Elsevier

Free Radical Biology and Medicine

Volume 129, December 2018, Pages 97-106
Free Radical Biology and Medicine

Original article
Lack of superoxide dismutase in a rad51 mutant exacerbates genomic instability and oxidative stress-mediated cytotoxicity in Saccharomyces cerevisiae

https://doi.org/10.1016/j.freeradbiomed.2018.09.015Get rights and content

Highlights

  • SOD1 inhibition can be used for selective killing of cancer cells deficient in homologous recombination pathway.

  • Deletion of RAD51 and SOD1 does not show synthetic lethality in budding yeast.

  • Sod1 deficiency aggravates genomic instability in conjunction with the absence of Rad51.

  • Accumulation of Rad51 deficiency-mediated DSB lesions increases intracellular ROS levels in the absence of Sod1.

  • DSB repair pathways and ROS response signaling have significant mutual genetic crosstalk.

Abstract

A genetic analysis of synthetic lethal interactions in yeast revealed that the mutation of SOD1, encoding an antioxidant enzyme that scavenges superoxide anion radical, impaired the growth of a set of mutants defective in homologous recombination (HR) pathway. Hence, SOD1 inhibition has been proposed as a promising approach for the selective killing of HR-deficient cancer cells. However, we show that the deletion of RAD51 and SOD1 is not synthetic lethal but displays considerably slow growth and synergistic sensitivity to both reactive oxygen species (ROS)- and DNA double-strand break (DSB)-generating drugs in the budding yeast Saccharomyces cerevisiae. The function of Sod1 in regard to Rad51 is dependent on Ccs1, a copper chaperone for Sod1. Sod1 deficiency aggravates genomic instability in conjunction with the absence of Rad51 by inducing DSBs and an elevated mutation frequency. Inversely, lack of Rad51 causes a Sod1 deficiency-derived increase of intracellular ROS levels. Taken together, our results indicate that there is a significant and specific crosstalk between two major cellular damage response pathways, ROS signaling and DSB repair, for cell survival.

Introduction

Aerobic organisms are unavoidably exposed to intrinsic and extrinsic stress that threatens genomic integrity and cell viability throughout their lifespan. These insults include direct DNA strand breaks and the frequent occurrence of cytotoxic damage mediated by reactive oxygen species (ROS). Although physical DNA damage, such as single-strand breaks (SSBs) and double-strand breaks (DSBs), is mostly responsible for cell lethality, oxidative DNA lesions created by endogenous ROS and oxidizing chemicals are also implicated in a large proportion of structural mutations and genomic instability that might eventually cause cancer, aging, and a variety of degenerative disorders [1], [2], [3]. To deal with these adverse conditions, cells have evolved highly coordinated processes such as various oxidative stress signaling systems and DNA damage response (DDR) pathways.

Mounting evidence suggests a highly involved mutual influence between ROS signaling and DDR pathways. ROS-induced DNA adducts, such as modified bases and abasic DNA sites, occur as often as ~10,000 times per cell every day [4]. Oxidative DNA damage, including the occasional production of SSBs, is highly mutagenic but lesions are not considered fatal because they would be efficiently processed with comparative ease by base excision repair (BER) and nucleotide excision repair (NER) [5], [6]. Cells deficient in the BER and NER pathway show not only elevated genomic instability with a higher mutation frequency but also greatly induced intracellular ROS levels, indicating that accumulated DNA damage is inversely capable of causing oxidative stress in the cell [7], [8]. Unrepaired alkylating DNA damage induces the nuclear accumulation of Yap1, a ROS-responsive transcription factor, which reveals a mechanistic link between BER-mediated DNA damage repair and oxidative stress signaling [9].

DNA DSBs arise approximately 10–50 per cell each day [10]. Although much less frequent than oxidative DNA damage or SSBs, DSBs are highly toxic to genome fidelity eventually leading to cell death because they can cause detrimental deletions, translocations, and critical fusions in the chromosome. DSBs in the G1 phase can be repaired by non-homologous end joining (NHEJ) mainly due to the compact chromatin and lack of sister chromatids. During relatively long S and G2 phases, however, replication-associated homologous recombination (HR) becomes predominant and crucial for DSB repair in yeast and especially in mammalian cancer cells, which are typified with uncontrolled cell division [11], [12]. Rad51, the homolog of E. coli RecA recombinase and a member of Rad52 epistasis group, is a central player in the HR pathway, searching for homology and strand pairing for recombination. Replication defects and chromosomal instability are induced in rad51 mutants [13], [14].

A genome-wide analysis of synthetic lethal interactions in budding yeast has disclosed novel genetic interactions among functionally independent genes of key proteins in the HR pathway and responders to oxidative stress such as Sod1 and Tsa1 [15]. Synthetic lethality refers to a type of genetic interaction that occurs when a combination of loss-of-function perturbations in two or more genes results in cell death, whereas a mutation in only one of these genes does not affect viability at all [16]. The synthetic lethal paradigm between the Rad52 epistasis group members and Sod1 has been proposed as a promising anticancer therapeutic since selective apoptosis was induced in RAD54B-deficient human colorectal cancer cells by Sod1 inhibition [17], [18]. In addition to its conventional action as a superoxide scavenger, Sod1 plays an important role as a transcription factor in the nucleus through direct phosphorylation by Dun1, an effector kinase in damage checkpoint signaling [19]. Moreover, cells with no functional ataxia-telangiectasia mutated (ATM) protein kinase are hypersensitive to oxidative stress, and the damage checkpoint activation mediated by Mec1, a yeast ATR homolog, is downregulated by the lack of Sod1, suggesting the existence of genetic and biochemical connections between cellular redox regulators and DDR pathways [20], [21], [22]. In a previous report, we revealed that Yap1 and Skn7, two transcription factors in charge of the oxidative stress response with various antioxidant enzymes, are highly implicated in the Rad51-mediated HR pathway in terms of a strategic defense mechanism against oxidative stress and DNA damaging insults [23].

In this study, unlike in previous reports derived from a high-throughput dataset, we show that the additional deletion of SOD1, a downstream target of Yap1 and Skn7, in an HR-defective rad51 mutant background is not synthetic lethal, but adversely affects the growth and genomic integrity of yeast cells with increased mutation frequency and activated damage checkpoint signaling. Importantly, we find that this novel role of Sod1 associated with Rad51 might be dependent on its conventional catalytic activity as an antioxidant and on the capability of intracellular translocation between cytosol and the nucleus. Accumulation of DSB damage in the rad51 sod1 double mutant inversely induces elevation of intracellular ROS levels. Altogether, our results suggest a specific genetic association between antioxidant defense signaling and the DNA DSB repair pathway, which provides a cooperative mechanism to maintain genome integrity and cell survival.

Section snippets

Strains, plasmids and growth media

All of the strain set used in this study are isogenic derivatives of S. cerevisiae BY4741 (MATa his3Δ1 leu2Δ0 lys2Δ0 ura3Δ0) obtained from Yeast Knockout (YKO) collection (YSC1053 glycerol stock, Thermo Scientific). The genotypes of all strains used in this study are listed in Supplementary Table 1. The strains with C-terminally GFP-fused proteins, Rad52-GFP and Sod1-GFP, were constructed by oligonucleotide-directed in-frame tagging method as previously described [24]. The strains with Sod1AA

Deletion of RAD51 and SOD1 is not synthetic lethal but shows severely delayed growth and synergistic drug sensitivity

Global genetic analyses of synthetic lethal interactions in yeast have reported that a sod1 mutant impaired the growth of mutants defective in HR such as rad50, rad51, rad52, and rad54 [15], [18]. To analyze the terminal phenotype of cells with neither Sod1 nor HR activity, we created a conditional mutant in which RAD51 is removed and the SOD1 ORF is put under the control of a tetO7 promoter so that the expression could be shut down by doxycycline treatment [29]. Unexpectedly, we found that

Discussion

An extensive search in the yeast genome has been suggested for a highly linked set of synthetic lethal partners whose human counterparts are frequently mutated in human cancer to develop potential drugs for anticancer therapy [42], [43]. Sod1 inhibition in RAD54B-deficient human colorectal cancer cells is a critical approach of selective killing of cancer cells exploiting the synthetic lethal principle between representative genes in the HR pathway and the oxidative stress response system [15],

Acknowledgements

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2016R1A6A1A03007648 and 2017R1D1A1A09000197).

References (47)

  • S.B. Haase et al.

    Flow cytometric analysis of DNA content in budding yeast

    Methods Enzymol.

    (1997)
  • V.C. Culotta et al.

    The copper chaperone for superoxide dismutase

    J. Biol. Chem.

    (1997)
  • L.A. Sturtz et al.

    Superoxide dismutase null mutants of baker's yeast, Saccharomyces cerevisiae

    Methods Enzymol.

    (2002)
  • A. Ciccia et al.

    The DNA damage response: making it safe to play with knives

    Mol. Cell

    (2010)
  • F.D. Sweeney et al.

    Saccharomyces cerevisiae Rad9 acts as a Mec1 adaptor to allow Rad53 activation

    Curr. Biol.

    (2005)
  • V. Pawar et al.

    Checkpoint kinase phosphorylation in response to endogenous oxidative DNA damage in repair-deficient stationary-phase Saccharomyces cerevisiae

    Mech. Ageing Dev.

    (2009)
  • W.H. Chung

    Mechanisms of a novel anticancer therapeutic strategy involving atmospheric pressure plasma-mediated apoptosis and DNA strand break formation

    Arch. Pharm. Res.

    (2016)
  • P.A. Savitsky et al.

    Redox regulation of Cdc25C

    J. Biol. Chem.

    (2002)
  • A. Ashworth et al.

    Genetic interactions in cancer progression and treatment

    Cell

    (2011)
  • S.M.B. Nijman

    Synthetic lethality: general principles, utility and detection using genetic screens in human cells

    FEBS Lett.

    (2011)
  • J.H.J. Hoeijmakers

    DNA damage, aging, and cancer

    N. Engl. J. Med.

    (2009)
  • J.P. Melis et al.

    Oxidative DNA damage and nucleotide excision repair

    Antioxid. Redox Signal.

    (2013)
  • A. Mehta et al.

    Sources of DNA double-strand breaks and models of recombinational DNA repair

    Cold Spring Harb. Perspect. Biol.

    (2014)
  • Cited by (12)

    • SOD1 mutations cause hypersensitivity to high-pressure-induced oxidative stress in Saccharomyces cerevisiae

      2022, Biochimica et Biophysica Acta - General Subjects
      Citation Excerpt :

      To examine whether failure to maintain the genome impaired sod1∆ mutant growth, we compared mutation rates between wild type and sod1∆ mutants using exponentially growing cells (OD600 = 0.1–0.5) cultured in SC medium under 0.1 MPa or 25 MPa for 24 h. After decompression, the mutation rates were measured by a forward mutation assay of the CAN1 locus based on detecting the CAN1 loss-of-function mutation on canavanine-containing agar plates [51–53]. The spontaneous mutation rate in the sod1∆ mutant was ~2.3-fold higher than that in the wild type (Fig. 7).

    • Integrative transcriptomic-proteomic analysis revealed the flavor formation mechanism and antioxidant activity in rice-acid inoculated with Lactobacillus paracasei and Kluyveromyces marxianus

      2021, Journal of Proteomics
      Citation Excerpt :

      The protein CCS1 was important for superoxide dismutase. A recent report demonstrated that the function of superoxide dismutase (SOD1) in regard to Rad51 was dependent on CCS1, a copper chaperone for SOD1 [47]. In addition, we found that the gene FMP46/BAO41318 encoding putative redox protein was 2.943-fold up-regulated, perhaps indicating that in addition to CCS1, another protein could increase the antioxidant ability.

    • Response of Trametes hirsuta to hexavalent chromium promotes laccase-mediated decolorization of reactive black 5

      2020, Ecotoxicology and Environmental Safety
      Citation Excerpt :

      To evaluate the influence of oxidative stress on laccase expression, DNA-damaging reagent MMS was continuously added into actively growing 6-day-old cultures of T. hirsuta TH315 for 7 days at a concentration of 0.02% (w/v) without Cr(VI) exposure. The selection of the chemical reagent in this experiment was designed based on the previous studies (Choi et al., 2018; Rowe et al., 2008), as well as the drug dosage of MMS (Tsang et al., 2014). The supernatants were collected for laccase activity determination.

    View all citing articles on Scopus
    View full text