Abstract
There is substantial evidence that mitochondria are involved in the aging process. Mitochondrial function requires the coordinated expression of hundreds of nuclear genes and a few dozen mitochondrial genes, many of which have been associated with either extended or shortened life span. Impaired mitochondrial function resulting from mtDNA and nuclear DNA variation is likely to contribute to an imbalance in cellular energy homeostasis, increased vulnerability to oxidative stress, and an increased rate of cellular senescence and aging. The complex genetic architecture of mitochondria suggests that there may be an equally complex set of gene interactions (epistases) involving genetic variation in the nuclear and mitochondrial genomes. Results from Drosophila suggest that the effects of mtDNA haplotypes on longevity vary among different nuclear allelic backgrounds, which could account for the inconsistent associations that have been observed between mitochondrial DNA (mtDNA) haplogroups and survival in humans. A diversity of pathways may influence the way mitochondria and nuclear – mitochondrial interactions modulate longevity, including: oxidative phosphorylation; mitochondrial uncoupling; antioxidant defenses; mitochondrial fission and fusion; and sirtuin regulation of mitochondrial genes. We hypothesize that aging and longevity, as complex traits having a significant genetic component, are likely to be controlled by nuclear gene variants interacting with both inherited and somatic mtDNA variability.
Keywords: mitochondria, epistasis, genetics, polymorphism, longevity, aging
Mitochondria and aging
The vast majority (90%) of the energy needs of the human body are met by mitochondrial oxidative phosphorylation (OXPHOS). OXPHOS takes place entirely in mitochondria and is a highly efficient system for producing the energy required to maintain the structure and function of the body. OXPHOS enzyme activities decline with age in human and primate muscle (Boffoli et al., 1994; Cooper et al., 1992; Trounce et al., 1989), liver (Yen et al., 1989), and brain (Bowling et al., 1993; Jazin et al., 1996) and correlate with the accumulation of somatic mtDNA deletions (Arnheim and Cortopassi, 1992; Bender et al., 2006; Bua et al., 2006; Chang et al., 2005; Corral-Debrinski et al., 1992a; Corral-Debrinski et al., 1992b; Cortopassi et al., 1992; Hattori et al., 1991; Hayakawa et al., 1993; Herbst et al., 2007; Kraytsberg et al., 2006; Linnane et al., 1990; Liu et al., 1998a; Mann et al., 1992; Melov et al., 1995; Nagley et al., 1992; Piko et al., 1988; Reeve et al., 2008; Sciacco et al., 1994; Simonetti et al., 1992; Soong et al., 1992; Sugiyama et al., 1991; Wallace et al., 1995; Wei, 1992; Yang et al., 1994; Yen et al., 1994; Yen et al., 1992; Yen et al., 1991; Zhang et al., 1992; Zhang et al., 1999; Zhang et al., 1998; Zhang et al., 2002) and base substitutions (Chinnery et al., 2001; Greaves et al., 2006; Kadenbach et al., 1995; Liu et al., 1998b; Michikawa et al., 1999; Munscher et al., 1993a; Munscher et al., 1993b; Murdock et al., 2000; Nekhaeva et al., 2002; Soong et al., 1992; Taylor et al., 2003; Wang et al., 2001; Zhang et al., 1993). For example, skeletal muscle mtDNA deletions localize to fibers that are also deficient in electron transport activity, and these defective fibers increase with age in humans and rodents (Bua et al., 2006; Herbst et al., 2007). High levels of somatic mtDNA deletions have been described in substantia nigra neurons from both elderly control subjects and patients with Parkinson disease (Bender et al., 2006; Kraytsberg et al., 2006; Reeve et al., 2008). High levels of mtDNA deletions were associated with respiratory chain deficiency (Bender et al., 2006) with these mutations being significantly higher in cytochrome c oxidase (COX)-deficient neurons than in COX-positive neurons, suggesting that mtDNA deletions may be directly responsible for impaired cellular respiration (Kraytsberg et al., 2006). High levels of somatic mtDNA mutations may also result in low COX activity observed in substantia nigra and muscle fibers from elderly humans (Itoh et al., 1996; Sciacco et al., 1994). An age-related decline in mtDNA content in skeletal muscle from humans (Short et al., 2005) and mice (Li et al.) has been related to decreases in both mitochondrial ATP production rate (Short et al., 2005) and oxidative phosphorylation coupling (Li et al.).
At present, it is unknown what mechanism is generating mtDNA deletions and mutations and the ideas concerning the role of mitochondria and mtDNA in aging continue to be in flux. While the mitochondrial theory of aging‘ hypothesis is attractive, in which somatic mutation of mtDNA leads respiratory chain dysfunction, enhancing the production of DNA-damaging oxygen radicals that in turn result in the accumulation of further mtDNA mutations and bioenergetic crisis, there is no evidence to support this process. Indeed, a rigorous test of the vicious cycle‘ remains to be undertaken.
The link between mtDNA mutations and reactive oxygen species (ROS) production is in question and remains to be tested. Recent results suggest that respiratory chain dysfunction is the primary inducer of premature aging in mtDNA mutator mice, independent of ROS (Trifunovic et al., 2005). MtDNA mutator mice accumulate mtDNA mutations in an approximately linear manner over their lifetime (Trifunovic et al., 2005). Despite the profound respiratory chain deficiency and premature aging phenotypes observed in mtDNA mutator mice, the amount of ROS produced is normal (Trifunovic et al., 2005). While ROS are toxic and may damage a variety of cellular components, there are also data to suggest that the organism may cope with increased ROS damage without developing premature aging (Trifunovic et al., 2005). Transgenic mice with reduced mitochondrially generated oxidative damage had a modest but significant increase in life span (Lapointe and Hekimi, 2008; Schriner et al., 2005).
The majority of genes encoded by the mtDNA are crucial for the machinery that converts metabolic energy into ATP. Human mtDNA is a 16,569 base pair loop that contains 37 genes coding for two rRNAs, 22 tRNAs and 13 polypeptides. These genes include NADH dehydrogenase, cytochrome c oxidase, ubiquinol/cytochrome c oxidoreductase, and ATP synthase, as well as the genes for unique ribosomal RNA and transfer RNA particles that are required for translating these genes into proteins. A major, unresolved issue remains that of the relationship between overall mutation load and its physiological effects at the tissue and organism level. Somatic (acquired) MtDNA mutations occur in postmitotic tissues (i.e., heart, brain, nerve). Cells in these high-energy tissues have a higher degree of heteroplasmy than cells in rapidly dividing, low-energy requiring tissues. However, the assertion that mtDNA is uniquely vulnerable to attack by ROS is not completely supported by current evidence (Jacobs, 2003). To date, no general consensus has been reached on the age related changes of mtDNA content (Santoro et al., 2006).
The role of mitochondria and mtDNA in aging is still being elucidated. Energy metabolism is generally preserved in long-living subjects and centenarians, suggesting an important role in human longevity for mitochondria (Salvioli et al., 2008). Mutations in mitochondrial DNA cause profound changes in many systems due to mitochondrial malfunction and may be an indication of the fundamental role that these organelles play in everyday processes. Even though mitochondrial deletions and mutations accumulate with age, their effects on tissue function have not been clearly demonstrated (Krishnan et al., 2007). In addition, the role of mtDNA and nuclear-encoded mitochondrial gene variants has not been investigated in sufficiently large populations to yield information on how mitochondrial and nuclear genes interact in promoting longevity. While much is known about the role of mitochondrial dysfunction in many diseases, significant advances in our understanding of the role of mitochondria in aging will continue to be made for years to come.
Mitochondrial DNA
Inherited variants and human longevity
Several small and somewhat underpowered studies have associated variations in mtDNA with human longevity. These studies have described associations between specific inherited mitochondrial variants and extended lifespan in Japanese (Niemi et al., 2005; Tanaka et al., 2004), Chinese Uygur (Ren et al., 2008), Italian (De Benedictis et al., 1999), French (Ivanova et al., 1998), Irish (Ross et al., 2001) and Finnish (Niemi et al., 2003; Niemi et al., 2005) populations. On the whole, these results support the idea that the effect of mtDNA inherited variants on longevity is population- and possibly sex-dependent, perhaps due to differences in nuclear genetic background.
Associations between mtDNA and longevity differ from the —usual SNP-based associations seen with the nuclear genome. Unlike nuclear DNA, the mitochondrial genome does not recombine, so that the DNA sequence remains together as a 16,569-bp segment. Studies of human populations have revealed ancestral-associated polymorphisms whose combination defines groups of mtDNA types (called haplogroups) that can be used to reconstruct human evolution lineages. The European population is almost exclusively distributed among the nine haplogroups designated as H, I, J, K, T, U, V, W and X, whereas haplogroups A, B, C, D, F, G and certain subclusters of macrohaplogroups M and N are characteristic to Asian populations, haplogroups A, B, C and D to native Americans and haplogroups L0, L1, L2 and L3 to African populations (Niemi et al., 2005).
New sub-haplogroups are being identified (Achilli et al., 2004). For instance, haplogroup H, the most common in Europe, can be subdivided into at least 15 sub-haplogroups (Kivisild et al., 2006), and formerly haplogroup K has been lately recognized as a sub-haplogroup of haplogroup U. These variants are likely non-neutral. In particular, a series of experiments suggesting that some mtDNA haplogroups are associated with longevity (De Benedictis et al., 1999; Niemi et al., 2003; Ren et al., 2008; Ross et al., 2001; Tanaka et al., 1998), as well as with mitochondrial diseases (Brown et al., 2002; Howell et al., 2003; Sadun et al., 2004), and complex diseases (Wallace, 2005). For example, in Caucasians such as northern Italians, haplogroup J is over-represented in long-living people and centenarians (De Benedictis et al., 1999). However, this association was seen only in male centenarians (De Benedictis et al., 1999). An over-representation of haplogroup J in nonagenarians and centenarians has been replicated in Irish and Finnish long-living people (Niemi et al., 2003; Ross et al., 2001), but not in southern Italians (Dato et al., 2004).
By contrast, the J haplogroup was underrepresented in Chinese Uygur nonagenarians (Ren et al., 2008). SNPs in the D-loop region also occurred in lower frequencies in Chinese Uygur nonagenarians (Ren et al., 2008). The 150T polymorphism in the noncoding region was associated with longevity in Finnish and Japanese subjects (Niemi et al., 2005). Interestingly, a stratified analysis revealed that mtDNA mutations characteristic of the J2 subhaplogroup (489C and 10398G) modified the association between the 150T mutation and longevity (Niemi and Majamaa, 2005). These findings suggest that longevity is partly determined by epistatic interactions among these mtDNA loci.
Oxidative phosphorylation
A key feature of the aging process is that the mitochondrial respiratory capacity declines and the production of reactive oxygen species increases in the later part of life span. The most commonly observed age-related changes in mitochondrial activity include an elevation in the rates of generation of superoxide anion radical and hydrogen peroxide, both progenitors for other intracellular ROS (Marnett et al., 2003; Sohal and Weindruch, 1996), and a decline in the rate of maximal respiration (Chiu and Richardson, 1980; Ferguson et al., 2005; Trounce et al., 1989). Drosophila aging is also associated with changes in mitochondrial structure and a decline in mitochondrial function (Ferguson et al., 2005; Walker and Benzer, 2004). Cytochrome c oxidase (COX; complex IV) is the only mitochondrial respiratory complex which shows an age- related decline in activity in Drosophila (Ferguson et al., 2005; Sohal et al., 1995). The decline in COX activity is accompanied by a decrease in ADP-stimulated respiration, and elevation of mitochondrial superoxide and hydrogen peroxide production (Ferguson et al., 2005; Sohal et al., 1995). Decreased COX activity (~30–50%) and increased superoxide generation are among the most consistent age-related alterations in mammalian tissues (Benzi et al., 1992; Cooper et al., 1992; Desai et al., 1996; Kwong and Sohal, 2000; Martinez et al., 1996). As in mammals, complex IV activity appears to be particularly vulnerable to both aging (Ferguson et al., 2005) and oxidative stress (Walker and Benzer, 2004) in flies. In Drosophila, two of the three COX subunits encoded in mitochondrial DNA show age-related decreases in protein abundance (43% and 75%, respectively) which could explain the age-related decrease in mitochondrial respiratory activity and an increase in ROS production (Sohal et al., 2008). Another likely explanation behind the age-related decline in OXPHOS function is the decline in expression of nuclear-encoded genes. For example, age-related changes in a large set of nuclear-encoded genes involved in ATP synthesis and mitochondrial respiration have been observed for both Caenorhabditis elegans and Drosophila (McCarroll et al., 2004). RNA interference of five genes encoding components of OXPHOS complexes I, III, IV, and V leads to increased life span in Drosophila (Copeland et al., 2009). However, reduced expression of OXPHOS genes was not consistently associated with reduced assembly of the complexes or reduced ATP levels. In addition, extended longevity was not correlated with energy consumption and accumulation of damage. Targeted RNAi of two complex I genes in adult tissues or in neurons alone was sufficient to extend life span (Copeland et al., 2009). Further support for the key role of specific OXPHOS-related genes in lifespan comes from mouse models where a knockout of SURF1 (Dell'agnello et al., 2007), a gene encoding a putative complex IV assembly factor, or reduced activity of murine CLK1 (Lapointe and Hekimi, 2008; Liu et al., 2005), a mitochondrial enzyme necessary for ubiquinone biosynthesis, lead to substantial increases in life span.
It has been proposed that the geographic distribution of human mtDNA lineages resulted from selection mainly driven by adaptation to climate and nutrition (Mishmar et al., 2003; Ruiz-Pesini et al., 2004; Ruiz-Pesini and Wallace, 2006; Wallace et al., 2003). According to this hypothesis, certain ancient mtDNA variants permitted humans to adapt to colder climates resulting in the regional enrichment of specific lineages. Underlying this selection were functional mtDNA variants that altered OXPHOS coupling efficiency, shifting the energetic balance from ATP generation to heat production consequently allowing Homo sapiens to adapt to colder environments after leaving Africa (Mishmar et al., 2003; Ruiz-Pesini et al., 2004).
While there is strong evidence supporting selection as an important factor in the evolution of human mtDNA (Balloux et al., 2009; Elson et al., 2004; Kivisild et al., 2006; Marcuello et al., 2009; Martinez-Redondo et al., ; Mishmar et al., 2003; Moilanen et al., 2003; Moilanen and Majamaa, 2003; Montiel-Sosa et al., 2006; Ruiz-Pesini et al., 1998; Ruiz-Pesini et al., 2000; Ruiz-Pesini et al., 2004; Ruiz-Pesini and Wallace, 2006), not all studies support climate as the driving force for human mtDNA evolution (Amo and Brand, 2007; Amo et al., 2008; Elson et al., 2004; Kivisild et al., 2006; Moilanen et al., 2003). Evidence that climatic adaptation has influenced the geographic distribution of mtDNA diversity was obtained by examining patterns of genetic variation across the mtDNA coding region, including the 13 mtDNA OXPHOS genes (Balloux et al., 2009; Mishmar et al., 2003; Ruiz-Pesini et al., 2004). An examination of regional (tropical, temperate and arctic) gene-specific variation in mitochondrial OXPHOS genes provided support for adaptive selection influencing mtDNA diversity (Mishmar et al., 2003). ATP6 was highly variable in the mtDNAs from the arctic, cytb was more variable in temperate Europe, and COI was highly variable in tropical Africa (Mishmar et al., 2003). These genes were largely invariant in the regions outside of their high adaptation zones (e.g. ATP6 was strongly conserved in the temperate and tropical zones). These results were interpreted as evidence for regional gene-specific selection since this pattern of variation would not be expected if all mtDNA mutations were random and neutral. The frequency of conserved, non-synonymous (missense) mutations across the mtDNA coding region was also found to increase from tropical Africa to temperate Europe and arctic northeastern Siberia (Ruiz-Pesini et al., 2004). This excess of non-synonymous mutations in the colder latitudes was interpreted as evidence for adaptive selection playing an important role as people migrated out of Africa into temperate and arctic Eurasia. However, other analyses do not support a simple model in which climatic adaptation has been a major force during human mtDNA evolution (Elson et al., 2004; Kivisild et al., 2006; Moilanen et al., 2003). For example, the excess non-synonymous substitutions observed in some OXPHOS genes may not reflect positive selection but the relaxation of negative selection in specific populations (Elson et al., 2004) or may be a feature of the terminal branches of the phylogenetic tree, independent of geographical region (Kivisild et al., 2006). Others have observed significant differences in the frequency of non-synonymous mutations among the European haplogroups (Moilanen et al., 2003), suggesting some mutations may be non-neutral within specific phylogenetic lineages but neutral within others.
Functional evidence supporting metabolic differences between haplogroups is equally inconsistent (Amo and Brand, 2007; Amo et al., 2008; Marcuello et al., 2009; Martinez-Redondo et al., ; Montiel-Sosa et al., 2006; Ruiz-Pesini et al., 1998; Ruiz-Pesini et al., 2000). Comparisons of spermatozoa motility among several European haplogroups revealed that sperm from haplogroups H subjects swam significantly faster than those from haplogroups T subjects (Ruiz-Pesini et al., 2000). Human spermatozoa motility is fully dependent on the functionality of the OXPHOS system and the haplogroup T samples showed 23% and 29% reductions, respectively, in complexes I and IV activity compared with haplogroup H samples (Ruiz-Pesini et al., 2000). Interestingly, no differences in complex II activity were observed between haplogroups H and T (complex II is exclusively encoded by the nuclear genome). Spermatozoa motility is directly correlated with activities of OXPHOS complexes I-IV (Ruiz-Pesini et al., 1998). Within the broadly distributed European haplogroup U, sublineages of the group exhibited differences in sperm motility (Montiel-Sosa et al., 2006). Conserved cytb mutations were enriched in northern Europe and less prevalent in southern Europe, which is suggestive of selection allowing adaptation to a colder northern climate (Montiel-Sosa et al., 2006). Maximal oxygen uptake (VO2 max) and mitochondrial oxidative damage have been shown to be higher in human subjects from European haplogroup H compared with haplogroup J (Marcuello et al., 2009; Martinez-Redondo et al.). By contrast, studies of cybrids harboring mitochondria with either haplogroup H or haplogroup T in cultured cells with identical nuclear backgrounds show no functionally important differences in bioenergetic capacities and coupling efficiencies (Amo et al., 2008) and results were similar for both isolated mitochondria and mitochondria within cells. Furthermore, cybrid studies comparing arctic (A, C and D) or tropical (L1, L2 and L3) haplogroups yielded no overall differences between arctic and tropical mtDNA haplogroups with regard to the overall kinetics of substrate oxidation (Amo and Brand, 2007). Intriguingly, mitochondria from Arctic haplogroups had similar or greater coupling efficiency than mitochondria from tropical haplogroups, which is contrary to the hypothesis that mitochondrial haplogroups with lower coupling efficiency were positively selected during radiations of modern humans (Amo and Brand, 2007).
There is likely a larger role for nuclear and mitochondrial interactions in determining the effect of mtDNA variation on OXPHOS function. In particular, nuclear-encoded ROS-scavenging mechanisms may interact with mtDNA haplotype to influence ROS homeostasis and affect OXPHOS capacity. Early studies in cultured primary cells with different human or mouse mtDNA haplotypes concluded that the respiratory capacity is not substantially influenced by any of the mtDNA variants tested (Battersby and Shoubridge, 2001; Carelli et al., 2002). More recently, a study examining OXPHOS capacity in mouse cell lines carrying a homogeneous nucleus but mtDNA derived from four crosses and NIH3T3 mouse cells also showed little variation in respiration (Moreno-Loshuertos et al., 2006), a coarse indicator of OXPHOS function. However, further examination revealed that differences in OXPHOS performance were detectable but masked by a specific upregulation in mitochondrial biogenesis, triggered by an increase in generation of ROS. The genetic element underlying the functional differences was an 'A' track polymorphism in tRNAArg. Cell lines with ten adenines had higher ROS production and increased amounts of mtDNA (Moreno-Loshuertos et al., 2006). This study demonstrated that some common and 'non-pathological' mtDNA variants in mice can reduce OXPHOS function, but this reduction can be compensated by enhanced ROS production.
Mitochondrial-nuclear epistasis
More than 90% of the functional mitochondrial genome is encoded in the nucleus. These nuclear-encoded mitochondrial genes (mitonuclear genes) arose either by transfer events from the mitochondrial to the nuclear genome or by recruitment of nuclear genes to a novel mitochondrial function through the acquisition of mitochondrial targeting sequences (Rand et al., 2004). Mildly deleterious mtDNA polymorphisms are a general property of animal populations (Nachman, 1998; Rand and Kann, 1998; Weinreich and Rand, 2000); as a result, the mitochondrial genome, which has high levels of genetic variation (Parsons et al., 1997), presents a large pool of potential variants that could affect aging and provide a broad opportunity for mitochondrial-nuclear interactions. As previously described, in humans there is statistical evidence that mtDNA haplotypes are associated with variation in longevity. However, some long-lived haplotypes also carry mutations associated with disease, suggesting that genetic background is an important modulator of mtDNA effects on aging (Rose et al., 2001).
While there is a wealth of research on mitochondrial aspects of aging, studies that manipulate nuclear and mitochondrial genetic variation to explore nuclear – mitochondrial epistatic effects on longevity are generally lacking (Blier et al., 2001; Dowling et al., 2007; Etterson et al., 2007; Rand et al., 2001; Rhode and Cruzan, 2005; Tang et al., 2007; Wade and Goodnight, 2006). Because mitochondria play a central role in energy metabolism it is likely that mitochondrial-nuclear epistasis has important fitness effects (Dowling et al., 2007; Hutter and Rand, 1995; Rand et al., 2001; Rawson and Burton, 2002; Rhode and Cruzan, 2005; Schmidt et al., 2001; Willett and Burton, 2003). and is evolutionarily important (Rand et al., 2004). Coadaptation among mitochondrial and mitonuclear genes predicts that the phenotypic effects of alternative mtDNAs should increase with increasing levels of DNA sequence divergence between native and foreign mtDNAs (from different populations or species, for example) (Rand et al., 2004). Studies in Drosophila have established that nuclear – mitochondrial interactions (epistases) are important in the fitness effects of mtDNA variation (Clark and Lyckegaard, 1988; James and Ballard, 2003; Kilpatrick and Rand, 1995; Rand et al., 2001). In particular, effects of the foreign Drosophila mtDNA on longevity depend strongly on the nuclear genetic background demonstrating that there is a nuclear – mtDNA epistatic effect for longevity (Rand et al., 2006). These previous studies have shown that the nuclear – mitochondrial epistatic effects are often stronger than mtDNA main effects on Drosophila longevity. One explanation for this observation is that the disruption of coadapted mitonuclear genotypes (e.g. coordination of protein synthesis in the nucleus and meitochondria) reduces mitochondrial performance (Blier et al., 2001). Evidence from a variety of systems including Drosophila, marine copepods and mouse cells (McKenzie et al., 2003; Rawson and Burton, 2002; Sackton et al., 2003) has supported this explanation. It might follow that such genotypes would have reduced longevity due to disrupted OXPHOS functions (detailed in the next section).
Results from several animal systems demonstrate that epistatic interactions with nuclear genetic background are a significant component of the mitochondrial genetics of aging. This epistasis may explain why some mtDNA mutations have very different phenotypic effects in different individuals, possibly obscuring the mtDNA effects in human aging and disease. Examples of nuclear – mitochondrial coadaptation in humans are limited (Bykhovskaya et al., 2000; De Benedictis et al., 2000; Shankar et al., 2008). De Benedictis et al. (De Benedictis et al., 2000) analyzed the distribution of the mtDNA inherited variants by tyrosine hydroxylase (THO) genotypes in three sample groups of increasing ages (20–49 years; 50–80 years; centenarians). The mtDNA haplogroups and THO genotypes were associated randomly in the first group, while in the second group, and particularly in the centenarians, a non-random association was observed between the mtDNA and nuclear DNA variability. Moreover, the U haplogroup was over-represented in centenarians carrying a THO genotype unfavorable to longevity. Maternally inherited deafness associated with the A1555G mutation in the mitochondrial 12S ribosomal RNA (rRNA) gene appears to require additional environmental or genetic changes for phenotypic expression (Bykhovskaya et al., 2000). Linkage results in several families with the A1555G mutation suggest that the chromosomal region around marker D8S277 harbors a nuclear modifier gene for this mitochondrial DNA disease mutation (Bykhovskaya et al., 2000). Leber Hereditary Optic Neuropathy (LHON) is a maternally inherited blinding disease caused by missense mutations in the mitochondrial DNA (mtDNA). Incomplete penetrance and a predominance of male patients presenting with vision loss suggest that both nuclear modifier genes and environmental factors play an important role in the development of the disease (Shankar et al., 2008). Linkage analysis in a large family harboring a homoplasmic G11778A mtDNA mutation on a haplogroup J background identified a novel LHON susceptibility locus on chromosome Xq25-27.2 (Shankar et al., 2008). These findings support the hypothesis that some human aging traits and diseases require particular interactions between mtDNA and nuclear DNA.
Consequences for oxidative phosphorylation
OXPHOS proteins are jointly encoded by nuclear and mitochondrial genes (Table 1). The enzyme complexes of the electron transport chain and OXPHOS are particularly attractive models for the analysis of cytonuclear co-adaptation (Rand et al., 2004). Several lines of evidence have shown that interactions between nDNA- and mtDNA-encoded OXPHOS proteins are functionally important, including: backcross analyses in whole organisms; cell cultures with mixed nuclei and mitochondria from evolutionarily diverged organisms; and comparisons of nuclear DNA- and mtDNA- substitution rates across multiple taxa. Structural models for complexes II – V are available that increase the power of molecular evolutionary analyses, and phenotypes can be studied with the use of enzyme assays. At a molecular level, co-adaptation should be evident from coordinated amino acid changes on gene trees of interacting OXPHOS proteins. A simple prediction of co-adaptation is that an experimental transplant‘ of interacting partners results in diminished OXPHOS performance, and this disruption should increase as the level of evolutionary divergence increases. We have additionally compiled a list of currently known interactions among nDNA- and mtDNA-encoded OXPHOS proteins from the Human Protein Reference Database (Mishra et al., 2006) and the IntAct database (Hermjakob et al., 2004) (Table 2).
Table 1.
Complex I | Complex II | Complex III | Complex IV | Complex V | |
---|---|---|---|---|---|
Nuclear genes | 39 | 4 | 10 | 10 | 12 |
Mitochondrial genes | 7 | 0 | 1 | 3 | 2 |
Table 2.
Complex I |
Complex III |
Complex IV |
|||
---|---|---|---|---|---|
Nuclear | Mitochondrial | Nuclear | Mitochondrial | Nuclear | Mitochondrial |
NDUFAB1 | ND1 | CYC1 | CYTB | COX4I1 | COX1 |
NDUFAB1 | ND2 | COX5A | COX1 | ||
NDUFAB1 | ND3 | ||||
NDUFAB1 | ND4 | ||||
NDUFAB1 | ND5 | ||||
NDUFS3 | ND6 | ||||
NDUFS7 | ND1 |
Backcross analyses in whole organisms
Maternal inheritance of organelle DNA enables one to transplant‘ the cytoplasmic genome from one strain or species onto the nuclear background of the paternal line. With control backcrosses to the maternal line, one can compare phenotypes of disrupted‘ (mtDNA on foreign nuclear background) and reconstituted‘ (mtDNA on original nuclear background) genotypes. Studies of the intertidal copepod Tigriopus californicus provide evidence for cytonuclear co-adaptation using enzyme assays of COX activity (Burton et al., 1999; Willett and Burton, 2003). In backcross genotypes between different geographical populations, COX activity is significantly reduced relative to control backcrosses. Sequence polymorphism surveys among Tigriopus populations show evidence for positive selection at COX but negative selection at mitochondrial and nuclear genes of complex III (plus other enzymes) (Willett and Burton, 2003).
Similar analyses among strains and species of Drosophila (James and Ballard, 2003; Sackton et al., 2003) provide important contrasts for the outcome of co-adaptation. Deleterious mutations should accumulate more rapidly in small (Tigriopus) than in large (Drosophila) populations; hence, disruption of co-adapted gene complexes might be detected among geographical populations in Tigriopus, but not Drosophila. More studies are needed to establish the generality of this population size effect on cytonuclear co-adaptation.
Cell cultures with mixed nuclei and mitochondria
In vitro models have been developed to demonstrate that mtDNA inherited variants modulate biological functions. The best known in vitro model is represented by cytoplasmic hybrids also known as cybrids. This technique allows the analysis of mtDNA mutations or inherited variants by minimizing the effect of the nuclear genome that is kept constant. Cybrid cell lines are relatively easy to obtain and easy to maintain (cells can be frozen and stored for years). This model can be used to test the effect of specific mtDNA mutations. In brief, cybrids are constructed by preparing mitochondria from human platelets and fusing them with mtDNA free cells. Several metabolic and other tests can be performed on the cybrid cells including: respiratory flux and membrane potential, modular kinetic analysis of OXPHOS, and catalase activity. Amo and Brand (Amo and Brand, 2007) examined the bioenergetic importance of mtDNA variants using modular kinetic analysis of oxidative phosphorylation in mitochondria from cybrid cells with constant nuclear DNA but different mtDNA. They found that there were no functionally-important bioenergetic differences between mitochondria bearing different mtDNA haplogroups using either isolated mitochondria or mitochondria within cells (Amo and Brand, 2007).
Mitochondrial-nuclear epistasis was examined in cardiomyocytes from unrelated cardiomyopathy patients (Davidson et al., 2009). The A4300G mutation in the mitochondrial tRNA gene is a known hotspot for mutations associated with cardiomyopathy. Biochemical analyses have shown decreased Complex I and IV activity (encoded by both nuclear and mitochondrial subunits) with normal Complex II activity (exclusively nuclear encoded) among A4300G mutation carriers (Davidson et al., 2009). Cybrids were constructed from cardiomyocytes to determine pathogenicity of the A4300G mtDNA mutation in different nuclear backgrounds. Four transnuclear cardiomyocyte cell lines were created with normal or patient nuclei and containing wild type or mutant A4300G mtDNA. Of the four cell lines analyzed, COX activity was low only in patient cardiomyocytes containing both the patient's nucleus and mitochondria. COX activity was normal in cells with either wild type nucleus or wild type mtDNA. These results strongly suggest that a (tissue specific) nuclear modifier gene may interact synergistically with the A4300G mtDNA mutation to cause COX deficiency (Davidson et al., 2009).
Mixing nuclei and mitochondria from organisms that have evolutionarily diverged over time results in reduced mitochondrial OXPHOS function (Kenyon and Moraes, 1997; McKenzie et al., 2003). Cell cultures have been established in which mitochondria from one species are placed in a cell with a foreign‘ nucleus. In primate models, cells carrying a human nucleus with mitochondria from chimpanzee or gorilla showed normal cellular respiration, but mitochondria from the orangutan or more distant primate species did not restore respiration (Kenyon and Moraes, 1997). In a mouse model, cell lines carrying mitochondria from six different species spanning 2–12 million years of divergence revealed a nearly linear disruption of respiratory chain function with evolutionary distance (McKenzie et al., 2003); complex II, which lacks mtDNA subunits, failed to show disruption (McKenzie et al., 2003). These results are consistent with the transplant‘ prediction of cytonuclear co-adaptation, and the complex II result provides an internal control: the disruption is only seen in those complexes involving both nuclear and mitochondrial subunits (McKenzie et al., 2003).
Comparisons of nuclear DNA and mtDNA-substitution rates
The evolution of nDNA-mtDNA-encoded protein interactions has been explored to determine whether rates of nonsynonymous substitutions have been higher, the same, or lower for nuclear- and mitochondrial-encoded residues in close proximity (Schmidt et al., 2001). Using evolutionary and crystallographic data for COX, Schmidt et al. (Schmidt et al., 2001) demonstrated that: (a) mtDNA-encoded residues in close physical proximity to nuclear DNA-encoded residues mutated at a faster (optimizing) rate than the other mitochondrial-encoded residues, and (b) nuclear DNA-encoded residues in close physical proximity to the mtDNA-encoded residues evolve more slowly (constraining) than the other nuclear-encoded residues in the complex.
These results suggest that the faster mtDNA mutation rate, which allows sampling of more residues in the interacting region, makes mtDNA the predominant partner in accommodating mutations important for subunit interaction. The Schmidt et al. (Schmidt et al., 2001) data suggest that polymorphisms affecting interactions would be biased towards mitochondrial subunits. Since there are >70 nuclear genes encoding four of the OXPHOS complexes we believe that genetic approaches that focus on common variation in the nDNA-encoded residues for interaction analysis will improve power and minimize false positives when we examine mitochondrial-nuclear epistasis.
Nuclear mitochondrial genes
A diversity of pathways may influence the way mitochondria and nuclear – mitochondrial interactions modulate longevity. Likely candidate pathways include OXPHOS, mitochondrial uncoupling, antioxidant defenses, mitochondrial fission and fusion, transport and degradation, and sirtuin regulation of mitochondrial genes.
Uncoupling protein gene variants
Human genetic variation in uncoupling proteins, UCP1-3, has been associated with several metabolic phenotypes and specific diseases but has not been examined with regard to lifespan or mortality. Variation in UCP1-3 has been associated with obesity (Alonso et al., 2005; Esterbauer et al., 1998; Evans et al., 2000; Lentes et al., 1999; Oh et al., 2004; Shin et al., 2005; Urhammer et al., 1998; van Abeelen et al., 2008), BMI (Heilbronn et al., 2000; Nakano et al., 2006), weight change (Berentzen et al., 2005), waist-to-hip ratio (Herrmann et al., 2003), body fat (Oppert et al., 1994) and body fat accumulation (Kim et al., 2005). UCP2-3 genetic polymorphisms have also been shown to modify the effect of a low calorie diet on body fat reduction (Cha et al., 2006; Yoon et al., 2007). Although the mechanism behind these associations is not clear, a UCP1 promoter variant has been shown to affect adipocyte transcription factor binding (Schaffler et al., 1999). Polymorphisms in UCP1-3 have also been associated with high-density lipoprotein cholesterol (Cha et al., 2007; Kotani et al., 2008) and UCP2 variants with increased carbohydrate and decreased lipid oxidation in juvenile obesity (Le Fur et al., 2004). UCP2 polymorphisms have been associated with insulin resistance (D'Adamo et al., 2004), type 2 diabetes (Bulotta et al., 2005; D'Adamo et al., 2004), and UCP2 genetic variation affects peripheral nerve dysfunction (Yamasaki et al., 2006) and dehydroepiandrosterone levels (Zietz et al., 2001) in type 2 diabetics. Other UCP genetic associations include: UCP1 and hypertension (Kotani et al., 2007); UCP2 and schizophrenia (Yasuno et al., 2007); UCP2-3 and anorexia nervosa(Campbell et al., 1999); UCP2 and exercise efficiency (Buemann et al., 2001); and UCP2-3 with diabetic neuropathy in patients with type 1 diabetes (Rudofsky et al., 2006)
Sirtuin decatylases
Sirtuins are —class 3 histone deacetylases (HDAC) (Gregoretti et al., 2004) that regulate the acetylation of histones and ultimately alter gene transcription. In general, histone acetylation is associated with gene transcription, while histone deacetylation silences genes; however, deacetylase activity may sometimes activate genes (Grozinger and Schreiber, 2002; Johnstone, 2002; Kurdistani and Grunstein, 2003). The evolutionary conservation of an NAD+ dependent deacetylase mechanism extends from yeast to mammals, and the SIR2 gene and its orthologs extend lifespan in yeast (Kaeberlein et al., 1999; Tissenbaum and Guarente, 2001), nematodes (Kaeberlein et al., 1999; Tissenbaum and Guarente, 2001), and flies (Rogina and Helfand, 2004). Lifespan in S. cerevisiae can be extended by overexpression of SIR2 (Kaeberlein et al., 1999) or through the addition of an extra copy of the SIR2, which extends lifespan up to 30% (Tissenbaum and Guarente, 2001). In C. elegans, gene duplication of sir-2.1 extends lifespan by up to 50% (Tissenbaum and Guarente, 2001) while dSir2 overexpression in D. melanogaster increases lifespan by 57% (Rogina and Helfand, 2004).
Studies involving several model organisms suggest that SIR2 deacetylase enzymes (sirtuins) may mediate the connection between energetics and lifespan (Guarente and Kenyon, 2000; Kaeberlein et al., 2004; Kaeberlein et al., 1999; Lamming et al., 2004; Rogina and Helfand, 2004; Tissenbaum and Guarente, 2001; Wood et al., 2004). While sirtuins suppress the transcription of a wide range of genes, their nicotinamide adenine dinucleotide (NAD+) dependent deacetylase activity (Imai et al., 2000; Landry et al., 2000) may allow them to act as sensors of nutrient fluctuation (Revollo et al., 2004). Moreover, the SIR2 gene and its orthologs mediate the life-extending effects of caloric restriction in yeast (Blander and Guarente, 2004; Kaeberlein et al., 2004; Lamming et al., 2004; Lin et al., 2000) and flies (Rogina and Helfand, 2004; Wood et al., 2004). In S. cerevisiae, SIR2 is upregulated by changes in metabolic activity due to calorie restriction, which leads to extended lifespan (Kaeberlein et al., 2004; Lin et al., 2000). Similarly, CR extends lifespan in D. melanogaster by increasing dSir2 expression (Rogina and Helfand, 2004). Both S. cerevisiae SIR2 and D. melanogaster dSir2 mutants do not live longer with caloric restriction (Kaeberlein et al., 2004; Lin et al., 2000), implying that caloric restriction-mediated lifespan extension involves SIR2.
The seven mammalian sirtuins, including human sirtuins SIRT 1-7 (Frye, 2000), are currently undergoing functional characterization. Sirtuins are expressed in a wide variety of tissues and each sirtuin has a unique expression profile (McBurney et al., 2003; Michishita et al., 2005). In general, SIRT1-7 exhibit abundant expression in brain and testes, and most sirtuins show higher expression in fetal brain than adult brain, suggesting a role in development (Michishita et al., 2005). Human sirtuins have adapted specialized cellular roles involving minimal functional redundancy (Michishita et al., 2005).
Sirtuin decatylases and mitochondrial function
SIRT3 (Onyango et al., 2002; Schwer et al., 2002), SIRT4, and SIRT5 (Michishita et al., 2005) localize to the mitochondria. SIRT3 activates several mitochondrial functions including mitochondrial uncoupling and respiration (Shi et al., 2005). Although initially described as a mitochondrial protein, recent studies suggest that SIRT3 can also be a nuclear protein that transfers to the mitochondria during cellular stress (Onyango et al., 2002; Scher et al., 2007; Schwer et al., 2002). Caloric restriction activates SIRT3 and enhances the expression of mitochondrial genes ATP-synthetase and cytochrome-c oxidase (Shi et al., 2005). The absence of SIRT3 results in the increased acetylation of multiple components of Complex I of the electron transport chain. In particular, SIRT3 physically interacts with the NDUFA9 protein of Complex I which may provide a mechanism for how SIRT3 functions in vivo to regulate and maintain basal ATP levels (Ahn et al., 2008). SIRT4 interacts with glutamate dehydrogenase (GDH) to promote mitochondrial activation and increases the ATP/ADP ratio (Haigis et al., 2006; Kelly and Stanley, 2001). Human SIRT1 regulates genes that are critical to lipid mobilization and glucose homeostasis (Picard et al., 2004; Rodgers et al., 2005). SIRT1 also regulates insulin secretion by repressing UCP2 in pancreatic beta cells (Bordone et al., 2006) and in the liver (Rodgers et al., 2005).
Sirtuin decatylase gene variants
To date, studies of the effects of genetic variants in human sirtuins on lifespan are limited. Two studies in Calabria, Italy (Bellizzi et al., 2005; Rose et al., 2003) demonstrated an association between two linked SIRT3 polymorphisms and lifespan. In the first study, Rose et al. (Rose et al., 2003) reported the effects of a silent G477T polymorphism in exon 3 (Ser159Ser). In this study of 120 centenarians (36 men and 84 women), the TT genotype was associated with increased survival in men — but not women. Bellizzi et al. (Bellizzi et al., 2005) later reported that this polymorphism was in complete linkage with a specific allele in a variable number tandem repeat (VNTR) in intron 5 that exhibits enhanced enzyme activity (Bellizzi et al., 2005). In this study, the allele lacking enhanced activity was nearly absent in the 86 men more than 90 years old (3% allele prevalence), but not in the 156 women in this age group (10% allele prevalence) (Bellizzi et al., 2005). These studies suggest that underexpression of SIRT3 may be detrimental for longevity in men. However, the small sample size leaves open the possibility that these findings are due to chance since the risk‘ allele occurs in only 14% of men and 10% of women under age 80. The lack of concordant results between men and women is also unexplained. The authors also assessed linkage in the region of SIRT3, which is located near the telomeric terminal of chromosome 11p15. Four genes potentially associated with longevity are found in this region: tyrosine hydroxylase (TH), proinsulin (INS), IGF2, and HRAS1 (Rose et al., 2003). Rose et al. (Rose et al., 2003) however, found no evidence of linkage disequilibrium (LD) between the SIRT3 G477T polymorphism and SNPs in these four genes (Rose et al., 2003). A meta-analysis of SIRT3 SNPs was recently carried out in four European populations (Lescai et al., 2009). One SIRT3 SNP (rs939915) was associated with longevity among Italian, French and German centenarians. Additional studies of SIRT1 SNPs did not identify associations with longevity (Flachsbart et al., 2005; Kuningas et al., 2007).
Lagouge et al. (Lagouge et al., 2006) recently examined the effect of SIRT1 genetic variants on energy expenditure in Finnish subjects. Three common SIRT1 SNPs were significantly associated with whole body energy expenditure in a cohort of healthy, normal-weight, nondiabetic offspring of type 2 diabetic patients. Higher free-living activity energy expenditure has demonstrated a strong association with lower risk of mortality among older adults (Manini et al., 2006). Energy expenditure was evaluated either during fasting or during a hyperinsulinemic clamp. The three SNPs included a promoter A/G (rs3740051), an intron 3 A/G (rs2236319), and the synonymous L322L C/T polymorphism (rs 2273773); however, these three SNPs are in high LD in European populations. The variant alleles were associated with a 6% increase in energy expenditure. Two other SIRT1 SNPs were not associated with energy expenditure. While these data indicate that humans SIRT1 polymorphisms influence energy expenditure, a more thorough assessment of SIRT1 variation, including all haplotype tagging SNPs and the six remaining sirtuin genes would strongly support the direct involvement of sirtuins in modulating energy homeostasis in humans.
Antioxidant defenses
The primary antioxidant proteins that defend against ROS damage are the mitochondrial Mn-superoxide dismutase (SOD2), and the cytoplasmic CuZn-superoxide dismutase (SOD1), catalase (CAT), and glutathione peroxidase (GPX1). The SOD2 gene encodes an intramitochondrial free radical scavenging enzyme that is the first line of defense against superoxide produced as a byproduct of OXPHOS. Most SOD2-knock-out mice die soon after birth as a consequence of lung damage and those animals that do survive suffer severe neurodegeneration (Lebovitz et al., 1996). SOD2 mutant mice also exhibit a tissue-specific inhibition of the OXPHOS complexes I and II and accumulation of oxidative DNA damage (Melov et al., 1999a). SOD1 is a major cytoplasmic antioxidant enzyme that metabolizes superoxide radicals to molecular oxygen and hydrogen peroxide, thus providing a defense against oxygen toxicity. Overexpression of the human SOD1 gene in Drosophila motor neurons extended normal life span of the animals by up to 40% and rescued the life span of a short-lived Sod null mutant (Parkes et al., 1998). SOD1 knock-out mice appear phenotypically normal, although female homozygous mice exhibit markedly reduced fertility (Ho et al., 1998).
GPX1 functions in the detoxification of hydrogen peroxide, and is one of the most important antioxidant enzymes in humans. Paraquat has been shown to upregulate Gpx1 in normal cells and Gpx1 knockout mice are highly sensitive to this oxidant (de Haan et al., 1998). Cortical neurons from Gpx1 knockout mice are more susceptible to peroxide (de Haan et al., 1998).
CAT is a common enzyme found in nearly all living organisms, where it functions to catalyze the decomposition of hydrogen peroxide to water and oxygen. CAT has one of the highest turnover numbers of all enzymes; one molecule of catalase can convert millions of molecules of hydrogen peroxide to water and oxygen per second. CAT largely determines the functional antioxidant capacity of mitochondria and is the enzyme that is most affected in aging (Terlecky et al., 2006). Transgenic mice that overexpress mitochondrial CAT have increased median and maximum life spans (Schriner et al., 2005). In these animals, cardiac pathology and cataract development were delayed, oxidative damage was reduced, and the development of mitochondrial deletions was reduced (Schriner et al., 2005). In C. elegans, loss of CAT results in the organism manifesting a progeric phenotype (Petriv and Rachubinski, 2004). Processing of reactive oxygen species becomes altered, peroxisome morphology is changed, and the organism's lifespan is shortened. Similarly, lifespan of the yeast S. cerevisiae is significantly reduced when its CAT is knocked out (Petriv and Rachubinski, 2004). In rats and mice cellular CAT levels drop with age, which is accompanied by an increase in reactive oxygen species and resultant oxidative stress (Beier et al., 1993; Ishii et al., 1996; Ito et al., 1986). Calorically restricted animals reverse this trend – they express elevated levels of CAT and are more long-lived (Rao et al., 1990).
Chronically reducing catalase activity causes cells to display a cascade of accelerated aging reactions (Koepke et al., 2008). In particular, hydrogen peroxide and related reactive oxygen species are produced, protein and DNA are oxidatively damaged, and mitochondrial biogenesis is corrupted (Koepke et al., 2008). In addition, mitochondria are functionally impaired, losing their ability to maintain a membrane potential and synthesize reactive oxygen species (Koepke et al., 2008). Sirt1 has also been shown to affect CAT expression and be a determinant of cell apoptosis by regulating cellular ROS levels (Hasegawa et al., 2008). Sirt1 maintains cell survival by regulating CAT expression and by preventing the depletion of ROS required for cell survival (Hasegawa et al., 2008). In contrast, excess ROS upregulates Sirt1, which activates CAT leading to rescuing apoptosis (Hasegawa et al., 2008).
Antioxidant defense gene variants
Variants in SOD2 and CAT have been associated with aging and age-related outcomes. In the Framingham Study, polymorphisms in SOD2 were associated with age at death and biological age scored determined using the osseographic scoring system (Lunetta et al., 2007). The most extensively studied polymorphism in SOD2 is the Ala16Val substitution (Rosenblum et al., 1996). This polymorphism may alter the leader signal and affect the import of SOD2 into mitochondria (Shimoda-Matsubayashi et al., 1996). The alanine variant of SOD2 has been associated with an increased risk for breast cancer (Ambrosone et al., 1999) and Parkinson‘s disease (Shimoda-Matsubayashi et al., 1996).
A common functional -262C/T substitution polymorphism in the promoter region of the human CAT gene influences transcription factor binding, reporter gene transcription and is correlated to blood catalase levels (Forsberg et al., 2001). The -262T allele may protect against neurodegenerative and physical decline (Christiansen et al., 2004). In a study of 2223 Danish individuals, aged 45–93 years, the CAT -262 TT genotype was associated with improved physical function and the T allele with improved cognitive functioning (Christiansen et al., 2004).
Mitochondrial fission and fusion
A typical mammalian cell can have hundreds of mitochondria. However, each mitochondrion is not autonomous, because fusion and fission events mix mitochondrial membranes and contents (including mtDNA) (Chan, 2006a; Chen and Chan, 2005; Detmer and Chan, 2007). As a result, mitochondrial fusion, fission, and trafficking control mitochondrial shape, number, size, distribution, and physiology. The dynamic equilibrium between fusion and fission has major implications for mitochondrial morphology and function of the mitochondrial population. In normal cells, high rates of fusion and fission enable mitochondria to cooperate with each other through continual exchange of contents. Individual mitochondria can stochastically lose essential components, but such defects are short-lived, because mitochondrial fusion will restore the missing components from neighboring mitochondria. In cells lacking mitochondrial fusion, such restoration of activity cannot occur, and defective mitochondria accumulate. In the absence of fusion, a large population of mitochondria lack mtDNA. Therefore, mitochondrial fusion is essential to allow defective mitochondria a pathway to recover mtDNA. This defect has been shown to account for the respiratory and membrane potential aberrations found in fusion-deficient cells.
Mitochondrial fusion is a membrane-remodeling process that coordinately merges the outer and inner membranes between two mitochondria. Mitochondrial fusion is important not only for maintenance of mitochondrial morphology, but also for cell growth, mitochondrial membrane potential, and respiration. It requires three large GTPases: the mitofusins MFN1 and MFN2 (Chen et al., 2003a), and the dynamin-related protein OPA1 (Song et al., 2007). Mitochondrial fusion is likely to be more complicated than most other intracellular membrane fusion events, because four lipid bilayers must be coordinately fused. MFN1 and MFN2 are transmembrane GTPases embedded in the outer membrane of mitochondria and are essential for fusion of mitochondria (Chen et al., 2003a). MFN1 and MFN2 form homotypic and heterotypic complexes that are capable of tethering mitochondria and are the only conserved mitochondrial outer membrane proteins involved in fusion. Mouse embryonic fibroblasts lacking Mfn1 or Mfn2 display fragmented mitochondria, a phenotype due to a severe reduction in mitochondrial fusion (Chen et al., 2003a). Cells lacking both Mfn1 and Mfn2 have completely fragmented mitochondria and show no detectable mitochondrial fusion activity (Chen et al., 2005). OPA1 is associated with the inner membrane and interactions with the mitofusins are still being elucidated. Mitochondrial fission requires the recruitment and assembly of the dynamin-related GTPase DNM1/DRP1 (Chan, 2006b), which constricts the diameter of mitochondria. The mitochondrial outer membrane protein FIS1 (Zhang and Chan, 2007) mediates DNM1/DRP1 recruitment to the mitochondrial surface. The machineries mediating mitochondrial fusion and fission are being elucidated, however little is known about how mitochondrial dynamics is regulated.
PINK1 and Parkin
Oxidative stress and mitochondrial dysfunction occur early in the pathogenesis of both sporadic and familial forms of Parkinson's disease (Bueler, 2009). Loss-of-function mutations in the PTEN-induced kinase 1 (PINK1) or Parkin genes, which encode a mitochondrially localized serine/threonine kinase and an ubiquitin-protein ligase, respectively, result in recessive familial forms of Parkinsonism (Poole et al., 2008; Whitworth and Pallanck, 2009). PINK1 and Parkin maintain mitochondrial integrity by regulating diverse aspects of mitochondrial function, including membrane potential, calcium homeostasis, cristae structure, respiratory activity, and mtDNA integrity (Bueler, 2010; Whitworth and Pallanck, 2009). Mutations in the PINK1 and Parkin genes result in enlarged or swollen mitochondria, and in the absence of PINK1 or Parkin cells often develop fragmented mitochondria (Bueler, 2010), suggesting a possible regulatory role for the PINK1/Parkin pathway in mitochondrial morphology (Poole et al., 2008). PINK1 is required to recruit Parkin to dysfunctional mitochondria (Ziviani et al., 2010) where PINK1 and Parkin promote mitochondrial fragmentation by targeting core components of the mitochondrial morphogenesis machinery for ubiquitination (Poole et al., 2010) leading to their degradation by autophagy. The PINK1/Parkin pathway also regulates the mitochondrial remodeling process by promoting mitochondrial fission (Park et al., 2009; Poole et al., 2008). The loss of mitochondrial integrity and the accumulation of defective mitochondria in PINK1 and Parkin mutants derives from reduced mitochondrial fission and autophagy which promote neurodegeneration in Parkinson disease (Poole et al., 2008; Poole et al., 2010; Vives-Bauza et al., 2010).
Mitochondrial DNA somatic mutations
While most aging-related studies to date have focused on inherited mtDNA mutations or deletions, somatic mutations in mtDNA lead to a condition called mtDNA heteroplasmy: a mixture of —normal and mutant mtDNA molecules in a cell. With the typical cell containing hundreds of mitochondria and each mitochondrion housing 2 to 7 mtDNA molecules, there are potentially thousands of copies of the mitochondrial genome per cell (Shuster et al., 1988). In addition, mtDNA has a mutation rate that is 10–20 times higher than that of nuclear DNA (Merriwether et al., 1991; Neckelmann et al., 1987; Wallace et al., 1997) . Both mutation and genetic drift within a mitochondrial population lead to heteroplasmy; they can can also drive a mutant mtDNA to become the dominant form(Jones et al., 2001). Somatic mtDNA mutations are common in postmitotic tissues (ie. heart, brain, nerve) and have been identified in many human tumors (Alonso et al., 1997; Bianchi et al., 1995; Chen et al., 2003b; Chen et al., 2002; Chinnery et al., 2002; Copeland et al., 2002; Fliss et al., 2000; Habano et al., 1999; Horton et al., 1996; Jeronimo et al., 2001; Jones et al., 2001; Kirches et al., 2001; Kurtz et al., 2004; Lee et al., 2004; Liu et al., 2001; Maitra et al., 2004; Nomoto et al., 2002; Parrella et al., 2001; Polyak et al., 1998; Sanchez-Cespedes et al., 2001; Sui et al., 2006; Tong et al., 2003; Wu et al., 2005; Yeh et al., 2000; Zhou et al., 2007; Zhou et al., 2006). In addition to somatically acquired mtDNA mutations, sequence deletions have also been reported in various tissues (Lee et al., 1994; Linnane et al., 1989; Melov et al., 1999b; Melov et al., 1995; Nagley et al., 1992; von Wurmb et al., 1998) including mitotic tissue (Fellous et al., 2009; Greaves et al., 2006; McDonald et al., 2008; Taylor et al., 2003), and tumors (Alonso et al., 1997; Bianchi et al., 1995; Horton et al., 1996; Lee et al., 2004; Maitra et al., 2004; Nomoto et al., 2002; Parrella et al., 2001; Wu et al., 2005)
Most individuals inherit intact healthy mitochondria at birth. Age-related somatic mtDNA mutations accumulate in postmitotic tissues until a certain tissue-specific threshold in the level of mutant to normal mtDNA molecules is surpassed and cells become compromised energetically (Hayashi et al., 1991; Rossignol et al., 1999; Wallace, 1994; Wallace et al., 1997). Heteroplasmic mutations and rearrangements of mtDNA have been reported in various tissues of elderly individuals (Hayashi et al., 1991; Melov et al., 1995; Wei, 1992; Wei, 1998a; Wei, 1998b; Zhang et al., 1998) and large-scale mtDNA deletions increase with age in skeletal muscle, heart, brain and central nervous system (Arnheim and Cortopassi, 1992; Cortopassi et al., 1992; Melov et al., 1999b; Pesce et al., 2001). The age-related accumulation of mtDNA mutations leads to impaired capacity for energy generation by OXPHOS (Melov et al., 1995; Wallace, 1995; Wallace et al., 1992; Wallace et al., 1995), decreased cellular stress resistance, and accelerated cellular mortality (Driggers et al., 1996; Ozawa, 1995a; Ozawa, 1995b; Simonetti et al., 1992; Trounce et al., 1989). Moreover, elderly adults develop more mtDNA damage and exhibit reduced activity of OXPHOS enzymes in postmitotic tissues compared to young and middle aged adults (Lu et al., 1999; Trounce et al., 1989). In general, organs with the highest ATP requirements and the lowest regenerative capacities, such as the brain, heart and skeletal muscle, are the most sensitive to the effects of mtDNA mutations (Wallace, 1994; Wallace et al., 1995).
There is evidence that heteroplasmic mtDNA for specific mutations are also associated with aging (Lee et al., 1997; Linnane et al., 1990; Linnane et al., 1989; von Wurmb-Schwark et al., 2003; Yen et al., 1994; Yen et al., 1992; Yen et al., 1991), degenerative diseases (Linnane et al., 1990; Linnane et al., 1989; Wallace, 2001) and tumors of the breast, colon, liver, head and neck, lung and prostate (Copeland et al., 2002; Horton et al., 1996; Mithani et al., 2007; Penta et al., 2001; Petros et al., 2005; Polyak et al., 1998). A common deletion of 4,977 base pairs has accumulates with age in several human postmitotic tissues (Arnheim and Cortopassi, 1992; Corral-Debrinski et al., 1992a; Corral-Debrinski et al., 1994; Cortopassi and Arnheim, 1990; Cortopassi et al., 1992; Linnane et al., 1990; Shenkar et al., 1996; Soong et al., 1992; von Wurmb-Schwark et al., 2003). When the proportion of deletion-positive mitochondria within a cell exceeds 50–60%, skeletal muscle fibers manifest a reduction in cytochrome c oxidase activity, and mitochondrial gene translation is inhibited (Hayashi et al., 1991). With the 8344A/G mutation, which causes the syndrome of myoclonic epilepsy and ragged-red fibers, the heteroplasmic threshold level is about 85% mutated DNA (Chomyn, 1998). Once this is exceeded, large changes in the phenotype can be observed with minor increases in the proportion of the mutant mtDNA. Another mutation, A3243A/G, may be related to age of onset of diabetes mellitus depending on the degree of mtDNA heteroplasmy (Guttman et al., 2001; Kato et al., 2002)and levels of mtDNA 3243A/G heteroplasmy are higher in diabetics than non-diabetics(Coon et al., 2006) (Majamaa-Voltti et al., 2006). Additional heteroplasmic mutations related to diabetes, hyperglycemia, insulin dependence and obesity include 3398T/C (Chen et al., 2000), 3254C/A (Chen et al., 2000), 3316G/A (Chen et al., 2000), 3156A/G (Ohkubo et al., 2001), 3357G/A (Ohkubo et al., 2001), 3375C/A (Ohkubo et al., 2001), and 3394T/C (Chen et al., 2000; Ohkubo et al., 2001). Several mutations in mitochondrial tRNA genes show age-related variation: 3243A/G (Poulton and Morten, 1993), 8344A/G(Lertrit et al., 1992) and 12320A/G(Weber et al., 1997), and the 3243A/G mutation is also associated with diabetes mellitus.
Oxidative stress has been implicated in the pathogenesis of neurodegenerative diseases such as Alzheimer‘s disease (AD) and Parkinson‘s disease. In an analysis of the mtDNA control region (CR), 65% of AD brains harbored a 414T/G mutation (Coskun et al., 2004). Moreover, AD brains had an average 63% increase in heteroplasmic mtDNA CR mutations; those from patients 80 years and older had a 130% increase in heteroplasmic CR mutations. The reported CR mutations preferentially altered known mtDNA regulatory elements. Certain AD brains harbored the disease-specific CR mutations at levels up to 70–80% heteroplasmy (Coskun et al., 2004). The MitoChip was recently used to assess heteroplasmy in the platelets of 19 AD patients and 18 matched controls (Coon et al., 2006).
Somatic mutations and longevity
Several studies support the importance of the acquired (Iwata et al., 2007; Rose et al., 2007; Zhang et al., 2003) and inherited (Niemi et al., 2005) C150T mutant for longevity. Rose et al. (Rose et al., 2007) set out to determine whether the accumulation of C150T heteroplasmy in leukocytes is a phenotypic consequence of extreme ageing or a genetically controlled event that may favor longevity. Centenarians and their descendants, despite the different ages, showed similar levels of C150T heteroplasmy which were significantly higher than levels in controls. In addition, heteroplasmy levels were significantly correlated in parent-offspring pairs but were independent of mtDNA inherited variability (haplogroup and sequence analyses). These findings suggest that the high degree of C150T heteroplasmy observed in centenarians is genetically controlled, and that such genetic control is independent of mtDNA variability and likely due to the nuclear genome. Iwata et al. (Iwata et al., 2007) examined leukocyte mtDNA from three groups of an Ashkenazi Jew population, including 124 95+ year old female participants, their mixed gender offspring, and mixed gender control subjects to examine the association of the C150T mutation with longevity. This analysis revealed a very low incidence of the C150T transition in the centenarians and near- centenarians and the other two groups. By contrast, a fairly high frequency of a homoplasmic T152C transition and of a homoplasmic T195C transition was seen in all three groups of subjects. An aging-related increase in incidence of the heteroplasmic T152C transition, presumably resulting from somatic events, was demonstrated in the Ashkenazi Jews (Iwata et al., 2007). Zhang et al. (Zhang et al., 2003) carried out a large-scale screening of the mtDNA CR in leukocytes from centenarians and younger controls. They found that the C150T mutation was significantly more common in centenarians than in younger controls, and provided evidence that somatic events, probably under nuclear genome control, contribute to the selective accumulation of this mutation in centenarians.
Conclusion
There is substantial evidence that mitochondria and mtDNA are involved in the aging process and possibly achieving a long life. Potential explanations include mtDNA abundance and the accumulation of somatic mutations, inherited variability, and cross-talk with the nuclear genome. We hypothesize that aging and longevity, as complex traits having a significant genetic component, are likely to be controlled by nuclear gene variants interacting with both inherited and somatic mtDNA variability. However, most previous studies of mtDNA in human subjects have had low statistical power due to the complexity of inherited lineages (haplogroups and sub-haplogroups) and small sample sizes. Perhaps more importantly, they have been unable to examine mitonuclear genetic interactions. In order to fully examine the complexity of mitochondrial-nuclear epistatsis, future studies will require: 1.) Large numbers of aged subjects; 2.) The entire sequence of mtDNA in order to consider genome-wide complexity; and 3.) An assessment of interacting mitochondrial and nuclear genes encoding mitochondrial proteins. With regard to human longevity, it is critical to examine the oldest old since there is considerable evidence that the extreme phenotype of human longevity (e.g., survival to centenarianship) is strongly heritable, even more so than survival to ages 90 or 95 (Perls and Terry, 2003; Tan et al., 2008). Thus, examining the genomes of centenarians is likely to identify genetic pathways that affect human aging and longevity.
Identifying mitochondrial genetic variants and the effects of interacting mitochondrial and nuclear genes that impact human longevity may provide insight into our understanding of aging and have relevance for many age-related diseases such as cardiovascular disease, diabetes, and Alzheimer‘s disease.
Acknowledgments
This work was supported by NIH grant U19AG023122
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Literature Cited
- Achilli A, Rengo C, Magri C, Battaglia V, Olivieri A, Scozzari R, Cruciani F, Zeviani M, Briem E, Carelli V, Moral P, Dugoujon JM, Roostalu U, Loogvali EL, Kivisild T, Bandelt HJ, Richards M, Villems R, Santachiara-Benerecetti AS, Semino O, Torroni A. The molecular dissection of mtDNA haplogroup H confirms that the Franco-Cantabrian glacial refuge was a major source for the European gene pool. Am J Hum Genet. 2004;75:910–8. doi: 10.1086/425590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahn BH, Kim HS, Song S, Lee IH, Liu J, Vassilopoulos A, Deng CX, Finkel T. A role for the mitochondrial deacetylase Sirt3 in regulating energy homeostasis. Proc Natl Acad Sci U S A. 2008;105:14447–52. doi: 10.1073/pnas.0803790105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alonso A, Marti A, Corbalan MS, Martinez-Gonzalez MA, Forga L, Martinez JA. Association of UCP3 gene -55C>T polymorphism and obesity in a Spanish population. Ann Nutr Metab. 2005;49:183–8. doi: 10.1159/000086883. [DOI] [PubMed] [Google Scholar]
- Alonso A, Martin P, Albarran C, Aquilera B, Garcia O, Guzman A, Oliva H, Sancho M. Detection of somatic mutations in the mitochondrial DNA control region of colorectal and gastric tumors by heteroduplex and single-strand conformation analysis. Electrophoresis. 1997;18:682–5. doi: 10.1002/elps.1150180504. [DOI] [PubMed] [Google Scholar]
- Ambrosone CB, Freudenheim JL, Thompson PA, Bowman E, Vena JE, Marshall JR, Graham S, Laughlin R, Nemoto T, Shields PG. Manganese superoxide dismutase (MnSOD) genetic polymorphisms, dietary antioxidants, and risk of breast cancer. Cancer Res. 1999;59:602–6. [PubMed] [Google Scholar]
- Amo T, Brand MD. Were inefficient mitochondrial haplogroups selected during migrations of modern humans? A test using modular kinetic analysis of coupling in mitochondria from cybrid cell lines. Biochem J. 2007;404:345–51. doi: 10.1042/BJ20061609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amo T, Yadava N, Oh R, Nicholls DG, Brand MD. Experimental assessment of bioenergetic differences caused by the common European mitochondrial DNA haplogroups H and T. Gene. 2008;411:69–76. doi: 10.1016/j.gene.2008.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arnheim N, Cortopassi G. Deleterious mitochondrial DNA mutations accumulate in aging human tissues. Mutat Res. 1992;275:157–67. doi: 10.1016/0921-8734(92)90020-p. [DOI] [PubMed] [Google Scholar]
- Balloux F, Handley LJ, Jombart T, Liu H, Manica A. Climate shaped the worldwide distribution of human mitochondrial DNA sequence variation. Proc Biol Sci. 2009;276:3447–55. doi: 10.1098/rspb.2009.0752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Battersby BJ, Shoubridge EA. Selection of a mtDNA sequence variant in hepatocytes of heteroplasmic mice is not due to differences in respiratory chain function or efficiency of replication. Hum Mol Genet. 2001;10:2469–79. doi: 10.1093/hmg/10.22.2469. [DOI] [PubMed] [Google Scholar]
- Beier K, Volkl A, Fahimi HD. The impact of aging on enzyme proteins of rat liver peroxisomes: quantitative analysis by immunoblotting and immunoelectron microscopy. Virchows Arch B Cell Pathol Incl Mol Pathol. 1993;63:139–46. doi: 10.1007/BF02899254. [DOI] [PubMed] [Google Scholar]
- Bellizzi D, Rose G, Cavalcante P, Covello G, Dato S, De Rango F, Greco V, Maggiolini M, Feraco E, Mari V, Franceschi C, Passarino G, De Benedictis G. A novel VNTR enhancer within the SIRT3 gene, a human homologue of SIR2, is associated with survival at oldest ages. Genomics. 2005;85:258–63. doi: 10.1016/j.ygeno.2004.11.003. [DOI] [PubMed] [Google Scholar]
- Bender A, Krishnan KJ, Morris CM, Taylor GA, Reeve AK, Perry RH, Jaros E, Hersheson JS, Betts J, Klopstock T, Taylor RW, Turnbull DM. High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat Genet. 2006;38:515–7. doi: 10.1038/ng1769. [DOI] [PubMed] [Google Scholar]
- Benzi G, Pastoris O, Marzatico F, Villa RF, Dagani F, Curti D. The mitochondrial electron transfer alteration as a factor involved in the brain aging. Neurobiol Aging. 1992;13:361–8. doi: 10.1016/0197-4580(92)90109-b. [DOI] [PubMed] [Google Scholar]
- Berentzen T, Dalgaard LT, Petersen L, Pedersen O, Sorensen TI. Interactions between physical activity and variants of the genes encoding uncoupling proteins -2 and -3 in relation to body weight changes during a 10-y follow-up. Int J Obes (Lond) 2005;29:93–9. doi: 10.1038/sj.ijo.0802841. [DOI] [PubMed] [Google Scholar]
- Bianchi MS, Bianchi NO, Bailliet G. Mitochondrial DNA mutations in normal and tumor tissues from breast cancer patients. Cytogenet Cell Genet. 1995;71:99–103. doi: 10.1159/000134072. [DOI] [PubMed] [Google Scholar]
- Blander G, Guarente L. The Sir2 family of protein deacetylases. Annu Rev Biochem. 2004;73:417–35. doi: 10.1146/annurev.biochem.73.011303.073651. [DOI] [PubMed] [Google Scholar]
- Blier PU, Dufresne F, Burton RS. Natural selection and the evolution of mtDNA-encoded peptides: evidence for intergenomic co-adaptation. Trends Genet. 2001;17:400–6. doi: 10.1016/s0168-9525(01)02338-1. [DOI] [PubMed] [Google Scholar]
- Boffoli D, Scacco SC, Vergari R, Solarino G, Santacroce G, Papa S. Decline with age of the respiratory chain activity in human skeletal muscle. Biochim Biophys Acta. 1994;1226:73–82. doi: 10.1016/0925-4439(94)90061-2. [DOI] [PubMed] [Google Scholar]
- Bordone L, Motta MC, Picard F, Robinson A, Jhala US, Apfeld J, McDonagh T, Lemieux M, McBurney M, Szilvasi A, Easlon EJ, Lin SJ, Guarente L. Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic beta cells. PLoS Biol. 2006;4:e31. doi: 10.1371/journal.pbio.0040031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bowling AC, Mutisya EM, Walker LC, Price DL, Cork LC, Beal MF. Age-dependent impairment of mitochondrial function in primate brain. J Neurochem. 1993;60:1964–7. doi: 10.1111/j.1471-4159.1993.tb13430.x. [DOI] [PubMed] [Google Scholar]
- Brown MD, Starikovskaya E, Derbeneva O, Hosseini S, Allen JC, Mikhailovskaya IE, Sukernik RI, Wallace DC. The role of mtDNA background in disease expression: a new primary LHON mutation associated with Western Eurasian haplogroup J. Hum Genet. 2002;110:130–8. doi: 10.1007/s00439-001-0660-8. [DOI] [PubMed] [Google Scholar]
- Bua E, Johnson J, Herbst A, Delong B, McKenzie D, Salamat S, Aiken JM. Mitochondrial DNA-deletion mutations accumulate intracellularly to detrimental levels in aged human skeletal muscle fibers. Am J Hum Genet. 2006;79:469–80. doi: 10.1086/507132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bueler H. Impaired mitochondrial dynamics and function in the pathogenesis of Parkinson's disease. Exp Neurol. 2009;218:235–46. doi: 10.1016/j.expneurol.2009.03.006. [DOI] [PubMed] [Google Scholar]
- Bueler H. Mitochondrial dynamics, cell death and the pathogenesis of Parkinson's disease. Apoptosis. 2010 doi: 10.1007/s10495-010-0465-0. [DOI] [PubMed] [Google Scholar]
- Buemann B, Schierning B, Toubro S, Bibby BM, Sorensen T, Dalgaard L, Pedersen O, Astrup A. The association between the val/ala-55 polymorphism of the uncoupling protein 2 gene and exercise efficiency. Int J Obes Relat Metab Disord. 2001;25:467–71. doi: 10.1038/sj.ijo.0801564. [DOI] [PubMed] [Google Scholar]
- Bulotta A, Ludovico O, Coco A, Di Paola R, Quattrone A, Carella M, Pellegrini F, Prudente S, Trischitta V. The common -866G/A polymorphism in the promoter region of the UCP-2 gene is associated with reduced risk of type 2 diabetes in Caucasians from Italy. J Clin Endocrinol Metab. 2005;90:1176–80. doi: 10.1210/jc.2004-1072. [DOI] [PubMed] [Google Scholar]
- Burton RS, Rawson PD, Edmands S. Genetic Architecture of Physiological Phenotypes: Empirical Evidence for Coadapted Gene Complexes. Amer Zool. 1999;39:451–462. [Google Scholar]
- Bykhovskaya Y, Estivill X, Taylor K, Hang T, Hamon M, Casano RA, Yang H, Rotter JI, Shohat M, Fischel-Ghodsian N. Candidate locus for a nuclear modifier gene for maternally inherited deafness. Am J Hum Genet. 2000;66:1905–10. doi: 10.1086/302914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campbell DA, Sundaramurthy D, Gordon D, Markham AF, Pieri LF. Association between a marker in the UCP-2/UCP-3 gene cluster and genetic susceptibility to anorexia nervosa. Mol Psychiatry. 1999;4:68–70. doi: 10.1038/sj.mp.4000477. [DOI] [PubMed] [Google Scholar]
- Carelli V, Vergani L, Bernazzi B, Zampieron C, Bucchi L, Valentino M, Rengo C, Torroni A, Martinuzzi A. Respiratory function in cybrid cell lines carrying European mtDNA haplogroups: implications for Leber's hereditary optic neuropathy. Biochim Biophys Acta. 2002;1588:7–14. doi: 10.1016/s0925-4439(02)00097-2. [DOI] [PubMed] [Google Scholar]
- Cha MH, Kim IC, Kim KS, Kang BK, Choi SM, Yoon Y. Association of UCP2 and UCP3 gene polymorphisms with serum high-density lipoprotein cholesterol among Korean women. Metabolism. 2007;56:806–13. doi: 10.1016/j.metabol.2007.01.023. [DOI] [PubMed] [Google Scholar]
- Cha MH, Shin HD, Kim KS, Lee BH, Yoon Y. The effects of uncoupling protein 3 haplotypes on obesity phenotypes and very low-energy diet-induced changes among overweight Korean female subjects. Metabolism. 2006;55:578–86. doi: 10.1016/j.metabol.2005.11.012. [DOI] [PubMed] [Google Scholar]
- Chan DC. Dissecting mitochondrial fusion. Dev Cell. 2006a;11:592–4. doi: 10.1016/j.devcel.2006.10.009. [DOI] [PubMed] [Google Scholar]
- Chan DC. Mitochondrial fusion and fission in mammals. Annu Rev Cell Dev Biol. 2006b;22:79–99. doi: 10.1146/annurev.cellbio.22.010305.104638. [DOI] [PubMed] [Google Scholar]
- Chang MC, Hung SC, Chen WY, Chen TL, Lee CF, Lee HC, Wang KL, Chiou CC, Wei YH. Accumulation of mitochondrial DNA with 4977-bp deletion in knee cartilage--an association with idiopathic osteoarthritis. Osteoarthritis Cartilage. 2005;13:1004–11. doi: 10.1016/j.joca.2005.06.011. [DOI] [PubMed] [Google Scholar]
- Chen H, Chan DC. Emerging functions of mammalian mitochondrial fusion and fission. Hum Mol Genet. 2005;14(Spec No 2):R283–9. doi: 10.1093/hmg/ddi270. [DOI] [PubMed] [Google Scholar]
- Chen H, Chomyn A, Chan DC. Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J Biol Chem. 2005;280:26185–92. doi: 10.1074/jbc.M503062200. [DOI] [PubMed] [Google Scholar]
- Chen H, Detmer SA, Ewald AJ, Griffin EE, Fraser SE, Chan DC. Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J Cell Biol. 2003a;160:189–200. doi: 10.1083/jcb.200211046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen JZ, Gokden N, Greene GF, Green B, Kadlubar FF. Simultaneous generation of multiple mitochondrial DNA mutations in human prostate tumors suggests mitochondrial hyper-mutagenesis. Carcinogenesis. 2003b;24:1481–7. doi: 10.1093/carcin/bgg102. [DOI] [PubMed] [Google Scholar]
- Chen JZ, Gokden N, Greene GF, Mukunyadzi P, Kadlubar FF. Extensive somatic mitochondrial mutations in primary prostate cancer using laser capture microdissection. Cancer Res. 2002;62:6470–4. [PubMed] [Google Scholar]
- Chen Y, Liao WX, Roy AC, Loganath A, Ng SC. Mitochondrial gene mutations in gestational diabetes mellitus. Diabetes Res Clin Pract. 2000;48:29–35. doi: 10.1016/s0168-8227(99)00138-2. [DOI] [PubMed] [Google Scholar]
- Chinnery PF, Samuels DC, Elson J, Turnbull DM. Accumulation of mitochondrial DNA mutations in ageing, cancer, and mitochondrial disease: is there a common mechanism? Lancet. 2002;360:1323–5. doi: 10.1016/S0140-6736(02)11310-9. [DOI] [PubMed] [Google Scholar]
- Chinnery PF, Taylor GA, Howell N, Brown DT, Parsons TJ, Turnbull DM. Point mutations of the mtDNA control region in normal and neurodegenerative human brains. Am J Hum Genet. 2001;68:529–32. doi: 10.1086/318204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chiu YJ, Richardson A. Effect of age on the function of mitochondria isolated from brain and heart tissue. Exp Gerontol. 1980;15:511–7. doi: 10.1016/0531-5565(80)90003-0. [DOI] [PubMed] [Google Scholar]
- Chomyn A. The myoclonic epilepsy and ragged-red fiber mutation provides new insights into human mitochondrial function and genetics. Am J Hum Genet. 1998;62:745–51. doi: 10.1086/301813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christiansen L, Petersen HC, Bathum L, Frederiksen H, McGue M, Christensen K. The catalase -262C/T promoter polymorphism and aging phenotypes. J Gerontol A Biol Sci Med Sci. 2004;59:B886–9. doi: 10.1093/gerona/59.9.b886. [DOI] [PubMed] [Google Scholar]
- Clark AG, Lyckegaard EM. Natural selection with nuclear and cytoplasmic transmission. III. Joint analysis of segregation and mtDNA in Drosophila melanogaster. Genetics. 1988;118:471–81. doi: 10.1093/genetics/118.3.471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coon KD, Valla J, Szelinger S, Schneider LE, Niedzielko TL, Brown KM, Pearson JV, Halperin R, Dunckley T, Papassotiropoulos A, Caselli RJ, Reiman EM, Stephan DA. Quantitation of heteroplasmy of mtDNA sequence variants identified in a population of AD patients and controls by array-based resequencing. Mitochondrion. 2006;6:194–210. doi: 10.1016/j.mito.2006.07.002. [DOI] [PubMed] [Google Scholar]
- Cooper JM, Mann VM, Schapira AH. Analyses of mitochondrial respiratory chain function and mitochondrial DNA deletion in human skeletal muscle: effect of ageing. J Neurol Sci. 1992;113:91–8. doi: 10.1016/0022-510x(92)90270-u. [DOI] [PubMed] [Google Scholar]
- Copeland JM, Cho J, Lo T, Jr, Hur JH, Bahadorani S, Arabyan T, Rabie J, Soh J, Walker DW. Extension of Drosophila life span by RNAi of the mitochondrial respiratory chain. Curr Biol. 2009;19:1591–8. doi: 10.1016/j.cub.2009.08.016. [DOI] [PubMed] [Google Scholar]
- Copeland WC, Wachsman JT, Johnson FM, Penta JS. Mitochondrial DNA alterations in cancer. Cancer Invest. 2002;20:557–69. doi: 10.1081/cnv-120002155. [DOI] [PubMed] [Google Scholar]
- Corral-Debrinski M, Horton T, Lott MT, Shoffner JM, Beal MF, Wallace DC. Mitochondrial DNA deletions in human brain: regional variability and increase with advanced age. Nat Genet. 1992a;2:324–9. doi: 10.1038/ng1292-324. [DOI] [PubMed] [Google Scholar]
- Corral-Debrinski M, Horton T, Lott MT, Shoffner JM, McKee AC, Beal MF, Graham BH, Wallace DC. Marked changes in mitochondrial DNA deletion levels in Alzheimer brains. Genomics. 1994;23:471–6. doi: 10.1006/geno.1994.1525. [DOI] [PubMed] [Google Scholar]
- Corral-Debrinski M, Shoffner JM, Lott MT, Wallace DC. Association of mitochondrial DNA damage with aging and coronary atherosclerotic heart disease. Mutat Res. 1992b;275:169–80. doi: 10.1016/0921-8734(92)90021-g. [DOI] [PubMed] [Google Scholar]
- Cortopassi GA, Arnheim N. Detection of a specific mitochondrial DNA deletion in tissues of older humans. Nucleic Acids Res. 1990;18:6927–33. doi: 10.1093/nar/18.23.6927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cortopassi GA, Shibata D, Soong NW, Arnheim N. A pattern of accumulation of a somatic deletion of mitochondrial DNA in aging human tissues. Proc Natl Acad Sci U S A. 1992;89:7370–4. doi: 10.1073/pnas.89.16.7370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coskun PE, Beal MF, Wallace DC. Alzheimer's brains harbor somatic mtDNA control-region mutations that suppress mitochondrial transcription and replication. Proc Natl Acad Sci U S A. 2004;101:10726–31. doi: 10.1073/pnas.0403649101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- D'Adamo M, Perego L, Cardellini M, Marini MA, Frontoni S, Andreozzi F, Sciacqua A, Lauro D, Sbraccia P, Federici M, Paganelli M, Pontiroli AE, Lauro R, Perticone F, Folli F, Sesti G. The -866A/A genotype in the promoter of the human uncoupling protein 2 gene is associated with insulin resistance and increased risk of type 2 diabetes. Diabetes. 2004;53:1905–10. doi: 10.2337/diabetes.53.7.1905. [DOI] [PubMed] [Google Scholar]
- Dato S, Passarino G, Rose G, Altomare K, Bellizzi D, Mari V, Feraco E, Franceschi C, De Benedictis G. Association of the mitochondrial DNA haplogroup J with longevity is population specific. Eur J Hum Genet. 2004;12:1080–2. doi: 10.1038/sj.ejhg.5201278. [DOI] [PubMed] [Google Scholar]
- Davidson MM, Walker WF, Hernandez-Rosa E, Nesti C. Evidence for nuclear modifier gene in mitochondrial cardiomyopathy. J Mol Cell Cardiol. 2009;46:936–42. doi: 10.1016/j.yjmcc.2009.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Benedictis G, Carrieri G, Garasto S, Rose G, Varcasia O, Bonafe M, Franceschi C, Jazwinski SM. Does a retrograde response in human aging and longevity exist? Exp Gerontol. 2000;35:795–801. doi: 10.1016/s0531-5565(00)00169-8. [DOI] [PubMed] [Google Scholar]
- De Benedictis G, Rose G, Carrieri G, De Luca M, Falcone E, Passarino G, Bonafe M, Monti D, Baggio G, Bertolini S, Mari D, Mattace R, Franceschi C. Mitochondrial DNA inherited variants are associated with successful aging and longevity in humans. Faseb J. 1999;13:1532–6. doi: 10.1096/fasebj.13.12.1532. [DOI] [PubMed] [Google Scholar]
- de Haan JB, Bladier C, Griffiths P, Kelner M, O'Shea RD, Cheung NS, Bronson RT, Silvestro MJ, Wild S, Zheng SS, Beart PM, Hertzog PJ, Kola I. Mice with a homozygous null mutation for the most abundant glutathione peroxidase, Gpx1, show increased susceptibility to the oxidative stress-inducing agents paraquat and hydrogen peroxide. J Biol Chem. 1998;273:22528–36. doi: 10.1074/jbc.273.35.22528. [DOI] [PubMed] [Google Scholar]
- Dell'agnello C, Leo S, Agostino A, Szabadkai G, Tiveron C, Zulian A, Prelle A, Roubertoux P, Rizzuto R, Zeviani M. Increased longevity and refractoriness to Ca(2+)-dependent neurodegeneration in Surf1 knockout mice. Hum Mol Genet. 2007;16:431–44. doi: 10.1093/hmg/ddl477. [DOI] [PubMed] [Google Scholar]
- Desai VG, Weindruch R, Hart RW, Feuers RJ. Influences of age and dietary restriction on gastrocnemius electron transport system activities in mice. Arch Biochem Biophys. 1996;333:145–51. doi: 10.1006/abbi.1996.0375. [DOI] [PubMed] [Google Scholar]
- Detmer SA, Chan DC. Functions and dysfunctions of mitochondrial dynamics. Nat Rev Mol Cell Biol. 2007;8:870–9. doi: 10.1038/nrm2275. [DOI] [PubMed] [Google Scholar]
- Dowling DK, Friberg U, Hailer F, Arnqvist G. Intergenomic epistasis for fitness: within-population interactions between cytoplasmic and nuclear genes in Drosophila melanogaster. Genetics. 2007;175:235–44. doi: 10.1534/genetics.105.052050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Driggers WJ, Grishko VI, LeDoux SP, Wilson GL. Defective repair of oxidative damage in the mitochondrial DNA of a xeroderma pigmentosum group A cell line. Cancer Res. 1996;56:1262–6. [PubMed] [Google Scholar]
- Elson JL, Turnbull DM, Howell N. Comparative genomics and the evolution of human mitochondrial DNA: assessing the effects of selection. Am J Hum Genet. 2004;74:229–38. doi: 10.1086/381505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Esterbauer H, Oberkofler H, Liu YM, Breban D, Hell E, Krempler F, Patsch W. Uncoupling protein-1 mRNA expression in obese human subjects: the role of sequence variations at the uncoupling protein-1 gene locus. J Lipid Res. 1998;39:834–44. [PubMed] [Google Scholar]
- Etterson JR, Keller SR, Galloway LF. Epistatic and cytonuclear interactions govern outbreeding depression in the autotetraploid Campanulastrum americanum. Evolution. 2007;61:2671–83. doi: 10.1111/j.1558-5646.2007.00234.x. [DOI] [PubMed] [Google Scholar]
- Evans D, Minouchehr S, Hagemann G, Mann WA, Wendt D, Wolf A, Beisiegel U. Frequency of and interaction between polymorphisms in the beta3-adrenergic receptor and in uncoupling proteins 1 and 2 and obesity in Germans. Int J Obes Relat Metab Disord. 2000;24:1239–45. doi: 10.1038/sj.ijo.0801402. [DOI] [PubMed] [Google Scholar]
- Fellous TG, Islam S, Tadrous PJ, Elia G, Kocher HM, Bhattacharya S, Mears L, Turnbull DM, Taylor RW, Greaves LC, Chinnery PF, Taylor G, McDonald SA, Wright NA, Alison MR. Locating the stem cell niche and tracing hepatocyte lineages in human liver. Hepatology. 2009;49:1655–63. doi: 10.1002/hep.22791. [DOI] [PubMed] [Google Scholar]
- Ferguson M, Mockett RJ, Shen Y, Orr WC, Sohal RS. Age-associated decline in mitochondrial respiration and electron transport in Drosophila melanogaster. Biochem J. 2005;390:501–11. doi: 10.1042/BJ20042130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flachsbart F, Croucher PJ, Nikolaus S, Hampe J, Cordes C, Schreiber S, Nebel A. Sirtuin 1 (SIRT1) sequence variation is not associated with exceptional human longevity. Exp Gerontol. 2005 doi: 10.1016/j.exger.2005.09.008. [DOI] [PubMed] [Google Scholar]
- Fliss MS, Usadel H, Caballero OL, Wu L, Buta MR, Eleff SM, Jen J, Sidransky D. Facile detection of mitochondrial DNA mutations in tumors and bodily fluids. Science. 2000;287:2017–9. doi: 10.1126/science.287.5460.2017. [DOI] [PubMed] [Google Scholar]
- Forsberg L, Lyrenas L, de Faire U, Morgenstern R. A common functional C-T substitution polymorphism in the promoter region of the human catalase gene influences transcription factor binding, reporter gene transcription and is correlated to blood catalase levels. Free Radic Biol Med. 2001;30:500–5. doi: 10.1016/s0891-5849(00)00487-1. [DOI] [PubMed] [Google Scholar]
- Frye RA. Phylogenetic classification of prokaryotic and eukaryotic Sir2-like proteins. Biochem Biophys Res Commun. 2000;273:793–8. doi: 10.1006/bbrc.2000.3000. [DOI] [PubMed] [Google Scholar]
- Greaves LC, Preston SL, Tadrous PJ, Taylor RW, Barron MJ, Oukrif D, Leedham SJ, Deheragoda M, Sasieni P, Novelli MR, Jankowski JA, Turnbull DM, Wright NA, McDonald SA. Mitochondrial DNA mutations are established in human colonic stem cells, and mutated clones expand by crypt fission. Proc Natl Acad Sci U S A. 2006;103:714–9. doi: 10.1073/pnas.0505903103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gregoretti IV, Lee YM, Goodson HV. Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis. J Mol Biol. 2004;338:17–31. doi: 10.1016/j.jmb.2004.02.006. [DOI] [PubMed] [Google Scholar]
- Grozinger CM, Schreiber SL. Deacetylase enzymes: biological functions and the use of small-molecule inhibitors. Chem Biol. 2002;9:3–16. doi: 10.1016/s1074-5521(02)00092-3. [DOI] [PubMed] [Google Scholar]
- Guarente L, Kenyon C. Genetic pathways that regulate ageing in model organisms. Nature. 2000;408:255–62. doi: 10.1038/35041700. [DOI] [PubMed] [Google Scholar]
- Guttman A, Gao HG, Haas R. Rapid analysis of mitochondrial DNA heteroplasmy in diabetes by gel-microchip electrophoresis. Clin Chem. 2001;47:1469–72. [PubMed] [Google Scholar]
- Habano W, Sugai T, Yoshida T, Nakamura S. Mitochondrial gene mutation, but not large-scale deletion, is a feature of colorectal carcinomas with mitochondrial microsatellite instability. Int J Cancer. 1999;83:625–9. doi: 10.1002/(sici)1097-0215(19991126)83:5<625::aid-ijc10>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
- Haigis MC, Mostoslavsky R, Haigis KM, Fahie K, Christodoulou DC, Murphy AJ, Valenzuela DM, Yancopoulos GD, Karow M, Blander G, Wolberger C, Prolla TA, Weindruch R, Alt FW, Guarente L. SIRT4 inhibits glutamate dehydrogenase and opposes the effects of calorie restriction in pancreatic beta cells. Cell. 2006;126:941–54. doi: 10.1016/j.cell.2006.06.057. [DOI] [PubMed] [Google Scholar]
- Hasegawa K, Wakino S, Yoshioka K, Tatematsu S, Hara Y, Minakuchi H, Washida N, Tokuyama H, Hayashi K, Itoh H. Sirt1 protects against oxidative stress-induced renal tubular cell apoptosis by the bidirectional regulation of catalase expression. Biochem Biophys Res Commun. 2008;372:51–6. doi: 10.1016/j.bbrc.2008.04.176. [DOI] [PubMed] [Google Scholar]
- Hattori K, Tanaka M, Sugiyama S, Obayashi T, Ito T, Satake T, Hanaki Y, Asai J, Nagano M, Ozawa T. Age-dependent increase in deleted mitochondrial DNA in the human heart: possible contributory factor to presbycardia. Am Heart J. 1991;121:1735–42. doi: 10.1016/0002-8703(91)90020-i. [DOI] [PubMed] [Google Scholar]
- Hayakawa M, Sugiyama S, Hattori K, Takasawa M, Ozawa T. Age-associated damage in mitochondrial DNA in human hearts. Mol Cell Biochem. 1993;119:95–103. doi: 10.1007/BF00926859. [DOI] [PubMed] [Google Scholar]
- Hayashi J, Ohta S, Kikuchi A, Takemitsu M, Goto Y, Nonaka I. Introduction of disease-related mitochondrial DNA deletions into HeLa cells lacking mitochondrial DNA results in mitochondrial dysfunction. Proc Natl Acad Sci U S A. 1991;88:10614–8. doi: 10.1073/pnas.88.23.10614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heilbronn LK, Kind KL, Pancewicz E, Morris AM, Noakes M, Clifton PM. Association of -3826 G variant in uncoupling protein-1 with increased BMI in overweight Australian women. Diabetologia. 2000;43:242–4. doi: 10.1007/s001250050036. [DOI] [PubMed] [Google Scholar]
- Herbst A, Pak JW, McKenzie D, Bua E, Bassiouni M, Aiken JM. Accumulation of mitochondrial DNA deletion mutations in aged muscle fibers: evidence for a causal role in muscle fiber loss. J Gerontol A Biol Sci Med Sci. 2007;62:235–45. doi: 10.1093/gerona/62.3.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hermjakob H, Montecchi-Palazzi L, Lewington C, Mudali S, Kerrien S, Orchard S, Vingron M, Roechert B, Roepstorff P, Valencia A, Margalit H, Armstrong J, Bairoch A, Cesareni G, Sherman D, Apweiler R. IntAct: an open source molecular interaction database. Nucleic Acids Res. 2004;32:D452–5. doi: 10.1093/nar/gkh052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herrmann SM, Wang JG, Staessen JA, Kertmen E, Schmidt-Petersen K, Zidek W, Paul M, Brand E. Uncoupling protein 1 and 3 polymorphisms are associated with waist-to-hip ratio. J Mol Med. 2003;81:327–32. doi: 10.1007/s00109-003-0431-1. [DOI] [PubMed] [Google Scholar]
- Ho YS, Gargano M, Cao J, Bronson RT, Heimler I, Hutz RJ. Reduced fertility in female mice lacking copper-zinc superoxide dismutase. J Biol Chem. 1998;273:7765–9. doi: 10.1074/jbc.273.13.7765. [DOI] [PubMed] [Google Scholar]
- Horton TM, Petros JA, Heddi A, Shoffner J, Kaufman AE, Graham SD, Jr, Gramlich T, Wallace DC. Novel mitochondrial DNA deletion found in a renal cell carcinoma. Genes Chromosomes Cancer. 1996;15:95–101. doi: 10.1002/(SICI)1098-2264(199602)15:2<95::AID-GCC3>3.0.CO;2-Z. [DOI] [PubMed] [Google Scholar]
- Howell N, Herrnstadt C, Shults C, Mackey DA. Low penetrance of the 14484 LHON mutation when it arises in a non-haplogroup J mtDNA background. Am J Med Genet A. 2003;119A:147–51. doi: 10.1002/ajmg.a.20135. [DOI] [PubMed] [Google Scholar]
- Hutter CM, Rand DM. Competition between mitochondrial haplotypes in distinct nuclear genetic environments: Drosophila pseudoobscura vs. D. persimilis. Genetics. 1995;140:537–48. doi: 10.1093/genetics/140.2.537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Imai S, Armstrong CM, Kaeberlein M, Guarente L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature. 2000;403:795–800. doi: 10.1038/35001622. [DOI] [PubMed] [Google Scholar]
- Ishii K, Zhen LX, Wang DH, Funamori Y, Ogawa K, Taketa K. Prevention of mammary tumorigenesis in acatalasemic mice by vitamin E supplementation. Jpn J Cancer Res. 1996;87:680–4. doi: 10.1111/j.1349-7006.1996.tb00277.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ito A, Watanabe H, Aoyama H, Nakagawa Y, Mori M. Effect of 1,2-dimethylhydrazine and hydrogen peroxide for the duodenal tumorigenesis in relation to blood catalase activity in mice. Hiroshima J Med Sci. 1986;35:197–200. [PubMed] [Google Scholar]
- Itoh K, Weis S, Mehraein P, Muller-Hocker J. Cytochrome c oxidase defects of the human substantia nigra in normal aging. Neurobiol Aging. 1996;17:843–8. doi: 10.1016/s0197-4580(96)00168-6. [DOI] [PubMed] [Google Scholar]
- Ivanova R, Lepage V, Charron D, Schachter F. Mitochondrial genotype associated with French Caucasian centenarians. Gerontology. 1998;44:349. doi: 10.1159/000022041. [DOI] [PubMed] [Google Scholar]
- Iwata N, Zhang J, Atzmon G, Leanza S, Cho J, Chomyn A, Burk RD, Barzilai N, Attardi G. Aging-related occurrence in Ashkenazi Jews of leukocyte heteroplasmic mtDNA mutation adjacent to replication origin frequently remodeled in Italian centenarians. Mitochondrion. 2007;7:267–72. doi: 10.1016/j.mito.2007.03.001. [DOI] [PubMed] [Google Scholar]
- Jacobs HT. The mitochondrial theory of aging: dead or alive? Aging Cell. 2003;2:11–7. doi: 10.1046/j.1474-9728.2003.00032.x. [DOI] [PubMed] [Google Scholar]
- James AC, Ballard JW. Mitochondrial genotype affects fitness in Drosophila simulans. Genetics. 2003;164:187–94. doi: 10.1093/genetics/164.1.187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jazin EE, Cavelier L, Eriksson I, Oreland L, Gyllensten U. Human brain contains high levels of heteroplasmy in the noncoding regions of mitochondrial DNA. Proc Natl Acad Sci U S A. 1996;93:12382–7. doi: 10.1073/pnas.93.22.12382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeronimo C, Nomoto S, Caballero OL, Usadel H, Henrique R, Varzim G, Oliveira J, Lopes C, Fliss MS, Sidransky D. Mitochondrial mutations in early stage prostate cancer and bodily fluids. Oncogene. 2001;20:5195–8. doi: 10.1038/sj.onc.1204646. [DOI] [PubMed] [Google Scholar]
- Johnstone RW. Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat Rev Drug Discov. 2002;1:287–99. doi: 10.1038/nrd772. [DOI] [PubMed] [Google Scholar]
- Jones JB, Song JJ, Hempen PM, Parmigiani G, Hruban RH, Kern SE. Detection of mitochondrial DNA mutations in pancreatic cancer offers a "mass"-ive advantage over detection of nuclear DNA mutations. Cancer Res. 2001;61:1299–304. [PubMed] [Google Scholar]
- Kadenbach B, Munscher C, Frank V, Muller-Hocker J, Napiwotzki J. Human aging is associated with stochastic somatic mutations of mitochondrial DNA. Mutat Res. 1995;338:161–72. doi: 10.1016/0921-8734(95)00021-w. [DOI] [PubMed] [Google Scholar]
- Kaeberlein M, Kirkland KT, Fields S, Kennedy BK. Sir2-independent life span extension by calorie restriction in yeast. PLoS Biol. 2004;2:E296. doi: 10.1371/journal.pbio.0020296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaeberlein M, McVey M, Guarente L. The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. Genes Dev. 1999;13:2570–80. doi: 10.1101/gad.13.19.2570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kato Y, Miura Y, Inagaki A, Itatsu T, Oiso Y. Age of onset possibly associated with the degree of heteroplasmy in two male siblings with diabetes mellitus having an A to G transition at 3243 of mitochondrial DNA. Diabet Med. 2002;19:784–6. doi: 10.1046/j.1464-5491.2002.00777.x. [DOI] [PubMed] [Google Scholar]
- Kelly A, Stanley CA. Disorders of glutamate metabolism. Ment Retard Dev Disabil Res Rev. 2001;7:287–95. doi: 10.1002/mrdd.1040. [DOI] [PubMed] [Google Scholar]
- Kenyon L, Moraes CT. Expanding the functional human mitochondrial DNA database by the establishment of primate xenomitochondrial cybrids. Proc Natl Acad Sci U S A. 1997;94:9131–5. doi: 10.1073/pnas.94.17.9131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kilpatrick ST, Rand DM. Conditional hitchhiking of mitochondrial DNA: frequency shifts of Drosophila melanogaster mtDNA variants depend on nuclear genetic background. Genetics. 1995;141:1113–24. doi: 10.1093/genetics/141.3.1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim KS, Cho DY, Kim YJ, Choi SM, Kim JY, Shin SU, Yoon YS. The finding of new genetic polymorphism of UCP-1 A-1766G and its effects on body fat accumulation. Biochim Biophys Acta. 2005;1741:149–55. doi: 10.1016/j.bbadis.2004.11.026. [DOI] [PubMed] [Google Scholar]
- Kirches E, Krause G, Warich-Kirches M, Weis S, Schneider T, Meyer-Puttlitz B, Mawrin C, Dietzmann K. High frequency of mitochondrial DNA mutations in glioblastoma multiforme identified by direct sequence comparison to blood samples. Int J Cancer. 2001;93:534–8. doi: 10.1002/ijc.1375. [DOI] [PubMed] [Google Scholar]
- Kivisild T, Shen P, Wall DP, Do B, Sung R, Davis K, Passarino G, Underhill PA, Scharfe C, Torroni A, Scozzari R, Modiano D, Coppa A, de Knijff P, Feldman M, Cavalli-Sforza LL, Oefner PJ. The role of selection in the evolution of human mitochondrial genomes. Genetics. 2006;172:373–87. doi: 10.1534/genetics.105.043901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koepke JI, Wood CS, Terlecky LJ, Walton PA, Terlecky SR. Progeric effects of catalase inactivation in human cells. Toxicol Appl Pharmacol. 2008;232:99–108. doi: 10.1016/j.taap.2008.06.004. [DOI] [PubMed] [Google Scholar]
- Kotani K, Sakane N, Saiga K, Adachi S, Shimohiro H, Mu H, Kurozawa Y. Relationship between A-3826G polymorphism in the promoter of the uncoupling protein-1 gene and high-density lipoprotein cholesterol in Japanese individuals: a cross-sectional study. Arch Med Res. 2008;39:142–6. doi: 10.1016/j.arcmed.2007.07.002. [DOI] [PubMed] [Google Scholar]
- Kotani K, Sakane N, Saiga K, Tsuzaki K, Shimohiro H, Tabata M, Kurozawa Y. The uncoupling protein-1 gene -3826A/G polymorphism and hypertension in Japanese subjects. Clin Chem Lab Med. 2007;45:1186–9. doi: 10.1515/CCLM.2007.253. [DOI] [PubMed] [Google Scholar]
- Kraytsberg Y, Kudryavtseva E, McKee AC, Geula C, Kowall NW, Khrapko K. Mitochondrial DNA deletions are abundant and cause functional impairment in aged human substantia nigra neurons. Nat Genet. 2006;38:518–20. doi: 10.1038/ng1778. [DOI] [PubMed] [Google Scholar]
- Krishnan KJ, Greaves LC, Reeve AK, Turnbull DM. Mitochondrial DNA mutations and aging. Ann N Y Acad Sci. 2007;1100:227–40. doi: 10.1196/annals.1395.024. [DOI] [PubMed] [Google Scholar]
- Kuningas M, Putters M, Westendorp RG, Slagboom PE, van Heemst D. SIRT1 gene, age-related diseases, and mortality: the Leiden 85-plus study. J Gerontol A Biol Sci Med Sci. 2007;62:960–5. doi: 10.1093/gerona/62.9.960. [DOI] [PubMed] [Google Scholar]
- Kurdistani SK, Grunstein M. Histone acetylation and deacetylation in yeast. Nat Rev Mol Cell Biol. 2003;4:276–84. doi: 10.1038/nrm1075. [DOI] [PubMed] [Google Scholar]
- Kurtz A, Lueth M, Kluwe L, Zhang T, Foster R, Mautner VF, Hartmann M, Tan DJ, Martuza RL, Friedrich RE, Driever PH, Wong LJ. Somatic mitochondrial DNA mutations in neurofibromatosis type 1-associated tumors. Mol Cancer Res. 2004;2:433–41. [PubMed] [Google Scholar]
- Kwong LK, Sohal RS. Age-related changes in activities of mitochondrial electron transport complexes in various tissues of the mouse. Arch Biochem Biophys. 2000;373:16–22. doi: 10.1006/abbi.1999.1495. [DOI] [PubMed] [Google Scholar]
- Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin C, Daussin F, Messadeq N, Milne J, Lambert P, Elliott P, Geny B, Laakso M, Puigserver P, Auwerx J. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell. 2006;127:1109–22. doi: 10.1016/j.cell.2006.11.013. [DOI] [PubMed] [Google Scholar]
- Lamming DW, Wood JG, Sinclair DA. Small molecules that regulate lifespan: evidence for xenohormesis. Mol Microbiol. 2004;53:1003–9. doi: 10.1111/j.1365-2958.2004.04209.x. [DOI] [PubMed] [Google Scholar]
- Landry J, Sutton A, Tafrov ST, Heller RC, Stebbins J, Pillus L, Sternglanz R. The silencing protein SIR2 and its homologs are NAD-dependent protein deacetylases. Proc Natl Acad Sci U S A. 2000;97:5807–11. doi: 10.1073/pnas.110148297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lapointe J, Hekimi S. Early mitochondrial dysfunction in long-lived Mclk1+/− mice. J Biol Chem. 2008;283:26217–27. doi: 10.1074/jbc.M803287200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Fur S, Le Stunff C, Dos Santos C, Bougneres P. The common -866 G/A polymorphism in the promoter of uncoupling protein 2 is associated with increased carbohydrate and decreased lipid oxidation in juvenile obesity. Diabetes. 2004;53:235–9. doi: 10.2337/diabetes.53.1.235. [DOI] [PubMed] [Google Scholar]
- Lebovitz RM, Zhang H, Vogel H, Cartwright J, Jr, Dionne L, Lu N, Huang S, Matzuk MM. Neurodegeneration, myocardial injury, and perinatal death in mitochondrial superoxide dismutase-deficient mice. Proc Natl Acad Sci U S A. 1996;93:9782–7. doi: 10.1073/pnas.93.18.9782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee CM, Weindruch R, Aiken JM. Age-associated alterations of the mitochondrial genome. Free Radic Biol Med. 1997;22:1259–69. doi: 10.1016/s0891-5849(96)00546-1. [DOI] [PubMed] [Google Scholar]
- Lee HC, Li SH, Lin JC, Wu CC, Yeh DC, Wei YH. Somatic mutations in the D-loop and decrease in the copy number of mitochondrial DNA in human hepatocellular carcinoma. Mutat Res. 2004;547:71–8. doi: 10.1016/j.mrfmmm.2003.12.011. [DOI] [PubMed] [Google Scholar]
- Lee HC, Pang CY, Hsu HS, Wei YH. Differential accumulations of 4,977 bp deletion in mitochondrial DNA of various tissues in human ageing. Biochim Biophys Acta. 1994;1226:37–43. doi: 10.1016/0925-4439(94)90056-6. [DOI] [PubMed] [Google Scholar]
- Lentes KU, Tu N, Chen H, Winnikes U, Reinert I, Marmann G, Pirke KM. Genomic organization and mutational analysis of the human UCP2 gene, a prime candidate gene for human obesity. J Recept Signal Transduct Res. 1999;19:229–44. doi: 10.3109/10799899909036648. [DOI] [PubMed] [Google Scholar]
- Lertrit P, Noer AS, Byrne E, Marzuki S. Tissue segregation of a heteroplasmic mtDNA mutation in MERRF (myoclonic epilepsy with ragged red fibers) encephalomyopathy. Hum Genet. 1992;90:251–4. doi: 10.1007/BF00220072. [DOI] [PubMed] [Google Scholar]
- Lescai F, Blanche H, Nebel A, Beekman M, Sahbatou M, Flachsbart F, Slagboom E, Schreiber S, Sorbi S, Passarino G, Franceschi C. Human longevity and 11p15.5: a study in 1321 centenarians. Eur J Hum Genet. 2009 doi: 10.1038/ejhg.2009.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Li HZ, Hu P, Deng J, Banoei MM, Sharma LK, Bai Y. Generation and bioenergetic analysis of cybrids containing mitochondrial DNA from mouse skeletal muscle during aging. Nucleic Acids Res. 38:1913–21. doi: 10.1093/nar/gkp1162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin SJ, Defossez PA, Guarente L. Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae. Science. 2000;289:2126–8. doi: 10.1126/science.289.5487.2126. [DOI] [PubMed] [Google Scholar]
- Linnane AW, Baumer A, Maxwell RJ, Preston H, Zhang CF, Marzuki S. Mitochondrial gene mutation: the ageing process and degenerative diseases. Biochem Int. 1990;22:1067–76. [PubMed] [Google Scholar]
- Linnane AW, Marzuki S, Ozawa T, Tanaka M. Mitochondrial DNA mutations as an important contributor to ageing and degenerative diseases. Lancet. 1989;1:642–5. doi: 10.1016/s0140-6736(89)92145-4. [DOI] [PubMed] [Google Scholar]
- Liu VW, Shi HH, Cheung AN, Chiu PM, Leung TW, Nagley P, Wong LC, Ngan HY. High incidence of somatic mitochondrial DNA mutations in human ovarian carcinomas. Cancer Res. 2001;61:5998–6001. [PubMed] [Google Scholar]
- Liu VW, Zhang C, Nagley P. Mutations in mitochondrial DNA accumulate differentially in three different human tissues during ageing. Nucleic Acids Res. 1998a;26:1268–75. doi: 10.1093/nar/26.5.1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu VW, Zhang C, Pang CY, Lee HC, Lu CY, Wei YH, Nagley P. Independent occurrence of somatic mutations in mitochondrial DNA of human skin from subjects of various ages. Hum Mutat. 1998b;11:191–6. doi: 10.1002/(SICI)1098-1004(1998)11:3<191::AID-HUMU2>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
- Liu X, Jiang N, Hughes B, Bigras E, Shoubridge E, Hekimi S. Evolutionary conservation of the clk-1-dependent mechanism of longevity: loss of mclk1 increases cellular fitness and lifespan in mice. Genes Dev. 2005;19:2424–34. doi: 10.1101/gad.1352905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu CY, Lee HC, Fahn HJ, Wei YH. Oxidative damage elicited by imbalance of free radical scavenging enzymes is associated with large-scale mtDNA deletions in aging human skin. Mutat Res. 1999;423:11–21. doi: 10.1016/s0027-5107(98)00220-6. [DOI] [PubMed] [Google Scholar]
- Lunetta KL, D'Agostino RB, Sr, Karasik D, Benjamin EJ, Guo CY, Govindaraju R, Kiel DP, Kelly-Hayes M, Massaro JM, Pencina MJ, Seshadri S, Murabito JM. Genetic correlates of longevity and selected age-related phenotypes: a genome-wide association study in the Framingham Study. BMC Med Genet. 2007;8(Suppl 1):S13. doi: 10.1186/1471-2350-8-S1-S13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maitra A, Cohen Y, Gillespie SE, Mambo E, Fukushima N, Hoque MO, Shah N, Goggins M, Califano J, Sidransky D, Chakravarti A. The Human MitoChip: a high-throughput sequencing microarray for mitochondrial mutation detection. Genome Res. 2004;14:812–9. doi: 10.1101/gr.2228504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Majamaa-Voltti KA, Winqvist S, Remes AM, Tolonen U, Pyhtinen J, Uimonen S, Karppa M, Sorri M, Peuhkurinen K, Majamaa K. A 3-year clinical follow-up of adult patients with 3243A>G in mitochondrial DNA. Neurology. 2006;66:1470–5. doi: 10.1212/01.wnl.0000216136.61640.79. [DOI] [PubMed] [Google Scholar]
- Manini TM, Everhart JE, Patel KV, Schoeller DA, Colbert LH, Visser M, Tylavsky F, Bauer DC, Goodpaster BH, Harris TB. Daily activity energy expenditure and mortality among older adults. Jama. 2006;296:171–9. doi: 10.1001/jama.296.2.171. [DOI] [PubMed] [Google Scholar]
- Mann VM, Cooper JM, Schapira AH. Quantitation of a mitochondrial DNA deletion in Parkinson's disease. FEBS Lett. 1992;299:218–22. doi: 10.1016/0014-5793(92)80118-z. [DOI] [PubMed] [Google Scholar]
- Marcuello A, Martinez-Redondo D, Dahmani Y, Casajus JA, Ruiz-Pesini E, Montoya J, Lopez-Perez MJ, Diez-Sanchez C. Human mitochondrial variants influence on oxygen consumption. Mitochondrion. 2009;9:27–30. doi: 10.1016/j.mito.2008.10.002. [DOI] [PubMed] [Google Scholar]
- Marnett LJ, Riggins JN, West JD. Endogenous generation of reactive oxidants and electrophiles and their reactions with DNA and protein. J Clin Invest. 2003;111:583–93. doi: 10.1172/JCI18022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martinez-Redondo D, Marcuello A, Casajus JA, Ara I, Dahmani Y, Montoya J, Ruiz-Pesini E, Lopez-Perez MJ, Diez-Sanchez C. Human mitochondrial haplogroup H: the highest VO2max consumer--is it a paradox? Mitochondrion. 10:102–7. doi: 10.1016/j.mito.2009.11.005. [DOI] [PubMed] [Google Scholar]
- Martinez M, Hernandez AI, Martinez N, Ferrandiz ML. Age-related increase in oxidized proteins in mouse synaptic mitochondria. Brain Res. 1996;731:246–8. doi: 10.1016/0006-8993(96)00708-1. [DOI] [PubMed] [Google Scholar]
- McBurney MW, Yang X, Jardine K, Bieman M, Th'ng J, Lemieux M. The absence of SIR2alpha protein has no effect on global gene silencing in mouse embryonic stem cells. Mol Cancer Res. 2003;1:402–9. [PubMed] [Google Scholar]
- McCarroll SA, Murphy CT, Zou S, Pletcher SD, Chin CS, Jan YN, Kenyon C, Bargmann CI, Li H. Comparing genomic expression patterns across species identifies shared transcriptional profile in aging. Nat Genet. 2004;36:197–204. doi: 10.1038/ng1291. [DOI] [PubMed] [Google Scholar]
- McDonald SA, Greaves LC, Gutierrez-Gonzalez L, Rodriguez-Justo M, Deheragoda M, Leedham SJ, Taylor RW, Lee CY, Preston SL, Lovell M, Hunt T, Elia G, Oukrif D, Harrison R, Novelli MR, Mitchell I, Stoker DL, Turnbull DM, Jankowski JA, Wright NA. Mechanisms of field cancerization in the human stomach: the expansion and spread of mutated gastric stem cells. Gastroenterology. 2008;134:500–10. doi: 10.1053/j.gastro.2007.11.035. [DOI] [PubMed] [Google Scholar]
- McKenzie M, Chiotis M, Pinkert CA, Trounce IA. Functional respiratory chain analyses in murid xenomitochondrial cybrids expose coevolutionary constraints of cytochrome b and nuclear subunits of complex III. Mol Biol Evol. 2003;20:1117–24. doi: 10.1093/molbev/msg132. [DOI] [PubMed] [Google Scholar]
- Melov S, Coskun P, Patel M, Tuinstra R, Cottrell B, Jun AS, Zastawny TH, Dizdaroglu M, Goodman SI, Huang TT, Miziorko H, Epstein CJ, Wallace DC. Mitochondrial disease in superoxide dismutase 2 mutant mice. Proc Natl Acad Sci U S A. 1999a;96:846–51. doi: 10.1073/pnas.96.3.846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melov S, Schneider JA, Coskun PE, Bennett DA, Wallace DC. Mitochondrial DNA rearrangements in aging human brain and in situ PCR of mtDNA. Neurobiol Aging. 1999b;20:565–71. doi: 10.1016/s0197-4580(99)00092-5. [DOI] [PubMed] [Google Scholar]
- Melov S, Shoffner JM, Kaufman A, Wallace DC. Marked increase in the number and variety of mitochondrial DNA rearrangements in aging human skeletal muscle. Nucleic Acids Res. 1995;23:4122–6. doi: 10.1093/nar/23.20.4122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Merriwether DA, Clark AG, Ballinger SW, Schurr TG, Soodyall H, Jenkins T, Sherry ST, Wallace DC. The structure of human mitochondrial DNA variation. J Mol Evol. 1991;33:543–55. doi: 10.1007/BF02102807. [DOI] [PubMed] [Google Scholar]
- Michikawa Y, Mazzucchelli F, Bresolin N, Scarlato G, Attardi G. Aging-dependent large accumulation of point mutations in the human mtDNA control region for replication. Science. 1999;286:774–9. doi: 10.1126/science.286.5440.774. [DOI] [PubMed] [Google Scholar]
- Michishita E, Park JY, Burneskis JM, Barrett JC, Horikawa I. Evolutionarily Conserved and Nonconserved Cellular Localizations and Functions of Human SIRT Proteins. Mol Biol Cell. 2005 doi: 10.1091/mbc.E05-01-0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mishmar D, Ruiz-Pesini E, Golik P, Macaulay V, Clark AG, Hosseini S, Brandon M, Easley K, Chen E, Brown MD, Sukernik RI, Olckers A, Wallace DC. Natural selection shaped regional mtDNA variation in humans. Proc Natl Acad Sci U S A. 2003;100:171–6. doi: 10.1073/pnas.0136972100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mishra GR, Suresh M, Kumaran K, Kannabiran N, Suresh S, Bala P, Shivakumar K, Anuradha N, Reddy R, Raghavan TM, Menon S, Hanumanthu G, Gupta M, Upendran S, Gupta S, Mahesh M, Jacob B, Mathew P, Chatterjee P, Arun KS, Sharma S, Chandrika KN, Deshpande N, Palvankar K, Raghavnath R, Krishnakanth R, Karathia H, Rekha B, Nayak R, Vishnupriya G, Kumar HG, Nagini M, Kumar GS, Jose R, Deepthi P, Mohan SS, Gandhi TK, Harsha HC, Deshpande KS, Sarker M, Prasad TS, Pandey A. Human protein reference database--2006 update. Nucleic Acids Res. 2006;34:D411–4. doi: 10.1093/nar/gkj141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mithani SK, Taube JM, Zhou S, Smith IM, Koch WM, Westra WH, Califano JA. Mitochondrial mutations are a late event in the progression of head and neck squamous cell cancer. Clin Cancer Res. 2007;13:4331–5. doi: 10.1158/1078-0432.CCR-06-2613. [DOI] [PubMed] [Google Scholar]
- Moilanen JS, Finnila S, Majamaa K. Lineage-specific selection in human mtDNA: lack of polymorphisms in a segment of MTND5 gene in haplogroup J. Mol Biol Evol. 2003;20:2132–42. doi: 10.1093/molbev/msg230. [DOI] [PubMed] [Google Scholar]
- Moilanen JS, Majamaa K. Phylogenetic network and physicochemical properties of nonsynonymous mutations in the protein-coding genes of human mitochondrial DNA. Mol Biol Evol. 2003;20:1195–210. doi: 10.1093/molbev/msg121. [DOI] [PubMed] [Google Scholar]
- Montiel-Sosa F, Ruiz-Pesini E, Enriquez JA, Marcuello A, Diez-Sanchez C, Montoya J, Wallace DC, Lopez-Perez MJ. Differences of sperm motility in mitochondrial DNA haplogroup U sublineages. Gene. 2006;368:21–7. doi: 10.1016/j.gene.2005.09.015. [DOI] [PubMed] [Google Scholar]
- Moreno-Loshuertos R, Acin-Perez R, Fernandez-Silva P, Movilla N, Perez-Martos A, Rodriguez de Cordoba S, Gallardo ME, Enriquez JA. Differences in reactive oxygen species production explain the phenotypes associated with common mouse mitochondrial DNA variants. Nat Genet. 2006;38:1261–8. doi: 10.1038/ng1897. [DOI] [PubMed] [Google Scholar]
- Munscher C, Muller-Hocker J, Kadenbach B. Human aging is associated with various point mutations in tRNA genes of mitochondrial DNA. Biol Chem Hoppe Seyler. 1993a;374:1099–104. doi: 10.1515/bchm3.1993.374.7-12.1099. [DOI] [PubMed] [Google Scholar]
- Munscher C, Rieger T, Muller-Hocker J, Kadenbach B. The point mutation of mitochondrial DNA characteristic for MERRF disease is found also in healthy people of different ages. FEBS Lett. 1993b;317:27–30. doi: 10.1016/0014-5793(93)81484-h. [DOI] [PubMed] [Google Scholar]
- Murdock DG, Christacos NC, Wallace DC. The age-related accumulation of a mitochondrial DNA control region mutation in muscle, but not brain, detected by a sensitive PNA-directed PCR clamping based method. Nucleic Acids Res. 2000;28:4350–5. doi: 10.1093/nar/28.21.4350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nachman MW. Deleterious mutations in animal mitochondrial DNA. Genetica. 1998;102–103:61–9. [PubMed] [Google Scholar]
- Nagley P, Mackay IR, Baumer A, Maxwell RJ, Vaillant F, Wang ZX, Zhang C, Linnane AW. Mitochondrial DNA mutation associated with aging and degenerative disease. Ann N Y Acad Sci. 1992;673:92–102. doi: 10.1111/j.1749-6632.1992.tb27440.x. [DOI] [PubMed] [Google Scholar]
- Nakano T, Shinka T, Sei M, Sato Y, Umeno M, Sakamoto K, Nomura I, Nakahori Y. A/G heterozygote of the A-3826G polymorphism in the UCP-1 gene has higher BMI than A/A and G/G homozygote in young Japanese males. J Med Invest. 2006;53:218–22. doi: 10.2152/jmi.53.218. [DOI] [PubMed] [Google Scholar]
- Neckelmann N, Li K, Wade RP, Shuster R, Wallace DC. cDNA sequence of a human skeletal muscle ADP/ATP translocator: lack of a leader peptide, divergence from a fibroblast translocator cDNA, and coevolution with mitochondrial DNA genes. Proc Natl Acad Sci U S A. 1987;84:7580–4. doi: 10.1073/pnas.84.21.7580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nekhaeva E, Bodyak ND, Kraytsberg Y, McGrath SB, Van Orsouw NJ, Pluzhnikov A, Wei JY, Vijg J, Khrapko K. Clonally expanded mtDNA point mutations are abundant in individual cells of human tissues. Proc Natl Acad Sci U S A. 2002;99:5521–6. doi: 10.1073/pnas.072670199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niemi AK, Hervonen A, Hurme M, Karhunen PJ, Jylha M, Majamaa K. Mitochondrial DNA polymorphisms associated with longevity in a Finnish population. Hum Genet. 2003;112:29–33. doi: 10.1007/s00439-002-0843-y. [DOI] [PubMed] [Google Scholar]
- Niemi AK, Majamaa K. Mitochondrial DNA and ACTN3 genotypes in Finnish elite endurance and sprint athletes. Eur J Hum Genet. 2005;13:965–9. doi: 10.1038/sj.ejhg.5201438. [DOI] [PubMed] [Google Scholar]
- Niemi AK, Moilanen JS, Tanaka M, Hervonen A, Hurme M, Lehtimaki T, Arai Y, Hirose N, Majamaa K. A combination of three common inherited mitochondrial DNA polymorphisms promotes longevity in Finnish and Japanese subjects. Eur J Hum Genet. 2005;13:166–70. doi: 10.1038/sj.ejhg.5201308. [DOI] [PubMed] [Google Scholar]
- Nomoto S, Yamashita K, Koshikawa K, Nakao A, Sidransky D. Mitochondrial D-loop mutations as clonal markers in multicentric hepatocellular carcinoma and plasma. Clin Cancer Res. 2002;8:481–7. [PubMed] [Google Scholar]
- Oh HH, Kim KS, Choi SM, Yang HS, Yoon Y. The effects of uncoupling protein-1 genotype on lipoprotein cholesterol level in Korean obese subjects. Metabolism. 2004;53:1054–9. doi: 10.1016/j.metabol.2004.02.014. [DOI] [PubMed] [Google Scholar]
- Ohkubo K, Yamano A, Nagashima M, Mori Y, Anzai K, Akehi Y, Nomiyama R, Asano T, Urae A, Ono J. Mitochondrial gene mutations in the tRNA(Leu(UUR)) region and diabetes: prevalence and clinical phenotypes in Japan. Clin Chem. 2001;47:1641–8. [PubMed] [Google Scholar]
- Onyango P, Celic I, McCaffery JM, Boeke JD, Feinberg AP. SIRT3, a human SIR2 homologue, is an NAD-dependent deacetylase localized to mitochondria. Proc Natl Acad Sci U S A. 2002;99:13653–8. doi: 10.1073/pnas.222538099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oppert JM, Vohl MC, Chagnon M, Dionne FT, Cassard-Doulcier AM, Ricquier D, Perusse L, Bouchard C. DNA polymorphism in the uncoupling protein (UCP) gene and human body fat. Int J Obes Relat Metab Disord. 1994;18:526–31. [PubMed] [Google Scholar]
- Ozawa T. Mechanism of somatic mitochondrial DNA mutations associated with age and diseases. Biochim Biophys Acta. 1995a;1271:177–89. doi: 10.1016/0925-4439(95)00026-z. [DOI] [PubMed] [Google Scholar]
- Ozawa T. Mitochondrial DNA mutations associated with aging and degenerative diseases. Exp Gerontol. 1995b;30:269–90. doi: 10.1016/0531-5565(94)00057-a. [DOI] [PubMed] [Google Scholar]
- Park J, Lee G, Chung J. The PINK1-Parkin pathway is involved in the regulation of mitochondrial remodeling process. Biochem Biophys Res Commun. 2009;378:518–23. doi: 10.1016/j.bbrc.2008.11.086. [DOI] [PubMed] [Google Scholar]
- Parkes TL, Elia AJ, Dickinson D, Hilliker AJ, Phillips JP, Boulianne GL. Extension of Drosophila lifespan by overexpression of human SOD1 in motorneurons. Nat Genet. 1998;19:171–4. doi: 10.1038/534. [DOI] [PubMed] [Google Scholar]
- Parrella P, Xiao Y, Fliss M, Sanchez-Cespedes M, Mazzarelli P, Rinaldi M, Nicol T, Gabrielson E, Cuomo C, Cohen D, Pandit S, Spencer M, Rabitti C, Fazio VM, Sidransky D. Detection of mitochondrial DNA mutations in primary breast cancer and fine-needle aspirates. Cancer Res. 2001;61:7623–6. [PubMed] [Google Scholar]
- Parsons TJ, Muniec DS, Sullivan K, Woodyatt N, Alliston-Greiner R, Wilson MR, Berry DL, Holland KA, Weedn VW, Gill P, Holland MM. A high observed substitution rate in the human mitochondrial DNA control region. Nat Genet. 1997;15:363–8. doi: 10.1038/ng0497-363. [DOI] [PubMed] [Google Scholar]
- Penta JS, Johnson FM, Wachsman JT, Copeland WC. Mitochondrial DNA in human malignancy. Mutat Res. 2001;488:119–33. doi: 10.1016/s1383-5742(01)00053-9. [DOI] [PubMed] [Google Scholar]
- Perls T, Terry D. Genetics of exceptional longevity. Exp Gerontol. 2003;38:725–30. doi: 10.1016/s0531-5565(03)00098-6. [DOI] [PubMed] [Google Scholar]
- Pesce V, Cormio A, Fracasso F, Vecchiet J, Felzani G, Lezza AM, Cantatore P, Gadaleta MN. Age-related mitochondrial genotypic and phenotypic alterations in human skeletal muscle. Free Radic Biol Med. 2001;30:1223–33. doi: 10.1016/s0891-5849(01)00517-2. [DOI] [PubMed] [Google Scholar]
- Petriv OI, Rachubinski RA. Lack of peroxisomal catalase causes a progeric phenotype in Caenorhabditis elegans. J Biol Chem. 2004;279:19996–20001. doi: 10.1074/jbc.M400207200. [DOI] [PubMed] [Google Scholar]
- Petros JA, Baumann AK, Ruiz-Pesini E, Amin MB, Sun CQ, Hall J, Lim S, Issa MM, Flanders WD, Hosseini SH, Marshall FF, Wallace DC. mtDNA mutations increase tumorigenicity in prostate cancer. Proc Natl Acad Sci U S A. 2005;102:719–24. doi: 10.1073/pnas.0408894102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Picard F, Kurtev M, Chung N, Topark-Ngarm A, Senawong T, Machado De Oliveira R, Leid M, McBurney MW, Guarente L. Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-gamma. Nature. 2004;429:771–6. doi: 10.1038/nature02583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piko L, Hougham AJ, Bulpitt KJ. Studies of sequence heterogeneity of mitochondrial DNA from rat and mouse tissues: evidence for an increased frequency of deletions/additions with aging. Mech Ageing Dev. 1988;43:279–93. doi: 10.1016/0047-6374(88)90037-1. [DOI] [PubMed] [Google Scholar]
- Polyak K, Li Y, Zhu H, Lengauer C, Willson JK, Markowitz SD, Trush MA, Kinzler KW, Vogelstein B. Somatic mutations of the mitochondrial genome in human colorectal tumours. Nat Genet. 1998;20:291–3. doi: 10.1038/3108. [DOI] [PubMed] [Google Scholar]
- Poole AC, Thomas RE, Andrews LA, McBride HM, Whitworth AJ, Pallanck LJ. The PINK1/Parkin pathway regulates mitochondrial morphology. Proc Natl Acad Sci U S A. 2008;105:1638–43. doi: 10.1073/pnas.0709336105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poole AC, Thomas RE, Yu S, Vincow ES, Pallanck L. The mitochondrial fusion-promoting factor mitofusin is a substrate of the PINK1/parkin pathway. PLoS One. 2010;5:e10054. doi: 10.1371/journal.pone.0010054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poulton J, Morten K. Noninvasive diagnosis of the MELAS syndrome from blood DNA. Ann Neurol. 1993;34:116. doi: 10.1002/ana.410340124. [DOI] [PubMed] [Google Scholar]
- Rand DM, Clark AG, Kann LM. Sexually antagonistic cytonuclear fitness interactions in Drosophila melanogaster. Genetics. 2001;159:173–87. doi: 10.1093/genetics/159.1.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rand DM, Fry A, Sheldahl L. Nuclear-mitochondrial epistasis and drosophila aging: introgression of Drosophila simulans mtDNA modifies longevity in D. melanogaster nuclear backgrounds. Genetics. 2006;172:329–41. doi: 10.1534/genetics.105.046698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rand DM, Haney RA, Fry AJ. Cytonuclear coevolution: the genomics of cooperation. Trends Ecol Evol. 2004;19:645–53. doi: 10.1016/j.tree.2004.10.003. [DOI] [PubMed] [Google Scholar]
- Rand DM, Kann LM. Mutation and selection at silent and replacement sites in the evolution of animal mitochondrial DNA. Genetica. 1998;102–103:393–407. [PubMed] [Google Scholar]
- Rao G, Xia E, Nadakavukaren MJ, Richardson A. Effect of dietary restriction on the age-dependent changes in the expression of antioxidant enzymes in rat liver. J Nutr. 1990;120:602–9. doi: 10.1093/jn/120.6.602. [DOI] [PubMed] [Google Scholar]
- Rawson PD, Burton RS. Functional coadaptation between cytochrome c and cytochrome c oxidase within allopatric populations of a marine copepod. Proc Natl Acad Sci U S A. 2002;99:12955–8. doi: 10.1073/pnas.202335899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reeve AK, Krishnan KJ, Elson JL, Morris CM, Bender A, Lightowlers RN, Turnbull DM. Nature of mitochondrial DNA deletions in substantia nigra neurons. Am J Hum Genet. 2008;82:228–35. doi: 10.1016/j.ajhg.2007.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ren WH, Li XH, Zhang HG, Deng FM, Liao WQ, Pang Y, Liu YH, Qiu MJ, Zhang GY, Zhang YG. Mitochondrial DNA haplogroups in a Chinese Uygur population and their potential association with longevity. Clin Exp Pharmacol Physiol. 2008;35:1477–81. doi: 10.1111/j.1440-1681.2008.05028.x. [DOI] [PubMed] [Google Scholar]
- Revollo JR, Grimm AA, Imai S. The NAD biosynthesis pathway mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian cells. J Biol Chem. 2004;279:50754–63. doi: 10.1074/jbc.M408388200. [DOI] [PubMed] [Google Scholar]
- Rhode JM, Cruzan MB. Contributions of heterosis and epistasis to hybrid fitness. Am Nat. 2005;166:E124–39. doi: 10.1086/491798. [DOI] [PubMed] [Google Scholar]
- Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P. Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature. 2005;434:113–8. doi: 10.1038/nature03354. [DOI] [PubMed] [Google Scholar]
- Rogina B, Helfand SL. Sir2 mediates longevity in the fly through a pathway related to calorie restriction. Proc Natl Acad Sci U S A. 2004;101:15998–6003. doi: 10.1073/pnas.0404184101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rose G, Dato S, Altomare K, Bellizzi D, Garasto S, Greco V, Passarino G, Feraco E, Mari V, Barbi C, BonaFe M, Franceschi C, Tan Q, Boiko S, Yashin AI, De Benedictis G. Variability of the SIRT3 gene, human silent information regulator Sir2 homologue, and survivorship in the elderly. Exp Gerontol. 2003;38:1065–70. doi: 10.1016/s0531-5565(03)00209-2. [DOI] [PubMed] [Google Scholar]
- Rose G, Passarino G, Carrieri G, Altomare K, Greco V, Bertolini S, Bonafe M, Franceschi C, De Benedictis G. Paradoxes in longevity: sequence analysis of mtDNA haplogroup J in centenarians. Eur J Hum Genet. 2001;9:701–7. doi: 10.1038/sj.ejhg.5200703. [DOI] [PubMed] [Google Scholar]
- Rose G, Passarino G, Scornaienchi V, Romeo G, Dato S, Bellizzi D, Mari V, Feraco E, Maletta R, Bruni A, Franceschi C, De Benedictis G. The mitochondrial DNA control region shows genetically correlated levels of heteroplasmy in leukocytes of centenarians and their offspring. BMC Genomics. 2007;8:293. doi: 10.1186/1471-2164-8-293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosenblum JS, Gilula NB, Lerner RA. On signal sequence polymorphisms and diseases of distribution. Proc Natl Acad Sci U S A. 1996;93:4471–3. doi: 10.1073/pnas.93.9.4471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ross OA, McCormack R, Curran MD, Duguid RA, Barnett YA, Rea IM, Middleton D. Mitochondrial DNA polymorphism: its role in longevity of the Irish population. Exp Gerontol. 2001;36:1161–78. doi: 10.1016/s0531-5565(01)00094-8. [DOI] [PubMed] [Google Scholar]
- Rossignol R, Malgat M, Mazat JP, Letellier T. Threshold effect and tissue specificity. Implication for mitochondrial cytopathies. J Biol Chem. 1999;274:33426–32. doi: 10.1074/jbc.274.47.33426. [DOI] [PubMed] [Google Scholar]
- Rudofsky G, Jr, Schroedter A, Schlotterer A, Voron'ko OE, Schlimme M, Tafel J, Isermann BH, Humpert PM, Morcos M, Bierhaus A, Nawroth PP, Hamann A. Functional polymorphisms of UCP2 and UCP3 are associated with a reduced prevalence of diabetic neuropathy in patients with type 1 diabetes. Diabetes Care. 2006;29:89–94. doi: 10.2337/diacare.29.01.06.dc05-0757. [DOI] [PubMed] [Google Scholar]
- Ruiz-Pesini E, Diez C, Lapena AC, Perez-Martos A, Montoya J, Alvarez E, Arenas J, Lopez-Perez MJ. Correlation of sperm motility with mitochondrial enzymatic activities. Clin Chem. 1998;44:1616–20. [PubMed] [Google Scholar]
- Ruiz-Pesini E, Lapena AC, Diez-Sanchez C, Perez-Martos A, Montoya J, Alvarez E, Diaz M, Urries A, Montoro L, Lopez-Perez MJ, Enriquez JA. Human mtDNA haplogroups associated with high or reduced spermatozoa motility. Am J Hum Genet. 2000;67:682–96. doi: 10.1086/303040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ruiz-Pesini E, Mishmar D, Brandon M, Procaccio V, Wallace DC. Effects of purifying and adaptive selection on regional variation in human mtDNA. Science. 2004;303:223–6. doi: 10.1126/science.1088434. [DOI] [PubMed] [Google Scholar]
- Ruiz-Pesini E, Wallace DC. Evidence for adaptive selection acting on the tRNA and rRNA genes of human mitochondrial DNA. Hum Mutat. 2006;27:1072–81. doi: 10.1002/humu.20378. [DOI] [PubMed] [Google Scholar]
- Sackton TB, Haney RA, Rand DM. Cytonuclear coadaptation in Drosophila: disruption of cytochrome c oxidase activity in backcross genotypes. Evolution. 2003;57:2315–25. doi: 10.1111/j.0014-3820.2003.tb00243.x. [DOI] [PubMed] [Google Scholar]
- Sadun F, De Negri AM, Carelli V, Salomao SR, Berezovsky A, Andrade R, Moraes M, Passos A, Belfort R, da Rosa AB, Quiros P, Sadun AA. Ophthalmologic findings in a large pedigree of 11778/Haplogroup J Leber hereditary optic neuropathy. Am J Ophthalmol. 2004;137:271–7. doi: 10.1016/j.ajo.2003.08.010. [DOI] [PubMed] [Google Scholar]
- Salvioli S, Capri M, Santoro A, Raule N, Sevini F, Lukas S, Lanzarini C, Monti D, Passarino G, Rose G, De Benedictis G, Franceschi C. The impact of mitochondrial DNA on human lifespan: a view from studies on centenarians. Biotechnol J. 2008;3:740–9. doi: 10.1002/biot.200800046. [DOI] [PubMed] [Google Scholar]
- Sanchez-Cespedes M, Parrella P, Nomoto S, Cohen D, Xiao Y, Esteller M, Jeronimo C, Jordan RC, Nicol T, Koch WM, Schoenberg M, Mazzarelli P, Fazio VM, Sidransky D. Identification of a mononucleotide repeat as a major target for mitochondrial DNA alterations in human tumors. Cancer Res. 2001;61:7015–9. [PubMed] [Google Scholar]
- Santoro A, Salvioli S, Raule N, Capri M, Sevini F, Valensin S, Monti D, Bellizzi D, Passarino G, Rose G, De Benedictis G, Franceschi C. Mitochondrial DNA involvement in human longevity. Biochim Biophys Acta. 2006;1757:1388–99. doi: 10.1016/j.bbabio.2006.05.040. [DOI] [PubMed] [Google Scholar]
- Schaffler A, Palitzsch KD, Watzlawek E, Drobnik W, Schwer H, Scholmerich J, Schmitz G. Frequency and significance of the A-->G (-3826) polymorphism in the promoter of the gene for uncoupling protein-1 with regard to metabolic parameters and adipocyte transcription factor binding in a large population-based Caucasian cohort. Eur J Clin Invest. 1999;29:770–9. doi: 10.1046/j.1365-2362.1999.00529.x. [DOI] [PubMed] [Google Scholar]
- Scher MB, Vaquero A, Reinberg D. SirT3 is a nuclear NAD+-dependent histone deacetylase that translocates to the mitochondria upon cellular stress. Genes Dev. 2007;21:920–8. doi: 10.1101/gad.1527307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmidt TR, Wu W, Goodman M, Grossman LI. Evolution of nuclear- and mitochondrial-encoded subunit interaction in cytochrome c oxidase. Mol Biol Evol. 2001;18:563–9. doi: 10.1093/oxfordjournals.molbev.a003836. [DOI] [PubMed] [Google Scholar]
- Schriner SE, Linford NJ, Martin GM, Treuting P, Ogburn CE, Emond M, Coskun PE, Ladiges W, Wolf N, Van Remmen H, Wallace DC, Rabinovitch PS. Extension of murine life span by overexpression of catalase targeted to mitochondria. Science. 2005;308:1909–11. doi: 10.1126/science.1106653. [DOI] [PubMed] [Google Scholar]
- Schwer B, North BJ, Frye RA, Ott M, Verdin E. The human silent information regulator (Sir)2 homologue hSIRT3 is a mitochondrial nicotinamide adenine dinucleotide-dependent deacetylase. J Cell Biol. 2002;158:647–57. doi: 10.1083/jcb.200205057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sciacco M, Bonilla E, Schon EA, DiMauro S, Moraes CT. Distribution of wild-type and common deletion forms of mtDNA in normal and respiration-deficient muscle fibers from patients with mitochondrial myopathy. Hum Mol Genet. 1994;3:13–9. doi: 10.1093/hmg/3.1.13. [DOI] [PubMed] [Google Scholar]
- Shankar SP, Fingert JH, Carelli V, Valentino ML, King TM, Daiger SP, Salomao SR, Berezovsky A, Belfort R, Jr, Braun TA, Sheffield VC, Sadun AA, Stone EM. Evidence for a novel x-linked modifier locus for leber hereditary optic neuropathy. Ophthalmic Genet. 2008;29:17–24. doi: 10.1080/13816810701867607. [DOI] [PubMed] [Google Scholar]
- Shenkar R, Navidi W, Tavare S, Dang MH, Chomyn A, Attardi G, Cortopassi G, Arnheim N. The mutation rate of the human mtDNA deletion mtDNA4977. Am J Hum Genet. 1996;59:772–80. [PMC free article] [PubMed] [Google Scholar]
- Shi T, Wang F, Stieren E, Tong Q. SIRT3, a mitochondrial sirtuin deacetylase, regulates mitochondrial function and thermogenesis in brown adipocytes. J Biol Chem. 2005;280:13560–7. doi: 10.1074/jbc.M414670200. [DOI] [PubMed] [Google Scholar]
- Shimoda-Matsubayashi S, Matsumine H, Kobayashi T, Nakagawa-Hattori Y, Shimizu Y, Mizuno Y. Structural dimorphism in the mitochondrial targeting sequence in the human manganese superoxide dismutase gene. A predictive evidence for conformational change to influence mitochondrial transport and a study of allelic association in Parkinson's disease. Biochem Biophys Res Commun. 1996;226:561–5. doi: 10.1006/bbrc.1996.1394. [DOI] [PubMed] [Google Scholar]
- Shin HD, Kim KS, Cha MH, Yoon Y. The effects of UCP-1 polymorphisms on obesity phenotypes among Korean female subjects. Biochem Biophys Res Commun. 2005;335:624–30. doi: 10.1016/j.bbrc.2005.07.096. [DOI] [PubMed] [Google Scholar]
- Short KR, Bigelow ML, Kahl J, Singh R, Coenen-Schimke J, Raghavakaimal S, Nair KS. Decline in skeletal muscle mitochondrial function with aging in humans. Proc Natl Acad Sci U S A. 2005;102:5618–23. doi: 10.1073/pnas.0501559102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shuster RC, Rubenstein AJ, Wallace DC. Mitochondrial DNA in anucleate human blood cells. Biochem Biophys Res Commun. 1988;155:1360–5. doi: 10.1016/s0006-291x(88)81291-9. [DOI] [PubMed] [Google Scholar]
- Simonetti S, Chen X, DiMauro S, Schon EA. Accumulation of deletions in human mitochondrial DNA during normal aging: analysis by quantitative PCR. Biochim Biophys Acta. 1992;1180:113–22. doi: 10.1016/0925-4439(92)90059-v. [DOI] [PubMed] [Google Scholar]
- Sohal RS, Sohal BH, Orr WC. Mitochondrial superoxide and hydrogen peroxide generation, protein oxidative damage, and longevity in different species of flies. Free Radic Biol Med. 1995;19:499–504. doi: 10.1016/0891-5849(95)00037-x. [DOI] [PubMed] [Google Scholar]
- Sohal RS, Toroser D, Bregere C, Mockett RJ, Orr WC. Age-related decrease in expression of mitochondrial DNA encoded subunits of cytochrome c oxidase in Drosophila melanogaster. Mech Ageing Dev. 2008;129:558–61. doi: 10.1016/j.mad.2008.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sohal RS, Weindruch R. Oxidative stress, caloric restriction, and aging. Science. 1996;273:59–63. doi: 10.1126/science.273.5271.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song Z, Chen H, Fiket M, Alexander C, Chan DC. OPA1 processing controls mitochondrial fusion and is regulated by mRNA splicing, membrane potential, and Yme1L. J Cell Biol. 2007;178:749–55. doi: 10.1083/jcb.200704110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soong NW, Hinton DR, Cortopassi G, Arnheim N. Mosaicism for a specific somatic mitochondrial DNA mutation in adult human brain. Nat Genet. 1992;2:318–23. doi: 10.1038/ng1292-318. [DOI] [PubMed] [Google Scholar]
- Sugiyama S, Hattori K, Hayakawa M, Ozawa T. Quantitative analysis of age-associated accumulation of mitochondrial DNA with deletion in human hearts. Biochem Biophys Res Commun. 1991;180:894–9. doi: 10.1016/s0006-291x(05)81149-0. [DOI] [PubMed] [Google Scholar]
- Sui G, Zhou S, Wang J, Canto M, Lee EE, Eshleman JR, Montgomery EA, Sidransky D, Califano JA, Maitra A. Mitochondrial DNA mutations in preneoplastic lesions of the gastrointestinal tract: a biomarker for the early detection of cancer. Mol Cancer. 2006;5:73. doi: 10.1186/1476-4598-5-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan Q, Zhao JH, Zhang D, Kruse TA, Christensen K. Power for genetic association study of human longevity using the case-control design. Am J Epidemiol. 2008;168:890–6. doi: 10.1093/aje/kwn205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanaka M, Gong JS, Zhang J, Yoneda M, Yagi K. Mitochondrial genotype associated with longevity. Lancet. 1998;351:185–6. doi: 10.1016/S0140-6736(05)78211-8. [DOI] [PubMed] [Google Scholar]
- Tanaka M, Takeyasu T, Fuku N, Li-Jun G, Kurata M. Mitochondrial genome single nucleotide polymorphisms and their phenotypes in the Japanese. Ann N Y Acad Sci. 2004;1011:7–20. doi: 10.1007/978-3-662-41088-2_2. [DOI] [PubMed] [Google Scholar]
- Tang Z, Wang X, Hu Z, Yang Z, Xu C. Genetic dissection of cytonuclear epistasis in line crosses. Genetics. 2007;177:669–72. doi: 10.1534/genetics.107.074302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taylor RW, Barron MJ, Borthwick GM, Gospel A, Chinnery PF, Samuels DC, Taylor GA, Plusa SM, Needham SJ, Greaves LC, Kirkwood TB, Turnbull DM. Mitochondrial DNA mutations in human colonic crypt stem cells. J Clin Invest. 2003;112:1351–60. doi: 10.1172/JCI19435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terlecky SR, Koepke JI, Walton PA. Peroxisomes and aging. Biochim Biophys Acta. 2006;1763:1749–54. doi: 10.1016/j.bbamcr.2006.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tissenbaum HA, Guarente L. Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans. Nature. 2001;410:227–30. doi: 10.1038/35065638. [DOI] [PubMed] [Google Scholar]
- Tong BC, Ha PK, Dhir K, Xing M, Westra WH, Sidransky D, Califano JA. Mitochondrial DNA alterations in thyroid cancer. J Surg Oncol. 2003;82:170–3. doi: 10.1002/jso.10202. [DOI] [PubMed] [Google Scholar]
- Trifunovic A, Hansson A, Wredenberg A, Rovio AT, Dufour E, Khvorostov I, Spelbrink JN, Wibom R, Jacobs HT, Larsson NG. Somatic mtDNA mutations cause aging phenotypes without affecting reactive oxygen species production. Proc Natl Acad Sci U S A. 2005;102:17993–8. doi: 10.1073/pnas.0508886102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trounce I, Byrne E, Marzuki S. Decline in skeletal muscle mitochondrial respiratory chain function: possible factor in ageing. Lancet. 1989;1:637–9. doi: 10.1016/s0140-6736(89)92143-0. [DOI] [PubMed] [Google Scholar]
- Urhammer SA, Dalgaard LT, Sorensen TI, Tybjaerg-Hansen A, Echwald SM, Andersen T, Clausen JO, Pedersen O. Organisation of the coding exons and mutational screening of the uncoupling protein 3 gene in subjects with juvenile-onset obesity. Diabetologia. 1998;41:241–4. doi: 10.1007/s001250050897. [DOI] [PubMed] [Google Scholar]
- van Abeelen AF, de Krom M, Hendriks J, Grobbee DE, Adan RA, van der Schouw YT. Variations in the uncoupling protein-3 gene are associated with specific obesity phenotypes. Eur J Endocrinol. 2008;158:669–76. doi: 10.1530/EJE-07-0834. [DOI] [PubMed] [Google Scholar]
- Vives-Bauza C, de Vries RL, Tocilescu M, Przedborski S. PINK1/Parkin direct mitochondria to autophagy. Autophagy. 2010;6:315–6. doi: 10.4161/auto.6.2.11199. [DOI] [PubMed] [Google Scholar]
- von Wurmb-Schwark N, Schwark T, Meissner C, Oehmichen M. Mitochondrial mutagenesis in the brain in forensic and pathological research. Leg Med (Tokyo) 2003;5:1–6. doi: 10.1016/s1344-6223(03)00003-8. [DOI] [PubMed] [Google Scholar]
- von Wurmb N, Oehmichen M, Meissner C. Demonstration of the 4977 bp deletion in human mitochondrial DNA from intravital and postmortem blood. Mutat Res. 1998;422:247–54. doi: 10.1016/s0027-5107(98)00205-x. [DOI] [PubMed] [Google Scholar]
- Wade MJ, Goodnight CJ. Cyto-nuclear epistasis: two-locus random genetic drift in hermaphroditic and dioecious species. Evolution. 2006;60:643–59. [PubMed] [Google Scholar]
- Walker DW, Benzer S. Mitochondrial "swirls" induced by oxygen stress and in the Drosophila mutant hyperswirl. Proc Natl Acad Sci U S A. 2004;101:10290–5. doi: 10.1073/pnas.0403767101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wallace DC. Mitochondrial DNA mutations in diseases of energy metabolism. J Bioenerg Biomembr. 1994;26:241–50. doi: 10.1007/BF00763096. [DOI] [PubMed] [Google Scholar]
- Wallace DC. 1994 William Allan Award Address. Mitochondrial DNA variation in human evolution, degenerative disease, and aging. Am J Hum Genet. 1995;57:201–23. [PMC free article] [PubMed] [Google Scholar]
- Wallace DC. A mitochondrial paradigm for degenerative diseases and ageing. Novartis Found Symp. 2001;235:247–63. doi: 10.1002/0470868694.ch20. discussion 263–6. [DOI] [PubMed] [Google Scholar]
- Wallace DC. A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev Genet. 2005;39:359–407. doi: 10.1146/annurev.genet.39.110304.095751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wallace DC, Lott MT, Shoffner JM, Brown MD. Diseases resulting from mitochondrial DNA point mutations. J Inherit Metab Dis. 1992;15:472–9. doi: 10.1007/BF01799605. [DOI] [PubMed] [Google Scholar]
- Wallace DC, Ruiz-Pesini E, Mishmar D. mtDNA variation, climatic adaptation, degenerative diseases, and longevity. Cold Spring Harb Symp Quant Biol. 2003;68:479–86. doi: 10.1101/sqb.2003.68.471. [DOI] [PubMed] [Google Scholar]
- Wallace DC, Shoffner JM, Trounce I, Brown MD, Ballinger SW, Corral-Debrinski M, Horton T, Jun AS, Lott MT. Mitochondrial DNA mutations in human degenerative diseases and aging. Biochim Biophys Acta. 1995;1271:141–51. doi: 10.1016/0925-4439(95)00021-u. [DOI] [PubMed] [Google Scholar]
- Wallace DC, Stugard C, Murdock D, Schurr T, Brown MD. Ancient mtDNA sequences in the human nuclear genome: a potential source of errors in identifying pathogenic mutations. Proc Natl Acad Sci U S A. 1997;94:14900–5. doi: 10.1073/pnas.94.26.14900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y, Michikawa Y, Mallidis C, Bai Y, Woodhouse L, Yarasheski KE, Miller CA, Askanas V, Engel WK, Bhasin S, Attardi G. Muscle-specific mutations accumulate with aging in critical human mtDNA control sites for replication. Proc Natl Acad Sci U S A. 2001;98:4022–7. doi: 10.1073/pnas.061013598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weber K, Wilson JN, Taylor L, Brierley E, Johnson MA, Turnbull DM, Bindoff LA. A new mtDNA mutation showing accumulation with time and restriction to skeletal muscle. Am J Hum Genet. 1997;60:373–80. [PMC free article] [PubMed] [Google Scholar]
- Wei YH. Mitochondrial DNA alterations as ageing-associated molecular events. Mutat Res. 1992;275:145–55. doi: 10.1016/0921-8734(92)90019-l. [DOI] [PubMed] [Google Scholar]
- Wei YH. Mitochondrial DNA mutations and oxidative damage in aging and diseases: an emerging paradigm of gerontology and medicine. Proc Natl Sci Counc Repub China B. 1998a;22:55–67. [PubMed] [Google Scholar]
- Wei YH. Oxidative stress and mitochondrial DNA mutations in human aging. Proc Soc Exp Biol Med. 1998b;217:53–63. doi: 10.3181/00379727-217-44205. [DOI] [PubMed] [Google Scholar]
- Weinreich DM, Rand DM. Contrasting patterns of nonneutral evolution in proteins encoded in nuclear and mitochondrial genomes. Genetics. 2000;156:385–99. doi: 10.1093/genetics/156.1.385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whitworth AJ, Pallanck LJ. The PINK1/Parkin pathway: a mitochondrial quality control system? J Bioenerg Biomembr. 2009;41:499–503. doi: 10.1007/s10863-009-9253-3. [DOI] [PubMed] [Google Scholar]
- Willett CS, Burton RS. ENVIRONMENTAL INFLUENCES ON EPISTATIC INTERACTIONS: VIABILITIES OF CYTOCHROME C GENOTYPES IN INTERPOPULATION CROSSES. Evolution. 2003;57:2286–2292. doi: 10.1111/j.0014-3820.2003.tb00240.x. [DOI] [PubMed] [Google Scholar]
- Wood JG, Rogina B, Lavu S, Howitz K, Helfand SL, Tatar M, Sinclair D. Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature. 2004;430:686–9. doi: 10.1038/nature02789. [DOI] [PubMed] [Google Scholar]
- Wu CW, Yin PH, Hung WY, Li AF, Li SH, Chi CW, Wei YH, Lee HC. Mitochondrial DNA mutations and mitochondrial DNA depletion in gastric cancer. Genes Chromosomes Cancer. 2005;44:19–28. doi: 10.1002/gcc.20213. [DOI] [PubMed] [Google Scholar]
- Yamasaki H, Sasaki H, Ogawa K, Shono T, Tamura S, Doi A, Sasahara M, Kawashima H, Nakao T, Furuta H, Nishi M, Nanjo K. Uncoupling protein 2 promoter polymorphism -866G/A affects peripheral nerve dysfunction in Japanese type 2 diabetic patients. Diabetes Care. 2006;29:888–94. doi: 10.2337/diacare.29.04.06.dc05-1984. [DOI] [PubMed] [Google Scholar]
- Yang JH, Lee HC, Lin KJ, Wei YH. A specific 4977-bp deletion of mitochondrial DNA in human ageing skin. Arch Dermatol Res. 1994;286:386–90. doi: 10.1007/BF00371798. [DOI] [PubMed] [Google Scholar]
- Yasuno K, Ando S, Misumi S, Makino S, Kulski JK, Muratake T, Kaneko N, Amagane H, Someya T, Inoko H, Suga H, Kanemoto K, Tamiya G. Synergistic association of mitochondrial uncoupling protein (UCP) genes with schizophrenia. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:250–3. doi: 10.1002/ajmg.b.30443. [DOI] [PubMed] [Google Scholar]
- Yeh JJ, Lunetta KL, van Orsouw NJ, Moore FD, Jr, Mutter GL, Vijg J, Dahia PL, Eng C. Somatic mitochondrial DNA (mtDNA) mutations in papillary thyroid carcinomas and differential mtDNA sequence variants in cases with thyroid tumours. Oncogene. 2000;19:2060–6. doi: 10.1038/sj.onc.1203537. [DOI] [PubMed] [Google Scholar]
- Yen TC, Chen YS, King KL, Yeh SH, Wei YH. Liver mitochondrial respiratory functions decline with age. Biochem Biophys Res Commun. 1989;165:944–1003. doi: 10.1016/0006-291x(89)92701-0. [DOI] [PubMed] [Google Scholar]
- Yen TC, King KL, Lee HC, Yeh SH, Wei YH. Age-dependent increase of mitochondrial DNA deletions together with lipid peroxides and superoxide dismutase in human liver mitochondria. Free Radic Biol Med. 1994;16:207–14. doi: 10.1016/0891-5849(94)90145-7. [DOI] [PubMed] [Google Scholar]
- Yen TC, Pang CY, Hsieh RH, Su CH, King KL, Wei YH. Age-dependent 6kb deletion in human liver mitochondrial DNA. Biochem Int. 1992;26:457–68. [PubMed] [Google Scholar]
- Yen TC, Su JH, King KL, Wei YH. Ageing-associated 5 kb deletion in human liver mitochondrial DNA. Biochem Biophys Res Commun. 1991;178:124–31. doi: 10.1016/0006-291x(91)91788-e. [DOI] [PubMed] [Google Scholar]
- Yoon Y, Park BL, Cha MH, Kim KS, Cheong HS, Choi YH, Shin HD. Effects of genetic polymorphisms of UCP2 and UCP3 on very low calorie diet-induced body fat reduction in Korean female subjects. Biochem Biophys Res Commun. 2007;359:451–6. doi: 10.1016/j.bbrc.2007.05.110. [DOI] [PubMed] [Google Scholar]
- Zhang C, Baumer A, Maxwell RJ, Linnane AW, Nagley P. Multiple mitochondrial DNA deletions in an elderly human individual. FEBS Lett. 1992;297:34–8. doi: 10.1016/0014-5793(92)80321-7. [DOI] [PubMed] [Google Scholar]
- Zhang C, Lee A, Liu VW, Pepe S, Rosenfeldt F, Nagley P. Mitochondrial DNA deletions in human cardiac tissue show a gross mosaic distribution. Biochem Biophys Res Commun. 1999;254:152–7. doi: 10.1006/bbrc.1998.9914. [DOI] [PubMed] [Google Scholar]
- Zhang C, Linnane AW, Nagley P. Occurrence of a particular base substitution (3243 A to G) in mitochondrial DNA of tissues of ageing humans. Biochem Biophys Res Commun. 1993;195:1104–10. doi: 10.1006/bbrc.1993.2158. [DOI] [PubMed] [Google Scholar]
- Zhang C, Liu VW, Addessi CL, Sheffield DA, Linnane AW, Nagley P. Differential occurrence of mutations in mitochondrial DNA of human skeletal muscle during aging. Hum Mutat. 1998;11:360–71. doi: 10.1002/(SICI)1098-1004(1998)11:5<360::AID-HUMU3>3.0.CO;2-U. [DOI] [PubMed] [Google Scholar]
- Zhang J, Asin-Cayuela J, Fish J, Michikawa Y, Bonafe M, Olivieri F, Passarino G, De Benedictis G, Franceschi C, Attardi G. Strikingly higher frequency in centenarians and twins of mtDNA mutation causing remodeling of replication origin in leukocytes. Proc Natl Acad Sci U S A. 2003;100:1116–21. doi: 10.1073/pnas.242719399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J, Montine TJ, Smith MA, Siedlak SL, Gu G, Robertson D, Perry G. The mitochondrial common deletion in Parkinson's disease and related movement disorders. Parkinsonism Relat Disord. 2002;8:165–70. doi: 10.1016/s1353-8020(01)00041-4. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Chan DC. Structural basis for recruitment of mitochondrial fission complexes by Fis1. Proc Natl Acad Sci U S A. 2007;104:18526–30. doi: 10.1073/pnas.0706441104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou S, Kachhap S, Sun W, Wu G, Chuang A, Poeta L, Grumbine L, Mithani SK, Chatterjee A, Koch W, Westra WH, Maitra A, Glazer C, Carducci M, Sidransky D, McFate T, Verma A, Califano JA. Frequency and phenotypic implications of mitochondrial DNA mutations in human squamous cell cancers of the head and neck. Proc Natl Acad Sci U S A. 2007 doi: 10.1073/pnas.0610818104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou S, Kassauei K, Cutler DJ, Kennedy GC, Sidransky D, Maitra A, Califano J. An oligonucleotide microarray for high-throughput sequencing of the mitochondrial genome. J Mol Diagn. 2006;8:476–82. doi: 10.2353/jmoldx.2006.060008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zietz B, Watzlawek E, Palitzsch KD, Scholmerich J, Schaffler A. GG-genotype in the promotor region of uncoupling-protein-1 gene is associated with lower level of dehydroepiandrosterone in type 2 diabetes. Exp Clin Endocrinol Diabetes. 2001;109:102–6. doi: 10.1055/s-2001-14829. [DOI] [PubMed] [Google Scholar]
- Ziviani E, Tao RN, Whitworth AJ. Drosophila parkin requires PINK1 for mitochondrial translocation and ubiquitinates mitofusin. Proc Natl Acad Sci U S A. 2010;107:5018–23. doi: 10.1073/pnas.0913485107. [DOI] [PMC free article] [PubMed] [Google Scholar]