Sei sulla pagina 1di 14

Free Radical Biology & Medicine, Vol. 33, No. 1, pp.

114, 2002
Copyright 2002 Elsevier Science Inc.
Printed in the USA. All rights reserved
0891-5849/02/$see front matter

PII S0891-5849(02)00827-4

Serial Review: Oxidative DNA Damage and Repair


Guest Editor: Miral Dizdaroglu
BIOLOGICAL CONSEQUENCES OF FREE RADICAL-DAMAGED DNA BASES
SUSAN S. WALLACE
Department of Microbiology and Molecular Genetics, The Markey Center for Molecular Genetics, The University of Vermont,
Burlington, VT, USA
(Received 11 December 2001; Revised 12 February 2002; Accepted 1 March 2002)

AbstractThe principal oxidized cytosine bases, uracil glycol, 5-hydroxycytosine, and 5-hydroxyuracil, are readily
bypassed, miscode, and are thus important premutagenic lesions. Similarly the principal oxidation product of guanine,
8-oxoguanine, miscodes with A and is a premutagenic lesion. Most of the thymine and adenine products that retain their
ring structure primarily pair with their cognate bases and are not potent premutagenic lesions. Although thymine glycol
pairs with its cognate base and is not mutagenic it significantly distorts the DNA molecule and is a lethal lesion. Ring
fragmentation, ring contraction, and ring open products of both pyrimidines and purines block DNA polymerases and
are potentially lethal lesions. Although these breakdown products have the potential to mispair during translesion
synthesis, the mutational spectra of prokaryotic mutants defective in the pyrimidine-specific and/or purine-specific DNA
glycosylases do not reflect that expected of the breakdown products. Taken together, the data suggest that the principal
biological consequences of endogenously produced and unrepaired free radical-damaged DNA bases are
mutations. 2002 Elsevier Science Inc.
KeywordsFree radical DNA damage, Oxidized pyrimidines, Oxidized purines, Base excision repair, Oxidized bases
and DNA polymerases, Oxidized bases and mutations

INTRODUCTION

lethal lesion. If, on the other hand, the lesion is not


structurally distorting, pairs with a noncognate base and
is readily bypassed by DNA polymerases, it is taken to
be premutagenic. Some lesions are neither mutagenic nor
lethal while others are lethal and mutagenic by virtue of
being able to be bypassed during translesion synthesis by
a specialized polymerase. To verify biochemical predictions, individual lesions have been introduced into unrepairable single-stranded viral DNA molecules and assessed for their lethal and mutagenic potential [3 6].
This approach is advantageous because lesion structure
and in vitro interactions with DNA polymerases can be
directly related to biological consequences; the disadvantage is that only a single sequence context is usually
investigated. Finally, sensitivity to DNA-damaging
agents and mutational spectra determined in DNA repair
mutants defective in the processing of individual lesions
give further support to potential lethality and mutagenicity of those lesions. The disadvantage here is that multiple lesions are present in the DNA at the same time.
Taken together, however, these data have provided the
underpinning for our understanding of the biological

The biological consequences of individual damaged


bases has been deduced by a combination of experimental approaches. Structural studies of individual model
lesions have provided information as to the potential
coding properties of that damage and whether it is structurally distorting to the DNA molecule. In vitro, biochemical studies with lesions randomly (older studies) or
site-specifically into template DNA molecules have been
employed to assess their interactions with various DNA
polymerases (for reviews see [1,2]). If the lesion is
structurally distorting and blocks the progression of the
DNA polymerase molecule, it is taken as a potentially
This article is part of a series of reviews on Oxidative DNA
Damage and Repair. The full list of papers may be found on the
homepage of the journal.
Susan S. Wallace, Ph.D., is Professor and Chair of the Department of
Microbiology and Molecular Genetics at the University of Vermont.
Address correspondence to: Dr. Susan S. Wallace, Department of
Microbiology and Molecular Genetics, The Markey Center for Molecular Genetics, The University of Vermont, 95 Carrigan Drive, Stafford
Hall, Burlington, VT 05405-0068, USA; Tel: (802) 656-2164; Fax:
(802) 656-8749; E-Mail: swallace@zoo.uvm.edu.
1

S. S. WALLACE

consequences of free radical-induced DNA base damages.


FREE RADICAL-DAMAGED THYMINE BASES

Historically the most well-studied oxidized pyrimidine base is 5,6-dihydro-5,6-dihydroxythymine (thymine


glycol). Thymine glycol is the most common thymine
lesion found after treatment by oxidizing agents such as
hydrogen peroxide, ionizing radiation, potassium permanganate, and osmium tetroxide [79]. Thymine glycol
exerts significant distortion on the duplex DNA molecule. Proton NMR spectroscopy on a duplex oligonucleotide containing thymine glycol shows that the base
portion of thymine glycol and the adenine opposite appear to be extrahelical [10]. Molecular dynamics simulations of 5-hydroxy-5,6-dihydrothymine, 6-hydroxy-5,6dihydrothymine, 5,6-dihydrothymine (dihydrothymine),
and thymine glycol support the idea that the hydroxy group
at the 5 position causes a pseudo-axial oriented methyl
group that results in distortion of the base pair 5 to both the
thymine glycol and the 5-hydroxy-5,6-dihydrothymine [11,
12]. In fact, primer extension studies with several DNA
polymerases using a template containing thymine glycol
show that extension occurs until the lesion site with adenine
inserted opposite the thymine glycol [1316]. In the presence of proofreading, extension can proceed no further,
supporting the idea that it is the base pair 5 to the thymine
glycol that is disturbed and suggesting that thymine glycol
is a lethal lesion. Further support for the conclusion from
the molecular dynamics calculations comes from studies
showing that 5,6-dihydro-5-hydroxythymidine is also a
strong block to Escherichia coli DNA polymerase I Klenow
fragment (exo-) [17]. As predicted from the biochemical
studies, thymine glycol is lethal having an inactivation
efficiency of one in biologically active single-stranded
DNA [3,5]; thymine glycol is also lethal in duplex DNA
[4,18]. There are, however, several sequence contexts
where thymine glycol is bypassed by certain DNA polymerases in vitro [16,19] and in vivo [20].
Because thymine glycol pairs with adenine it is a poor
premutagenic lesion. In SOS induced E. coli where translesion synthesis is upregulated, no mutations are found at
thymine glycol sites [5]. However, when thymine glycol
is inserted into a bypass sequence, a low level of misincorporation of G opposite thymine glycol occurs, giving
rise to mutation [20].
Another ring saturation product of thymine, dihydrothymine, has been well studied. Dihydrothymine does
not inhibit DNA polymerases [21] and it correctly pairs
with cognate adenine; accordingly it is neither lethal nor
mutagenic [6].
The 5-methyl oxidation products, 5-hydroxymethyluracil (5-HMU) [9,22,23] and 5-formyluracil (5-fU) [24]

are also formed by free radicals in DNA following


gamma irradiation as well as during Fenton reactions
[24,25]. X ray crystallographic and NMR studies of
DNA containing 5-HMU show it to assume normal Bform geometry when paired with A [26]. Although the
global conformation of the duplexes is normal and no
distortion is observed, the pucker at the C1 position of
the 2-deoxyribose is changed to exo from the normal
endo pucker [27]. Despite this, 5-HMU pairs correctly in
vitro and is a poor premutagenic lesion [28]. DNA polymerases are not blocked by 5-fU; however, misincorporation of guanine [29] and cytosine [30] opposite the
lesion is observed. When placed into a plasmid vector
and assayed in E. coli, 5-fU causes targeted T 3 C
transitions and T 3 A transversions [31].
DNA containing thymine ring fragmentation, contraction, or ring-open products of thymine have been used as
a template for DNA polymerases. Urea, a fragmentation
product of oxidized thymine, like sites of base loss, lacks
coding and stacking. NMR analysis of a duplex dodecamer containing urea opposite thymine shows that both
are stacked into the helix [32]. In vitro studies with DNA
polymerases have shown that urea is a poorly coding and
stacking lesion and a strong block to DNA synthesis
[13]. In keeping with this observation, urea is also lethal
[18]. When urea is bypassed, purines are the preferred
inserted bases in vitro [21] and mutational studies of urea
in SOS-induced E. coli show that guanine is the major
residue inserted [33]. The ring-open product of dihydrothymine, -ureidoisobutyric acid [21,34], and thymidine
C5-hydrate [34] are also strong blocks to DNA polymerases. Purines and T are inserted opposite the DHT
ring-open product when bypassed in vitro [21] and T 3
A transversions predominate in SOS-induced E. coli
[33]. The phosphoramidite of 5-hydroxy-5-methylhydantoin (hydantoin) was recently synthesized, site specifically incorporated into an oligonucleotide and used as a
template for DNA polymerases [35]. With the three
polymerases tested, E. coli DNA polymerase I Klenow
fragment, Taq DNA polymerase, and Pol , hydantoin
was a strong blocking lesion and with Kf and Taq, dAMP
was the preferred inserted base.
REPAIR OF THYMINE LESIONS

Removal of free radical-damaged thymine products is


initiated by a DNA glycosylase, the first enzyme in the
base excision repair pathway (see [36,37] for reviews).
In vitro, the thymine ring saturation products, the ring
fragmentation, contraction, and ring-open products of
DNA thymine are recognized and removed by the products of the E. coli nth gene (endonuclease III)[38 48],
the nei gene (endonuclease VIII) [49,50], and to a much
lesser extent, the fpg gene (formamidopyrimidine DNA

Consequences of oxidized DNA bases

glycosylase, also called MutM) [35,45,46,51], although


hydantoin and the ring-open form of thymidine C5-hydrate
are efficiently removed by Fpg [35,46]. The ring fragmentation products can also be directly recognized by several
apurinic (AP) endonucleases [52], and thymine glycol is a
substrate for the UvrABC nucleotide [53]. 5-fU paired with
A is removed by AlkA [54,55]; mammalian cells contain a
glycosylase that recognizes 5-HMU [56].
E. coli devoid of both Nth and Nei are spontaneous
mutators [57], but none of the mutations observed are
targeted to thymine, which is in keeping with the observation that the majority of free radical-damaged thymine
bases still correctly pair with adenine. A nth nei fpg alkA
quadruple mutant does enhance mutations in a 5-fUcontaining plasmid [58]. Double nth nei mutants, however, are sensitive to the lethal effects of ionizing radiation and hydrogen peroxide [57,59], which also agrees
with the in vitro studies demonstrating that thymine
glycols, and the thymine breakdown products, are distorting, replication blocking, and lethal lesions. The nth
and nei genes are transcribed as the terminal genes in
operons containing apparently unrelated genes [60], but
do not appear to be induced by agents that produce the
damages the enzymes recognize, although they are induced by a shift from anaerobic to aerobic growth [61].
There are two Saccharomyces cerevisiae Nth orthologs that recognize thymine products [62,63], NTG1,
a mitochondrial activity, and NTG2, primarily present in
the nucleus [64]. There are conflicting data in the literature as to the phenotypes of S. cerevisiae devoid of Ntg1
and Ntg2. In one study [65] double mutants appear to be
spontaneous mutators and sensitive to hydrogen peroxide, while another study [66] shows the double mutants
to have no observable phenotype unless in combination
with mutants defective in recombination, nucleotide excision repair, or translesion synthesis. NTG1 is induced
by menadione [67] and repair of thymine glycol in yeast
has been shown to be coupled to transcription [68]. The
Schizosaccharomyces pombe Nth has a similar substrate
specificity to its orthologs in S. cerevisiae [69,70].
Mammalian Nth orthologs from bovine, mouse, and
human have also been cloned and studied [48,7177].
Several studies have reported human NTH1 to localize
both to the nucleus and the mitochondria [75,78], while
another showed hNTH1 [79] to be exclusively sorted to
the nucleus of HeLa cells and to be cell cycle regulated.
hNTH1 transcription does not appear to be induced by
low doses of ionizing radiation (Inoue and Wallace,
unpublished observations).
FREE RADICAL-DAMAGED CYTOSINE BASES

The major product resulting from hydroxyl radical


attack on DNA cytosine is cytosine glycol, which is

highly unstable and readily deaminates to form uracil


glycol or dehydrates to form 5-hydroxycytosine
(5-OHC) [25,80,81]. Uracil glycol can also dehydrate to
form 5-hydroxyuracil (5-OHU). These latter three products are the major stable free radical-damaged cytosine
products. Several studies have shown that uracil glycol,
5-OHC, and 5-OHU are readily bypassed by DNA polymerases [82,83]. The uracil derivatives, uracil glycol and
5-OHU always pair with adenine, and thus are potentially potent premutagenic lesions [82,83]. 5-OHC pairs
with cognate guanine as well as with adenine and to a
much lesser extent, cytosine [82]. Because they are bypassed by DNA polymerases the oxidized cytosines
should not be lethal lesions. Again, the in vitro studies
with DNA polymerases predict the in vivo results. Studies with single-stranded DNA containing a unique 5-hydroxyuracil or uracil glycol show these lesions not to be
cytotoxic and to induce C 3 T transitions at very high
efficiencies [84]. 5-hydroxycytosine, which is also not
cytotoxic, is much less mutagenic than the uracil derivatives giving C 3 T transitions as well as some C 3 G
transversions [84,85].

REPAIR OF CYTOSINE LESIONS

Free radical-damaged cytosines are repaired by base


excision repair and are recognized by the same glycosylases that remove free radical-damaged thymines, in E.
coli Nth and Nei (for reviews see [86,87]). Fpg also
recognizes 5-OHC and 5-OHU in vitro [45]. Double
mutants deficient in both Nth and Nei are strong spontaneous mutators, about 20-fold above background and
all of the observed mutations are C 3 T transitions
[57,88]. This is in keeping with the substrate specificities
of Nth and Nei, which recognize and remove premutagenic cytosine lesions from DNA. A cellular role for Fpg
in the repair of premutagenic oxidized cytosines is not
supported by the mutation spectrum of nth nei fpg triple
mutants where no increase in C 3 T transitions is found
over that observed in nth nei double mutants (Blaisdell
and Wallace, unpublished observations). As described
above, yeast mutants defective in the Nth orthologs that
recognize oxidized cytosines [67] are at best mild mutators [65]. It should be noted that C 3 T transitions are
one of the major mutational events found in the oxidative
mutational spectra.

CONSEQUENCES OF FREE RADICAL-DAMAGED


PYRIMIDINES

Table 1 summarizes the in vitro and in vivo properties


of representative free radical-damaged thymines and cytosines, and Fig. 1 gives their structures. Of the thymine

S. S. WALLACE
Table 1. Consequences of Representative Free Radical Damaged Pyrimidinesa

Lesion
Thymine glycol
Dihydrothymine
5-Hydroxymethyluracil
5-Formyluracil
Urea
-Ureidoisobutyric acid
Hydantoin
Uracil glycol
5-Hydroxycytosine
5-Hydroxyuracil
Dihydrouracil

Block to DNA
polymerases

Lethal

Base inserted opposite


in vitro

Mutagenic

Yes
No
No
No
Yes
Yes
Yes
No
No
No
No

Yes
No
No
No
Yes
Yes
n.d.
No
No
No
n.d.

AG
A
A
AGC
AG
AGT
AG
A
GA
A
A

Poor
No
Poor
T3C, T3A
T3C
T3A
n.d.
C3T
C3T, C3G
C3T
n.d.

Referenced in text.
n.d. not determined.

moieties that retain intact ring structure, only 5-fU mispairs in vitro and in vivo. When spontaneous mutagenesis is assessed in E. coli lacking the oxidative DNA
glycosylases that recognize free radical-damaged thymines, no mutations at thymine sites are observed. In fact,
mutants devoid of the three oxidative DNA glycosylases,
nth, nei fpg, do not show any mutations at T (Blaisdell
and Wallace; unpublished), even though FPG can also
recognize thymine lesions in vitro. Quadruple mutants,
nth nei fpg alkA, do show an increase in mutations in a
plasmid containing 5-fU in keeping with the in vitro
observation that 5-fU is a substrate for AlkA. See Table
2 for a list of the oxidative DNA glycosylases and their
principal substrates.
When bypassed, purines, principally A, are inserted opposite the thymine ring fragmentation, ring open, and contraction products, most often producing a nonmutagenic
pair. Also, the rate of production of the thymine breakdown products during normal metabolism is considered to
be very low. It is possible that the breakdown products of
thymine might play an as yet undetermined role in spontaneous mutagenesis because in addition to being recognized
by the above-mentioned DNA glycosylases, urea is recognized by the AP endonucleases. Taken together, however,
these data support the idea that oxidized thymines do not
play an important role in mutation induction. On the other
hand, thymine glycol and the thymine breakdown products
are lethal. This is supported by studies with nth nei double
mutants showing them to be hypersensitive to agents that
produce these lesions in DNA. In contrast to free radical
damaged thymine, the cytosine derivatives that retain their
ring structure are all readily bypassed, miscode, and are
potent premutagenic lesions. This is supported by the observation that E. coli mutants devoid of the activities that
recognize oxidized cytosines are mutators with all of the
mutations targeted at cytosine.
In eukaryotes redundant systems apparently play an
important role in processing oxidized pyrimidines. For

example, in S. cerevisiae, mutants defective in the pyrimidine-specific DNA glycosylases do not have strong
phenotype [66]. It remains to be determined what role the
oxidized pyrimidine lesions play in mutagenesis and
carcinogenesis in mammalian systems.
FREE RADICAL-DAMAGED GUANINE BASES

The eighth position on the imidazoyl ring of guanine is


the most readily oxidized leading to the formation of 7,8dihydro-8-oxoguanine (8-oxoG). 8-oxoG has been found in
DNA following treatment with various oxidizing agents,
metals, and ionizing radiation, and is commonly used as a
biomarker of oxidative stress [89,90]. The 8-oxoG adduct
has been extensively structurally characterized both by
NMR and x-ray crystallography. When paired with cytosine, the overall duplex structure is very similar to an
unmodified duplex with the 8-oxoguanine, assuming an
anti confirmation and appropriately base paired to cytosine
[91,92]. Analysis of 8-oxoG opposite adenine shows the
8-oxoG to assume a syn conformation forming a stable
Hoogsteen base pair with adenine in the anti alignment
[93,94]. These structural observations provide the framework for both biochemical studies with DNA polymerases
and the mutagenic analysis of DNA containing 8-oxoG.
Like the oxidized cytosines studied thus far, 8-oxoguanine is readily bypassed by all DNA polymerases
tested [9598]. Depending on which polymerase is used,
there is preferential insertion of cognate C or the noncognate A. Thus the in vitro data predict that because of
its ability to mispair with A, 8-oxoguanine would not be
a lethal lesion but a mutagenic lesion. Interestingly, the
replicative polymerases tested showed significant incorporation of dATP opposite 8-oxoG, while the repair
polymerases preferentially insert dCTP [96].
In keeping with its ability to mispair with A in vitro, an
8-oxoG residue site specifically introduced into a plasmid
vector and replicated in E. coli gives rise exclusively to G

Consequences of oxidized DNA bases

Fig. 1. Structures of the representative pyrimidine base lesions described in Table 1.

3 T transversions at the site of the adduct [95,97,98]. In


HeLa cells a site-specific 8-oxoG adduct in a doublestranded shuttle vector also induced targeted G 3 T transversions [99]. Similar results were found with the singlestranded shuttle vector in COS cells [100]. Of interest, a
sequence context surrounding 8-oxoG that reduced Fpg
cleavage and promoted mispairing with A in vitro was
recapitulated surrounding G 3 T transversions in bacterial
and human oxidative DNA databases [101].
Ionizing radiation also produces the imidazoyl ring frag-

mentation product of guanine 2,6-diamino-4-hydroxy-5formamidopyrimidine (Fapy-G). Although the Fapy-G


product has not been synthesized, methyl Fapy-G has been
examined in vitro and shown to be a strong block to DNA
polymerases [102], and a lethal lesion [103].
REPAIR OF GUANINE LESIONS

Protection of E. coli cells from the potential mutagenic effects of 8-oxoG is multifaceted [104]. E. coli

S. S. WALLACE
Table 2. Oxidative DNA Glycosylases and their Preferred Natural
Substrates

Glycosylase
Nth (endo III) orthologs
MutY orthologs
Ogg orthologs
Fpg orthologs
Nei (endo VIII) orthologs

Principal substrate
Oxidized pyrimidines, Fapy-G
A 8-oxoG
8-oxoG C
8-oxoG C
Oxidized pyrimidines, Fapy-G

formamidopyrimidine DNA glycosylase (Fpg, MutM)


efficiently removes 8-oxoG from DNA when paired with
C. If this repair does not take place prior to DNA replication, A may be inserted opposite the 8-oxoG. To
counteract this potentially mutagenic event, the MutY
protein removes the A mispaired with 8-oxoG. To further
avoid any potential mutations from 8-oxoG adducts, the
MutT protein hydrolyzes 8-oxodGTP in the nucleotide
pools to 8-oxodGMP preventing it from being incorporated opposite A during DNA replication. E. coli cells
defective in Fpg are not sensitive to oxidizing agents but
are mild mutators [105,106]. Addition of the mutY mutation to fpg mutants confers a strong spontaneous mutator phenotype, about 400-fold over that of wild type
[106]. Biologically active DNA exposed extracellularly
to ionizing radiation also shows an increase in G 3 T
transversions in fpg and fpg mutY mutants [107109].
Interestingly, addition of the nei mutation (endo VIII) to
the fpg mutY double mutant confers an additional 3-fold
increase in spontaneous mutation frequency [88]. Single
nei mutants have no observable phenotype and when the
nei mutation is combined with an nth mutation, all the
spontaneous mutations are at cytosines [88]. However, in
the absence of Fpg and MutY, Nei must be able to
remove some potentially premutagenic 8-oxoG lesions
from DNA. In vitro, endo VIII does have activity against
8-oxoG paired with C, albeit weak [88]. (Others have
observed Nei activity on 8-oxoG paired with A [110], but
this in vitro observation is not consistent with the biological result that in the absence of MutY, Fpg and Nei,
all the mutations are G 3 T transversions.) Methyl
Fapy-G is removed by Fpg [111,112], Ogg1, the eukaryotic functional homolog of Fpg [113115], and is a
substrate for the Nth orthologs and Nei [116].
In E. coli, fpg and mutY are transcribed as part of
operons containing genes apparently unrelated to repair
of oxidative damage [117]. Using RNAase protection,
fpg and mutY transcripts were shown not to be induced
by DNA-damaging agents [61], but are increased after
shift from anaerobic to aerobic growth [61,118]. Fpg
activity does appear to be downregulated by metal chelators [119]; however, no differences in fpg and mutY
transcript levels are found in arcA, fnr and fur mutants
[61].

Fpg is found across the bacterial kingdom but is


absent in the Archaea and Eukaryota, with the exception
of the yeast Candida albicans and the plant Arabidopsis
thaliana [120,121]. In eukaryotic cells, the primary activity that removes 8-oxoG from DNA is not an ortholog
of Fpg protein, but rather a member of the Nth superfamily. This protein, called Ogg1, is found across the
eukaryotic kingdom and has a similar substrate specificity to Fpg, that is, it preferentially removes 8-oxoG
opposite C as well as the Fapy products [113115,122].
Like Fpg in E. coli, Ogg in Saccharomyces cerevisiae is
not essential [123]. S. cerevisiae Ogg1 mutants are not
sensitive to the oxidizing agent hydrogen peroxide, but
they do exhibit a mutator phenotype and accumulate G
3 T transversions [123]. Interestingly the yeast S. cerevisiae does not possess an ortholog of the MutY protein,
and 8-oxoG-A mispairs formed during replication are
processed by mismatch repair [124]. However, a MutY
ortholog is present in the yeast Schizosaccharomyces
pombe.
Interestingly, in Drosophila melanogaster, the ribosomal protein S3 has both DNA glycosylase activity
against 8-oxoG as well as an AP lyase activity [125]. S3
also catalyzes the release of Fapy-G from gamma-irradiated DNA [126] and expression of S3 protein in E. coli
fpg mutants suppresses the spontaneous mutator phenotype [125]. Drosophila also have a functional Ogg protein [127].
Orthologs of the yeast Ogg1 are also found in mammalian cells [128 136]. -hOGG1 appears to localize to
the nucleus while -hOGG1 localizes to mitochondria
[78]. Ogg1 activity has been shown to be induced in rat
kidneys by potassium bromate, a renal oxidative carcinogen [137], by peroxisome proliferators [138], crocidolite asbestos [139], and upon administration of diesel
exhaust particles [140]. Increased Ogg activity has also
been demonstrated in ischemic reperfused mouse brain
[141] and rat hearts [142], hOGG1 transcripts are not
induced by low doses of ionizing radiation (Inoue and
Wallace, unpublished observations) and expression of
hOGG1 does not appear to vary during cell cycle [143].
These data suggest that OGG is a housekeeping enzyme
that is induced under some conditions of oxidative stress.
The hOGG1 gene is localized to chromosome 3p26.2
[132], a frequent site of loss of heterozygosity in many
types of human tumors, including clear cell carcinomas
of the kidney [144], head and neck cancers [145], and
small cell lung carcinomas [146,147]. In carcinomas of
the kidney, loss of heterozygosity for the Ogg gene
occurs in 85% of the cases examined [144]. A number of
mutations are also found in the hOGG1 gene of tumor
cells [144,147]. The most common variant amino acid
substitution is Ser326Cys, which does not alter the activity of hOGG1 [148]. Rarer polymorphisms including

Consequences of oxidized DNA bases

substitution of glutamine for arginine at position 46 and


histidine for arginine at position 154 do lead to hOGG1
activity that is less efficient and selective than that of
wild type [149]. Interestingly, a leukemic cell line,
KG-1, has a homozygous Arg209Glu mutation that renders hOGG1 nonfunctional [150]. Mice nullizygous for
Ogg1 exhibit only a mild spontaneous mutator phenotype in certain tissues [151,152], which is similar to the
mutator phenotype observed in ogg mutant yeast and fpg
mutant bacteria.
Orthologs of the MutY protein MYH [153155] are
found in mammalian cells. Human MYH is sorted to the
nucleus and mitochondria as determined by alternative
splicing [156 158]. However, the mice nullizygous for
Myh and both OGG1 and MYH also appear to have no
significant phenotype [159], suggesting that alternative
routes for repairing this potent premutagenic lesion exist
in mammalian cells.
FREE RADICAL-DAMAGED ADENINE BASES

The 8 position on the imidazoyl ring of adenine is also


susceptible to oxidation to 7,8-dihydro-8-oxoadenine (8oxoA). 8-oxoA has been observed in DNA treated by
ionizing radiation and other oxidizing agents [160 162].
Structural studies show that an 8-oxoA paired with T
causes little disturbance of a duplex nonomer and remains in the anti position [163]. In crystals, 8-oxoA
assumes the syn position paired with G anti [164]. In
vitro studies with DNA polymerase I Klenow exo and
Taq DNA polymerase show that 8-oxoA is not a blocking lesion and that thymine is exclusively incorporated
opposite 8-oxoA [163]. In keeping with these observations with bacterial polymerases, 8-oxoA site specifically
incorporated into a phage genome and replicated in E.
coli cells is not cytotoxic or mutagenic [165]. However
with mammalian DNA polymerase , some dGTP is
incorporated opposite 8-oxoA and pol can incorporate
both dGTP and dATP [166,167]. Single 8-oxoA lesions
have been shown to cause A 3 G and A 3 C mutations
in mammalian cells [168].
2-Hydroxyadenine (2-OHA) is observed in DNA of
cells oxidized by metals or ionizing radiation [169 172].
Primer extension studies using templates containing
2-OHA show it to be bypassed and all bases to be
misincorporated [173,174]. With single- and doublestranded vectors containing a single 2-OHA lesion and
replicated in E. coli no cytotoxicity is observed, but A 3
T and A 3 G substitutions are found [175]. With an
SV40 vector containing a single 2-OHA transfected into
COS-7 cells, a targeted 1 deletion was the most frequent mutation observed followed by A 3 G transitions
[176].
4,6-Diamino-5-formamidopyrimidine (Fapy-A) is

readily produced by oxidizing agents and ionizing radiation [177179]. Fapy-A blocks DNA synthesis in vitro
by T7 DNA polymerase, the Klenow fragment of DNA
pol I and polymerase from calf thymus, and thus is a
potentially lethal lesion [180]. -Deoxyadenosine (dA) is a major adenine lesion produced by ionizing
radiation [181] and is a moderate block to DNA synthesis in vitro [182] and in vivo [183]. Targeted mutations
are all one base deletions [183], suggesting that -dA
does not remain stacked in the helix.

REPAIR OF ADENINE LESIONS

8-oxoA is a relatively poor substrate for Fpg [184]


and endo VIII (Dizdaroglu and Wallace unpublished
observations), however, it is efficiently removed by S.
cerevisiae Ogg1 when paired with C [185], which is not
the biological substrate. It may not be important to have
efficient repair of 8-oxoA since it is readily bypassed by
polymerases and most often pairs with T; it is not a lethal
lesion or mutagenic lesion in E. coli and only a weak
mutagenic lesion in mammalian cells.
Little is known about the repair of 2-OHA although
activity against this lesion was found in rat organs [186].
Also, hMYH removes 2-OHA paired with G [157].
Fapy-A, a cytotoxic lesion, is recognized by Fpg protein
and presumably by OGG1 and some orthologs of Nth.

CONSEQUENCES OF FREE
RADICAL-DAMAGED PURINES

Table 3 summarizes the properties of the most wellstudied free radical-damaged purines and Fig. 2 gives
their structures. Of these, 8-oxoG has been the most
frequently studied and is an important premutagenic lesion causing G 3 T transversions. Both E. coli and S.
cerevisiae mutants devoid of the enzymes that process
8-oxoG are strong spontaneous mutators. OGG1 that
removes 8-oxoG is induced in mammalian cells in response to a number of oxidative stressors. Also polymorphisms and loss of heterozygosity of hOGG1 have been
observed in a number of human tumors. However, mice
lacking the glycosylases that process 8-oxoguanine do
not appear to exhibit a major phenotype, although they
do accumulate small numbers of 8-oxoG in the DNA of
certain tissues. With the exception of -adenine, the
oxidized adenines that retain their ring structure primarily pair with cognate T, although the 2-oxoadenine has
been shown to be mutagenic. Both Fapy-G and Fapy-A
are polymerase-blocking lesions; Fapy-G is lethal but not
mutagenic.

S. S. WALLACE
Table 3. Consequences of Free Radical-Damaged Purinesa

Lesion

Block to DNA
polymerases

Lethal

Base inserted opposite


in vitro

Mutagenic

No
Yes
No
No
Yes
Yes

No
Yes
No
No
Yes
n.d.

CA
n.d.
TG
TA
Deletion
n.d.

G3T
No
Poor
A3T, A3G
1 deletion
n.d.

8-Oxoguanine
Fapy-G
8-Oxoadenine
2-Oxoadenine
-Adenine
Fapy-A
a

Referenced in text.
n.d. not determined.

FREE RADICAL DAMAGES OCCURING IN CLUSTERS

In contrast to the free radical damage produced in DNA


from the byproducts of oxidative metabolism, which are
distributed along the DNA molecule, ionizing radiation
produces a burst of hydroxyl radicals during the radiolysis
of water, that can form damage clusters in both DNA
strands [187,188]. When DNA molecules containing
closely opposed clustered base damages are used as substrates for the oxidative DNA glycosylases that recognize
them, [189 193], cleavage of the first strand containing two
opposed damages, is relatively unaffected by the position of
the opposing lesion and a single strand break is formed by
the enzyme that recognizes the damage in the first strand.
However, once a strand break is introduced into one strand
by a DNA glycosylase, cleavage of the damage in the
second strand depends on the position of the opposing
strand break. The excision of the second strand usually
occurs if the opposing strand break is more than one base
away. If excision of both strands does take place, a double
strand break results. In a completely reconstituted base
excision repair system [191] repair of individual strands

usually does not happen, rather a double strand break is


formed depending on whether the DNA glycosylase can
cleave the damage-containing strand closely opposed to the
strand break. These biochemical studies predicted that cells
devoid of the oxidative DNA glycosylases would be radioresistant, whereas cells overproducing an oxidative DNA
glycosylase would be radiosensitive. This was verified by
showing that E. coli triple mutant cells devoid of Nth, Nei,
and Fpg are about twice as radioresistant as wild-type cells
and that this increase in radioresistance could be attributed
to the reduced formation of double strand breaks [194].
Furthermore, the production of the additional double strand
breaks in wild-type cells occurs postirradiation, presumably
due to incisions at closely opposed base damages by the
oxidative DNA glycosylases. Also, cells overproducing
Fpg are radiosensitive and this increase in radiosensitivity is
correlated with an increased formation of double strand
breaks postirradiation. Thus abortive base excision repair
processing of closely opposed free radical-damaged DNA
bases can lead to the formation of lethal double strand
breaks.

Fig. 2. Structures of the representative purine base lesions described in Table 3. -Adenine, not shown, is the anomer of
deoxyadenosine.

Consequences of oxidized DNA bases


CONCLUSIONS

Study of the biological consequences of individual


free radical DNA lesions has been greatly facilitated by
their chemical synthesis and incorporation into DNA
molecules for use as enzyme substrates or as templates
for DNA polymerases. For the lesions studied so far, the
in vitro studies have predicted the cellular results which,
in turn, have been substantiated by studies with DNA
repair defective mutants.

[16]

Acknowledgements Work from the authors laboratory is supported


by NIH R37 CA 33657, R01 CA52040, awarded by the National
Cancer Institute, R01 AG17101-01 awarded by the National Institute
on Aging, and DE-FG02-99ER62861 awarded by the U.S. Department
of Energy. The author wishes to thank past and present students,
postdoctoral associates, and research staff members.

[20]

REFERENCES
Hatahet, Z.; Wallace, S. S. Translesion DNA synthesis. In:
Nickoloff, J. A.; Hoekstra, M. F., eds. DNA damage and repair,
vol. 1: DNA repair in prokaryotes and lower eukaryotes. Totowa, NJ: Humana Press, Inc.; 1998:229 262.
Wang, D.; Kreutzer, D. A.; Essigmann, J. M. Mutagenicity and
repair of oxidative DNA damage: insights from studies using
defined lesions. Mutat. Res. 400:99 115; 1998.
Achey, P. M.; Wright, C. F. Inducible repair of thymine ring
saturation damage in X174 DNA. Radiat. Res. 93:609 612;
1983.
Moran, E.; Wallace, S. S. The role of specific DNA base damages in the X-ray-induced inactivation of bacteriophage PM2.
Mutat. Res. 146:229 241; 1985.
Hayes, R. C.; Petrullo, L. A.; Huang, H. M.; Wallace, S. S.;
LeClerc, J. E. Oxidative damage in DNA. Lack of mutagenicity
by thymine glycol lesions. J. Mol. Biol. 201:239 246; 1988.
Evans, J.; Maccabee, M.; Hatahet, Z.; Courcelle, J.; Bockrath,
R.; Ide, H.; Wallace, S. Thymine ring saturation and fragmentation products: lesion bypass, misinsertion and implications for
mutagenesis. Mutat. Res. 299:147156; 1993.
Frenkel, K.; Goldstein, M. S.; Teebor, G. W. Identification of the
cis-thymine glycol moiety in chemically oxidized and gammairradiated deoxyribonucleic acid by high-pressure liquid chromatography analysis. Biochemistry 20:7566 7571; 1981.
Breimer, L. H.; Lindahl, T. Enzymatic excision of DNA bases
damaged by exposure to ionizing radiation or oxidizing agents.
Mutat. Res. 150:85 89; 1985.
Teoule, R. Radiation-induced DNA damage and its repair. Int. J.
Radiat. Biol. Relat. Stud. Phys. Chem. Med. 51:573589; 1987.
Kao, J. Y.; Goljer, I.; Phan, T. A.; Bolton, P. H. Characterization
of the effects of a thymine glycol residue on the structure,
dynamics, and stability of duplex DNA by NMR. J. Biol. Chem.
268:1778717793; 1993.
Miller, J.; Miaskiewicz, K.; Osman, R. Structure-function studies of DNA damage using ab initio quantum mechanics and
molecular dynamics simulation. Ann. N. Y. Acad. Sci. 726:71
91; 1994.
Miaskiewicz, K.; Miller, J.; Ornstein, R.; Osman, R. Molecular
dynamics simulations of the effects of ring-saturated thymine
lesions on DNA structure. Biopolymers 35:113124; 1995.
Ide, H.; Kow, Y. W.; Wallace, S. S. Thymine glycols and urea
residues in M13 DNA constitute replicative blocks in vitro.
Nucleic Acids Res. 13:8035 8052; 1985.
Rouet, P.; Essigmann, J. M. Possible role for thymine glycol in
the selective inhibition of DNA synthesis on oxidized DNA
templates. Cancer Res. 45:6113 6118; 1985.
Clark, J. M.; Beardsley, G. P. Thymine glycol lesions terminate

[22]

[1]

[2]
[3]
[4]
[5]
[6]

[7]

[8]
[9]
[10]

[11]

[12]
[13]
[14]
[15]

[17]

[18]

[19]

[21]

[23]
[24]

[25]

[26]

[27]

[28]

[29]

[30]

[31]

[32]

[33]

[34]

chain elongation by DNA polymerase I in vitro. Nucleic Acids


Res. 14:737749; 1986.
Hayes, R. C.; LeClerc, J. E. Sequence dependence for bypass of
thymine glycols in DNA by DNA polymerase I. Nucleic Acids
Res. 14:10451061; 1986.
Greenberg, M. M.; Matray, T. J. Inhibition of klenow fragment
(exo-) catalyzed DNA polymerization by (5R)-5,6-dihydro-5hydroxythymidine and structural analogue 5,6-dihydro- 5-methylthymidine. Biochemistry 36:1407114079; 1997.
Laspia, M. F.; Wallace, S. S. Excision repair of thymine glycols,
urea residues, and apurinic sites in Escherichia coli. J. Bacteriol.
170:3359 3366; 1988.
Clark, J.; Beardsley, G. Template length, sequence context, and
3-5 exonuclease activity modulate replicative bypass of thymine glycol lesions in vitro. Biochemistry 28:775779; 1989.
Basu, A. K.; Loechler, E. L.; Leadon, S. A.; Essigmann, J. M.
Genetic effects of thymine glycol: site-specific mutagenesis and
molecular modeling studies. Proc. Natl. Acad. Sci. USA 86:
76777681; 1989.
Ide, H.; Petrullo, L. A.; Hatahet, Z.; Wallace, S. S. Processing of
DNA base damage by DNA polymerases. Dihydrothymine and
-ureidoisobutyric acid as models for instructive and noninstructive lesions. J. Biol. Chem. 266:1469 1477; 1991.
Frenkel, K.; Cummings, A.; Solomon, J.; Cadet, J.; Steinberg,
J. J.; Teebor, G. W. Quantitative determination of the 5-(hydroxymethyl)uracil moiety in the DNA of -irradiated cells.
Biochemistry 24:4527 4533; 1985.
Hagen, U. Current aspects on the radiation induced base damage
in DNA. Radiat. Environ. Biophys. 25:261271; 1986.
Kasai, H.; Iida, A.; Yamaizumi, Z.; Nishimura, S.; Tanooka, H.
5-Formyldeoxyuridine: a new type of DNA damage induced by
ionizing radiation and its mutagenicity to Salmonella strain
TA102. Mutat. Res. 243:249 253; 1990.
Douki, T.; Delatour, T.; Paganon, F.; Cadet, J. Measurement of
oxidative damage at pyrimidine bases in -irradiated DNA.
Chem. Res. Toxicol. 9:11451151; 1996.
Mellac, S.; Fazakerley, G.; Sowers, L. Structures of base pairs
with 5-(hydroxymethyl)-2-deoxyuridine in DNA determined by
NMR spectroscopy. Biochemistry 32:7779 7786; 1993.
Birnbaum, G. I.; Deslauriers, R.; Lin, T. S.; Shiau, W. H.;
Prusoff, A. A novel intramolecular hydrogen bond in the crystal
structure of 5-hydromethyl-2-deoxyuridine, an antiviral and
antineoplastic nucleoside. Conformational analysis of the deoxyribose ring. J. Am. Chem. Soc. 102:4236 4241; 1980.
Levy, D. D.; Teebor, G. W. Site directed substitution of 5-hydroxymethyluracil for thymine in replicating X-174am3 DNA
via synthesis of 5-hydroxymethyl-2-deoxyuridine-5-triphosphate. Nucleic Acids Res. 19:33373343; 1991.
Masaoka, A.; Terato, H.; Kobayashi, M.; Ohyama, Y.; Ide, H.
Oxidation of thymine to 5-formyluracil in DNA promotes misincorporation of dGMP and subsequent elongation of a mismatched primer terminus by DNA polymerase. J. Biol. Chem.
276:1650116510; 2001.
Zhang, Q. M.; Sugiyama, H.; Miyabe, I.; Matsuda, S.; Saito, I.;
Yonei, S. Replication of DNA templates containing 5-formyluracil, a major oxidative lesion of thymine in DNA. Nucleic Acids
Res. 25:3969 3973; 1997.
Miyabe, I.; Zhang, Q.-M.; Sugiyama, H.; Kino, K.; Yonei, S.
Mutagenic effects of 5-formyluracil on a plasmid vector during
replication in Escherichia coli. Int. J. Radiat. Biol. 77:5358;
2001.
Gervais, V.; Guy, A.; Teoule, R.; Fazakerley, G. V. Solution
conformation of an oligonucleotide containing a urea deoxyribose residue in front of a thymine. Nucleic Acids Res. 20:6455
6460; 1992.
Maccabee, M.; Evans, J. S.; Glackin, M. P.; Hatahet, Z.; Wallace, S. S. Pyrimidine ring fragmentation products. Effects of
lesion structure and sequence context on mutagenesis. J. Mol.
Biol. 236:514 530; 1994.
Matray, T. J.; Haxton, K. J.; Greenberg, M. M. The effects of the
ring fragmentation product of thymidine C5-hydrate on phos-

10

[35]

[36]

[37]

[38]

[39]

[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47]

[48]

[49]

[50]

[51]

S. S. WALLACE
phodiesterases and klenow (exo-) fragment. Nucleic Acids Res.
23:4642 4648; 1995.
Gasparutto, D.; Ait-Abbas, M.; Jaquinod, M.; Boiteux, S.; Cadet, J. Repair and coding properties of 5-hydroxy-5-methylhydantoin nucleosides inserted into DNA oligomers. Chem. Res.
Toxicol. 13:575584; 2000.
Wallace, S. S. Oxidative damage to DNA and its repair. In:
Scandalios, J., ed. Oxidative stress and the molecular biology of
antioxidant defenses. Cold Spring Harbor, NY: Cold Spring
Harbor Laboratory Press; 1997:49 90.
Wallace, S. S. Enzymatic processing of radiation-induced free
radical damage in DNA. Radiat. Res. 150(Suppl. 5):S60 S79;
1998.
Armel, P. R.; Strniste, G.; Wallace, S. S. Studies on Escherichia
coli X-ray endonuclease specificity: roles of hydroxyl and reducing radicals in the production of DNA lesions. Radiat. Res.
69:328 338; 1977.
Gates, F. T.; Linn, S. Endonuclease from Escherichia coli that
acts specifically upon duplex DNA damages by ultraviolet light,
osmium tetroxide, acid or X-rays. J. Biol. Chem. 252:2802
2807; 1977.
Breimer, L. H.; Lindahl, T. A DNA glycosylase from Escherichia coli that releases free urea from a polydeoxyribonucleotide containing fragments of base residues. Nucleic Acids Res.
8:6199 6211; 1980.
Katcher, H. L.; Wallace, S. S. Characterization of the Escherichia coli x-ray endonuclease, endonuclease III. Biochemistry
22:4071 4081; 1983.
Breimer, L. H.; Lindahl, T. DNA glycosylase activities for
thymine residues damaged by ring saturation, fragmentation, or
ring contraction are functions of endonuclease III in Escherichia
coli. J. Biol. Chem. 259:55435548; 1984.
Boorstein, R. J.; Hilbert, T. P.; Cadet, J.; Cunningham, R. P.;
Teebor, G. W. UV-induced pyrimidine hydrates in DNA are
repaired by bacterial and mammalian DNA glycosylase activities. Biochemistry 28:6164 6170; 1989.
Dizdaroglu, M.; Laval, J.; Boiteux, S. Substrate specificity of the
Escherichia coli endonuclease III: excision of thymine- and
cytosine-derived lesions in DNA produced by radiation-generated free radicals. Biochemistry 32:1210512111; 1993.
Hatahet, Z.; Kow, Y. W.; Purmal, A. A.; Cunningham, R. P.;
Wallace, S. S. New substrates for old enzymes. 5-Hydroxy-2deoxycytidine and 5-hydroxy-2-deoxyuridine are substrates for
Escherichia coli endonuclease III and formamidopyrimidine
DNA N-glycosylase, while 5-hydroxy-2-deoxyuridine is a substrate for uracil DNA N-glycosylase. J. Biol. Chem. 269:18814
18820; 1994.
Jurado, J.; Saparbaev, M.; Matray, T. J.; Greenberg, M. M.;
Laval, J. The ring fragmentation product of thymidine C5hydrate when present in DNA is repaired by the Escherichia coli
Fpg and Nth proteins. Biochemistry 37:77577763; 1998.
Dizdaroglu, M.; Bauche, C.; Rodriguez, H.; Laval, J. Novel
substrates of Escherichia coli Nth protein and its kinetics for
excision of modified bases from DNA damaged by free radicals.
Biochemistry 39:5586 5592; 2000.
Asagoshi, K.; Odawara, H.; Nakano, H.; Miyano, T.; Terato, H.;
Ohyama, Y.; Seki, S.; Ide, H. Comparison of substrate specificities of Escherichia coli endonuclease III and its mouse homologue (mNTH1) using defined oligonucleotide substrates. Biochemistry 39:11389 11398; 2000.
Melamede, R. J.; Hatahet, Z.; Kow, Y. W.; Ide, H.; Wallace,
S. S. Isolation and characterization of endonuclease VIII from
Escherichia coli. Biochemistry 33:12551264; 1994.
Jiang, D.; Hatahet, Z.; Melamede, R. J.; Kow, Y. W.; Wallace,
S. S. Characterization of Escherichia coli endonuclease VIII.
J. Biol. Chem. 272:32230 32239; 1997.
DHam, C.; Romieu, A.; Jaquinod, M.; Gasparutto, D.; Cadet, J.
Excision of 5,6-dihydroxy-5,6-dihydrothymine, 5,6-dihydrothymine, and 5-hydroxycytosine from defined sequence oligonucleotides by Escherichia coli endonuclease III and Fpg proteins:

[52]

[53]

[54]

[55]

[56]

[57]

[58]

[59]

[60]

[61]

[62]

[63]

[64]

[65]

[66]

kinetic and mechanistic aspects. Biochemistry 38:33353344;


1999.
Kow, Y. W.; Wallace, S. S. Exonuclease III recognizes urea
residues in oxidized DNA. Proc. Natl. Acad. Sci. USA 82:8354
8358; 1985.
Kow, Y. W.; Wallace, S. S.; Van Houten, B. UvrABC nuclease
complex repairs thymine glycol, an oxidative DNA base damage. Mutat. Res. 235:147156; 1990.
Terato, H.; Masaoka, A.; Kobayashi, M.; Fukushima, S.;
Ohyama, Y.; Yoshida, M.; Ide, H. Enzymatic repair of 5-formyluracil. II. Mismatch formation between 5-formyluracil and guanine during DNA replication and its recognition by two proteins
involved in base excision repair (AlkA) and mismatch repair
(MutS). J. Biol. Chem. 274:25144 25150; 1999.
Masaoka, A.; Terato, H.; Kobayashi, M.; Honsho, A.; Ohyama,
Y.; Ide, H. Enzymatic repair of 5-formyluracil. I. Excision of
5-formyluracil site-specifically incorporated into oligonucleotide
substrates by AlkA protein (Escherichia coli 3-methyladenine
DNA glycosylase II). J. Biol. Chem. 274:25136 25143; 1999.
Cannon-Carlson, S. V.; Gokhale, H.; Teebor, G. W. Purification
and characterization of 5-hydroxymethyluracil-DNA glycosylase from calf thymus. Its possible role in the maintenance of
methylated cytosine residues. J. Biol. Chem. 264:13306 13312;
1989.
Jiang, D.; Hatahet, Z.; Blaisdell, J. O.; Melamede, R. J.; Wallace,
S. S. Escherichia coli endonuclease VIII: cloning, sequencing,
and overexpression of the nei structural gene and characterization of nei and nei nth mutants. J. Bacteriol. 179:37733782;
1997.
Zhang, Q. M.; Miyabe, I.; Matsumoto, Y.; Kino, K.; Sugiyama,
H.; Yonei, S. Identification of repair enzymes for 5-formyluracil
in DNA. Nth, Nei, and MutM proteins of Escherichia coli.
J. Biol. Chem. 275:3547135477; 2000.
Saito, Y.; Uraki, F.; Nakajima, S.; Asaeda, A.; Ono, K.; Kubo,
K.; Yamamoto, K. Characterization of endonuclease III (nth)
and endonuclease VIII (nei) mutants of Escherichia coli K-12. J.
Bacteriol. 179:37833785; 1997.
Gifford, C. M.; Wallace, S. S. The genes encoding endonuclease
VIII and endonuclease III in Escherichia coli are transcribed as
the terminal genes in operons. Nucleic Acids Res. 28:762769;
2000.
Gifford, C. M.; Blaisdell, J. O.; Wallace, S. S. Multiprobe RNase
protection assay analysis of mRNA levels for the Escherichia
coli oxidative DNA glycosylase genes under conditions of oxidative stress. J. Bacteriol. 182:5416 5424; 2000.
Eide, L.; Bjrs, M.; Pirovano, M.; Alseth, I.; Berdal, K. G.;
Seeberg, E. Base excision of oxidative purine and pyrimidine
DNA damage in Saccharomyces cerevisiae by a DNA glycosylase with sequence similarity to endonuclease III from Escherichia coli. Proc. Natl. Acad. Sci. USA 93:1073510740; 1996.
Augeri, L.; Lee, Y.-M.; Barton, A. B.; Doetsch, P. W. Purification, characterization, gene cloning, and expression of Saccharomyces cerevisiae redoxyendonuclease, a homolog of Escherichia coli endonuclease III. Biochemistry 36:721729; 1997.
You, H. J.; Swanson, R. L.; Harrington, C.; Corbett, A. H.;
Jinks-Robertson, S.; Senturker, S.; Wallace, S. S.; Boiteux, S.;
Dizdaroglu, M.; Doetsch, P. W. Saccharomyces cerevisiae
Ntg1p and Ntg2p: broad specificity N-glycosylases for the repair
of oxidative DNA damage in the nucleus and mitochondria.
Biochemistry 38:11298 11306; 1999.
Alseth, I.; Eide, L.; Pirovano, M.; Rognes, T.; Seeberg, E.;
Bjrs, M. The Saccharomyces cerevisiae homologues of endonuclease III from Escherichia coli, Ntg1 and Ntg2, are both
required for efficient repair of spontaneous and induced oxidative DNA damage in yeast. Mol. Cell. Biol. 19:3779 3787;
1999.
Swanson, R. L.; Morey, N. J.; Doetsch, P. W.; Jinks-Robertson,
S. Overlapping specificities of base excision repair, nucleotide
excision repair, recombination, and translesion synthesis pathways for DNA base damage in Saccharomyces cerevisiae. Mol.
Cell. Biol. 19:2929 2935; 1999.

Consequences of oxidized DNA bases


[67] You, H. J.; Swanson, R. L.; Doetsch, P. W. Saccharomyces
cerevisiae possesses two functional homologues of Escherichia
coli endonuclease III. Biochemistry 37:6033 6040; 1998.
[68] Leadon, S. A.; Barbee, S. L.; Dunn, A.-B. The yeast RAD2, but
not RAD1, gene is involved in the transcription-coupled repair of
thymine glycols. Mutat. Res. 337:169 178; 1995.
[69] Rolda n-Arjona, T.; Anselmino, C.; Lindahl, T. Molecular cloning and functional analysis of a Schizosaccharomyces pombe
homologue of Escherichia coli endonuclease III. Nucleic Acids
Res. 24:33073312; 1996.
[70] Karahalil, B.; Rolda n-Arjona, T.; Dizdaroglu, M. Substrate specificity of Schizosaccharomyces pombe Nth protein for products
of oxidative DNA damage. Biochemistry 37:590 595; 1998.
[71] Hilbert, T. P.; Boorstein, R. J.; Kung, H. C.; Bolton, P. H.; Xing,
D.; Cunningham, R. P.; Teebor, G. W. Purification of a mammalian homologue of Escherichia coli endonuclease III: identification of a bovine pyrimidine hydrate-thymine glycol DNAglycosylase/AP lyase by irreversible cross linking to a thymine
glycol-containing oligoxynucleotide. Biochemistry 35:2505
2511; 1996.
[72] Aspinwall, R.; Rothwell, D. G.; Rolda n-Arjona, T.; Anselmino,
C.; Ward, C. J.; Cheadle, J. P.; Sampson, J. R.; Lindahl, T.;
Harris, P. C.; Hickson, I. D. Cloning and characterization of a
functional human homolog of Escherichia coli endonuclease III.
Proc. Natl. Acad. Sci. USA 94:109 114; 1997.
[73] Hilbert, T. P.; Chaung, W.; Boorstein, R. J.; Cunningham, R. P.;
Teebor, G. W. Cloning and expression of the cDNA encoding
the human homologue of the DNA repair enzyme, Escherichia
coli endonuclease III. J. Biol. Chem. 272:6733 6740; 1997.
[74] Sarker, A. H.; Ikeda, S.; Nakano, H.; Terato, H.; Ide, H.; Imai,
K.; Akiyama, K.; Tsutsui, K.; Bo, Z.; Kubo, K.; Yamamoto, K.;
Yasui, A.; Yoshida, M. C.; Seki, S. Cloning and characterization
of a mouse homologue (mNthl1) of Escherichia coli endonuclease III. J. Mol. Biol. 282:761774; 1998.
[75] Ikeda, S.; Biswas, T.; Roy, R.; Izumi, T.; Boldogh, I.; Kurosky,
A.; Sarker, A. H.; Seki, S.; Mitra, S. Purification and characterization of human NTH1, a homolog of Escherichia coli endonuclease III. Direct identification of Lys-212 as the active nucleophilic residue. J. Biol. Chem. 273:2158521593; 1998.
[76] Asagoshi, K.; Yamada, T.; Okada, Y.; Terato, H.; Ohyama, Y.;
Seki, S.; Ide, H. Recognition of formamidopyrimidine by Escherichia coli and mammalian thymine glycol glycosylases. Distinctive paired base effects and biological and mechanistic implications. J. Biol. Chem. 275:2478124786; 2000.
[77] Eide, L.; Luna, L.; Gustad, E. C.; Henderson, P. T.; Essigmann,
J. M.; Demple, B.; Seeberg, E. Human endonuclease III acts
preferentially on DNA damage opposite guanine residues in
DNA. Biochemistry 40:6653 6659; 2001.
[78] Takao, M.; Aburatani, H.; Kobayashi, K.; Yasui, A. Mitochondrial targeting of human DNA glycosylases for repair of oxidative DNA damage. Nucleic Acids Res. 26:29172922; 1998.
[79] Luna, L.; Bjrs, M.; Hoff, E.; Rognes, T.; Seeberg, E. Cellcycle regulation, intracellular sorting and induced overexpression of the human NTH1 DNA glycosylase involved in removal
of formamidopyrimidine residues from DNA. Mutat. Res. 460:
95104; 2000.
[80] von Sonntag, C. The chemical basis of radiation biology. London: Taylor and Francis; 1987.
[81] Wagner, J. R.; Hu, C.-C.; Ames, B. N. Endogenous oxidative
damage of deoxycytidine in DNA. Proc. Natl. Acad. Sci. USA
89:3380 3384; 1992.
[82] Purmal, A. A.; Kow, Y. W.; Wallace, S. S. Major oxidative
products of cytosine, 5-hydroxycytosine and 5-hydroxyuracil,
exhibit sequence context-dependent mispairing in vitro. Nucleic
Acids Res. 22:7278; 1994.
[83] Purmal, A. A.; Lampman, G. W.; Bond, J. P.; Hatahet, Z.;
Wallace, S. S. Enzymatic processing of uracil glycol, a major
oxidative product of DNA cytosine. J. Biol. Chem. 273:10026
10035; 1998.
[84] Kreutzer, D. A.; Essigmann, J. M. Oxidized, deaminated cy-

[85]

[86]
[87]

[88]

[89]

[90]
[91]

[92]

[93]

[94]

[95]

[96]

[97]

[98]

[99]

[100]

[101]

[102]

[103]

11

tosines are a source of C 3 T transitions in vivo. Proc. Natl.


Acad. Sci. USA 95:3578 3582; 1998.
Feig, D. I.; Sowers, L. C.; Loeb, L. A. Reverse chemical mutagenesis: identification of the mutagenic lesions resulting from
reactive oxygen species-mediated damage to DNA. Proc. Natl.
Acad. Sci. USA 91:6609 6613; 1994.
Wallace, S. S. The biological consequences of oxidized DNA
bases. Br. J. Cancer Suppl. 8:118 128; 1987.
Wallace, S. S. AP endonucleases and DNA glycosylases that
recognize oxidative DNA damage. Environ. Mol. Mutagen. 12:
431 477; 1988.
Blaisdell, J. O.; Hatahet, Z.; Wallace, S. S. A novel role for
Escherichia coli endonuclease VIII in the prevention of spontaneous G 3 T transversions. J. Bacteriol. 181:6396 6402; 1999.
Kasai, H.; Crain, P. F.; Kuchino, Y.; Nishimura, S.; Ootsuyama,
A.; Tanooka, H. Formation of 8-hydroxyguanine moiety in cellular DNA by agents producing oxygen radicals and evidence for
its repair. Carcinogenesis 7:1849 1851; 1986.
Floyd, R. A. The role of 8-hydroxyguanine in carcinogenesis.
Carcinogenesis 11:14471450; 1990.
Oda, Y.; Uesugi, S.; Ikehara, M.; Nishimura, S.; Kawase, Y.;
Ishikawa, H.; Inoue, H.; Ohtsuka, E. NMR studies of a DNA
containing 8-hydroxydeoxyguanosine. Nucleic Acids Res. 19:
14071412; 1991.
Lipscomb, L.; Peek, M.; Morningstar, M.; Verghis, S.; Miller,
E.; Rich, A.; Essigmann, J.; Williams, L. X-ray structure of a
DNA decamer containing 7,8-dihydro-8-oxoguanine. Proc. Natl.
Acad. Sci. USA 92:719 723; 1995.
Kouchakdjian, M.; Bodepudi, V.; Shibutani, S.; Eisenberg, M.;
Johnson, F.; Grollman, A. P.; Patel, D. J. NMR structural studies
of the ionizing radiation adduct 7-hydro-8-oxodeoxyguanosine
(8-oxo-7H-dG) opposite deoxyadenosine in a DNA duplex.
8-Oxo-7H-dG(syn).dA(anti) alignment at lesion site. Biochemistry 30:14031412; 1991.
McAuley-Hecht, K.; Leonard, G.; Gibson, N.; Thomson, J.;
Watson, W.; Hunter, W.; Brown, T. Crystal structure of a DNA
duplex containing 8-hydroxydeoxyguanine-adenine base pairs.
Biochemistry 33:10266 10270; 1994.
Wood, M. L.; Dizdaroglu, M.; Gajewski, E.; Essigmann, J. M.
Mechanistic studies of ionizing radiation and oxidative mutagenesis: genetic effects of a single 8-hydroxyguanine (7-hydro-8oxoguanine) residue inserted at a unique site in a viral genome.
Biochemistry 29:7024 7032; 1990.
Shibutani, S.; Takeshita, M.; Grollman, A. P. Insertion of specific bases during DNA synthesis past the oxidation-damaged
base 8-oxodG. Nature 349:431 434; 1991.
Moriya, M.; Ou, C.; Bodepudi, V.; Johnson, F.; Takeshita, M.;
Grollman, A. P. Site-specific mutagenesis using a gapped duplex
vector: a study of translesion synthesis past 8-oxodeoxyguanosine in E. coli. Mutat. Res. 254:281288; 1991.
Cheng, K. C.; Cahill, D. S.; Kasai, H.; Nishimura, S.; Loeb,
L. A. 8-Hydroxyguanine, an abundant form of oxidative DNA
damage, causes G 3 T and A 3 C substitutions. J. Biol. Chem.
267:166 172; 1992.
Klein, J. C.; Bleeker, M. J.; Saris, C. P.; Roelen, H. C.; Brugghe,
H. F.; van den Elst, H.; van der Marel, G. A.; van Boom, J. H.;
Westra, J. G.; Kriek, E.; Berns, A. J. M. Repair and replication
of plasmids with site-specific 8-oxodG and 8-AAFdG residues in
normal and repair-deficient human cells. Nucleic Acids Res.
20:4437 4443; 1992.
Moriya, M. Single-stranded shuttle phagemid for mutagenesis
studies in mammalian cells: 8-oxo-guanine in DNA induces
targeted G:C 3 T:A transversions in simian kidney cells. Proc.
Natl. Acad. Sci. USA 90:11221126; 1993.
Hatahet, Z.; Zhou, M.; Reha-Krantz, L. J.; Morrical, S. W.;
Wallace, S. S. In search of a mutational hotspot. Proc. Natl.
Acad. Sci. USA 95:8556 8561; 1998.
OConnor, T. R.; Boiteux, S.; Laval, J. Ring-opened 7-methylguanine residues in DNA are a block to in vitro DNA synthesis.
Nucleic Acids Res. 16:5879 5894; 1988.
Tudek, B.; Boiteux, S.; Laval, J. Biological properties of imida-

12

[104]

[105]

[106]

[107]

[108]

[109]

[110]

[111]

[112]

[113]

[114]

[115]

[116]

[117]

[118]

[119]

S. S. WALLACE
zole ring-opened N7-methylguanine in M13mp18 phage DNA.
Nucleic Acids Res. 20:3079 3084; 1992.
Michaels, M. L.; Miller, J. H. The GO system protects organisms
from the mutagenic effect of the spontaneous lesion 8-hydroxyguanine (7,8-dihydro-8-oxoguanine). J. Bacteriol. 174:6321
6325; 1992.
Cabrera, M.; Nghiem, Y.; Miller, J. H. mutM, a second mutator
locus in Escherichia coli that generates G T 3 T A transversions. J. Bacteriol. 170:54055407; 1988.
Michaels, M. L.; Cruz, C.; Grollman, A. P.; Miller, J. H. Evidence that MutY and MutM combine to prevent mutations by an
oxidatively damaged form of guanine in DNA. Proc. Natl. Acad.
Sci. USA 89:70227025; 1992.
Kuipers, G. K.; Poldervaart, H. A.; Slotman, B. J.; Lafleur, M. V.
The influence of formamidopyrimidine-DNA glycosylase on the
spontaneous and -radiation-induced mutation spectrum of the
lacZ gene. Mutat. Res. 435:141150; 1999.
Konevega, L. V.; Kalinin, V. L. Mutagenic effects of gammarays and incorporated 8-3H-purines on extracellular lambda
phage: influence of mutY and mutM host mutations. Mutat. Res.
459:229 235; 2000.
Kuipers, G. K.; Slotman, B. J.; Poldervaart, H. A.; ReitsmaWijker, C. A.; Lafleur, M. V. The influence of combined Fpgand MutY-deficiency on the spontaneous and -radiation-induced mutation spectrum in the lacZ gene of M13mp10. Mutat.
Res. 461:189 195; 2000.
Hazra, T. K.; Izumi, T.; Venkataraman, R.; Kow, Y. W.; Dizdaroglu, M.; Mitra, S. Characterization of a novel 8-oxoguanineDNA glycosylase activity in Escherichia coli and identification
of the enzyme as endonuclease VIII. J. Biol. Chem. 275:27762
27767; 2000.
Chetsanga, C. J.; Lindahl, T. Release of 7-methylguanine residues whose imidazole rings have been opened from damaged
DNA by a DNA glycosylase from Escherichia coli. Nucleic
Acids Res. 6:36733684; 1979.
Chetsanga, C. J.; Lozon, M.; Makaroff, C.; Savage, L. Purification and characterization of Escherichia coli formamidopyrimidine-DNA glycosylase that excises damaged 7-methylguanine
from deoxyribonucleic acid. Biochemistry 20:52015207; 1981.
Nash, H. M.; Bruner, S. D.; Scharer, O. D.; Kawate, T.; Addona,
T. A.; Spooner, E.; Lane, W. S.; Verdine, G. L. Cloning of a
yeast 8-oxoguanine DNA glycosylase reveals the existence of a
base-excision DNA-repair protein superfamily. Curr. Biol.
6:968 980; 1996.
van der Kemp, P. A.; Thomas, D.; Barbey, R.; de Oliveira, R.;
Boiteux, S. Cloning and expression in Escherichia coli of the
OGG1 gene of Saccharomyces cerevisiae, which codes for a
DNA glycosylase that excises 7,8-dihydro-8-oxoguanine and
2,6-diamino-4-hydroxy-5-N-methylformamidopyrimidine. Proc.
Natl. Acad. Sci. USA 93:51975202; 1996.
Girard, P.-M.; Guibourt, N.; Boiteux, S. The Ogg1 protein of
Saccharomyces cerevisiae: a 7,8-dihydro-8-oxoguanine DNA
glycosylase/AP lyase whose lysine 241 is a critical residue for
catalytic activity. Nucleic Acids Res. 25:3204 3211; 1997.
Asagoshi, K.; Yamada, T.; Okada, Y.; Terato, H.; Ohyama, Y.;
Seki, S.; Ide, H. Recognition of formamidopyrimidine by Escherichia coli and mammalian thymine glycol glycosylases. Distinctive paired base effects and biological and mechanistic implications. J. Biol. Chem. 275:2478124786; 2000.
Gifford, C. M.; Wallace, S. S. The genes encoding formamidopyrimidine and MutY DNA glycosylases in Escherichia coli
are transcribed as part of complex operons. J. Bacteriol. 181:
4223 4236; 1999.
Kim, H. S.; Park, Y. W.; Kasai, H.; Nishimura, S.; Park, C. W.;
Choi, K. H.; Chung, M. H. Induction of E. coli oh8Gua endonuclease by oxidative stress: its significance in aerobic life.
Mutat. Res. 363:115123; 1996.
Lee, H. S.; Lee, Y. S.; Kim, H. S.; Choi, J. Y.; Hassan, H. M.;
Chung, M. H. Mechanism of regulation of 8-hydroxyguanine
endonuclease by oxidative stress: roles of FNR, ArcA, and Fur.
Free Radic. Biol. Med. 24:11931201; 1998.

[120] Gao, M. J.; Murphy, T. M. Alternative forms of formamidopyrimidine-DNA glycosylase from Arabidopsis thaliana. Photochem. Photobiol. 73:128 134; 2001.
[121] Ohtsubo, T.; Matsuda, O.; Iba, K.; Terashima, I.; Sekiguchi, M.;
Nakabeppu, Y. Molecular cloning of AtMMH, an Arabidopsis
thaliana ortholog of the Escherichia coli mutM gene, and analysis of functional domains of its product. Mol. Gen. Genet.
259:577590; 1998.
[122] Karahalil, B.; Girard, P. M.; Boiteux, S.; Dizdaroglu, M. Substrate specificity of the Ogg1 protein of Saccharomyces cerevisiae: excision of guanine lesions produced in DNA by ionizing
radiation- or hydrogen peroxide/metal ion-generated free radicals. Nucleic Acids Res. 26:1228 1232.
[123] Thomas, D.; Scot, A. D.; Barbey, R.; Padula, M.; Boiteux, S.
Inactivation of OGG1 increases the incidence of G C 3 T A
transversions in Saccharomyces cerevisiae: evidence for endogenous oxidative damage to DNA in eukaryotic cells. Mol. Gen.
Genet. 254:171178; 1997.
[124] Ni, T. T.; Marsischky, G. T.; Kolodner, R. D. MSH2 and MSH6
are required for removal of adenine misincorporated opposite
8-oxo-guanine in S. cerevisiae. Mol. Cell 4:439 444; 1999.
[125] Yacoub, A.; Augeri, L.; Kelley, M. R.; Doetsch, P. W.; Deutsch,
W. A. A Drosophila ribosomal protein contains 8-oxoguanine
and abasic site DNA repair activities. EMBO J. 15:2306 2312;
1996.
[126] Deutsch, W. A.; Yacoub, A.; Jaruga, P.; Zastawny, T. H.;
Dizdaroglu, M. Characterization and mechanism of action of
Drosophila ribosomal protein S3 DNA glycosylase activity for
the removal of oxidatively damaged DNA bases. J. Biol. Chem.
272:3285732860; 1997.
[127] Dherin, C.; Dizdaroglu, M.; Doerflinger, H.; Boiteux, S.; Radicella, J. P. Repair of oxidative DNA damage in Drosophila
melanogaster: identification and characterization of dOgg1, a
second DNA glycosylase activity for 8-hydroxyguanine and
formamidopyrimidines. Nucleic Acids Res. 28:4583 4592;
2000.
[128] Radicella, J. P.; Dherin, C.; Desmaze, C.; Fox, M. S.; Boiteux,
S. Cloning and characterization of hOGG1, a human homolog of
the OGG1 gene of Saccharomyces cerevisiae. Proc. Natl. Acad.
Sci. USA 94:8010 8015; 1997.
[129] Aburatani, H.; Hippo, Y.; Ishida, T.; Takashima, R.; Matsuba,
C.; Kodama, T.; Takao, M.; Yasui, A.; Yamamoto, K.; Asano,
M.; Fukasawa, K.; Yoshinari, T.; Inoue, H.; Ohtsuka, E.; Nishimura, S. Cloning and characterization of mammalian 8-hydroxyguanine-specific DNA glycosylase/apurinic, apyrimidinic
lyase, a functional mutM homologue. Cancer Res. 57:2151
2156; 1997.
[130] Arai, K.; Morishita, K.; Shinmura, K.; Kohno, T.; Kim, S.-R.;
Nohmi, T.; Taniwaki, S.; Ohwada, S.; Yokota, J. Cloning of a
human homolog of the yeast OGG1 gene that is involved in the
repair of oxidative DNA damage. Oncogene 14:28572861;
1997.
[131] Bjrs, M.; Luna, L.; Johnsen, B.; Hoff, E.; Haug, T.; Rognes,
T.; Seeberg, E. Opposite base-dependent reactions of a human
base excision repair enzyme on DNA containing 7,8-dihydro-8oxoguanine and abasic sites. EMBO J. 16:6314 6322; 1997.
[132] Lu, R.; Nash, H. M.; Verdine, G. L. A mammalian DNA repair
enzyme that excises oxidatively damaged guanines maps to a
locus frequently lost in lung cancer. Curr. Biol. 7:397 407;
1997.
[133] Rolda n-Arjona, T.; Wei, Y.-F.; Carter, K. C.; Klungland, A.;
Anselmino, C.; Wang, R.-P.; Augustus, M.; Lindahl, T. Molecular cloning and functional expression of a human cDNA encoding the antimutator enzyme 8-hydroxyguanine-DNA glycosylase. Proc. Natl. Acad. Sci. USA 94:8016 8020; 1997.
[134] Rosenquist, T. A.; Zharkov, D. O.; Grollman, A. P. Cloning and
characterization of a mammalian 8-oxoguanine DNA glycosylase. Proc. Natl. Acad. Sci. USA 94:7429 7434; 1997.
[135] Tani, M.; Shinmura, K.; Kohno, T.; Shiroishi, T.; Wakana, S.;
Kim, S. R.; Nohmi, T.; Kasai, H.; Takenoshita, S.; Nagamachi,
Y.; Yokota, J. Genomic structure and chromosomal localization

Consequences of oxidized DNA bases

[136]

[137]

[138]

[139]

[140]

[141]

[142]

[143]

[144]

[145]

[146]

[147]

[148]

[149]

[150]

of the mouse Ogg1 gene that is involved in the repair of 8-hydroxyguanine in DNA damage. Mamm. Genome 9:3237; 1998.
Boiteux, S.; Dherin, C.; Reille, F.; Apiou, F.; Dutrillaux, B.;
Radicella, J. P. Excision repair of 8-hydroxyguanine in mammalian cells: the mouse Ogg1 protein as a model. Free Radic.
Res. 29:487 497; 1998.
Lee, Y. S.; Choi, J. Y.; Park, M. K.; Choi, E. M.; Kasai, H.;
Chung, M. H. Induction of oh8Gua glycosylase in rat kidneys by
potassium bromate (KBrO3), a renal oxidative carcinogen. Mutat. Res. 364:227233; 1996.
Rusyn, I.; Denissenko, M. F.; Wong, V. A.; Butterworth, B. E.;
Cunningham, M. L.; Upton, P. B.; Thurman, R. G.; Swenberg,
J. A. Expression of base excision repair enzymes in rat and
mouse liver is induced by peroxisome proliferators and is dependent upon carcinogenic potency. Carcinogenesis 21:2141
2145; 2000.
Kim, H.-N.; Morimoto, Y.; Tsuda, T.; Ootsuyama, Y.; Hirohashi, M.; Hirano, T.; Tanaka, I.; Lim, Y.; Yun, I.-G.; Kasai, H.
Changes in DNA 8-hydroxyguanine levels, 8-hydroxyguanine
repair activity, and hOGG1 and hMTH1 mRNA expression in
human lung alveolar epithelial cells induced by crocidolite asbestos. Carcinogenesis 22:265269; 2001.
Tsurudome, Y.; Hirano, T.; Yamato, H.; Tanaka, I.; Sagai, M.;
Hirano, H.; Nagata, N.; Itoh, H.; Kasai, H. Changes in levels of
8-hydroxyguanine in DNA, its repair and OGG1 mRNA in rat
lungs after intratracheal administration of diesel exhaust particles. Carcinogenesis 20:15731576; 1999.
Lin, L. H.; Cao, S.; Yu, L.; Cui, J.; Hamilton, W. J.; Liu, P. K.
Up-regulation of base excision repair activity for 8-hydroxy-2deoxyguanosine in the mouse brain after forebrain ischemiareperfusion. J. Neurochem. 74:1098 1105; 2000.
You, H.; Kim, G.; Kim, Y.; Chun, Y.; Park, J.; Chung, M. H.;
Kim, M. Increased 8-hydroxyguanine formation and endonuclease activity for its repair in ischemic-reperfused hearts of rats. J.
Mol. Cell. Cardiol. 32:10531059; 2000.
Dhenaut, A.; Boiteux, S.; Radicella, J. P. Characterization of the
hOGG1 promoter and its expression during the cell cycle. Mutat.
Res. 461:109 118; 2000.
Audebert, M.; Chevillard, S.; Levalois, C.; Gyapay, G.; Vieillefond, A.; Klijanienko, J.; Vielh, P.; El Naggar, A. K.; Oudard, S.;
Boiteux, S.; Radicella, J. P. Alterations of the DNA repair gene
OGG1 in human clear cell carcinomas of the kidney. Cancer
Res. 60:4740 4644; 2000.
Blons, H.; Radicella, J. P.; Laccourreye, O.; Brasnu, D.; Beaune,
P.; Boiteux, S.; Laurent-Puig, P. Frequent allelic loss at chromosome 3p distinct from genetic alterations of the 8-oxoguanine
DNA glycosylase 1 gene in head and neck cancer. Mol. Carcinog. 26:254 260; 1999.
Chevillard, S.; Radicella, J. P.; Levalois, C.; Lebeau, J.; Poupon,
M. F.; Oudard, S.; Dutrillaux, B.; Boiteux, S. Mutations in
OGG1, a gene involved in the repair of oxidative DNA damage,
are found in human lung and kidney tumours. Oncogene 16:
30833086; 1998.
Kohno, T.; Shinmura, K.; Tosaka, M.; Tani, M.; Kim, S. R.;
Sugimura, H.; Nohmi, T.; Kasai, H.; Yokota, J. Genetic polymorphisms and alternative splicing of the hOGG1 gene, that is
involved in the repair of 8-hydroxyguanine in damaged DNA.
Oncogene 16:3219 3225; 1998.
Dherin, C.; Radicella, J. P.; Dizdaroglu, M.; Boiteux, S. Excision
of oxidatively damaged DNA bases by the human -hOgg1
protein and the polymorphic -hOgg1(Ser326Cys) protein
which is frequently found in human populations. Nucleic Acids
Res. 27:4001 4007; 1999.
Audebert, M.; Radicella, J. P.; Dizdaroglu, M. Effect of single
mutations in the OGG1 gene found in human tumors on the
substrate specificity of the Ogg1 protein. Nucleic Acids Res.
28:26722678; 2000.
Hyun, J. H.; Choi, J. Y.; Zeng, H. H.; Lee, Y. S.; Kim, H. S.;
Yoon, S. H.; Chung, M. H. Leukemic cell line, KG-1 has a
functional loss of hOGG1 enzyme due to a point mutation and

[151]

[152]

[153]

[154]

[155]

[156]

[157]

[158]

[159]

[160]
[161]

[162]

[163]

[164]

[165]

[166]

[167]

13

8-hydroxydeoxyguanosine can kill KG-1. Oncogene 19:4476


4479; 2000.
Klungland, A.; Rosewell, I.; Hollenbach, S.; Larsen, E.; Daly,
G.; Epe, B.; Seeberg, E.; Lindahl, T.; Barnes, D. E. Accumulation of premutagenic DNA lesions in mice defective in removal
of oxidative base damage. Proc. Natl. Acad. Sci. USA 96:
13300 13305; 1999.
Minowa, O.; Arai, T.; Hirano, M.; Monden, Y.; Nakai, S.;
Fukuda, M.; Itoh, M.; Takano, H.; Hippou, Y.; Aburatani, H.;
Masumura, K.; Nohmi, T.; Nishimura, S.; Noda, T. Mmh/Ogg1
gene inactivation results in accumulation of 8-hydroxyguanine
in mice. Proc. Natl. Acad. Sci. USA 97:4156 4161; 2000.
McGoldrick, J. P.; Yeh, Y.-C.; Solomon, M.; Essigmann, J. M.;
Lu, A.-L. Characterization of a mammalian homolog of the
Escherichia coli MutY mismatch repair protein. Mol. Cell. Biol.
15:989 996; 1995.
Slupska, M. M.; Baikalov, C.; Luther, W. M.; Chiang, J.-H.;
Wei, Y.-F.; Miller, J. H. Cloning and sequencing a human
homolog (hMYH) of the Escherichia coli mutY gene whose
function is required for the repair of oxidative DNA damage. J.
Bacteriol. 178:38853892; 1996.
Slupska, M. M.; Luther, W. M.; Chiang, J. H.; Yang, H.; Miller,
J. H. Functional expression of hMYH, a human homolog of the
Escherichia coli MutY protein. J. Bacteriol. 181:6210 6213;
1999.
Takao, M.; Zhang, Q. M.; Yonei, S.; Yasui, A. Differential
subcellular localization of human MutY homolog (hMYH) and
the functional activity of adenine:8-oxoguanine DNA glycosylase. Nucleic Acids Res. 27:3638 3644; 1999.
Ohtsubo, T.; Nishioka, K.; Imaiso, Y.; Iwai, S.; Shimokawa, H.;
Oda, H.; Fujiwara, T.; Nakabeppu, Y. Identification of human
MutY homolog (hMYH) as a repair enzyme for 2- hydroxyadenine in DNA and detection of multiple forms of hMYH located
in nuclei and mitochondria. Nucleic Acids Res. 28:13551364;
2000.
Tsai-Wu, J. J.; Su, H. T.; Wu, Y. L.; Hsu, S. M.; Wu, C. H.
Nuclear localization of the human mutY homologue hMYH. J.
Cell. Biochem. 77:666 677; 2000.
Friedberg, E. C.; Meira, L. B. Database of mouse strains carrying targeted mutations in genes affecting cellular responses to
DNA damage. Version 4. Mutat. Res. 459:243274; 2000.
Van Hemmen, J. J.; Bleichrodt, J. F. The decomposition of
adenine by ionizing radiation. Radiat. Res. 46:444 456; 1971.
Stillwell, W. G.; Xu, H. X.; Adkins, J. A.; Wishnok, J. S.;
Tannenbaum, S. R. Analysis of methylated and oxidized purines
in urine by capillary gas chromatography-mass spectrometry.
Chem. Res. Toxicol. 2:94 99; 1989.
Malins, D. C.; Haimanot, R. 4,6-Diamino-5-formamidopyrimidine, 8-hydroxyguanine and 8-hydroxyadenine in DNA from
neoplastic liver of English sole exposed to carcinogens. Biochem. Biophys. Res. Commun. 173:614 619; 1990.
Guschlbauer, W.; Duplaa, A. M.; Guy, A.; Teoule, R.; Fazakerley, G. V. Structure and in vitro replication of DNA templates
containing 7,8-dihydro-8-oxoadenine. Nucleic Acids Res. 19:
17531758; 1991.
Leonard, G. A.; Guy, A.; Brown, T.; Teoule, R.; Hunter, W. N.
Conformation of guanine-8-oxoadenine base pairs in the crystal
structure of d(CGCGAATT(O8A)GCG). Biochemistry 31:
8415 8420; 1992.
Wood, M.; Esteve, A.; Morningstar, M.; Kuziemko, G.; Essigmann, J. Genetic effects of oxidative DNA damage: comparative
mutagenesis of 7,8-dihydro-8-oxoguanine and 7,8-dihydro-8oxoadenine in Escherichia coli. Nucleic Acids Res. 20:6023
6032; 1992.
Shibutani, S.; Bodepudi, V.; Johnson, F.; Grollman, A. P. Translesional synthesis on DNA templates containing 8-oxo-7,8-dihydrodeoxyadenosine. Biochemistry 32:4615 4621; 1993.
Kamiya, H.; Miura, H.; Murata-Kamiya, N.; Ishikawa, H.; Sakaguchi, T.; Inoue, H.; Sasaki, T.; Masutani, C.; Hanaoka, F.;
Nishimura, S. 8-Hydroxyadenine (7,8-dihydro-8-oxoadenine)

14

[168]

[169]

[170]

[171]

[172]

[173]

[174]

[175]

[176]

[177]

[178]

[179]

[180]

S. S. WALLACE
induces misincorporation in in vitro DNA synthesis and mutations in NIH 3T3 cells. Nucleic Acids Res. 23:28932899; 1995.
Kamiya, H.; Murata-Kamiya, N.; Koizume, S.; Inoue, H.; Nishimura, S.; Ohtsuka, E. 8-Hydroxyguanine (7,8-dihydro-8-oxoguanine) in hot spots of the c-Ha-ras gene: effects of sequence
contexts on mutation spectra. Carcinogenesis 16:883 889;
1995.
Nackerdien, Z.; Kasprzak, K. S.; Rao, G.; Halliwell, B.; Dizdaroglu, M. Nickel(II)- and cobalt(II)-dependent damage by hydrogen peroxide to the DNA bases in isolated human chromatin.
Cancer Res. 51:58375842; 1991.
Olinski, R.; Zastawny, T.; Budzbon, J.; Skokowski, J.; Zegarski,
W.; Dizdaroglu, M. DNA base modifications in chromatin of
human cancerous tissues. FEBS Lett. 309:193198; 1992.
Mori, T.; Hori, Y.; Dizdaroglu, M. DNA base damage generated
in vivo in hepatic chromatin of mice upon whole body -irradiation. Int. J. Radiat. Biol. 64:645 650; 1993.
Kamiya, H.; Kasai, H. Formation of 2-hydroxydeoxyadenosine
triphosphate, an oxidatively damaged nucleotide, and its incorporation by DNA polymerases. Steady-state kinetics of the incorporation. J. Biol. Chem. 270:19446 19450; 1995.
Kamiya, H.; Ueda, T.; Ohgi, T.; Matsukage, A.; Kasai, H.
Misincorporation of dAMP opposite 2-hydroxyadenine, an oxidative form of adenine. Nucleic Acids Res. 23:761766; 1995.
Kamiya, H.; Kasai, H. Effects of sequence contexts on misincorporation of nucleotides opposite 2-hydroxyadenine. FEBS
Lett. 391:113116; 1996.
Kamiya, H.; Kasai, H. Substitution and deletion mutations induced by 2-hydroxyadenine in Escherichia coli: effects of sequence contexts in leading and lagging strands. Nucleic Acids
Res. 25:304 311; 1997.
Kamiya, H.; Kasai, H. Mutations induced by 2-hydroxyadenine
on a shuttle vector during leading and lagging strand syntheses
in mammalian cells. Biochemistry 36:1112511130; 1997.
Dizdaroglu, M.; Nackerdien, Z.; Chao, B. C.; Gajewski, E.; Rao,
G. Chemical nature of in vivo DNA base damage in hydrogen
peroxide-treated mammalian cells. Arch. Biochem. Biophys.
285:388 390; 1991.
Olinski, R.; Zastawny, T.; Budzbon, J.; Skokowski, J.; Zegarski,
W.; Dizdaroglu, M. Oxidative DNA base damage and antioxidant enzyme activities in human lung cancer. FEBS Lett. 309:
193198; 1992.
Zastawny, T. H.; Czerwinska, B.; Drzewiecka, B.; Olinski, R.
Radiation-induced oxidative DNA base damage and its repair in
nuclear matrix-associated DNA and in bulk DNA in hepatic
chromatin of rat upon whole-body gamma-irradiation. Free
Radic. Biol. Med. 22:101107; 1997.
Graziewicz, M. A.; Zastawny, T. H.; Olinski, R.; Speina, E.;
Siedlecki, J.; Tudek, B. FAPYadenine is a moderately efficient

[181]
[182]

[183]

[184]

[185]

[186]

[187]
[188]
[189]
[190]
[191]
[192]

[193]
[194]

chain terminator for prokaryotic DNA polymerases. Free Radic.


Biol. Med. 28:75 83; 2000.
Lesiak, K. B.; Wheeler, K. T. Formation of -deoxyadenosine in
polydeoxynucleotides exposed to ionizing radiation under anoxic conditions. Radiat. Res. 121:328 337; 1990.
Ide, H.; Yamaoka, T.; Kimura, Y. Replication of DNA templates
containing the -anomer of deoxyadenosine, a major adenine
lesion produced by hydroxyl radicals. Biochemistry 33:7127
7133; 1994.
Shimizu, H.; Yagi, R.; Kimura, Y.; Makino, K.; Terato, H.;
Ohyama, Y.; Ide, H. Replication bypass and mutagenic effect of
-deoxyadenosine site-specifically incorporated into singlestranded vectors. Nucleic Acids Res. 25:597 603; 1997.
Boiteux, S.; Gajewski, E.; Laval, J.; Dizdaroglu, M. Substrate
specificity of the Escherichia coli Fpg protein (formamidopyrimidine-DNA glycosylase): excision of purine lesions in DNA
produced by ionizing radiation or photosensitization. Biochemistry 31:106 110; 1992.
Girard, P. M.; DHam, C.; Cadet, J.; Boiteux, S. Opposite
base-dependent excision of 7,8-dihydro-8-oxoadenine by the
Ogg1 protein of Saccharomyces cerevisiae. Carcinogenesis 19:
1299 1305; 1998.
Tsurudome, Y.; Hirano, T.; Kamiya, H.; Yamaguchi, R.; Asami,
S.; Itoh, H.; Kasai, H. 2-Hydroxyadenine, a mutagenic form of
oxidative DNA damage, is not repaired by a glycosylase type
mechanism in rat organs. Mutat. Res. 408:121127; 1998.
Ward, J. F. Biochemistry of DNA lesions. Radiat. Res. 104:
S103S111; 1985.
Ward, J. F. DNA damage produced by ionizing radiation in
mammalian cells: identities, mechanisms of formation, and reparability. Prog. Nucleic Acid Res. Mol. Biol. 35:95125; 1988.
Chaudhry, M. A.; Weinfeld, M. The action of Escherichia coli
endonuclease III on multiply damaged sites in DNA. J. Mol.
Biol. 249:914 922; 1995.
Harrison, L.; Hatahet, Z.; Purmal, A. A.; Wallace, S. S. Multiply
damaged sites in DNA: interactions with Escherichia coli endonucleases III and VIII. Nucleic Acids Res. 26:932941; 1998.
Harrison, L.; Hatahet, Z.; Wallace, S. S. In vitro repair of
synthetic ionizing radiation-induced multiply damaged DNA
sites. J. Mol. Biol. 290:667 684; 1999.
David-Cordonnier, M. H.; Laval, J.; ONeill, P. Clustered DNA
damage, influence on damage excision by XRS5 nuclear extracts
and Escherichia coli Nth and Fpg proteins. J. Biol. Chem.
275:1186511873; 2000.
David-Cordonnier, M. H.; Boiteux, S.; ONeill, P. Excision of
8-oxoguanine within clustered damage by the yeast OGG1 protein. Nucleic Acids Res. 29:11071113; 2001.
Blaisdell, J. O.; Wallace, S. S. Abortive base-excision repair of
radiation-induced clustered DNA lesions in Escherichia coli.
Proc. Natl. Acad. Sci. USA 98:7426 7430; 2001.

Potrebbero piacerti anche