Elsevier

DNA Repair

Volume 31, July 2015, Pages 80-90
DNA Repair

Association of the Rad9–Rad1–Hus1 checkpoint clamp with MYH DNA glycosylase and DNA

https://doi.org/10.1016/j.dnarep.2015.05.004Get rights and content

Highlights

  • Individual Rad9–Rad1–Hus1 (9–1–1) subunits play distinct roles in BER.

  • The interdomain connecting loop of Hus1 is a key determinant of MYH binding.

  • The K136A mutant demonstrated that Hus1 binding is uncoupled from MYH activation.

  • The order of DNA binding affinity is: 91−266–1–1  Rad91−266

  • Hus1  Rad1.

  • Preferential binding of 9–1–1 to 5′-recessed DNA is independent of clamp loader.

Abstract

Cell cycle checkpoints provide surveillance mechanisms to activate the DNA damage response, thus preserving genomic integrity. The heterotrimeric Rad9–Rad1–Hus1 (9–1–1) clamp is a DNA damage response sensor and can be loaded onto DNA. 9–1–1 is involved in base excision repair (BER) by interacting with nearly every enzyme in BER. Here, we show that individual 9–1–1 components play distinct roles in BER directed by MYH DNA glycosylase. Analyses of Hus1 deletion mutants revealed that the interdomain connecting loop (residues 134–155) is a key determinant of MYH binding. Both the N-(residues 1–146) and C-terminal (residues 147–280) halves of Hus1, which share structural similarity, can interact with and stimulate MYH. The Hus1K136A mutant retains physical interaction with MYH but cannot stimulate MYH glycosylase activity. The N-terminal domain, but not the C-terminal half of Hus1 can also bind DNA with moderate affinity. Intact Rad9 expressed in bacteria binds to and stimulates MYH weakly. However, Rad91−266 (C-terminal truncated Rad9) can stimulate MYH activity and bind DNA with high affinity, close to that displayed by heterotrimeric 91−266–1–1 complexes. Conversely, Rad1 has minimal roles in stimulating MYH activity or binding to DNA. Finally, we show that preferential recruitment of 91−266–1–1 to 5′-recessed DNA substrates is an intrinsic property of this complex and is dependent on complex formation. Together, our findings provide a mechanistic rationale for unique contributions by individual 9–1–1 subunits to MYH-directed BER based on subunit asymmetry in protein–protein interactions and DNA binding events.

Introduction

Cell cycle checkpoints provide surveillance mechanisms to activate the DNA damage response (DDR), thus preserving genomic integrity [1], [2]. Activation of DDR leads to cell cycle arrest (which allows time for DNA repair) and enhances DNA repair. When DNA damage is extreme, apoptosis is triggered. The checkpoint system includes an array of proteins that function as sensors, transducers, and effectors [3], [4], [5], [6]. The heterotrimeric Rad9/Rad1/Hus1 (9–1–1) complex is a DDR sensor [7], [8] and is loaded onto DNA by the Rad17-RFC2–5 clamp loader [9], [10], [11], [12]. 9–1–1 is essential for embryonic development, genomic stability, and telomere integrity [13], [14], [15], [16]. Besides serving as a damage sensor [17], 9–1–1 is involved in many DNA metabolisms [14] including base excision repair (BER) (reviewed in [18]). Remarkably, 9–1–1 interacts with nearly every enzyme in BER and is proposed to constitute a platform to coordinate BER. Many BER proteins interact selectively with specific subunit(s) of 9–1–1 [19], [20], [21].

The first step in BER is carried out by a DNA glycosylase, which cleaves damaged or mismatched bases. MYH (also called MUTYH) DNA glycosylase excises adenine when it is mispaired with 8-oxo-7,8-dihydroguanine (G0) or guanine and thus reduces G:C to T:A mutations [22], [23], [24]. The resulting apurinic/apyrimidinic (AP) site is processed by AP-endonuclease 1 (APE1), allowing the downstream BER enzymes to complete the DNA repair process. Mutations in the human MYH (hMYH) gene can lead to colorectal cancer (as in MYH-associated polyposis or MAP) [25], while APE1 is essential for cell viability [26]. MYH contains unique motifs that mediate interactions with partner proteins involved in DNA replication, mismatch repair, and DDR (reviewed in [22], [23]). We have shown that the interdomain connector (IDC) located between the N- and C-terminal domains of hMYH is uniquely oriented [27] to interact with Hus1 [21] and APE1 [28].

The ring structure of 9–1–1 [29], [30], [31] is remarkably similar to that of the proliferating cell nuclear antigen (PCNA) [32], [33], [34]. Each 9–1–1 subunit folds into two globular domains linked by an interdomain connecting loop (IDCL) (Fig. 1A). According to the PCNA–DNA structure [35], the 9–1–1 ring is supposed to encircle double-stranded DNA [30]. Although the three subunits of the 9–1–1 complex are structurally similar, they exhibit key differences. These differences are most pronounced in the IDCLs between their N- and C-terminal domains [29], [30], [31]. These structural distinctions between 9–1–1 components have been suggested to dictate protein-binding specificity for individual subunits. For example, hMYH and hAPE1 bind preferentially to the Hus1 subunit [20], [21]. Functionally, the Hus1 subunit alone can stimulate MYH activity [21]. In this paper, we constructed a panel of Hus1 mutant proteins to identify domains required for binding MYH, stimulating MYH glycosylase activity, and binding DNA. Subsequently we tested the roles of other 9–1–1 components in MYH activation and DNA binding. This systematic and quantitative biochemical strategy revealed key differences in the ability of 9–1–1 subunits to bind DNA and functionally interact with MYH, thus supporting a model whereby each subunit of 9–1–1 plays a distinct and directed role in BER.

Section snippets

Glutathione-S-transferase (GST)-tagged Hus1 protein constructs

The plasmid pGEX-3X-hHus1 containing GST-tagged hHus1 was obtained from Dr. A.E. Tomkinson at the University of New Mexico. GST fusions incorporating hHus1 deletion constructs were made by polymerase chain reaction (PCR) using primers listed in Table S1 in the Supplementary material. The PCR products were digested with BamHI and SalI and ligated into the BamHI-XhoI-digested pGEX-4T-2 vector (GE Healthcare). The K136A and V137A mutants of the hHus1 gene were constructed by QuickChange

MYH binds to the interdomain connecting loop of Hus1

We have shown that hMYH physically interacts with 9–1–1 mainly via the Hus1 subunit [21]. The Hus1 binding site is located within the IDC region (residues 295–350) of hMYH [21]. However, the regions of hHus1 protein engaged in the physical interaction with MYH have not been determined. Thus, we generated a panel of hHus1 deletion constructs fused to GST (Fig. 3A and B) to map the MYH interacting region. Because MYH binds preferentially to the Hus1 subunit over Rad1 and Rad9 [21] and the IDCL

Discussion

In the present study, we reveal the differential roles played by the 9–1–1 subunits in physical and functional interactions with the MYH glycosylase and DNA binding. The three subunits of 9–1–1 are structurally similar, but exhibit key differences [29], [30], [31] (Fig. 1A). This structural asymmetry correlates with an asymmetry in protein–protein interactions (reviewed in [18]). In particular, hMYH and hAPE1 bind preferentially to the Hus1 subunit [20], [21]. The IDCL domain of Hus1 is

Conflict of interest statement

The authors declare that there is no conflict of interest.

Acknowledgements

We thank Dr. Yusaku Nakabeppu (Kyushu University, Japan) for the mouse Myh clone and Dr. Alan Tomkinson (University of New Mexico) for the cDNA of hRad9 and hRad1. We thank Dr. Eric Toth (University of Maryland Medical School) for critical reading of this manuscript and constructing Fig. 1B. This work was supported by the National Cancer Institute of the National Institute of Health grants R01-CA78391 and S10-OD011969 to A.L. and R01-CA102428 to G.M.W.

References (51)

  • M. Bradford

    A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein–dye binding

    Anal. Biochem.

    (1976)
  • S. Hirano et al.

    Mutator phenotype of MUTYH-null mouse embryonic stem cells

    J. Biol. Chem.

    (2003)
  • C.Y. Lee et al.

    An Escherichia coli MutY mutant without the six-helix barrel domain is a dimer in solution and assembles cooperatively into multisubunit complexes with DNA

    J. Biol. Chem.

    (2004)
  • D.M. Jameson et al.

    Fluorescence anisotropy applied to biomolecular interactions

    Methods Enzymol.

    (1995)
  • P. Roos-Mattjus et al.

    Phosphorylation of human Rad9 is required for genotoxin-activated checkpoint signaling

    J. Biol. Chem.

    (2003)
  • R.E. Georgescu et al.

    Structure of a sliding clamp on DNA

    Cell

    (2008)
  • D.Y. Chang et al.

    The role of MutY homolog (Myh1) in controlling the histone deacetylase Hst4 in the fission yeast Schizosaccharomyces pombe

    J. Mol. Biol.

    (2011)
  • B.J. Hwang et al.

    Mammalian MutY homolog (MYH or MUTYH) protects cells from oxidative DNA damage

    DNA Repair (Amst.)

    (2014)
  • J. Bartek et al.

    Checking on DNA damage in S phase

    Nat. Rev. Mol. Cell Biol.

    (2004)
  • A. Sancar et al.

    Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints

    Annu. Rev. Biochem.

    (2004)
  • S. Matsuoka et al.

    ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage

    Science

    (2007)
  • D. Shechter et al.

    ATR and ATM regulate the timing of DNA replication origin firing

    Nat. Cell Biol.

    (2004)
  • L. Zou et al.

    Sensing DNA damage through ATRIP recognition of RPA–ssDNA complexes

    Science

    (2003)
  • B.B. Zhou et al.

    The DNA damage response: putting checkpoints in perspective

    Nature

    (2000)
  • R.P. St Onge et al.

    The human G2 checkpoint control protein hRAD9 is a nuclear phosphoprotein that forms complexes with hRAD1 and hHUS1

    Mol. Biol. Cell

    (1999)
  • Cited by (0)

    View full text