Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Mar 24.
Published in final edited form as: Sci Total Environ. 2022 Oct 1;856(Pt 2):159192. doi: 10.1016/j.scitotenv.2022.159192

Nucleic Acid Adductomics – the Next Generation of Adductomics towards Assessing Environmental Health Risks

Marcus S Cooke a,1,*, Yuan-Jhe Chang b,1, Yet-Ran Chen c, Chiung-Wen Hu d,*, Mu-Rong Chao b,e,*
PMCID: PMC11932045  NIHMSID: NIHMS2061899  PMID: 36195140

Abstract

This Discussion article aims to explore the potential for a new generation of assay to emerge from cellular and urinary DNA adductomics which brings together DNA- RNA- and, to some extent, protein adductomics, to better understand the role of the exposome in environmental health. Components of the exposome have been linked to an increased risk of various, major diseases, and to identify the precise nature, and size, of risk, in this complex mixture of exposures, powerful tools are needed. Modification of nucleic acids (NA) is a key consequence of environmental exposures, and a goal of cellular DNA adductomics is to evaluate the totality of DNA modifications in the genome, on the basis that this will be most informative. Consequently, an approach which encompasses modifications of all nucleic acids (NA) would be potentially yet more informative. This article focuses on NA adductomics, which brings together the assessment of both DNA and RNA modifications, including modified (2’-deoxy)ribonucleosides (2’-dN/rN), modified nucleobases (nB), plus: DNA-DNA, RNA-RNA, DNA-RNA, DNA-protein, and RNA-protein crosslinks (DDCL, RRCL, DRCL, DPCL, and RPCL, respectively). We discuss the need for NA adductomics, plus the pros and cons of cellular vs. urinary NA adductomics, and present some evidence for the feasibility of this approach. We propose that NA adductomics provides a more comprehensive approach to the study of nucleic acid modifications, which will facilitate a range of advances, including the identification of novel, unexpected modifications e.g., RNA-RNA, and DNA-RNA crosslinks; key modifications associated with mutagenesis; agent-specific mechanisms; and adductome signatures of key environmental agents, leading to the dissection of the exposome, and its role in human health/disease, across the life course.

Keywords: adductomics, nucleic acids, repair, exposome, damage, mass spectrometry

Graphical Abstract

graphic file with name nihms-2061899-f0007.jpg

1. Introduction to the Exposome and Biomarkers

Components of the Environment contribute 80-90% of the risk of developing cancer, and degenerative diseases 1, 2. Specifically, this risk of disease is driven by components of the exposome i.e., all the internal and external, environmental factors to which humans are exposed, across the life course, including diet, air pollution, social interactions, living environment, and lifestyle choices 3 (summarized in Figure 1). Exposome-related health issues therefore represent a significant challenge to public health 4. Interactions between the exposome and the genome impact individual health, and contribute to the risk of disease 5. However, the exposome represents a complex mix of exposures, processes, and factors, and therefore to identify the precise nature, and size, of risk powerful tools are needed to link exposure, cellular consequences, and health/disease 6. Considerable effort is focused currently on methodology for estimating the specific and general external exposomes (the concepts of which are illustrated Figure 1) i.e., the origins of exposure lie exclusively outside the body. From a health perspective, the focus is on the agents which reach the body’s cells, and may then lead to disease 7. In this regard, it is the internal exposome which is of primary interest as it represents (i) the total target (which could be a particular cell type, organ, or whole-body burden), and (ii) dose of physico-chemical agents in the body (irrespective of their source i.e., being external or internal to the body, or how they are derived e.g., processes such as inflammation, metabolism, and microbiome8 (Figure 1) 6, 7. (We have added ‘physico-’ to the definition to include agents such as radiation.) A biomarker can be described as “a measurable substance in an organism whose presence is indicative of some phenomenon such as disease, infection, or environmental exposure” 9. Therefore, biomarkers of the internal exposome e.g., modifications of cellular biomolecules may be particularly biologically informative, as they may represent the internal dose, effect(s) on the biological system, and possess a role in disease 10 (see Figure 1). On this basis, a major goal of toxicology is to use biomarkers of the internal exposome to (i) identify and (ii) act as proxies for key health-modifying elements of the exposome without the need for detailed measurements of the external environment 6.

Figure 1. Role of the exposome in the generation of cellular molecular modifications.

Figure 1.

The exposome (encompassing both external and internal exposomes) represents a wide, and diverse range of exposures, that may occur across the life course. Species arising from the exposome have the potential to damage, or otherwise modify, DNA, RNA, proteins. These modified molecules may result in adverse outcomes for the cell, and lead to disease in the organism. Example exposures from the exposome are illustrated.

Like other ‘omics techniques (e.g., genomics, metabolomics, proteomics), a major strength of DNA adductomics is the untargeted assessment of the totality of a target, in this case the DNA adductome i.e., all of the covalently modifications of DNA nucleobases (DNA adducts) derived from environmental and dietary genotoxicants and endogenously produced electrophiles 11. In this Discussion, we have summarized some of the recent advances in DNA adductomics, and then indicated how DNA adductomics may advance to become yet more comprehensive through the detection of the totality of nucleic acid (NA)-derived adducts, and how NA adductomics may thereby better inform on both exposures and their biological consequences.

2. Consequences and causes of nucleic acid modifications

There is continual exposure to agents, and processes which damage 12, or otherwise modify NA [which includes DNA, RNA, and the 2’-deoxyribonucleotide and ribonucleotide i.e., (d)NTP pools] 13, and their associated proteins (represented by “molecular modifications” in Figure 1). The definition of a genotoxin is a chemical or agent that can cause DNA or chromosomal damage 14. Currently there is not an equivalent term for RNA damage, although analogy clearly exists 15. We propose that DNA and RNA can, and should, be considered together when adopting a holistic view of the effect of genotoxin exposure, particularly as damage to RNA, like DNA, is increasingly being linked to disease (reviewed in 1619). As discussed elsewhere, whether a particular alteration of NA is referred to as damage or a modification depends upon the modification, and context 20. Modifications induced intentionally by normal cellular process, such as epigenetic programming (e.g., DNA methylation), may be viewed as modifications, whereas those formed unintentionally are damage 20, although a grey area may exist in which context is key to interpretation 21.

To date most focus has been upon damage to DNA (and 2’-dNTP pool) as this impacts cell function 22, and plays a role in the pathogenesis of, arguably, all major human diseases, in particular cancer, but also neurodegeneration, and cardiovascular disease, plus aging 23. However, there is increasing evidence that damage to, or modification of, RNA (and the ribonucleotide pool) also affects cell function, and plays a role in pathogenesis 17, 18, 24. Although DNA and RNA have distinctly different functions within a cell, and hence damage to them has different implications, they both represent targets for agents which can damage nucleic acids 25, 26. Damage to DNA can interfere with fundamental cellular processes, such as DNA replication, and transcription, and can thereby lead to a variety of outcomes, for example mutation, microsatellite instability, alterations in transcription factor binding, loss of heterozygosity 22, and cytotoxicity 27. Damage to RNA (both coding and non-coding) accumulates at much higher levels than DNA 28, 29, alters the chemical properties and hence the function of RNA leading to downstream consequences, such as defects in protein synthesis 3032. The consequences of damage to non-coding RNAs (which represent ~95% of the cellular RNA) is largely unknown, but the presence of mechanisms to detect modified rRNA and tRNA suggest their presence could have detrimental outcomes for the cell (reviewed in 33). As they are the origin of the nucleobases in DNA and RNA, damage to the 2’-dNTP and NTP pools is a potentially significant source of damaged nucleobases in these biomolecules 3437, resulting in similar cellular consequences as listed above. The presence of mechanisms to sanitize damage from the 2’-dNTP and NTP pools (reviewed in 38) supports the significance of these as sources of damage to NA.

Processes which alter NA can have exogenous and/or endogenous origins. An example of an endogenous origin is the “leakage” of electrons from normal cellular metabolism, which may lead to the generation of reactive oxygen species, and can damage NA resulting in background levels of oxidatively modified NA 39. Another example is DNA methylation, where specialised enzymes add or remove methyl groups from DNA nucleobases to control gene expression, chromosomal structure, and stability 40. In contrast, environmental stressors have exogenous origins, and while the damage that they cause to NA can be specific, and hence characteristic, to a particular stressor e.g., cyclobutane pyrimidine dimers (from UV radiation 41), 8,9-dihydro-8-(N7-guanyl)-9-hydroxyaflatoxin B1 (from dietary aflatoxin B1 42), 7-(2’-deoxyadenosin-N6-yl)aristolactam I (from Aristolochic acid 43), they may also impact endogenous processes, and influence levels of endogenously-derived NA modifications. For example, exposure to UVR or ionising radiation, and metabolism of certain xenobiotics may all lead to the generation of free electrons, impact redox homeodynamics, and increase levels of oxidatively modified NA above baseline levels e.g., 8-oxo-7,8-dihydro-2’-deoxyguanosine and 8-oxo-7,8-dihydroguanosine, from DNA and RNA, respectively 44. Environmental factors such as UVR and ozone can transform chemicals, which will also lead to the production of reactive species with the potential to modify DNA 45. Similarly, some exogenous stressors e.g., benzo(a)pyrene 46, polychlorinated biphenyls, methylmercury, and organochlorine pesticides 47, can influence methylation, and other epigenetic processes. Consequently, there is some overlap in the types of damage arising from both endogenous and exogenous sources. While not every stressor may cause a characteristic single form of damage, like those noted above, we propose that it might be possible to determine unique ‘signatures’ of a particular stressor if multiple forms of damage are considered, or this might identify the precise mechanisms by which a stressor may act 46. In both cases, the greater the types of damage considered, the more precise the signature or mechanism.

Of the processes which induce DNA damage, oxidation is a prime example of the potential to induce a multiplicity of different forms of damage. Recently defined as a hallmark of environmental insult 48, oxidative eustress/distress 49 can lead to the generation over 24 DNA nucleobase (nB) products, and the total number of adducts exceeds 100, when 2-deoxyribose (2-dR), and phosphate backbone modifications are considered 23, and this does not include DNA-DNA and DNA-protein crosslinks, or the adducts derived from secondary processes, such as lipid peroxidation 50. These numbers are increased further, when RNA-derived modifications, and epigenetic modifications of DNA and RNA are also included, bringing the total number of potential oxidatively-generated modifications/adducts into the high hundreds, if not greater.

Most of the literature describes the targeted analysis of DNA damage, rather than RNA, and the measurement of a single, or a few adducts 51. While not precluding the use of targeted analysis, this approach cannot assess the spectrum of NA adducts present in the cell, which gives rise to several weaknesses: e.g., potential biomarkers of exposure may be excluded, it is impossible to identify modifications which act in concert to increase disease risk, and it precludes the identification of yet-to-be-discovered modifications with a key role in disease. Indeed, there is growing evidence that RNA modifications are involved in a growing number of human diseases 24, 38, 52 (see also the MODOMICS website for a comprehensive review, https://iimcb.genesilico.pl/modomics/), and RNA modifications have a broad range of functions, including RNA processing, splicing, polyadenylation, editing, structure, stability, localization, translation initiation, and gene expression regulation 53. To date, approximately 151 modified RNA nucleobase residues have been identified, increasing to ~334 as nucleoside and nucleotide forms are included 5459 however their precise effects upon cellular function remain to be discovered 13. Other types of NA-associated adducts also have significant biological consequences e.g., DNA-DNA, and DNA-protein crosslinks (DDCL, DPCL) interfere with DNA replication and transcription 60, 61, RNA-protein crosslinks (RPCL) can modulate, or stabilize, RNA, interfering with function 62 [DNA-RNA crosslinks (DRCL), and RNA-RNA crosslinks (RRCL) may also occur, but given that their existence has not yet been reported, their function is unknown]. The breadth of NA modifications is therefore significant and, just as DNA adductomics aims to be most informative by reporting on the totality of DNA adducts 63, we propose that a non-targeted NA adductomics will be yet more informative by aiming to comprehensively inform on the totality of NA modifications, simultaneously.

3. Cellular DNA adductomics - towards cellular NA adductomics.

Kanaly et al. 64 first proposed a cellular DNA adductomic strategy to simultaneously measure multiple DNA adducts using liquid chromatography-triple quadrupole tandem mass spectrometry (LC-QqQ-MS/MS) with electrospray ionization (ESI), operating in the selected reaction monitoring (SRM) mode 65. Due to the labile nature of the 2-dN glycosidic bond, this method detects a number of contiguous SRM of [M+H]+ to [M+H-116]+ transitions, in which 116 Da corresponds to the neutral loss of the 2-dR moiety. Lately, high resolution MS (HRMS)66, 67 has also been adopted for DNA adductomics analysis, for example, the hybrid quadrupole-linear ion trap Orbitrap MS (Q-LIT-OT-MS) 68. The mass of the ions detected by OT can be measured with ppm accuracy (≤ 1-5 ppm mass error). This high mass accuracy significantly improves/enhances confidence in DNA adduct identification, compared to the nominal mass accuracy obtained by QqQ-MS/MS. The advent of DNA adductomics by HRMS has led to the proposal of a “top-down” approach, by which patterns of adducts may be used to trace, and identify the originating exposure source 69, 70 (summarised in Figure 2).

Figure 2. The concept of a top-down approach for HRMS-based DNA adductomics for linking human, environmental carcinogen exposure, and cancer risk.

Figure 2.

Cellular DNA adductome maps are developed, using HRMS, from exposed and unexposed individuals. DNA adductome maps are compared, and potential candidate adducts, reflective of exposure, are short-listed. Expected (known) adducts are identified from characteristics held within an DNA adductome library 140, and unexpected (unknown) adducts are identified by in silico prediction. Precursor reactive species/metabolites are identified from the profile of adducts formed, and the responsible environmental agents, to which the individuals were exposure, are then proposed.

Previously, we reported a cellular DNA adductomics approach 71, then a HRMS approach 72, and broadened the range of adducts to include DDCL (i.e., crosslinkomics) 73. Recently, RNA adductomics was reported, which uses neutral loss targeting of the [M+H]+ > [M+H-132]+ transitions, in which 132 Da corresponds to the neutral loss of the 2-ribose (R) 25. This reveals the potential for genotoxins to cause analogous forms of damage to both DNA, and RNA 25, further emphasising the importance of studying NA, more broadly, as a target for genotoxins. However, to date there are no adductomics methods for the combined detection of DNA, and RNA modifications, let alone in conjunction with DDCL, RRCL, DRCL, DPCL, and RPCL.

4. Cellular nucleic acid adductomics

We propose that NA adductomics will offer the most comprehensive approach for the study of nucleic acid modifications, to date, by combining the assessment of DNA, and RNA modifications, plus DDCL, RRCL, DRCL, DPCL, and RPCL. To illustrate the feasibility of NA adductomics we present supportive data. For example, we illustrated that different NA modifications produce their own specific fragmentation patterns in collision-induced dissociation (CID; Figure 3) by HRMS (i.e., Q-LIT-OT-MS). The experimental details and method applied are given in the Supplementary information. For example: modified 2’-dN tend to lose dR, resulting in the product ion of [M+H-dR]+; modified ribonucleosides (rN) tend to lose ribose (R), resulting in the product ion of [M+H-R]+ or [M+H-MeR]+, respectively. The origin of [M+H-MeR]+ can be ascribed to ribose 2’-O-methylation, which is one of the most common and widespread types of RNA modification, commonly found in rRNA, tRNA, small nucleolar RNA (snRNA) and, as reported recently, in mRNA 74. Modified nB produce specific product ions including [B+H]+ and [B+H-NH3]+; DDCL generate product ions of [M+H-2dR]+, and [M+H-dR]+; RRCL have product ions of [M+H-2R]+, and [M+H-R]+; DRCL have product ions of [M+H-dR]+, [M+H-R]+, and [M+H-dR-R]+; DPCL give [M+H-dR]+, [M+H-Cys ]+ and [M+H-dR-Cys]+; RPCL generate [M+H-R]+, [M+H-Cys]+, and [M+H-R-Cys]+. The cysteine moieties arise from the proteins with which the nucleic acids were crosslinked 73. The above exact mass of neutral loss (Da) or product ions (m/z) monitored by HRMS are provided in the Supplementary Table S1 together with the supporting studies reported in the literature. In general, for modified 2’-dN, rN, nB, DDCL and DPCL, the fragmentation patterns observed are consistent with the previous studies (see Table S1). However, to the best of our knowledge, no study has been reported the others (RRCL, DRCL and RPCL) in the literature. It is worth noting that adductomics is not limited solely to adducts formed via damaging reactions i.e., the products of which are unintentional. For example, intentional products including those arising from endogenous processes, such as epigenetic modifications 21, are also detected by NA adductomics. The goal is to evaluate the maximal number of modifications, in a single analytical run, to provide the most comprehensive characterization of the role of NA adducts in environmental exposures, and disease risk. Additionally, this could lead to the generation of stressor-specific, signature patterns of adducts (characteristic NA adductome maps), and help inform on the mode of action of stressors.

Figure. 3. Fragmentation patterns of (A) modified 2’-dN, (B) modified rN, (C) modified nB nB , (D) DDCL, (E) RRCL, (F) DRCL, (G) DPCL, and (H) RPCL, by LC-Q-LIT-OT-MS with CID fragmentation mode.

Figure. 3.

Test solutions were prepared by dissolving various commercially available standards in deionized water, including modified 2’-dN, modified rN, modified nB. For the DDCL, RRCL, DPCL and RPCL, these were synthesized by incubating native 2’-deoxyribonucleosides (i.e., dGuo, dAdo, dCyd and dThd), ribonucleosides (i.e., Guo, Ado, Cyd and Urd), and/or cysteine, with formaldehyde 73. The fragmentation patterns were obtained using a LC-Q-LIT-OT-MS equipped with a heated electrospray ionization source, operated in positive ESI-CID mode. All the informative, and specific product ions for each type of NA modification were generated in MS2. The experimental details are provided in the Supplementary information.

While cellular DNA, and hence NA, adductomics have multiple, potential applications 69, these are likely to be particularly valuable in identifying environmental exposures associated with increased cancer risk in humans. However, to date, there have been few reports of cellular DNA adductomics analysis of human tissue 64, 75, although more are beginning to appear 65, but this has not yet extended to human populations per se. Human tissue can represent a severe analysis challenge due to the need for high sensitivity to detect multiple adducts 76. Recent advances in linear ion trap, and Orbitrap instruments offer improved sensitivity 51, but the availability of such instrumentation is limited, not least due to cost. Furthermore, cellular DNA adductomics requires a significant amount of DNA, typically 50-500 μg, depending upon adduct frequency 67, 77, which is more than that needed for targeted analyses 51. Obtaining tissue is necessarily invasive, can be challenging logistically, adds additional ethics review board scrutiny, may discourage volunteer participation 78, 79, and limits access to vulnerable individuals (e.g., the young, or elderly). Isolating DNA from exfoliated epithelial cells (e.g., from the urogenital tract or buccal cavity) represents a potential way forward, and a means to circumvent some of these issues 80, albeit while restricting the investigation to a particular organ. In contrast, biomonitoring NA damage products in urine may be a viable alternative, with a number of advantages over cellular NA 50 (see below).

5. Biomonitoring NA adducts in urine.

The presence of DNA adducts in urine is generally considered to be the consequence of DNA repair 81, 82 i.e., base excision repair (BER) is the source of nB adducts 83, and global genome-, and transcriptional coupled-nucleotide excision repair (GG-NER, and TCR-NER, respectively), results in 2’-dN adducts 8487 (Figure 4). More specific evidence for this comes from the reported presence of certain 2’-dN adducts in urine, such as (5’R)-, (5’S)-8,5-cyclo-2’deoxyadenosines 8890, and 3-(2-deoxy-β-d-erythro-pentofuranosyl)pyrimido[1,2-α]purin-10(3H)-one 91, 92, all of which appear to be repaired by NER 9294. Additionally, sanitization of the (2’-deoxy)ribonucleotide pools represents another logical source of urinary 2’-dN (and rN) adducts (Figure 4) 95. Spontaneous depurination may also be a contributor to urinary adduct levels 96 e.g., unstable N7-guanine adducts which have been reported to occur dose-dependently in the urine of rodents, and which appear proportionally to levels in the liver (reviewed in 97). Given on-going exposure to environmental agents which may induce DNA damage, and the usual steady-state repair, 98 the urinary NA adductome will be reflective of the body burden of adducts. While it may be argued that these repaired adducts are unlikely to be the ones specifically responsible for inducing mutations, or other adverse cellular events, they can reflect the risk of mutations occurring i.e., the larger the whole organism adductome burden, the greater the risk of mutation occurring within that organism 99. Furthermore, defects in the post- adduct excision steps of DNA repair may also have detrimental consequences for the cell 100.

Figure 4. Representative sources of NA modifications in urine.

Figure 4.

Broadly, base excision repair (BER) of DNA leads to the presence of modified nucleobases in urine, and there is some evidence that urinary, modified 2’-deoxyribonucleosides are derived from the activity of global genome, and/or transcription-coupled, nucleotide excision repair (NER). The activity of enzymes, such as the Nudix hydrolases (e.g., NUDT1), which hydrolyse modified (2’-d)NTPs to NMPs, may also contribute to urinary levels of modified (2’-deoxy)ribonucleosides. The figure uses oxidatively modified DNA and RNA to illustrate examples of the processes. The repair products of DNA-DNA crosslinks (DDCL), DNA-protein crosslinks (DPCL), RNA-RNA crosslinks (RRCL), and RNA-protein crosslinks (RPCL) are less well defined.

There is a growing number of reports where targeted approaches have been used to successfully detect specific, environmentally induced DNA adducts in urine. Examples of urinary adducts include: 3-(4-dihydroxyphenyl)adenine, following benzene exposure of mice 101; aflatoxin N7-guanine, and 8,9-dihydro-8-(2,6-diamino-4-oxo-3,4-dihydropyrimid-5-yl-formamido)-9-hydroxy-aflatoxin B1, following exposure of rats to aflatoxin 102; N7-(1-hydroxy-3-buten-2-yl)guanine following exposure of mice to 1,3-butadiene (BD) 103, and occupationally- or recreationally- (tobacco smoke) exposed humans 104; N7-(1-hydroxy-3-buten-2-yl) guanine adducts were also detected in the same human urine samples from the previous citation 103, and bis-N7-guanine DNA-DNA crosslinks in mice exposed to BD 105. The above reports, using targeted approaches, are beginning to successfully link exposure to environmental agents to specific urinary DNA adducts.

The literature describes a growing number of reports describing processes for the removal of RNA-derived adducts, the activity of which is supported by the presence of RNA products in urine (reviewed in 38; Figure 4). The processes for the repair of crosslinks (NA-NA, and NA-protein) are more complex, and less well understood than for mono-adducts. DDCL are repaired by processes that involves NER 106, with the potential involvement of BER 107, 108. It has been suggested that the crosslink is removed in the context of an oligonucleotide, which is subsequently digested down to a smaller, lesion-containing product, prior to excretion into the urine 87. There are reports of multiple mechanisms for cells to cope with DNA-protein crosslinks 109, which may be broadly defined as: direct hydrolysis of the covalent linkage between protein and DNA; excision of the lesion-containing DNA fragment; and proteolysis of the protein component 110. Given these possibilities, the post-repair processing of the DNA-protein adduct itself is less clear than for DDCL (and for RRCL, and RPCL, even less so) 111. Nevertheless, unless the DNA adduct is reversed 112, some form of adduct-derived product will remain, which needs to be removed 61, and excreted (as evidenced from our data, presented here). Taken together, the above findings are highly supportive of the potential of urinary NA adducts to inform on whole body environmental exposures.

Measuring adducts in urine is not without its limitations. For example, while urine may inform on the body burden of adducts, to date it is not always possible to relate adducts in urine to a particular organ (skin and UVR-induced cyclobutane pyrimidine dimers may be a notable exception113, 114, although contributions from peripheral blood mononuclear cells are possible). In contrast, cellular DNA adductomics can report on the adducts that are not repaired, and enables a direct link to be formed with the target tissue. Early reports examining urinary 8-oxoGua, 8-oxodGuo, and thymine glycol suggested possible contributions from the diet to urinary modified nucleobase concentrations, whereas the modified 2’-deoxynucleoside was unaffected 115, 116. However, careful review of these studies revealed limitations in their study design 117, and more robust studies, in both mice and humans, subsequently revealed no contribution from diet to either 8-oxoGua or 8-oxodGuo (or indeed uric acid, a major purine breakdown product) in urine 81, 118, 119, suggesting that this might be true for other adducts. Repair of adducts from the gut microbiome might also be considered a potential contributor to urinary adduct levels, although given that there appears to be no uptake of adducts from the diet, uptake of adducts from the microbiome is also unlikely to be a source; whether or not this is the case has yet to be reported in the literature. Normalization, or correcting for urine concentration can be a challenge, and may prevent comparisons between study groups. However, this has been well-studied for the targeted analysis of adducts such 8-oxodGuo, with 24 h collections being described as the gold standard, and spot urines corrected for creatinine a suitable alternative 120. Additionally, we have proposed the use of a cocktail of stable isotopically labelled standards in DNA adductomics which will aid quantification, and correction for matrix effects 71.

Urine has numerous benefits as a matrix in which to study biomarkers. It is obtained non-invasively, easily collected, transported, and stored, with low biological hazard. Furthermore, no pre-processing is needed prior to storage, small volumes are required for analysis (typically less than 500 μL), and adduct stability is maintained for >15 y at −80 °C (in the case of the representative lesion, 8-oxodGuo, at least) 121, allowing the use of previously banked samples 117, 122. Urine sample workup is generally simpler than for DNA. It is a well-established matrix in which to study biomarkers of exposure 123, including a wide variety of DNA adducts e.g., oxidized nucleic acid products (including RNA) 124, 125, N-nitroso-derived DNA adducts 126, alkylated purines 127, and aristolochic acid-derived DNA adducts 128, illustrating the breadth of adducts in urine, which parallels that in NA. However, as noted above, such targeted analyses severely restrict the amount of information that can be gained. Applying adductomics to the study of urine therefore represents a convenient means to non-invasively assess the totality of NA modifications, and therefore exposures. Like most assays, cellular and urinary DNA adductomics both possess strengths and weaknesses but, as the literature for cellular and urinary targeted assays show, their appropriate application can be highly informative, and there is growing evidence that this is particularly true for adductomics, where a strength is being as comprehensive as possible, and applicable to a wide range of in vivo and in vitro systems 129132.

6. Why aim for the most comprehensive approach to studying nucleic acid damage?

While cellular and urinary DNA adductomics aim to assess the totality of DNA adducts in the genome, these approaches are not entirely comprehensive for genomic damage, as they have excluded RNA modifications (i.e., damage, and intentional modifications, such as epigenetic changes), and other classes of adducts (e.g., crosslinks), all of which have downstream consequences for the cell, and are consequently implicated in pathogenesis 38, 60, 62, 133135. We propose that the NA adductome represents an improved, functional, multi-faceted biomarker of the interaction between exposome, and genome. Furthermore, comprehensively assessing the totality of damage to all NA moieties, is critical to quantifying, and characterizing exposures, along with defining the role of nucleic acid damage in disease. The strengths of this approach include consideration of: (i) the factors which influence adduct formation, arising from the physicochemical nature of the NA moiety, e.g., due to abundance, conformation (double-, single-stranded, or free nucleoside/nucleobase), physical associations (e.g., with proteins, or metal ions), location (nucleus, cytoplasm, proximity to mitochondria), and propensity to modification (e.g., RNA is more easily oxidised than DNA, 28, 33 and probably more easily damaged generally26); (ii) the variety of downstream effects that modified nucleic acids can cause, and their relevance to pathogenesis, e.g., mutations (DNA), epigenetic changes (DNA/RNA), error-containing proteins, and altered protein synthesis (RNA) 38; and (iii) the ability to perform non-invasive assessments via urine.

7. Is NA adductomics feasible?

Cellular DNA adductomics of biological samples is becoming increasingly well-established 11, 25, 130, 136, 137. However, we propose that, in line with the goal of ‘omics techniques to “determine the totality of a target”, the next advance for DNA adductomics is to evaluate the totality of modifications to nucleic acids, in cells, tissues, and urine. We therefore propose the development of cellular and urinary NA adductomics, extending our cellular 71, and urinary 138 DNA adductomics assays to include modified 2’-dN, nB, rN, DDCL, RRCL, DRCL, DPCL, and RPCL. Urine is a convenient matrix in which to assess the NA adductome as it contains the totality of adducts following the repair of cells in the entire body, producing levels significantly higher than in individual tissues, and reflecting the totality of exposures. We therefore propose an NA adductomics approach, using a LC-Q-LIT-OT-MS with CID fragmentation, to achieve the detection of the widest, and most comprehensive, range of nucleic acid modifications. To support the feasibility of our proposed NA adductomics approach, our preliminary, proof-of-principle results revealed that at least six types of NA modifications were present in the urine of healthy subjects (Figure 5), including modified 2’-dN, modified rN, modified nB, DRCL, RRCL and RPCL. DDCL and DPCL were not detected in the urine, from healthy human subjects. Of the NA modifications detected, three out of 61 modified 2’-dN, 16 out of 265 modified rN, six out of 325 modified nB were fully identified, and confirmed using commercially available standards (summarized in Table S2). The detailed LC-Q-LIT-OT-MS analysis is provided in the Supplementary information. Among these fully identified NA modifications, it is worth noting that several are novel NA modifications not previously identified in the human urine, such as N4-methyl-2’-deoxycytidine (N4-mdCyd). At present, we cannot identify whether they derive from exogenous, or endogenous sources (or both). As expected, several well-known NA modifications were also successfully detected such as 8-oxodGuo and 5-methyl-2’-deoxycytidine (5-mdCyd; see Figure 5A), N6-methyladenine (m6A) and N6,2′-O-dimethyladenosine (m6Am; see Figure 5B), together with N7-methylguanine (N7-mGua), N3-methyladenine (N3-mAde) and 8-oxoGua (Figure 5C). In addition to the modified purine nucleobases (Figure 5C), a large number of modified pyrimidine nucleobases were also observed (> 400 adducts; data not shown). Regarding NA-associated crosslinks, for the first time we identified one DRCL (Figure 5D) and six RRCL (Figure 5E), which we propose to be AP apurinic/apyrimidinic (AP) site-derived crosslinks 139 (DRCL: Ion 1 = 2’-deoxyadenosine-AP (dAdo-AP); RRCL: Ion 2 = 2’-O-methylguanosine-AP (Gm-AP); Ion 3 = 2’-O-methyluridine-AP (Um-AP); Ion 4 = deazaguanosine-AP (dzGuo-AP); Ion 5 = adenosine-AP (Ado-AP); Ion 6 = 2’-O-methyladenosine-AP (Am-AP); Ion 7 = adenosine-di-AP (Ado-Di-AP). Moreover, a novel RPCL (Ion 8) was newly identified, which we propose is adenosine-CH2-cysteine (Ado-CH2-Cys). These assumptions were supported by the presence of their protonated precursor ions [M + H]+, as well as their specific fragmentation features. The proposed chemical structures, and product ion spectra of NA-associated crosslinks are provided in Supplementary Figure S1S3, and Table S3. In addition to the above NA modifications, we detected numerous other NA modifications in human urine, but these are yet to be fully identified (e.g., Figures 5A5C). The structure elucidation of these NA modifications requires a searchable mass spectral database, akin to those currently under development for DNA adductomics 140, 141. In total, we detected over 1,000 modified nucleic acid moieties in urine, giving the first indication of the significant breadth of modifications present in the cellular, and urinary, adductomes.

Figure 5. Proof-of-principle: successful generation of NA adductome maps for (A) 2’-dN adducts, (B) rN adducts, (C) nB nB adducts (modified purines), (D) DRCL, (E) RRCL and (F) RPCL, from a pooled urine derived from six healthy subjects, by LC-Q-LIT-OT-MS with CID fragmentation.

Figure 5.

Data are reported as accurate mass-to-charge ratios (m/z; Y axis, left) with retention times (RT; X axis) and associated peak intensities (spot color; Y axis, right), using OriginPro. Eight NA-associated crosslinks (Ions 1-8) were newly identified, and the corresponding isomers of RRCL were further labelled with letter “a” to “d”. Additionally, over 300 ions were noted corresponding to pyrimidine nB (data not shown). The identification of each type of NA modification was achieved by the observation of their protonated precursor ions [M + H]+, and their specific fragmentation features in MS2. The detailed HRMS monitoring parameters are given in the Supplementary information.

HRMS-based ‘omics analysis always generates a massive dataset of product ion spectra. In the present study, the above NA modifications were characterized, and identified one by one using an ion-map function of Thermo Scientific Xcalibur software (ver. 4.1, Thermo Fisher Scientific Inc., USA) combined with extensive manual inspection (as described in Supplementary Experimental Information), which is a complicated and extremely time-consuming process. This laborious process highlights the need of dedicated, post-data analysis software which can characterize, and identify all the specific fragmentation features derived from all types of NA adducts. To date, post-data analysis software is only available for the DNA adductomics 76, 142, 143. Although several data processing methods have been developed for proteomics or metabolomics (e.g., the software XCMS 144), there is still a lack of open-access/freely available automated data analysis methods that can detect compounds through tandem reactions143, such as by tracking specific product ions or a neutral loss MS2 fragmentation, in untargeted NA adductomic approaches.

Despite of the lack of post-data processing software for NA adductomics, we demonstrated the ability to detect novel, and/or unexpected NA modifications such as the modified 2’-dN, rN, DRCL, RRCL and RPCL noted here, and several of the crosslinks we reported previously 73. Combined, this represents a major strength of NA adductomics, and offers a means to fully characterise exposures based upon the spectra of adducts that they induce, together with providing a better understanding of the modifications which lead to mutagenicity.

8. Challenges

There are several challenges to successfully performing NA adductomics, using HRMS, these include: (1) Analytical software. There is a need for high-throughput post-data processing software, as the data sets generated are massive and complex, consisting of various features derived from different types of NA modifications. (2) A comprehensive NA adductome database. Such a database would assist in identification of expected and unexpected adducts. Unlike proteomics or metabolomics, a NA adductome database is not available yet, although DNA adductome databases are in preparation 140, 141. This would allow for searches based upon exact mass, fragmentation pattern, and chemical structures, as well as offering visual representations of the 3D structure, all of which will aid in the identification of NA modifications. (3) Harmonization of LC–HRMS-based DNA, RNA, and NA adductomics approaches. NA adductomics can be performed using a range of HRMS instruments. Many pre-analytical, analytical, and post-analytical factors can impact NA adduct detection, identification, and quantification. Strategies are needed to account for these differences including optimal sample work-up and extraction, removing matrix effects, data normalization, and ion suppression, or using pooled quality control samples and internal standards, for harmonizing cross-platform and cross-laboratory data collected from untargeted HRMS-based NA adductome studies. For example, a reference standardization protocol has been recently proposed for harmonizing large-scale metabolomics data collected across different studies or different HRMS methods 145. Analogous issues have been addressed for the measurement of cellular and urinary nucleic biomarkers of oxidative stress via the formation of international consortia, such as the European Standards Committee on Oxidative DNA Damage, the European Standards Committee on Urinary Lesion Analysis, and the European Comet Assay Validation Group 146. We propose that an equivalent consortium, encompassing all that NA adductomics represents (perhaps in conjunction with protein adductomics), would be the beginning of a strategy towards addressing the above challenges. (4) Biological meaning. Adductomics offers the potential for detecting unexpected adducts whose identity and role is unknown, but their presence potentially significant. As has been seen from other ‘omics approaches, this kind of unbiased, untargeted approach has the potential to reveal new features for mechanistic investigation, but the feature needs to be detected before a biological meaning can be sought. Achieving this requires effort across multiple fields of study. However, a biological meaning is not a prerequisite for use. For example, a particular pattern of gene expression can be ascribed to be the consequence of exposure to a particular agent, without knowing the function of all the genes. The equivalent is likely to be true for NA modifications.

9. Future perspectives, and conclusions

We suggest that the NA adductomic approach (cellular, urinary, and other biomatrices) to biomonitoring environmental exposure will offer a valuable means to contribute to evaluating the role of damage to all nucleic acids in health and disease. For the first time, through the detection of over 1,000 modified NA moieties, we illustrated the scale of the NA adductome further emphasising the need, and potential benefits of the NA adductomic approach, which is analogous to the other ‘omics approaches. The impact of this methodology extends to the study of any disease for which there is an environmental component, and for which our approach will facilitate Adductome-Wide Association Studies (AWAS; an untargeted, agnostic, hypothesis-generating approach for exploring nucleic acid/protein modifications associated with health outcomes, complementary to GWAS, Epigenome-WAS, and Exposome-WAS). As show in Figure 6, we propose applying our approach to the creation of a library of NA adductome maps, consisting of the integrative and informative NA adductome profiles for environmental agents. More broadly, this can be combined with the study of associations between DNA, and RNA modifications, and the relationships between environment-induced chemical modifications, and epigenomic modification (which we term the “NA adductome network”). Our approach also has utility for mechanistic studies e.g., for more comprehensively identifying the processes responsible for genotoxicity; elucidating the spectrum of physiologically relevant NA modifications that are targeted by discrete repair pathways; improving our understanding of how environmental agents impact epigenetics; and performing more detailed exploration of the relationship between NA adductome signatures, mutational signatures 77, 147, 148, epigenome/epitranscriptome signatures 53, 149, and metabolome signatures 150. Combined, this will provide the most comprehensive, multi-adductomic characterizations of environmental agents to date, vital to identifying the components of the exposome which are associated with disease, their mechanisms, and a potential utility in disease risk assessment.

Figure 6. Illustration of how Adductome-Wide Association Studies (AWAS) may be used to explore the environmental causes of cancer.

Figure 6.

The proposed approach may be used to investigate the link between exposure to environmental agents, and health risk. Top panel. The premise is that the exposome results in modification of all types of nucleic acids [DNA, RNA, (d)NTPs], which is associated with an increased risk of developing major diseases, such as cancer. Main figure. In vitro and in vivo models are exposed to a panel of environmental toxicants, individually, and cellular and urinary NA adductome maps are created. These are used to develop a library of NA adductome maps and a network of inter-related NA modifications. Associations are then investigated between NA adductomic “signatures” of specific exposures and mutational, epigenomic, metabolomic, and epitranscriptomic which, together, may elucidate the underlying etiology and/or mechanism(s) of disease.

Supplementary Material

Supplementary data

Acknowledgements

The research reported in this publication was supported, in part, by the National Institute of Environmental Health Sciences of the National Institutes of Health under award number R01ES030557 (to MSC and C-WH). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. This work was also supported by the Ministry of Science and Technology, Taiwan (grant numbers MOST 109-2314-B-040-018-MY3 to M-RC; MOST 110-2628-B-040-002 to C-WH). The sponsors had no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; and in the decision to submit the article for publication.

Footnotes

CRediT author statement

Marcus S. Cooke: Conceptualisation, writing - original draft preparation, reviewing and editing; Yuan-Jhe Chang: investigation, data curation; Yet-Ran Chen: methodology, data curation; Chiung-Wen Hu: conceptualisation, methodology, writing - reviewing and editing; and Mu-Rong Chao: conceptualisation, methodology, data curation, writing - reviewing and editing.

Declaration of competing financial interests

The authors declare they have no actual or potential competing financial interests.

Abbreviations1

1

(2’-deoxy)ribonucleosides (2’-dN/rN), nucleobases (nB), DNA-DNA crosslinks (DDCL); DNA-protein crosslinks (DPCL); RNA-protein crosslinks (RPCL); DNA-RNA crosslinks (DRCL); RNA-RNA crosslinks (RRCL); liquid chromatography-triple quadrupole tandem mass spectrometry (LC-QqQ-MS/MS); high resolution mass spectrometry (HRMS); hybrid quadrupole-linear ion trap Orbitrap MS (Q-LIT-OT-MS); adductome-wide association studies (AWAS; an untargeted, agnostic, hypothesis-generating approach for exploring nucleic acid/protein modifications associated with health outcomes, complementary to GWAS, Epigenome-WAS, and Exposome-WAS)

References

  • 1.Willett WC, Balancing life-style and genomics research for disease prevention. Science 2002, 296, (5568), 695–8. [DOI] [PubMed] [Google Scholar]
  • 2.Smith MT; Rappaport SM, Building exposure biology centers to put the E into “G x E” interaction studies. Environ Health Perspect 2009, 117, (8), A334–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Wild CP; Scalbert A; Herceg Z, Measuring the exposome: a powerful basis for evaluating environmental exposures and cancer risk. Environ Mol Mutagen 2013, 54, (7), 480–99. [DOI] [PubMed] [Google Scholar]
  • 4.Vermeulen R; Schymanski EL; Barabasi AL; Miller GW, The exposome and health: Where chemistry meets biology. Science 2020, 367, (6476), 392–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wild CP, Complementing the genome with an “exposome”: the outstanding challenge of environmental exposure measurement in molecular epidemiology. Cancer Epidemiol Biomarkers Prev 2005, 14, (8), 1847–50. [DOI] [PubMed] [Google Scholar]
  • 6.David A; Chaker J; Price EJ; Bessonneau V; Chetwynd AJ; Vitale CM; Klanova J; Walker DI; Antignac JP; Barouki R; Miller GW, Towards a comprehensive characterisation of the human internal chemical exposome: Challenges and perspectives. Environ Int 2021, 156, 106630. [DOI] [PubMed] [Google Scholar]
  • 7.Smith MT, The Exposome. In Encyclopedia of Toxicology (Third Edition), Wexler P, Ed. Academic Press: 2014. [Google Scholar]
  • 8.Wilson MR; Jiang Y; Villalta PW; Stornetta A; Boudreau PD; Carra A; Brennan CA; Chun E; Ngo L; Samson LD; Engelward BP; Garrett WS; Balbo S; Balskus EP, The human gut bacterial genotoxin colibactin alkylates DNA. Science 2019, 363, (6428). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Biomarkers Definitions Working, G., Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 2001, 69, (3), 89–95. [DOI] [PubMed] [Google Scholar]
  • 10.Gallo V; Egger M; McCormack V; Farmer PB; Ioannidis JP; Kirsch-Volders M; Matullo G; Phillips DH; Schoket B; Stromberg U; Vermeulen R; Wild C; Porta M; Vineis P, STrengthening the reporting of OBservational studies in Epidemiology-Molecular Epidemiology (STROBE-ME): an extension of the STROBE statement. Eur J Epidemiol 2011, 26, (10), 797–810. [DOI] [PubMed] [Google Scholar]
  • 11.Guo J; Turesky RJ, Emerging Technologies in Mass Spectrometry-Based DNA Adductomics. High Throughput 2019, 8, (2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Reddy MC; Vasquez KM, Repair of genome destabilizing lesions. Radiat Res 2005, 164, (4 Pt 1), 345–56. [DOI] [PubMed] [Google Scholar]
  • 13.Zhao LY; Song J; Liu Y; Song CX; Yi C, Mapping the epigenetic modifications of DNA and RNA. Protein Cell 2020, 11, (11), 792–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Phillips DH; Arlt VM, Genotoxicity: damage to DNA and its consequences. Exs 2009, 99, 87–110. [DOI] [PubMed] [Google Scholar]
  • 15.Bellacosa A; Moss EG, RNA repair: damage control. Curr Biol 2003, 13, (12), R482–4. [DOI] [PubMed] [Google Scholar]
  • 16.Xu Z; Huang J; Gao M; Guo G; Zeng S; Chen X; Wang X; Gong Z; Yan Y, Current perspectives on the clinical implications of oxidative RNA damage in aging research: challenges and opportunities. Geroscience 2021, 43, (2), 487–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Poulsen HE; Specht E; Broedbaek K; Henriksen T; Ellervik C; Mandrup-Poulsen T; Tonnesen M; Nielsen PE; Andersen HU; Weimann A, RNA modifications by oxidation: a novel disease mechanism? Free Radic Biol Med 2012, 52, (8), 1353–61. [DOI] [PubMed] [Google Scholar]
  • 18.Kong Q; Lin CL, Oxidative damage to RNA: mechanisms, consequences, and diseases. Cell Mol Life Sci 2010, 67, (11), 1817–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Li Z; Chen X; Liu Z; Ye W; Li L; Qian L; Ding H; Li P; Aung LHH, Recent Advances: Molecular Mechanism of RNA Oxidation and Its Role in Various Diseases. Frontiers in Molecular Biosciences 2020, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Amente S; Scala G; Majello B; Azmoun S; Tempest HG; Premi S; Cooke MS, Genome-wide mapping of genomic DNA damage: methods and implications. Cell Mol Life Sci 2021, 78, (21-22), 6745–6762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gorini F; Scala G; Cooke MS; Majello B; Amente S, Towards a comprehensive view of 8-oxo-7,8-dihydro-2’-deoxyguanosine: Highlighting the intertwined roles of DNA damage and epigenetics in genomic instability. DNA Repair (Amst) 2021, 97, 103027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Evans MD; Cooke MS, Factors contributing to the outcome of oxidative damage to nucleic acids. Bioessays 2004, 26, (5), 533–42. [DOI] [PubMed] [Google Scholar]
  • 23.Evans MD; Dizdaroglu M; Cooke MS, Oxidative DNA damage and disease: induction, repair and significance. Mutat Res 2004, 567, (1), 1–61. [DOI] [PubMed] [Google Scholar]
  • 24.Fimognari C, Role of Oxidative RNA Damage in Chronic-Degenerative Diseases. Oxid Med Cell Longev 2015, 2015, 358713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Takeshita T; Kanaly RA, In vitro DNA/RNA Adductomics to Confirm DNA Damage Caused by Benzo[a]pyrene in the Hep G2 Cell Line. Front Chem 2019, 7, 491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Simms CL; Zaher HS, Quality control of chemically damaged RNA. Cell Mol Life Sci 2016, 73, (19), 3639–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Owiti NA; Nagel ZD; Engelward BP, Fluorescence Sheds Light on DNA Damage, DNA Repair, and Mutations. Trends Cancer 2021, 7, (3), 240–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hofer T; Badouard C; Bajak E; Ravanat JL; Mattsson A; Cotgreave IA, Hydrogen peroxide causes greater oxidation in cellular RNA than in DNA. Biol Chem 2005, 386, (4), 333–7. [DOI] [PubMed] [Google Scholar]
  • 29.Wurtmann EJ; Wolin SL, RNA under attack: cellular handling of RNA damage. Crit Rev Biochem Mol Biol 2009, 44, (1), 34–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Shan X; Chang Y; Lin CL, Messenger RNA oxidation is an early event preceding cell death and causes reduced protein expression. FASEB J 2007, 21, (11), 2753–64. [DOI] [PubMed] [Google Scholar]
  • 31.Tanaka M; Chock PB; Stadtman ER, Oxidized messenger RNA induces translation errors. Proc Natl Acad Sci U S A 2007, 104, (1), 66–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Dai DP; Gan W; Hayakawa H; Zhu JL; Zhang XQ; Hu GX; Xu T; Jiang ZL; Zhang LQ; Hu XD; Nie B; Zhou Y; Li J; Zhou XY; Li J; Zhang TM; He Q; Liu DG; Chen HB; Yang N; Zuo PP; Zhang ZX; Yang HM; Wang Y; Wilson SH; Zeng YX; Wang JY; Sekiguchi M; Cai JP, Transcriptional mutagenesis mediated by 8-oxoG induces translational errors in mammalian cells. Proc Natl Acad Sci U S A 2018, 115, (16), 4218–4222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yan LL; Zaher HS, How do cells cope with RNA damage and its consequences? J Biol Chem 2019, 294, (41), 15158–15171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Rai P; Sobol RW, Mechanisms of MTH1 inhibition-induced DNA strand breaks: The slippery slope from the oxidized nucleotide pool to genotoxic damage. DNA Repair (Amst) 2019, 77, 18–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Nakabeppu Y; Ohta E; Abolhassani N, MTH1 as a nucleotide pool sanitizing enzyme: Friend or foe? Free Radic Biol Med 2017, 107, 151–158. [DOI] [PubMed] [Google Scholar]
  • 36.Rudd SG; Valerie NCK; Helleday T, Pathways controlling dNTP pools to maintain genome stability. DNA Repair (Amst) 2016, 44, 193–204. [DOI] [PubMed] [Google Scholar]
  • 37.Henderson PT; Evans MD; Cooke MS, Salvage of oxidized guanine derivatives in the (2’-deoxy)ribonucleotide pool as source of mutations in DNA. Mutat Res 2010, 703, (1), 11–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chao MR; Evans MD; Hu CW; Ji Y; Moller P; Rossner P; Cooke MS, Biomarkers of nucleic acid oxidation - A summary state-of-the-art. Redox Biol 2021, 42, 101872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Cooke MS; Evans MD; Dizdaroglu M; Lunec J, Oxidative DNA damage: mechanisms, mutation, and disease. FASEB J 2003, 17, (10), 1195–214. [DOI] [PubMed] [Google Scholar]
  • 40.Suganuma T; Workman JL, Nucleotide Metabolism Behind Epigenetics. Front Endocrinol (Lausanne) 2021, 12, 731648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cadet J; Grand A; Douki T, Solar UV radiation-induced DNA Bipyrimidine photoproducts: formation and mechanistic insights. Top Curr Chem 2015, 356, 249–75. [DOI] [PubMed] [Google Scholar]
  • 42.Coskun E; Jaruga P; Vartanian V; Erdem O; Egner PA; Groopman JD; Lloyd RS; Dizdaroglu M, Aflatoxin-Guanine DNA Adducts and Oxidatively Induced DNA Damage in Aflatoxin-Treated Mice in Vivo as Measured by Liquid Chromatography-Tandem Mass Spectrometry with Isotope Dilution. Chem Res Toxicol 2019, 32, (1), 80–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bellamri M; Brandt K; Brown CV; Wu MT; Turesky RJ, Cytotoxicity and genotoxicity of the carcinogen aristolochic acid I (AA-I) in human bladder RT4 cells. Arch Toxicol 2021, 95, (6), 2189–2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ziech D; Franco R; Georgakilas AG; Georgakila S; Malamou-Mitsi V; Schoneveld O; Pappa A; Panayiotidis MI, The role of reactive oxygen species and oxidative stress in environmental carcinogenesis and biomarker development. Chem Biol Interact 2010, 188, (2), 334–9. [DOI] [PubMed] [Google Scholar]
  • 45.Sun K; Song Y; He F; Jing M; Tang J; Liu R, A review of human and animals exposure to polycyclic aromatic hydrocarbons: Health risk and adverse effects, photo-induced toxicity and regulating effect of microplastics. Science of The Total Environment 2021, 773, 145403. [DOI] [PubMed] [Google Scholar]
  • 46.Bukowska B; Sicinska P, Influence of Benzo(a)pyrene on Different Epigenetic Processes. Int J Mol Sci 2021, 22, (24). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Desaulniers D; Xiao GH; Lian H; Feng YL; Zhu J; Nakai J; Bowers WJ, Effects of mixtures of polychlorinated biphenyls, methylmercury, and organochlorine pesticides on hepatic DNA methylation in prepubertal female Sprague-Dawley rats. Int J Toxicol 2009, 28, (4), 294–307. [DOI] [PubMed] [Google Scholar]
  • 48.Peters A; Nawrot TS; Baccarelli AA, Hallmarks of environmental insults. Cell 2021, 184, (6), 1455–1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sies H, Oxidative eustress: On constant alert for redox homeostasis. Redox Biol 2021, 41, 101867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chan SW; Dedon PC, The biological and metabolic fates of endogenous DNA damage products. J Nucleic Acids 2010, 2010, 929047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Hwa Yun B; Guo J; Bellamri M; Turesky RJ, DNA adducts: Formation, biological effects, and new biospecimens for mass spectrometric measurements in humans. Mass Spectrom Rev 2020, 39, (1-2), 55–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Gao X; Holleczek B; Cuk K; Zhang Y; Anusruti A; Xuan Y; Xu Y; Brenner H; Schottker B, Investigation on potential associations of oxidatively generated DNA/RNA damage with lung, colorectal, breast, prostate and total cancer incidence. Sci Rep 2019, 9, (1), 7109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Esteve-Puig R; Bueno-Costa A; Esteller M, Writers, readers and erasers of RNA modifications in cancer. Cancer Lett 2020, 474, 127–137. [DOI] [PubMed] [Google Scholar]
  • 54.Boccaletto P; Baginski B, MODOMICS: An Operational Guide to the Use of the RNA Modification Pathways Database. Methods Mol Biol 2021, 2284, 481–505. [DOI] [PubMed] [Google Scholar]
  • 55.Boccaletto P; Machnicka MA; Purta E; Piatkowski P; Baginski B; Wirecki TK; de Crecy-Lagard V; Ross R; Limbach PA; Kotter A; Helm M; Bujnicki JM, MODOMICS: a database of RNA modification pathways. 2017 update. Nucleic Acids Res 2018, 46, (D1), D303–D307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Boccaletto P; Stefaniak F; Ray A; Cappannini A; Mukherjee S; Purta E; Kurkowska M; Shirvanizadeh N; Destefanis E; Groza P; Avsar G; Romitelli A; Pir P; Dassi E; Conticello SG; Aguilo F; Bujnicki JM, MODOMICS: a database of RNA modification pathways. 2021 update. Nucleic Acids Res 2022, 50, (D1), D231–D235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Czerwoniec A; Dunin-Horkawicz S; Purta E; Kaminska KH; Kasprzak JM; Bujnicki JM; Grosjean H; Rother K, MODOMICS: a database of RNA modification pathways. 2008 update. Nucleic Acids Res 2009, 37, (Database issue), D118–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Dunin-Horkawicz S; Czerwoniec A; Gajda MJ; Feder M; Grosjean H; Bujnicki JM, MODOMICS: a database of RNA modification pathways. Nucleic Acids Res 2006, 34, (Database issue), D145–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Machnicka MA; Milanowska K; Osman Oglou O; Purta E; Kurkowska M; Olchowik A; Januszewski W; Kalinowski S; Dunin-Horkawicz S; Rother KM; Helm M; Bujnicki JM; Grosjean H, MODOMICS: a database of RNA modification pathways--2013 update. Nucleic Acids Res 2013, 41, (Database issue), D262–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Groehler A. t.; Degner A; Tretyakova NY, Mass Spectrometry-Based Tools to Characterize DNA-Protein Cross-Linking by Bis-Electrophiles. Basic Clin Pharmacol Toxicol 2017, 121 Suppl 3, 63–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Weickert P; Stingele J, DNA-Protein Crosslinks and Their Resolution. Annu Rev Biochem 2022. [DOI] [PubMed] [Google Scholar]
  • 62.Kramer K; Sachsenberg T; Beckmann BM; Qamar S; Boon KL; Hentze MW; Kohlbacher O; Urlaub H, Photo-cross-linking and high-resolution mass spectrometry for assignment of RNA-binding sites in RNA-binding proteins. Nat Methods 2014, 11, (10), 1064–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Himmelstein MW; Boogaard PJ; Cadet J; Farmer PB; Kim JH; Martin EA; Persaud R; Shuker DE, Creating context for the use of DNA adduct data in cancer risk assessment: II. Overview of methods of identification and quantitation of DNA damage. Crit Rev Toxicol 2009, 39, (8), 679–94. [DOI] [PubMed] [Google Scholar]
  • 64.Kanaly RA; Hanaoka T; Sugimura H; Toda H; Matsui S; Matsuda T, Development of the adductome approach to detect DNA damage in humans. Antioxid Redox Signal 2006, 8, (5-6), 993–1001. [DOI] [PubMed] [Google Scholar]
  • 65.Ohnishi I; Iwashita Y; Matsushita Y; Ohtsuka S; Yamashita T; Inaba K; Fukazawa A; Ochiai H; Matsumoto K; Kurono N; Matsushima Y; Mori H; Suzuki S; Suzuki S; Tanioka F; Sugimura H, Mass spectrometric profiling of DNA adducts in the human stomach associated with damage from environmental factors. Genes Environ 2021, 43, (1), 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.La Barbera G; Dragsted LO, Development of an untargeted DNA adductomics method by ultra-high performance liquid chromatography coupled to high-resolution mass spectrometry. . ChemRxiv. 2022. [DOI] [PubMed] [Google Scholar]
  • 67.Balbo S; Hecht SS; Upadhyaya P; Villalta PW, Application of a high-resolution mass-spectrometry-based DNA adductomics approach for identification of DNA adducts in complex mixtures. Anal Chem 2014, 86, (3), 1744–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Guo J; Villalta PW; Weight CJ; Bonala R; Johnson F; Rosenquist TA; Turesky RJ, Targeted and Untargeted Detection of DNA Adducts of Aromatic Amine Carcinogens in Human Bladder by Ultra-Performance Liquid Chromatography-High-Resolution Mass Spectrometry. Chem Res Toxicol 2018, 31, (12), 1382–1397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Villalta PW; Balbo S, The Future of DNA Adductomic Analysis. Int J Mol Sci 2017, 18, (9). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hemeryck LY; Decloedt AI; Vanden Bussche J; Geboes KP; Vanhaecke L, High resolution mass spectrometry based profiling of diet-related deoxyribonucleic acid adducts. Anal Chim Acta 2015, 892, 123–31. [DOI] [PubMed] [Google Scholar]
  • 71.Chang YJ; Cooke MS; Hu CW; Chao MR, Novel approach to integrated DNA adductomics for the assessment of in vitro and in vivo environmental exposures. Arch Toxicol 2018, 92, (8), 2665–2680. [DOI] [PubMed] [Google Scholar]
  • 72.Chang YJ; Cooke MS; Chen YR; Yang SF; Li PS; Hu CW; Chao MR, Is high resolution a strict requirement for mass spectrometry-based cellular DNA adductomics? Chemosphere 2021, 274, 129991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Hu CW; Chang YJ; Cooke MS; Chao MR, DNA Crosslinkomics: A Tool for the Comprehensive Assessment of Interstrand Crosslinks Using High Resolution Mass Spectrometry. Anal Chem 2019, 91, (23), 15193–15203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Ayadi L; Galvanin A; Pichot F; Marchand V; Motorin Y, RNA ribose methylation (2’-O-methylation): Occurrence, biosynthesis and biological functions. Biochim Biophys Acta Gene Regul Mech 2019, 1862, (3), 253–269. [DOI] [PubMed] [Google Scholar]
  • 75.Kanaly RA; Matsui S; Hanaoka T; Matsuda T, Application of the adductome approach to assess intertissue DNA damage variations in human lung and esophagus. Mutat Res 2007, 625, (1-2), 83–93. [DOI] [PubMed] [Google Scholar]
  • 76.Walmsley SJ; Guo J; Murugan P; Weight CJ; Wang J; Villalta PW; Turesky RJ, Comprehensive Analysis of DNA Adducts Using Data-Independent wSIM/MS(2) Acquisition and wSIM-City. Anal Chem 2021, 93, (16), 6491–6500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Totsuka Y; Lin Y; He Y; Ishino K; Sato H; Kato M; Nagai M; Elzawahry A; Totoki Y; Nakamura H; Hosoda F; Shibata T; Matsuda T; Matsushima Y; Song G; Meng F; Li D; Liu J; Qiao Y; Wei W; Inoue M; Kikuchi S; Nakagama H; Shan B, DNA Adductome Analysis Identifies N-Nitrosopiperidine Involved in the Etiology of Esophageal Cancer in Cixian, China. Chem Res Toxicol 2019, 32, (8), 1515–1527. [DOI] [PubMed] [Google Scholar]
  • 78.Randell RL; Gulati AS; Cook SF; Martin CF; Chen W; Jaeger EL; Schoenborn AA; Basta PV; Dejong H; Luo J; Gallant M; Sandler RS; Long MD; Kappelman MD, Collecting Biospecimens From an Internet-Based Prospective Cohort Study of Inflammatory Bowel Disease (CCFA Partners): A Feasibility Study. JMIR Res Protoc 2016, 5, (1), e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hansen TV; Simonsen MK; Nielsen FC; Hundrup YA, Collection of blood, saliva, and buccal cell samples in a pilot study on the Danish nurse cohort: comparison of the response rate and quality of genomic DNA. Cancer Epidemiol Biomarkers Prev 2007, 16, (10), 2072–6. [DOI] [PubMed] [Google Scholar]
  • 80.Yun BH; Bellamri M; Rosenquist TA; Turesky RJ, Method for Biomonitoring DNA Adducts in Exfoliated Urinary Cells by Mass Spectrometry. Anal Chem 2018, 90, (16), 9943–9950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Cooke MS; Evans MD; Dove R; Rozalski R; Gackowski D; Siomek A; Lunec J; Olinski R, DNA repair is responsible for the presence of oxidatively damaged DNA lesions in urine. Mutat Res 2005, 574, (1-2), 58–66. [DOI] [PubMed] [Google Scholar]
  • 82.Rozalski R; Siomek A; Gackowski D; Foksinski M; Gran C; Klungland A; Olinski R, Substantial decrease of urinary 8-oxo-7,8-dihydroguanine, a product of the base excision repair pathway, in DNA glycosylase defective mice. Int J Biochem Cell Biol 2005, 37, (6), 1331–6. [DOI] [PubMed] [Google Scholar]
  • 83.Foksinski M; Rozalski R; Guz J; Ruszkowska B; Sztukowska P; Piwowarski M; Klungland A; Olinski R, Urinary excretion of DNA repair products correlates with metabolic rates as well as with maximum life spans of different mammalian species. Free Radic Biol Med 2004, 37, (9), 1449–54. [DOI] [PubMed] [Google Scholar]
  • 84.Evans MD; Saparbaev M; Cooke MS, DNA repair and the origins of urinary oxidized 2’-deoxyribonucleosides. Mutagenesis 2010, 25, (5), 433–42. [DOI] [PubMed] [Google Scholar]
  • 85.Jumpathong W; Chan W; Taghizadeh K; Babu IR; Dedon PC, Metabolic fate of endogenous molecular damage: Urinary glutathione conjugates of DNA-derived base propenals as markers of inflammation. Proc Natl Acad Sci U S A 2015, 112, (35), E4845–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kemp MG; Reardon JT; Lindsey-Boltz LA; Sancar A, Mechanism of release and fate of excised oligonucleotides during nucleotide excision repair. J Biol Chem 2012, 287, (27), 22889–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Cooke MS; Harry EL; Liljendahl TS; Segerback D, DNA nucleotide excision repair, where do all the cyclobutane pyrimidine dimers go? Cell Cycle 2013, 12, (10), 1642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Jaruga P; Dizdaroglu M, Identification and quantification of (5’R)- and (5’S)-8,5’-cyclo-2’-deoxyadenosines in human urine as putative biomarkers of oxidatively induced damage to DNA. Biochem Biophys Res Commun 2010, 397, (1), 48–52. [DOI] [PubMed] [Google Scholar]
  • 89.Jaruga P; Rozalski R; Jawien A; Migdalski A; Olinski R; Dizdaroglu M, DNA damage products (5’R)- and (5’S)-8,5’-cyclo-2’-deoxyadenosines as potential biomarkers in human urine for atherosclerosis. Biochemistry 2012, 51, (9), 1822–4. [DOI] [PubMed] [Google Scholar]
  • 90.Kant M; Akis M; Calan M; Arkan T; Bayraktar F; Dizdaroglu M; Islekel H, Elevated urinary levels of 8-oxo-2’-deoxyguanosine, (5’R)- and (5’S)-8,5’-cyclo-2’-deoxyadenosines, and 8-iso-prostaglandin F2alpha as potential biomarkers of oxidative stress in patients with prediabetes. DNA Repair (Amst) 2016, 48, 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Hoberg AM; Otteneder M; Marnett LJ; Poulsen HE, Measurement of the malondialdehyde-2’-deoxyguanosine adduct in human urine by immuno-extraction and liquid chromatography/atmospheric pressure chemical ionization tandem mass spectrometry. J Mass Spectrom 2004, 39, (1), 38–42. [DOI] [PubMed] [Google Scholar]
  • 92.Otteneder MB; Knutson CG; Daniels JS; Hashim M; Crews BC; Remmel RP; Wang H; Rizzo C; Marnett LJ, In vivo oxidative metabolism of a major peroxidation-derived DNA adduct, M1dG. Proc Natl Acad Sci U S A 2006, 103, (17), 6665–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.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, (29), 22355–62. [DOI] [PubMed] [Google Scholar]
  • 94.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, (8), 3832–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Cooke MS; Evans MD; Herbert KE; Lunec J, Urinary 8-oxo-2’-deoxyguanosine--source, significance and supplements. Free Radic Res 2000, 32, (5), 381–97. [DOI] [PubMed] [Google Scholar]
  • 96.Mueller N; Risenbrand G, The influence of N7 substituents on the stability of N7-alkylated guanosines. Chem Biol Interact 1985, 53, 173–181. [DOI] [PubMed] [Google Scholar]
  • 97.Smith JW; Groopman JD, Aflatoxins. In Encyclopedia of Cancer (Third Edition), Boffetta P; Hainaut P, Eds. Academic Press: Oxford, 2019; pp 30–43. [Google Scholar]
  • 98.Loft S; Poulsen HE, Markers of oxidative damage to DNA: antioxidants and molecular damage. Methods Enzymol 1999, 300, 166–84. [DOI] [PubMed] [Google Scholar]
  • 99.Basu AK, DNA Damage, Mutagenesis and Cancer. Int J Mol Sci 2018, 19, (4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Kemp MG; Hu J, PostExcision Events in Human Nucleotide Excision Repair. Photochem Photobiol 2017, 93, (1), 178–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Mikes P; Sistek V; Krouzelka J; Kralik A; Frantik E; Mraz J; Linhart I, 3-(3,4-Dihydroxyphenyl)adenine, a urinary DNA adduct formed in mice exposed to high concentrations of benzene. J Appl Toxicol 2013, 33, (6), 516–20. [DOI] [PubMed] [Google Scholar]
  • 102.Walton M; Egner P; Scholl PF; Walker J; Kensler TW; Groopman JD, Liquid chromatography electrospray-mass spectrometry of urinary aflatoxin biomarkers: characterization and application to dosimetry and chemoprevention in rats. Chem Res Toxicol 2001, 14, (7), 919–26. [DOI] [PubMed] [Google Scholar]
  • 103.Erber L; Goodman S; Wright FA; Chiu WA; Tretyakova NY; Rusyn I, Intra- and Inter-Species Variability in Urinary N7-(1-Hydroxy-3-buten-2-yl)guanine Adducts Following Inhalation Exposure to 1,3-Butadiene. Chem Res Toxicol 2021, 34, (11), 2375–2383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Sangaraju D; Boldry EJ; Patel YM; Walker V; Stepanov I; Stram D; Hatsukami D; Tretyakova N, Isotope Dilution nanoLC/ESI(+)-HRMS(3) Quantitation of Urinary N7-(1-Hydroxy-3-buten-2-yl) Guanine Adducts in Humans and Their Use as Biomarkers of Exposure to 1,3-Butadiene. Chem Res Toxicol 2017, 30, (2), 678–688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Erber L; Goodman S; Jokipii Krueger CC; Rusyn I; Tretyakova N, Quantitative NanoLC/NSI(+)-HRMS Method for 1,3-Butadiene Induced bis-N7-guanine DNA-DNA Cross-Links in Urine. Toxics 2021, 9, (10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Abdullah UB; McGouran JF; Brolih S; Ptchelkine D; El-Sagheer AH; Brown T; McHugh PJ, RPA activates the XPF-ERCC1 endonuclease to initiate processing of DNA interstrand crosslinks. EMBO J 2017, 36, (14), 2047–2060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Yang Z; Nejad MI; Varela JG; Price NE; Wang Y; Gates KS, A role for the base excision repair enzyme NEIL3 in replication-dependent repair of interstrand DNA cross-links derived from psoralen and abasic sites. DNA Repair (Amst) 2017, 52, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kothandapani A; Patrick SM, Evidence for base excision repair processing of DNA interstrand crosslinks. Mutat Res 2013, 743-744, 44–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Kojima Y; Machida YJ, DNA-protein crosslinks from environmental exposure: Mechanisms of formation and repair. Environ Mol Mutagen 2020, 61, (7), 716–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Stingele J; Bellelli R; Boulton SJ, Mechanisms of DNA-protein crosslink repair. Nat Rev Mol Cell Biol 2017, 18, (9), 563–573. [DOI] [PubMed] [Google Scholar]
  • 111.Saha S; Sun Y; Huang SN; Baechler SA; Pongor LS; Agama K; Jo U; Zhang H; Tse-Dinh YC; Pommier Y, DNA and RNA Cleavage Complexes and Repair Pathway for TOP3B RNA- and DNA-Protein Crosslinks. Cell Rep 2020, 33, (13), 108569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Yi C; He C, DNA Repair by Reversal of DNA Damage. Cold Spring Harbor Perspectives in Biology 2013, 5, (1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Cooke MS; Evans MD; Burd RM; Patel K; Barnard A; Lunec J; Hutchinson PE, Induction and excretion of ultraviolet-induced 8-oxo-2’-deoxyguanosine and thymine dimers in vivo: implications for PUVA. J Invest Dermatol 2001, 116, (2), 281–5. [DOI] [PubMed] [Google Scholar]
  • 114.Shih BB; Farrar MD; Cooke MS; Osman J; Langton AK; Kift R; Webb AR; Berry JL; Watson REB; Vail A; de Gruijl FR; Rhodes LE, Fractional Sunburn Threshold UVR Doses Generate Equivalent Vitamin D and DNA Damage in Skin Types I-VI but with Epidermal DNA Damage Gradient Correlated to Skin Darkness. J Invest Dermatol 2018, 138, (10), 2244–2252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Fraga CG; Shigenaga MK; Park JW; Degan P; Ames BN, Oxidative damage to DNA during aging: 8-hydroxy-2’-deoxyguanosine in rat organ DNA and urine. Proc Natl Acad Sci U S A 1990, 87, (12), 4533–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Park EM; Shigenaga MK; Degan P; Korn TS; Kitzler JW; Wehr CM; Kolachana P; Ames BN, Assay of excised oxidative DNA lesions: isolation of 8-oxoguanine and its nucleoside derivatives from biological fluids with a monoclonal antibody column. Proc Natl Acad Sci U S A 1992, 89, (8), 3375–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Cooke MS; Lunec J; Evans MD, Progress in the analysis of urinary oxidative DNA damage. Free Radic Biol Med 2002, 33, (12), 1601–14. [DOI] [PubMed] [Google Scholar]
  • 118.Gackowski D; Rozalski R; Roszkowski K; Jawien A; Foksinski M; Olinski R, 8-Oxo-7,8-dihydroguanine and 8-oxo-7,8-dihydro-2’-deoxyguanosine levels in human urine do not depend on diet. Free Radic Res 2001, 35, (6), 825–32. [DOI] [PubMed] [Google Scholar]
  • 119.Rozalski R; Siomek A; Gackowski D; Foksinski M; Gran C; Klungland A; Olinski R, Diet is not responsible for the presence of several oxidatively damaged DNA lesions in mouse urine. Free Radic Res 2004, 38, (11), 1201–5. [DOI] [PubMed] [Google Scholar]
  • 120.Barregard L; Moller P; Henriksen T; Mistry V; Koppen G; Rossner P Jr.; Sram RJ; Weimann A; Poulsen HE; Nataf R; Andreoli R; Manini P; Marczylo T; Lam P; Evans MD; Kasai H; Kawai K; Li YS; Sakai K; Singh R; Teichert F; Farmer PB; Rozalski R; Gackowski D; Siomek A; Saez GT; Cerda C; Broberg K; Lindh C; Hossain MB; Haghdoost S; Hu CW; Chao MR; Wu KY; Orhan H; Senduran N; Smith RJ; Santella RM; Su Y; Cortez C; Yeh S; Olinski R; Loft S; Cooke MS, Human and methodological sources of variability in the measurement of urinary 8-oxo-7,8-dihydro-2’-deoxyguanosine. Antioxid Redox Signal 2013, 18, (18), 2377–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Loft S; Svoboda P; Kasai H; Tjonneland A; Vogel U; Moller P; Overvad K; Raaschou-Nielsen O, Prospective study of 8-oxo-7,8-dihydro-2’-deoxyguanosine excretion and the risk of lung cancer. Carcinogenesis 2006, 27, (6), 1245–50. [DOI] [PubMed] [Google Scholar]
  • 122.Cooke MS; Olinski R; Loft S; European Standards Committee on Urinary Lesion, A., Measurement and meaning of oxidatively modified DNA lesions in urine. Cancer Epidemiol Biomarkers Prev 2008, 17, (1), 3–14. [DOI] [PubMed] [Google Scholar]
  • 123.Aylward LL; Hays SM; Zidek A, Variation in urinary spot sample, 24 h samples, and longer-term average urinary concentrations of short-lived environmental chemicals: implications for exposure assessment and reverse dosimetry. J Expo Sci Environ Epidemiol 2017, 27, (6), 582–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Joergensen A; Broedbaek K; Weimann A; Semba RD; Ferrucci L; Joergensen MB; Poulsen HE, Association between urinary excretion of cortisol and markers of oxidatively damaged DNA and RNA in humans. PLoS One 2011, 6, (6), e20795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Broedbaek K; Weimann A; Stovgaard ES; Poulsen HE, Urinary 8-oxo-7,8-dihydro-2’-deoxyguanosine as a biomarker in type 2 diabetes. Free Radic Biol Med 2011, 51, (8), 1473–9. [DOI] [PubMed] [Google Scholar]
  • 126.Da Pieve C; Sahgal N; Moore SA; Velasco-Garcia MN, Development of a liquid chromatography/tandem mass spectrometry method to investigate the presence of biomarkers of DNA damage in urine related to red meat consumption and risk of colorectal cancer. Rapid Commun Mass Spectrom 2013, 27, (21), 2493–503. [DOI] [PubMed] [Google Scholar]
  • 127.Hu K; Zhao G; Liu J; Jia L; Xie F; Zhang S; Liu H; Liu M, Simultaneous quantification of three alkylatedpurine adducts in human urine using sulfonic acid poly(glycidyl methacrylatedivinylbenzene)-based microspheres as sorbent combined with LC-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2018, 1081-1082, 15–24. [DOI] [PubMed] [Google Scholar]
  • 128.Leung EM; Chan W, Noninvasive measurement of aristolochic acid-DNA adducts in urine samples from aristolochic acid-treated rats by liquid chromatography coupled tandem mass spectrometry: evidence for DNA repair by nucleotide-excision repair mechanisms. Mutat Res 2014, 766-767, 1–6. [DOI] [PubMed] [Google Scholar]
  • 129.Solivio MJ; Stornetta A; Gilissen J; Villalta PW; Deschoemaeker S; Heyerick A; Dubois L; Balbo S, In Vivo Identification of Adducts from the New Hypoxia-Activated Prodrug CP-506 Using DNA Adductomics. Chem Res Toxicol 2022, 35, (2), 275–282. [DOI] [PubMed] [Google Scholar]
  • 130.Guidolin V; Carlson ES; Carra A; Villalta PW; Maertens LA; Hecht SS; Balbo S, Identification of New Markers of Alcohol-Derived DNA Damage in Humans. Biomolecules 2021, 11, (3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Alexander EM; Kreitler DF; Guidolin V; Hurben AK; Drake E; Villalta PW; Balbo S; Gulick AM; Aldrich CC, Biosynthesis, Mechanism of Action, and Inhibition of the Enterotoxin Tilimycin Produced by the Opportunistic Pathogen Klebsiella oxytoca. ACS Infect Dis 2020, 6, (7), 1976–1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Kanaly RA; Micheletto R; Matsuda T; Utsuno Y; Ozeki Y; Hamamura N, Application of DNA adductomics to soil bacterium Sphingobium sp. strain KK22. Microbiologyopen 2015, 4, (5), 841–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Nakano T; Xu X; Salem AMH; Shoulkamy MI; Ide H, Radiation-induced DNA-protein cross-links: Mechanisms and biological significance. Free Radic Biol Med 2017, 107, 136–145. [DOI] [PubMed] [Google Scholar]
  • 134.Hentze MW; Castello A; Schwarzl T; Preiss T, A brave new world of RNA-binding proteins. Nat Rev Mol Cell Biol 2018, 19, (5), 327–341. [DOI] [PubMed] [Google Scholar]
  • 135.Karmakar S; Harcourt EM; Hewings DS; Scherer F; Lovejoy AF; Kurtz DM; Ehrenschwender T; Barandun LJ; Roost C; Alizadeh AA; Kool ET, Organocatalytic removal of formaldehyde adducts from RNA and DNA bases. Nat Chem 2015, 7, (9), 752–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Carra A; Guidolin V; Dator RP; Upadhyaya P; Kassie F; Villalta PW; Balbo S, Targeted High Resolution LC/MS(3) Adductomics Method for the Characterization of Endogenous DNA Damage. Front Chem 2019, 7, 658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Takeshita T; Tao F; Kojima N; Kanaly RA, Triple quadrupole mass spectrometry comparative DNA adductomics of Hep G2 cells following exposure to safrole. Toxicol Lett 2019, 300, 92–104. [DOI] [PubMed] [Google Scholar]
  • 138.Cooke MS; Hu CW; Chang YJ; Chao MR, Urinary DNA adductomics - A novel approach for exposomics. Environ Int 2018, 121, (Pt 2), 1033–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Housh K; Jha JS; Yang Z; Haldar T; Johnson KM; Yin J; Wang Y; Gates KS, Formation and Repair of an Interstrand DNA Cross-Link Arising from a Common Endogenous Lesion. J Am Chem Soc 2021, 143, (37), 15344–15357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Guo J; Turesky RJ; Tarifa A; DeCaprio AP; Cooke MS; Walmsley SJ; Villalta PW, Development of a DNA Adductome Mass Spectral Database. Chem Res Toxicol 2020, 33, (4), 852–854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.La Barbera G; Nommesen KD; Cuparencu C; Stanstrup J; Dragsted LO, A Comprehensive Database for DNA Adductomics. Frontiers in Chemistry 2022, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Murray KJ; Carlson ES; Stornetta A; Balskus EP; Villalta PW; Balbo S, Extension of Diagnostic Fragmentation Filtering for Automated Discovery in DNA Adductomics. Anal Chem 2021, 93, (14), 5754–5762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Sousa PFM; Martella G; Aberg KM; Esfahani B; Motwani HV, nLossFinder-A Graphical User Interface Program for the Nontargeted Detection of DNA Adducts. Toxics 2021, 9, (4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Gadara D; Coufalikova K; Bosak J; Smajs D; Spacil Z, Systematic Feature Filtering in Exploratory Metabolomics: Application toward Biomarker Discovery. Anal Chem 2021, 93, (26), 9103–9110. [DOI] [PubMed] [Google Scholar]
  • 145.Liu KH; Nellis M; Uppal K; Ma C; Tran V; Liang Y; Walker DI; Jones DP, Reference Standardization for Quantification and Harmonization of Large-Scale Metabolomics. Anal Chem 2020, 92, (13), 8836–8844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Moller P; Cooke MS; Collins A; Olinski R; Rozalski R; Loft S, Harmonising measurements of 8-oxo-7,8-dihydro-2’-deoxyguanosine in cellular DNA and urine. Free Radic Res 2012, 46, (4), 541–53. [DOI] [PubMed] [Google Scholar]
  • 147.Kucab JE; Zou X; Morganella S; Joel M; Nanda AS; Nagy E; Gomez C; Degasperi A; Harris R; Jackson SP; Arlt VM; Phillips DH; Nik-Zainal S, A Compendium of Mutational Signatures of Environmental Agents. Cell 2019, 177, (4), 821–836 e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Totsuka Y; Watanabe M; Lin Y, New horizons of DNA adductome for exploring environmental causes of cancer. Cancer Sci 2021, 112, (1), 7–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Kan RL; Chen J; Sallam T, Crosstalk between epitranscriptomic and epigenetic mechanisms in gene regulation. Trends Genet 2022, 38, (2), 182–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Deng P; Li X; Petriello MC; Wang C; Morris AJ; Hennig B, Application of metabolomics to characterize environmental pollutant toxicity and disease risks. Rev Environ Health 2019, 34, (3), 251–259. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary data

RESOURCES