Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Sep 1.
Published in final edited form as: Biochim Biophys Acta. 2012 Jun 19;1819(9-10):979–991. doi: 10.1016/j.bbagrm.2012.06.002

Mitochondrial DNA Damage and its Consequences for Mitochondrial Gene Expression

Susan D Cline 1
PMCID: PMC3412069  NIHMSID: NIHMS388439  PMID: 22728831

Abstract

How mitochondria process DNA damage and whether a change in the steady-state level of mitochondrial DNA damage (mtDNA) contributes to mitochondrial dysfunction are questions that fuel burgeoning areas of research into aging and disease pathogenesis. Over the past decade, researchers have identified and measured various forms of endogenous and environmental mtDNA damage and have elucidated mtDNA repair pathways. Interestingly, mitochondria do not appear to contain the full range of DNA repair mechanisms that operate in the nucleus, although mtDNA contains types of damage that are targets of each nuclear DNA repair pathway. The reduced repair capacity may, in part, explain the high mutation frequency of the mitochondrial chromosome. Since mtDNA replication is dependent on transcription, mtDNA damage may alter mitochondrial gene expression at three levels: by causing DNA polymerase γ nucleotide incorporation errors leading to mutations, by interfering with the priming of mtDNA replication by the mitochondrial RNA polymerase, or by inducing transcriptional mutagenesis or premature transcript termination. This review summarizes our current knowledge of mtDNA damage, its repair, and its effects on mtDNA integrity and gene expression.

1. Introduction

Mitochondria harbor the small, circular genome in human cells that is essential for life, but the environment of the mitochondrial matrix is rather inhospitable to the macromolecules that reside there. The oxidative production of ATP required for cellular function also generates reactive oxygen species (ROS) that damage mitochondrial DNA (mtDNA), membrane lipids, and protein [16]. Additionally, environmental chemicals that enter the body can induce mtDNA damage, both by amplifying the production of endogenous DNA lesions and by generating unique DNA adducts [7]. Unlike nuclei, which have multiple mechanisms for DNA repair, mitochondria appear limited in their ability to rectify all of the possible forms of mtDNA damage [8]. Persistent chromosomal damage can impair mitochondrial DNA polymerase γ (pol γ) and RNA polymerase (POLRMT) activity and threaten mtDNA stability and gene expression. The past decade has brought a surge in research efforts to characterize the variety of biomolecular damage in mitochondria, to elucidate the mechanisms of mitochondrial DNA repair, and to determine the effects of DNA damage on the function of mitochondrial polymerases.

2. Oxidative damage in mitochondria

A host of reactive molecules are generated within mitochondria due to the escape of electrons from the transport chain during oxidative phosphorylation [1]. Approximately 0.15% of the molecular oxygen consumed by mitochondria is reduced to superoxide anion (O2·−) [9]. Seven potential sites for matrix O2·− generation exist, with complexes I and III exhibiting the highest rate of O2·− production [3]. Superoxide cannot diffuse through the inner mitochondrial membrane (IMM), so it is confined to the matrix where it can react with Fe-S proteins to liberate iron or be converted to hydrogen peroxide by manganese-superoxide dismutase, Mn-SOD. Hydrogen peroxide may diffuse through the IMM, but if it remains in the matrix, it can undergo Fenton chemistry with Fe(II) to yield hydroxyl radicals, which are highly reactive toward DNA and polyunsaturated fatty acids (PUFAs) [2]. Free radical attack on the DNA bases and phosphodiester backbone produce a number of oxidative lesions to the genetic material and release reactive base aldehydes, called base propenals. Polyunsaturated lipid peroxidation (LPO) in the membrane is a major source of electrophilic aldehydes that, along with base propenals, react with cysteine, lysine, arginine, and histidine side chains in proteins and with amino groups of DNA bases [2,4,6,1013].

Polyunsaturated fatty acyl chains comprise over 50% of the total fatty acids in the IMM and are subject to free radical chain reactions that are triggered by ROS [14]. The auto-oxidation of the unsaturated bonds in the PUFAs leads to the formation of distinctive aldehyde products that are routinely neutralized by glutathione conjugation or aldehyde dehydrogenase activity in mitochondria [6,11]. However, cellular conditions that cause an increase in mitochondrial ROS may overwhelm these mechanisms. A recent study has shown that the oxidation of linoleate cardiolipin, which is the predominant cardiolipin species in the IMM, liberates 4-hydroxynonenal (4-HNE) [15,16]. Due to differences in the lipid compositions of the IMM, the mitochondrial outer membrane and the cell plasma membrane, nuclear and mitochondrial DNA may be exposed to a different mixture of the α,β-unsaturated aldehydes, the most prevalent of which are 4-HNE, malondialdehyde (MDA), acrolein and crotonaldehyde [11,13]. Each of these non-radical oxidants can induce protein and DNA modifications in the matrix and is capable of diffusing outward through the IMM [4]. Therefore, reactive aldehydes may compromise mitochondrial gene expression at multiple levels: by damaging mtDNA to induce genetic instability or interfere with polymerase function, by inactivating pol γ, POLRMT or their associated factors involved in gene expression, and by modifying proteins required for the mitochondrial import of RNA and cytosolic proteins. Mitochondrial protein modification by free radical and non-radical endogenous agents and its ramifications on cellular function are discussed in more detail in recent reviews from the Jones laboratory [4,17]. This article will continue with a focus on mitochondrial chromosome damage and its consequences for gene expression.

3. Mitochondrial DNA damage

Mitochondrial DNA polymorphisms and somatic mutations have been studied in the pursuit to understand human disease inheritance, longevity, and population migration [1822]. Certain sequences in the mtDNA chromosome, such as the hypervariable segments of the D-loop regulatory region, appear to be unstable in the natural course of aging [21]. Many single nucleotide variations in mtDNA are benign, but some promote disease [20]. Cells may contain thousands of copies of the mitochondrial chromosome, and a threshold level of the mtDNA population must be compromised before biochemical phenotypes will arise in a cell [22,23]. There appears to be an orchestrated control of the mitochondrial matrix conditions, mitochondrial dynamics, and mtDNA maintenance mechanisms that allow natural genetic change, yet prevent pathogenic mtDNA mutations and degradation [24].

Mitochondrial DNA is susceptible to reactive species generated from cellular metabolism and to environmental agents, including industrial byproducts, therapeutic drugs, and radiation, as will be discussed in detail below. The need for physiological measurements of mtDNA damage and for studies of mtDNA turnover in response to damage is increasing as medical science seeks to correlate the presence of modified nucleic acid metabolites in patient specimens with disease or health risks [25,26]. This is a challenge considering that mtDNA makes up a small fraction of the total cellular DNA and the relative amount of mtDNA to nuclear DNA varies between tissues. Advances in liquid chromatography and mass spectrometry techniques, specifically HPLC-MS/MS, quantitative PCR methods, and repair enzyme-based assays have allowed sensitive detection of a plethora of DNA modifications by eliminating the artifacts that commonly plagued earlier measurements [7,2739]. In recent years, studies have focused on identifying and measuring mtDNA damage to determine whether particular lesions may play a significant role in the mtDNA mutagenesis or depletion observed in clinical disease and chemical toxicology (Table 1). The following sections discuss some of the types of DNA damage detected in human cells that are likely to impact mitochondrial gene expression.

Table 1.

Mitochondrial DNA Damage

Type of Damage mtDNA Source Reference
8-oxo-2′-deoxyguanosine (8-oxo-dG) Alzheimer’s and non-diseased human brain [40,41, 46,49]
HeLa cells [42]
Rat kidney [48]
Rat liver [45, 52]
Rat heart [44]
Mouse liver [47]
8-hydroxy-2′-deoxyadenine Alzheimer’s and non-diseased human brain [49]
5-hydroxy-2′-deoxycytosine Alzheimer’s and non-diseased human brain [49]
5-hydroxy-2′-deoxyuracil Alzheimer’s and non-diseased human brain [49]
2,6-diamino-4-hydroxy-5- formamidopyrimidine (FapyG) Alzheimer’s and non-diseased human brain [49]
4,6-diamino-5-formamido-pyrimidine (FapyA) Alzheimer’s and non-diseased human brain [49]
Fpg Sensitive Sites (includes 8-oxo-dG, 5- hydroxy-2′-purines, and formamidopyrimidines) Mouse liver [30]
CHO cells [43]
1, N6-ethenodeoxyadenosineb Rat liver [51]
3, N4-ethenodeoxycytidineb Rat liver [51]
M1dG Rat liver [50]
BaPDEb C3H10T ½ mouse embryonic cells [94]
Rat hepatocytes [95]
Rat liver, lung, & kidney [96]
Atlantic killifish liver, brain & muscle [182]
N-nitrosamine adductsb Rat liver & lung [97]
Aflatoxin B1 adductsb Rat liver [83]
Cisplatin adductsb CHO cells [153]
Mouse dorsal root ganglion neurons [121]
Maternal and fetal rat tissues [119]
Fetal monkey tissues [120]
a

DNA damage detected by damage-specific methods.

b

Exposure conditions given in the corresponding reference.

3.1 Endogenous mtDNA damage

Free-radical and non-radical oxidants generated inside of cells produce a spectrum of DNA damage including small modifications to bases, such as 8-oxo-2′-deoxyguanosine (8-oxo-dG), formamidopyrimidines, and 8,5′-cyclo-2′-deoxynucleosides, insults to the sugar backbone, such as abasic sites, oxidized deoxyribose rings, and DNA strand breaks, and chemical adducts of bases, such as aldehyde modifications, just to name a few [29]. To date, only a limited set of these have been specifically detected in mtDNA, but most are suspected to occur in mitochondria due to the oxidative conditions of the matrix [5,13,29,30,4052] (Table 1). It has been known for over a decade that the steady-state level of oxidative damage in mtDNA is higher than that in nDNA; however, detection methods for oxidative lesions have required improvements to prevent artifacts and to allow the necessary specificity and sensitivity for measurements [53]. Several different types of oxidative lesions may form under the same cellular conditions, complicating the effort to obtain a complete profile of metabolic DNA damage with a single technique. Another difficulty in mtDNA damage detection is that lesions may undergo additional chemistry in vivo that alters their structure. For example, the 8-oxo-dG in DNA is more susceptible to oxidation than the native dG, and 8-oxo-dG oxidation produces a variety of more stable and mutagenic forms guanine damage, such as hydantoin derivatives [54, 55]. Unlike 8-oxo-dG, hydantoins of DNA bases may not be readily removed from mtDNA, giving rise to their greater mutagenic potential [8, 56]. Therefore, methods that allow for the characterization of multiple lesions and their derivatives would provide the most accurate account of mtDNA damage. In addition to directly damaging the mitochondrial chromosome, biological oxidants can react with free nucleotides producing aberrant substrates for mitochondrial DNA and RNA synthesis [57, 58]. While mitochondria possess an enzyme that can clear 8-oxo-dGTP from the substrate pool, other nucleotide damage also may occur and persist. To compensate for the nucleotide pool imbalance, mtDNA degradation may occur in order to restore the ratios of nucleotides needed for proper synthesis [5860].

Abasic sites, due primarily to purine loss from DNA, occur at a steady-state level of >50,000 sites per cell in mammalian tissues [6163]. Brain, which is rich in mitochondria, has the greatest amount of abasic sites [62]. The 2′-deoxyribose of the abasic site may be oxidized to form modified sugars and single strand breaks (SSBs). In fact, a recent study found that abasic sites and SSBs appear to be the predominant forms of mtDNA damage produced by H2O2 and rotenone-induced oxidative stress in cultured cells [64]. The C4′-oxidation of an abasic sugar will result in the release of the adjacent base in the strand as a base propenal [25, 65]. The base propenals act as electrophiles to generate DNA adducts also produced by the LPO-derived aldehydes [12].

The metabolic capacity and membrane composition of mitochondria make the matrix of these organelles a likely site for the generation of a host of reactive aldehydes [11]. The formation of DNA adducts by biological aldehydes has been reviewed recently in detail; therefore, a brief summary will be offered here [13,66]. In general, aldehydes react primarily with the N2-amine of deoxyguanosine (dG), but they also attack the amino groups of other bases. Aldehyde adducts of DNA bases may undergo additional chemistry depending on their structure. Formaldehyde, which is produced through amino acid metabolism, initiates covalent crosslinks between protein and DNA, but also produces N2-hydroxymethyl-dG adducts [67]. Although it is produced physiologically, formaldehyde is rapidly oxidized to formate and may not be a substantial threat to DNA, unless it is encountered at higher concentrations through environmental exposure [68,69]. The α, β-unsaturated aldehydes, resulting from PUFA and DNA oxidation, each produce a characteristic set of adducts by a Michael addition to the amino group of DNA bases followed by a cyclization to form either a five-membered (etheno) or a six-membered (propano) ring on the DNA base [13]. The LPO-induced, etheno adducts, 1, N6-ethenodeoxyadenosine and 3, N4-ethenodeoxycytidine, are produced in the liver mtDNA of Long-Evans Cinnamon rats, animals with a genetic background resulting in the accumulation of copper and ultimately in the formation of hepatocellular carcinomas [51]. Malondialdehyde (MDA), generated from both prostaglandin biosynthesis and LPO, creates the six-membered, exocyclic adducts, M1dG, M1dA and M1dC, of which M1dG is most prevalent and occurs at a higher frequency in mtDNA than nDNA [50]. The cardiolipin LPO product, 4-HNE, forms 1, N2- and N2,3-etheno-dG adducts, which are also produced by acrolein and crotonaldehyde, l, N2-propano-dG, and four unique stereoisomeric l, N2-propano-dG adducts carrying a fatty acyl group [70]. The DNA adducts of MDA and 4-HNE, and metabolites of these adducts, are of particular interest clinically, due to their elevated levels in patients with various pathologies [25]. Acrolein creates the hydroxy-l, N2-propano-dG adducts, α-and γ-OH-PdG, with γ-OH-PdG being the major product [13]. The γ-OH-PdG adduct may further react with nearby functional groups to form DNA interstrand crosslinks and protein-DNA crosslinks [7072]. Considering the PUFA enrichment of the IMM, α,β-unsaturated aldehydes are likely a major threat to mtDNA stability and gene expression during tissue oxidative stress.

Interestingly, protein-DNA covalent crosslinks are also produced through the action of DNA topoisomerases, enzymes required for replication, transcription, and separation of chromosomes. The characterization of mitochondrial type I topoisomerases, Top1mt and Top 3αmt, and the subsequent detection of Top1mt cleavage sites in the D-loop of mtDNA raises the possibility that aberrant topoisomerase activity may be a cause of mtDNA deletions and chromosome degradation [7375]. As part of their mechanism in mediating DNA topology during transcription and replication, the type I topoisomerases cut a DNA strand and create a covalent phosphotyrosyl bond with either the 5′ or 3′ end of the nick [76]. These enzyme-DNA complexes, which are normally transient intermediates in the catalytic cycle, can be stabilized by cancer chemotherapy drugs and by many forms of oxidative and environmental DNA damage [76, 77]. Extending the lifetime of the intermediates can be deleterious, as they can be broken by replication and transcription complexes leaving unique tyrosyl-DNA linkages that must be resolved before DNA strand continuity can be restored. Tdp1, a phospholipase D family member that resides in both nuclei and mitochondria, recognizes the 3′-phosphotyrosyl-DNA ends created by the disruption of type I topoisomerase catalytic intermediates [78, 79]. A loss of Tdp1 function results in spinocerebellar ataxia with axonal neuropathy (SCAN1), which resembles Friedreich ataxia, a disease that is known to be associated with mtDNA instability [26,80]. This suggests that Tdp1 plays a critical role in the processing of Top1mt-mediated DNA strand breaks in order to maintain mitochondrial function in neurons [79,81]. Since Top1mt alleviates torsional stress in mtDNA during replication and transcription, studies of the response of this enzyme to DNA damage and further studies of Tdp1 function in mtDNA repair may provide insight into the mechanisms for mtDNA deletions and the side effects of cancer chemotherapies targeted against topoisomerases [82].

3.2 Environmentally-induced mtDNA damage

Chemicals in the environment, metabolites of dietary components, drugs in clinical therapies, and radiation from sunlight or medical procedures are external sources of DNA damage. The benzo[a]pyrene and acrolein components of cigarette smoke, the fungal toxin aflatoxin B1, platinum-based chemotherapy agents, antiviral nucleoside analogs, and ultraviolet (UV) radiation all induce DNA adducts that inhibit mitochondrial transcription or interfere with DNA pol γ function [8386](R. Kasiviswanathan and W. C. Copeland, personal communication). Therefore, disruption of mitochondrial gene expression or mutagenesis of mtDNA should be considered in assessing environmental exposure or drug toxicities.

The aldehydes, produced physiologically in our tissues, are increased during the metabolism of alcohol and can also enter our bodies from the environment, where they are present with other reactive agents [7, 87]. Acetaldehyde, also called ethanal, is generated at a high concentration in the liver following the ingestion of alcohol and is converted to acetate in mitochondria by aldehyde dehydrogenase 2, ALDH2. If ALDH2 cannot efficiently oxidize acetaldehyde, it forms a number of adducts, including N2-ethylidene-dG, 1, N2-propano-2′-dG and their derivatives [8891]. The 1, N2 -propano-2′-dG adduct can combine with a second aldehyde molecule to make an interstrand DNA crosslink that will inhibit strand separation by helicases [92]. In industrial pollutants, formaldehyde coexists with alkylating agents, a combination that forms N2-hydroxymethyl-dG and its derivatives from subsequent methylation events [69]. Cigarette smoke contains acrolein, crotonaldehyde and formaldehyde along with polycyclic aromatic hydrocarbons (PAH), such as benzo[a]pyrene, and several tobacco-specific N-nitrosamines (TSNA). Benzo[a]pyrene (BaP) is metabolized by the liver cytochrome P450 CYP1A1 to benzo[a]pyrene 7,8-dihydrodiol-9,10-epoxide (BaPDE), and both BaP and BaPDE have been shown to accumulate in mitochondria and inhibit mtDNA synthesis [9396]. The TSNA compounds, also activated by CYP enzymes, form DNA adducts that have been detected in the lung and liver mtDNA of carcinogen exposed rats, however the effects of TSNA adducts on mtDNA synthesis and gene expression have not been determined [97]. The BaP effects on mtDNA replication likely stem from BaPDE modifications of dG and dA, which block DNA pol γ and inhibit topoisomerase activity. However, in smoke exposed tissues, the coexisting aldehyde and TSNA-induced damage may act synergistically with BaPDE adducts to hinder mitochondrial polymerases [85, 98103].

Hepatic toxicity and carcinogenesis from environmental exposures may be due in part to mtDNA damage by xenobiotic derivatives, as in the case of BaP or aflatoxin B1, or by ROS generated during CYP activity. Polychlorinated biphenyls (PCBs), which are widespread in our environment, increase ROS production during their metabolism by liver CYP enzymes [104]. Rats chronically exposed to PCBs were found to have elevated levels of M1dG in their livers, possibly due to the release of MDA and base propenals by lipid and DNA sugar oxidation, respectively [105]. In fact, this finding suggests that toxic environmental agents that increase ROS by the disruption of mitochondrial membrane potential, such as dioxins in herbicides, may also induce oxidative mtDNA damage that inhibits mitochondrial gene expression [106].

The nucleoside analogs designed as antiviral drugs and some DNA damaging agents employed in cancer therapies have off-target effects on mitochondria that may be attributed to direct and indirect damage to mtDNA and its RNA transcripts. Nucleotide reverse transcriptase inhibitors (NRTIs) against HIV have common side effects that resemble symptoms of mitochondrial diseases, including skeletal muscle and heart myopathies, peripheral neuropathy, lactic acidosis, lipodystrophy and hepatic steatosis [107]. The mitochondrial pol γ is the only human DNA polymerase that effectively uses NRTIs as substrates, and many of the clinically utilized NRTIs are incorporated into mtDNA and cause the termination of strand synthesis [108, 109]. Other ribonucleoside analogs intended for the treatment of RNA viral infections, but found to be highly toxic to patients in clinical trials, may be substrates for POLRMT and inhibit mitochondrial transcription [110]. Indeed, the future development of therapeutic nucleoside analogs may benefit from initial testing of substrate utilization by mitochondrial polymerases now that purified recombinant systems, as described in Section 5, make these assessments possible.

Many anticancer agents directly or indirectly damage DNA to inhibit tumor growth and induce apoptosis. A few of these have side effects that suggest that they may have effects on mitochondrial function. Bleomycin and neocarzinostatin cause oxidative damage to DNA through ROS and reactive aldehyde generation [12, 111]. These drugs are known to cause pulmonary fibrosis, and bleomycin is used to generate fibrosis in animal models for the study of this disease process [112,113]. While no direct evidence exists, it is possible that mtDNA damage may contribute to the cellular transformation and apoptosis observed in the lung tissue of bleomycin-treated patients [25,65]. The platinum (Pt)-containing chemotherapy drugs, such as cisplatin, carboplatin, and oxaliplatin, bind directly to DNA to form single base adducts and intra- and interstrand crosslinks (ICL) between guanine bases [114]. For cisplatin, the intrastrand l,2-d(GpG) crosslink appears to be the most prevalent modification following drug exposure; however, even in minor amounts, ICL adducts are catastrophic to replication and transcription as they prevent the strand separation required for polymerase function [115118]. Cisplatin adducts appear in the mtDNA of fetal tissues following the treatment of pregnant rats and monkeys, indicating that the drug may alter development through inhibition of mitochondrial gene expression [119,120]. Patients treated with Pt agents often experience peripheral neuropathy, which may relate to the observation of cisplatin-induced mtDNA damage in dorsal root ganglion neurons [121]. Cisplatin exposure reduced mtDNA replication and transcription in the cultured neurons, which correlates with the in vitro finding that pol γ nucleotide insertion is inhibited by GG intrastrand crosslinks [84,122]. In the nucleus, repair of the full spectrum of Pt-DNA adducts requires a concerted effort by the Fanconi pathway for ICL repair, homologous recombination, mismatch repair and nucleotide excision repair mechanisms [118,123125]. Cells of the body that are reliant on oxidative metabolism, such as neurons and muscle cells, may be highly susceptible to Pt-drug chemotherapy, because mitochondria do not have the full complement of these repair pathways [126].

4. Mitochondrial DNA repair mechanisms

Historically, the high DNA mutation rate in mitochondria has been attributed to oxidative damage in the context of limited mtDNA repair. However, in the last two decades, studies have revealed that mitochondria possess several mechanisms for genetic maintenance, including a robust base excision repair (BER) mechanism that utilizes both mitochondrial proteins and nuclear proteins that translocate into mitochondria. Additionally, mitochondrial fusion and fission events and mtDNA degradation may allow the organelles to lessen the toxic effects of damage left unresolved by mitochondrial repair pathways [64,127,128]. Several reviews published in the last two years have described in detail the known repair mechanisms in mitochondria, so only a brief synopsis of mtDNA repair and the latest findings are reported here [8,129131].

The DNA repair repertoire of mitochondria currently includes single-nucleotide base excision repair (SN-BER) [43,126,132,133], long-patch BER (LP-BER) [134138], single-strand break repair (SSBR) [131], YB-1-mediated mismatch repair [139,140], removal of adenine opposite 8-oxo-dG [141,142], and MTH1 removal of 8-oxo-dGTP and 8-oxo-2′-dATP from the mitochondrial nucleotide pool [143,144]. Nonhomologous end joining and homologous recombination activities, which can repair double-stranded DNA breaks, have been detected in mammalian mitochondria, and recent studies suggest a role for the RecQ helicase, RECQL4, in mitochondrial double-strand break repair [145150]. Strikingly, mitochondria lack nucleotide excision repair activity, which resolves bulky DNA damage, such as UV photoproducts, BaPDE adducts, cisplatin intrastrand crosslinks, and the oxidative lesions, 8,5′-cyclo-2′-deoxypurine and M1dG [98100,124,126,151156]. The mitochondrial limitations for excision repair are discussed below, along with some recent findings that add insight into the relationship between nuclear and mitochondrial DNA damage in preserving cellular function.

4.1 Base Excision Repair

Base excision repair (BER) is primarily responsible for the removal of DNA bases altered by oxidation, alkylation, or deamination, and the repair of abasic sites and single-strand breaks resulting from spontaneous hydrolysis and oxidation, respectively (reviewed in [130]). The mitochondrial BER mechanisms, SN-BER and LP-BER, closely resemble the nuclear pathways and utilize some of the same proteins, but DNA pol γ conducts the 5′-deoxyribose phosphate lyase and polymerase activities performed by DNA polymerase β, in the nucleus. Several nuclear DNA glycosylases, which catalyze damage-specific N-glycosylic bond cleavage, have been found in mitochondria, including uracil DNA glycosylase, UNG 1, nth endonuclease III-like 1 glycolysase, NTHL1, MutY homolog glycosylase, MUTYH, and the bifunctional glycosylases OGG1, NEIL1 and NEIL2 ([130] and references therein). The latter three enzymes have overlapping specificity for 8-oxo-dG, but differ in their recognition of other oxidative lesions. Together, they rectify most of the prevalent forms of free radical induced oxidative DNA damage. In fact, the level of 8-oxo-dG in the intact mtDNA isolated from the liver of OGG1−/− mice is only slightly elevated above that in mtDNA from the liver of wild type animals, suggesting the efficient removal of 8-oxo-dG by the NEIL enzymes or the degradation of highly damaged mitochondrial chromosomes in the absence of OGG1 [30]. While mitochondria have redundant glycosylase activity for 8-oxo-dG removal, they appear to lack the monofunctional N-methylpurine DNA glycosylase, MPG, that is responsible for the BER processing of alkylpurines, including methylated bases and larger alkylations, such as the l, N6-etheno-dA and l, N2-etheno-dG adducts generated by reactive aldehydes [157159]. Therefore, it appears that these forms of base damage pose a persistent threat to mtDNA gene expression and stability.

OGG1, NEIL1 and NEIL2 all conduct N-glycosylic bond cleavage at oxidized bases, but NEIL1 and 2 catalyze both β- and δ-elimination reactions to excise the abasic sugar without recruiting AP endonuclease 1 (APE1). OGG1 performs only β-elimination at the sugar and relies on APE1 to complete sugar excision from the DNA strand. Nuclear BER initiated by either the OGG1 or NEIL glycosylases converge at the recruitment of XRCC1/ligase III and DNA pol β for restoring continuity to the gapped DNA strand produced by damage removal [130]. The recognition of the single-base, gapped intermediate is believed to involve a poly(ADP-ribose) polymerase-1 (PARP1) complex [160], and in the case of the NEIL-initiated BER pathway, also requires the activity of polynucleotide kinase 3′-phosphatase, PNKP, an enzyme that prepares the DNA ends at a strand gap for extension by pol β [161, 162]. Interestingly, mitochondria do not contain the XRCC1 protein, which acts as a scaffold for assembly of nuclear BER factors and brings in ligase III to complete repair. Mitochondrial ligase III is essential for mouse embryo viability; therefore, it must be recruited into the BER process by a surrogate for XRCC1 [163,164].

Recently, aprataxin, PARP1, and PNKP have been located in mitochondria [160,165167]. Mutations in the gene for aprataxin, APTX, is associated with the disorder ataxia oculomotor apraxia 1, or AOA1, which is an autosomal recessive cerebellar ataxia with peripheral neuropathy and eye movement limitations [168171]. Aprataxin shares homology with PNKP, histidine-triad (HIT) proteins, and zinc-finger DNA binding proteins and interacts with PARP1 at single-strand breaks where it removes 5′-adenylation damage to prepare DNA ends for strand ligation by DNA ligase III [160,171,172]. Based on the mitochondrial localization and the molecular interactions of aprataxin, this protein is a candidate for the recognition and processing of single-stranded breaks in a mitochondrial BER mechanism [173]. While the ADP ribosylation of mitochondrial proteins has been reported by several groups, the mitochondrial presence of PARP1 and its role in mtDNA expression is still a matter of debate [165,174176]. PARP1 appears to enter the organelle through an interaction with the mitochondrial import protein, mitofillin; however, a recent report suggests that PARP1 affects mitochondrial repair and transcription through its regulation of nuclear genes for mitochondrial proteins, instead of by mitochondrial interactions [165,176]. PNKP, on the other hand, appears to participate in mitochondrial BER through a direct association with NEIL2 [167]. The mitochondrial PNKP is identical to the nuclear protein, which contains a C-terminal mitochondrial localization sequence. The depletion of PNKP from human cells causes an increase in both mitochondrial and nuclear DNA damage following H2O2 treatment [166]. Since aprataxin, PARP-1, and PNKP appear to act in restoring continuity to DNA strands, these proteins may play a broad role in recruiting pol γ and ligase III for the repair of strand breaks formed by many insults to mtDNA.

The discrepancies found in the mitochondrial localization of repair proteins may be due to a tissue-specificity for their function in mitochondrial DNA repair. Equally as plausible is the possibility that DNA damaging agents may trigger the mitochondrial localization of specific repair proteins, such as PARP1. While this idea is merely speculation, a recent paper showing that PARP-1 binds to DNA containing cisplatin l, 2-d(GpG) intrastrand crosslinks, taken along with the reports of mitochondrial PARP1, suggests that this hypothesis may be a valid explanation for inconsistencies in PARP1 localization studies [177,178]. PARP-1 may enter the mitochondria in response to mtDNA damage in order to facilitate mtDNA repair or to initiate a signal for mtDNA destruction. Further studies in this area may bring to light the link between the DNA damage binding and ADP ribosylation properties of PARP1 in mitochondria.

4.2 Nucleotide Excision Repair

It was established in 1974, and reaffirmed in two later studies, that mitochondria lack the NER mechanism responsible for the removal of UV-induced pyrimidine dimers [126,151,152]. Consistent with these findings, mitochondria display very poor repair of cisplatin intrastrand crosslinks but efficiently remove interstrand crosslinks, which are repaired by recombination instead of NER [126, 153]. Since both types of cisplatin adducts are efficiently removed by nuclear repair mechanisms, mitochondrial damage is likely to be the major mechanism for cisplatin-induced neurotoxicity [121,123,124,179]. In light of the apparent mitochondrial deficiency in NER, 8,5′-cyclo-2′-deoxypurines (cPu), M1dG and BaPDE adducts, would also pose problems for mtDNA gene expression in many tissues of the body [98100,124,154156]. While cPu have yet to be detected in mtDNA, these oxidative lesions accumulate in the liver, kidney, and brain of aging mice and are known to inhibit DNA transcription [155,180]. M1dG occurs at a two-fold higher frequency in mtDNA than nuclear chromosomes and likely co-exists with other DNA damage requiring NER removal, including endogenous aldehyde adducts and adducts generated by polyaromatic hydrocarbons (PAH) [13, 50, 105]. Benzo[a]pyrene and BaPDE accumulate in mitochondria, and a 40- to 90-fold higher level of BaP covalent adducts has been observed in the mtDNA relative to the nDNA of mammalian cells upon exposure to BaPDE [93, 94, 96]. This difference may be due to a higher binding affinity of PAH compounds for mtDNA [181]. A more recent study of Atlantic killifish confirmed that BaP-induced adducts accumulate in the mtDNA of liver, brain and muscle tissue following intraperitoneal BaP injection, but no significant difference between mtDNA and nDNA levels were observed [182]. Although there are inconsistencies between cellular and whole animal studies of PAH adducts, both systems revealed that mtDNA is highly susceptible to PAH damage due to the absence of mitochondrial NER.

The only evidence to date for an NER-like mechanism in mitochondria is the presence of the Cockayne syndrome proteins, CSA and CSB, which are required for nuclear transcription-coupled NER (TC-NER) [183185]. TC-NER is detected in nuclear chromosomes as the more rapid repair of the transcribed strands of genes relative to non-transcribed DNA and is initiated by the arrest of an elongating RNA polymerase II (RNAP II) complex at a site of DNA damage [186]. The CSB protein recruits CSA and chromatin remodeling proteins to the stalled RNAP II complex, while CSA, a ubiquitin E3 ligase, brings in other essential factors that assist in reversing RNAP II and processing the RNA transcript to clear the site of damage for template strand repair [185]. The CSB protein has been implicated in the repair of 8-oxo-dG, and the loss of CSB in an Ogg1−/− background increases mutation rates [187, 188]. However, Ogg1−/− Csb−/− mice have no significant increase in 8-oxo-dG in their liver mtDNA relative to wild type animals [30]. The NEIL glycosylases in mitochondra likely compensate for Ogg1 loss in removing 8-oxo-dG, and the elevated mutation frequencies with CSB loss may be due to the persistence of other oxidative lesions. Recently, the NEIL1 glycosylase, which is found both in the nucleus and in mitochondria, has been shown to mediate nuclear NER of 8′, 5′-cyclo-2′ deoxyadenosine (cyclo-dA), an endogenous oxidative DNA lesion formed by intramolecular cyclization between C5′ of the deoxyribose and C8 of the purine ring [189]. Interestingly, mice lacking CSB accumulate cyclo-dA, indicating a role for CSB in its repair. While cyclo-dA has yet to be detected in mtDNA, the finding that CSB participates in the removal of this oxidative lesion suggests that mitochondria have an NER-like mechanism [184,189]. Another intriguing aspect of the Ogg1−/− Csb−/− mouse study is that a portion of the mtDNA oxidative damage in the mutant animals appears to be due to 4′-oxidation resulting in base loss, a process that would generate base propenals and aldehyde-induced DNA adducts [65]. Since aldehydes primarily damage dG, the formation of aldehyde adducts in mtDNA may, in part, explain the prevalent G→T transversions and mtDNA deletions observed in Ogg1−/− Csb−/− mice [187]. Further studies of mtDNA from Ogg1−/− Csb−/− mice are required to determine whether the persistence of mtDNA damage due to the lack of a CSB-dependent mechanism may contribute to the enhanced mutation frequency in these animals.

Mitochondria may have NER-like mechanism that resolves a different spectrum of DNA damage than that handled by the nuclear NER pathway, while some nuclear targets for NER may trigger mtDNA deletion or degradation. Indeed, the recent finding of PARP-1 recognition of 1,2-d(GpG) platinum crosslinks hints at the possibility for alternative processing of NER-targeted adducts in mtDNA [178]. As discussed below, the arrest of POLRMT by the aldehyde adduct, M1dG, also suggests that a mitochondrial transcription-coupled mechanism for mtDNA damage signaling may exist. Our current knowledge of mtDNA repair, however, leaves us to conclude that metabolically- and environmentally-induced adducts requiring NER for their removal simply will persist in mitochondrial chromosomes where they can disrupt mtDNA replication and gene expression, cause mtDNA mutation, and may ultimately lead to mitochondrial chromosome degradation.

5. DNA damage effects on gene expression

Many types of DNA damage have been studied for their effects on mammalian nuclear DNA polymerases and RNAP II [185,190194]. These studies are the basis for understanding the process of translesion DNA synthesis, which serves as a tolerance mechanism for DNA damage, and for the mechanism of TC-NER, which was discussed above. To date, we have only a minimal knowledge of how DNA damage affects pol γ or POLRMT function. Additionally, proteins that support mtDNA replication and transcription, namely mtTop1 and the nucleoid/transcription factor, TFAM, interact with DNA damage, and those interactions may alter mtDNA processing [73, 102, 179, 195]. The structural and biochemical properties of pol γ and POLRMT are discussed by R. Kasiviswanathan and J. J. Arnold, respectively, elsewhere in this issue [82, 110]. The following sections of this review summarize the behavior of these enzymes when encountering DNA damage and the possible ramifications of mtDNA on mitochondrial gene expression.

5.1 Mitochondrial DNA replication

DNA pol γ performs all of the replicative and repair-associated DNA synthesis in mitochondria. In all of the proposed models for mtDNA replication, DNA strand synthesis begins from an RNA primer generated by POLRMT (reviewed by [82] in this issue). This requirement intimately links the functions of these enzymes and presents two avenues by which DNA damage may affect mtDNA replication, by interfering with POLRMT RNA primer synthesis or by disrupting pol γ processivity and fidelity in DNA synthesis. In the nucleus, a plethora of DNA polymerases are part of the ‘tool kit’ for lesion bypass mechanisms, which are activated through ubiquitinylation of the PCNA sliding clamp [192, 196]. However, it appears that pol γ acts alone when negotiating DNA damage in mitochondrial chromosomes, and any replication-mediated signals that may trigger the repair or bypass of mutagenic damage have yet to be elucidated.

For mtDNA replication, DNA pol γ forms a highly processive holoenzyme complex with two molecules of its accessory subunit, p55. The polymerase possesses 3′ → 5′ proofreading ability that confers fidelity for nucleotide insertion, but the activity is reduced when pol γ is bound with p55 [197200]. This property indicates that pol γ may bypass some forms of DNA damage through either correct or errant nucleotide incorporation. The proofreading activity of the enzyme competes with its nucleotide insertion activity, complicating the kinetic analysis of either process in isolation. Therefore, an exonuclease-deficient (exo p140) pol γ has been purified for the study of nucleotide insertion opposite DNA adducts and of nucleotide analog utilization during strand synthesis [201]. The exo p140 enzyme retains its ability to associate with p55 in a holoenzyme complex and has been characterized relative to wild-type pol γ for fidelity and nucleotide insertion kinetics. For studies of pol γ interaction with DNA damage, primer:template duplex substrates are used that contain a single lesion in the template strand either at the position immediately past the end of the primer or at a position that is a few residues downstream of the primer terminus. The placement of the damaged nucleotide in these locations allows for the assessment either single nucleotide insertion kinetics or kinetics after a ‘running start’ to DNA synthesis, respectively. Additionally, undamaged primer:template substates are employed to analyze pol γ binding and incorporation of damaged nucleotide triphosphates that may be present in the mitochondrial dNTP pool [202]. These reconstituted systems have provided the basis for our understanding of pol γ interaction with DNA damage and of therapeutic drug effects on mtDNA synthesis.

Among the forms of DNA damage investigated for their effects on pol γ are abasic sites, the oxidation adducts, 8-oxo-dG, M1dG, and γ-OH-PdG, the UV-induced CPDs, the carcinogenic adducts of benzo[a]pyrene- and benzo[c]phenanthrene diol epoxide, and the intrastrand crosslinks induced by platinum-based chemotherapy agents ([8486]; R. Kasiviswanathan and W. C. Copeland, personal communication; S. D. Cline, unpublished) (see Table 2). Abasic sites are likely to be the single greatest threat to mtDNA stability, based on their prevalence in human cells [62]. Early studies with Xenopus laevis pol γ showed that abasic sites inhibited 80% of strand synthesis and caused nearly exclusive insertion of dATP when translesion synthesis did occur [203]. Abasic sites frequently arise from depurination, so most of these events would be mutagenic. The same study showed a similar degree of pol γ inhibition by 8-oxo-dG; however, lesion bypass occurred primarily through dCTP insertion, indicating that coding errors would be avoided during replication of a damaged chromosome. Following these studies, a correlation was made between a mtDNA 4977 bp deletion and elevated levels of 8-oxo-dG in aging human brain tissue suggesting that oxidative lesions in DNA may trigger substantial loss of genetic material through their effects on DNA pol γ [46]. MTH1 expression is increased in Parkinson disease patients, indicating that oxidation of the nucleotide pool is part of the neurodegenerative mechanism [204]. While mitochondrial MTH1 can effectively clear 8-oxo-dGTP from the nucleotide pool and 8-oxo-dG is efficiently removed from mtDNA by BER, pol γ likely encounters these damaged substrates in tissues under oxidative stress [143]. Compared to wild-type mice, MTH1−/− animals show an accumulation of 8-oxo-dG in the mtDNA of their dopaminergic neurons following treatment with 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP), an agent used to mimic the effects of Parkinson’s disease. MPTP-induced 8-oxo-dG accumulation presumably occurs through pol γ incorporation of 8-oxo-dGTP from the dNTP pool, since BER is not compromised in the MTH1 deficient mice [58, 202, 205, 206]. This finding indicates that the levels of ROS generated by MPTP may overwhelm BER capacity to remove 8-oxo-dG from mtDNA. Studies such as these suggest that the neurodegeneration observed in aging and disease may arise due to multiple insults on mtDNA, some of which may inhibit pol γ synthesis to induce mutagenesis and mtDNA deletions [22, 46, 207, 208].

Table 2.

DNA Pol γ Responses to DNA Damage

Damage Inhibitiona Insertion Preferenceb Reference
Abasic sitesc n.d.d dATP [203]
8-oxo-dG + dCTP≫dGTP>dATP>dTTP [202, 203, 206]
γ-OH-PdG + dATP>dGTP≫dTTP>dCTP R. Kasiviswanathan and W.C. Copeland, personal communication
M1dG n.d. dATP>dGTP≫dCTP>dTTP Cline, unpublished
CPD (T^T)e + [86]
 T-T 3′-T + dATP≫dGTP≫dTTP>dCTP
 T-T 5′-T ++ dATP
PAH [85]
 BaP trans R dG +++ dATP>dGTP>dTTP>dCTP
 BaP trans S dG +++ dATP>dGTP>dTTP>dCTP
 BaP trans R dA ++ dATP>dTTP
 BaP trans S dA ++ dATP>dTTP
 BaP cis R dA ++ dATP > dTTP>dGTP>dCTP
 BaP cis S dA ++ dATP > dGTP>dTTP>dCTP
 BcPh cis R dA ++ dATP≫dTTP>dGTP>dCTP
 BcPh cis S dA ++ dATP≫dCTP>dGTP>dTTP
Pt-GG intrastrandf [84]
 oxaliplatin 85% n.d.
 cisplatin 91% n.d.
 JM216 (satraplatin) 95% n.d.
a

Based on the translesion insertion efficiency which is the ratio: (kcat/Km) preferred dNTP at normal base/(kcat/Km) preferred dNTP at the damaged base. Translesion insertion efficiency indicates the extent of synthesis inhibition and is expressed as: + = up to 1000-fold; ++ = 1000 to 10,000-fold; and +++ = 10,000 to 100,000-fold inhibition.

b

Preference for nucleotide insertion opposite the adduct; ≫ indicates at least an order of magnitude difference in either Vmax/Km or kcat/Km for insertion. Insertion was not detectable for dNTPs not listed.

c

Results only for Xenopus laevis pol γ

d

n.d. indicates ‘not determined’.

e

Information provided for insertion opposite the 3′-T or 5′-T of the TT dimer. Only dATP was studied for insertion at the 5′-T.

f

Insertion kinetics for individual dNTPs were not determined; therefore, the percentage inhibition of primer extension by the Pt adduct relative to the undamaged GG in the presence of all four dNTPs and a 1:1 enzyme-to-template ratio is shown.

Several oxidative lesions have been shown to disrupt DNA synthesis by human DNA pol γ using the reconstituted system described above (see Table 2). An 8-oxo-dG in the template strand prevents 95% of pol γ molecules from proceeding past insertion at the lesion [202, 206]. As seen with the Xenopus enzyme, human pol γ inserts dCTP opposite 8-oxo-dG, so that the rare bypass events are error-free. This suggests that 8-oxo-dG primarily inhibits replication, and other types of damage, produced concurrently with 8-oxo-dG, may be responsible for mtDNA mutagenesis. The adducts γ-OH-PdG and M1dG, produced by the aldehyde byproducts of lipid peroxidation, likely form along with 8-oxo-dG in human tissues. Although these adducts are much less prevalent in mtDNA than 8-oxo-dG, they have substantial effects on pol γ activity. Human pol γ shows a strong preference for the incorporation of purines over the correct insertion of dC opposite both γ-OH-PdG and M1dG. Interestingly, the enzyme can continue strand synthesis from γ-OH-PdG:purine pairs making this adduct highly mutagenic, while M1dG:purine formation seems to terminate replication (R. Kasiviswanathan and W.C. Copeland, personal communication; S. D. Cline, unpublished). These findings indicate that endogenous adducts formed in mtDNA during oxidative stress have very specific effects on DNA integrity, and that less prevalent, but persistent aldehyde adducts, may contribute to mtDNA instability.

Environmental agents and therapeutic drugs also have substantial effects on pol γ function. While UV-induced CPDs form only in sun exposed tissues, their impact on mtDNA replication may contribute to dermatological aging, neoplasia and cancer [209213]. Recently, it was shown that pol γ incorporates adenines opposite thymine-thymine CPDs; however, the observed 800-fold decrease in efficiency for adenine insertion suggests that the majority of pol γ replication will stall at CPDs, preventing replication of UV-damaged chromosomes [86]. These data point to CPD encounter by pol γ as a source of the mtDNA instability associated with UV exposure. For other human tissues, ingestion, inhalation or injection are the most likely delivery mechanisms for environmental mtDNA damaging agents. Two prevalent PAH compounds, BaP and benzo[c] phenanthrene (BcPh), are converted by liver cytochromes P450 and epoxide hydrolyases to their respective diol epoxides (BaPDE and BcPhDE), which react with purines to generate a host of stereisomeric DNA adducts [214216]. The BaPDE and BcPhDE adducts of guanine and adenine are substantial blocks to pol γ strand extension, findings consistent with the inhibition of mtDNA synthesis observed in cultured hepatocytes treated with BaP [95,217]. When translesion synthesis does occur opposite these PAH adducts, pol γ primarily inserts purine nucleotides resulting in strand mutagenesis or in a base pair that cannot be extended [217]. Therefore, the instability of mtDNA is likely to be a part of the carcinogenic mechanism of PAH exposure.

In the therapeutic realm, two drug classes appear to have effects on mtDNA, the Pt-based anticancer agents and the NRTIs used against HIV. As mentioned above, cisplatin can cause peripheral neuropathy in patients and reduced mtDNA replication and transcription in cultured dorsal root ganglion neurons [121]. Previous in vitro studies had shown that cisplatin or oxaliplatin intrastrand d(GpG) crosslinks arrest pol γ synthesis either just prior to or immediately after nucleotide incorporation opposite the 3′-G in the d(GpG) [84]. Taken together, these findings indicate that termination of mtDNA replication by pol γ is the underlying mechanism for the side effects of cisplatin.

Many NRTIs utilized clinically for HIV therapy have the off-target effect of mitochondrial toxicity resulting in lactic acidosis, liver failure and granolocytopenia in patients. These nucleoside analogs are incorporated into mtDNA by pol γ and cause the termination of DNA synthesis [108, 109]. NRTIs display the following order of pol γ inhibition: zalcitabine (ddC) = didanosine (ddI) = stavudine (D4T) ≫ lamivudine (3TC) > tenofovir (PMPA or TDF) > zidovudine (AZT) > carbovir (CBV, the active form of abacavir, ABC) [108, 109]. Consistent with these findings, a high level of mtDNA depletion was observed in peripheral blood mononuclear cells from children treated with ddI, and the agents ddC, ddI, and D4T induced mtDNA depletion in cultured human skeletal muscle cells [218, 219]. In the muscle cells, ddI decreased the RNA for mitochondrial-encoded MTCYB and MTC03 genes without altering mRNA levels for POLG and TFAM [219]. A loss of mtDNA was also seen in lymphocytes treated with ddC, ddI, and D4T, while lymphocytes exposed to AZT, which has lesser effects on pol γ, retained their mtDNA [107]. From this body of work, it may be concluded that NRTI mitochondrial toxicity results from mtDNA depletion triggered by the direct effects of the agents on DNA synthesis by pol γ.

5.2 Mitochondrial DNA transcription

Less is known about the response of POLRMT to DNA or nucleotide pool damage than about that of pol γ or other RNA polymerases. The RNA polymerases of the T7 bacteriophage, Escherichia coli, yeast, and human cells have been characterized for their ability to facilitate TC-NER and for their response to a wide variety of DNA damage [185, 186, 193, 220, 221]. Although NER of UV damage is absent from mitochondria, making the existence of TC-NER unlikely, the presence of CSA and CSB suggests that other transcription-coupled processes may exist. Even in the nucleus, the exact mechanism that triggers repair following CSA and CSB interaction with a damage-arrested RNAP II elongation complex has not been elucidated [185]. Mitochondrial CSA and CSB have yet to be analyzed for their association with POLRMT, so whether they play a similar role in a mitochondrial transcriptional response to mtDNA damage remains uncertain. Indeed, the polycistronic nature of mitochondrial transcription on both mtDNA strands, along with the dependence of replication priming on transcription, make the study of POLRMT interaction with DNA damage essential to our understanding of mitochondrial chromosome maintenance and gene expression.

If POLRMT arrest at DNA damage does not trigger repair, the POLRMT elongation complex may abort transcription, preventing efficient gene expression, or may remain stalled at the site of damage, potentially emanating a signal for mitochondrial chromosome degradation or for mitophagy [222]. Such a “self-destruct” signal is issued by the sustained arrest of nuclear RNAP II complexes, which triggers apoptosis, and recently, p53 has been found in mitochondria and shown to control mtDNA copy number [223226]. So, in a similar way, POLRMT arrest might protect the cell by starting a signaling cascade that would eliminate populations of defective mtDNA, in lieu of their repair, and prevent the expression of aberrant mtDNA transcripts [226, 227]. DNA damage that does not halt transcript elongation may cause POLRMT to insert incorrect nucleotides, creating an errant template for protein translation. This outcome has been termed ‘transcriptional mutagenesis’ and has been documented for E. coli RNA polymerase and eukaryotic RNAP II enzymes in their interactions with several forms of DNA damage [228230]. As discussed above for DNA pol γ, oxidative damage to the mitochondrial nucleotide pool or the presence of nucleotides derived from therapeutic nucleosides might also foul RNA synthesis, as the POLRMT binds and inserts the modified NTPs. Since transcriptional disruption could have both detrimental and protective consequences, experimental systems that allow for the isolation and sequencing of POLRMT transcripts and for the isolation of factors that bind to damage-arrested POLRMT are needed in order to elucidate transcription-mediated events resulting from mtDNA damage. Reconstituted in vitro systems with purified human mitochondrial proteins have been employed for the analysis of POLRMT initiation, elongation and nucleotide utilization [231235]. The few studies completed thus far have provided insight not only by showing the effects of DNA and nucleotide pool damage on POLRMT transcript elongation but also by revealing functional differences between POLRMT and the T7RNAP and RNAP II enzymes.

The in vitro arrest of POLRMT by DNA damage is the most tangible evidence that transcription-coupled mechanisms for mtDNA maintenance might exist and that the inhibition of POLRMT by mtDNA damage may contribute to mitochondrial dysfunction in human aging and disease. Since the mechanisms of POLRMT initiation and elongation are still being refined and the mitochondrial promoters appear to impart different characteristics to POLRMT initiation, the study of POLRMT reaction to DNA damage must be conducted in a way that allows the findings to be placed into context of promoter-driven POLRMT function on the mtDNA chromosome [232, 233]. With the apparent absence of NER-like mechanisms in mitochondria, mtDNA damage by endogenous aldehydes may pose a significant threat to mtDNA gene expression, as most other oxidative lesions are repaired by BER.

The M1dG adduct formed by MDA nearly halts transcription by RNAP II and is a substantial impediment to T7RNAP [236]. To characterize the encounter of POLRMT with the M1dG, our group employed purified human POLRMT, its transcription factors, TFAM and TFB2M, and DNA templates containing a site-specific adduct placed in either the transcribed or non-transcribed strand downstream of either the LSP or HSP1 mitochondrial chromosome promoter sequences [234]. The exocyclic ring of M1dG opens to form an acyclic adduct, N2-OPdG, when it is paired with C; therefore, the adduct was placed in the template opposite either C or T in order to elucidate the structural effects of N2-OPdG and M1dG, respectively, on POLRMT transcript elongation. The acyclic N2-OPdG arrested >60% of POLRMT complexes, which was at least two-fold greater than T7RNAP arrest in a similar promoter-driven context [236]. Likewise, M1dG blocked >80% of POLRMT complexes, again twice exceeding the extent of T7RNAP arrest. These findings indicate that structural differences in the active sites of elongating T7RNAP and POLRMT result in different responses of the enzymes to DNA damage [237]. In fact, POLRMT sensitivity to the N2-OPdG and M1dG adduct conformations more closely resembled that of RNAP II, a multi-subunit enzyme with transcription factor requirements for initiation and elongation [236]. Interestingly, the sensitivity of POLRMT to the aldehyde adducts appeared to be dependent upon the initiating promoter [232234]. POLRMT displayed a 10% greater degree of transcriptional arrest at both N2-OPdG and M1dG following initiation at LSP than after HSP1 initiation. This finding, in conjunction with the recent studies showing that LSP and HSP1 differ in their response to TFAM during POLRMT initiation, suggests that POLRMT may adopt promoter-specific conformations for elongation. This hypothesis may be addressed through experiments using templates containing PdG, a saturated analog of M1dG, that arrests >95% of POLRMT in stable elongation complexes with DNA [234]. Biochemical mapping of arrested POLRMT complexes at PdG following initiation at LSP or HSP1 should provide structural and mechanistic insight into POLRMT elongation. The fact that POLRMT complexes can bypass N2-OPdG, the form of the aldehyde adduct that would be present in double-stranded mtDNA, suggests that transcriptional mutagenesis is likely during mitochondrial RNA synthesis. The characteristics of the errant transcripts generated from POLRMT insertion at N2-OPdG may also be readily addressed using the reconstituted system.

As discussed above, oxidized nucleotides, such as 8-oxoGTP, are likely the most prevalent type of aberrant nucleotides in mitochondria. However, NRTIs and other therapeutic nucleoside analogs infiltrate the substrate pools for mitochondrial replication and transcription. Indeed, analogs that were developed for the treatment of RNA viral infections, but were found to have unacceptable toxicity for clinical use, are substrates for POLRMT. The antivirals inhibit transcript elongation or completely terminate RNA synthesis in a manner that correlates with their structure (J. J. Arnold, personal communication). These findings may guide the design of new nucleoside analogs, which may be readily tested in the POLRMT reconstituted system, in order to develop therapies with fewer undesired effects on patients.

In addition to its effects on POLRMT, DNA damage also alters the nucleic acid binding properties of TFAM, a dual HMG-box transcription factor that forms nucleoids and interacts with a host of other proteins involved in DNA transactions [23]. It preferentially binds to DNA containing either an 8-oxo-dG lesion or a cisplatin intrastrand crosslink and appears to inhibit the excision repair of these forms of DNA damage [84, 179, 195, 238]. A purpose for TFAM inhibition of excision repair is unclear at this time; however, TFAM may serve to balance mtDNA repair and gene expression on mitochondrial chromosomes for maintenance of the total mtDNA population. TFAM dissociates from DNA in the presence of p53, which has been shown to translocate to mitochondria during cell stress [225]. Since p53 also participates in mitochondrial BER, an interplay between p53 and TFAM may determine whether damaged mtDNA chromosomes are repaired or degraded [239]. TFAM may associate more strongly with oxidatively damaged chromosomes to sequester them into nucleoids and prevent their transcription until p53 releases TFAM from the DNA allowing BER to occur, thus explaining why TFAM inhibits 8-oxodG incision. This idea is plausible based on the nucleoid heterogeneity of mammalian cells and the observation that nucleoids with lesser amounts of TFAM have greater replication activity. A threshold level of TFAM per chromosome may exist for an optimal mtDNA topology that allows polymerase binding and translocation. Therefore, in addition to directly inhibiting RNA synthesis by POLRMT, DNA damage could alter mitochondrial gene expression through its effects on the distribution of TFAM.

6. Conclusion

Over three decades ago researchers began to consider the adverse effects of DNA damage on mitochondria and the ramifications of drug therapies on the mitochondrial nucleotide pool. Today, advances in biophysical techniques allow us to detect and even measure the levels of damaged DNA bases in mitochondria. We now know that both endogenous and environmental agents, including drug therapies, pose a threat to mitochondrial health by their chemical modification of mtDNA or through their incorporation by mitochondrial polymerases. Recent discoveries in mitochondrial biology include the elucidation of the repair pathways in and the analysis of DNA damage effects on DNA pol γ and POLRMT. This research has shown numerous insults to mtDNA may interfere with mitochondrial biogenesis and gene expression in a tissue-specific way. For tissues that are highly dependent on oxidative phosphorylation, such as brain, muscle and kidney, the metabolic processes in mitochondria generate DNA lesions that cannot be repaired and interfere with mtDNA replication and transcription. Other polymerase-disrupting adducts are produced by environmental agents that are likely to have greatest effects in the epithelium of the respiratory tract, liver and gut. Additionally, therapies targeted against viruses and human cancers also interfere with mitochondrial polymerase function and inadvertently produce harmful side effects. Research focused on understanding the prevalence and repair of the many forms of DNA damage in human tissues will not only advance our understanding of DNA repair, but also of environmental and drug toxicology and of the process of human aging and disease progression. Findings in this field, taken together with advances in the study of nuclear-mitochondrial signaling in response to cellular redox state and energetics, will provide the most complete picture of how mitochondrial DNA repair, dynamics, biogenesis and destruction act together to maintain mtDNA integrity and gene expression to support cellular function.

Highlights.

  • Mitochondrial DNA is susceptible to endogenous and environmental damage.

  • Mitochondrial lack the full cohort of nuclear DNA repair mechanisms.

  • Persistent mtDNA damage poses a threat to mitochondrial gene expression.

  • Mitochondrial polymerase disruption by mtDNA damage may underlie human disease and environmental toxicity.

Acknowledgments

The author is supported by the NIH/National Institute of General Medical Sciences with grant R15GM087681 and is grateful to Dr. Richard McCann for his critical reading of the manuscript and to Drs. Bill Copeland and Rajesh Kasiviswanathan for their assistance in preparing Table 2.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Boveris A. Mitochondrial production of superoxide radical and hydrogen peroxide. Adv Exp Med Biol. 1977;78:67–82. doi: 10.1007/978-1-4615-9035-4_5. [DOI] [PubMed] [Google Scholar]
  • 2.Halliwell B, Gutteridge JM. Free radicals, lipid peroxidation, and cell damage. Lancet. 1984;2:1095. doi: 10.1016/s0140-6736(84)91530-7. [DOI] [PubMed] [Google Scholar]
  • 3.Brand MD. The sites and topology of mitochondrial superoxide production. Exp Gerontol. 2010;45:466–472. doi: 10.1016/j.exger.2010.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Roede JR, Jones DP. Reactive species and mitochondrial dysfunction: mechanistic significance of 4-hydroxynonenal. Environ Mol Mutagen. 2010;51:380–390. doi: 10.1002/em.20553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Cadet J, Douki T, Ravanat JL. Oxidatively generated base damage to cellular DNA. Free Radic Biol Med. 2010;49:9–21. doi: 10.1016/j.freeradbiomed.2010.03.025. [DOI] [PubMed] [Google Scholar]
  • 6.Yin H, Xu L, Porter NA. Free radical lipid peroxidation: mechanisms and analysis. Chem Rev. 2011;111:5944–5972. doi: 10.1021/cr200084z. [DOI] [PubMed] [Google Scholar]
  • 7.Swenberg JA, Lu K, Moeller BC, Gao L, Upton PB, Nakamura J, Starr TB. Endogenous versus exogenous DNA adducts: their role in carcinogenesis, epidemiology, and risk assessment. Toxicol Sci. 2011;120(Suppl 1):S130–145. doi: 10.1093/toxsci/kfq371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Liu P, Demple B. DNA repair in mammalian mitochondria: Much more than we thought? Environ Mol Mutagen. 2010;51:417–426. doi: 10.1002/em.20576. [DOI] [PubMed] [Google Scholar]
  • 9.St-Pierre J, Buckingham JA, Roebuck SJ, Brand MD. Topology of superoxide production from different sites in the mitochondrial electron transport chain. J Biol Chem. 2002;277:44784–44790. doi: 10.1074/jbc.M207217200. [DOI] [PubMed] [Google Scholar]
  • 10.Basu AK, O’Hara SM, Valladier P, Stone K, Mols O, Marnett LJ. Identification of adducts formed by reaction of guanine nucleosides with malondialdehyde and structurally related aldehydes. Chem Res Toxicol. 1988;1:53–59. doi: 10.1021/tx00001a010. [DOI] [PubMed] [Google Scholar]
  • 11.Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med. 1991;11:81–128. doi: 10.1016/0891-5849(91)90192-6. [DOI] [PubMed] [Google Scholar]
  • 12.Dedon PC, Plastaras JP, Rouzer CA, Marnett LJ. Indirect mutagenesis by oxidative DNA damage: formation of the pyrimidopurinone adduct of deoxyguanosine by base propenal. Proc Natl Acad Sci U S A. 1998;95:11113–11116. doi: 10.1073/pnas.95.19.11113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Voulgaridou GP, Anestopoulos I, Franco R, Panayiotidis MI, Pappa A. DNA damage induced by endogenous aldehydes: current state of knowledge. Mutat Res. 2011;711:13–27. doi: 10.1016/j.mrfmmm.2011.03.006. [DOI] [PubMed] [Google Scholar]
  • 14.Colbeau A, Nachbaur J, Vignais PM. Enzymic characterization and lipid composition of rat liver subcellular membranes. Biochim Biophys Acta. 1971;249:462–492. doi: 10.1016/0005-2736(71)90123-4. [DOI] [PubMed] [Google Scholar]
  • 15.Schlame M, Ren M, Xu Y, Greenberg ML, Haller I. Molecular symmetry in mitochondrial cardiolipins. Chem Phys Lipids. 2005;138:38–49. doi: 10.1016/j.chemphyslip.2005.08.002. [DOI] [PubMed] [Google Scholar]
  • 16.Liu W, Porter NA, Schneider C, Brash AR, Yin H. Formation of 4-hydroxynonenal from cardiolipin oxidation: Intramolecular peroxyl radical addition and decomposition. Free Radic Biol Med. 2011;50:166–178. doi: 10.1016/j.freeradbiomed.2010.10.709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Jones DP. Radical-free biology of oxidative stress. Am J Physiol Cell Physiol. 2008;295:C849–868. doi: 10.1152/ajpcell.00283.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Pakendorf B, Stoneking M. Mitochondrial DNA and human evolution. Annu Rev Genomics Hum Genet. 2005;6:165–183. doi: 10.1146/annurev.genom.6.080604.162249. [DOI] [PubMed] [Google Scholar]
  • 19.Torroni A, Achilli A, Macaulay V, Richards M, Bandelt HJ. Harvesting the fruit of the human mtDNA tree. Trends Genet. 2006;22:339–345. doi: 10.1016/j.tig.2006.04.001. [DOI] [PubMed] [Google Scholar]
  • 20.Poole JC, Procaccio V, Brandon MC, Merrick G, Wallace DC. Multiplex analysis of mitochondrial DNA pathogenic and polymorphic sequence variants. Biol Chem. 2010;391:1115–1130. doi: 10.1515/BC.2010.125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rose G, Romeo G, Dato S, Crocco P, Bruni AC, Hervonen A, Majamaa K, Sevini F, Franceschi C, Passarino G. Somatic point mutations in mtDNA control region are influenced by genetic background and associated with healthy aging: a GEHA study. PLoS One. 2010;5:e13395. doi: 10.1371/journal.pone.0013395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Greaves LC, Reeve AK, Taylor RW, Turnbull DM. Mitochondrial DNA and disease. J Pathol. 2012;226:274–286. doi: 10.1002/path.3028. [DOI] [PubMed] [Google Scholar]
  • 23.Bogenhagen DF. Mitochondrial DNA nucleoid structure. Biochim Biophys Acta. 2012 doi: 10.1016/j.bbagrm.2011.11.005. [DOI] [PubMed] [Google Scholar]
  • 24.Holt IJ. Zen and the art of mitochondrial DNA maintenance. Trends Genet. 2010;26:103–109. doi: 10.1016/j.tig.2009.12.011. [DOI] [PubMed] [Google Scholar]
  • 25.Chan SW, Dedon PC. The biological and metabolic fates of endogenous DNA damage products. J Nucleic Acids. 2010;2010:929047. doi: 10.4061/2010/929047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Haugen AC, Di Prospero NA, Parker JS, Fannin RD, Chou J, Meyer JN, Halweg C, Collins JB, Durr A, Fischbeck K, Van Houten B. Altered gene expression and DNA damage in peripheral blood cells from Friedreich’s ataxia patients: cellular model of pathology. PLoS Genet. 2010;6:el000812. doi: 10.1371/journal.pgen.1000812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Beckman KB, Ames BN. Endogenous oxidative damage of mtDNA. Mutat Res. 1999;424:51–58. doi: 10.1016/s0027-5107(99)00007-x. [DOI] [PubMed] [Google Scholar]
  • 28.Anson RM, Senturker S, Dizdaroglu M, Bohr VA. Measurement of oxidatively induced base lesions in liver from Wistar rats of different ages. Free Radic Biol Med. 1999;27:456–462. doi: 10.1016/s0891-5849(99)00091-x. [DOI] [PubMed] [Google Scholar]
  • 29.De Bont R, van Larebeke N. Endogenous DNA damage in humans: a review of quantitative data. Mutagenesis. 2004;19:169–185. doi: 10.1093/mutage/geh025. [DOI] [PubMed] [Google Scholar]
  • 30.Trapp C, McCullough AK, Epe B. The basal levels of 8-oxoG and other oxidative modifications in intact mitochondrial DNA are low even in repair-deficient (Oggl(−/−)/Csb(−/−)) mice. Mutat Res. 2007;625:155–163. doi: 10.1016/j.mrfmmm.2007.06.006. [DOI] [PubMed] [Google Scholar]
  • 31.Jaruga P, Dizdaroglu M. 8,5′-Cyclopurine-2′-deoxynucleosides in DNA: mechanisms of formation, measurement, repair and biological effects. DNA Repair (Amst) 2008;7:1413–1425. doi: 10.1016/j.dnarep.2008.06.005. [DOI] [PubMed] [Google Scholar]
  • 32.Jaruga P, Kirkali G, Dizdaroglu M. Measurement of formamidopyrimidines in DNA. Free Radic Biol Med. 2008;45:1601–1609. doi: 10.1016/j.freeradbiomed.2008.09.019. [DOI] [PubMed] [Google Scholar]
  • 33.Swenberg JA, Fryar-Tita E, Jeong YC, Boysen G, Starr T, Walker VE, Albertini RJ. Biomarkers in toxicology and risk assessment: informing critical dose-response relationships. Chem Res Toxicol. 2008;21:253–265. doi: 10.1021/tx700408t. [DOI] [PubMed] [Google Scholar]
  • 34.Taghizadeh K, McFaline JL, Pang B, Sullivan M, Dong M, Plummer E, Dedon PC. Quantification of DNA damage products resulting from deamination, oxidation and reaction with products of lipid peroxidation by liquid chromatography isotope dilution tandem mass spectrometry. Nat Protoc. 2008;3:1287–1298. doi: 10.1038/nprot.2008.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Dizdaroglu M, Kirkali G, Jaruga P. Formamidopyrimidines in DNA: mechanisms of formation, repair, and biological effects. Free Radic Biol Med. 2008;45:1610–1621. doi: 10.1016/j.freeradbiomed.2008.07.004. [DOI] [PubMed] [Google Scholar]
  • 36.Hunter SE, Jung D, Di Giulio RT, Meyer JN. The QPCR assay for analysis of mitochondrial DNA damage, repair, and relative copy number. Methods. 2010;51:444–451. doi: 10.1016/j.ymeth.2010.01.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Garcia CC, Freitas FP, Di Mascio P, Medeiros MH. Ultrasensitive simultaneous quantification of 1,N2-etheno-2′-deoxyguanosine and 1,N2-propano-2′-deoxyguanosine in DNA by an online liquid chromatography-electrospray tandem mass spectrometry assay. Chem Res Toxicol. 2010;23:1245–1255. doi: 10.1021/tx1001018. [DOI] [PubMed] [Google Scholar]
  • 38.Nair J, Nair UJ, Sun X, Wang Y, Arab K, Bartsch H. Quantifying etheno-DNA adducts in human tissues, white blood cells, and urine by ultrasensitive (32)P-postlabeling and immunohistochemistry. Methods Mol Biol. 2011;682:189–205. doi: 10.1007/978-1-60327-409-8_14. [DOI] [PubMed] [Google Scholar]
  • 39.Cadet J, Douki T, Ravanat JL. Measurement of oxidatively generated base damage in cellular DNA. Mutat Res. 2011;711:3–12. doi: 10.1016/j.mrfmmm.2011.02.004. [DOI] [PubMed] [Google Scholar]
  • 40.Mecocci P, MacGarvey U, Kaufman AE, Koontz D, Shoffner JM, Wallace DC, Beal MF. Oxidative damage to mitochondrial DNA shows marked age-dependent increases in human brain. Ann Neurol. 1993;34:609–616. doi: 10.1002/ana.410340416. [DOI] [PubMed] [Google Scholar]
  • 41.Mecocci P, MacGarvey U, Beal MF. Oxidative damage to mitochondrial DNA is increased in Alzheimer’s disease. Ann Neurol. 1994;36:747–751. doi: 10.1002/ana.410360510. [DOI] [PubMed] [Google Scholar]
  • 42.Higuchi Y, Linn S. Purification of all forms of HeLa cell mitochondrial DNA and assessment of damage to it caused by hydrogen peroxide treatment of mitochondria or cells. J Biol Chem. 1995;270:7950–7956. doi: 10.1074/jbc.270.14.7950. [DOI] [PubMed] [Google Scholar]
  • 43.Taffe BG, Larminat F, Laval J, Croteau DL, Anson RM, Bohr VA. Gene-specific nuclear and mitochondrial repair of formamidopyrimidine DNA glycosylase-sensitive sites in Chinese hamster ovary cells. Mutat Res. 1996;364:183–192. doi: 10.1016/s0921-8777(96)00031-6. [DOI] [PubMed] [Google Scholar]
  • 44.Palmeira CM, Serrano J, Kuehl DW, Wallace KB. Preferential oxidation of cardiac mitochondrial DNA following acute intoxication with doxorubicin. Biochim Biophys Acta. 1997;1321:101–106. doi: 10.1016/s0005-2728(97)00055-8. [DOI] [PubMed] [Google Scholar]
  • 45.Suter M, Richter C. Fragmented mitochondrial DNA is the predominant carrier of oxidized DNA bases. Biochemistry. 1999;38:459–464. doi: 10.1021/bi9811922. [DOI] [PubMed] [Google Scholar]
  • 46.Lezza AM, Mecocci P, Cormio A, Beal MF, Cherubini A, Cantatore P, Senin U, Gadaleta MN. Mitochondrial DNA 4977 bp deletion and OH8dG levels correlate in the brain of aged subjects but not Alzheimer’s disease patients. FASEB J. 1999;13:1083–1088. doi: 10.1096/fasebj.13.9.1083. [DOI] [PubMed] [Google Scholar]
  • 47.de Souza-Pinto NC, Eide L, Hogue BA, Thybo T, Stevnsner T, Seeberg E, Klungland A, Bohr VA. Repair of 8-oxodeoxyguanosine lesions in mitochondrial dna depends on the oxoguanine dna glycosylase (OGG1) gene and 8-oxoguanine accumulates in the mitochondrial dna of OGGl-defective mice. Cancer Res. 2001;61:5378–5381. [PubMed] [Google Scholar]
  • 48.Kakimoto M, Inoguchi T, Sonta T, Yu HY, Imamura M, Etoh T, Hashimoto T, Nawata H. Accumulation of 8-hydroxy-2′-deoxyguanosine and mitochondrial DNA deletion in kidney of diabetic rats. Diabetes. 2002;51:1588–1595. doi: 10.2337/diabetes.51.5.1588. [DOI] [PubMed] [Google Scholar]
  • 49.Wang J, Xiong S, Xie C, Markesbery WR, Lovell MA. Increased oxidative damage in nuclear and mitochondrial DNA in Alzheimer’s disease. J Neurochem. 2005;93:953–962. doi: 10.1111/j.1471-4159.2005.03053.x. [DOI] [PubMed] [Google Scholar]
  • 50.Jeong YC, Nakamura J, Upton PB, Swenberg JA. Pyrimido[1,2-a]-purin-10(3H)-one, M1G, is less prone to artifact than base oxidation. Nucleic Acids Res. 2005;33:6426–6434. doi: 10.1093/nar/gki944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Nair J, Strand S, Frank N, Knauft J, Wesch H, Galle PR, Bartsch H. Apoptosis and age-dependant induction of nuclear and mitochondrial etheno-DNA adducts in Long-Evans Cinnamon (LEC) rats: enhanced DNA damage by dietary curcumin upon copper accumulation. Carcinogenesis. 2005;26:1307–1315. doi: 10.1093/carcin/bgi073. [DOI] [PubMed] [Google Scholar]
  • 52.Nakamoto H, Kaneko T, Tahara S, Hayashi E, Naito H, Radak Z, Goto S. Regular exercise reduces 8-oxodG in the nuclear and mitochondrial DNA and modulates the DNA repair activity in the liver of old rats. Exp Gerontol. 2007;42:287–295. doi: 10.1016/j.exger.2006.11.006. [DOI] [PubMed] [Google Scholar]
  • 53.Yakes FM, Van Houten B. Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc Natl Acad Sci U S A. 1997;94:514–519. doi: 10.1073/pnas.94.2.514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Henderson PT, Delaney JC, Gu F, Tannenbaum SR, Essigmann JM. Oxidation of 7,8-dihydro-8-oxoguanine affords lesions that are potent sources of replication errors in vivo. Biochemistry. 2002;41:914–921. doi: 10.1021/bi0156355. [DOI] [PubMed] [Google Scholar]
  • 55.Henderson PT, Delaney JC, Muller JG, Neeley WL, Tannenbaum SR, Burrows CJ, Essigmann JM. The hydantoin lesions formed from oxidation of 7,8-dihydro-8-oxoguanine are potent sources of replication errors in vivo. Biochemistry. 2003;42:9257–9262. doi: 10.1021/bi0347252. [DOI] [PubMed] [Google Scholar]
  • 56.Redrejo-Rodriguez M, Saint-Pierre C, Couve S, Mazouzi A, Ishchenko AA, Gasparutto D, Saparbaev M. New insights in the removal of the hydantoins, oxidation product of pyrimidines, via the base excision and nucleotide incision repair pathways. PLoS One. 2011;6:e21039. doi: 10.1371/journal.pone.0021039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Song S, Pursell ZF, Copeland WC, Longley MJ, Kunkel TA, Mathews CK. DNA precursor asymmetries in mammalian tissue mitochondria and possible contribution to mutagenesis through reduced replication fidelity. Proc Natl Acad Sci USA. 2005;102:4990–4995. doi: 10.1073/pnas.0500253102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Pursell ZF, McDonald JT, Mathews CK, Kunkel TA. Trace amounts of 8-oxo-dGTP in mitochondrial dNTP pools reduce DNA polymerase gamma replication fidelity. Nucleic Acids Res. 2008;36:2174–2181. doi: 10.1093/nar/gkn062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Nakabeppu Y, Sakumi K, Sakamoto K, Tsuchimoto D, Tsuzuki T, Nakatsu Y. Mutagenesis and carcinogenesis caused by the oxidation of nucleic acids. Biol Chem. 2006;387:373–379. doi: 10.1515/BC.2006.050. [DOI] [PubMed] [Google Scholar]
  • 60.Rampazzo C, Miazzi C, Franzolin E, Pontarin G, Ferraro P, Frangini M, Reichard P, Bianchi V. Regulation by degradation, a cellular defense against deoxyribonucleotide pool imbalances. Mutat Res. 2010;703:2–10. doi: 10.1016/j.mrgentox.2010.06.002. [DOI] [PubMed] [Google Scholar]
  • 61.Lindahl T, Nyberg B. Rate of depurination of native deoxyribonucleic acid. Biochemistry. 1972;11:3610–3618. doi: 10.1021/bi00769a018. [DOI] [PubMed] [Google Scholar]
  • 62.Nakamura J, Swenberg JA. Endogenous apurinic/apyrimidinic sites in genomic DNA of mammalian tissues. Cancer Res. 1999;59:2522–2526. [PubMed] [Google Scholar]
  • 63.Atamna H, Cheung I, Ames BN. A method for detecting abasic sites in living cells: age-dependent changes in base excision repair. Proc Natl Acad Sci U S A. 2000;97:686–691. doi: 10.1073/pnas.97.2.686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Shokolenko I, Venediktova N, Bochkareva A, Wilson GL, Alexeyev MF. Oxidative stress induces degradation of mitochondrial DNA. Nucleic Acids Res. 2009;37:2539–2548. doi: 10.1093/nar/gkp100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Dedon PC. The chemical toxicology of 2-deoxyribose oxidation in DNA. Chem Res Toxicol. 2008;21:206–219. doi: 10.1021/tx700283c. [DOI] [PubMed] [Google Scholar]
  • 66.Nair U, Bartsch H, Nair J. Lipid peroxidation-induced DNA damage in cancer-prone inflammatory diseases: a review of published adduct types and levels in humans. Free Radic Biol Med. 2007;43:1109–1120. doi: 10.1016/j.freeradbiomed.2007.07.012. [DOI] [PubMed] [Google Scholar]
  • 67.Lu K, Ye W, Zhou L, Collins LB, Chen X, Gold A, Ball LM, Swenberg JA. Structural characterization of formaldehyde-induced cross-links between amino acids and deoxynucleosides and their oligomers. J Am Chem Soc. 2010;132:3388–3399. doi: 10.1021/ja908282f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wang M, Cheng G, Balbo S, Carmella SG, Villalta PW, Hecht SS. Clear differences in levels of a formaldehyde-DNA adduct in leukocytes of smokers and nonsmokers. Cancer Res. 2009;69:7170–7174. doi: 10.1158/0008-5472.CAN-09-1571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lu K, Moeller B, Doyle-Eisele M, McDonald J, Swenberg JA. Molecular dosimetry of N2-hydroxymethyl-dG DNA adducts in rats exposed to formaldehyde. Chem Res Toxicol. 2011;24:159–161. doi: 10.1021/tx1003886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Minko IG, Kozekov ID, Harris TM, Rizzo CJ, Lloyd RS, Stone MP. Chemistry and biology of DNA containing 1,N(2)-deoxyguanosine adducts of the alpha,beta-unsaturated aldehydes acrolein, crotonaldehyde, and 4-hydroxynonenal. Chem Res Toxicol. 2009;22:759–778. doi: 10.1021/tx9000489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Stone MP, Cho YJ, Huang H, Kim HY, Kozekov ID, Kozekova A, Wang H, Minko IG, Lloyd RS, Harris TM, Rizzo CJ. Interstrand DNA cross-links induced by alpha,beta-unsaturated aldehydes derived from lipid peroxidation and environmental sources. Acc Chem Res. 2008;41:793–804. doi: 10.1021/ar700246x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Huang H, Wang H, Voehler MW, Kozekova A, Rizzo CJ, McCullough AK, Lloyd RS, Stone MP. gamma-Hydroxy-1, N2-propano-2′-deoxyguanosine DNA adduct conjugates the N-terminal amine of the KWKK peptide via a carbinolamine linkage. Chem Res Toxicol. 2011;24:1123–1133. doi: 10.1021/tx200113n. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zhang H, Barcelo JM, Lee B, Kohlhagen G, Zimonjic DB, Popescu NC, Pommier Y. Human mitochondrial topoisomerase I. Proc Natl Acad Sci U S A. 2001;98:10608–10613. doi: 10.1073/pnas.191321998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Wang Y, Lyu YL, Wang JC. Dual localization of human DNA topoisomerase Illalpha to mitochondria and nucleus. Proc Natl Acad Sci U S A. 2002;99:12114–12119. doi: 10.1073/pnas.192449499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zhang H, Pommier Y. Mitochondrial topoisomerase I sites in the regulatory D-loop region of mitochondrial DNA. Biochemistry. 2008;47:11196–11203. doi: 10.1021/bi800774b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Pommier Y. DNA topoisomerase I inhibitors: chemistry, biology, and interfacial inhibition. Chem Rev. 2009;109:2894–2902. doi: 10.1021/cr900097c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Sordet O, Khan QA, Pommier Y. Apoptotic topoisomerase I-DNA complexes induced by oxygen radicals and mitochondrial dysfunction. Cell Cycle. 2004;3:1095–1097. [PubMed] [Google Scholar]
  • 78.Debethune L, Kohlhagen G, Grandas A, Pommier Y. Processing of nucleopeptides mimicking the topoisomerase I-DNA covalent complex by tyrosyl-DNA phosphodiesterase. Nucleic Acids Res. 2002;30:1198–1204. doi: 10.1093/nar/30.5.1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Das BB, Dexheimer TS, Maddali K, Pommier Y. Role of tyrosyl-DNA phosphodiesterase (TDP1) in mitochondria. Proc Natl Acad Sci USA. 2010;107:19790–19795. doi: 10.1073/pnas.1009814107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Takashima H, Boerkoel CF, John J, Saifi GM, Salih MA, Armstrong D, Mao Y, Quiocho FA, Roa BB, Nakagawa M, Stockton DW, Lupski JR. Mutation of TDP1, encoding a topoisomerase I-dependent DNA damage repair enzyme, in spinocerebellar ataxia with axonal neuropathy. Nat Genet. 2002;32:267–272. doi: 10.1038/ng987. [DOI] [PubMed] [Google Scholar]
  • 81.El-Khamisy SF, Saifi GM, Weinfeld M, Johansson F, Helleday T, Lupski JR, Caldecott KW. Defective DNA single-strand break repair in spinocerebellar ataxia with axonal neuropathy-1. Nature. 2005;434:108–113. doi: 10.1038/nature03314. [DOI] [PubMed] [Google Scholar]
  • 82.Kasiviswanathan R, Collins TR, Copeland WC. The interface of transcription and DNA replication in the mitochondria. Biochim Biophys Acta. 2011 doi: 10.1016/j.bbagrm.2011.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Niranjan BG, Bhat NK, Avadhani NG. Preferential attack of mitochondrial DNA by aflatoxin Bl during hepatocarcinogenesis. Science. 1982;215:73–75. doi: 10.1126/science.6797067. [DOI] [PubMed] [Google Scholar]
  • 84.Vaisman A, Lim SE, Patrick SM, Copeland WC, Hinkle DC, Turchi JJ, Chaney SG. Effect of DNA polymerases and high mobility group protein 1 on the carrier ligand specificity for translesion synthesis past platinum-DNA adducts. Biochemistry. 1999;38:11026–11039. doi: 10.1021/bi9909187. [DOI] [PubMed] [Google Scholar]
  • 85.Graziewicz MA, Sayer JM, Jerina DM, Copeland WC. Nucleotide incorporation by human DNA polymerase gamma opposite benzo[a]pyrene and benzo[c]phenanthrene diol epoxide adducts of deoxyguanosine and deoxyadenosine. Nucleic Acids Res. 2004;32:397–405. doi: 10.1093/nar/gkh213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kasiviswanathan R, Gustafson MA, Copeland WC, Meyer JN. Human mitochondrial DNA polymerase gamma exhibits potential for bypass and mutagenesis at UV-induced cyclobutane thymine dimers. J Biol Chem. 2012;287:9222–9229. doi: 10.1074/jbc.M111.306852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Swenberg JA, Bogdanffy MS, Ham A, Holt S, Kim A, Morinello EJ, Ranasinghe A, Scheller N, Upton PB. Formation and repair of DNA adducts in vinyl chloride- and vinyl fluoride-induced carcinogenesis. IARC Sci Publ. 1999:29–43. [PubMed] [Google Scholar]
  • 88.Wang M, Mclntee EJ, Cheng G, Shi Y, Villalta PW, Hecht SS. Identification of DNA adducts of acetaldehyde. Chem Res Toxicol. 2000;13:1149–1157. doi: 10.1021/tx000118t. [DOI] [PubMed] [Google Scholar]
  • 89.Inagaki S, Esaka Y, Goto M, Deyashiki Y, Sako M. LC-MS study on the formation of cyclic 1,N2-propano guanine adduct in the reactions of DNA with acetaldehyde in the presence of histone. Biol Pharm Bull. 2004;27:273–276. doi: 10.1248/bpb.27.273. [DOI] [PubMed] [Google Scholar]
  • 90.Matsuda T, Yabushita H, Kanaly RA, Shibutani S, Yokoyama A. Increased DNA damage in ALDH2-deficient alcoholics. Chem Res Toxicol. 2006;19:1374–1378. doi: 10.1021/tx060113h. [DOI] [PubMed] [Google Scholar]
  • 91.Yu HS, Oyama T, Isse T, Kitagawa K, Pham TT, Tanaka M, Kawamoto T. Formation of acetaldehyde-derived DNA adducts due to alcohol exposure. Chem Biol Interact. 2010;188:367–375. doi: 10.1016/j.cbi.2010.08.005. [DOI] [PubMed] [Google Scholar]
  • 92.Lao Y, Hecht SS. Synthesis and properties of an acetaldehyde-derived oligonucleotide interstrand cross-link. Chem Res Toxicol. 2005;18:711–721. doi: 10.1021/tx0497292. [DOI] [PubMed] [Google Scholar]
  • 93.Backer JM, Weinstein LB. Mitochondrial DNA is a major cellular target for a dihydrodiol-epoxide derivative of benzo[a]pyrene. Science. 1980;209:297–299. doi: 10.1126/science.6770466. [DOI] [PubMed] [Google Scholar]
  • 94.Backer JM, Weinstein IB. Interaction of benzo(a)pyrene and its dihydrodiol-epoxide derivative with nuclear and mitochondrial DNA in C3H10T 1/2 cell cultures. Cancer Res. 1982;42:2764–2769. [PubMed] [Google Scholar]
  • 95.Stairs PW, Guzelian PS, Van Tuyle GC. Benzo[a]pyrene differentially alters mitochondrial and nuclear DNA synthesis in primary hepatocyte cultures. Res Commun Chem Pathol Pharmacol. 1983;42:95–106. [PubMed] [Google Scholar]
  • 96.De Flora S, Balansky R, Scatolini L, Di Marco C, Gasparini L, Orlando M, Izzotti A. Adducts to nuclear DNA and mitochondrial DNA as biomarkers in chemoprevention. IARC Sci Publ. 1996:291–301. [PubMed] [Google Scholar]
  • 97.Stepanov I, Hecht SS. Mitochondrial DNA adducts in the lung and liver of F344 rats chronically treated with 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and (S)-4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol. Chem Res Toxicol. 2009;22:406–414. doi: 10.1021/tx800398x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Wei D, Maher VM, McCormick JJ. Site-specific rates of excision repair of benzo[a]pyrene diol epoxide adducts in the hypoxanthine phosphoribosyltransferase gene of human fibroblasts: correlation with mutation spectra. Proc Natl Acad Sci U S A. 1995;92:2204–2208. doi: 10.1073/pnas.92.6.2204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Hess MT, Gunz D, Luneva N, Geacintov NE, Naegeli H. Base pair conformation-dependent excision of benzo[a]pyrene diol epoxide-guanine adducts by human nucleotide excision repair enzymes. Mol Cell Biol. 1997;17:7069–7076. doi: 10.1128/mcb.17.12.7069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Lloyd DR, Hanawalt PC. p53-dependent global genomic repair of benzo[a]pyrene-7,8-diol-9,10-epoxide adducts in human cells. Cancer Res. 2000;60:517–521. [PubMed] [Google Scholar]
  • 101.Pommier Y, Kohlhagen G, Pourquier P, Sayer JM, Kroth H, Jerina DM. Benzo[a]pyrene diol epoxide adducts in DNA are potent suppressors of a normal topoisomerase I cleavage site and powerful inducers of other topoisomerase I cleavages. Proc Natl Acad Sci U S A. 2012;97:2040–2045. doi: 10.1073/pnas.040397497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Pommier Y, Laco GS, Kohlhagen G, Sayer JM, Kroth H, Jerina DM. Position-specific trapping of topoisomerase I-DNA cleavage complexes by intercalated benzo[a]-pyrene diol epoxide adducts at the 6-amino group of adenine. Proc Natl Acad Sci U S A. 2000;97:10739–10744. doi: 10.1073/pnas.190312697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Khan QA, Kohlhagen G, Marshall R, Austin CA, Kalena GP, Kroth H, Sayer JM, Jerina DM, Pommier Y. Position-specific trapping of topoisomerase II by benzo[a]pyrene diol epoxide adducts: implications for interactions with intercalating anticancer agents. Proc Natl Acad Sci USA. 2003;100:12498–12503. doi: 10.1073/pnas.2032456100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Schlezinger JJ, White RD, Stegeman JJ. Oxidative inactivation of cytochrome P-450 1A (CYP1A) stimulated by 3,3′,4,4′-tetrachlorobiphenyl: production of reactive oxygen by vertebrate CYP1As. Mol Pharmacol. 1999;56:588–597. doi: 10.1124/mol.56.3.588. [DOI] [PubMed] [Google Scholar]
  • 105.Jeong YC, Walker NJ, Burgin DE, Kissling G, Gupta M, Kupper L, Birnbaum LS, Swenberg JA. Accumulation of MldG DNA adducts after chronic exposure to PCBs, but not from acute exposure to polychlorinated aromatic hydrocarbons. Free Radic Biol Med. 2008;45:585–591. doi: 10.1016/j.freeradbiomed.2008.04.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Biswas G, Srinivasan S, Anandatheerthavarada HK, Avadhani NG. Dioxin-mediated tumor progression through activation of mitochondria-to-nucleus stress signaling. Proc Natl Acad Sci USA. 2008;105:186–191. doi: 10.1073/pnas.0706183104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Setzer B, Schlesier M, Thomas AK, Walker UA. Mitochondrial toxicity of nucleoside analogues in primary human lymphocytes. Antivir Ther. 2005;10:327–334. [PubMed] [Google Scholar]
  • 108.Lim SE, Copeland WC. Differential incorporation and removal of antiviral deoxynucleotides by human DNA polymerase gamma. J Biol Chem. 2001;276:23616–23623. doi: 10.1074/jbc.M101114200. [DOI] [PubMed] [Google Scholar]
  • 109.Lewis W, Day BJ, Copeland WC. Mitochondrial toxicity of NRTI antiviral drugs: an integrated cellular perspective. Nat Rev Drug Discov. 2003;2:812–822. doi: 10.1038/nrd1201. [DOI] [PubMed] [Google Scholar]
  • 110.Arnold JJ, Smidansky ED, Moustafa IM, Cameron CE. Human mitochondrial RNA polymerase: Structure-function, mechanism and inhibition. Biochim Biophys Acta. 2012 doi: 10.1016/j.bbagrm.2012.04.002. [DOI] [PubMed] [Google Scholar]
  • 111.Dedon PC, Goldberg IH. Free-radical mechanisms involved in the formation of sequence-dependent bistranded DNA lesions by the antitumor antibiotics bleomycin, neocarzinostatin, and calicheamicin. Chem Res Toxicol. 1992;5:311–332. doi: 10.1021/tx00027a001. [DOI] [PubMed] [Google Scholar]
  • 112.Kasper M, Barth K. Bleomycin and its role in inducing apoptosis and senescence in lung cells - modulating effects of caveolin-1. Curr Cancer Drug Targets. 2009;9:341–353. doi: 10.2174/156800909788166501. [DOI] [PubMed] [Google Scholar]
  • 113.Degryse AL, Lawson WE. Progress toward improving animal models for idiopathic pulmonary fibrosis. Am J Med Sci. 2011;341:444–449. doi: 10.1097/MAJ.0b013e31821aa000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Wang D, Lippard SJ. Cellular processing of platinum anticancer drugs. Nat Rev Drug Discov. 2005;4:307–320. doi: 10.1038/nrd1691. [DOI] [PubMed] [Google Scholar]
  • 115.Ayala-Torres S, Chen Y, Svoboda T, Rosenblatt J, Van Houten B. Analysis of gene-specific DNA damage and repair using quantitative polymerase chain reaction. Methods. 2000;22:135–147. doi: 10.1006/meth.2000.1054. [DOI] [PubMed] [Google Scholar]
  • 116.Todd RC, Lippard SJ. Inhibition of transcription by platinum antitumor compounds. Metallomics. 2009;1:280–291. doi: 10.1039/b907567d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Todd RC, Lippard SJ. Consequences of cisplatin binding on nucleosome structure and dynamics. Chem Biol. 2010;17:1334–1343. doi: 10.1016/j.chembiol.2010.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zhu G, Myint M, Ang WH, Song L, Lippard SJ. Monofunctional Platinum-DNA Adducts Are Strong Inhibitors of Transcription and Substrates for Nucleotide Excision Repair in Live Mammalian Cells. Cancer Res. 2012;72:790–800. doi: 10.1158/0008-5472.CAN-11-3151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Giurgiovich AJ, Diwan BA, Olivero OA, Anderson LM, Rice JM, Poirier MC. Elevated mitochondrial cisplatin-DNA adduct levels in rat tissues after transplacental cisplatin exposure. Carcinogenesis. 1997;18:93–96. doi: 10.1093/carcin/18.1.93. [DOI] [PubMed] [Google Scholar]
  • 120.Giurgiovich AJ, Anderson LM, Jones AB, Dove LF, Moskal TJ, Rice JM, Olivero OA, Poirier MC. Transplacental cisplatin exposure induces persistent fetal mitochondrial and genomic DNA damage in patas monkeys. Reprod Toxicol. 1997;11:95–100. doi: 10.1016/s0890-6238(96)00201-8. [DOI] [PubMed] [Google Scholar]
  • 121.Podratz JL, Knight AM, Ta LE, Staff NP, Gass JM, Genelin K, Schlattau A, Lathroum L, Windebank AJ. Cisplatin induced mitochondrial DNA damage in dorsal root ganglion neurons. Neurobiol Dis. 2011;41:661–668. doi: 10.1016/j.nbd.2010.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Ang WH, Myint M, Lippard SJ. Transcription inhibition by platinum-DNA cross-links in live mammalian cells. J Am Chem Soc. 2010;132:7429–7435. doi: 10.1021/ja101495v. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Zamble DB, Mu D, Reardon JT, Sancar A, Lippard SJ. Repair of cisplatin-- DNA adducts by the mammalian excision nuclease. Biochemistry. 1996;35:10004–10013. doi: 10.1021/bi960453+. [DOI] [PubMed] [Google Scholar]
  • 124.Reardon JT, Vaisman A, Chaney SG, Sancar A. Efficient nucleotide excision repair of cisplatin, oxaliplatin, and Bis-aceto-ammine-dichloro-cyclohexylamine-platinum(IV) (JM216) platinum intrastrand DNA diadducts. Cancer Res. 1999;59:3968–3971. [PubMed] [Google Scholar]
  • 125.Wang D, Hara R, Singh G, Sancar A, Lippard SJ. Nucleotide excision repair from site-specifically platinum-modified nucleosomes. Biochemistry. 2003;42:6747–6753. doi: 10.1021/bi034264k. [DOI] [PubMed] [Google Scholar]
  • 126.LeDoux SP, Wilson GL, Beecham EJ, Stevnsner T, Wassermann K, Bohr VA. Repair of mitochondrial DNA after various types of DNA damage in Chinese hamster ovary cells. Carcinogenesis. 1992;13:1967–1973. doi: 10.1093/carcin/13.11.1967. [DOI] [PubMed] [Google Scholar]
  • 127.Liu D, Croteau DL, Souza-Pinto N, Pitta M, Tian J, Wu C, Jiang H, Mustafa K, Keijzers G, Bohr VA, Mattson MP. Evidence that OGG1 glycosylase protects neurons against oxidative DNA damage and cell death under ischemic conditions. J Cereb Blood Flow Metab. 2011;31:680–692. doi: 10.1038/jcbfm.2010.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Elachouri G, Vidoni S, Zanna C, Pattyn A, Boukhaddaoui H, Gaget K, Yu-Wai-Man P, Gasparre G, Sarzi E, Delettre C, Olichon A, Loiseau D, Reynier P, Chinnery PF, Rotig A, Carelli V, Hamel CP, Rugolo M, Lenaers G. OPA1 links human mitochondrial genome maintenance to mtDNA replication and distribution. Genome Res. 2011;21:12–20. doi: 10.1101/gr.108696.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Gredilla R. DNA damage and base excision repair in mitochondria and their role in aging. J Aging Res. 2011;2010:257093. doi: 10.4061/2011/257093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Svilar D, Goellner EM, Almeida KH, Sobol RW. Base excision repair and lesion-dependent subpathways for repair of oxidative DNA damage. Antioxid Redox Signal. 2011;14:2491–2507. doi: 10.1089/ars.2010.3466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Sykora P, Wilson DM, 3rd, Bohr VA. Repair of persistent strand breaks in the mitochondrial genome. Mech Ageing Dev. 2012;133:169–175. doi: 10.1016/j.mad.2011.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Anson RM, Croteau DL, Stierum RH, Filburn C, Parsell R, Bohr VA. Homogenous repair of singlet oxygen-induced DNA damage in differentially transcribed regions and strands of human mitochondrial DNA. Nucleic Acids Res. 1998;26:662–668. doi: 10.1093/nar/26.2.662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Stierum RH, Dianov GL, Bohr VA. Single-nucleotide patch base excision repair of uracil in DNA by mitochondrial protein extracts. Nucleic Acids Res. 1999;27:3712–3719. doi: 10.1093/nar/27.18.3712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Akbari M, Visnes T, Krokan HE, Otterlei M. Mitochondrial base excision repair of uracil and AP sites takes place by single-nucleotide insertion and long-patch DNA synthesis. DNA Repair (Amst) 2008;7:605–616. doi: 10.1016/j.dnarep.2008.01.002. [DOI] [PubMed] [Google Scholar]
  • 135.Liu P, Qian L, Sung JS, de Souza-Pinto NC, Zheng L, Bogenhagen DF, Bohr VA, Wilson DM, 3rd, Shen B, Demple B. Removal of Oxidative DNA Damage via FENl-Dependent Long-Patch Base Excision Repair in Human Cell Mitochondria. Mol Cell Biol. 2008;28:4975–4987. doi: 10.1128/MCB.00457-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Szczesny B, Tann AW, Longley MJ, Copeland WC, Mitra S. Long patch base excision repair in mammalian mitochondrial genomes. J Biol Chem. 2008;283:26349–26356. doi: 10.1074/jbc.M803491200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Zheng L, Zhou M, Guo Z, Lu H, Qian L, Dai H, Qiu J, Yakubovskaya E, Bogenhagen DF, Demple B, Shen B. Human DNA2 is a mitochondrial nuclease/helicase for efficient processing of DNA replication and repair intermediates. Mol Cell. 2008;32:325–336. doi: 10.1016/j.molcel.2008.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Tann AW, Boldogh I, Meiss G, Qian W, Van Houten B, Mitra S, Szczesny B. Apoptosis induced by persistent single-strand breaks in mitochondrial genome: critical role of EXOG (5′-EXO/endonuclease) in their repair. J Biol Chem. 2011;286:31975–31983. doi: 10.1074/jbc.M110.215715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Mason PA, Matheson EC, Hall AG, Lightowlers RN. Mismatch repair activity in mammalian mitochondria. Nucleic Acids Res. 2003;31:1052–1058. doi: 10.1093/nar/gkg167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.de Souza-Pinto NC, Mason PA, Hashiguchi K, Weissman L, Tian J, Guay D, Lebel M, Stevnsner TV, Rasmussen LJ, Bohr VA. Novel DNA mismatch-repair activity involving YB-1 in human mitochondria. DNA Repair (Amst) 2009;8:704–719. doi: 10.1016/j.dnarep.2009.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Takao M, Zhang QM, Yonei S, Yasui A. Differential subcellular localization of human MutY homolog (hMYH) and the functional activity of adenine:8-oxoguanine DNA glycosylase. Nucleic Acids Res. 1999;27:3638–3644. doi: 10.1093/nar/27.18.3638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Ohtsubo T, Nishioka K, Imaiso Y, Iwai S, Shimokawa H, Oda H, Fujiwara T, Nakabeppu Y. Identification of human MutY homolog (hMYH) as a repair enzyme for 2-hydroxyadenine in DNA and detection of multiple forms of hMYH located in nuclei and mitochondria. Nucleic Acids Res. 2000;28:1355–1364. doi: 10.1093/nar/28.6.1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Kang D, Nishida J, Iyama A, Nakabeppu Y, Furuichi M, Fujiwara T, Sekiguchi M, Takeshige K. Intracellular localization of 8-oxo-dGTPase in human cells, with special reference to the role of the enzyme in mitochondria. J Biol Chem. 1995;270:14659–14665. doi: 10.1074/jbc.270.24.14659. [DOI] [PubMed] [Google Scholar]
  • 144.Nakabeppu Y. Molecular genetics and structural biology of human MutT homolog, MTH1. Mutat Res. 2001;477:59–70. doi: 10.1016/s0027-5107(01)00096-3. [DOI] [PubMed] [Google Scholar]
  • 145.Thyagarajan B, Padua RA, Campbell C. Mammalian mitochondria possess homologous DNA recombination activity. J Biol Chem. 1996;271:27536–27543. doi: 10.1074/jbc.271.44.27536. [DOI] [PubMed] [Google Scholar]
  • 146.Lakshmipathy U, Campbell C. Double strand break rejoining by mammalian mitochondrial extracts. Nucleic Acids Res. 1999;27:1198–1204. doi: 10.1093/nar/27.4.1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Bacman SR, Williams SL, Moraes CT. Intra- and inter-molecular recombination of mitochondrial DNA after in vivo induction of multiple double-strand breaks. Nucleic Acids Res. 2009;37:4218–4226. doi: 10.1093/nar/gkp348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Fukui H, Moraes CT. Mechanisms of formation and accumulation of mitochondrial DNA deletions in aging neurons. Hum Mol Genet. 2009;18:1028–1036. doi: 10.1093/hmg/ddn437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Singh DK, Ghosh AK, Croteau DL, Bohr VA. RecQ helicases in DNA double strand break repair and telomere maintenance. Mutat Res. 2011 doi: 10.1016/j.mrfmmm.2011.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Croteau DL, Rossi ML, Canugovi C, Tian J, Sykora P, Ramamoorthy M, Wang Z, Singh DK, Akbari M, Kasiviswanathan R, Copeland WC, Bohr VA. RECQL4 localizes to mitochondria and preserves mitochondrial DNA integrity. Aging Cell. 2012;11:456–466. doi: 10.1111/j.1474-9726.2012.00803.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Clayton DA, Doda JN, Friedberg EC. The absence of a pyrimidine dimer repair mechanism in mammalian mitochondria. Proc Natl Acad Sci U S A. 1974;71:2777–2781. doi: 10.1073/pnas.71.7.2777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Pascucci B, Versteegh A, van Hoffen A, van Zeeland AA, Mullenders LH, Dogliotti E. DNA repair of UV photoproducts and mutagenesis in human mitochondrial DNA. J Mol Biol. 1997;273:417–427. doi: 10.1006/jmbi.1997.1268. [DOI] [PubMed] [Google Scholar]
  • 153.Olivero OA, Chang PK, Lopez-Larraza DM, Semino-Mora MC, Poirier MC. Preferential formation and decreased removal of cisplatin-DNA adducts in Chinese hamster ovary cell mitochondrial DNA as compared to nuclear DNA. Mutat Res. 1997;391:79–86. doi: 10.1016/s0165-1218(97)00037-2. [DOI] [PubMed] [Google Scholar]
  • 154.Johnson KA, Fink SP, Marnett LJ. Repair of propanodeoxyguanosine by nucleotide excision repair in vivo and in vitro. J Biol Chem. 1997;272:11434–11438. doi: 10.1074/jbc.272.17.11434. [DOI] [PubMed] [Google Scholar]
  • 155.Brooks PJ, Wise DS, Berry DA, Kosmoski JV, Smerdon MJ, Somers RL, Mackie H, Spoonde AY, Ackerman EJ, Coleman K, Tarone RE, Robbins JH. The oxidative DNA lesion 8,5′-(S)-cyclo-2′-deoxyadenosine is repaired by the nucleotide excision repair pathway and blocks gene expression in mammalian cells. J Biol Chem. 2000;275:22355–22362. doi: 10.1074/jbc.M002259200. [DOI] [PubMed] [Google Scholar]
  • 156.Kuraoka I, Bender C, Romieu A, Cadet J, Wood RD, Lindahl T. Removal of oxygen free-radical-induced 5′,8-purine cyclodeoxynucleosides from DNA by the nucleotide excision-repair pathway in human cells. Proc Natl Acad Sci U S A. 2000;97:3832–3837. doi: 10.1073/pnas.070471597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Hang B, Chenna A, Rao S, Singer B. 1,N6-ethenoadenine and 3,N4-ethenocytosine are excised by separate human DNA glycosylases. Carcinogenesis. 1996;17:155–157. doi: 10.1093/carcin/17.1.155. [DOI] [PubMed] [Google Scholar]
  • 158.Saparbaev M, Langouet S, Privezentzev CV, Guengerich FP, Cai H, Elder RH, Laval J. 1,N(2)-ethenoguanine, a mutagenic DNA adduct, is a primary substrate of Escherichia coli mismatch-specific uracil-DNA glycosylase and human alkylpurine-DNA-N-glycosylase. J Biol Chem. 2002;277:26987–26993. doi: 10.1074/jbc.M111100200. [DOI] [PubMed] [Google Scholar]
  • 159.Lee CY, Delaney JC, Kartalou M, Lingaraju GM, Maor-Shoshani A, Essigmann JM, Samson LD. Recognition and processing of a new repertoire of DNA substrates by human 3-methyladenine DNA glycosylase (AAG) Biochemistry. 2009;48:1850–1861. doi: 10.1021/bi8018898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Harris JL, Jakob B, Taucher-Scholz G, Dianov GL, Becherel OJ, Lavin MF. Aprataxin, poly-ADP ribose polymerase 1 (PARP-1) and apurinic endonuclease 1 (APE1) function together to protect the genome against oxidative damage. Hum Mol Genet. 2009;18:4102–4117. doi: 10.1093/hmg/ddp359. [DOI] [PubMed] [Google Scholar]
  • 161.Schellenberg MJ, Williams RS. DNA end processing by polynucleotide kinase/phosphatase. Proc Natl Acad Sci USA. 2011;108:20855–20856. doi: 10.1073/pnas.1118214109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Coquelle N, Havali-Shahriari Z, Bernstein N, Green R, Glover JN. Structural basis for the phosphatase activity of polynucleotide kinase/phosphatase on single-and double-stranded DNA substrates. Proc Natl Acad Sci USA. 2011;108:21022–21027. doi: 10.1073/pnas.1112036108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Puebla-Osorio N, Lacey DB, Alt FW, Zhu C. Early embryonic lethality due to targeted inactivation of DNA ligase III. Mol Cell Biol. 2006;26:3935–3941. doi: 10.1128/MCB.26.10.3935-3941.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Simsek D, Furda A, Gao Y, Artus J, Brunet E, Hadjantonakis AK, Van Houten B, Shuman S, McKinnon PJ, Jasin M. Crucial role for DNA ligase III in mitochondria but not in Xrcc1-dependent repair. Nature. 2011;471:245–248. doi: 10.1038/nature09794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Rossi MN, Carbone M, Mostocotto C, Mancone C, Tripodi M, Maione R, Amati P. Mitochondrial localization of PARP-1 requires interaction with mitofilin and is involved in the maintenance of mitochondrial DNA integrity. J Biol Chem. 2009;284:31616–31624. doi: 10.1074/jbc.M109.025882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Tahbaz N, Subedi S, Weinfeld M. Role of polynucleotide kinase/phosphatase in mitochondrial DNA repair. Nucleic Acids Res. 2012;40:3484–3495. doi: 10.1093/nar/gkr1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Mandal SM, Hegde ML, Chatterjee A, Hegde PM, Szczesny B, Banerjee D, Boldogh I, Gao R, Falkenberg M, Gustafsson CM, Sarkar PS, Hazra TK. Role of human DNA glycosylase Nei-like 2 (NEIL2) and single strand break repair protein polynucleotide kinase 3′-phosphatase in maintenance of mitochondrial genome. J Biol Chem. 2012;287:2819–2829. doi: 10.1074/jbc.M111.272179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Aicardi J, Barbosa C, Andermann E, Andermann F, Morcos R, Ghanem Q, Fukuyama Y, Awaya Y, Moe P. Ataxia-ocular motor apraxia: a syndrome mimicking ataxia-telangiectasia. Ann Neurol. 1988;24:497–502. doi: 10.1002/ana.410240404. [DOI] [PubMed] [Google Scholar]
  • 169.Date H, Onodera O, Tanaka H, Iwabuchi K, Uekawa K, Igarashi S, Koike R, Hiroi T, Yuasa T, Awaya Y, Sakai T, Takahashi T, Nagatomo H, Sekijima Y, Kawachi I, Takiyama Y, Nishizawa M, Fukuhara N, Saito K, Sugano S, Tsuji S. Early-onset ataxia with ocular motor apraxia and hypoalbuminemia is caused by mutations in a new HIT superfamily gene. Nat Genet. 2001;29:184–188. doi: 10.1038/ng1001-184. [DOI] [PubMed] [Google Scholar]
  • 170.Moreira MC, Barbot C, Tachi N, Kozuka N, Uchida E, Gibson T, Mendonca P, Costa M, Barros J, Yanagisawa T, Watanabe M, Ikeda Y, Aoki M, Nagata T, Coutinho P, Sequeiros J, Koenig M. The gene mutated in ataxia-ocular apraxia 1 encodes the new HIT/Zn-finger protein aprataxin. Nat Genet. 2001;29:189–193. doi: 10.1038/ng1001-189. [DOI] [PubMed] [Google Scholar]
  • 171.Seidle HF, Bieganowski P, Brenner C. Disease-associated mutations inactivate AMP-lysine hydrolase activity of Aprataxin. J Biol Chem. 2005;280:20927–20931. doi: 10.1074/jbc.M502889200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Tumbale P, Appel CD, Kraehenbuehl R, Robertson PD, Williams JS, Krahn J, Ahel I, Williams RS. Structure of an aprataxin-DNA complex with insights into AOA1 neurodegenerative disease. Nat Struct Mol Biol. 2011;18:1189–1195. doi: 10.1038/nsmb.2146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Sykora P, Croteau DL, Bohr VA, Wilson DM., 3rd Aprataxin localizes to mitochondria and preserves mitochondrial function. Proc Natl Acad Sci USA. 2011;108:7437–7442. doi: 10.1073/pnas.1100084108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Scovassi AI. Mitochondrial poly(ADP-ribosylation): from old data to new perspectives. FASEB J. 2004;18:1487–1488. doi: 10.1096/fj.04-1841rev. [DOI] [PubMed] [Google Scholar]
  • 175.Pankotai E, Lacza Z, Muranyi M, Szabo C. Intra-mitochondrial poly(ADP-ribosyDation: potential role for alpha-ketoglutarate dehydrogenase. Mitochondrion. 2009;9:159–164. doi: 10.1016/j.mito.2009.01.013. [DOI] [PubMed] [Google Scholar]
  • 176.Lapucci A, Pittelli M, Rapizzi E, Felici R, Moroni F, Chiarugi A. Poly(ADP-ribose) polymerase-1 is a nuclear epigenetic regulator of mitochondrial DNA repair and transcription. Mol Pharmacol. 2011;79:932–940. doi: 10.1124/mol.110.070110. [DOI] [PubMed] [Google Scholar]
  • 177.Zhang CX, Chang PV, Lippard SJ. Identification of nuclear proteins that interact with platinum-modified DNA by photoaffinity labeling. J Am Chem Soc. 2004;126:6536–6537. doi: 10.1021/ja049533o. [DOI] [PubMed] [Google Scholar]
  • 178.Zhu G, Chang P, Lippard SJ. Recognition of platinum-DNA damage by poly(ADP-ribose) polymerase-1. Biochemistry. 2010;49:6177–6183. doi: 10.1021/bi100775t. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Huang JC, Zamble DB, Reardon JT, Lippard SJ, Sancar A. HMG-domain proteins specifically inhibit the repair of the major DNA adduct of the anticancer drug cisplatin by human excision nuclease. Proc Natl Acad Sci U S A. 1994;91:10394–10398. doi: 10.1073/pnas.91.22.10394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Marietta C, Gulam H, Brooks PJ. A single 8,5′-cyclo-2′-deoxyadenosine lesion in a TATA box prevents binding of the TATA binding protein and strongly reduces transcription in vivo. DNA Repair (Amst) 2002;1:967–975. doi: 10.1016/s1568-7864(02)00148-9. [DOI] [PubMed] [Google Scholar]
  • 181.Allen JA, Coombs MM. Covalent binding of polycyclic aromatic compounds to mitochondrial and nuclear DNA. Nature. 1980;287:244–245. doi: 10.1038/287244a0. [DOI] [PubMed] [Google Scholar]
  • 182.Jung D, Cho Y, Meyer JN, Di Giulio RT. The long amplicon quantitative PCR for DNA damage assay as a sensitive method of assessing DNA damage in the environmental model, Atlantic killifish (Fundulus heteroclitus) Comp Biochem Physiol C Toxicol Pharmacol. 2009;149:182–186. doi: 10.1016/j.cbpc.2008.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Aamann MD, Sorensen MM, Hvitby C, Berquist BR, Muftuoglu M, Tian J, de Souza-Pinto NC, Scheibye-Knudsen M, Wilson DM, 3rd, Stevnsner T, Bohr VA. Cockayne syndrome group B protein promotes mitochondrial DNA stability by supporting the DNA repair association with the mitochondrial membrane. FASEB J. 2010;24:2334–2346. doi: 10.1096/fj.09-147991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Kamenisch Y, Fousteri M, Knoch J, von Thaler AK, Fehrenbacher B, Kato H, Becker T, Dolle ME, Kuiper R, Majora M, Schaller M, van der Horst GT, van Steeg H, Rocken M, Rapaport D, Krutmann J, Mullenders LH, Berneburg M. Proteins of nucleotide and base excision repair pathways interact in mitochondria to protect from loss of subcutaneous fat, a hallmark of aging. J Exp Med. 2010;207:379–390. doi: 10.1084/jem.20091834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Lagerwerf S, Vrouwe MG, Overmeer RM, Fousteri MI, Mullenders LH. DNA damage response and transcription. DNA Repair (Amst) 2011;10:743–750. doi: 10.1016/j.dnarep.2011.04.024. [DOI] [PubMed] [Google Scholar]
  • 186.Mellon I, Spivak G, Hanawalt PC. Selective removal of transcription-blocking DNA damage from the transcribed strand of the mammalian DHFR gene. Cell. 1987;51:241–249. doi: 10.1016/0092-8674(87)90151-6. [DOI] [PubMed] [Google Scholar]
  • 187.Osterod M, Larsen E, Le Page F, Hengstler JG, Van Der Horst GT, Boiteux S, Klungland A, Epe B. A global DNA repair mechanism involving the Cockayne syndrome B (CSB) gene product can prevent the in vivo accumulation of endogenous oxidative DNA base damage. Oncogene. 2002;21:8232–8239. doi: 10.1038/sj.onc.1206027. [DOI] [PubMed] [Google Scholar]
  • 188.Trapp C, Reite K, Klungland A, Epe B. Deficiency of the Cockayne syndrome B (CSB) gene aggravates the genomic instability caused by endogenous oxidative DNA base damage in mice. Oncogene. 2007;26:4044–4048. doi: 10.1038/sj.onc.1210167. [DOI] [PubMed] [Google Scholar]
  • 189.Jaruga P, Xiao Y, Vartanian V, Lloyd RS, Dizdaroglu M. Evidence for the involvement of DNA repair enzyme NEIL1 in nucleotide excision repair of (5′R)- and (5′S)-8,5′-cyclo-2′-deoxyadenosines. Biochemistry. 2010;49:1053–1055. doi: 10.1021/bi902161f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Einolf HJ, Guengerich FP. Fidelity of nucleotide insertion at 8-oxo-7,8-dihydroguanine by mammalian DNA polymerase delta. Steady-state and pre-steady-state kinetic analysis. J Biol Chem. 2001;276:3764–3771. doi: 10.1074/jbc.M006696200. [DOI] [PubMed] [Google Scholar]
  • 191.McCulloch SD, Kokoska RJ, Garg P, Burgers PM, Kunkel TA. The efficiency and fidelity of 8-oxo-guanine bypass by DNA polymerases delta and eta. Nucleic Acids Res. 2009;37:2830–2840. doi: 10.1093/nar/gkp103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Waters LS, Minesinger BK, Wiltrout ME, D’Souza S, Woodruff RV, Walker GC. Eukaryotic translesion polymerases and their roles and regulation in DNA damage tolerance. Microbiol Mol Biol Rev. 2009;73:134–154. doi: 10.1128/MMBR.00034-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Tornaletti S. DNA repair in mammalian cells: Transcription-coupled DNA repair: directing your effort where it’s most needed. Cell Mol Life Sci. 2009;66:1010–1020. doi: 10.1007/s00018-009-8738-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Garcia-Diaz M, Murray MS, Kunkel TA, Chou KM. Interaction between DNA Polymerase lambda and anticancer nucleoside analogs. J Biol Chem. 2010;285:16874–16879. doi: 10.1074/jbc.M109.094391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Canugovi C, Maynard S, Bayne AC, Sykora P, Tian J, de Souza-Pinto NC, Croteau DL, Bohr VA. The mitochondrial transcription factor A functions in mitochondrial base excision repair. DNA Repair (Amst) 2010;9:1080–1089. doi: 10.1016/j.dnarep.2010.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Ulrich HD. Timing and spacing of ubiquitin-dependent DNA damage bypass. FEBS Lett. 2011;585:2861–2867. doi: 10.1016/j.febslet.2011.05.028. [DOI] [PubMed] [Google Scholar]
  • 197.Longley MJ, Ropp PA, Lim SE, Copeland WC. Characterization of the native and recombinant catalytic subunit of human DNA polymerase gamma: identification of residues critical for exonuclease activity and dideoxynucleotide sensitivity. Biochemistry. 1998;37:10529–10539. doi: 10.1021/bi980772w. [DOI] [PubMed] [Google Scholar]
  • 198.Johnson AA, Tsai Y, Graves SW, Johnson KA. Human mitochondrial DNA polymerase holoenzyme: reconstitution and characterization. Biochemistry. 2000;39:1702–1708. doi: 10.1021/bi992104w. [DOI] [PubMed] [Google Scholar]
  • 199.Johnson AA, Johnson KA. Exonuclease proofreading by human mitochondrial DNA polymerase. J Biol Chem. 2001;276:38097–38107. doi: 10.1074/jbc.M106046200. [DOI] [PubMed] [Google Scholar]
  • 200.Lee HR, Johnson KA. Fidelity of the human mitochondrial DNA polymerase. J Biol Chem. 2006;281:36236–36240. doi: 10.1074/jbc.M607964200. [DOI] [PubMed] [Google Scholar]
  • 201.Longley MJ, Nguyen D, Kunkel TA, Copeland WC. The fidelity of human DNA polymerase gamma with and without exonucleolytic proofreading and the p55 accessory subunit. J Biol Chem. 2001;276:38555–38562. doi: 10.1074/jbc.M105230200. [DOI] [PubMed] [Google Scholar]
  • 202.Hanes JW, Thai DM, Johnson KA. Incorporation and replication of 8-oxo-deoxyguanosine by the human mitochondrial DNA polymerase. J Biol Chem. 2006;281:36241–36248. doi: 10.1074/jbc.M607965200. [DOI] [PubMed] [Google Scholar]
  • 203.Pinz KG, Shibutani S, Bogenhagen DF. Action of mitochondrial DNA polymerase gamma at sites of base loss or oxidative damage. J Biol Chem. 1995;270:9202–9206. doi: 10.1074/jbc.270.16.9202. [DOI] [PubMed] [Google Scholar]
  • 204.Shimura-Miura H, Hattori N, Kang D, Miyako K, Nakabeppu Y, Mizuno Y. Increased 8-oxo-dGTPase in the mitochondria of substantia nigral neurons in Parkinson’s disease. Ann Neurol. 1999;46:920–924. [PubMed] [Google Scholar]
  • 205.Yamaguchi H, Kajitani K, Dan Y, Furuichi M, Ohno M, Sakumi K, Kang D, Nakabeppu Y. MTH1, an oxidized purine nucleoside triphosphatase, protects the dopamine neurons from oxidative damage in nucleic acids caused by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Cell Death Differ. 2006;13:551–563. doi: 10.1038/sj.cdd.4401788. [DOI] [PubMed] [Google Scholar]
  • 206.Graziewicz MA, Bienstock RJ, Copeland WC. The DNA polymerase gamma Y955C disease variant associated with PEO and parkinsonism mediates the incorporation and translesion synthesis opposite 7,8-dihydro-8-oxo-2′-deoxyguanosine. Hum Mol Genet. 2007;16:2729–2739. doi: 10.1093/hmg/ddm227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Krishnan KJ, Greaves LC, Reeve AK, Turnbull D. The ageing mitochondrial genome. Nucleic Acids Res. 2007;35:7399–7405. doi: 10.1093/nar/gkm635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.de Souza-Pinto NC, Wilson DM, 3rd, Stevnsner TV, Bohr VA. Mitochondrial DNA, base excision repair and neurodegeneration. DNA Repair (Amst) 2008;7:1098–1109. doi: 10.1016/j.dnarep.2008.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Hixon S, Moustacchi E. The fate of yeast mitochondrial DNA after ultraviolet irradiation. I. Degradation during post-UV dark liquid holding in non-nutrient medium. Biochem Biophys Res Commun. 1978;81:288–296. doi: 10.1016/0006-291x(78)91531-0. [DOI] [PubMed] [Google Scholar]
  • 210.Ray AJ, Turner R, Nikaido O, Rees JL, Birch-Machin MA. The spectrum of mitochondrial DNA deletions is a ubiquitous marker of ultraviolet radiation exposure in human skin. J Invest Dermatol. 2000;115:674–679. doi: 10.1046/j.1523-1747.2000.00092.x. [DOI] [PubMed] [Google Scholar]
  • 211.Birket MJ, Birch-Machin MA. Ultraviolet radiation exposure accelerates the accumulation of the aging-dependent T414G mitochondrial DNA mutation in human skin. Aging Cell. 2007;6:557–564. doi: 10.1111/j.1474-9726.2007.00310.x. [DOI] [PubMed] [Google Scholar]
  • 212.Birch-Machin MA, Swalwell H. How mitochondria record the effects of UV exposure and oxidative stress using human skin as a model tissue. Mutagenesis. 2010;25:101–107. doi: 10.1093/mutage/gep061. [DOI] [PubMed] [Google Scholar]
  • 213.Reimann V, Kramer U, Sugiri D, Schroeder P, Hoffmann B, Medve-Koenigs K, Jockel KH, Ranft U, Krutmann J. Sunbed use induces the photoaging-associated mitochondrial common deletion. J Invest Dermatol. 2008;128:1294–1297. doi: 10.1038/sj.jid.5701151. [DOI] [PubMed] [Google Scholar]
  • 214.Conney AH. Induction of microsomal enzymes by foreign chemicals and carcinogenesis by polycyclic aromatic hydrocarbons: G. H. A. Clowes Memorial Lecture. Cancer Res. 1982;42:4875–4917. [PubMed] [Google Scholar]
  • 215.Wood AW, Chang RL, Levin W, Yagi H, Tada M, Vyas KP, Jerina DM, Conney AH. Mutagenicity of the optical isomers of the diastereomeric bay-region chrysene 1,3-diol-3,4-epoxides in bacterial and mammalian cells. Cancer Res. 1982;42:2972–2976. [PubMed] [Google Scholar]
  • 216.Jerina DM, Chadha A, Cheh AM, Schurdak ME, Wood AW, Sayer JM. Covalent bonding of bay-region diol epoxides to nucleic acids. Adv Exp Med Biol. 1991;283:533–553. doi: 10.1007/978-1-4684-5877-0_70. [DOI] [PubMed] [Google Scholar]
  • 217.Graziewicz MA, Longley MJ, Bienstock RJ, Zeviani M, Copeland WC. Structure-function defects of human mitochondrial DNA polymerase in autosomal dominant progressive external ophthalmoplegia. Nat Struct Mol Biol. 2004;11:770–776. doi: 10.1038/nsmb805. [DOI] [PubMed] [Google Scholar]
  • 218.Saitoh A, Fenton T, Alvero C, Fletcher CV, Spector SA. Impact of nucleoside reverse transcriptase inhibitors on mitochondria in human immunodeficiency virus type 1-infected children receiving highly active antiretroviral therapy. Antimicrob Agents Chemother. 2007;51:4236–4242. doi: 10.1128/AAC.00893-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Saitoh A, Haas RH, Naviaux RK, Salva NG, Wong JK, Spector SA. Impact of nucleoside reverse transcriptase inhibitors on mitochondrial DNA and RNA in human skeletal muscle cells. Antimicrob Agents Chemother. 2008;52:2825–2830. doi: 10.1128/AAC.00434-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Mellon I. Transcription-coupled repair: a complex affair. Mutat Res. 2005;577:155–161. doi: 10.1016/j.mrfmmm.2005.03.016. [DOI] [PubMed] [Google Scholar]
  • 221.Ganesan AK, Hanawalt PC. Transcription-coupled nucleotide excision repair of a gene transcribed by bacteriophage T7 RNA polymerase in Escherichia coli. DNA Repair (Amst) 2010;9:958–963. doi: 10.1016/j.dnarep.2010.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol. 2011;12:9–14. doi: 10.1038/nrm3028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Ljungman M, Zhang F. Blockage of RNA polymerase as a possible trigger for u.v. light-induced apoptosis. Oncogene. 1996;13:823–831. [PubMed] [Google Scholar]
  • 224.Ljungman M, O’Hagan HM, Paulsen MT. Induction of ser15 and lys382 modifications of p53 by blockage of transcription elongation. Oncogene. 2001;20:5964–5971. doi: 10.1038/sj.onc.1204734. [DOI] [PubMed] [Google Scholar]
  • 225.Sansome C, Zaika A, Marchenko ND, Moll UM. Hypoxia death stimulus induces translocation of p53 protein to mitochondria. Detection by immunofluorescence on whole cells. FEBS Lett. 2001;488:110–115. doi: 10.1016/s0014-5793(00)02368-1. [DOI] [PubMed] [Google Scholar]
  • 226.Lebedeva MA, Eaton JS, Shadel GS. Loss of p53 causes mitochondrial DNA depletion and altered mitochondrial reactive oxygen species homeostasis. Biochim Biophys Acta. 2009;1787:328–334. doi: 10.1016/j.bbabio.2009.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Seidel-Rogol BL, Shadel GS. Modulation of mitochondrial transcription in response to mtDNA depletion and repletion in HeLa cells. Nucleic Acids Res. 2002;30:1929–1934. doi: 10.1093/nar/30.9.1929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Saxowsky TT, Doetsch PW. RNA polymerase encounters with DNA damage: transcription-coupled repair or transcriptional mutagenesis? Chem Rev. 2006;106:474–488. doi: 10.1021/cr040466q. [DOI] [PubMed] [Google Scholar]
  • 229.Marietta C, Brooks PJ. Transcriptional bypass of bulky DNA lesions causes new mutant RNA transcripts in human cells. EMBO Rep. 2007;8:388–393. doi: 10.1038/sj.embor.7400932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Saxowsky TT, Meadows KL, Klungland A, Doetsch PW. 8-Oxoguanine-mediated transcriptional mutagenesis causes Ras activation in mammalian cells. Proc Natl Acad Sci USA. 2008;105:18877–18882. doi: 10.1073/pnas.0806464105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Wang Z, Cotney J, Shadel GS. Human mitochondrial ribosomal protein MRPL12 interacts directly with mitochondrial RNA polymerase to modulate mitochondrial gene expression. J Biol Chem. 2007;282:12610–12618. doi: 10.1074/jbc.M700461200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Lodeiro MF, Uchida AU, Arnold JJ, Reynolds SL, Moustafa IM, Cameron CE. Identification of multiple rate-limiting steps during the human mitochondrial transcription cycle in vitro. J Biol Chem. 2010;285:16387–16402. doi: 10.1074/jbc.M109.092676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Shutt TE, Lodeiro MF, Cotney J, Cameron CE, Shadel GS. Core human mitochondrial transcription apparatus is a regulated two-component system in vitro. Proc Natl Acad Sci USA. 2010;107:12133–12138. doi: 10.1073/pnas.0910581107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Cline SD, Lodeiro MF, Marnett LJ, Cameron CE, Arnold JJ. Arrest of human mitochondrial RNA polymerase transcription by the biological aldehyde adduct of DNA, M1dG. Nucleic Acids Res. 2010;38:7546–7557. doi: 10.1093/nar/gkq656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Smidansky ED, Arnold JJ, Reynolds SL, Cameron CE. Human mitochondrial RNA polymerase: evaluation of the single-nucleotide-addition cycle on synthetic RNA/DNA scaffolds. Biochemistry. 2011;50:5016–5032. doi: 10.1021/bi200350d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Cline SD, Riggins JN, Tornaletti S, Marnett LJ, Hanawalt PC. Malondialdehyde adducts in DNA arrest transcription by T7 RNA polymerase and mammalian RNA polymerase II. Proc Natl Acad Sci USA. 2004;101:7275–7280. doi: 10.1073/pnas.0402252101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Ringel R, Sologub M, Morozov YI, Litonin D, Cramer P, Temiakov D. Structure of human mitochondrial RNA polymerase. Nature. 2011;478:269–273. doi: 10.1038/nature10435. [DOI] [PubMed] [Google Scholar]
  • 238.Bogenhagen DF, Rousseau D, Burke S. The layered structure of human mitochondrial DNA nucleoids. J Biol Chem. 2008;283:3665–3675. doi: 10.1074/jbc.M708444200. [DOI] [PubMed] [Google Scholar]
  • 239.de Souza-Pinto NC, Harris CC, Bohr VA. p53 functions in the incorporation step in DNA base excision repair in mouse liver mitochondria. Oncogene. 2004;23:6559–6568. doi: 10.1038/sj.onc.1207874. [DOI] [PubMed] [Google Scholar]

RESOURCES