ArticlePDF AvailableLiterature Review

Protein Adductomics: Analytical Developments and Applications in Human Biomonitoring

Authors:

Abstract and Figures

Proteins contain many sites that are subject to modification by electrophiles. Detection and characterisation of these modifications can give insights into environmental agents and endogenous processes that may be contributing factors to chronic human diseases. An untargeted approach, utilising mass spectrometry to detect modified amino acids or peptides, has been applied to blood proteins haemoglobin and albumin, focusing in particular on the N-terminal valine residue of haemoglobin and the cysteine-34 residue in albumin. Technical developments to firstly detect simultaneously multiple adducts at these sites and then subsequently to identify them are reviewed here. Recent studies in which the methods have been applied to biomonitoring human exposure to environmental toxicants are described. With advances in sensitivity, high-throughput handling of samples and robust quality control, these methods have considerable potential for identifying causes of human chronic disease and of identifying individuals at risk.
Content may be subject to copyright.
toxics
Review
Protein Adductomics: Analytical Developments and
Applications in Human Biomonitoring
George W. Preston and David H. Phillips *
Environmental Research Group, Department of Analytical, Environmental and Forensic Science,
School of Population Health and Environmental Sciences, King’s College London, Franklin-Wilkins Building,
150 Stamford Street, London SE1 9NH, UK; george.preston@kcl.ac.uk
*Correspondence: david.phillips@kcl.ac.uk
Received: 22 March 2019; Accepted: 20 May 2019; Published: 25 May 2019


Abstract:
Proteins contain many sites that are subject to modification by electrophiles. Detection and
characterisation of these modifications can give insights into environmental agents and endogenous
processes that may be contributing factors to chronic human diseases. An untargeted approach,
utilising mass spectrometry to detect modified amino acids or peptides, has been applied to blood
proteins haemoglobin and albumin, focusing in particular on the N-terminal valine residue of
haemoglobin and the cysteine-34 residue in albumin. Technical developments to firstly detect
simultaneously multiple adducts at these sites and then subsequently to identify them are reviewed
here. Recent studies in which the methods have been applied to biomonitoring human exposure to
environmental toxicants are described. With advances in sensitivity, high-throughput handling of
samples and robust quality control, these methods have considerable potential for identifying causes
of human chronic disease and of identifying individuals at risk.
Keywords: haemoglobin; albumin; mass spectrometry; biomarkers; protein adducts
1. The Exposome and Adductomics
Many decades of epidemiological observations have indicated that incidences of chronic human
diseases are likely to result from a combination of environmental exposures to chemical and physical
stressors, and predispositions inherent in human genetics. The wide geographical variation of many
such diseases implies that it is environmental factors that play the dominant role, and not inherited
predisposition, in disease causation [
1
], but knowledge of what the environmental factors are is often
far from complete. As a consequence, estimations of overall risks associated with these factors are
inaccurate and important associations may go undetected. These limitations have recently been
framed within the context of the exposome, which can be thought of as the environmental counterpart
of the genome. Conceptually, the exposome aims to reflect the totality of environmental exposures
throughout the human lifespan, and to take into account both external components (e.g., exogenous
environmental agents) and internal ones (e.g., endogenous cellular processes that give rise to altered
stasis or function) [
2
5
]. For strategies to improve human health to be eective, it is essential to
unravel the causes of chronic human diseases and to assess accurately their risks. The goal of
studying the exposome (i.e., of exposomics) is disease prevention through the acquisition of a broad
scientific perspective that encompasses health, environmental, educational, socioeconomic and political
factors [69].
Two recent collaborative projects have applied the exposome concept to investigating
environmental impacts on human health by assessing environmental exposure at personal and
population levels within existing short- and long-term population studies. In the EXPOsOMICS
project the emphasis has been on the measurement and impact of air and water pollution, studied
Toxics 2019,7, 29; doi:10.3390/toxics7020029 www.mdpi.com/journal/toxics
Toxics 2019,7, 29 2 of 17
in a number of adult and child study populations [
10
]. In the HELIX project the focus has been on
early-life events, examining exposure to a range of chemicals and physical agents in existing birth
cohorts [
11
]. Both these projects utilised a combination of exposure monitoring, using mobile and static
monitors, smartphone and satellite data, and omics techniques to investigate biomarkers associated
with exposures. The multi-omic approach has included metabolome, proteome, transcriptome,
epigenome and adductome profiles. While many of the results of these interrelated analyses have
yet to emerge, it is anticipated that new insights into the importance of environmental factors in the
aetiology of human diseases will ensue and that the studies will point the way to improved strategies
for monitoring human exposures and their health consequences.
While these projects have focused on human exposures and health outcomes, broader ecological
issues may also be addressed by the exposome concept. The adverse outcome pathway (AOP) concept
seeks to define the initial molecular events that culminate in adverse (toxicological) endpoints [
12
].
There is currently much discussion of how to assess the properties of complex mixtures of chemicals,
taking into consideration possible positive and negative interactions between their components,
in order to refine hazard identification and risk assessment. It has been proposed that considering the
relative contributions of components of the exposome in relation to complex mixtures combined with a
mechanistic understanding of the induced adverse eects, may improve the integrated risk assessment
for both human and environmental health [13].
Electrophiles have long been suspected in the causality of cancer and other chronic diseases.
Because they are reactive, they can be measured indirectly through the adducts they form with
protein and DNA. Indeed, damage to, or modification of, DNA by reactive intermediates of chemical
carcinogens or by ionising and non-ionising radiation is a key early event in the carcinogenic process.
The exposome concept encompasses a “top-down” approach to identifying environmental factors
that determine susceptibility to disease throughout the entire lifespan. In parallel, a “bottom-up”
approach can investigate biomarkers specific for certain environmental exposures, based on knowledge
of environmental carcinogens and their pathways of metabolic activation. As part of this approach,
protein adductomics constitute the untargeted investigation of modification of proteins by endogenous
or exogenous agents.
2. Approaches to Protein Adductomics
The concept of an adductome (that is, a collection of additional products) implicates two types
of reactant: Those that add, and those to which are added. In the context of the present discussion,
these are nucleophilic protein sites (amino acid residues) and electrophilic toxicants, respectively
1
.
Reactants of either type are potentially diverse, meaning that the adductome could be vast. From an
analytical standpoint, this potential vastness (i.e., structural diversity) is problematic because of the
lack of a common ‘handle’ or ‘signature’ by which to purify and identify the adducts. Accordingly,
investigators have focused on adducts of either specific nucleophiles or specific electrophiles. If the
investigator’s aim is to discover biomarkers of exposure, a nucleophile is selected and the electrophiles
to which it adds are captured; if the aim is instead to discover targets, an electrophile is selected and
the nucleophiles that add to it are captured. Given that the focus of this review is on biomarkers
of environmental exposure, we will concentrate on the former approach. The latter approach is
also important, however, because it is a route by which novel adducts could be accessed, either
directly [14,15] or indirectly [16].
1
A note regarding language. For the reaction of a nucleophile with an electrophile, the view of the chemist is that the
nucleophile is the active participant, providing electrons for the chemical bond (‘nucleophilic addition’, ‘nucleophilic attack’,
and so on). Toxicologists, on the other hand, tend to speak of the toxicant as active (toxicant ‘binds’ to target), and since the
toxicant is usually an electrophile the roles would seem to switch. This second interpretation is equally logical because the
nucleophilic targets are often endogenous and less mobile (e.g., DNA or protein) and therefore seem to be passive entities.
Toxics 2019,7, 29 3 of 17
Of the methods that capture electrophiles, the most advanced methods are based on haemoglobin
(Hb) and human serum albumin (HSA). There have been a number of important methodological
developments since Rappaport et al. reviewed the subject in 2012 [
17
]. Another, related review [
18
]
was published during the preparation of the present review.
2.1. Hb as A Target of Electrophiles
Hb is found in the erythrocytes, where it functions as an oxygen carrier. Its high concentration
and reactivity (see below) make it a likely target of electrophiles, and its long lifetime
in vivo
(126 days,
the lifetime of an erythrocyte [
19
]) presumably gives the resulting adducts an opportunity to accumulate.
Human Hb A, the major form of Hb in adults, is a tetramer composed of two
α
-chains and two
β
-chains. The four chains, each of which binds one molecule of haem, all adopt similar folds in the
tetramer. The
α
- and
β
-chains have several amino acid residues in common, including the N-terminal
valine residues [
20
]. The
α
-amino groups of these terminal residues are nucleophilic, and have been
observed to react with toxicologically-relevant electrophiles [
21
]. The N-terminal
α
-amino groups of
the α- and β-chains have similar pKavalues and similar reactivity towards certain electrophiles (e.g.,
the acetylating agent acetic anhydride), but not necessarily towards all electrophiles [
22
]. For example,
another acetylating agent, methyl acetyl phosphate, has been observed to modify the N-terminus of
only the β-chain [23].
The
β
-chain of Hb possesses a cysteine residue (Cys-
β
93) for which there is no equivalent in the
α
-chain [
20
]. Adducts of Hb Cys-
β
93 have been the subject of both targeted and, to a lesser extent,
untargeted adductomic analyses (see below). A targeted adductomic method (i.e., a method involving
simultaneous monitoring of multiple known/hypothesised adducts) was used to monitor Hb adducts
of 15 dierent aromatic amines (e.g., 4-aminobiphenyl) in tobacco smokers’ blood [
24
]. These, it should
be pointed out, are not adducts of the amines themselves, but rather of the corresponding arylnitroso
compounds [
25
]. Arylnitroso compounds form via oxidation of the amines’ N-hydroxy metabolites,
in a reaction for which, in the erythrocyte at least, the oxidant is the oxy form of Hb itself. The Cys-
β
93
adducts of arylnitroso compounds are N-arylsulfinamides, which hydrolyse under acidic conditions to
regenerate their corresponding aromatic amines [
26
]. On this basis, detection of the aromatic amines
liberated by acid hydrolysis of N-arylsulfinamides has been used as an indirect way of detecting the
adducts [24].
2.2. The N-alkyl Edman Method
The analytical tractability of Hb N-terminal adducts is due to a general property of N-terminal
amino acid residues, namely their ability to be detached from the rest of the protein via Edman
degradation. This is a procedure that was originally developed for protein sequencing, but which was
modified in the 1980s by Ehrenberg and co-workers for the analysis of Hb N-terminal adducts [
27
].
Ehrenberg and co-workers’ procedure has been referred to as the ‘N-alkyl Edman method’ because of
its ability to detect, for example, N
α
-methyl and N
α
-ethyl substituents [
28
,
29
]. In fact, the observed
N
α
-substituents have not been limited to simple alkyl groups, but for convenience the modified
N-terminal amino acid is referred to as N-alkylvaline. Edman’s original procedure involved
reacting the
α
-amino group of a peptide with phenyl isothiocyanate, which rendered an acid-labile
product [
30
]. Treatment of this product with anhydrous acid liberates the terminal amino acid as an
anilinothiazolinone, which is then isomerised in aqueous acid to a phenylthiohydantoin (PTH) [
31
].
Ehrenberg and co-workers found that Hb with N-terminal N-alkylvaline (i.e., a secondary amine)
reacted with isothiocyanate reagents in the same way as unmodified Hb, but that the resulting
derivatives were labile even under neutral conditions [
27
]. The final product, a substituted PTH,
could therefore be isolated using conditions under which unmodified Hb remained intact.
In subsequent iterations of the N-alkyl Edman method, the isothiocyanate reagent was varied so as
to generate analytes appropriate for particular analytical methods. The most recent iteration, the ‘FIRE
procedure’, uses fluorescein isothiocyanate (‘FIRE’ being a contraction of ‘
f
luorescein
i
sothiocyanate’,
Toxics 2019,7, 29 4 of 17
R
-group’ and ‘
E
dman degradation’) [
32
]. The FIRE procedure was initially developed with targeted
analysis in mind, but was later adapted for untargeted analyses (‘FIRE screening procedure’ [28]).
2.3. The Role of Tandem Mass Spectrometry in Protein Adductomics
Like most other adductomic methodologies, the FIRE screening procedure utilises tandem mass
spectrometry (MS/MS) for the detection of adducts. MS/MS, as its name suggests, involves two stages
of mass analysis. The first stage is for intact precursor ions (e.g., protonated molecules) and the second
stage is for product ions (i.e., fragments of precursor ions). A process of fragmentation takes place
in between the two stages. Mass analysis can be performed in either a static mode, whereby ions of
specified mass-to-charge ratio (m/z) are isolated, or a dynamic mode, whereby a continuous range of
m/zvalues is scanned. Either stage can be performed in either mode, meaning that a number of dierent
types of experiment are possible. In selected reaction monitoring (SRM), a technique commonly used
for targeted analyses, ions of pre-specified m/zare isolated at both stages. Isolation is achieved by
defining a narrow window of permissible m/zvalues and is often done using a quadrupole mass filter.
An apparatus commonly used for SRM is the triple quadrupole mass spectrometer, which consists
of two quadrupole mass filters, with a collision cell between them, connected in series. The first and
second stages of mass analysis take place in the first and second filters, respectively, with fragmentation
taking place in the collision cell. Other MS/MS techniques of relevance to this review are precursor ion
scanning, data-dependent acquisition (DDA) and data-independent acquisition (DIA). These will be
covered in more detail in the sections concerning HSA adductomics.
2.4. Stepped MS/MS Methods
Several adductomic studies have employed stepped methods, which can be thought of as hybrids
of SRM and scanning. A stepped method consists of a sequence of SRM experiments that collectively
resemble a scan. In considering how the methods work, it is instructive to think of adducts’ structures in
terms of two distinct parts: A constant part that derives from the nucleophile (common to all precursor
ions) and a variable part that derives from the electrophile (variable among precursor ions). It follows,
therefore, that a given product ion (or neutral fragment) will be either constant or variable depending on
how the precursor ion becomes broken up into fragments. Given that the variable parts of the precursor
ions are unlikely to be known a priori, the constituent SRM experiments of a stepped method must be
necessarily arbitrary. For this reason, it is common to see lists of equally-spaced integer or half-integer
m/zvalues [
28
]. We have referred to these arbitrary values as sampling points [
33
]. The idea of an
arbitrary SRM experiment might strike the reader as odd, since SRM is traditionally used for targeted
analyses, but for untargeted analyses it does not matter where the sampling points fall. The important
thing is that, collectively, they are able to capture all relevant adducts. The limitation of stepped
methods is their low resolution, which means that they are unable to identify adducts unambiguously
purely on the basis of mass. Their value, therefore, tends to be in providing a quantitative description
of the distribution of adducts.
2.5. The FIRE Screening Procedure
The FIRE screening procedure [
28
] is a method for untargeted detection of Hb adducts (Figure 1).
It is a stepped method akin to the ‘adductome approach to detect DNA damage’ developed by
Kanaly et al. [
34
]. In the FIRE screening procedure, dierent precursor ions (protonated fluorescein
thiohydantoins, FTHs) are captured at the first stage of mass analysis via one of 136 dierent windows.
Each window is approximately 0.7 m/zunits wide, and the m/zvalues on which the windows are
centred are 1 Da apart. Thus, by cycling through all 136 windows, the method can capture a wide range
of precursor ions and can, therefore, detect the corresponding range of mass shifts (between +14 and
+149 Da). Once captured, a precursor ion is fragmented, and its products are passed to the second stage
of mass analysis. Here, a set of fixed windows permit only constant product ions to pass to the detector
(implicates loss of variable neutral fragments), and a variable window permits only variable product
Toxics 2019,7, 29 5 of 17
ions to pass (implicates loss of constant neutral fragments). If the right combination of constant and
variable product ions is detected, then the presence of a corresponding FTH,
and therefore
Hb adduct,
can be inferred. This MS/MS is done ‘online’ following the chromatographic separation of the FTHs,
and the data thus generated are, like those reported by Kanaly et al., visualised as an ‘adductome map’,
usually a plot of m/zagainst retention time [28,34,35].
Toxics 2019, 7, x FOR PEER REVIEW 5 of 17
Figure 1. Main steps of the FIRE (‘fluorescein isothiocyanate’, ‘R-group’ and ‘Edman degradation’)
screening procedure for Hb N-terminal adductomics. The procedure detects ‘R’ groups, which are
generated when an N-terminus of Hb reacts with an electrophile in vivo. The N-termini are
derivatised with fluorescein isothiocyanate, and derivatives with ‘R’ groups are selectively
decomposed to the corresponding fluorescein thiohydantoins. The thiohydantoins are analysed using
LC and online ‘stepped’ triple quadrupole mass spectrometry.
Carlsson et al. used their procedure to screen the blood of smokers and non-smokers and
detected 26 features of interest; this study is described below in Section 3.
2.6. HSA as A Target of Electrophiles
HSA is the major protein in human plasma. Its lifetime in vivo, whilst shorter than that of Hb, is
presumably still long enough for adducts to accumulate. In vivo, HSA binds fatty acids, scavenges
metal ions, and contributes to the oncotic pressure of blood [22]. Extensive use of HSA has been made
for the biological monitoring of toxicants, and a detailed account of this can be found in the recent
review by Sabbioni and Turesky [36]. For the purposes of the present review, we focus on providing
a background to the untargeted HSA adductomics studies.
Thus far, HSA contains a number of nucleophilic sites, including (but not limited to) histidine
residues, lysine residues and a single reduced cysteine residue (Cys-34). Notably, histidine residues
in HSA, as in Hb, are targets of epoxides [37,38]. Lysine residues in serum albumins are notable
targets of aflatoxin B1 dialdehyde [39,40].
Cys-34 is the only site in HSA for which untargeted adductomic methods have been developed.
The motivation to look at this particular site is related to the unique chemistry of thiol groups, and
the fact that HSA Cys-34 accounts for the majority of such groups in human plasma [41]. Given that
the reacting species is a thiolate anion rather than a thiol group proper [41], adduct formation should
be promoted by alkaline conditions and/or basic groups within the local protein environment. The
pKa of the HSA Cys-34 thiol group is controversial, but is generally regarded to be lower than that of
a typical thiol group [41]. In the three-dimensional structure of HSA, as determined by X-ray
crystallography, the side chain of Cys-34 is partially buried [42]. On this basis, it has been inferred
that there might be a limit to the size of the electrophiles that HSA Cys-34 can add to. It has also been
recognised, however, that the tertiary structure of HSA is dynamic and that Cys-34 may become less
buried upon deprotonation of the thiol group [17,43]. HSA Cys-34 is reactive towards a variety of
toxicologically-relevant electrophiles, including sulphur mustard and metabolites of aromatic amines
Figure 1.
Main steps of the FIRE (‘fluorescein isothiocyanate’, ‘R-group’ and ‘Edman degradation’)
screening procedure for Hb N-terminal adductomics. The procedure detects ‘R’ groups, which are
generated when an N-terminus of Hb reacts with an electrophile
in vivo
. The N-termini are derivatised
with fluorescein isothiocyanate, and derivatives with ‘R’ groups are selectively decomposed to the
corresponding fluorescein thiohydantoins. The thiohydantoins are analysed using LC and online
‘stepped’ triple quadrupole mass spectrometry.
Carlsson et al. used their procedure to screen the blood of smokers and non-smokers and detected
26 features of interest; this study is described below in Section 3.
2.6. HSA as A Target of Electrophiles
HSA is the major protein in human plasma. Its lifetime
in vivo
, whilst shorter than that of Hb, is
presumably still long enough for adducts to accumulate.
In vivo
, HSA binds fatty acids, scavenges
metal ions, and contributes to the oncotic pressure of blood [
22
]. Extensive use of HSA has been made
for the biological monitoring of toxicants, and a detailed account of this can be found in the recent
review by Sabbioni and Turesky [
36
]. For the purposes of the present review, we focus on providing a
background to the untargeted HSA adductomics studies.
Thus far, HSA contains a number of nucleophilic sites, including (but not limited to) histidine
residues, lysine residues and a single reduced cysteine residue (Cys-34). Notably, histidine residues in
HSA, as in Hb, are targets of epoxides [
37
,
38
]. Lysine residues in serum albumins are notable targets of
aflatoxin B1dialdehyde [39,40].
Cys-34 is the only site in HSA for which untargeted adductomic methods have been developed.
The motivation to look at this particular site is related to the unique chemistry of thiol groups,
and the
fact
that HSA Cys-34 accounts for the majority of such groups in human plasma [
41
]. Given that the reacting
species is a thiolate anion rather than a thiol group proper [
41
], adduct formation should be promoted
by alkaline conditions and/or basic groups within the local protein environment. The pK
a
of the HSA
Cys-34 thiol group is controversial, but is generally regarded to be lower than that of a typical thiol
group [
41
]. In the three-dimensional structure of HSA, as determined by X-ray crystallography, the side
Toxics 2019,7, 29 6 of 17
chain of Cys-34 is partially buried [
42
]. On this basis, it has been inferred that there might be a limit to
the size of the electrophiles that HSA Cys-34 can add to. It has also been recognised, however, that the
tertiary structure of HSA is dynamic and that Cys-34 may become less buried upon deprotonation of the
thiol group [
17
,
43
]. HSA Cys-34 is reactive towards a variety of toxicologically-relevant electrophiles,
including sulphur mustard and metabolites of aromatic amines [
44
,
45
], and can also undergo oxidative
transformations [
46
,
47
]. It appears that,
in vivo
,
a substantial
proportion of the HSA Cys-34 thiol
groups is S-thiolated (S-[cystein-S-yl], S-[glutathion-S-yl] and so on), and a smaller, but appreciable
proportion is found as the corresponding sulfenic, sulfinic or sulfonic acids [41,47].
2.7. HSA Cys-34 Adductomics
To date, methods for HSA Cys-34 adductomics have been based exclusively on peptide analytes
(Figure 2). When HSA is digested with trypsin, and no cleavages are missed, Cys-34 and its adducts
are found in a 21-amino-acid peptide [
48
,
49
]. This peptide, which Rappaport’s group has referred
to as ‘T3’ (i.e., the third-heaviest tryptic peptide [
49
]), has been used as an analyte in a number of
studies [
49
51
]. When a combination of trypsin and chymotrypsin is used, the Cys-34-containing
peptide is instead the LQQCPF hexapeptide [
43
]. The use of Pronase, suggested by Sabbioni and
Turesky as a means of generating lower-molecular-weight analytes, has not to our knowledge been
implemented for untargeted HSA Cys-34 adductomics [
36
]. When Noort et al. [
44
,
52
] used Pronase to
digest HSA adducts of either sulphur mustard or acrylamide, the respective modifications were found
in the CPF tripeptide.
Figure 2.
Main steps of published HSA Cys-34 adductomic workflows. The reaction of HSA with an
electrophile in the blood plasma installs an ‘R’ group at the Cys-34 site. The HSA is isolated from
plasma or serum and digested—usually with trypsin—to produce a mixture of peptides. Some of
the peptides contain ‘R’ groups and others do not (the introduction of an enrichment step prior to
digestion can limit the number of those that do not). Peptides are then separated chromatographically
and analysed using MS/MS. One of the MS/MS methods, a stepped triple-quadrupole method termed
FS-SRM, is depicted. This method monitors three variable product ions of the tryptic ‘T3’ peptide
(y15,y16 and y17 ).
Some of the first untargeted HSA adductomic analyses were performed by Aldini et al. using the
technique of precursor ion scanning [
43
] (see also the ‘chemical modificomics’ method proposed by
Goto et al. [
53
]). Precursor ion scanning is an MS/MS technique involving a scan at the first stage of
mass analysis and the isolation of a constant product ion at the second stage. The result is a spectrum
of the dierent precursor ions that give rise to a given product. Aldini et al. [
43
] reacted purified HSA
with a mixture of
α
,
β
-unsaturated aldehydes (4-hydroxy-2-nonenal, 4-hydroxy-2-hexenal and acrolein),
and digested the products with trypsin and chymotrypsin. Analysis of the digestion products, using
liquid chromatography (LC) and online precursor ion scanning, revealed peaks corresponding to
Toxics 2019,7, 29 7 of 17
substituted LQQCPF peptides. These, in turn, corresponded to HSA Cys-34 Michael adducts of the
α,β-unsaturated aldehyde reactants.
2.8. Fixed-Step SRM of HSA Adducts
An important development, reported by Li et al. in 2011, was the demonstration of a stepped
method called fixed-step SRM (FS-SRM [
49
]). FS-SRM consists of a sequence of SRM experiments that
collectively resemble a linked scan [
54
]. In developing the method, Li et al. drew on elements of the
‘adductome approach to detect DNA damage’ described by Kanaly et al. [
34
,
55
], and also a method
of analysing mercapturic acids described by Wagner et al. [
56
]. Being a stepped method, FS-SRM
is broadly analogous to the FIRE screening procedure (which, in fact, it pre-dates). The analytes in
FS-SRM are substituted T3 peptides, and the precursor ions captured in the first stage of mass analysis
are triply-protonated peptides. The product ions isolated in the second stage are doubly-charged
variable y-ions and a singly-charged constant b-ion. Together, these precursor and product ions
constitute what is eectively a peptide sequence tag [
57
]. The sampling points used for FS-SRM are
4.5 Da apart and, in the Li and co-workers’ study, there were 77 of them. FS-SRM diers from the other
stepped methods in that, for FS-SRM, the sample is infused into the mass spectrometer as a mixture of
adducts rather than as a series of eluted components. There is still an LC step but it is disconnected
from the mass spectrometry, and it serves to capture the entire population of adducts rather than to
separate them. The method is therefore freed from a major constraint imposed by LC, namely the need
for a full set of SRM experiments to be done within the width of a chromatographic peak.
Our personal experience with protein adductomics has been in the implementation of FS-SRM for
epidemiological studies [
10
,
33
]. Such studies, which typically involve tens or hundreds of samples,
pose challenges that are not necessarily encountered in smaller pilot studies. In implementing the
method of Li et al., the main challenge that we faced was the need for higher throughput. This was
addressed by evaluating the various stages of sample preparation (HSA purification, adduct enrichment,
digestion and peptide clean-up) and optimising these where possible. Notably, we deleted the adduct
enrichment step, and we changed the method of sample clean-up from HPLC (serial) to solid-phase
extraction (SPE; eectively parallel). A model adduct, prepared by treating HSA with N-ethylmaleimide,
proved useful for evaluating the performance of the methods.
In parallel with our work on FS-SRM, Grigoryan et al. [
50
] developed a new analytical workflow
based on LC with on-line DDA mass spectrometry. In DDA, the data on which the acquisition is
dependent are precursor ions’ m/zvalues, and they are obtained via a high-resolution scan—using,
for example, an Orbitrap mass analyser. The data are used to direct the isolation of precursor ions,
and so only these precursor ions are fragmented. The acquisition is the scan via which the resulting
product ions are detected. In addition to their analytical method, Grigoryan et al. [
50
] also developed
methods for sample preparation and data analysis (the ‘adductomics pipeline’). The method of sample
preparation is essentially a streamlined version of the one developed by Li et al. [
49
]. One major
dierence with respect to the earlier method, however, was the omission of a reducing agent, which
had previously been used to reduce protein disulphide bonds prior to tryptic digestion. The eect
of omitting the reducing agent was to preserve S-thiolated forms of Cys-34. The method of data
analysis begins with the detection of a tag (a combination of constant and variable product ions)
in the product-ion scan data. The corresponding precursor ion is then identified, and an ion count
chromatogram for this precursor ion is extracted. A particularly innovative part of the pipeline is
the method by which the peptide analytes are quantified. Each analyte is quantified relative to a
‘housekeeping peptide’, which is another tryptic peptide of HSA. In this way, the method is able to
control for variation in the quantity of digested HSA. Grigoryan et al. [
50
] used their pipeline to analyse
samples of plasma from smokers and non-smokers, and found a total of 43 putative adducts (see
Section 3below).
Toxics 2019,7, 29 8 of 17
2.9. Multiplex Adduct Peptide Profiling
Another promising method for HSA Cys-34 adductomics (and potentially also Hb Cys-
β
93
adductomics) is ‘multiplex adduct peptide profiling’ (MAPP [
51
]). MAPP utilises DIA mass
spectrometry, which is perhaps the least prescriptive of all MS/MS techniques. Similar to a stepped
SRM-based method, DIA captures precursor ions via a series of contiguous windows. The windows
are, however, rather wider than those used for SRM, and it is therefore likely that a given window will
capture multiple precursor ions (in MAPP, for example, the width of each window is 10 m/zunits).
As in DDA mass spectrometry, the second stage of mass analysis is a scan, and a high-resolution scan
is done as an alternative first stage.
The MAPP method, like the ‘adductomics pipeline’, requires prior knowledge of the peptide
analyte’s sequence and the site of modification. Series of constant product ions (e.g., b-ions from
backbone scission near the N-terminus) are recognised and are linked back to their respective precursor
ions via common chromatographic retention times. The substituted peptide’s mass shift is then
confirmed by the presence of corresponding variable product ions. Although the authors were only
able to identify oxidised and S-thiolated forms of HSA Cys-34, their method has the potential to detect
toxicologically-relevant adducts (e.g., if the samples could be further enriched for these adducts prior
to analysis).
2.10. Hb and HSA Compared
Given that Hb and HSA contain some of the same nucleophilic functional groups, these proteins
might be expected to have overlapping reactivity towards electrophiles. The observation that cysteine
residues in HSA and Hb can add to comparable amounts of benzene oxide
in vivo
, for example,
is evidence of such overlap [
58
]. On the other hand, Dingley et al. [
59
] found that dietary exposure
to 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP; see Section 3.3) caused the formation of
substantially larger amounts of HSA adducts than Hb adducts. A similar fate has been observed
for aflatoxin B
1
in rats: of a given dose of this toxicant, a substantially higher proportion is found
bound to serum albumin than to Hb [
60
,
61
]. This might also be expected to be the case in humans,
and indeed assays for HSA adducts of aflatoxin B
1
dialdehyde have been developed [
62
]. Possible
reasons for dierences in the amount or type of adducts include (i) the fact that Hb and HSA are
synthesised at dierent sites in the body (in dierent cell types), and as a result could be exposed
to dierent electrophiles [
36
]; (ii) the fact that Hb resides inside the erythrocyte, whereas HSA is
secreted [
18
]; (iii) the influence of neighbouring amino acid side chains and cofactors on the reactivity
of the nucleophilic groups (see Sections 2.1 and 2.6); and (iv) the possibility that the erythrocyte
membrane could shield Hb from electrophiles, or even sequester electrophiles [
63
]. It is also worth
considering that apparent dierences in the extent of adduct formation could reflect dierences in
chemical and biological stability of the proteins and/or modifications.
2.11. Other Target Proteins
Few proteins other than Hb and HSA have been discussed as candidates for untargeted adductomic
analyses, and fewer still have been investigated experimentally. Hb and HSA adducts are probably
two of the richest and most accessible sources of potential biomarkers, but this is not to say that other
proteins could not provide additional and unique information. Three other proteins of relevance to the
present review have been discussed: Collagen, histones and apolipoproteins. Collagen is mentioned
by Scheepers in his workshop report [
19
], presumably because of its abundance in the body and its
extremely long lifespan in certain tissues [
64
]. However, there have been few attempts to use collagen
adducts for biological monitoring, probably because of the heterogeneity, physical properties and
limited accessibility of collagen [
65
67
]. Histones, which are also mentioned by Scheepers, represent
a more promising source of biomarkers. Work on histone adducts has not been extensive, but some
interesting results have been obtained. N-Terminal segments of histones are of particular interest
Toxics 2019,7, 29 9 of 17
because they protrude from nucleosomal core particles, and, on this basis, it is plausible that they could
be accessible to electrophiles. Consistent with this idea, SooHoo et al. [
68
] observed modifications
near the N-termini of histones isolated from cultured human lymphoblasts that had been exposed
to anti-benzo[a]pyrene 7,8-dihydrodiol-9,10-oxide (BPDE). Fabrizi et al. [
69
] used a model peptide to
infer the reactivity of an N-terminal segment of histone H2B towards phosgene, and observed the
incorporation of carbonyl groups into the peptide.
Apolipoproteins have been investigated as targets of endogenous electrophiles, such as the lipid
oxidation product 4-hydroxy-2-nonenal. By definition, endogenous adducts cannot be biomarkers
of exposure in the strict sense, but they could potentially be biomarkers of eect. We mention them
here because they have been the subject of a recent untargeted adductomics study. This study focused
on adducts of histidine and lysine residues in human low density lipoprotein [
35
]. Unlike the FIRE
screening procedure or FS-SRM, the method is not site-specific; rather, it detects modifications to any
and all residues of particular amino acid. The analytes are ‘free’ amino acids, which are prepared from
lipoprotein by acid hydrolysis. Consequently, they may represent a mixture of sites, and perhaps a
mixture of proteins. The analytical method, like others described elsewhere in this article, involves
ultraperformance LC and triple quadrupole mass spectrometry. Apparently it is a stepped method,
in which a constant product ion is isolated at the second stage of mass analysis. For adducts of histidine
residues, the constant product is the immonium ion of histidine, and for adducts of lysine residues,
it is a deaminated immonium ion of lysine. Shibata et al. [
35
] used their method to analyse low density
lipoprotein that had been first purified from human plasma, and then oxidised
in vitro
. The oxidised
lipoprotein was treated with sodium borohydride to reduce imine linkages (as in, for example, a lysine
residue adducts of 9-oxononanoic acid), before being hydrolysed and the resulting amino acids
analysed. The authors produced adductome maps for lipoprotein with and without oxidation, and by
comparing these maps they were able to attribute the formation of the aforementioned 9-oxononanoic
acid adduct to the oxidising condition.
2.12. Adduct Enrichment
Enrichment, in the context of untargeted adductomics, entails depletion of the unmodified
nucleophile and possibly also other substances that might interfere with the detection of the adducts.
In the FIRE screening procedure for Hb adducts, enrichment is facilitated by the detachment of the
N-alkylvaline residues. This exaggerates the relatively minor dierence in structure between Hb
and its adducts, thereby allowing the unmodified Hb to be removed readily [
28
]. For HSA Cys-34
adductomics, methods of enrichment have mainly exploited the reactivity of the Cys-34 thiol group,
which is present in the unmodified HSA but not in the adducts. Funk et al. [
70
] demonstrated the
use of a disulfide-functionalised resin for scavenging unmodified HSA, and this method was later
used in adductomic workflows [
49
,
51
,
71
]. The main limitation of the thiol scavenging method is that
it does not remove S-thiolated HSA: If a reducing agent is later added to reduce the other disulfide
bonds in HSA (i.e., those of the cystine residues) then the S-thiolation is reversed and the Cys-34 thiol
would seem to reappear. Funk et al. [
70
] sought to limit this eect by removing the S-thiolation prior to
the scavenging step. In our hands, the thiol scavenging method proved dicult to implement in a
high-throughput setting, and so we deleted it from our workflow [
33
]. Chung et al. [
71
] used thiol
scavenging as the first of two stages of enrichment, the second stage being an antibody-mediated
purification of the substituted T3 peptides using a polyclonal antibody raised against the T3 peptide
but having cross-reactivity with adducts.
3. Human Biomonitoring
3.1. Methodological Considerations
Human biomonitoring refers to the quantification of xenobiotics or their derivatives (and sometimes
their early eects) in human biospecimens [
72
]. As well as confirming the nature of the exposure,
Toxics 2019,7, 29 10 of 17
biomonitoring aims to measure the internal dose of the xenobiotic(s). The biomonitoring of protein
adducts is usually done as part of the ‘bottom up’ (targeted) approach (see Section 1). A typical targeted
method might involve isotope dilution (i.e., the addition of a known amount of an isotopically-labelled
standard) followed by LC-MS/MS. This would require prior characterisation of the adduct and synthesis
of a suitable standard.
In principle, data collected via the untargeted approach (e.g., peak areas from LC-MS/MS) could
be used in the same way as those collected in targeted studies. However, this would depend on the
untargeted method achieving an acceptable accuracy, precision and dynamic range for each relevant
adduct. At some stage, a synthetic reference compound would be needed to confirm a particular
adduct’s identity, and to implicate the corresponding electrophile [
18
]. For hitherto unknown adducts,
possible identities must first be proposed. Methods that have assisted in this endeavour have included
database searching, the use of calculator software, and the comparison of measured and predicted
physicochemical properties [
50
,
73
]. The characterisation of novel adducts—a challenging aspect of the
research—has been reviewed in detail by Carlsson et al. [18].
Accuracy, in practice, may suer as a consequence of the need to capture a range of adducts.
It is likely that the use of generic standards (e.g., the S-carbamidomethylated T3 peptide for FS-SRM)
aects accuracy, and therefore precludes absolute quantification [
33
]. Dynamic ranges are dependent
on the analytical method, and presumably also on the ability to enrich adducts. As judged from
lowest reported adduct concentrations, the detection limits of Grigoryan and co-workers’ LC-MS-based
method, and of the FIRE screening procedure, are good (<7 and <0.1 adduct molecules per million
HSA molecules or Hb chains, respectively [
28
,
50
]). The methods should, therefore, be able to detect
some xenobiotic adducts, although in practice relatively few such adducts have been observed [
50
].
For FS-SRM (our implementation), the detection and quantification limits are in the region of one adduct
molecule per thousand HSA molecules, and are probably too high to detect xenobiotic adducts [
33
].
Putative adducts detected by FS-SRM and the other methods may, however, relate to the early eects
of exposure.
At the present time, the role of the untargeted methods is to complement the targeted methods,
rather than to replace them. Indeed, approaches that combine both methods have been proposed [
7
].
Some authors advocate a more pragmatic ‘fit-for-purpose’ approach, which balances methodological
rigour with cost. Dennis et al. draw a distinction between regulatory endeavours, which require
maximal rigour, and exploratory studies whose aims might be achievable without a fully validated
method [7].
While the discipline of untargeted protein adductomics is still a relatively young one, there have
been a number of pilot studies that have sought to demonstrate its utility. Additionally, some targeted
investigations have looked for adduct formation at the same sites (e.g., Cys-34 of HSA) and these will
also be mentioned here.
3.2. Human Biomonitoring of Hb Adducts
In the first adductomic application of the FIRE method (see Section 2.5), Hb samples from smokers
and non-smokers were analysed and compared [
28
]. In all samples seven adducts at the N-terminal
valine residue were identified; these were the addition of methyl and ethyl groups, and adducts
formed by ethylene oxide, acrylonitrile, methyl vinyl ketone, acrylamide and glycidamide; in addition,
a further 19 unknown adducts were detected in all samples. Subsequently, one of these unknown
adducts has been identified as derived from ethyl methyl ketone [
74
]. A further four have been
attributed to the precursor electrophiles glyoxal, methylglyoxal, acrylic acid and 1-octen-3-one [
73
];
and recently another adduct, detected in smokers and non-smokers at similar levels, has been identified
as N-(4-hydroxybenzyl)valine, postulated to have arisen from either 4-quinone methide, which could
form the valine adduct via a Michael addition, or 4-hydroxybenzaldehyde, which could form the same
adduct via a Schibase formation followed by reduction [75].
Toxics 2019,7, 29 11 of 17
Applying their untargeted Hb adductomic approach to a larger study population, Carlsson et
al. [
76
] analysed blood samples from healthy children about 12 years old (n=51). In this cohort,
a total of 24 adducts (12 of them previously identified; see above) were observed and their levels
quantified. Relatively large interindividual variations in adduct levels were observed. The frequencies
of micronuclei in erythrocytes were also determined. Analysis using a partial least-squares regression
model showed that as much as 60% of the micronucleus variation could be explained by the adduct
levels. This indicates the ability of such studies to align measurements of internal dose (protein
adducts) with endpoints of genotoxicity (micronucleus formation).
3.3. Human Biomonitoring of HSA Adducts
An early study that demonstrated the utility of monitoring HSA for alkylated cysteine involved
exposure of human blood to
14
C-labelled sulfur mustard (the chemical warfare agent mustard gas) [
44
].
Isolation and tryptic digestion of albumin produced the 21-amino acid fragment containing a sulfur
mustard-cysteine adduct, detected by micro-LC-MS/MS. An alternative method, which employed
Pronase for the digestion, yielded a modified tripeptide (Cys-Pro-Phe), which was detected with
greater sensitivity than the 21-amino acid fragment. The method was used to analyse samples of
blood from nine Iranians exposed to sulfur mustard during the Iran-Iraq war of 1986. In all nine cases,
the sulfur mustard-adducted tripeptide was detected.
Application of the FS-SRM method to analyses of archived plasma protein that had been pooled
according to subjects’ ethnicities and tobacco smoking habits demonstrated dierences between
pools [
49
] and suggested that FS-SRM might be able to detect statistically significant dierences
between groups of individual samples that had not been pooled.
A pilot study of 20 smokers and 20 never-smokers provided evidence of the eect of smoking
on levels of putative HSA adducts. Dierences between smokers and never-smokers were most
apparent in putative adducts with net gains in mass between 105 Da and 114 Da (relative to unmodified
HSA) [33].
Further investigations of the eects of tobacco smoking have revealed around 43 adduct features,
some of which are positively associated with smoking and but also some that are negatively associated.
The former result from genotoxic constituents of tobacco smoke, such as ethylene oxide and acrylonitrile,
while the latter, which include Cys-34 oxidation products and disulfides, may reflect alterations in the
serum redox state of smokers, resulting in lower adduct levels [50].
Grigoryan et al. used LC and high-resolution mass spectrometry to investigate interactions
between the Cys-34 and reactive oxygen species (ROS) [
47
]. Chronic exposure to ROS is linked to
many chronic diseases and, in this study, a number of adducts originating from ROS were detected in
human serum: Sulfinic acid, sulfonic acid and a proposed sulfinamide structure (a mono-oxygenated
moiety also with the loss of two hydrogen atoms).
Antibody enrichment may pave the way to a more sensitive assay. Using a polyclonal antibody,
raised against the T3 peptide, but with cross-reactivity to the peptide containing adducts (see
Section 2.12), ten modified T3 peptides were detected in human plasma samples; eight of them were
characterised and they included Cys-34 oxidation products, modification involving loss of water or
lysine, cysteinylation, and transpeptidation of arginine [68].
In a study of women from the Xuanwei and Fuyuan counties in China, where extensive use of
smoky coal for heating and cooking has resulted in very high rates of lung cancer among non-smokers,
HSA Cys-34 adducts were compared in 29 females who used smoky coal and 10 controls using other
energy sources [
77
]. Fifty dierent modified T3 peptides were identified, including oxidation products,
mixed disulfides, rearrangements and truncations. Two peptides that were detected at significantly
lower levels in the smoky coal group were adducts of glutathione and
γ
-glutamylcysteine. The results
are interpreted as evidence that exposure to the indoor combustion products results in depletion of
glutathione, an essential antioxidant, as well as its precursor γ-glutamylcysteine [77].
Toxics 2019,7, 29 12 of 17
A recent study on the health eects of urban air pollution, the Oxford Street II study [
78
], involved a
randomised crossover design whereby three groups of volunteers (healthy subjects, chronic obstructive
pulmonary disease (COPD) suerers and patients with ischaemic heart disease (IHD)) walked for two
hours along a busy street in London where trac is restricted to diesel buses and taxis. The volunteers
also spent two hours walking in a London park on a separate occasion. They were monitored for
respiratory and cardiovascular function in both environments and, in addition, two studies have
analysed their HSA samples for adducts. In the first report, Liu et al. [
79
] analysed 50 HSA samples
by high-resolution mass spectrometry to determine whether protein modifications dier between
COPD or IHD patients and healthy subjects. The untargeted analysis of adducts at the Cys-34 locus
of HSA detected 39 adducts with sucient data, and these adducts were examined for associations
with estimated exposures to air pollution and health status. Multivariate linear regression revealed
21 significant
associations, mainly with the underlying diseases, but also with air-pollution exposures.
Interestingly, most of the associations indicated that adduct levels decreased with the presence of
disease or increased pollutant concentrations. Negative associations of COPD and IHD with the Cys-34
disulfide of glutathione and two Cys-34 sulfoxidations were consistent with results from smokers and
non-smokers [
50
] and from non-smoking women exposed to indoor combustion of coal and wood [
74
].
In the second study, Preston et al. [
80
] examined a larger number of Oxford Street II samples by
the FS-SRM method. Associations between amounts of putative adducts and two types of measure
were tested: Pollution (e.g., ambient concentrations of nitrogen dioxide and particulate matter) and
health outcome (e.g., measures of lung health and arterial stiness). There were 11 instances of a
response variable being associated with a pollution measurement and eight instances of a response
variable being associated with a health outcome measure. However, no two measures of dierent
types were associated with the same adduct amount, suggesting that the internal changes responsible
for health outcomes may dier from those that eect changes in adduct amounts.
In a more targeted study, Bellamri et al. [
81
] investigated the formation in human subjects of HSA
adducts at Cys-34 by PhIP, which is formed in cooked meats and may be associated with colorectal,
prostate and mammary cancer. Volunteers abstained from eating cooked well-done meat or fish for
three weeks, then ate a semi-controlled diet that included cooked beef containing known quantities of
PhIP for four weeks. The volunteers then returned to their regular diets, but with the exclusion of
cooked well-done meat and fish for a further four weeks. The authors found that an adduct of oxidised
PhIP, which was below the limit of detection (LOD) (10 femtograms PhIP/mg HSA) in most subjects
before the meat feeding, increased by up to 560-fold at week 4 in subjects who ate meat containing 8.0
to 11.7
µ
g of PhIP per 150–200 g serving. In contrast, the adduct remained below the LOD in subjects
who ingested 1.2 or 3.0
µ
g PhIP per serving, and PhIP-HSA adduct levels did not correlate with PhIP
intake levels across four exposure groups (p=0.76). There were also indications that the PhIP adduct
was unstable, having a half-life of fewer than two weeks. Nevertheless, the study demonstrates that
the Cys-34 site in HSA is accessible by a relatively large molecule like PhIP, despite concerns about
possible steric hindrance (see Section 2.6.).
4. Prospects
A key advantage of monitoring proteins for adducts is the abundance of material that can be
obtained from tissue banks; for example, red blood cells are an abundant source of Hb and blood
plasma or serum is an abundant source of HSA. The proteins’ lifespans in blood mean that there
is a substantial “capture period” for monitoring exposure to genotoxicants; and protein adducts,
unlike DNA adducts, are not subject to loss through repair processes. Full implementation of the
exposome concept requires monitoring individuals or populations at several points in time over the
course of their lives [
3
,
4
]. This is achievable if biobanks collect material from individuals not just once
but multiple times, and such biobanks already exist.
Dried blood spots can also be a suitable source of protein for investigation [
82
]. If obtained
from neonatal blood spots (i.e., Guthrie spots) then the material provides a valuable opportunity for
Toxics 2019,7, 29 13 of 17
investigating exposures in utero. A single blood spot of about 50
µ
L is estimated to contain about
9.6 mg of protein, of which about 7.7 mg will be Hb and 1.2 mg HSA [
79
]. In a proof-of-principle study,
Yano et al. [
83
] identified 26 Cys-34 adducts (oxidation and S-thiolation products) in HSA isolated from
dried blood spots of 49 newborn babies and were able to distinguish between newborns of smoking
and non-smoking mothers on the basis of the levels of a putative cyano modification to Cys-34.
There is also the potential to broaden the scope of adductomics by investigating novel modifiable
loci in blood proteins. Cys-34 of HSA and the N-terminus of Hb are undoubtedly major targets for
electrophiles, but the literature hints at wider reactivity within the blood proteome. Consequently,
a hitherto-untapped source of analytes for protein adductomics can be envisaged. Mapping the loci at
which adducts can form will be beneficial and, for this purpose, new chemical tools will be required.
Identifying adducts detected by the top-down approaches will be a challenge, necessitating chemical
synthesis of candidate structures for unequivocal characterisation.
Protein adductomics is a component of the exposome concept that is still relatively novel, but it
is one that has already demonstrated the ability to capture electrophiles of both endogenous and
exogenous origin; this suggests the potential to contribute meaningfully to the aims of the exposome
concept—to describe the totality of all biologically relevant exposures. Rapid advances in mass
spectrometry instrumentation, with significant increases in sensitivity and resolution, will drive further
advances in protein adductomics methodology. When coupled with other omics approaches, such as
proteomics, transcriptomics and metabolomics, all of which have the potential for high-throughput
screening of populations, a future can be envisaged in which it will be possible to capture snapshots
of human exposure to genotoxicants and the resultant biological consequences at multiple stages
throughout life. Building this comprehensive picture should shed significant light on the causes and
courses of chronic diseases in humans. Such knowledge will provide new opportunities for early
intervention to reduce potentially harmful human exposure, to monitor the eectiveness of intervention
strategies and, ultimately, to prevent diseases before they occur.
Author Contributions:
Both authors critically reviewed the literature and contributed equally to writing
the manuscript.
Funding:
The authors’ research was funded by the European Community’s Seventh Framework Programme
(FP7/2007-2013) under grant agreement number 308610 (the EXPOsOMICS project). Additional funding was from
Cancer Research UK (Programme Grant CRUK/A14329) and the MRC-PHE Centre for Environment and Health
(MRC grant number G0801056/1).
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Rappaport, S.M. Genetic Factors Are Not the Major Causes of Chronic Diseases. PLoS ONE
2016
,11, e0154387.
[CrossRef] [PubMed]
2.
Rappaport, S.M.; Smith, M.T. Epidemiology. Environment and disease risks. Science
2010
,330, 460–461.
[CrossRef] [PubMed]
3.
Wild, C.P. Complementing the genome with an “exposome”: The outstanding challenge of environmental
exposure measurement in molecular epidemiology. Cancer Epidemiol. Biomark. Prev.
2005
,14, 1847–1850.
[CrossRef] [PubMed]
4. Wild, C.P. The exposome: From concept to utility. Int. J. Epidemiol. 2012,41, 24–32. [CrossRef] [PubMed]
5.
Rappaport, S.M.; Barupal, D.K.; Wishart, D.; Vineis, P.; Scalbert, A. The blood exposome and its role in
discovering causes of disease. Environ. Health Perspect. 2014,122, 769–774. [CrossRef]
6. Rappaport, S.M. Biomarkers intersect with the exposome. Biomarkers 2012,17, 483–489. [CrossRef]
7.
Dennis, K.K.; Marder, E.; Balshaw, D.M.; Cui, Y.; Lynes, M.A.; Patti, G.J.; Rappaport, S.M.; Shaughnessy, D.T.;
Vrijheid, M.; Barr, D.B. Biomonitoring in the Era of the Exposome. Environ. Health Perspect.
2017
,125, 502–510.
[CrossRef]
8.
Stewart, B.W.; Bray, F.; Forman, D.; Ohgaki, H.; Straif, K.; Ullrich, A.; Wild, C.P. Cancer prevention as part of
precision medicine: ‘Plenty to be done’. Carcinogenesis 2016,37, 2–9. [CrossRef] [PubMed]
Toxics 2019,7, 29 14 of 17
9.
Wild, C.P.; Scalbert, A.; Herceg, Z. Measuring the exposome: A powerful basis for evaluating environmental
exposures and cancer risk. Environ. Mol. Mutagen. 2013,54, 480–499. [CrossRef] [PubMed]
10.
Vineis, P.; Chadeau-Hyam, M.; Gmuender, H.; Gulliver, J.; Herceg, Z.; Kleinjans, J.; Kogevinas, M.;
Kyrtopoulos, S.; Nieuwenhuijsen, M.; Phillips, D.H.; et al. The exposome in practice: Design of the
EXPOsOMICS project. Int. J. Hyg. Environ Health 2017,220, 142–151. [CrossRef]
11.
Vrijheid, M.; Slama, R.; Robinson, O.; Chatzi, L.; Coen, M.; van den Hazel, P.; Thomsen, C.; Wright, J.;
Athersuch, T.J.; Avellana, N.; et al. The human early-life exposome (HELIX): Project rationale and design.
Environ. Health Perspect. 2014,122, 535–544. [CrossRef]
12.
Ankley, G.T.; Bennett, R.S.; Erickson, R.J.; Ho, D.J.; Hornung, M.W.; Johnson, R.D.; Mount, D.R.; Nichols, J.W.;
Russom, C.L.; Schmieder, P.K.; et al. Adverse outcome pathways: A conceptual framework to support
ecotoxicology research and risk assessment. Environ. Toxicol. Chem.
2010
,29, 730–741. [CrossRef] [PubMed]
13.
Escher, B.I.; Hackermüller, J.; Polte, T.; Scholz, S.; Aigner, A.; Altenburger, R.; Böhme, A.; Bopp, S.K.; Brack, W.;
Busch, W.; et al. From the exposome to mechanistic understanding of chemical-induced adverse eects.
Environ. Int. 2017,99, 97–106. [CrossRef] [PubMed]
14.
Dennehy, M.K.; Richards, K.A.M.; Wernke, G.R.; Shyr, Y.; Liebler, D.C. Cytosolic and Nuclear Protein Targets
of Thiol-Reactive Electrophiles. Chem. Res. Toxicol. 2006,19, 20–29. [CrossRef] [PubMed]
15.
Weerapana, E.; Simon, G.M.; Cravatt, B.F. Disparate proteome reactivity profiles of carbon electrophiles.
Nat. Chem. Biol. 2008,4, 405–407. [CrossRef]
16.
Medina-Cleghorn, D.; Bateman, L.A.; Ford, B.; Heslin, A.; Fisher, K.J.; Dalvie, E.D.; Nomura, D.K. Mapping
Proteome-Wide Targets of Environmental Chemicals Using Reactivity-Based Chemoproteomic Platforms.
Chem. Biol. 2015,22, 1394–1405. [CrossRef] [PubMed]
17. Rappaport, S.M.; Li, H.; Grigoryan, H.; Funk, W.E.; Williams, E.R. Adductomics: Characterizing exposures
to reactive electrophiles. Toxicol. Lett. 2012,213, 83–90. [CrossRef]
18.
Carlsson, H.; Rappaport, S.M.; Tornqvist, M. Protein Adductomics: Methodologies for Untargeted Screening
of Adducts to Serum Albumin and Hemoglobin in Human Blood Samples. High Throughput
2019
,8, 6.
[CrossRef] [PubMed]
19.
Scheepers, P.T.J. The use of biomarkers for improved retrospective exposure assessment in epidemiological
studies: Summary of an ECETOC workshop. Biomarkers 2008,13, 734–748. [CrossRef]
20. Dickerson, R.E.; Geis, I. Hemoglobin; Benjamin Cummings: Menlo Park, CA, USA, 1983.
21.
Rubino, F.M.; Pitton, M.; Di Fabio, D.; Colombi, A. Toward an “omic” physiopathology of reactive chemicals:
Thirty years of mass spectrometric study of the protein adducts with endogenous and xenobiotic compounds.
Mass Spectrom. Rev. 2009,28, 725–784. [CrossRef] [PubMed]
22.
Kaplan, A.; Jack, R.; Opheim, K.E.; Toivola, B.; Lyon, A.W. Clinical Chemistry; Williams & Wilkins: Malvern,
UK, 1995.
23.
Ueno, H.; Pospischil, M.A.; Manning, J.M.; Kluger, R. Site-specific modification of hemoglobin by methyl
acetyl phosphate. Arch. Biochem. Biophys. 1986,244, 795–800. [CrossRef]
24.
Bryant, M.S.; Vineis, P.; Skipper, P.L.; Tannenbaum, S.R. Hemoglobin adducts of aromatic amines: Associations
with smoking status and type of tobacco. Proc. Natl. Acad. Sci. USA 1988,85, 9788. [CrossRef] [PubMed]
25.
Turesky, R.J.; Le Marchand, L. Metabolism and Biomarkers of Heterocyclic Aromatic Amines in Molecular
Epidemiology Studies: Lessons Learned from Aromatic Amines. Chem. Res. Toxicol.
2011
,24, 1169–1214.
[CrossRef] [PubMed]
26.
Ringe, D.; Turesky, R.J.; Skipper, P.L.; Tannenbaum, S.R. Structure of the single stable hemoglobin adduct
formed by 4-aminobiphenyl in vivo. Chem. Res. Toxicol. 1988,1, 22–24. [CrossRef] [PubMed]
27.
Törnqvist, M.; Mowrer, J.; Jensen, S.; Ehrenberg, L. Monitoring of environmental cancer initiators through
hemoglobin adducts by a modified Edman degradation method. Anal. Biochem.
1986
,154, 255–266.
[CrossRef]
28.
Carlsson, H.; von Stedingk, H.; Nilsson, U.; Tornqvist, M. LC-MS/MS screening strategy for unknown
adducts to N-terminal valine in hemoglobin applied to smokers and nonsmokers. Chem. Res. Toxicol. 2014,
27, 2062–2070. [CrossRef] [PubMed]
29.
Tornqvist, M. Epoxide adducts to N-terminal valine of hemoglobin. Methods Enzymol.
1994
,231, 650–657.
[CrossRef]
30.
Edman, P. Method for Determination of the Amino Acid Sequence in Peptides. Acta Chem. Scand.
1950
,4,
283–293. [CrossRef]
Toxics 2019,7, 29 15 of 17
31.
Price, N.C.; Stevens, L. Fundamentals of Enzymology, 3rd ed.; Oxford University Press: New York, NY,
USA, 1999.
32.
Von Stedingk, H.; Rydberg, P.; Törnqvist, M. A new modified Edman procedure for analysis of N-terminal
valine adducts in hemoglobin by LC–MS/MS. J. Chromatogr. B 2010,878, 2483–2490. [CrossRef]
33.
Preston, G.W.; Plusquin, M.; Sozeri, O.; van Veldhoven, K.; Bastian, L.; Nawrot, T.S.; Chadeau-Hyam, M.;
Phillips, D.H. Refinement of a Methodology for Untargeted Detection of Serum Albumin Adducts in Human
Populations. Chem. Res. Toxicol. 2017,30, 2120–2129. [CrossRef]
34.
Kanaly, R.A.; 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, 993–1001. [CrossRef]
35.
Shibata, T.; Shimizu, K.; Hirano, K.; Nakashima, F.; Kikuchi, R.; Matsushita, T.; Uchida, K. Adductome-based
identification of biomarkers for lipid peroxidation. J. Biol. Chem. 2017,292, 8223–8235. [CrossRef]
36.
Sabbioni, G.; Turesky, R.J. Biomonitoring Human Albumin Adducts: The Past, the Present, and the Future.
Chem. Res. Toxicol. 2017,30, 332–366. [CrossRef] [PubMed]
37.
Day, B.W.; Skipper, P.L.; Zaia, J.; Tannenbaum, S.R. Benzo[a]pyrene anti-diol epoxide covalently modifies
human serum albumin carboxylate side chains and imidazole side chain of histidine(146). J. Am. Chem. Soc.
1991,113, 8505–8509. [CrossRef]
38.
Lindh, C.H.; Kristiansson, M.H.; Berg-Andersson, U.A.; Cohen, A.S. Characterization of adducts
formed between human serum albumin and the butadiene metabolite epoxybutanediol. Rapid Commun.
Mass Spectrom. 2005,19, 2488–2496. [CrossRef]
39.
Sabbioni, G. Chemical and physical properties of the major serum albumin adduct of aflatoxin B
1
and their
implications for the quantification in biological samples. Chemico-Biol. Interact. 1990,75, 1–15. [CrossRef]
40.
Guengerich, F.P.; Arneson, K.O.; Williams, K.M.; Deng, Z.; Harris, T.M. Reaction of Aflatoxin B
1
Oxidation
Products with Lysine. Chem. Res. Toxicol. 2002,15, 780–792. [CrossRef]
41.
Turell, L.; Radi, R.; Alvarez, B. The thiol pool in human plasma: The central contribution of albumin to redox
processes. Free Radic. Biol. Med. 2013,65, 244–253. [CrossRef]
42.
He, X.M.; Carter, D.C. Atomic structure and chemistry of human serum albumin. Nature
1992
,358, 209–215.
[CrossRef]
43.
Aldini, G.; Regazzoni, L.; Orioli, M.; Rimoldi, I.; Facino, R.M.; Carini, M. A tandem MS precursor-ion scan
approach to identify variable covalent modification of albumin Cys34: A new tool for studying vascular
carbonylation. J. Mass Spectrom. 2008,43, 1470–1481. [CrossRef]
44.
Noort, D.; Hulst, A.G.; de Jong, L.P.; Benschop, H.P. Alkylation of human serum albumin by sulfur mustard
in vitro and in vivo: Mass spectrometric analysis of a cysteine adduct as a sensitive biomarker of exposure.
Chem. Res. Toxicol. 1999,12, 715–721. [CrossRef] [PubMed]
45.
Peng, L.; Dasari, S.; Tabb, D.L.; Turesky, R.J. Mapping Serum Albumin Adducts of the Food-Borne Carcinogen
2-Amino-1-methyl-6-phenylimidazo[4,5-b]pyridine by Data-Dependent Tandem Mass Spectrometry.
Chem. Res. Toxicol. 2012,25, 2179–2193. [CrossRef] [PubMed]
46.
Pathak, K.V.; Bellamri, M.; Wang, Y.; Langouët, S.; Turesky, R.J. 2-Amino-9H-pyrido[2,3-b]indole (A
α
C)
Adducts and Thiol Oxidation of Serum Albumin as Potential Biomarkers of Tobacco Smoke. J. Biol. Chem.
2015,290, 16304–16318. [CrossRef]
47.
Grigoryan, H.; Li, H.; Iavarone, A.T.; Williams, E.R.; Rappaport, S.M. Cys34 adducts of reactive oxygen
species in human serum albumin. Chem. Res. Toxicol. 2012,25, 1633–1642. [CrossRef]
48.
Dong, Q.; Yan, X.; Kilpatrick, L.E.; Liang, Y.; Mirokhin, Y.A.; Roth, J.S.; Rudnick, P.A.; Stein, S.E. Tandem Mass
Spectral Libraries of Peptides in Digests of Individual Proteins: Human Serum Albumin (HSA). Mol. Cell.
Proteom. 2014,13, 2435. [CrossRef]
49.
Li, H.; Grigoryan, H.; Funk, W.E.; Lu, S.S.; Rose, S.; Williams, E.R.; Rappaport, S.M. Profiling Cys34 adducts
of human serum albumin by fixed-step selected reaction monitoring. Mol. Cell. Proteom. MCP
2011
,
10, M110.004606. [CrossRef]
50.
Grigoryan, H.; Edmands, W.; Lu, S.S.; Yano, Y.; Regazzoni, L.; Iavarone, A.T.; Williams, E.R.; Rappaport, S.M.
Adductomics Pipeline for Untargeted Analysis of Modifications to Cys34 of Human Serum Albumin.
Anal. Chem. 2016,88, 10504–10512. [CrossRef]
51.
Porter, C.J.; Bereman, M.S. Data-independent-acquisition mass spectrometry for identification of
targeted-peptide site-specific modifications. Anal. Bioanal. Chem. 2015,407, 6627–6635. [CrossRef]
Toxics 2019,7, 29 16 of 17
52.
Noort, D.; Fidder, A.; Hulst, A.G. Modification of human serum albumin by acrylamide at cysteine-34:
A basis for a rapid biomonitoring procedure. Arch. Toxicol. 2003,77, 543–545. [CrossRef]
53.
Goto, T.; Kojima, S.; Shitamichi, S.; Lee, S.H.; Oe, T. Chemical modificomics: A novel strategy for ecient
biomarker discovery through chemical modifications on a target peptide. Anal. Methods
2012
,4, 1945–1952.
[CrossRef]
54.
Todd, J.F.J. Recommendations for Nomenclature and Symbolism for Mass Spectroscopy. Pure Appl. Chem.
1991,63, 1541–1566. [CrossRef]
55.
Kanaly, R.A.; 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, 83–93. [CrossRef] [PubMed]
56.
Wagner, S.; Scholz, K.; Donegan, M.; Burton, L.; Wingate, J.; Völkel, W. Metabonomics and Biomarker
Discovery: LC
MS Metabolic Profiling and Constant Neutral Loss Scanning Combined with Multivariate
Data Analysis for Mercapturic Acid Analysis. Anal. Chem. 2006,78, 1296–1305. [CrossRef]
57.
Mann, M.; Wilm, M. Error-Tolerant Identification of Peptides in Sequence Databases by Peptide Sequence
Tags. Anal. Chem. 1994,66, 4390–4399. [CrossRef]
58.
Yeowell-O’Connell, K.; Rothman, N.; Smith, M.T.; Hayes, R.B.; Li, G.; Waidyanatha, S.; Dosemeci, M.;
Zhang, L.; Yin, S.; Titenko-Holland, N.; et al. Hemoglobin and albumin adducts of benzene oxide among
workers exposed to high levels of benzene. Carcinogenesis 1998,19, 1565–1571. [CrossRef] [PubMed]
59.
Dingley, K.H.; Curtis, K.D.; Nowell, S.; Felton, J.S.; Lang, N.P.; Turteltaub, K.W. DNA and protein
adduct formation in the colon and blood of humans after exposure to a dietary-relevant dose of
2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine. Cancer Epidemiol. Biomark. Prev. 1999,8, 507–512.
60.
Tannenbaum, S.R.; Skipper, P.L. Biological aspects to the evaluation of risk: dosimetry of carcinogens in man.
Fundam. Appl. Toxicol. 1984,4, S367–S373. [CrossRef]
61.
Sabbioni, G.; Skipper, P.L.; Buchi, G.; Tannenbaum, S.R. Isolation and characterization of the major serum
albumin adduct formed by aflatoxin B1in vivo in rats. Carcinogenesis 1987,8, 819–824. [CrossRef]
62.
Wild, C.P.; Jiang, Y.Z.; Sabbioni, G.; Chapot, B.; Montesano, R. Evaluation of methods for quantitation
of aflatoxin-albumin adducts and their application to human exposure assessment. Cancer Res.
1990
,50,
245–251.
63.
Noort, D.; Fidder, A.; Degenhardt-Langelaan, C.E.; Hulst, A.G. Retrospective detection of sulfur mustard
exposure by mass spectrometric analysis of adducts to albumin and hemoglobin: An
in vivo
study. J. Anal.
Toxicol. 2008,32, 25–30. [CrossRef]
64.
Verzijl, N.; DeGroot, J.; Thorpe, S.R.; Bank, R.A.; Shaw, J.N.; Lyons, T.J.; Bijlsma, J.W.J.; Lafeber, F.P.J.G.;
Baynes, J.W.; TeKoppele, J.M. Eect of Collagen Turnover on the Accumulation of Advanced Glycation End
Products. J. Biol. Chem. 2000,275, 39027–39031. [CrossRef] [PubMed]
65.
Jonsson, B.A.G.; Wishnok, J.S.; Skipper, P.L.; Stillwell, W.G.; Tannenbaum, S.R. Lysine Adducts Between
Methyltetrahydrophthalic Anhydride and Collagen in Guinea Pig Lung. Toxicol. Appl. Pharmacol.
1995
,135,
156–162. [CrossRef]
66. Miller, E.J.; Gay, S. Collagen: An overview. Methods Enzymol. 1982,82, 3–32. [CrossRef] [PubMed]
67.
Miller, E.J.; Kent Rhodes, R. Preparation and characterization of the dierent types of collagen.
Methods Enzymol. 1982,82, 33–64. [CrossRef] [PubMed]
68.
SooHoo, C.K.; Singh, K.; Skipper, P.L.; Tannenbaum, S.R.; Dasari, R.R. Characterization of benzo[a]pyrene
anti-diol epoxide adducts to human histones. Chem. Res. Toxicol. 1994,7, 134–138. [CrossRef]
69.
Fabrizi, L.; Taylor, G.W.; Cañas, B.; Boobis, A.R.; Edwards, R.J. Adduction of the Chloroform Metabolite
Phosgene to Lysine Residues of Human Histone H2B. Chem. Res. Toxicol.
2003
,16, 266–275. [CrossRef]
[PubMed]
70.
Funk, W.E.; Li, H.; Iavarone, A.T.; Williams, E.R.; Riby, J.; Rappaport, S.M. Enrichment of cysteinyl adducts
of human serum albumin. Anal. Biochem. 2010,400, 61–68. [CrossRef] [PubMed]
71.
Chung, M.K.; Grigoryan, H.; Iavarone, A.T.; Rappaport, S.M. Antibody enrichment and mass spectrometry
of albumin-Cys34 adducts. Chem. Res. Toxicol. 2014,27, 400–407. [CrossRef]
72.
Angerer, J.; Ewers, U.; Wilhelm, M. Human biomonitoring: State of the art. Int. J. Hyg. Environ Health
2007
,
210, 201–228. [CrossRef]
73.
Carlsson, H.; Tornqvist, M. Strategy for identifying unknown hemoglobin adducts using adductome
LC-MS/MS data: Identification of adducts corresponding to acrylic acid, glyoxal, methylglyoxal,
and 1-octen-3-one. Food Chem. Toxicol. 2016,92, 94–103. [CrossRef]
Toxics 2019,7, 29 17 of 17
74.
Carlsson, H.; Motwani, H.V.; Osterman Golkar, S.; Tornqvist, M. Characterization of a Hemoglobin Adduct
from Ethyl Vinyl Ketone Detected in Human Blood Samples. Chem. Res. Toxicol.
2015
,28, 2120–2129.
[CrossRef] [PubMed]
75.
Degner, A.; Carlsson, H.; Karlsson, I.; Eriksson, J.; Pujari, S.S.; Tretyakova, N.Y.; Törnqvist, M. Discovery
of Novel N-(4-Hydroxybenzyl)valine Hemoglobin Adducts in Human Blood. Chem. Res. Toxicol.
2018
,31,
1305–1314. [CrossRef] [PubMed]
76.
Carlsson, H.; Aasa, J.; Kotova, N.; Vare, D.; Sousa, P.F.M.; Rydberg, P.; Abramsson-Zetterberg, L.; Tornqvist, M.
Adductomic Screening of Hemoglobin Adducts and Monitoring of Micronuclei in School-Age Children.
Chem. Res. Toxicol. 2017,30, 1157–1167. [CrossRef]
77.
Lu, S.S.; Grigoryan, H.; Edmands, W.M.; Hu, W.; Iavarone, A.T.; Hubbard, A.; Rothman, N.; Vermeulen, R.;
Lan, Q.; Rappaport, S.M. Profiling the Serum Albumin Cys34 Adductome of Solid Fuel Users in Xuanwei
and Fuyuan, China. Environ. Sci. Technol. 2017,51, 46–57. [CrossRef] [PubMed]
78.
Sinharay, R.; Gong, J.; Barratt, B.; Ohman-Strickland, P.; Ernst, S.; Kelly, F.J.; Zhang, J.J.; Collins, P.; Cullinan, P.;
Chung, K.F. Respiratory and cardiovascular responses to walking down a trac-polluted road compared
with walking in a trac-free area in participants aged 60 years and older with chronic lung or heart disease
and age-matched healthy controls: A randomised, crossover study. Lancet
2018
,391, 339–349. [CrossRef]
[PubMed]
79.
Liu, S.; Grigoryan, H.; Edmands, W.M.B.; Dagnino, S.; Sinharay, R.; Cullinan, P.; Collins, P.; Chung, K.F.;
Barratt, B.; Kelly, F.J.; et al. Cys34 Adductomes Dier between Patients with Chronic Lung or Heart Disease
and Healthy Controls in Central London. Environ. Sci. Technol. 2018,52, 2307–2313. [CrossRef]
80.
Preston, G.W.; Dagnino, S.; Ponzi, E.; Sozeri, O.; van Veldhoven, K.; Barratt, B.; Liu, S.; Grigoryan, H.; Lu, S.S.;
Rappaport, S.; et al. Relationships between airborne pollutants, serum albumin adducts and short-term
health outcomes in an experimental crossover study. Chemosphere 2019, submitted.
81.
Bellamri, M.; Wang, Y.; Yonemori, K.; White, K.K.; Wilkens, L.R.; Le Marchand, L.; Turesky, R.J. Biomonitoring
an albumin adduct of the cooked meat carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine in
humans. Carcinogenesis 2018,39, 1455–1462. [CrossRef]
82.
Funk, W.E.; Waidyanatha, S.; Chaing, S.H.; Rappaport, S.M. Hemoglobin adducts of benzene oxide in
neonatal and adult dried blood spots. Cancer Epidemiol. Biomark. Prev. 2008,17, 1896–1901. [CrossRef]
83.
Yano, Y.; Grigoryan, H.; Schiman, C.; Edmands, W.; Petrick, L.; Hall, K.; Whitehead, T.; Metayer, C.;
Dudoit, S.; Rappaport, S. Untargeted adductomics of Cys34 modifications to human serum albumin in
newborn dried blood spots. Anal. Bioanal. Chem. 2019. [CrossRef]
©
2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... Biomonitoring of reactive metabolites regardless of their origin, whether exogenous or endogenous, is challenging due to their short-life in vivo and adductomics provided unique opportunity to detect covalent adducts that are relatively stable and long-lived. Adductomics studies broadly uses two approaches (1) targeted and (2) untargeted; former method focuses on the detection of specific adducts upon exposure to a specific chemical agents, and later category aims to characterization of total adducts through covalent bonding [2] [3]. ...
... Recent studies suggest that exposure to environmental toxicants plays crucial role in pathogenesis and progression of chronic inflammatory diseases and non-communicable diseases (neurological disorders, auto immune diseases, lung diseases, cardiovascular complications cancers and various cancers), and one of key mechanism is found to be the formation of adducts. Chronic diseases are the product of genetic factors (G) and exposures (E) and their associated interactions (GxE) [22] [3]. According to data of the World Health Organization (WHO) it is estimated that nearly half of the mortality is result of exposure to toxic chemicals, which includes particulate air pollution (comprising indoor air pollution and occupational exposure-14%), active and passive smoking (13%), Increased levels of sodium in plasma (6%), and consumption of alcohol (5%) [23]. ...
Article
Full-text available
One of the utmost frequently emerging neurodegenerative diseases, Parkinson's disease (PD), comprehend the forfeit of dopamine (DA)-generating nerve cells in the substantia nigra pars compacta (SN-PC). The etiology and pathogenesis underlying the emergence of PD is still obscure. However, expanding corroboration encourages the involvement of genetic and environmental factors in the etiology of PD. The destruction of numerous cellular components, namely oxidative stress, ubiquitin-proteasome system (UPS) dysfunction, autophagy-lysosome system dysfunction, neuroinflammation and programmed cell death, and mitochondrial dysfunction partake in the pathogenesis of PD. Present-day pharmacotherapy can alleviate the manifestations, but no therapy has been demonstrated to cease disease progression. Peroxisome proliferator-activated receptors (PPARs) are ligand-directed transcription factors pertaining to the class of nuclear hormone receptors (NHR), and are implicated in the modulation of mitochondrial operation, inflammation, wound healing, redox equilibrium, and metabolism of blood sugar and lipids. Numerous PPAR agonists have been recognized to safeguard nerve cells from oxidative destruction, inflammation, and programmed cell death in PD and other neurodegenerative diseases. Additionally, various investigations suggest that regular administration of PPAR-activating non-steroidal anti-inflammatory drugs (NSAIDs) (ibuprofen, indomethacin), and leukotriene receptor antagonists (montelukast) were related to the de-escalated evolution of neurodegenerative diseases. The present review elucidates the emerging evidence enlightening the neuroprotective outcomes of PPAR agonists in in vivo and in vitro models experiencing PD. Existing articles up to the present were procured through PubMed, MEDLINE, etc., utilizing specific keywords spotlighted in this review. Furthermore, the authors aim to provide insight into the neuroprotective actions of PPAR agonists by outlining the pharmacological mechanism. As a conclusion, PPAR agonists exhibit neuroprotection through modulating the expression of a group of genes implicated in cellular survival pathways, and may be a propitious target in the therapy of incapacitating neurodegenerative diseases like PD.
... Biomonitoring of reactive metabolites regardless of their origin, whether exogenous or endogenous, is challenging due to their short-life in vivo and adductomics provided unique opportunity to detect covalent adducts that are relatively stable and long-lived. Adductomics studies broadly uses two approaches (1) targeted and (2) untargeted; former method focuses on the detection of specific adducts upon exposure to a specific chemical agents, and later category aims to characterization of total adducts through covalent bonding [2] [3]. ...
... Recent studies suggest that exposure to environmental toxicants plays crucial role in pathogenesis and progression of chronic inflammatory diseases and non-communicable diseases (neurological disorders, auto immune diseases, lung diseases, cardiovascular complications cancers and various cancers), and one of key mechanism is found to be the formation of adducts. Chronic diseases are the product of genetic factors (G) and exposures (E) and their associated interactions (GxE) [22] [3]. According to data of the World Health Organization (WHO) it is estimated that nearly half of the mortality is result of exposure to toxic chemicals, which includes particulate air pollution (comprising indoor air pollution and occupational exposure-14%), active and passive smoking (13%), Increased levels of sodium in plasma (6%), and consumption of alcohol (5%) [23]. ...
Article
Full-text available
Adductomics novel and emerging discipline in the toxicological research emphasizes on adducts formed by reactive chemical agents with biological molecules in living organisms. Development in analytical methods propelled the application and utility of adductomics in interdisciplinary sciences. This review endeavors to add a new dimension where comprehensive insights into di-verse applications of adductomics in addressing some of society’s pressing challenges are pro-vided. Also focuses on diverse applications of adductomics include: forecasting risk of chronic diseases triggered by reactive agents and predicting carcinogenesis induced by tobacco smoking; assessing chemical agents’ toxicity and supplementing genotoxicity studies; designing personal-ized medication and precision treatment in cancer chemotherapy; appraising environmental quality or extent of pollution using biological systems; crafting tools and techniques for diagnosis of diseases and detecting food contaminants; furnishing exposure profile of the individual to electrophiles; and assisting regulatory agencies in risk assessment of reactive chemical agents. Characterizing adducts that are present in extremely low concentrations is an exigent task and more over absence of dedicated database to identify adducts is further exacerbating the problem of adduct diagnosis. In addition, there is scope of improvement in sample preparation methods and data processing software and algorithms for accurate assessment of adducts.
... The principle of adductomics involves the quantification of covalent adducts bound to tissues, thereby typifying the electrophilic characteristics of a drug or its reactive metabolites [175]. Throughout everyday life humans are exposed to reactive physiochemical agents with both endogenous and exogenous origins [176]. ...
Article
Full-text available
Definition of the relationship between drug protein adduct formation (haptenation) and development of immunological adverse drug reactions (drug hypersensitivity) has been an area of active research for over 80 years. The hapten hypothesis which states that “immunogenicity of low molecular weight organic chemicals is dependent on modification of self-proteins,” evolved from Landsteiner and Jacob’s discovery of a correlation between the reactivity of dinitro-halogenated benzenes and their sensitization potential. The hypothesis rapidly evolved to encompass drugs that often require metabolic activation to generate electrophilic, protein-reactive intermediates. As tissue culture methods advanced, the importance of drug hapten-specific T-cells in the disease pathogenesis was defined. This led to a plethora of studies describing the uptake and processing of drug(metabolite) protein adducts by antigen presenting cells, and the subsequent surface display of hapten-modified peptides in the context of MHC molecules. Although the pathway of hapten-specific T-cell activation is now well established, several questions need to be addressed: first, what is the nature of the hapten-modified peptides displayed by MHC? Second, how many of these peptides stimulate T-cells?; third, what are the critical protein modifications involved in T-cell activation; and finally, what is the role of hapten-specific T-cells in the iatrogenic disease? These questions will become increasingly important as more and more targeted covalent binding inhibitor drugs are approved for human use. In this review, we provide a brief synopsis of hapten research and then describe the approaches used by Pharma and academia to study hapten covalent binding and the role of drug protein adducts in the activation of human T-cells.
... Protein adductomics is useful in gaining insights into the mechanisms of action and toxicological effects of these reactive molecules on living organisms. Recent advances in MS and MS-based proteomics have advanced the analysis of protein adducts (Carlsson et al., 2019;Preston & Phillips, 2019) (Figure 7). However, the study of protein adducts presents several analytical challenges, such as low percentages of formation of adducts with tagless reactive molecules, lack of widely applicable methods for their enrichment, and uncertain incremental masses of the adducts. ...
Article
Full-text available
Chemical proteomics, which involves studying the covalent modifications of proteins by small molecules, has significantly contributed to our understanding of protein function and has become an essential tool in drug discovery. Mass spectrometry (MS) is the primary method for identifying and quantifying protein-small molecule adducts. In this review, we discuss various methods for measuring proteomic reactivity using MS and covalent proteomics probes that engage through reactivity-driven and proximity-driven mechanisms. We highlight the applications of these methods and probes in live-cell measurements, drug target identification and validation, and characterizing protein-small molecule interactions. We conclude the review with current developments and future opportunities in the field, providing our perspectives on analytical considerations for MS-based analysis of the proteomic reactivity landscape.
... Therefore, identification of xenobiotics adducted to key proteins and identification of the sites of adduction within the protein are important to better understand the events underlying diseases and chemically induced adverse reactions. Generally, xenobiotic compounds can react with multiple proteins at multiple sites, and identification and characterization of adducted proteins are complicated and present significant analytical challenges [1,14,15]. ...
Article
Full-text available
Histones are the major proteinaceous components of chromatin in eukaryotic cells and an important part of the epigenome. The broad-spectrum herbicide atrazine (2-chloro-4-[ethylamino]-6-[isopropylamino]-1, 3, 5-triazine) and its metabolites are known to form protein adducts, but the formation of atrazine–histone adducts has not been studied. In this study, a bottom-up proteomics analysis method was optimized and applied to identify histone adduction by atrazine in vitro . Whole histones of calf thymus or human histone H3.3 were incubated with atrazine. After solvent-based protein precipitation, the protein was digested by trypsin/Glu-C and the resulting peptides were analyzed by high-resolution mass spectrometry using an ultra-high-performance liquid chromatograph interfaced with a quadrupole Exactive-Orbitrap mass spectrometer. The resulting tryptic/Glu-C peptide of DTNLCAIHAK from calf thymus histone H3.1 or human histone H3.3 was identified with an accurate mass shift of +179.117 Da in atrazine incubated samples. It is deduced that a chemical group with an elemental composition of C 8 H 13 N 5 (179.1171 Da) from atrazine adducted with calf thymus histone H3.1 or human histone H3.3. It was confirmed by MS/MS analysis that the adduction position was at its cysteine 110 residue. Time- and concentration-dependent assays also confirmed the non-enzymatic covalent modification of histone H3.3 by atrazine in vitro. Thus, the potential exists that atrazine adduction may lead to the alteration of histones that subsequently disturbs their normal function. Graphical abstract
... In the newest studies, LC-MS/MS analyses after trypsin digestion is the method of choice to perform targeted and untargeted analyses (Grigoryan et al. 2016;Preston and Phillips 2019;Yano et al. 2020). However, it seems that applications are not going beyond small studies, since the detection levels of small molecules cannot be matched (Table 3). ...
Article
Full-text available
Chemicals are measured regularly in air, food, the environment, and the workplace. Biomonitoring of chemicals in biological fluids is a tool to determine the individual exposure. Blood protein adducts of xenobiotics are a marker of both exposure and the biologically effective dose. Urinary metabolites and blood metabolites are short term exposure markers. Stable hemoglobin adducts are exposure markers of up to 120 days. Blood protein adducts are formed with many xenobiotics at different sites of the blood proteins. Newer methods apply the techniques developed in the field of proteomics. Larger adducted peptides with 20 amino acids are used for quantitation. Unfortunately, at present the methods do not reach the limits of detection obtained with the methods looking at single amino acid adducts or at chemically cleaved adducts. Therefore, to progress in the field new approaches are needed.
... Chemical modifications provide important information regarding biological events: molecular dosimeters for chemical exposure (e.g., smokers vs. nonsmokers [4]), biomarkers to evaluate cancer risks (e.g., estrogen quinone adduct vs. breast cancer [5]), and triggers for structural and/or activity changes (e.g., modification on a-synuclein vs. aggregation [6]). The screening of chemical modifications on proteins has been performed as chemical modificomics [7] and adductomics [8]. Furthermore, an alternative approach focuses on only N-terminal modifications, both because of the biological significance of the modifications, such as the relationship between modification and half-life/activity [9], and to reduce the complicity of the analysis compared with comprehensive screening. ...
Article
Proteins are continuously exposed to reactive chemical species owing to physiological and chemical stresses, resulting in various chemical modifications such as oxidation, nitration, glycation/glycoxidation, lipidation/lipoxidation, and adduct formation with drugs/chemicals. Hemoglobin (Hb) is the most abundant protein in blood (∼150 mg/mL) with a long half-life (turnover: 126 days). Hb is thus believed to be a major target of reactive chemical species and those modifications reflect biological events. Chemical modifications on Hb have been analyzed as N-terminal valine (Val) adducts by the Edman degradation reaction because Val is the N-terminal amino acid in both the α- and β-subunits of Hb and is exposed on the surface of the protein. However, this strategy is limited to N-terminal alkylated Val and overlooks N-terminal acylated and deaminated Val. Here, we describe a proof-of-concept study to develop an alternative screening approach based on liquid chromatography-tandem mass spectrometry with peptide-specific constant neutral loss/precursor ion scanning to include all types of N-terminal modifications on Hb.
Article
Air pollution consists of complex mixtures of chemicals with serious deleterious health effects from acute and chronic exposure. To help understand the mechanisms by which adverse effects occur, the present work examines the responses of cultured human epidermal keratinocytes to specific chemicals commonly found in woodsmoke. Our earlier findings with liquid smoke flavoring (aqueous extract of charred wood) revealed that such extracts stimulated the expression of genes associated with oxidative stress and proinflammatory response, activated the aryl hydrocarbon receptor, thereby inducing cytochrome P4501A1 activity, and induced cross-linked envelope formation, a lethal event ordinarily occurring during terminal differentiation. The present results showed that furfural produced transcriptional responses resembling those of liquid smoke, cyclohexanedione activated the aryl hydrocarbon receptor, and several chemicals induced envelope formation. Of these, syringol permeabilized the cells to the egress of lactate dehydrogenase at a concentration close to that yielding envelope formation, while furfural induced envelope formation without permeabilization detectable in this way. Furfural (but not syringol) stimulated the incorporation of amines into cell proteins in extracts in the absence of transglutaminase activity. Nevertheless, both chemicals substantially increased the amount of cellular protein incorporated into envelopes and greatly altered the envelope protein profile. Moreover, the proportion of keratin in the envelopes was dramatically increased. These findings are consistent with the chemically induced protein cross-linking in the cells. Elucidating mechanisms by which this phenomenon occurs may help understand how smoke chemicals interact with proteins to elicit cellular responses, interpret bioassays of complex pollutant mixtures, and suggest additional sensitive ways to monitor exposures.
Article
Full-text available
Pesticides are increasingly used in combinations in crop protection, resulting in enhanced toxicities for various organisms. Although protein adductomics is challenging, it remains a powerful bioanalytical tool to check environmental exposure and characterize xenobiotic adducts as putative toxicity biomarkers with high accuracy, facilitated by recent advances in proteomic methodologies and a mass spectrometry high-throughput technique. The present study aims to predict the potential neurotoxicity effect of imidacloprid and λ-cyhalothrin insecticides on human neural cells. Our protocol consisted first of 3D in vitro developing neurospheroids derived from human brain tumors and then treatment by pesticide mixture. Furthermore, we adopted a bottom-up proteomic-based approach using nanoflow ultraperformance liquid chromatography coupled with a high-resolution mass spectrometer for protein-adduct analysis with prediction of altered sites. Two proteins were selected, namely, calcium-calmodulin-dependent protein kinase-II (CaMK2) and annexin-A1 (ANXA1), as key targets endowed with primordial roles. De novo sequencing revealed several adduct formations in the active site of 82-ANXA1 and 228-CaMK2 as a result of neurotoxicity, predicted by the added mass shifts for the structure of electrophilic precursors. To the best of our knowledge, our study is the first to adopt a proteomic-based approach to investigate in depth pesticide molecular interactions and their potential to adduct proteins which play a crucial role in the neurotoxicity mechanism.
Article
Full-text available
The reaction products of electrophiles in vivo can be measured as adducts to the abundant proteins, hemoglobin (Hb), and human serum albumin (HSA), in human blood samples. During the last decade, methods for untargeted screening of such adducts, called “adductomics”, have used liquid chromatography-mass spectrometry to detect large numbers of previously unknown Hb and HSA adducts. This review presents methodologies that were developed and used in our laboratories for Hb and HSA adductomics, respectively. We discuss critical aspects regarding choice of target protein, sample preparation, mass spectrometry, data evaluation, and strategies for identification of detected unknown adducts. With this review we give an overview of these two methodologies used for protein adductomics and the precursor electrophiles that have been elucidated from the adducts.
Article
Full-text available
Background: Long-term exposure to pollution can lead to an increase in the rate of decline of lung function, especially in older individuals and in those with chronic obstructive pulmonary disease (COPD), whereas shorter-term exposure at higher pollution levels has been implicated in causing excess deaths from ischaemic heart disease and exacerbations of COPD. We aimed to assess the effects on respiratory and cardiovascular responses of walking down a busy street with high levels of pollution compared with walking in a traffic-free area with lower pollution levels in older adults. Methods: In this randomised, crossover study, we recruited men and women aged 60 years and older with angiographically proven stable ischaemic heart disease or stage 2 Global initiative for Obstructive Lung Disease (GOLD) COPD who had been clinically stable for 6 months, and age-matched healthy volunteers. Individuals with ischaemic heart disease or COPD were recruited from existing databases or outpatient respiratory and cardiology clinics at the Royal Brompton & Harefield NHS Foundation Trust and age-matched healthy volunteers using advertising and existing databases. All participants had abstained from smoking for at least 12 months and medications were taken as recommended by participants' doctors during the study. Participants were randomly assigned by drawing numbered disks at random from a bag to do a 2 h walk either along a commercial street in London (Oxford Street) or in an urban park (Hyde Park). Baseline measurements of participants were taken before the walk in the hospital laboratory. During each walk session, black carbon, particulate matter (PM) concentrations, ultrafine particles, and nitrogen dioxide (NO2) concentrations were measured. Findings: Between October, 2012, and June, 2014, we screened 135 participants, of whom 40 healthy volunteers, 40 individuals with COPD, and 39 with ischaemic heart disease were recruited. Concentrations of black carbon, NO2, PM10, PM2.5, and ultrafine particles were higher on Oxford Street than in Hyde Park. Participants with COPD reported more cough (odds ratio [OR] 1·95, 95% CI 0·96-3·95; p<0·1), sputum (3·15, 1·39-7·13; p<0·05), shortness of breath (1·86, 0·97-3·57; p<0·1), and wheeze (4·00, 1·52-10·50; p<0·05) after walking down Oxford Street compared with Hyde Park. In all participants, irrespective of their disease status, walking in Hyde Park led to an increase in lung function (forced expiratory volume in the first second [FEV1] and forced vital capacity [FVC]) and a decrease in pulse wave velocity (PWV) and augmentation index up to 26 h after the walk. By contrast, these beneficial responses were attenuated after walking on Oxford Street. In participants with COPD, a reduction in FEV1 and FVC, and an increase in R5-20 were associated with an increase in during-walk exposure to NO2, ultrafine particles and PM2.5, and an increase in PWV and augmentation index with NO2 and ultrafine particles. In healthy volunteers, PWV and augmentation index were associated both with black carbon and ultrafine particles. Interpretation: Short-term exposure to traffic pollution prevents the beneficial cardiopulmonary effects of walking in people with COPD, ischaemic heart disease, and those free from chronic cardiopulmonary diseases. Medication use might reduce the adverse effects of air pollution in individuals with ischaemic heart disease. Policies should aim to control ambient levels of air pollution along busy streets in view of these negative health effects. Funding: British Heart Foundation.
Article
Full-text available
Lipid peroxidation is an endogenous source of aldehydes that give rise to covalent modification of proteins in various pathophysiological states. In the present study, a strategy for the comprehensive detection and comparison of adducts was applied to find a biomarker for lipid peroxidation-modified proteins in vivo. This adductome approach utilized liquid chromatography with electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS) methods designed to detect the specific product ions from positively ionized adducts in a selected reaction monitoring mode. Using this procedure, we comprehensively analyzed lysine and histidine adducts generated in the in vitro oxidized low-density lipoproteins (LDL) and observed a prominent increase in several adducts, including a major lysine adduct. Based on the high resolution ESI-MS of the adduct and on the LC-ESI-MS/MS analysis of the synthetic adduct candidates, the major lysine adduct detected in the oxidized LDL was identified as N?-(8-carboxyoctanyl)lysine (COL). Strikingly, a significantly higher amount of COL was detected in the sera from atherosclerosis-prone mice and from patients with hyperlipidemia compared to the controls. These data not only offer structural insights into protein modification by lipid peroxidation products, but also provide a platform for the discovery of biomarkers for human diseases.
Article
Exposure to air pollution can have both short-term and long-term effects on health. However, the relationships between specific pollutants and their effects can be obscured by characteristics of both the pollution and the exposed population. One way of elucidating the relationships is to link exposures and internal changes at the level of the individual. To this end, we combined personal exposure monitoring (59 individuals, Oxford Street II crossover study) with mass-spectrometry-based analyses of putative serum albumin adducts (fixed-step selected reaction monitoring). We attempted to infer adducts' identities using data from another, higher-resolution mass spectrometry method, and were able to detect a semi-synthetic standard with both methods. A generalised least squares regression method was used to test for associations between amounts of adducts and pollution measures (ambient concentrations of nitrogen dioxide and particulate matter), and between amounts of adducts and short-term health outcomes (measures of lung health and arterial stiffness). Amounts of some putative adducts (e.g., one with a positive mass shift of ∼143 Da) were associated with exposure to pollution (11 associations), and amounts of other adducts were associated with health outcomes (eight associations). Adducts did not appear to provide a link between exposures and short-term health outcomes.
Article
Metabolism of chemicals from the diet, exposures to xenobiotics, the microbiome, and lifestyle factors (e.g., smoking, alcohol intake) produce electrophiles that react with nucleophilic sites in circulating proteins, notably Cys34 of human serum albumin (HSA). To discover potential risk factors resulting from in utero exposures, we are investigating HSA-Cys34 adducts in archived newborn dried blood spots (DBS) that reflect systemic exposures during the last month of gestation. The workflow includes extraction of proteins from DBS, measurement of hemoglobin (Hb) to normalize for blood volume, addition of methanol to enrich HSA by precipitation of Hb and other interfering proteins, digestion with trypsin, and detection of HSA-Cys34 adducts via nanoflow liquid chromatography-high-resolution mass spectrometry. As proof-of-principle, we applied the method to 49 archived DBS collected from newborns whose mothers either actively smoked during pregnancy or were nonsmokers. Twenty-six HSA-Cys34 adducts were detected, including Cys34 oxidation products, mixed disulfides with low molecular weight thiols (e.g., cysteine, homocysteine, glutathione, cysteinylglycine), and other modifications. Data were normalized with a novel method (“scone”) to remove unwanted technical variation arising from HSA digestion, blood volume, DBS age, mass spectrometry analysis, and batch effects. Using an ensemble of linear and nonlinear models, the Cys34 adduct of cyanide was found to consistently discriminate between newborns of smoking and nonsmoking mothers with a mean fold change (smoking/nonsmoking) of 1.31. These results indicate that DBS adductomics is suitable for investigating in utero exposures to reactive chemicals and metabolites that may influence disease risks later in life.
Article
Humans are exposed to a wide range of electrophilic compounds present in our diet, environment, or formed endogenously as part of normal physiological processes. These electrophiles can modify nucleophilic sites of proteins and DNA to form covalent adducts. Recently, powerful untargeted adductomic approaches have been developed for systematic screening of these adducts in human blood. Our earlier untargeted adductomics study detected 19 unknown adducts to N-terminal valine in hemoglobin (Hb) in human blood. The present study describes the characterization of one of these adducts, which corresponds to the addition of a 4-hydroxybenzyl (4-OHBn) group to N-terminal valine in Hb that forms N-(4-hydroxybenzyl) valine (4-OHBn-Val). The adduct structure was determined by comparison of its accurate mass, HPLC retention time, and MS/MS fragmentation to that of authentic standards prepared by chemical synthesis. Average 4-OHBn-Val adduct concentrations in twelve human blood samples were estimated to 380 ± 160 pmol/g Hb. Two possible routes of 4-OHBnVal adduct formation are proposed using two different precursor electrophiles: 4-quinone methide (4-QM) and 4-hydroxybenzaldehyde (4-OHBA). We found that 4-QM reacts rapidly with valine to form the 4-OHBn-Val adduct; however, the quinone methide is unstable under physiological conditions due to hydrolysis. It was shown that 4-OHBA forms reversible Schiff base adducts with valine, which can be stabilized via in vivo reduction generating the 4-OHBn-Val adduct. In addition, trace amounts of isomeric 2-hydroxybenzyl-valine (2-OHBn-Val) adducts were detected in the twelve human blood samples (estimated mean adduct level, 5.0 ± 1.4 pmol/g Hb). Further studies are needed to quantify the contributions from identified possible precursor electrophile(s) to the observed hydroxybenzyl adducts in humans.
Article
2-Amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) is formed in cooked meats and may be linked to dietary associated colorectal, prostate, and mammary cancers. Genotoxic N-oxidized metabolites of PhIP react with the Cys 34 of albumin (Alb) to form a sulfinamide adduct, a biomarker of the biologically effective dose. We examined the kinetics of PhIP-Alb adduct formation in plasma of volunteers on a 4-week semi-controlled diet of cooked meat containing known quantities of PhIP. The adduct was below the limit of detection (LOD) (10 femtograms PhIP/mg Alb) in most subjects before the meat feeding, but increased by up to 560-fold at week four in subjects who ate meat containing 8.0 to 11.7 μg of PhIP per 150 - 200 g serving. In contrast, the adduct remained below the LOD in subjects who ingested 1.2 or 3.0 μg PhIP per serving. Correlations were not seen between PhIP-Alb adduct levels and PhIP intake levels (P = 0.76), the amount of PhIP accrued in hair (P = 0.13), the amounts of N-oxidized urinary metabolites of PhIP (P = 0.66), or caffeine CYP1A2 activity (P = 0.55), a key enzyme involved in the bioactivation of PhIP. The half-life of the PhIP-Alb adduct was less than 2 weeks, signifying that the adduct was not stable. PhIP-Alb adduct formation is direct evidence of bioactivation of PhIP in vivo. However, the PhIP hair biomarker is a longer-lived and more sensitive biomarker to assess exposure to this potential human carcinogen.
Article
Oxidative stress generates reactive species that modify proteins, deplete antioxidant defenses and contribute to chronic obstructive pulmonary disease (COPD) and ischemic heart disease (IHD). To determine whether protein modifications differ between COPD or IHD patients and healthy subjects, we performed untargeted analysis of adducts at the Cys34 locus of human serum albumin (HSA). Biospecimens were obtained from nonsmoking participants from London, U.K., including healthy subjects (n=20) and patients with COPD (n=20) or IHD (n=10). Serum samples were digested with trypsin and analyzed by liquid chromatography-high resolution mass spectrometry. Effects of air pollution on adduct levels were also investigated based on estimated residential exposures to PM2.5, O3 and NO2. For the 39 adducts with sufficient data, levels were essentially identical in blood samples collected from the same subjects on two consecutive days, consistent with the 28-d residence time of HSA. Multivariate linear regression revealed 21 significant associations, mainly with the underlying diseases but also with air-pollution exposures (p-value < 0.05). Interestingly, most of the associations indicated that adduct levels decreased with the presence of disease or increased pollutant concentrations. Negative associations of COPD and IHD with the Cys34 disulfide of glutathione and two Cys34 sulfoxidations, were consistent with previous results from smoking and non-smoking volunteers and nonsmoking women exposed to indoor combustion of coal and wood.
Article
Covalently-modified blood proteins (e.g., serum albumin adducts) are increasingly being viewed as potential biomarkers via which the environmental causes of human diseases may be understood. The notion that some (perhaps many) modifications have yet to be discovered has led to the development of untargeted adductomics methods, which attempt to capture entire populations of adducts. One such method is fixed-step selected reaction monitoring (FS-SRM), which analyses distributions of serum albumin adducts via shifts in the mass of a tryptic peptide [Li et al. (2011) Mol. Cell. Proteomics 10, M110 004606]. Working on the basis that FS-SRM might be able to detect biological variation due to environmental factors, we aimed to scale the methodology for use in an epidemiological setting. Development of sample preparation methods led to a batch workflow with increased throughput and provision for quality control. Challenges posed by technical and biological variation were addressed in the processing and interpretation of the data. A pilot study of 20 smokers and 20 never-smokers provided evidence of an effect of smoking on levels of putative serum albumin adducts. Differences between smokers and never-smokers were most apparent in putative adducts with net gains in mass between 105 Da and 114 Da (relative to unmodified albumin). The findings suggest that our implementation of FS-SRM could be useful for studying other environmental factors with relevance to human health.
Article
Electrophilic compounds/metabolites present in humans, originating from endogenous processes or exogenous exposure, pose a risk for health effects through their reactions with nucleophilic sites in proteins and DNA, forming adducts. Adductomic approaches are developed to screen for adducts to biomacromolecules in vivo by mass spectrometry (MS), with the aim to detect adducts corresponding to unknown exposures from electrophiles. In the present study, adductomic screening was performed using blood samples from healthy children about 12 years old (n=51). The frequencies of micronuclei (MN) in erythrocytes in peripheral blood were monitored as a measure of genotoxic effect/genotoxic exposure. The applied adductomic approach has earlier been reported by us, and is based on analysis of N-terminal valine adducts in hemoglobin (Hb) by liquid chromatography tandem mass spectrometry (LC-MS/MS). High resolution MS was introduced for refined screening of previously unknown N-terminal Hb adducts. Measured adduct levels were compared with MN frequencies using multivariate data analysis. In the 51 individuals a total of 24 adducts (whereof 12 were previously identified) were observed and their levels quantified. Relatively large inter-individual variations in adduct levels were observed. The data analysis (with partial least square regression) showed that as much as 60 % of the MN variation could be explained by the adduct levels. This study, for the first time applies the combination of these sensitive methods to measure internal dose of potentially genotoxic chemicals and genotoxic effect, respectively. The results indicate that this is a valuable approach for characterization of exposure to chemical risk factors for genotoxic effects present in individuals of the general population.