You are on page 1of 13

Mutation Research 424 Ž1999.

83–95

Lipid peroxidation—DNA damage by malondialdehyde


)
Lawrence J. Marnett
A.B. Hancock Jr. Memorial Laboratory for Cancer Research Center in Molecular Toxicology, Vanderbilt Cancer Center, Departments of
Biochemistry and Chemistry, Vanderbilt UniÕersity School of Medicine, NashÕille TN 37232, USA
Accepted 23 July 1998

Abstract

Malondialdehyde is a naturally occurring product of lipid peroxidation and prostaglandin biosynthesis that is mutagenic
and carcinogenic. It reacts with DNA to form adducts to deoxyguanosine and deoxyadenosine. The major adduct to DNA is
a pyrimidopurinone called M 1G. Site-specific mutagenesis experiments indicate that M 1G is mutagenic in bacteria and is
repaired by the nucleotide excision repair pathway. M 1G has been detected in liver, white blood cells, pancreas, and breast
from healthy human beings at levels ranging from 1–120 per 10 8 nucleotides. Several different assays for M 1G have been
described that are based on mass spectrometry, 32 P-postlabeling, or immunochemical techniques. Each technique offers
advantages and disadvantages based on a combination of sensitivity and specificity. Application of each of these techniques
to the analysis of M 1G is reviewed and future needs for improvements are identified. M 1G appears to be a major endogenous
DNA adduct in human beings that may contribute significantly to cancer linked to lifestyle and dietary factors. High
throughput methods for its detection and quantitation will be extremely useful for screening large populations. q 1999
Elsevier Science B.V. All rights reserved.

Keywords: Malondialdehyde; Pyrimidopurinone; M 1G; DNA adduct; Mass spectrometry; Postlabeling; Immunochemistry

1. Lipid peroxidation Some of these react with protein and DNA and as a
result are toxic and mutagenic. The basic reactions of
Lipid peroxidation generates a complex variety of lipid peroxidation are outlined in Scheme 1 w1x.
products, many of which are reactive electrophiles. Polyunsaturated fatty acids contain one or more
methylene groups positioned between cis double
bonds. The methylene groups are highly reactive to
oxidizing agents and their hydrogen atoms are re-
Abbreviations: MDA, malondialdehyde; M 1G, pyrimido-
w1,2 a xpurin-10Ž3 H .-one; M 1 A, N 6 -Ž3-oxo-propenyl.deoxyadeno- moved to form carbon-centered radicals Že.g., 1..
sine; M 1C, N 4-Ž3-oxopropenyl.deoxycytidine; 8-oxodG, 8-oxo- Carbon-centered radicals react with O 2 to form per-
7,8-dihydrodeoxyguanosine; PdG, 1, N 2-propanodeoxyguanosine; oxyl radicals at a rate that is limited only by diffu-
GCrEC NCIrMS, gas chromatographyrelectron capture negative sion. Thus, the initial products of polyunsaturared
chemical ionizationrmass spectrometry; GCrMS, gas chromatog- fatty acid oxidation are peroxyl radicals. The fate of
raphyrmass spectrometry; PFB, pentafluorobenzyl; LCrMS, liq-
uid chromatographyrmass spectrometry
the peroxyl radical depends on its position in the
)
Corresponding author. Tel.: q1-615-343-7329; Fax: q1-615- carbon chain of the fatty acid. If the peroxyl radical
343-7534; E-mail: marnett@toxicology.mc.vanderbilt.edu exists at one of the two ends of the double bond

0027-5107r99r$ - see front matter q 1999 Elsevier Science B.V. All rights reserved.
PII: S 0 0 2 7 - 5 1 0 7 Ž 9 9 . 0 0 0 1 0 - X
84 L.J. Marnettr Mutation Research 424 (1999) 83–95

Scheme 1. Pathways of lipid peroxidation.

system Že.g., 2., it is reduced to a hydroperoxide. another molecule of fatty acid reduces the peroxyl
Conjugated diene hydroperoxides such as 4 are the radical, a new carbon-centered radical is generated
simplest products of lipid peroxidation and are rela- which propagates the fatty acid oxidation. Through
tively stable in the absence of metals. However, this radical chain process, one oxidizing agent can
metal complexes and metalloproteins are abundant in cause many molecules of fatty acid to become oxi-
cells and they rapidly reduce all fatty acid hydro- dized. The actual lengths of the radical chains that
peroxides by one electron to alkoxyl radicals, which occur in cells are sensitive to many factors but in
undergo multiple chemical reactions to generate a pure chemical systems approximately 60 molecules
broad range of products w2x. Thus, even the simplest of linoleic acid and 200 molecules of arachidonic
initial lipid peroxidation products produce a complex acid are consumed per initiation event w3x. The major
range of epoxides, hydroperoxides, and carbonyl determinant of the length of radical chains in vivo is
compounds, inter alia. the concentration of vitamin E in the phospholipid
In phospholipid membranes, the molecules that bilayer w4x. Vitamin E reduces peroxyl radicals which
reduce peroxyl radicals to hydroperoxides are either normally breaks the radical chain and slows the rate
another molecule of fatty acid or vitamin E. If of lipid peroxidation. However, under conditions of
L.J. Marnettr Mutation Research 424 (1999) 83–95 85

very low oxidant, vitamin E has actually been shown radicals that decompose to a range of products.
to initiate another radical chain w5x. Among these are many carbonyl compounds includ-
If the peroxyl radical is at an internal position in ing hexanal, 4-hydroxynonenal, and other saturated
the fatty acid chain Že.g., 3., cyclization to an adja- and a ,b-unsaturated aldehydes and ketones w10x.
cent double bond will compete with reduction to a Many of these compounds react with protein and
hydroperoxide. The cyclization product Ž5. is a cyclic DNA and are toxic and mutagenic. MDA appears to
peroxide adjacent to a carbon-centered radical. This be the most mutagenic product of lipid peroxidation
carbon-centered radical has two fates. The radical whereas 4-hydroxynonenal is the most toxic w11x.
can couple with O 2 to form a peroxyl radical which
is reduced to a hydroperoxide Ž6. as described above.
Alternatively, carbon-centered radical 5 can undergo 2. Alternate pathways to aldehydes linked to
a second cyclization to form a bicyclic peroxide inflammation
which after coupling to O 2 and reduction yields a
molecule Ž7. structurally analogous to the prosta- Another pathway has been described recently for
glandin endoperoxide, PGG2 , albeit without the the formation of aldehydes that is independent of
stereochemical control exhibited in the enzymatic lipid peroxidation but which may be important for
reaction w6x. Compound 7 serves as a common inter- the generation of gentoxic compounds. Hazen et al.
mediate for the production of isoprostanes and have demonstrated that myeloperoxidase in the pres-
malondialdehyde ŽMDA. through chemical conver- ence of chlorine generates hypochlorous acid which
sion of the bicyclic peroxide group w6,7x. The frag- oxidizes amino acids to chloramines w12x. The latter
mentation that produces MDA generates a 17-carbon derivatives lose of chloride and CO 2 to form imines;
fatty acid as the other product w8x. MDA has been the imines hydrolyze to aldehydes ŽScheme 2.. In the
used for many years as a convenient biomarker for case of threonine, the hydroxyaldehyde product un-
lipid peroxidation because of its facile reaction with dergoes dehydration to form acrolein. This may be
thiobarbituric acid to form an intensely colored chro- an important source of the acrolein that damages
mogen w9x. However, the thiobarbituric acid reaction DNA endogenously in human beings and which has
is notoriously non-specific which has led to substan- been assumed to arise from lipid peroxidation w13x.
tial controversy over its use for quantitation of MDA
from in vivo samples. Recently, isoprostanes have
become popular biomarkers of lipid peroxidation and 3. Genotoxicity of MDA
their use has been validated in several in vivo mod-
els w7x. MDA was first shown to be carcinogenic in 1972
All of the hydroperoxide intermediates depicted in following topical administration to mice w14x. This
Scheme 1 and their many regioisomers and was an unusual result because the tumors arose in a
stereoisomers can be reduced by metal ions to alkoxyl very short time and were not the type of tumors

Scheme 2. Oxidation of amino acids to aldehydes by myeloperoxidase.


86 L.J. Marnettr Mutation Research 424 (1999) 83–95

typically seen in a mouse skin assay. Subsequent


attempts to confirm this skin carcinogenesis result
were unsuccessful w15x. Mukai and Goldstein re-
ported the mutagenic activity of MDA toward
Salmonella typhimurium in 1976 w16x. Since MDA is
unstable, the material used for the assay was pre-
pared by hydrolysis of tetraethoxypropane. Some
impurities generated during tetraethoxypropane hy-
drolysis are nearly 30-fold more mutagenic than
MDA and account for a significant part of the activ-
ity Ž; 50%. detected in the assay w17x. However,
assay of highly purified MDA prepared by three
independent means revealed that this compound is,
indeed, mutagenic w18x. The carcinogenic activity of
MDA was established in a 2-year rodent bioassay
w19x. Highly purified preparations of MDA induced
thyroid tumors in rats but not mice.
Several other aldehydes, including some a ,b-un-
saturated aldehydes are mutagenic in Salmonella if
steps are taken to minimize their toxicity w20x. This
consists of adding high concentrations of glutathione
shortly after administration of the aldehyde to the
bacteria to strip aldehydes from cysteinyl groups of
proteins which are putative targets for their toxic
action. In fact, 4-hydroxynonenal was originally dis-
covered as a highly toxic product of lipid peroxida-
tion and both it and its glutathione conjugate were
used as chemotherapeutic agents in some small hu-
man clinical trials w21x. The toxic activity of the
4-hydroxynonenal appeared to be due to its reactivity
with sulfhydryl proteins w22x.
Scheme 3. Formation of MDA–DNA adducts.

4. Reaction of MDA with DNA


the pyrimidow1,2 a xpurin-10Ž3 H .-one ŽM 1G. is the
MDA reacts with nucleic acid bases to form major adduct followed by N 6-Ž3-oxo-propenyl.
multiple adducts ŽScheme 3.. Both its carbonyl deoxyadenosine ŽM 1 A.. The amount of M 1G is ap-
equivalent adducts to N 2 and N 1 of dG with loss of proximately five times that of M 1 A. N 4-Ž3-
two water molecules to form a pyrimidopurinone oxopropenyl.deoxycytidine ŽM 1C. is formed in only
w23,24x. This compound is a planar aromatic molecule trace amounts w28x.
that is moderately fluorescent Ž; 3% quantum yield.. A complicating factor in the reaction of MDA
The condensation products with dA and dC arise by with nucleosides is its polymerization to form dimers
addition of one of the carbonyl equivalents of MDA and trimers that also react with DNA. The dimer of
with the exocyclic amino groups to form an oxo- MDA reacts with dG to form an oxadiazabi-
propenyl derivative w25–27x. No evidence for cy- cyclow3:3:1xnonene derivative whereas the trimer of
clization of these products has been observed. Com- MDA reacts with dA and dC to form Ž5X ,7X-bis-for-
parison of the yields of the various adducts produced myl-2X H-3X ,6X - dihydro-2X ,6X-methano-1X ,3X - oxazocin-
in the reaction of MDA with DNA in vitro indicates 3X-yl. derivatives of the exocyclic amino groups
L.J. Marnettr Mutation Research 424 (1999) 83–95 87

w26,27x. The oligomerization of MDA is relatively Base propenals are oxopropenyl base derivatives
slow at neutral pH so the monomeric adducts de- structurally analogous to acroleins substituted at the
picted in Scheme 3 are the major products generated b position with good-to-moderate leaving groups
under physiological conditions. However, there may ŽScheme 4.. These compounds Žincluding M 1 A and
be conditions in vitro or possibly in vivo where these M 1C. transfer their oxopropenyl group to dG to form
unusual multimeric adducts are formed. The ability M 1G. Treatment of DNA with bleomycin or
of MDA to form monomeric and oligomeric adducts calicheamicin in the complete absence of lipid leads
is a reflection of the nucleophilic reactivity of its to the formation of M 1G. M 1G also is formed by the
enol functional group whereas the electrophilic reac- direct reaction of adenine propenal with DNA. This
tivity is due to its aldehyde functional group. The provides another pathway linking oxidative stress to
existence of oligomeric deoxynucleoside adducts ne- the formation of M 1G in DNA and may explain the
cessitated the adoption of abbreviations for the indi- occurrence of M 1G at high levels in certain human
vidual adducts that designate the number of MDA tissues Žsee below..
units in each adduct ŽScheme 3..

6. Mutagenic potential and repair of MDA–DNA


5. Alternate routes to M 1G adducts

Dedon et al. have recently demonstrated that M 1G The mutagenic potential of MDA–DNA adducts
Žand presumably other MDA-derived adducts. can be has been evaluated using random and site-specific
formed independently of lipid peroxidation w29x. Di- approaches. Benamira et al. reacted MDA at neutral
rect oxidation of DNA by agents that abstract the 4X pH with a single-stranded M13 vector containing the
hydrogen atom of the sugar backbone Že.g., lacZ a gene and scored mutations to the lacZ ay
calicheamicin, bleomycin. initiates a cascade of reac- phenotype following replication in strains of Es-
tions that lead to the formation of base propenals. cherichia coli induced for the SOS response w30x.
Increasing concentrations of MDA led to an increase
in lacZ ay mutations coincident with an increase in
the level of the major MDA–deoxyguanosine adduct,
M 1G. The most common sequence changes induced
by MDA were base pair substitutions Ž76%.. Of
these, 43% Ž29r68. were transversions, most of
which were G ™ T Ž24r29.. Transitions accounted
for 57% of the base pair substitutions Ž39r68. and
were comprised exclusively of C ™ T Ž22r39. and
A ™ G Ž17r39.. Frameshift mutations were identi-
fied in 16% of the induced mutants and were com-
prised of mainly single base additions occurring in
runs of reiterated bases Ž11r14.. If one assumes that
all mutations are targeted to the site of adduction, the
ability of MDA to induce base pair substitution
mutations at dG, dA, and dC residues coincides with
its ability to form adducts at all three deoxynucleo-
sides. No mutations were detected at dT residues as
expected from the fact that MDA does not form
detectable adducts to dT.
Site-specific mutagenesis experiments have been
used to investigate the mutagenic potential of M 1G.
Scheme 4. Reaction of oxidizing agents with DNA to form M 1G. This necessitated modification of the synthetic reac-
88 L.J. Marnettr Mutation Research 424 (1999) 83–95

tions used for oligodeoxynucleotide synthesis to ac- by reconstituted bacterial and mammalian nucleoside
commodate the instability of M 1G to the base depro- excision repair complexes was confirmed by a series
tection conditions normally used in the last step of of in vitro experiments w33x. PdG appears to be
the synthetic pathway w31x. M 1G was positioned at a removed more efficiently by the mammalian nu-
defined site in a duplex M13 genome using a gapped cleotide excision repair complex than the bacterial
duplex approach. Unmodified and M 1G-modified complex.
genomes containing either a cytosine or thymine
opposite the lesion were transformed into repair-pro-
ficient and repair-deficient E. coli strains. Base pair 7. Occurrence of M 1G in genomic DNA
substitutions were quantitated by hybridization anal-
ysis. Modified genomes containing a cytosine oppo- 7.1. Postlabeling analysis
site M 1G resulted in roughly equal numbers of M 1G
™ A and M 1G ™ T mutations with few M 1G ™ C The high mutagenic potential of M 1G begs the
mutations w32x. The total mutation frequency was question of whether it is formed in human beings. As
approximately 1%, which represents a 500-fold in- with all endogenous DNA adducts, answering this
crease in mutations compared to unmodified question requires unusually robust and rigorous as-
M13MB102. Transformation of modified genomes says because of the lack of a control DNA from an
containing a thymine opposite M 1G allowed an esti- untreated animal or human sample. By definition, the
mate to be made of the ability of M 1G to block untreated DNA is what is being analyzed for the
replication. The Žy.-strand was replicated ) 80% of presence of ‘background DNA damage’. Thus, any
the time in the unadducted genome, but only 20% of assay used for analysis must be highly sensitive and
the time when M 1G was present. Correction of the very specific. One must be able to verify that the
mutation frequency for the strand bias of replication adduct detected is actually what it is presumed to be.
indicated that the actual frequency of mutations in- This is no simple task with any DNA adduct.
32
duced by M 1G was 18% w32x. The spectrum and P-Postlabeling was first employed for the detec-
frequency of mutations induced by M 1G were very tion and quantitation M 1G. Vaca and coworkers
similar to those observed with a structural analog developed a method using nuclease P1 treatment for
1, N 2-propanodeoxyguanosine ŽPdG. assayed in the enrichment that was used to detect the adduct in
same system. mouse liver DNA w34x. Treatment of mice with
Repair of M 1G was assessed by transformation of MDA Ž20 mmolrkg. caused an increase in adduct
M 1G-containing genomes into E. coli strains defi- level to 6 per 10 6 nucleotides. The levels of M 1G in
cient in individual genes of DNA repair based on the untreated animals was not reported. In subsequent
assumption that inactivation of a repair gene will studies, treatment of mice with 200 mmolrkg MDA
increase the half-life of the adduct and increase its stimulated the formation of MDA–hemoglobin
mutagenicity in site-specific experiments. No effect adducts in blood and M 1G in liver. The levels of
on mutation frequency was observed when cells M 1G declined slowly following MDA treatment and
were defective in formamidopyrimidine glycosylase demonstrated a half-life of 12.5 days w35x. This slow
or 3-methyladenine glycosylase. These two glycosy- half-life for removal is interesting considering the
lases have been shown previously to participate in efficient removal of M 1G and PdG by nucleotide
the repair of 8-oxo-7,8-dihydrodeoxyguanosine Ž8- excision repair described above. Interestingly, in
oxodG. and exocyclic adducts, respectively w32,33x. these experiments, the levels of M 1G in the liver
Approximately a fourfold increase in mutation fre- increased significantly up to 4 days after administra-
quency was observed when M 1G-containing genomes tion of MDA.
were transformed into cells deficient in nucleotide Wang and Liehr utilized 32 P-postlabeling to
excision repair Žeither uÕrAy or uÕrBy . w32x. In- demonstrate a 2- to 3-fold increase in the levels of
creases in mutation frequency of similar magnitude M 1G in the kidneys of Syrian hamsters treated with
were observed with PdG-containing genomes. The doses of diethylstilbestrol or estradiol that induce
ability of PdG Žand by analogy M 1G. to be removed cancer in this organ w36x. Wang and Liehr also
L.J. Marnettr Mutation Research 424 (1999) 83–95 89

provided evidence for the existence of MDA adducts


to dA as well as dG w37x. The identities of these
adducts have not yet been established but they have
been found in human white cells and breast Žsee
below..
The levels of M 1G in human white blood cells
and breast tissue have been determined by 32 P-post-
labeling. Values in white cells of 2.6 " 1.2 adducts
per 10 7 nucleotides Žrange 1–5 per 10 7 . were re-
ported from a study of 26 male and female volun-
teers w38x. Parallel analysis of 7 female volunteers
indicated levels of 3.0 " 1.3 M 1G per 10 7 nu-
cleotides in normal breast tissue. Fang et al. reported
a dramatic increase in the level of M 1G in the white
cells of women Žbut not men. fed a diet high in
sunflower oil compared to a group fed a high rape-
seed oil diet w39x. The high polyunsaturated fatty acid
diet stimulated nearly a 20-fold increase in the levels
of M 1G up to levels of 28 per 10 7 nucleotides. Wang
et al. reported that the levels of M 1G and putative
MDA–dA adducts were higher Ž2- to 3-fold. in the
normal breast tissue of women with breast cancer
compared to the normal tissue of women without
breast cancer w40x. The levels of M 1G in the tumor
tissue of women with breast cancer were lower than
those in the surrounding normal tissue. Scheme 5. Steps in the analysis of M 1G by GCrEC NCIrMS.

7.2. Mass spectrometric analysis mode enables quantitation by comparison of the


intensity of the base peak of the endogenously occur-
Our laboratory has employed gas chromatogra- ring M 1G Ž mrz s 186. to that of the internal stan-
phyrmass spectrometry ŽGCrMS. with electron dard Ž mrz s 188. ŽFig. 1.. The chromatographic
capture negative chemical ionization detection trace in Fig. 1 displays two separable M 1G-PFB
ŽGCrEC NCIrMS. for quantitation of M 1G w41,42x. derivatives corresponding to the N7 and N9 isomers.
Scheme 5 charts the course of the analytical method. The GCrEC NCIrMS assays display good linearity
Nuclear DNA is isolated, purified and digested to and quantitative recovery of M 1G when oligonucleo-
deoxynucleosides. During nuclease treatment, an in- tides containing this adduct are carried through the
ternal standard of M 1G-deoxyribose labeled with two analytical procedure w42x. The limit of sensitivity of
deuteriums is added. The mixture of deoxynucleo- the assay is approximately 1 M 1G per 10 8 nu-
sides is passed through an immunoaffinity column cleotides starting with 1 mg of DNA. Specificity is
containing an immobilized monoclonal antibody provided by the combination of immunoaffinity
raised against a periodate-cleaved M 1G-ribose conju- chromatography, chemical derivatization, chromato-
gated to albumin w43x. Unmodified deoxynucleosides graphic coelution, and selected ion monitoring.
pass through the immunoaffinity column then the Application of this technique to human liver and
M 1G-deoxyribose is eluted with methanol. The de- leukocyte DNA samples reveals that M 1G is present
oxyribose residue is removed by acid hydrolysis then at levels of ; 1 in 10 6 nucleosides and 6 in 10 8
the M 1G base is derivatized with pentafluorobenzyl nucleosides, respectively w41,42x Žsee Table 1.. The
ŽPFB. bromide. Analysis of the PFB derivatives by livers were unused transplant samples and the leuko-
GCrEC NCIrMS in the selected ion monitoring cyte samples were from human volunteers. The sam-
90 L.J. Marnettr Mutation Research 424 (1999) 83–95

Fig. 1. Selected ion monitoring profiles of PFB derivatives of M 1G. The upper trace represents the channel for the material endogenously
present in DNA Ž mrz s 186. whereas the lower trace represents the channel for the internal standard Ž mrz s 188..

ple size in the latter study was rather small but no chemical, immunochemical and spectroscopic pre-
obvious differences in M 1G levels were detected requisites. However, it is possible to directly verify
between the two smokers included in the study and a the presence of M 1G residues in DNA by liquid
very small but statistically significant difference was chromatographyrmass spectrometry ŽLCrMS.. A
seen between men and women. A study of M 1G sample of human liver DNA that exhibited a high
levels in unused human pancreas transplant samples concentration of M 1G by GCrEC NCIrMS analysis
demonstrated a bimodal distribution. The levels in was processed in the absence of an internal standard
approximately half the samples fell below the limit and the M 1G-containing fractions were isolated w45x.
of detection of the assay and the levels in the other The isolated material was then injected into a triple
half ranged from 2–50 per 10 8 nucleosides w44x. quadrupole LCrMS with electrospray detection. As
As indicated above, the specificity of the GCrEC observed with most deoxynucleosides, source-in-
NCIrMS assay is ensured by a combination of duced fragmentation led to loss of the deoxyribose
L.J. Marnettr Mutation Research 424 (1999) 83–95 91

Table 1 quadrupole. The spectrum of ions generated was


Levels of M 1G in human tissues identical to that produced by tandem mass spectral
Tissue Averagea Range Technique Reference analysis of an authentic standard of M 1G ŽFig. 2..
Liver 9 Ž ns6. 5–12 GCrMS w41x This establishes unequivocally that M 1G residues are
White cells 0.6 Ž ns10. 0.5–0.8 GCrMS w42x present in human liver DNA. Similar verification has
Pancreas 3.2 Ž ns 27. - 0.1–5 GCrMS w47x
32 been carried out on human leukocyte DNA samples
White cells 2.6 Ž ns 26. 1–5 P w48x
White cells b 0.9 Ž ns 7. 0.2–2.5 32
P w39x but in that case the low levels of M 1G required a
White cells c 11 Ž ns6. 1.2–28 32
P w39x mass fragmentographic approach. The offset voltage
Breast d 0.2 Ž ns 51. 0.05–1.3 32 P w40x in the second quadrupole was programmed to pro-
Breast e 0.08 Ž ns 28. 0.02–0.19 32 P w40x gressively change the fragmentation pattern of the
32
Breast 3 Ž ns 7. 0.7–5.6 P w48x
protonated molecular ion and the expected alteration
a
All levels represent number of adducts per 10 7 nucleotides. in fragmentation pattern for M 1G was observed w42x.
b
Women fed high monounsaturated fat diet. A concern in the analysis of endogenous DNA
c
d
Women fed high polyunsaturated fat diet. adducts is the possibility that they are artifactually
Normal tissue from breast cancer patients. generated during the workup. This is especially seri-
e
Normal breast tissue from non-cancer bearing women.
ous if one is studying adducts that could be gener-
ated by oxidative processes that might occur post
mortem or ex vivo. We have used different ap-
moiety in the first quadrupole. The protonated proaches to minimize this problem in the analysis of
molecular ion corresponding to M 1G Ž mrz s 188. M 1G w41x. During tissue handling and DNA isola-
was then collisionally dissociated in the second tion, large amounts of antioxidants were added to all
quadrupole and the product ions analyzed in the third buffers to minimize post mortem lipid peroxidation.

Fig. 2. Mass spectrum of the collisionally dissociated mrz s 188 ion produced during the analysis of a human liver DNA sample.
92 L.J. Marnettr Mutation Research 424 (1999) 83–95

Interestingly, inclusion of butylated hydroxyanisole the levels of M 1G in liver increases by nearly 100%
Ž10 mM. or vitamin E Ž10 mM. did not lower the w41x.
levels of M 1G detected in liver tissue but inclusion
of vitamin C increased them approximately 2-fold.
We attribute this to the long-recognized ability of
ascorbic acid to support iron-catalyzed lipid per- 8. Perspective
oxidation.
The other approach we have employed to assess The available evidence indicates that M 1G, and
possible artifactual M 1G production is the inclusion possibly other MDA-derived adducts, is a highly
of isotopically labeled dG in the buffer used for mutagenic adduct that is present in the genomic
DNA digestion ŽScheme 6. w41x. Deoxyguanosine, DNA of human beings ŽTable 1.. The levels detected
chemically synthesized to contain four 13 C and one by various methods are reasonably similar consider-
15
N atoms, was added to the digestion mixture in ing the differences in the basic analytical procedures.
amounts comparable to the amounts of dG in the What is lacking at present is an understanding of the
DNA sample. If any MDA was present as a result of importance of the detection of this adduct in people.
lipid peroxidation during tissue manipulation, it This is a problem facing virtually everyone who
would react with the isotopically labeled dG to form studies the chemistry and biology of DNA adducts.
M 1G that would yield a PFB derivative in GCrEC How can one relate the level of a particular DNA
NCIrMS with a mrz s 191. No such material was adduct to a disease outcome? This is especially
detected in any of our analyses. difficult for a disease like cancer which is multifac-
Analysis of rat tissues for M 1G by GCrEC torial and involves not only mutations in multiple
NCIrMS reveals a level in liver Ž; 6 per 10 7 nucle- genes but also proliferation, clonal expansion, and
osides. comparable to that detected in human beings. escape from apoptosis.
The level in 2 year old rats is nearly double that in 6 Relating the level of an adduct to an outcome is a
week old rats. Interestingly, analysis of testis DNA challenge that will be approached primarily by cor-
samples from the same rats reveals that the levels of relative studies. Does the adduct level go up or down
M 1G are below the limit of detection of the assay Ž1 in a predictable or logical way in situations that
per 10 8 nucleosides.. When rats are exposed to the predispose or protect from development of the dis-
lipid peroxidation stimulus CCl 4 by oral intubation, ease? Given the variety of endogenous DNA dam-
age, answering this question for a single adduct will
require studies with large population groups. Thus,
the analytical assays will not only need to be robust
and specific but also amenable to high throughput
formats that can be carried out by interested but not
expert investigators. 32 P-Postlabeling and GCrEC
NCIrMS assays are time consuming and require
extensive sample manipulation by highly skilled per-
sonnel. 32 P-Postlabeling offers the advantage of high
sensitivity with minimal requirements for DNA Ž10
mg.. Immunochemical approaches offer considerable
promise for population-based studies because of their
speed and sensitivity. For example, Leuratti et al.
recently reported the development of an immunoslot
blot assay for M 1G that uses the previously described
monoclonal antibody raised against this adduct w46x.
The assay was used to examine a large number of
Scheme 6. Isotopic probe for artifactual generation on M 1G ex gastric biopsy samples to probe for a relationship
vivo. between inflammation and MDA–DNA adduction.
L.J. Marnettr Mutation Research 424 (1999) 83–95 93

Although there is always concern about cross-re- made by using either water-soluble or lipid-soluble chain-
activity in immunochemical reactions, the availabil- breaking antioxidants, J. Am. Chem. Soc. 106 Ž1984. 2479–
2481.
ity of mass spectrometric approaches to adduct iden- w5x A.R. Waldeck, R. Stocker, Radical-initiated lipid peroxida-
tification provides a backup that can be used to tion in low density lipoproteins: insights obtained from ki-
confirm adduct level and structure when an interest- netic modeling, Chem. Res. Toxicol. 9 Ž1996. 954–964.
ing trend is noted. GCrMS methods can be used to w6x W.A. Pryor, J.P. Stanley, A suggested mechanism for the
confirm adducts levels and LCrMS methods can be production of malondialdehyde during the autoxidation of
polyunsaturated fatty acids. Non-enzymatic production of
used to confirm structure. In fact, one might specu- prostaglandin endoperoxides during autoxidation, J. Org.
late that LCrMS methods will eventually evolve Chem. 40 Ž1975. 3615–3617.
into high throughput, robust assays. The sensitivity w7x J.D. Morrow, L.J. Roberts, The isoprostanes—current
of LCrMS has increased approximately three orders knowledge and directions for future research, Biochem. Phar-
of magnitude in the last 10 years to a point where it macol. 51 Ž1996. 1–9.
w8x M. Hamberg, B. Samuelsson, On the mechanism of the
is within an order of magnitude of GCrMS. For biosynthesis of prostaglandins E 1 and F1a , J. Biol. Chem.
example, the limit of detection of M 1G by LCrMS 242 Ž1967. 5336–5343.
is 3 per 10 7 from 1 mg of DNA. Impressive im- w9x D.R. Janero, Malondialdehyde and thiobarbituric acid-reac-
provements are being made in interfaces, detectors, tivity as diagnostic indices of lipid peroxidation and perox-
plumbing and data reduction that will certainly trans- idative tissue injury, Free Radic. Biol. Med. 9 Ž1990. 515–
540.
late into major increases in sensitivity in the next w10x H. Esterbauer, Lipid peroxidation products: formation, chem-
few years. Therefore, it seems only a matter of time ical properties and biological activities, in: G. Pli, K.H.
before DNA hydrolyzates are processed by LCrMS Cheeseman, M.U. Dianzani, T.F. Slater ŽEds.., Free Radicals
in high throughput mode for quantitation of a range in Liver Injury, IRL Press, Oxford, 1985, pp. 29–47.
w11x H. Esterbauer, P. Eckl, A. Ortner, Possible mutagens derived
of endogenously and exogenously derived adducts.
from lipids and lipid precursors, Mutat. Res. Rev. Genet.
The availability of a general method for adduct Toxicol. 238 Ž1990. 223–233.
identification and quantitation will help determine an w12x S.L. Hazen, F.F. Hsu, A. d’Avignon, J.W. Heinecke, Human
inventory of DNA damage in individual human be- neutrophils employ myeloperoxidase to convert a-amino
ings, regardless of the source. acids to a battery of reactive aldehydes: a pathway for
aldehyde generation at sites of inflammation, Biochemistry
37 Ž1998. 6864–6873.
w13x R.G. Nath, F.-L. Chung, Detection of exocyclic 1, N 2-pro-
Acknowledgements panodeoxyguanosine adducts as common DNA lesions in
rodents and humans, Proc. Natl. Acad. Sci. U.S.A. 91 Ž1994.
Research in the Marnett laboratory is supported 7491–7495.
w14x R.J. Shamberger, T.L. Andreone, C.E. Willis, Antioxidants
by research and center grants from the National and cancer: IV. Initiating activity of malonaldehyde as a
Institutes of Health ŽCA47479, ES00267, and carcinogen, J. Natl. Cancer Inst. 53 Ž1974. 1771–1773.
CA68485.. w15x S.M. Fischer, S. Olge, L.J. Marnett, S. Nesnow, T.J. Slaga,
The lack of initiating andror promoting activity of sodium
malondialdehyde on Sencar mouse skin, Cancer Lett. 19
Ž1983. 61–66.
References w16x F.H. Mukai, B.D. Goldstein, Mutagenicity of malondialde-
hyde, a decomposition product of peroxidized polyunsatu-
w1x N.A. Porter, Mechanisms for the autoxidation of polyunsatu- rated fatty acids, Science 191 Ž1976. 868–869.
rated lipids, Acc. Chem. Res. 19 Ž1986. 262–268. w17x L.J. Marnett, M.A. Tuttle, Comparison of the mutagenicities
w2x T.A. Dix, J. Aikens, Mechanisms and biological relevance of of malondialdehyde and side products formed during its
lipid peroxidation initiation, Chem. Res. Toxicol. 6 Ž1993. chemical synthesis, Cancer Res. 40 Ž1980. 276–282.
2–18. w18x A.K. Basu, L.J. Marnett, Unequivocal demonstration that
w3x W.A. Pryor, Oxy-radicals and related species: their forma- malondialdehyde is a mutagen, Carcinogenesis 4 Ž1983.
tion, lifetimes and reactions, Annu. Rev. Physiol. 48 Ž1986. 331–333.
657–667. w19x J.W. Spalding, Toxicology and carcinogenesis studies of
w4x L.R.C. Barclay, S.J. Locke, J.M. MacNeil, J. VanKessel, malondialdehyde sodium salt Ž3-hydroxy-2-propenal, sodium
G.W. Burton, K.U. Ingold, Autoxidation of micelles and salt. in F344rN rats and B6C3F1 mice, NTP Technical
model membranes. Quantitative kinetic measurements can be Report 331 Ž1988. 5–13.
94 L.J. Marnettr Mutation Research 424 (1999) 83–95

w20x L.J. Marnett, H.K. Hurd, M.C. Hollstein, D.E. Levin, H. w34x C.E. Vaca, P. Vodicka, K. Hemminki, Determination of
X X
Esterbauer, B.N. Ames, Naturally occurring carbonyl com- malonaldehyde-modified 2 -deoxyguanosine-3 -monophos-
32
pounds are mutagens in Salmonella tester strain TA104, phate and DNA by P-postlabeling, Carcinogenesis 13
Mutat. Res. 148 Ž1985. 25–34. Ž1992. 593–599.
w21x E. Schauenstein, H. Esterbauer, H. Zollner, Malondialde- w35x A. Kautiainen, C.E. Vaca, F. Granath, Studies on the rela-
hyde, in: E. Schauenstein, H. Esterbauer, H. Zollner ŽEds.., tionship between hemoglobin and DNA adducts of malon-
Aldehydes in Biological Systems, Pion, London, 1977, pp. aldehyde and their stability in vivo, Carcinogenesis 14 Ž1993.
133–140. 705–708.
w22x H. Krokan, R.C. Grafstrom, K. Sundqvist, H. Esterbauer, w36x M.-Y. Wang, J.G. Liehr, Induction by estrogens of lipid
C.C. Harris, Cytotoxicity, thiol depletion and inhibition of peroxidation and lipid peroxide-derived malonaldehyde–
O 6-methylguanine–DNA methyltransferase by various alde- DNA adducts in male Syrian hamsters: role of lipid peroxida-
hydes in cultured human bronchial fibroblasts, Carcinogene- tion in estrogen-induced kidney carcinogenesis, Carcinogene-
sis 6 Ž1985. 1755–1759. sis 16 Ž1995. 1941–1945.
w23x H. Seto, T. Okuda, T. Takesue, T. Ikemura, Reaction of w37x M.Y. Wang, J.G. Liehr, Lipid hydroperoxide-induced en-
malondialdehyde with nucleic acid: I. Formation of fluores- dogenous DNA adducts in hamsters: possible mechanism of
cent pyrimidow1,2-axpurin-10Ž3 H .-one nucleosides, Bull. lipid hydroperoxide-mediated carcinogenesis, Arch. Biochem.
Chem. Soc. Jpn. 56 Ž1983. 1799–1802. Biophys. 316 Ž1995. 38–46.
w24x L.J. Marnett, A.K. Basu, S.M. O’Hara, P.E. Weller, w38x C.E. Vaca, J.-L. Fang, M. Mutanen, L. Valsta, 32 P-Postlabel-
A.F.M.M. Rahman, J.P. Oliver, Reaction of malondialdehyde ing determination of DNA adducts of malonaldehyde in
with guanine nucleosides: formation of adducts containing humans: total white blood cells and breast tissue, Carcino-
oxadiazabicyclononene residues in the base-pairing region, J. genesis 16 Ž1995. 1847–1851.
Am. Chem. Soc. 108 Ž1986. 1348–1350. w39x J.L. Fang, C.E. Vaca, L.M. Valsta, M. Mutanen, Determina-
w25x V. Nair, G.A. Turner, R.J. Offerman, Novel adducts from the tion of DNA adducts of malonaldehyde in humans: effects of
modification of nucleic acid bases by malondialdehyde, J. dietary fatty acid composition, Carcinogenesis 17 Ž1996.
Am. Chem. Soc. 106 Ž1984. 3370–3371. 1035–1040.
w26x K. Stone, M. Ksebati, L.J. Marnett, Investigation of the w40x M. Wang, K. Dhingra, W.N. Hittleman, J.G. Liehr, M. de
adducts formed by reaction of malondialdehyde with adeno- Andrade, D. Li, Lipid peroxidation-induced putative malon-
sine, Chem. Res. Toxicol. 3 Ž1990. 33–38. dialdehyde–DNA adducts in human breast tissues, Cancer
w27x K. Stone, A. Uzieblo, L.J. Marnett, Studies of the reaction of Epidemiol. Biomarkers Prev. 5 Ž1996. 705–710.
malondialdehyde with cytosine nucleosides, Chem. Res. Tox- w41x A.K. Chaudhary, M. Nokubo, G.R. Reddy, S.N. Yeola, J.D.
icol. 3 Ž1990. 467–472. Morrow, I.A. Blair, L.J. Marnett, Detection of endogenous
w28x A.K. Chaudhary, G.R. Reddy, I.A. Blair, L.J. Marnett, Char- malondialdehyde–deoxyguanosine adducts in human liver,
X
acterization of an N 6-oxopropenyl-2 -deoxyadenosine adduct Science 265 Ž1994. 1580–1582.
in malondialdehyde-modified DNA using liquid chromatog- w42x C.A. Rouzer, A.K. Chaudhary, M. Nokubo, D.M. Ferguson,
raphy electrospray ionization tandem mass spectrometry, G.R. Reddy, I.A. Blair, L.J. Marnett, Analysis of the malon-
X
Carcinogenesis 17 Ž1996. 1167–1170. dialdehyde-2 -deoxyguanosine adduct, pyrimidopurinone, in
w29x P.C. Dedon, J.P. Plastaras, C.A. Rouzer, L.J. Marnett, Indi- human leukocyte DNA by gas chromatographyrelectron cap-
rect mutagenesis by oxidative DNA damage: formation of ture negative chemical ionizationrmass spectrometry, Chem.
the pyrimidopurinone adduct of deoxyguanosine by base Res. Toxicol. 10 Ž1997. 181–188.
propenal, Proc. Natl. Acad. Sci. U.S.A., 1998, submitted. w43x C.L. Sevilla, N.H. Mahle, N. Eliezer, A. Uzieblo, S.M.
w30x M. Benamira, K. Johnson, A. Chaudhary, K. Bruner, C. O’Hara, M. Nokubo, R. Miller, C.A. Rouzer, L.J. Marnett,
Tibbetts, L.J. Marnett, Induction of mutations by replication Development of monoclonal antibodies to the malondialde-
of malondialdehyde-modified M13 DNA in Escherichia coli: hyde–deoxyguanosine adduct, pyrimidopurinone, Chem. Res.
determination of the extent of DNA modification, genetic Toxicol. 10 Ž1997. 172–180.
requirements for mutagenesis, and types of mutations in- w44x F. Kadlubar, K. Anderson, N. Lang, P. Thompson, S.
duced, Carcinogenesis 16 Ž1995. 93–99. MacLeod, M. Mikhailova, M. Chou, J. Plastaras, L. Marnett,
w31x G.R. Reddy, L.J. Marnett, Synthesis of an oligodeoxynu- S. Haussermann, H. Bartsch, Comparison of endogenous
cleotide containing the alkaline labile malondialdehyde– DNA adducts levels in human pancreas, Proc. Am. Assoc.
deoxyguanosine adduct pyrimidow1,2-axpurin-10Ž3 H .-one, J. Cancer Res. 39 Ž1998. 286, Abstract.
Am. Chem. Soc. 117 Ž1995. 5007–5008. w45x A.K. Chaudhary, M. Nokubo, T.D. Oglesby, L.J. Marnett,
w32x S.P. Fink, G.R. Reddy, L.J. Marnett, Mutagenicity in Es- I.A. Blair, Characterization of endogenous DNA adducts by
cherichia coli of the major DNA adduct derived from the liquid chromatographyrelectrospray ionizationrtandem mass
endogenous mutagen malondialdehyde, Proc. Natl. Acad. spectrometry, J. Mass Spectrom. 30 Ž1995. 1157–1166.
Sci. U.S.A. 94 Ž1997. 8652–8657. w46x C. Leuratti, R. Singh, C. Lagneau, P.B. Farmer, L.J. Marnett,
w33x K.A. Johnson, S.P. Fink, L.J. Marnett, Repair of propan- D.E.G. Shuker, Quantitation of malondialdehyde–DNA dam-
odeoxyguanosine by nucleotide excision repair in vivo and in age in human tissues using a sensitive immunoslot blot assay,
vitro, J. Biol. Chem. 272 Ž1997. 11434–11438. Proc. Am. Assoc. Cancer Res. 39 Ž1998. 286, Abstract.
L.J. Marnettr Mutation Research 424 (1999) 83–95 95

w47x F.F. Kadlubar, K.E. Anderson, N.P. Lang, P.A. Thompson, w48x C.E. Vaca, J.-L. Fang, M. Mutanen, L. Valstar, 32 P-Post-
S.L. MacLeod, M.W. Chou, M. Mikhailova, J. Plastaras, L.J. labeling determination of DNA adducts of malonaldehyde in
Marnett, S. Haussermann, J. Nair, I. Velie, H. Bartsch, humans: total white blood cells and breast tissue, Carcino-
Comparison of endogenous DNA adduct levels in human genesis 16 Ž1995. 1847–1851.
pancreas, Mutat. Res., 1998, in press.

You might also like