Mitochondrial Na (1)

Embed Size (px)

Citation preview

  • 8/12/2019 Mitochondrial Na (1)

    1/10

    Clinical Science (2004) 107, 355364 (Printed in Great Britain) 355

    R E V I E W

    Mitochondrial DNA and aging

    Mikhail F. ALEXEYEV, Susan P. LEDOUX and Glenn L. WILSONDepartment of Cell Biology and Neuroscience, University of South Alabama, 307 University Blvd, Mobile, AL 36688, U.S.A.

    A B S T R A C T

    Among the numerous theories that explain the process of aging, the mitochondrial theory of aging

    has received the most attention. This theory states that electrons leaking from the ETC (electron

    transfer chain) reduce molecular oxygen to form O2 (superoxide anion radicals). O2

    , through

    both enzymic and non-enzymic reactions, can cause the generation of other ROS (reactive oxygen

    species). The ensuing state of oxidative stress results in damage to ETC components and mtDNA

    (mitochondrial DNA), thus increasing further the production of ROS. Ultimately, this vicious cycle

    leads to a physiological decline in function, or aging. This review focuses on recent developments in

    aging research related to the role played by mtDNA. Both supportive and contradictory evidence

    is discussed.

    INVOLVEMENT OF mtDNA

    (MITOCHONDRIAL DNA) IN AGING

    Aging can be defined as a multifactorial phenomenon

    characterized by a time-dependent decline in physio-

    logical function [1]. This physiological decline is believed

    to be associated with an accumulation of defects in the

    metabolic pathways. RNA, proteins and other cellularmacromolecules are rapidly turned over and, conse-

    quently, are poor candidates for progressively accumulat-

    ing damage over a lifetime. Therefore even early studies

    on mechanisms of aging focused on DNA (for example,

    see [2,3]). In mammalian cells, mitochondria and the

    nucleus are the only organelles that possess DNA. It

    appears obvious that thephysiological integrityof the cell

    must critically depend upon the integrity of its genome,

    whichis maintainedby DNArepairmachinery. However,

    although the organization, synthesisand repair of nuclear

    DNA have been the focus of intense studies, mtDNA has

    received much less attention until recently.

    BASIC mtDNA BIOLOGY

    Human mtDNA is a circular double-stranded molecule

    that is 16 569 bp long (other sequenced mammalian

    mitochondrial genomes have similar lengths; Figure 1).

    Key words:aging, DNA damage, mitochondrial DNA, oxidative stress, reactive oxygen species, repair.

    Abbreviations: BER, base excision repair; CAT, catalase; ETC, electron transfer chain; O2, superoxide anion radical;

    OH , hydroxyl radical; mtDNA, mitochondrial DNA; MLSP, maximum lifespan; 8-oxodG, 7,8-dihydro-8-oxoguanine; OGG1,

    8-oxoguanine-DNA glycosylase; ROS, reactive oxygen species; SOD, superoxide dismutase; TBA, thiobarbituric acid.

    Correspondence:Dr Mikhail F. Alexeyev (email [email protected]).

    It encodes two rRNAs, 22 tRNAs and 13 polypeptides,

    of which seven are components of complex I (NADH

    dehydrogenase), three are components of complex IV

    (cytochromecoxidase), two are subunits of complex V

    (ATP synthase) and cytochromeb(a subunit of complex

    III) [4]. The inheritance of mtDNA is almost exclusively

    maternal, although some important exceptions have been

    reported [57]. mtDNA is present in one to severalthousand copies per cell [8] and is encapsulated into

    mitochondria at 111 copies per mitochondrion with

    the mean being two genomes per organelle [9]. The two

    mtDNA strands can be separated by denaturing caesium

    chloride gradient centrifugation [10]. Most of the infor-

    mation is encoded in the heavy (purine-rich) strand

    (two rRNAs, 14 tRNAs and 12 polypeptides). The light

    (pyrimidine-rich) strand contains genetic information for

    only one polypeptide and eight tRNAs. Mitochondrial

    genes have no introns and intergenic sequences are absent

    or limited to a few bases. Some genes overlap and, in

    some instances, termination codons are not encoded, butare generated post-transcriptionally by polyadenylation

    [11]. mtDNA is totally dependent upon nuclear-encoded

    proteins for its maintenance and transcription. In fact,

    the mitochondrial proteome consists of an estimated

    1500 polypeptides [12] of which only 13 are encoded

    by its own DNA. mtDNA replication is conducted by

    C 2004 The Biochemical Society

  • 8/12/2019 Mitochondrial Na (1)

    2/10

    356 M. F. Alexeyev, S. P. LeDoux and G. L. Wilson

    Figure 1 Map of human mtDNA

    OH and OL , origins of heavy and light strand replication respectively; ND1

    ND6, NADH dehydrogenase (ETC complex I) subunits 16; Cox1Cox3, cytochrome

    oxidase subunits 13 (ETC complex IV); ATP6 and ATP8, subunits 6 and 8 of

    mitochondrial ATPase (complex V); Cyt b, cytochrome b (complex III).

    the heterodimeric DNA polymerase [13]. Replication

    of mtDNA continues throughout the lifespan of an or-

    ganism in both proliferating and post-mitotic cells. It oc-

    curs bidirectionally, initiated at two spatially and tempo-

    rally distinct origins of replication, OH and OL, for the

    heavy and light strand origins of replication respectively(for review, see [14]). The mitochondrial proteome is

    dynamic, i.e. both the precise polypeptide composition

    and the relative abundance of a given polypeptide may

    vary in mitochondrial proteomes of different tissues as

    well as in the same tissue over time [15]. With the dis-

    covery of mitochondrial diseases caused by mutations of

    mtDNA, it has been found that wild-type (normal) and

    mutated mtDNA may coexist in thesame cell, a condition

    called heteroplasmy [16].

    mtDNA DAMAGE AND THE MITOCHONDRIALTHEORY OF AGING

    Despite the fact that in animal cells mtDNA comprises

    only 13 % of genetic material, several lines of evidence

    suggest that its contribution to cellular physiology could

    be much greater than would be expected from its size

    alone. For instance, (i) it mutates at higher rates than

    nuclear DNA, which may be a consequence of its close

    proximity to the ETC (electron transfer chain); (ii) it

    encodes either polypeptides of ETC or components

    required for their synthesis and, therefore, any coding

    mutations in mtDNA will affect the ETC as a whole;

    this could affect both the assembly and function of the

    products of numerous nuclear genes in ETC complexes;

    (iii) defects in the ETC can have pleiotropic effects

    because they affect cellular energetics as a whole.

    Several lines of evidence indirectly implicate mtDNA

    in longevity. The Framingham Longevity Study of Coro-

    nary Heart Disease has indicated that longevity is morestrongly associated with age of maternal death than that

    of paternal death, suggesting that mtDNA inheritance

    might be involved [17]. On the other hand, longevity

    was shown to be associated with certain mtDNA poly-

    morphisms. ThusItalian male centenarianshave increased

    incidence of mtDNA haplogroup J [18], whereas French

    and Japanese centenarians have increased incidences of

    GA transition at mt9055 and CA transversion at

    mt5178 respectively [19,20]. However, a study of an Irish

    population failed to link longevity to any particular

    mitochondrial haplotype, suggesting that factors other

    than mtDNA polymorphismalso mayplay a role in aging[21].

    Mitochondria have been shown to accumulate high

    levels of lipophilic carcinogens such as polycyclic aro-

    matic hydrocarbons [22,23]. When cells are exposed to

    some of these compounds, mtDNA is damaged pre-

    ferentially [24]. Other mutagenic chemicals also have

    been shown to preferentially target mtDNA [23,2529].

    Therefore it is conceivable that life-long exposure to

    certain environmental toxins could result in a preferential

    accumulation of mtDNA damage and accelerate aging.

    However, perhaps the most relevant kind of insult to

    which mtDNA is exposed is oxidative damage. The lack

    of protective histones and close proximity to the ETC,

    whose complexes I and III are believed to be the pre-

    dominant sites of ROS (reactive oxygen species) pro-

    duction inside the cell, make mtDNA extremely

    vulnerable to oxidative stress. Indeed, the free radical

    theory of aging first put forward by Harman [3033]

    statesthatit is themitochondrial production ofROS, such

    as superoxide and H2O2, and the resulting accumulation

    of damage to macromolecules that causes aging. Cumul-

    ative damage to biological macromolecules was proposed

    to overwhelm the capacity of biological systems to repair

    themselves, resulting in an inevitable functional decline.

    The mitochondrial theory of aging can be considered asan extension and refinement of the free radical theory.

    Its major premise is that mtDNA mutations accumulate

    progressively during life and are directly responsible for

    a measurable deficiency in cellular oxidative phosphor-

    ylation activity, leading to an enhanced ROS production.

    In turn, increased ROS production results in an increased

    rate of mtDNA damage and mutagenesis, thus causing

    a vicious cycle of exponentially increasing oxidative

    damage and dysfunction, which ultimately culminates in

    death (Figure 2). Since Miquel et al. [34] first suggested

    that mtDNA might be damaged in aging, numerous

    studies over the past two decades have supplied a wealth

    C 2004 The Biochemical Society

  • 8/12/2019 Mitochondrial Na (1)

    3/10

    Mitochondrial DNA and aging 357

    Figure 2 Vicious cycle of mtDNA damage

    ROS produced at ETC lead to the inhibition of mtDNA transcription and ETC

    activity, resulting in even higher levels of ROS production.

    of information consistent with the predictions of the free

    radical/mitochondrial theory of aging.

    (i) Oxidative damage in cells and, in

    particular, in mtDNA, is ubiquitous,

    substantial and, like mortality rates,

    increases exponentially with age [35]It has been shown that exhalation of ethane and n-pen-

    thane, indicators of ROS-mediated lipid peroxidation

    increases with age [36]. mtDNA was shown to accu-

    mulate oxidative damage in an age-dependent manner in

    skeletal muscle [37,38], the diaphragm [39,40], cardiac

    muscle [4143] and the brain [44]. Additionally, an age-

    related increase in oxidative damage to mtDNA appears

    to be greater than oxidative damage to nuclear DNA

    in houseflies [45,46]. In rodents, an age-related increase in

    8-oxodG (7,8-dihydro-8-oxoguanine), a mutagenic

    DNA base lesion caused by oxidative stress, was obser-

    ved in mtDNA isolated from the livers of both rats and

    mice [47]. Dietary restriction, which is known to retard

    aging and increase the lifespan in rodents, has been found

    to significantly reduce the age-related accumulation of

    8-oxodG levels in nuclear DNA in all tissues of male

    B6D23F1 mice and in most tissues of male F344 rats. This

    study [47] also showed that dietary restriction prevented

    the age-related increase in 8-oxodG levels in mtDNA

    isolated from the livers of both rats and mice [47].Another study [48] has found that the activities of DNA

    repair enzymes OGG1 (8-oxoguanine-DNA glycosyl-

    ase), hypoxanthine- and uracil-DNA glycosylase in-

    crease in liver extracts of Wistar and OXYS rats with age.

    In both strains, OGG1 activities were approx. 10 times

    greater in nuclear extracts than in mitochondrial ex-

    tracts [48]. However, while OGG1 activity in nuclear

    extracts remained relatively constant during the study,

    this activity increased with age in mitochondrial extracts.

    Importantly, in OXYS rats, which are characterized by

    the overproduction of ROS, high levels of lipid per-

    oxidation, protein oxidation and decreased life span, the

    levels of mitochondrial OGG1 activity were greater than

    in normal Wistar rats, and an increase in this activity

    began earlier [48]. The increase in 8-oxodG levels in

    mtDNA with aging appears to be a general phenomenon

    and has been reported by de Souza-Pinto et al. [49] and

    Hudson et al. [50] The steady-state concentration of

    8-oxodG in mitochondrial DNA was shown to be in-versely correlated with MLSP (maximum lifespan) in the

    heart and brain of mammals, i.e., slowly aging mammals

    show lower 8-oxodG levels in mtDNA than rapidly

    aging ones [51]. Furthermore, these authors show that

    this inverse relationship is restricted to mtDNA, because

    8-oxodG levels in nuclear DNA were not significantly

    correlated with MLSP. The correlationbetween 8-oxodG

    and MLSP was better in the heart and the brain,

    possibly, in part, because these organs are composed

    predominantly of postmitotic cells [51].

    (ii) Longer life expectancy in organismsbelonging to the same cohort group

    is associated with relatively higher

    levels of antioxidants and lower

    concentrations of the products of

    oxygen free radical reactionsAll houseflies lose the ability to fly prior to death.

    Therefore, in an aging population, shorter-lived flies can

    be identified as flightless crawlers in contrast with their

    longer-lived cohorts, the fliers. The average lifespan of

    crawlers is about one-third shorter than the fliers. Levels

    of antioxidant defences [SOD (superoxide dismutase),

    CAT (catalase) and glutathione] and products of oxygen

    free radical reactions [inorganic peroxides and TBA(thio-

    barbituric acid) reactants] were compared between craw-

    lers and fliers. The fliers showed greater SOD and CAT

    activities and glutathione concentrations than crawlers,

    whereas the amount of inorganic peroxides (H2O2) and

    TBA reactants was higher in the crawlers than in fliers

    [52].

    (iii) An experimentally induced decrease

    in oxidative stress retards age-associated

    deterioration at the organelle andorganismic levels and extends

    chronological and metabolic life spanSimultaneous overexpression of Cu/ZnSOD and CAT in

    Drosophilawas shown [53] to increase the maximum and

    average lifespan by one-third, to retard the age-related

    accumulation of oxidative damage to DNA and protein,

    to increase resistance to the oxidative effects of X-ray ex-

    posure, to attenuate the age-related increase in the rate of

    mitochondrial H2O2 generation, to increase the speed

    of walking and to increase the metabolic potential defined

    as the total amount of oxygen consumed per unit body

    weight during the adult life. Moreover, others have been

    C 2004 The Biochemical Society

  • 8/12/2019 Mitochondrial Na (1)

    4/10

  • 8/12/2019 Mitochondrial Na (1)

    5/10

    Mitochondrial DNA and aging 359

    role in the aging process [47,50,90,91]. The important role

    played by DNA-repair enzymes in the accumulation of

    this lesion is underlined by the fact that liver mtDNA

    from knockout mice for OGG1 (the glycosylase that

    recognizes this lesion) accumulates 30 times as much

    8-oxodG as mtDNA of wild-type control mice [91].

    Interestingly, several studies have indicated that OGG1activity in mitochondrial extracts from old rats is higher

    compared with extracts from young animals, which is

    apparently at odds with the observed accumulation of

    8-oxodG in mtDNA from older animals [49,50,91,92].

    However, this controversy was recently resolved by

    Szczesny et al. [93], who have shown that, in both hepa-

    tocytes from oldmice and in senescenthuman fibroblasts,

    mitochondrial import of OGG1 is impaired and that a

    significant fraction of this enzyme remains localized in

    the outer membrane and intermembrane space in the pre-

    cursor form. Because uracil-DNA glycosylase, another

    BER enzyme, has a similar age-dependent impairment inmitochondrial import, these authors [93] conclude that

    there appears to be a general deficiency in the import of

    BER enzymes into mitochondria from senescent animals

    and cells. Therefore, although age-related impairment in

    mitochondrial protein import does not necessarily ex-

    tend beyond some DNA repair enzymes, it can explain

    the simultaneous accumulationof 8-oxodG and increased

    in vitroOGG1 activity in mitochondria from senescent

    animals in some settings. It should be noted, however,

    that other studies point out that the ability of mitochon-

    dria to import proteins is a dynamic function that can be

    modulated by various stimuli, such as thyroid hormone

    [94], which may provide an alternative explanation for

    the observed phenomena. Moreover, the rate of mito-

    chondrial import of matrix chaperonins GRP75(glucose-

    regulated protein 75) and HSP60 (heat-shock protein

    60), which are essential for the import of precursor pro-

    teins, in fact increases with age [95]. Also, there is an

    apparent lack of consensus in the literature regarding the

    increase in mitochondrial OGG1 activity with aging in

    both senescent cells and animals. Some groups have re-

    ported a decline in both activity and expression of several

    key mitochondrial repairenzymes, including OGG1[96

    98]. Therefore, to date, there is no consensus on the ques-

    tion of whether DNA repair declines with age [99]. Oneof the possible explanations for such discrepancies is in

    theintrinsic variability between cell types andtissues with

    respect of their reliance on antioxidant defences com-

    pared with DNA repair for the maintenance of struc-

    tural integrity of mtDNA. Results from our laboratory

    [100,101] suggest that the differential susceptibility of

    glial cell types to oxidative damage and apoptosis does

    not appear to be related to cellular antioxidant capacity,

    but rather to differences in their ability to repair oxida-

    tive mtDNA damage [100], and that different glial cell

    types differ in their capacity to repair oxidative damage

    [101].

    CHALLENGES TO THE MITOCHONDRIAL

    THEORY OF AGING

    Recently, the rate of ROS generation by mitochondria

    under physiological conditions, which is a cornerstone

    of the mitochondrial theory of aging, has been critically

    re-examined by several groups. Hansford et al. [102]have found that active H2O2 production (an indirect

    measure of O2 generation) requires both a high frac-

    tional reduction of complex I (indexed by NADH/

    NAD+:NADH ratio) and a high membrane potential,

    . These conditions are only achieved with supra-

    physiological concentrations of succinate. With physio-

    logical concentrations of NAD-linked substrates, rates of

    H2O2 formation are much lower (less than 0.1 % of re-

    spiratory chain electron flux). This H2O2 production

    may be stimulated by the complex III inhibitor antimy-

    cin A, but not by myxothiazol [102]. Staniek and Nohl

    [103,104] reported further that mitochondria respiringon complex I and complex II substrates generate detec-

    table H2O2 only in the presence of the complex III

    inhibitor antimycin. They also suggested that the rates

    of mitochondrial H2O2 production reported by others

    are artificially high due to flaws in experimental design

    [103,104]. Martin Brands group [105] capitalized on

    these findings and used an improved experimental design

    to show that mitochondria do not release measurable

    amounts of superoxide or H2O2 when respiring on

    complex I or complex II substrates, but release significant

    amounts of superoxide from complex I when respiring

    on palmitoyl carnitine. However, even at saturating con-

    centrations of palmitoyl carnitine, in their estimation,

    only0.15 % oftheelectron flow gives riseto H2O2under

    resting conditions with a respiration rate of 200 nmol

    electrons min1 mg1 of mitochondrial protein. Under

    physiological conditions, this rate should be even lower

    due to (i) lower partial oxygen pressure, (ii) lower

    concentration of palmitoyl carnitine and (iii) lower mito-

    chondrial membrane potential. Therefore, under physio-

    logical conditions in cells with uncompromised antioxi-

    dant defences, ROS are produced by ETC in quantities

    that can be efficiently scavenged by mitochondrial anti-

    oxidant systems. As a consequence, no significant oxi-

    dative damage should be inflicted on mitochondrialcomponents, including mtDNA due to electron leak

    from ETC, provided that cells have normal levels of

    antioxidants. This conclusion is in agreement with the

    observations of Orr et al. [106], who have recently re-

    examined their earlier findings and those of others on the

    effectof overexpressionof antioxidantenzymes on exten-

    sion of Drosophila lifespan. They have found that

    significant increases in the activities of both Cu/ZnSOD

    and CAT had no beneficial effect on survivorship in

    relatively long-livedy w mutant flies, and were associated

    with slightly decreased life spans in wild-type flies of

    the Oregon-R strain. The introduction of additional

    C 2004 The Biochemical Society

  • 8/12/2019 Mitochondrial Na (1)

    6/10

    360 M. F. Alexeyev, S. P. LeDoux and G. L. Wilson

    transgenes encoding MnSOD or thioredoxin reductase in

    the same genetic background also failed to cause life span

    extension. These authors [106] conclude that increasing

    the activities of major antioxidative enzymes above wild-

    type levels does not decrease the rate of aging in long-

    lived strains ofDrosophila, although there may be some

    effect in relatively short-lived strains [106]. In line withthis conclusion, Van Remmen et al. [107] in their study

    of mice heterozygous for the MnSOD gene knockout

    have found that, although life-long reduction of MnSOD

    activity leads to increased levels of oxidative damage to

    mitochondrial and nuclear DNA and increased cancer

    incidence, it does not appear to affect aging.

    A report on the requirement of cytosolic CAT for

    phenotypic lifespan extension inCaenorhabditis elegans

    daf-C and clk-1 mutants [108] was recently retracted

    [109], dealing another blow to the notion that lifespan can

    be routinely enhanced by increasingantioxidantdefences.

    Rasmussen et al. [110,111] assayed 13 different en-zyme activities using optimized preparation techniques

    and found that the central bioenergetic systems, includ-

    ing pyruvate dehydrogenase, tricarboxylic acid cycle,

    respiratory chain and ATP synthesis, appeared unaltered

    with age. Maklashina and Ackrell [112] have recently

    critically examined the literature regarding the role of

    ETC dysfunction in aging. They conclude that the

    available evidence for age-related inactivation of the res-

    piratory chain can be challenged because of the prepar-

    ation purity and the use of inadequate assay procedures,

    and that recent experimental evidence does not support

    the mitochondrial theory of aging.

    In contrast, Jacobs [113] does not so much challenge

    experimental evidence supporting the mitochondrial

    theory of aging as to point out that all the evidence

    available to date is indirect in its nature. He argues that

    studiesperformedsofardonotaddressthecriticalissueof

    cause andeffect, i.e. does thesomatic mutation of mtDNA

    result in oxidative phosphorylation dysfunction and

    increased oxidative stress? Does increased oxidative stress

    promote mtDNA mutagenesis? Finally, he points out that

    the results from his own laboratory [114] suggest that,

    at least in a tissue culture model, progressive accumul-

    ation of mtDNA mutations due to expression of proof-

    reading-deficient DNA polymerase does not lead tosignificant phenotypic changes, despite the accumulation

    of mtDNA mutations to the level three times higher than

    that found in aged tissues. However, very recently [115],

    the concerted effort of several groups (including that of

    Howard Jacobs) led by Nils-Goran Larsson resulted in

    the generationof homozygousknock-inmice thatexpress

    a proofreading-deficient catalytic subunit of mitochon-

    drial DNA polymerase . These mice develop an

    mtDNA mutator phenotype with a 35-fold increase

    in the levels of point mutations, as well as increased

    amounts of deleted mtDNA. This increase in somatic

    mtDNA mutations is associated with reduced lifespan

    and the premature onset of age-related phenotypes such

    as weight loss, reduced subcutaneous fat, alopecia (hair

    loss), kyphosis (curvature of the spine), osteoporosis,

    anaemia, reduced fertility and heart enlargement [115].

    Thus results of this study provide the best evidence so

    far for a causative link between mtDNA mutations and

    aging phenotypes in mammals. However, as these authorsconcede, the detailed kinetics of the accumulation of

    somatic mtDNA mutations remain to be elucidated. The

    mutation load in the brain of mutator mice at 2 months

    of age is already 23-fold greater than in 6-month-

    old wild-type littermates. This, and the rather uniform

    mutation loads between tissues, suggests that much of the

    accumulation of mutations may occur during embryonic

    and/or fetal development. Also, the onset of premature

    aging in this model is not accompanied, temporally, by a

    large de novo accumulation of mtDNA mutations

    around 6 months. Therefore it appears plausible that the

    premature onset of aging in this model is the resultof the cumulative physiological damage caused by the

    high mutation load present during adult life and/or to

    segregation or clonal expansion of specific mutations, as

    supported by the observed mosaicism for the respiratory

    chain deficiency found in the heart. However, since the

    effects of high mutational burden in mtDNA during

    embryonic and fetal development are poorly understood,

    one has to consider the possibility that premature aging

    in this model is predetermined at the prenatal, rather

    than postnatal, stage. If this holds true, then the issue of

    causative relationship between mtDNA mutations and

    normal aging remains open, since, in normal aging, accu-

    mulation of mtDNA mutations occurs postnatally. The

    observation that knock-in mice show some features (e.g.

    alopecia) that are more characteristic of human, rather

    than mouse aging lends some credibility to the notion

    that a high mutational load in mtDNA at the prenatal

    stage might be an important limitation of this model.

    Clearly, a model with inducible mtDNA mutator pheno-

    type should help to resolve many of these outstanding

    issues. Finally, mtDNA mutations in this study [115] are

    generated by a mutator DNA polymerase rather than

    by oxidative DNAdamage. Therefore this study does not

    address one of the central premises of the mitochondrial

    theory of aging, namely that oxidative mtDNA damageis the driving force behind the accumulation of mtDNA

    mutations.

    In the preceding paragraphs we have discussed support

    provided for the mitochondrial theory of aging by the

    discovery of mitochondrial disease. One might then re-

    asonably expect, based on the mitochondrial theory of

    aging, that mitochondrial ROS would be causative in

    a significant fraction of pathogenic mtDNA mutations.

    However, an analysis of 188 pathogenic mtDNA point

    mutations [65] revealed that the mutagenic effect of

    8-oxodG, widely regarded as a prime lesion resulting

    from an oxidative insult to DNA, can be implicated in

    C 2004 The Biochemical Society

  • 8/12/2019 Mitochondrial Na (1)

    7/10

    Mitochondrial DNA and aging 361

    the aetiology of only a few mutations. Indeed, unrepaired

    8-oxodG in mtDNA can pair with both C and A with

    almost equal efficiency resulting in GT (and CA

    on complementary strand) transversions, which account

    for only 5.9 % of pathogenic mtDNA mutations. Even

    when the potentially mutagenic pool of 8-oxodGTP

    (8-oxodeoxyGTP), the product of cytoplasmic/matrixdGTP pool oxidation) is taken in consideration (TG

    and AC transversions), the cumulative impact of both

    types of mutation is still only 8.5 %. For comparison,

    82 % (almost 10 times as many) of the pathogenic point

    mutations in mtDNA can be attributed to deamination of

    adenine and cytosine. 8-oxodG is not a prime oxidative

    mutagenic lesion, because it is efficiently repaired by

    BER pathways. Thus the exact factors required for the

    accumulation of point mutations have yet to be fully

    defined. Oxidative DNA damage can produce a variety

    of base lesions whose mutagenic potential has not been

    fully elucidated [116]. Therefore it is possible that other,less prominent and, at present, unidentified lesions are

    responsible for the bulk of ROS-mediated mutagenesis.

    Alternatively, it can be postulated that ROS do not play

    a major role in mtDNA mutagenesis.

    CONCLUSIONS

    Aging is a complex multifactorial process which we

    have only begun to understand. Although the available

    evidence strongly suggests that mitochondria play a role

    in this process, there appears to be a wide range of opi-

    nions as to the exact nature of the involvement ofmitochondria in aging. In this review, we have made an

    attempt to present in a balanced fashion theexisting views

    on the involvement of mtDNA in aging. Clearly, more

    research must be done to fully elucidate how damage to

    mtDNA contributes to the aging process. It is possible

    that the seemingly contradictory results of different

    studies will be reconciled or explained through the use of

    integrative approaches and unified model systems. The

    next few years of aging research should prove exciting

    as our knowledge expands dramatically as the results

    of studies from several laboratories, now experimentally

    testing the mitochondrial theory of aging, are published.

    REFERENCES

    1 Mandavilli, B. S., Santos, J. H. and Van Houten, B. (2002)Mitochondrial DNA repair and aging. Mutat. Res. 509,127151

    2 Price, G. B., Modak, S. P. and Makinodan, T. (1971)Age-associated changes in the DNA of mouse tissue.Science (Washington, D.C.)171, 917920

    3 Wheeler, K. T. and Lett, J. T. (1974) On the possibilitythat DNA repair is related to age in non-dividing cells.Proc. Natl. Acad. Sci. U.S.A.71, 18621865

    4 Anderson, S., Bankier, A. T., Barrell, B. G. et al. (1981)Sequence and organization of the human mitochondrialgenome. Nature (London)290, 457465

    5 Kvist, L., Martens, J., Higuchi, H., Nazarenko, A. A.,Valchuk, O. P. and Orell, M. (2003) Evolution and geneticstructure of the great tit (Parus major) complex.Proc. R. Soc. Lond. Ser. B.270, 14471454

    6 Schwartz, M. and Vissing, J. (2003) New patterns ofinheritance in mitochondrial disease. Biochem. Biophys.Res. Commun.310, 247251

    7 Gyllensten, U., Wharton, D., Josefsson, A. and Wilson,

    A. C. (1991) Paternal inheritance of mitochondrial DNAin mice. Nature (London)352, 255257

    8 Takamatsu, C., Umeda, S., Ohsato, T. et al. (2002)Regulation of mitochondrial D-loops by transcriptionfactor A and single-stranded DNA-binding protein.EMBO Rep.3, 451456

    9 Cavelier, L., Johannisson, A. and Gyllensten, U. (2000)Analysis of mtDNA copy number and composition ofsingle mitochondrial particles using flow cytometry andPCR. Exp. Cell Res.259, 7985

    10 Kasamatsu, H. and Vinograd, J. (1974) Replication ofcircular DNA in eukaryotic cells. Annu. Rev. Biochem.43, 695719

    11 Ojala, D., Montoya, J. and Attardi, G. (1981) tRNApunctuation model of RNA processing in humanmitochondria. Nature (London)290, 470474

    12 Taylor, S. W., Fahy, E. and Ghosh, S. S. (2003) Global

    organellar proteomics. Trends Biotechnol.21, 828813 Lim, S. E., Longley, M. J. and Copeland, W. C. (1999) The

    mitochondrial p55 accessory subunit of human DNApolymerase enhances DNA binding, promotesprocessive DNA synthesis, and confersN-ethylmaleimideresistance. J. Biol. Chem.274, 3819738203

    14 Taanman, J. W. (1999) The mitochondrial genome:structure, transcription, translation and replication.Biochim. Biophys. Acta1410, 103123

    15 Mootha, V. K., Bunkenborg, J., Olsen, J. V. et al. (2003)Integrated analysis of protein composition, tissuediversity, and gene regulation in mouse mitochondria.Cell (Cambridge, Mass.)115, 629640

    16 Wallace, D. C. (1992) Mitochondrial genetics:a paradigm for aging and degenerative diseases?Science (Washington, D.C.)256, 628632

    17 Brand, F. N., Kiely, D. K., Kannel, W. B. and Myers, R. H.(1992) Family patterns of coronary heart diseasemortality: the Framingham Longevity Study.

    J. Clin. Epidemiol.45, 16917418 De Benedictis, G., Rose, G., Carrieri, G. et al. (1999)

    Mitochondrial DNA inherited variants are associatedwith successful aging and longevity in humans.FASEB J.13, 15321536

    19 Tanaka, M., Gong, J. S., Zhang, J., Yoneda, M. andYagi, K. (1998) Mitochondrial genotype associated withlongevity. Lancet351, 185186

    20 Ivanova, R., Lepage, V., Charron, D. and Schachter, F.(1998) Mitochondrial genotype associated with FrenchCaucasian centenarians. Gerontology44, 349

    21 Ross, O. A., McCormack, R., Curran, M. D. et al. (2001)Mitochondrial DNA polymorphism: its role in longevityof the Irish population. Exp. Gerontol.36, 11611178

    22 Allen, J. A. and Coombs, M. M. (1980) Covalent bindingof polycyclic aromatic compounds to mitochondrial andnuclear DNA. Nature (London)287, 244245

    23 Wunderlich, V., Schutt, M., Bottger, M. and Graffi, A.(1970) Preferential alkylation of mitochondrialdeoxyribonucleic acid byN-methyl-N-nitrosourea.Biochem. J.118, 99109

    24 Backer, J. M. and Weinstein, I. B. (1980) MitochondrialDNA is a major cellular target for a dihydrodiol-epoxidederivative of benzo[a]pyrene. Science (Washington, D.C.)209, 297299

    25 Rossi, S. C., Gorman, N. and Wetterhahn, K. E. (1988)Mitochondrial reduction of the carcinogen chromate:formation of chromium(V). Chem. Res. Toxicol.1,101107

    26 Miyaki, M., Yatagai, K. and Ono, T. (1977) Strand breaksof mammalian mitochondrial DNA induced bycarcinogens. Chem. Biol. Interact.17, 321329

    C 2004 The Biochemical Society

  • 8/12/2019 Mitochondrial Na (1)

    8/10

    362 M. F. Alexeyev, S. P. LeDoux and G. L. Wilson

    27 Neubert, D., Hopfenmuller, W. and Fuchs, G. (1981)Manifestation of carcinogenesis as a stochastic processon the basis of an altered mitochondrial genome.Arch. Toxicol.48, 89125

    28 Niranjan, B. G., Bhat, N. K. and Avadhani, N. G. (1982)Preferential attack of mitochondrial DNA by aflatoxin B1during hepatocarcinogenesis. Science (Washington, D.C.)215, 7375

    29 Tomasi, A., Albano, E., Banni, S. et al. (1987) Free-radicalmetabolism of carbon tetrachloride in rat livermitochondria. A study of the mechanism of activation.Biochem. J.246, 313317

    30 Harman, D. (1972) Free radical theory of aging: dietaryimplications. Am. J. Clin. Nutr.25, 839843

    31 Harman, D. (1973) Free radical theory of aging. Triangle12, 153158

    32 Harman, D. (1972) The biologic clock: the mitochondria?J. Am. Geriatr. Soc.20, 145147

    33 Harman, D. (1956) Aging: a theory based on free radicaland radiation chemistry. J. Gerontol.11, 298300

    34 Miquel, J., Economos, A. C., Fleming, J. and Johnson, Jr,J. E. (1980) Mitochondrial role in cell aging.Exp. Gerontol.15, 575591

    35 Sohal, R. S. and Weindruch, R. (1996) Oxidative stress,caloric restriction, and aging. Science (Washington, D.C.)273, 5963

    36 Matsuo, M., Gomi, F., Kuramoto, K. and Sagai, M. (1993)Food restriction suppresses an age-dependent increase inthe exhalation rate of pentane from rats: a longitudinalstudy. J. Gerontol.48, B133B136

    37 Katayama, M., Tanaka, M., Yamamoto, H., Ohbayashi, T.,Nimura, Y. and Ozawa, T. (1991) Deleted mitochondrialDNA in the skeletal muscle of aged individuals.Biochem. Int.25, 4756

    38 Lee, C. M., Chung, S. S., Kaczkowski, J. M.,Weindruch, R. and Aiken, J. M. (1993) Multiplemitochondrial DNA deletions associated with age inskeletal muscle of rhesus monkeys. J. Gerontol.48,B201B205

    39 Hayakawa, M., Torii, K., Sugiyama, S., Tanaka, M. andOzawa, T. (1991) Age-associated accumulation of8-hydroxydeoxyguanosine in mitochondrial DNA ofhuman diaphragm. Biochem. Biophys. Res. Commun.

    179, 1023102940 Torii, K., Sugiyama, S., Tanaka, M. et al. (1992) Aging-associated deletions of human diaphragmaticmitochondrial DNA. Am. J. Respir. Cell. Mol. Biol.6,543549

    41 Hayakawa, M., Sugiyama, S., Hattori, K., Takasawa, M.and Ozawa, T. (1993) Age-associated damage inmitochondrial DNA in human hearts.Mol. Cell. Biochem.119, 95103

    42 Marin-Garcia, J., Zoubenko, O. and Goldenthal, M. J.(2002) Mutations in the cardiac mitochondrial DNAcontrol region associated with cardiomyopathy and aging.

    J. Cardiac Fail.8, 9310043 Lai, L. P., Tsai, C. C., Su, M. J. et al. (2003) Atrial

    fibrillation is associated with accumulation of aging-related common type mitochondrial DNA deletionmutation in human atrial tissue. Chest123, 539544

    44 Corral-Debrinski, M., Horton, T., Lott, M. T., Shoffner,

    J. M., Beal, M. F. and Wallace, D. C. (1992) MitochondrialDNA deletions in human brain: regional variability andincrease with advanced age. Nat. Genet.2, 324329

    45 Ames, B. N., Shigenaga, M. K. and Hagen, T. M. (1993)Oxidants, antioxidants, and the degenerative diseases ofaging. Proc. Natl. Acad. Sci. U.S.A.90, 79157922

    46 Agarwal, S. and Sohal, R. S. (1994) DNA oxidativedamage and life expectancy in houseflies. Proc. Natl.Acad. Sci. U.S.A.91, 1233212335

    47 Hamilton, M. L., Van Remmen, H., Drake, J. A. et al.(2001) Does oxidative damage to DNA increase with age?Proc. Natl. Acad. Sci. U.S.A.98, 1046910474

    48 Ishchenko, A., Sinitsyna, O., Krysanova, Z. et al. (2003)Age-dependent increase of 8-oxoguanine-,hypoxanthine-, and uracil-DNA glycosylase activities inliver extracts from OXYS rats with inheritedovergeneration of free radicals and Wistar rats.Med. Sci. Monit.9, BR16BR24

    49 de Souza-Pinto, N. C., Hogue, B. A. and Bohr, V. A.(2001) DNA repair and aging in mouse liver: 8-oxodGglycosylase activity increase in mitochondrial but not innuclear extracts. Free Radical Biol. Med.30, 916923

    50 Hudson, E. K., Hogue, B. A., Souza-Pinto, N. C. et al.(1998) Age-associated change in mitochondrial DNAdamage. Free Radical Res.29, 573579

    51 Barja, G. and Herrero, A. (2000) Oxidative damage to

    mitochondrial DNA is inversely related to maximum lifespan in the heart and brain of mammals. FASEB J. 14,312318

    52 Sohal, R. S., Toy, P. L. and Allen, R. G. (1986)Relationship between life expectancy, endogenousantioxidants and products of oxygen free radical reactionsin the housefly,Musca domestica.Mech. Ageing Dev.36,7177

    53 Orr, W. C. and Sohal, R. S. (1994) Extension of life-spanby overexpression of superoxide dismutase and catalase inDrosophila melanogaster.Science (Washington, D.C.)263,11281130

    54 Sun, J., Folk, D., Bradley, T. J. and Tower, J. (2002)Induced overexpression of mitochondrial Mn-superoxidedismutase extends the life span of adult Drosophilamelanogaster. Genetics161, 661672

    55 Parkes, T. L., Elia, A. J., Dickinson, D., Hilliker, A. J.,

    Phillips, J. P. and Boulianne, G. L. (1998) Extension ofDrosophilalifespan by overexpression of human SOD1 inmotorneurons. Nat. Genet.19, 171174

    56 Orr, W. C. and Sohal, R. S. (1993) Effects of Cu-Znsuperoxide dismutase overexpression of life span andresistance to oxidative stress in transgenicDrosophilamelanogaster. Arch. Biochem. Biophys.301, 3440

    57 Gallagher, I. M., Jenner, P., Glover, V. and Clow, A. (2000)CuZn-superoxide dismutase transgenic mice: no effect onlongevity, locomotor activity and3 H-mazindol and3H-spiperone binding over 19 months. Neurosci. Lett.289, 221223

    58 Harris, N., Costa, V., MacLean, M., Mollapour, M.,Moradas-Ferreira, P. and Piper, P. W. (2003) Mnsodoverexpression extends the yeast chronological (G0) lifespan but acts independently of Sir2p histone deacetylaseto shorten the replicative life span of dividing cells.Free Radical Biol. Med.34, 15991606

    59 Brunet-Rossinni, A. K. (2004) Reduced free-radicalproduction and extreme longevity in the little brown bat(Myotis lucifugus) versus two non-flying mammals.Mech. Ageing Dev.125, 1120

    60 Weindruch, R., Walford, R. L., Fligiel, S. and Guthrie, D.(1986) The retardation of aging in mice by dietaryrestriction: longevity, cancer, immunity and lifetimeenergy intake. J. Nutr.116, 641654

    61 Lee, D. W. and Yu, B. P. (1990) Modulation of freeradicals and superoxide dismutases by age and dietaryrestriction. Aging2, 357362

    62 Wallace, D. C., Singh, G., Lott, M. T. et al. (1988)Mitochondrial DNA mutation associated with Lebershereditary optic neuropathy. Science (Washington, D.C.)242, 14271430

    63 Holt, I. J., Harding, A. E. and Morgan-Hughes, J. A.(1988) Deletions of muscle mitochondrial DNA inpatients with mitochondrial myopathies.Nature (London)331, 717719

    64 Lestienne, P. and Ponsot, G. (1988) Kearns-Sayresyndrome with muscle mitochondrial DNA deletion.Lanceti, 885

    65 Reference deleted66 Geromel, V., Kadhom, N., Cebalos-Picot, I. et al. (2001)

    Superoxide-induced massive apoptosis in cultured skinfibroblasts harboring the neurogenic ataxia retinitispigmentosa (NARP) mutation in the ATPase-6 gene ofthe mitochondrial DNA. Hum. Mol. Genet.10,12211228

    67 Ohkoshi, N., Mizusawa, H., Shiraiwa, N., Shoji, S.,Harada, K. and Yoshizawa, K. (1995) Superoxidedismutases of muscle in mitochondrialencephalomyopathies. Muscle Nerve18, 12651271

    C 2004 The Biochemical Society

  • 8/12/2019 Mitochondrial Na (1)

    9/10

    Mitochondrial DNA and aging 363

    68 Filosto, M., Tonin, P., Vattemi, G., Spagnolo, M.,Rizzuto, N. and Tomelleri, G. (2002) Antioxidant agentshave a different expression pattern in muscle fibers ofpatients with mitochondrial diseases.Acta Neuropathol. (Berl.).103, 215220

    69 Lu, C. Y., Wang, E. K., Lee, H. C., Tsay, H. J. and Wei,Y. H. (2003) Increased expression of manganese-superoxide dismutase in fibroblasts of patients with

    CPEO syndrome. Mol. Genet. Metab.80, 32132970 Kunishige, M., Mitsui, T., Akaike, M. et al. (2003)

    Overexpressions of myoglobin and antioxidant enzymesin ragged-red fibers of skeletal muscle from patients withmitochondrial encephalomyopathy. Muscle Nerve 28,484492

    71 Geromel, V., Rotig, A., Munnich, A. and Rustin, P. (2002)Coenzyme Q10 depletion is comparatively lessdetrimental to human cultured skin fibroblasts thanrespiratory chain complex deficiencies. Free Radical Res.36, 375379

    72 Gross, N. J., Getz, G. S. and Rabinowitz, M. (1969)Apparent turnover of mitochondrial deoxyribonucleicacid and mitochondrial phospholipids in the tissues of therat. J. Biol. Chem.244, 15521562

    73 Lee, C. K., Klopp, R. G., Weindruch, R. and Prolla, T. A.(1999) Gene expression profile of aging and its retardation

    by caloric restriction. Science (Washington, D.C.)285,1390139374 Gadaleta, M. N., Petruzzella, V., Renis, M., Fracasso, F.

    and Cantatore, P. (1990) Reduced transcription ofmitochondrial DNA in the senescent rat. Tissuedependence and effect ofl-carnitine. Eur. J. Biochem.187, 501506

    75 Calleja, M., Pena, P., Ugalde, C., Ferreiro, C., Marco, R.and Garesse, R. (1993) Mitochondrial DNA remainsintact duringDrosophilaaging, but the levels ofmitochondrial transcripts are significantly reduced.

    J. Biol. Chem.268, 188911889776 Clayton, D. A., Doda, J. N. and Friedberg, E. C. (1975)

    Absence of a pyrimidine dimer repair mechanism formitochondrial DNA in mouse and human cells.Basic Life Sci.5B, 589591

    77 Clayton, D. A., Doda, J. N. and Friedberg, E. C. (1974)The absence of a pyrimidine dimer repair mechanism in

    mammalian mitochondria. Proc. Natl. Acad. Sci. U.S.A.71, 27772781

    78 Bohr, V. A., Smith, C. A., Okumoto, D. S. and Hanawalt,P. C. (1985) DNA repair in an active gene: removal ofpyrimidine dimers from the DHFR gene of CHO cellsis much more efficient than in the genome overall.Cell (Cambridge, Mass.)40, 359369

    79 LeDoux, S. P. and Wilson, G. L. (2001) Base excisionrepair of mitochondrial DNA damage in mammaliancells. Prog. Nucleic Acid Res. Mol. Biol.68, 273284

    80 LeDoux, S. P., Driggers, W. J., Hollensworth, B. S. andWilson, G. L. (1999) Repair of alkylation and oxidativedamage in mitochondrial DNA. Mutat. Res.434, 149159

    81 LeDoux, S. P., Patton, N. J., Avery, L. J. and Wilson, G. L.(1993) Repair of N-methylpurines in the mitochondrialDNA of xeroderma pigmentosum complementationgroup D cells. Carcinogenesis14, 913917

    82 Driggers, W. J., LeDoux, S. P. and Wilson, G. L. (1993)Repair of oxidative damage within the mitochondrialDNA of RINr 38 cells. J. Biol. Chem.268, 2204222045

    83 Shen, C. C., Wertelecki, W., Driggers, W. J., LeDoux, S. P.and Wilson, G. L. (1995) Repair of mitochondrial DNAdamage induced by bleomycin in human cells. Mutat. Res.337, 1923

    84 Driggers, W. J., Grishko, V. I., LeDoux, S. P. and Wilson,G. L. (1996) Defective repair of oxidative damage in themitochondrial DNA of a xeroderma pigmentosum groupA cell line. Cancer Res.56, 12621266

    85 Druzhyna, N., Nair, R. G., LeDoux, S. P. and Wilson,G. L. (1998) Defective repair of oxidative damage inmitochondrial DNA in Downs syndrome. Mutat. Res.409, 8189

    86 Grishko, V. I., Druzhyna, N., LeDoux, S. P. and Wilson,G. L. (1999) Nitric oxide-induced damage to mtDNA andits subsequent repair. Nucleic Acids Res.27, 45104516

    87 LeDoux, S. P., Wilson, G. L., Beecham, E. J., Stevnsner, T.,Wassermann, K. and Bohr, V. A. (1992) Repair ofmitochondrial DNA after various types of DNA damagein Chinese hamster ovary cells. Carcinogenesis13,19671973

    88 Pettepher, C. C., LeDoux, S. P., Bohr, V. A. and Wilson,G. L. (1991) Repair of alkali-labile sites within themitochondrial DNA of RINr 38 cells after exposure tothe nitrosourea streptozotocin. J. Biol. Chem.266,31133117

    89 Bohr, V. A., Stevnsner, T. and de Souza-Pinto, N. C.(2002) Mitochondrial DNA repair of oxidative damage inmammalian cells. Gene286, 127134

    90 Shigenaga, M. K., Hagen, T. M. and Ames, B. N. (1994)Oxidative damage and mitochondrial decay in aging.Proc. Natl. Acad. Sci. U.S.A.91, 1077110778

    91 de Souza-Pinto, N. C., Eide, L., Hogue, B. A. et al. (2001)Repair of 8-oxodeoxyguanosine lesions in mitochondrialDNA depends on the oxoguanine DNA glycosylase(OGG1) gene and 8-oxoguanine accumulates in themitochondrial DNA of OGG1-defective mice.Cancer Res.61, 53785381

    92 Souza-Pinto, N. C., Croteau, D. L., Hudson, E. K.,Hansford, R. G. and Bohr, V. A. (1999) Age-associated

    increase in 8-oxo-deoxyguanosine glycosylase/AP lyaseactivity in rat mitochondria. Nucleic Acids Res.27,19351942

    93 Szczesny, B., Hazra, T. K., Papaconstantinou, J., Mitra, S.and Boldogh, I. (2003) Age-dependent deficiency inimport of mitochondrial DNA glycosylases required forrepair of oxidatively damaged bases. Proc. Natl.Acad. Sci. U.S.A.100, 1067010675

    94 Craig, E. E., Chesley, A. and Hood, D. A. (1998) Thyroidhormone modifies mitochondrial phenotype byincreasing protein import without altering degradation.Am. J. Physiol.275, C1508C1515

    95 Craig, E. E. and Hood, D. A. (1997) Influence of aging onprotein import into cardiac mitochondria. Am. J. Physiol.272, H2983H2988

    96 Chen, D., Cao, G., Hastings, T. et al. (2002) Age-dependent decline of DNA repair activity for oxidativelesions in rat brain mitochondria. J. Neurochem.81,12731284

    97 Shen, G. P., Galick, H., Inoue, M. and Wallace, S. S. (2003)Decline of nuclear and mitochondrial oxidative baseexcision repair activity in late passage human diploidfibroblasts. DNA Repair2, 673693

    98 Kikuchi, H., Furuta, A., Nishioka, K., Suzuki, S. O.,Nakabeppu, Y. and Iwaki, T. (2002) Impairment ofmitochondrial DNA repair enzymes against accumulationof 8-oxo-guanine in the spinal motor neurons ofamyotrophic lateral sclerosis. Acta Neuropathol (Berl/).103, 408414

    99 Bohr, V. A. (2002) Repair of oxidative DNA damage innuclear and mitochondrial DNA, and some changes withaging in mammalian cells. Free Radical Biol. Med. 32,804812

    100 Hollensworth, S. B., Shen, C., Sim, J. E., Spitz, D. R.,Wilson, G. L. and LeDoux, S. P. (2000) Glial cell type-specific responses to menadione-induced oxidative stress.Free Radical Biol. Med.28, 11611174

    101 Druzhyna, N. M., Hollensworth, S. B., Kelley, M. R.,Wilson, G. L. and Ledoux, S. P. (2003) Targeting human8-oxoguanine glycosylase to mitochondria ofoligodendrocytes protects against menadione-inducedoxidative stress. Glia42, 370378

    102 Hansford, R. G., Hogue, B. A. and Mildaziene, V. (1997)Dependence of H2O2formation by rat heartmitochondria on substrate availability and donor age.

    J. Bioenerg. Biomembr.29, 8995103 Staniek, K. and Nohl, H. (1999) H2O2detection from

    intact mitochondria as a measure for one-electronreduction of dioxygen requires a non-invasive assaysystem. Biochim. Biophys. Acta1413, 7080

    C 2004 The Biochemical Society

  • 8/12/2019 Mitochondrial Na (1)

    10/10

    364 M. F. Alexeyev, S. P. LeDoux and G. L. Wilson

    104 Staniek, K. and Nohl, H. (2000) Are mitochondria apermanent source of reactive oxygen species?Biochim. Biophys. Acta1460, 268275

    105 St-Pierre, J., Buckingham, J. A., Roebuck, S. J. and Brand,M. D. (2002) Topology of superoxide production fromdifferent sites in the mitochondrial electron transportchain. J. Biol. Chem.277, 4478444790

    106 Orr, W. C., Mockett, R. J., Benes, J. J. and Sohal, R. S.

    (2003) Effects of overexpression of copper-zinc andmanganese superoxide dismutases, catalase, andthioredoxin reductase genes on longevity inDrosophilamelanogaster. J. Biol. Chem.278, 2641826422

    107 Van Remmen, H., Ikeno, Y., Hamilton, M. et al. (2003)Life-long reduction in MnSOD activity results inincreased DNA damage and higher incidence of cancerbut does not accelerate aging. Physiol. Genomics 16,2937

    108 Taub, J., Lau, J. F., Ma, C. et al. (1999) A cytosolic catalaseis needed to extend adult lifespan inC. elegansdaf-C andclk-1 mutants. Nature (London)399, 162166

    109 Taub, J., Lau, J. F., Ma, C. et al. (2003) A cytosolic catalaseis needed to extend adult lifespan inC. elegansdaf-C andclk-1 mutants. Nature (London)421, 764

    110 Rasmussen, U. F., Krustrup, P., Kjaer, M. and Rasmussen,H. N. (2003) Human skeletal muscle mitochondrialmetabolism in youth and senescence: no signs offunctional changes in ATP formation and mitochondrialoxidative capacity. Pflugers Arch.446, 270278

    111 Rasmussen, U. F., Krustrup, P., Kjaer, M. and Rasmussen,H. N. (2003) Experimental evidence against themitochondrial theory of aging. A study of isolated human

    skeletal muscle mitochondria. Exp. Gerontol.38, 877886112 Maklashina, E. and Ackrell, B. A. (2004) Is defectiveelectron transport at the hub of aging? Aging Cell 3, 2127

    113 Jacobs, H. T. (2003) The mitochondrial theory of aging:dead or alive? Aging Cell2, 1117

    114 Spelbrink, J. N., Toivonen, J. M., Hakkaart, G. A. et al.(2000)In vivofunctional analysis of the humanmitochondrial DNA polymerase POLG expressed incultured human cells. J. Biol. Chem.275, 2481824828

    115 Trifunovic, A., Wredenberg, A., Falkenberg, M. et al.(2004) Premature ageing in mice expressing defectivemitochondrial DNA polymerase. Nature (London)429,417423

    116 Cooke, M. S., Evans, M. D., Dizdaroglu, M. and Lunec, J.(2003) Oxidative DNA damage: mechanisms, mutation,and disease. FASEB J.17, 11951214

    Received 26 May 2004/22 July 2004; accepted 28 July 2004

    Published as Immediate Publication 28 July 2004, DOI 10.1042/CS20040148

    C 2004 The Biochemical Society