Elsevier

DNA Repair

Volume 9, Issue 10, 5 October 2010, Pages 1098-1111
DNA Repair

Stable interactions between DNA polymerase δ catalytic and structural subunits are essential for efficient DNA repair

https://doi.org/10.1016/j.dnarep.2010.07.013Get rights and content

Abstract

Eukaryotic DNA polymerase δ (Pol δ) activity is crucial for chromosome replication and DNA repair and thus, plays an essential role in genome stability. In Saccharomyces cerevisiae, Pol δ is a heterotrimeric complex composed of the catalytic subunit Pol3, the structural B subunit Pol31, and Pol32, an additional auxiliary subunit. Pol3 interacts with Pol31 thanks to its C-terminal domain (CTD) and this interaction is of functional importance both in DNA replication and DNA repair. Interestingly, deletion of the last four C-terminal Pol3 residues, LSKW, in the pol3-ct mutant does not affect DNA replication but leads to defects in homologous recombination and in break-induced replication (BIR) repair pathways. The defect associated with pol3-ct could result from a defective interaction between Pol δ and a protein involved in recombination. However, we show that the LSKW motif is required for the interaction between Pol3 C-terminal end and Pol31. This loss of interaction is relevant in vivo since we found that pol3-ct confers HU sensitivity on its own and synthetic lethality with a POL32 deletion. Moreover, pol3-ct shows genetic interactions, both suppression and synthetic lethality, with POL31 mutant alleles. Structural analyses indicate that the B subunit of Pol δ displays a major conserved region at its surface and that pol31 alleles interacting with pol3-ct, correspond to substitutions of Pol31 amino acids that are situated in this particular region. Superimposition of our Pol31 model on the 3D architecture of the phylogenetically related DNA polymerase α (Pol α) suggests that Pol3 CTD interacts with the conserved region of Pol31, thus providing a molecular basis to understand the defects associated with pol3-ct. Taken together, our data highlight a stringent dependence on Pol δ complex stability in DNA repair.

Introduction

Central to the chromosomal DNA replication apparatus in eukaryotic cells are three essential DNA polymerases, namely DNA polymerase α, δ and ɛ [1]. The primase subunit of the Pol α/primase complex initiates synthesis by oligomerizing short RNA primers on both leading and lagging strands. These primers are initially extended by Pol α and subsequently transferred to processive polymerases Pol ɛ and Pol δ. In contrast to the quite obvious role of Pol α, the specific roles for Pol ɛ and Pol δ in DNA replication were much debated for years. However, recent evidence strongly supports a model of the replication fork wherein the leading and the lagging strand templates are primarily copied by Pol ɛ and Pol δ, respectively [2], [3].

Pol α, Pol δ and Pol ɛ belong to the B class of DNA polymerases [4] and their catalytic subunits are phylogenetically related and conserved from yeast to humans [5], [6]. Similarly, among the different accessory subunits of the replicative Pols, only the so-called B subunit [7], is clearly conserved in eukaryotic organisms [8]. Interestingly, Pol α, Pol δ and Pol ɛ share unifying features of their subunit organization that again underlines their evolutionary relationship. Indeed, the sequence conservation of the catalytic subunit extends past the polymerase fold to a cysteine-rich C-terminal domain (CTD) that contains two zinc fingers. A large body of experimental evidence has highlighted that this CTD interacts with the B subunit and that this interaction is of functional importance in Pol α [9], Pol δ [10], [11] and Pol ɛ [12]. Recently, a 3D architecture of Pol α has been published that allows a clearer overall picture of the catalytic and B subunit interactions for all three replicative polymerases [13]. An interesting conclusion drawn from this structural analysis is that it defines the C-terminal region of the replicative Pols α, δ and ɛ as an independently folded protein domain, separate from the polymerase fold.

DNA polymerase δ from Saccharomyces cerevisiae, has three subunits: Pol3/Cdc2, the catalytic subunit of 125 kDa that contains the polymerase and 3′ to 5′ exonuclease active site domains; Pol31/Hys2, the essential structural B subunit of 55 kDa; Pol32, the additional C subunit of 40 kDa [14]. The enzymes from Schizosaccharomyces pombe and humans have an additional small fourth subunit that functions to stabilize the complex [15], [16], [17]. The enzymes from the three different sources show roughly similar structure characteristics [7] and contain one of each of the subunits, i.e., they are a monomeric catalytic complex [18], [19]. The catalytic and the B subunits form a stable complex (as discussed above), to which the C subunit is tethered solely via interactions with the B subunit [14], [20]. The interaction between the B subunit and the C subunit of Pol δ participates to the functional integrity of the whole complex. Indeed, while POL32 is dispensable for growth in S. cerevisiae, the POL32 deletion mutant nevertheless shows poor growth and synthetic lethality with numerous genes that function in DNA metabolism [14], [21]. In addition, the orthologous S. pombe Cdc27 gene is essential for growth [18], [22]. In both S. cerevisiae and S. pombe, the C subunit interacts with the B subunit via its N-terminal part, whereas its C-terminal part contains a conserved PCNA-binding motif and a motif that mediates interaction with Pol α [18], [23], [24]. It is noteworthy that deletion of the POL32 N-terminal end exhibits the same phenotype than the complete deletion [24]. Using two-hybrid screening, the human C subunit, p66, has been shown to contain p50 (the human Pol δ B subunit) and PCNA binding domains within the 144 N-terminal and 20 C-terminal amino acids, respectively [25]. Recently, the crystal structure of the complex between p50 and the 144 amino acids N-terminal domain of p66 of human Pol δ has been reported [26]. It appears that p66 makes an intricate complex with p50 and has a significantly large surface area buried at the heterodimer interface. Interestingly, in this study, it is noted that in spite of the formation of a very tight complex between p50 and p66 subunits, the residues involved in dimerization correspond to poorly conserved regions among eukaryotic sequences. The analysis of the secondary structure elements of p50 and p66 reveals, however, that this complex is likely to share the same overall fold with its orthologs [26].

The interaction between the Pol δ subunits plays a crucial role in the stability of the Pol δ complex and allows full proficiency of Pol δ in DNA replication. Interestingly, essential functions of Pol δ in DNA repair are even more sensitive to defective interactions within the complex. This observation comes essentially from the study of Pol δ mutants in S. cerevisiae. The pol3-13 mutation leads to the substitution of a Pol3 cysteine at position 1074 by a serine (C1074S). Pol3 contains a CTD with two zinc fingers, ZnF1 and ZnF2. Residue C1074 is in the last ZnF2 cysteine. Two-hybrid studies have shown that the mutated C1074S ZnF2 module can no longer interact with Pol31 [11]. In vivo, the pol3-13 allele confers thermosensitivity, but at the permissive temperature of 30 °C, it confers UV and γ-rays sensitivity and almost a complete defect in both γ- and UV-induced recombination and UV-induced mutagenesis [10]. Similarly, Pol32 interacts with Pol31 but also with Pol1, the catalytic subunit of Pol α [24], [27] and the translesion DNA polymerase Rev1 [28]. Deletion of POL32 is viable but confers UV sensitivity and abolishes UV-induced mutagenesis [29]. In addition, break-induced replication (BIR) repair is seriously impaired in the pol32Δ mutant and gives rise to aberrant double-strand break (DSB) repair resulting in half crossovers [30], [31], [32]. Interestingly, Rev1 is required for UV-induced mutagenesis [33] while Pol α is involved in BIR [31]. Thus, Pol32 interaction with Rev1 and Pol α could have provided a clear molecular basis to understand the role of Pol32 in UV-induced mutagenesis and BIR. However, the deletion of neither the Pol1 or Rev1 interacting domains of Pol32 leads to defects in the corresponding DNA repair pathway [24], [31]. By contrast, the essential functions of Pol32 in DNA repair are comprised in a small amino-terminal domain of Pol32, not much larger than that necessary to specify the interaction with Pol31, suggesting that the Pol32 subunit mediates its role in UV-induced mutagenesis and BIR through the Pol δ complex [24], [31].

In our previous studies, we described the pol3-ct allele that changes a Leu codon to a stop codon at position +1094 in S. cerevisiae resulting in the loss of the four last C-terminal amino acids (LSKW) of Pol3 [34], [35]. Yeast cells carrying this allele show no obvious defects in genome replication since they are neither thermosensitive (ts) nor mutators and they do not display high levels of spontaneous recombination. In addition, they are not sensitive to UV and γ-rays irradiation. We found, however, that the pol3-ct mutant, although repairing DSB as efficiently as WT, yields shorter recombination intermediates in both meiosis and mitosis. This interesting phenotype of the pol3-ct mutant implies that the role of Pol δ during HR is not restricted to gap filling but participates as well in recombination intermediates extension.

The phenotype associated with the pol3-ct allele raises the attractive possibility that in the pol3-ct mutant, Pol δ fails to interact with a protein involved in recombination intermediates extension. However, in our present study, we provide genetic and structural evidence showing that pol3-ct leads to a destabilized interaction between Pol3 CTD and Pol31. Therefore, we reach the alternative conclusion that DNA repair synthesis is clearly dependent on Pol δ complex stability.

Section snippets

Yeast strains, plasmids and media

All media were prepared as previously described [36]. Strains used in the present study are isogenic derivatives of W303-1A [37] and listed in Supplementary Table S1.

Yeast two-hybrid assay

Strain CTY10-5d (MATa, ade2-101, his3Δ200, leu2Δ1, trp1Δ901, gal4, gal80, URA3::lexAop-lacZ) was used for two-hybrid analysis. Pol31 was fused in frame to the lexA binding domain in plasmid pBTM116, while the Pol3 C-terminal amino acids 1032-1097 were fused to the Gal4 activating domain of pACT2 to yield plasmid pACT2-Pol3-ZnF2 [11]

Loss of the last four amino acids at the C-terminal end of Pol3 impairs the interaction with Pol31

While the pol3-ct mutant shows defects in HR and BIR, it does not display any noticeable dysfunction during genome replication, suggesting that Pol δ works efficiently in this mutant. However, Sanchez Garcia and colleagues demonstrated that the C-terminal end of Pol3 is responsible for a direct interaction with Pol31 [11]. Interestingly, they found that deleting the last 13 amino acids of Pol3 disrupts this interaction. Therefore, we used their two-hybrid assay to determine the role of the last

Pol δ stability is impaired in the pol3-ct mutant

pol3-ct has proven to be a useful allele of the essential POL3 gene to study the role of Pol δ in gene conversion and BIR [32], [34], [35]. The present study of pol3-ct sheds light on the role of the C-terminal end of Pol3 in Pol δ stability and allows comprehending the role of Pol δ in DNA repair. We found that the C-terminal end of Pol3 missing the last four LSKW amino acids poorly interacts with Pol31 in a yeast two-hybrid assay. This result is confirmed by the observation that Pol3 ZnF2

Funding

This work was supported by two grants from the Agence Nationale de la Recherche (ANR-07-BLAN-0350-01 and ANR-09-PIRI-0015-03), by INCa/Canceropole IdF and by the CEA and the CNRS. The funders had no role, data collection and analysis, decision to publish, or preparation of the manuscript.

Competing interests

The authors have declared that no competing interests exist.

Acknowledgements

We thank Dr. Stuart MacNeill for his generous gift of the strain and plasmids used in the yeast two-hybrid assay. We thank Dr. Akio Sugino for his generous gift of yeast strains YNN101 (hys2-2) and YNN102 (pol32::LEU2) and Dr. Dana Branzei for her generous gift of yeast strain KSH542 (hys2-1). We are grateful to S. Boiteux, X. Veaute and E. Coïc for their support and fruitful discussions.

References (53)

  • E. Johansson et al.

    Structure of DNA polymerase delta from Saccharomyces cerevisiae

    J. Biol. Chem.

    (2001)
  • M. Hanna et al.

    Pol32 is required for Pol zeta-dependent translesion synthesis and prevents double-strand breaks at the replication fork

    Mutat. Res.

    (2007)
  • E. Johansson et al.

    The Pol32 subunit of DNA polymerase delta contains separable domains for processive replication and proliferating cell nuclear antigen (PCNA) binding

    J. Biol. Chem.

    (2004)
  • S. Boiteux et al.

    Repair of 8-oxoguanine in Saccharomyces cerevisiae: interplay of DNA repair and replication mechanisms

    Free Radic. Biol. Med.

    (2002)
  • A. Fiser et al.

    Modeller: generation and refinement of homology-based protein structure models

    Methods Enzymol.

    (2003)
  • A. Koc et al.

    Hydroxyurea arrests DNA replication by a mechanism that preserves basal dNTP pools

    J. Biol. Chem.

    (2004)
  • P.M. Burgers et al.

    Structure and processivity of two forms of Saccharomyces cerevisiae DNA polymerase delta

    J. Biol. Chem.

    (1998)
  • T. Ohya et al.

    The DNA polymerase domain of pol(epsilon) is required for rapid, efficient, and highly accurate chromosomal DNA replication, telomere length maintenance, and normal cell senescence in Saccharomyces cerevisiae

    J. Biol. Chem.

    (2002)
  • P. Garg et al.

    DNA polymerases that propagate the eukaryotic DNA replication fork

    Crit. Rev. Biochem. Mol. Biol.

    (2005)
  • Z.F. Pursell et al.

    Yeast DNA polymerase epsilon participates in leading-strand DNA replication

    Science

    (2007)
  • J.C. Weill et al.

    DNA polymerases in adaptive immunity

    Nat. Rev. Immunol.

    (2008)
  • T. Mizuno et al.

    Molecular architecture of the mouse DNA polymerase alpha-primase complex

    Mol. Cell. Biol.

    (1999)
  • L. Giot et al.

    Involvement of the yeast DNA polymerase delta in DNA repair in vivo

    Genetics

    (1997)
  • J. Sanchez Garcia et al.

    The C-terminal zinc finger of the catalytic subunit of DNA polymerase delta is responsible for direct interaction with the B-subunit

    Nucleic Acids Res.

    (2004)
  • S. Klinge et al.

    3D architecture of DNA Pol alpha reveals the functional core of multi-subunit replicative polymerases

    EMBO J.

    (2009)
  • S. Zuo et al.

    DNA polymerase delta isolated from Schizosaccharomyces pombe contains five subunits

    Proc. Natl. Acad. Sci. U.S.A.

    (1997)
  • Cited by (26)

    • Underappreciated Roles of DNA Polymerase δ in Replication Stress Survival

      2021, Trends in Genetics
      Citation Excerpt :

      Although the loss of either of the accessory subunits POLD2 and POLD3 is lethal in mice and humans, only the POL31 deletion has a similarly fatal effect in yeast, at least in the absence of DNA damage. The essential role of Pol31 is unclear; yet, it may mainly have a structural role, ensuring that the catalytic subunit is stable, to facilitate both lagging strand synthesis and long-patch DNA repair [56]. Pol32 is implicated specifically in break-induced fork restart, although Pol31, being essential, is also likely involved in this pathway.

    • POLD1: Central mediator of DNA replication and repair, and implication in cancer and other pathologies

      2016, Gene
      Citation Excerpt :

      A Pol3 mutant (Pol3-ct) was proficient in strand invasion and synthesis of a short tract of DNA but unable to perform extensive DNA synthesis required to complete BIR, with a requirement for POLD3 in BIR also demonstrated in human cells (Smith et al., 2009; Costantino et al., 2014). In addition, deletion of just the four C-terminal residues in Pol3 (Pol3-ct) leads to defects in HR and BIR repair mechanisms (Brocas et al., 2010). In the HEK293 kidney cell line, depletion of p125 blocked cell cycle at G1 and G2/M phases, indicative of DNA damage checkpoint responses, and increased oxidative DNA damage (Song et al., 2015).

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text