle after depletion of CUL-2. (MOV 3596 kb)

GFP-PSF-1 associates with condensing chromatin during prophase in embryos depleted for LRR-1, but is then released from chromatin during late prophase.

Images were acquired and analysed as for Supplementary Movie 1. The association of GFP-PSF-1 with prophase chromatin can be seen in the first embryonic cell cycle (P0 cell) from 3’20” to 5’50” and during the second cell cycle from 24’30” to 26’10” for the AB cell (left side of embryo) or from 27’30” to 29’10” for the P1 cell (right side). (MOV 2271 kb)

GFP-PSF-1 remains on chromatin throughout mitosis in embryos depleted for both UBXN-3 and LRR-1.

Images were acquired and analysed as for Supplementary Movie 1. (MOV 3887 kb)

GFP-PSF-1 is released from chromatin before prophase in ubxn-3 RNAi embryos.

Images were acquired and analysed as for Supplementary Movie 1. (MOV 3219 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sonneville, R., Moreno, S., Knebel, A. et al. CUL-2LRR-1 and UBXN-3 drive replisome disassembly during DNA replication termination and mitosis. Nat Cell Biol 19, 468–479 (2017). https://doi.org/10.1038/ncb3500

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3500

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