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:

Mechanism of asymmetric polymerase assembly at the eukaryotic replication fork

Abstract

Eukaryotes use distinct polymerases for leading- and lagging-strand replication, but how they target their respective strands is uncertain. We reconstituted Saccharomyces cerevisiae replication forks and found that CMG helicase selects polymerase (Pol) ɛ to the exclusion of Pol δ on the leading strand. Even if Pol δ assembles on the leading strand, Pol ɛ rapidly replaces it. Pol δ–PCNA is distributive with CMG, in contrast to its high stability on primed ssDNA. Hence CMG will not stabilize Pol δ, instead leaving the leading strand accessible for Pol ɛ and stabilizing Pol ɛ. Comparison of Pol ɛ and Pol δ on a lagging-strand model DNA reveals the opposite. Pol δ dominates over excess Pol ɛ on PCNA-primed ssDNA. Thus, PCNA strongly favors Pol δ over Pol ɛ on the lagging strand, but CMG over-rides and flips this balance in favor of Pol ɛ on the leading strand.

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: Reconstitution of a leading-strand replisome with CMG, Pol ɛ, RFC, PCNA and RPA.
Figure 2: Comparison of Pol ɛ and Pol δ in leading-strand replication with CMG.
Figure 3: CMG selects Pol ɛ from a mixture of Pol ɛ and Pol δ.
Figure 4: Pol ɛ functions stably with CMG, whereas Pol δ does not.
Figure 5: Pol ɛ and Pol δ rapidly associate with a PCNA-primed template.
Figure 6: Pol δ is dominant over Pol ɛ on a lagging-strand model template.
Figure 7: Scheme of asymmetric polymerase assembly at a replication fork.

Similar content being viewed by others

References

  1. Méndez, J. & Stillman, B. Perpetuating the double helix: molecular machines at eukaryotic DNA replication origins. BioEssays 25, 1158–1167 (2003).

    Article  PubMed  Google Scholar 

  2. Benkovic, S.J., Valentine, A.M. & Salinas, F. Replisome-mediated DNA replication. Annu. Rev. Biochem. 70, 181–208 (2001).

    Article  CAS  PubMed  Google Scholar 

  3. O'Donnell, M., Langston, L.D. & Stillman, B. Principles and concepts of DNA replication in bacteria, archaea, and eukarya. Cold Spring Harb. Perspect. Biol. 5, a010108 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Kornberg, A. & Baker, T.A. DNA Replication (W.H. Freeman, New York, 1992).

  5. Kunkel, T.A. & Burgers, P.M. Dividing the workload at a eukaryotic replication fork. Trends Cell Biol. 18, 521–527 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pursell, Z.F., Isoz, I., Lundstrom, E.B., Johansson, E. & Kunkel, T.A. Yeast DNA polymerase ɛ participates in leading-strand DNA replication. Science 317, 127–130 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jeruzalmi, D., O'Donnell, M. & Kuriyan, J. Clamp loaders and sliding clamps. Curr. Opin. Struct. Biol. 12, 217–224 (2002).

    Article  CAS  PubMed  Google Scholar 

  8. Pan, M., Kelman, L.M. & Kelman, Z. The archaeal PCNA proteins. Biochem. Soc. Trans. 39, 20–24 (2011).

    Article  CAS  PubMed  Google Scholar 

  9. Hedglin, M., Kumar, R. & Benkovic, S.J. Replication clamps and clamp loaders. Cold Spring Harb. Perspect. Biol. 5, a010165 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  10. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Onesti, S. & MacNeill, S.A. Structure and evolutionary origins of the CMG complex. Chromosoma 122, 47–53 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Bochman, M.L. & Schwacha, A. The Saccharomyces cerevisiae Mcm6/2 and Mcm5/3 ATPase active sites contribute to the function of the putative Mcm2–7 'gate'. Nucleic Acids Res. 38, 6078–6088 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Costa, A. et al. The structural basis for MCM2–7 helicase activation by GINS and Cdc45. Nat. Struct. Mol. Biol. 18, 471–477 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bochman, M.L. & Schwacha, A. The Mcm2–7 complex has in vitro helicase activity. Mol. Cell 31, 287–293 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kang, Y.H., Galal, W.C., Farina, A., Tappin, I. & Hurwitz, J. Properties of the human Cdc45/Mcm2–7/GINS helicase complex and its action with DNA polymerase ɛ in rolling circle DNA synthesis. Proc. Natl. Acad. Sci. USA 109, 6042–6047 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bell, S.P. & Kaguni, J.M. Helicase loading at chromosomal origins of replication. Cold Spring Harb. Perspect. Biol. 5, a010124 (2013).

    PubMed  PubMed Central  Google Scholar 

  18. Boos, D., Frigola, J. & Diffley, J.F. Activation of the replicative DNA helicase: breaking up is hard to do. Curr. Opin. Cell Biol. 24, 423–430 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Fu, Y.V. et al. Selective bypass of a lagging strand roadblock by the eukaryotic replicative DNA helicase. Cell 146, 931–941 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Tanaka, S. & Araki, H. Helicase activation and establishment of replication forks at chromosomal origins of replication. Cold Spring Harb. Perspect. Biol. 5, a010371 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Sheu, Y.J., Kinney, J.B., Lengronne, A., Pasero, P. & Stillman, B. Domain within the helicase subunit Mcm4 integrates multiple kinase signals to control DNA replication initiation and fork progression. Proc. Natl. Acad. Sci. USA 111, E1899–E1908 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Evrin, C. et al. A double-hexameric MCM2–7 complex is loaded onto origin DNA during licensing of eukaryotic DNA replication. Proc. Natl. Acad. Sci. USA 106, 20240–20245 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Speck, C., Chen, Z., Li, H. & Stillman, B. ATPase-dependent cooperative binding of ORC and Cdc6 to origin DNA. Nat. Struct. Mol. Biol. 12, 965–971 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bermudez, V.P., Farina, A., Raghavan, V., Tappin, I. & Hurwitz, J. Studies on human DNA polymerase ɛ and GINS complex and their role in DNA replication. J. Biol. Chem. 286, 28963–28977 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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).

    Article  CAS  PubMed  Google Scholar 

  28. Takayama, Y. et al. GINS, a novel multiprotein complex required for chromosomal DNA replication in budding yeast. Genes Dev. 9, 1153–1165 (2003).

    Article  Google Scholar 

  29. Sowd, G.A. & Fanning, E. A wolf in sheep's clothing: SV40 co-opts host genome maintenance proteins to replicate viral DNA. PLoS Pathog. 8, e1002994 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Waga, S. & Stillman, B. The DNA replication fork in eukaryotic cells. Annu. Rev. Biochem. 67, 721–751 (1998).

    Article  CAS  PubMed  Google Scholar 

  31. Stillman, B. DNA polymerases at the replication fork in eukaryotes. Mol. Cell 30, 259–260 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zlotkin, T. et al. DNA polymerase epsilon may be dispensable for SV40- but not cellular-DNA replication. EMBO J. 15, 2298–2305 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sekedat, M.D. et al. GINS motion reveals replication fork progression is remarkably uniform throughout the yeast genome. Mol. Syst. Biol. 6, 353 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  34. On, K.F. et al. Prereplicative complexes assembled in vitro support origin-dependent and independent DNA replication. EMBO J. 33, 605–620 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Gros, J., Devbhandari, S. & Remus, D. Origin plasticity during budding yeast DNA replication in vitro. EMBO J. 33, 1284 (2014).

    Article  CAS  PubMed Central  Google Scholar 

  36. Lee, S.H., Pan, Z.Q., Kwong, A.D., Burgers, P.M. & Hurwitz, J. Synthesis of DNA by DNA polymerase ɛ in vitro. J. Biol. Chem. 266, 22707–22717 (1991).

    CAS  PubMed  Google Scholar 

  37. Mok, M. & Marians, K.J. The Escherichia coli preprimosome and DNA B helicase can form replication forks that move at the same rate. J. Biol. Chem. 262, 16644–16654 (1987).

    CAS  PubMed  Google Scholar 

  38. Kaplan, D.L. & O'Donnell, M. DnaB drives DNA branch migration and dislodges proteins while encircling two DNA strands. Mol. Cell 10, 647–657 (2002).

    Article  CAS  PubMed  Google Scholar 

  39. Wu, C.A., Zechner, E.L. & Marians, K.J. Coordinated leading- and lagging-strand synthesis at the Escherichia coli DNA replication fork. I. Multiple effectors act to modulate Okazaki fragment size. J. Biol. Chem. 267, 4030–4044 (1992).

    CAS  PubMed  Google Scholar 

  40. Dua, R., Levy, D.L. & Campbell, J.L. Analysis of the essential functions of the C-terminal protein/protein interaction domain of Saccharomyces cerevisiae pol ɛ and its unexpected ability to support growth in the absence of the DNA polymerase domain. J. Biol. Chem. 274, 22283–22288 (1999).

    Article  CAS  PubMed  Google Scholar 

  41. Kesti, T., Flick, K., Keranen, S., Syvaoja, J.E. & Wittenberg, C. DNA polymerase ɛ catalytic domains are dispensable for DNA replication, DNA repair, and cell viability. Mol. Cell 3, 679–685 (1999).

    Article  CAS  PubMed  Google Scholar 

  42. Burgers, P.M. Saccharomyces cerevisiae replication factor C. II. Formation and activity of complexes with the proliferating cell nuclear antigen and with DNA polymerases δ and ɛ. J. Biol. Chem. 266, 22698–22706 (1991).

    CAS  PubMed  Google Scholar 

  43. Johansson, E., Garg, P. & Burgers, P.M. The Pol32 subunit of DNA polymerase δ contains separable domains for processive replication and proliferating cell nuclear antigen (PCNA) binding. J. Biol. Chem. 279, 1907–1915 (2004).

    Article  CAS  PubMed  Google Scholar 

  44. Langston, L.D. & O'Donnell, M. DNA polymerase delta is highly processive with proliferating cell nuclear antigen and undergoes collision release upon completing DNA. J. Biol. Chem. 283, 29522–29531 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chilkova, O. et al. The eukaryotic leading and lagging strand DNA polymerases are loaded onto primer-ends via separate mechanisms but have comparable processivity in the presence of PCNA. Nucleic Acids Res. 35, 6588–6597 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Isoz, I., Persson, U., Volkov, K. & Johansson, E. The C-terminus of Dpb2 is required for interaction with Pol2 and for cell viability. Nucleic Acids Res. 40, 11545–11553 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Yoder, B.L. & Burgers, P.M. Saccharomyces cerevisiae replication factor C. I. Purification and characterization of its ATPase activity. J. Biol. Chem. 266, 22689–22697 (1991).

    CAS  PubMed  Google Scholar 

  48. Shibahara, K. & Stillman, B. Replication-dependent marking of DNA by PCNA facilitates CAF-1-coupled inheritance of chromatin. Cell 96, 575–585 (1999).

    Article  CAS  PubMed  Google Scholar 

  49. Balakrishnan, L. & Bambara, R.A. Okazaki fragment metabolism. Cold Spring Harb. Perspect. Biol. 5, a010173 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Garg, P., Stith, C.M., Sabouri, N., Johansson, E. & Burgers, P.M. Idling by DNA polymerase δ maintains a ligatable nick during lagging-strand DNA replication. Genes Dev. 18, 2764–2773 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kelman, Z., Naktinis, V. & O'Donnell, M. Radiolabeling of proteins for biochemical studies. Methods Enzymol. 262, 430–442 (1995).

    Article  CAS  PubMed  Google Scholar 

  52. Sadowski, I., Su, T.C. & Parent, J. Disintegrator vectors for single-copy yeast chromosomal integration. Yeast 24, 447–455 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. Henricksen, L.A., Umbricht, C.B. & Wold, M.S. Recombinant replication protein A: expression, complex formation, and functional characterization. J. Biol. Chem. 269, 11121–11132 (1994).

    CAS  PubMed  Google Scholar 

  54. Georgescu, R.E., Kurth, I. & O'Donnell, M.E. Single-molecule studies reveal the function of a third polymerase in the replisome. Nat. Struct. Mol. Biol. 19, 113–116 (2012).

    Article  CAS  Google Scholar 

  55. Yao, N. et al. Replication factor C clamp loader subunit arrangement within the circular pentamer and its attachment points to proliferating cell nuclear antigen. J. Biol. Chem. 278, 50744–50753 (2003).

    Article  CAS  PubMed  Google Scholar 

  56. Finkelstein, J., Antony, E., Hingorani, M.M. & O'Donnell, M. Overproduction and analysis of eukaryotic multiprotein complexes in Escherichia coli using a dual-vector strategy. Anal. Biochem. 319, 78–87 (2003).

    Article  CAS  PubMed  Google Scholar 

  57. Chilkova, O., Jonsson, B.H. & Johansson, E. The quaternary structure of DNA polymerase epsilon from Saccharomyces cerevisiae. J. Biol. Chem. 278, 14082–14086 (2003).

    Article  CAS  PubMed  Google Scholar 

  58. Langston, L.D. & O'Donnell, M. DNA polymerase δ is highly processive with proliferating cell nuclear antigen and undergoes collision release upon completing DNA. J. Biol. Chem. 283, 29522–29531 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors are grateful for support from the US National Institutes of Health (GM38839) and Howard Hughes Medical Institute to M.E.O. The authors thank A. Tackett and B. Chait (Rockefeller University) for yeast strain OY001.

Author information

Authors and Affiliations

Authors

Contributions

R.E.G., L.L. and M.E.O. conceived the project and designed the experiments. R.E.G., L.L., N.Y.Y. and T.A. executed the experiments and analyzed the results together with M.E.O. L.D.L., O.Y. and J.F. designed the yeast strain constructions, and O.Y. and J.F. carried them out. R.E.G., L.D.L., O.Y. and D.Z. carried out protein purifications. M.E.O. and R.E.G. wrote the manuscript with input from L.D.L.

Corresponding author

Correspondence to Mike E O'Donnell.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 SDS-PAGE of CMG and helicase activity.

(a) Purified CMG (4 μg) was analyzed in a 8% SDS polyacrylamide gel. (b) The bar graph indicates the stoichiometry of Mcm2-7, Cdc45 and GINS subunits within the CMG prep (MCM2-7 is set at 1.0), as determined by comparing their intensities with protein standards of known amounts of each of these complexes determined by Bradford stain and loaded in the same gel. (c) Helicase activity of CMG was determined using an end-labeled forked DNA substrate explained in Extended Experimental Procedures. The 144 μl helicase assay contained 1.9 pmol CMG and 72 fmol radiolabeled fork. Reactions were performed at 30 °C in the presence of 2 mM ATP. The 27% unwinding observed using this 26-fold excess of yeast CMG over DNA is comparable to the 24% unwinding observed using a 40-fold excess of Drosophila CMG over a similar DNA substrate (and in similar conditions). Thus the yeast CMG displays comparable specific activity to Drosophila CMG (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, 247-58 (2010)).

Supplementary Figure 2 Analysis of 3-kb forked DNA-replication products by CMG–Pol ɛ.

Reactions were performed using α − 32P-dCTP incorporation, and therefore the length of DNA products were corrected for the increase in 32P-dCMP incorporation with DNA length as in Kurth, I., Georgescu, R.E. & O'Donnell, M.E. A solution to release twisted DNA during chromosome replication by coupled DNA polymerases. Nature 496, 119-22 (2013). The analysis corresponds to the experiment of Fig. 1. Scans of 2′ (a) and 4′ (b) replication reactions are plotted at the same scale. The lane profiles were normalized to the corresponding molecular weight at each pixel in order to correct for the fact that longer products incorporate more radiolabel. (c) Normalized lane profiles for the 8′, 12′ and 20′ reactions; the inset is a plot of the total DNA synthesis (area under the scans) determined by fitting the data to a double Gaussian distribution (the error bars represent the standard errors of parameters obtained from the fitting). The dashed line denotes the end-labeled product due to polymerase/exonuclease idling activity of Pol ɛ (reactions missing CMG and ATP). (d) Lane profiles of replication reactions in which RFC/PCNA or RPA are omitted. The inset histogram shows the relative percentage of the accumulated 3kb product in the complete replication reaction versus the incomplete reactions.

Supplementary Figure 3 RPA inhibits fork unwinding if it is added before CMG.

Scheme of the unwinding reactions; CMG is either: (a) preincubated with the DNA fork for 10 min at 30 °C before adding RPA, or (b) added to the DNA fork after RPA. The unwinding reaction is initiated upon the addition of 1 mM ATP. (c) RPA inhibits binding but not unwinding by CMG. Native PAGE depicting time course reactions (2’ 10’ and 20’) performed as illustrated in the reaction schemes and described in Extended Experimental Procedures. In separate 55 μl reactions, 0.7 pmol CMG was pre-incubated with 27.5 fmol DNA at 30° for 20’ in the presence or absence of 0.55 pmol RPA. Unwinding was initiated by addition of ATP to 1 mM (and RPA where indicated) and 12 μl aliquots were removed at the indicated times after addition of ATP for analysis by native PAGE.

Supplementary Figure 4 Analysis of φX174 ssDNA replication products.

The analysis corresponds to the experiment of Fig. 5. Primed φX174 ssDNA is coated with a single-strand binding protein (E.coli SSB or S. cerevisiae RPA) and preincubated with RFC and PCNA to load the clamp on the primed site as well one of the DNA polymerases (Pol ɛ; or δ). Length of DNA at each time point was assessed by laser scans of the data in Panels A and B. The graphs illustrated in panel A and B are ImageQuant analysis scans (arbitrary units) of 1’ and 2’ (panel a – for Pol ɛ) and 20” and 40” (panel b for Pol δ) of replication reaction products. Scans were analyzed by fitting to a single Gaussian distribution (black lines) and the numbers shown (together with the standard errors of parameters) in either blue or red correspond to the usage of eSSB or yRPA proteins during the replication reactions. Panels (c) and (d): Rates of Pol ɛ and Pol δ extension using either E. coli SSB (blue) or RPA (red).

Supplementary Figure 5 Pol δ takes over PCNA-primed ssDNA from a moving Pol ɛ.

The reaction was performed using PCNA-primed φX174 ssDNA as described in the Extended Experimental Procedures with the following exceptions: 100 nM Pol ɛ was included in the preincubation, and 5 nM Pol δ was added 10 s after DNA synthesis was initiated by adding dNTPs. Time points reflect the time after addition of Pol δ.

Supplementary Figure 6 Purified yeast Pol δ and Pol ɛ.

Pol δ and Pol ɛ were purified as described in Extended Experimental Procedures. 5 μg of the purified protein complexes were separated on a 8 % SDS-PAGE gel and stained with Commassie Brilliant Blue. The positions of the four Pol ɛ, and three Pol δ subunits are indicated to the right of their respective bands in the gel.

Supplementary Figure 7 Uncropped gels for figures in main manuscript.

Uncropped gels of (A) Fig. 2, (B) Fig. 4 and (C) Fig. 5, panel B of the main manuscript. For the gels in Figures 2 and 4 we cropped out the Molecular Weight Markers since their intensities were overpowering the signal of the experimental reactions. The gel was cropped in Figure 5 due to losses that resulted in loading un-equivalent reaction product at the 10 min time points of Pol ɛ/SSB. However, the 1,2,5 min time points revealed the full time course to final product in these reactions.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Table 1 (PDF 35648 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Georgescu, R., Langston, L., Yao, N. et al. Mechanism of asymmetric polymerase assembly at the eukaryotic replication fork. Nat Struct Mol Biol 21, 664–670 (2014). https://doi.org/10.1038/nsmb.2851

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2851

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