mitochondrial dna mutations in human disease

9
Critical Review Mitochondrial DNA Mutations in Human Disease Laura C. Greaves and Robert W. Taylor Mitochondrial Research Group, School of Neurology, Neurobiology and Psychiatry, University of Newcastle upon Tyne, UK Summary Since their first association with human disease in 1988, more than 250 pathogenic point mutations and rearrangements of the 16.6 kb mitochondrial genome (mtDNA) have been reported in a spectrum of clinical disorders which exhibit prominent muscle and central nervous system involvement. With novel mutations and disease phenotypes still being described, mtDNA disorders are recognized collectively as common, inherited genetic diseases although relatively little is still known concerning the precise pathophysiological mechanisms that lead to cell dysfunction and pathology. This review considers the basic principles of mitochondrial genetics which govern both the behaviour and investigation of pathogenic mtDNA mutations summarizing recent advances in this area, and an assessment of the ongoing debate into the role of somatic mtDNA mutations in neurodegenerative disease, ageing and cancer. IUBMB Life, 58: 143 – 151, 2006 Keywords mtDNA disorders; mitochondrial genetics; neurodegen- erative disease; ageing; cancer. INTRODUCTION Mitochondria are ubiquitous organelles found in all nucleated cells and are the major generators of cellular ATP by oxidative phosphorylation (OXPHOS). Electrons gener- ated by oxidation of fat and carbohydrates are transferred to oxygen via the redox components of the respiratory chain (complexes I – IV), forming water. Protons are pumped across the inner membrane from the matrix to the intermembrane space forming an electrochemical gradient which is used by the ATP synthase (complex V) to synthesize ATP. Thirteen of the 90 or so protein subunits that make up the OXPHOS complexes are encoded by the mitochondrial genome (mtDNA), making this organelle the only location of extrachromosomal DNA within mammalian cells. Human mtDNA is a multicopy, circular double-stranded DNA molecule of 16.6 kb which encodes 37 contiguous genes – 13 polypeptides of the OXPHOS system and the necessary RNA machinery (2 rRNAs and 22 tRNAs) for their translation within the organelle (Fig. 1). MtDNA has no introns, the only non-coding region being the 1.1 kb displacement (D-) loop which contains transcriptional promoters and one of the proposed replication origins, O H . All the remaining respira- tory chain subunits, together with several hundred other proteins including the entire complement of enzymes and factors that comprise the mtDNA replisome and transcription machinery are encoded by nuclear genes. Mitochondrial function is therefore wholly dependent upon the coordination and cooperation of both genomes. Whilst this review will focus exclusively on disease-causing mtDNA mutations, the contribution of mtDNA replication, transcription and translation to mitochondrial disease pathol- ogy cannot be neglected, not only in terms of their role in the expression of specific mtDNA mutations and ensuing bio- chemical consequences, but also as mutations of proteins critical for the maintenance of mtDNA are increasingly recognized as causing specific clinical disorders. Perhaps the best example is that of the POLG1 gene, which encodes the 140-kDa catalytic (DNA polymerase and 3 0 -5 0 exonuclease activities) subunit of mtDNA polymerase g, forming a heterodimeric enzyme together with the 55-kDa accessory subunit, POLG2. The first pathogenic mutations of POLG1 were identified in families with autosomal dominant chronic progressive external ophthalmoplegia (adPEO) and an accu- mulation of multiple mtDNA deletions was demonstrated in affected tissues. Since then, both compound heterozygous and homozygous POLG1 mutations have been described in recessive PEO, adult-onset spinocerebellar ataxia with multi- ple mtDNA deletions and Alpers syndrome due to quantitative mtDNA depletion. POLG1 mutations have also been linked to Parkinsonism and premature ovarian failure (1), whilst mice expressing a mutator (proofreading-deficient) POLG1 enzyme accumulate mtDNA mutations at a higher Received 14 February 2006; accepted 2 March 2006 Address correspondence to: Dr R. W. Taylor, Mitochondrial Research Group, School of Neurology, Neurobiology and Psychiatry, University of Newcastle upon Tyne, Newcastle upon Tyne, NE2 4HH, UK. Tel: þ44 (0)191 2223685. Fax: þ44 (0)191 2228553. E-mail: [email protected] IUBMB Life, 58(3): 143 – 151, March 2006 ISSN 1521-6543 print/ISSN 1521-6551 online Ó 2006 IUBMB DOI: 10.1080/15216540600686888

Upload: luna-smith

Post on 15-Sep-2015

7 views

Category:

Documents


1 download

DESCRIPTION

Mitochondrial DNA Mutations in Human Diseases is an article wicht dissects about some problems that are risen when there are mutations in the DNA.

TRANSCRIPT

  • Critical Review

    Mitochondrial DNA Mutations in Human Disease

    Laura C. Greaves and Robert W. TaylorMitochondrial Research Group, School of Neurology, Neurobiology and Psychiatry,University of Newcastle upon Tyne, UK

    Summary

    Since their rst association with human disease in 1988, more than250 pathogenic point mutations and rearrangements of the 16.6 kbmitochondrial genome (mtDNA) have been reported in a spectrum ofclinical disorders which exhibit prominent muscle and central nervoussystem involvement. With novel mutations and disease phenotypesstill being described, mtDNA disorders are recognized collectively ascommon, inherited genetic diseases although relatively little is stillknown concerning the precise pathophysiological mechanismsthat lead to cell dysfunction and pathology. This review considersthe basic principles of mitochondrial genetics which govern boththe behaviour and investigation of pathogenic mtDNA mutationssummarizing recent advances in this area, and an assessment ofthe ongoing debate into the role of somatic mtDNA mutations inneurodegenerative disease, ageing and cancer.

    IUBMB Life, 58: 143 151, 2006

    Keywords mtDNA disorders; mitochondrial genetics; neurodegen-erative disease; ageing; cancer.

    INTRODUCTION

    Mitochondria are ubiquitous organelles found in all

    nucleated cells and are the major generators of cellular ATP

    by oxidative phosphorylation (OXPHOS). Electrons gener-

    ated by oxidation of fat and carbohydrates are transferred to

    oxygen via the redox components of the respiratory chain

    (complexes I IV), forming water. Protons are pumped across

    the inner membrane from the matrix to the intermembrane

    space forming an electrochemical gradient which is used by the

    ATP synthase (complex V) to synthesize ATP. Thirteen of the

    90 or so protein subunits that make up the OXPHOS

    complexes are encoded by the mitochondrial genome

    (mtDNA), making this organelle the only location of

    extrachromosomal DNA within mammalian cells. Human

    mtDNA is a multicopy, circular double-stranded DNA

    molecule of 16.6 kb which encodes 37 contiguous genes 13

    polypeptides of the OXPHOS system and the necessary RNA

    machinery (2 rRNAs and 22 tRNAs) for their translation

    within the organelle (Fig. 1). MtDNA has no introns, the only

    non-coding region being the 1.1 kb displacement (D-) loop

    which contains transcriptional promoters and one of the

    proposed replication origins, OH. All the remaining respira-

    tory chain subunits, together with several hundred other

    proteins including the entire complement of enzymes and

    factors that comprise the mtDNA replisome and transcription

    machinery are encoded by nuclear genes. Mitochondrial

    function is therefore wholly dependent upon the coordination

    and cooperation of both genomes.

    Whilst this review will focus exclusively on disease-causing

    mtDNA mutations, the contribution of mtDNA replication,

    transcription and translation to mitochondrial disease pathol-

    ogy cannot be neglected, not only in terms of their role in the

    expression of specic mtDNA mutations and ensuing bio-

    chemical consequences, but also as mutations of proteins

    critical for the maintenance of mtDNA are increasingly

    recognized as causing specic clinical disorders. Perhaps the

    best example is that of the POLG1 gene, which encodes the

    140-kDa catalytic (DNA polymerase and 30-50 exonucleaseactivities) subunit of mtDNA polymerase g, forming aheterodimeric enzyme together with the 55-kDa accessory

    subunit, POLG2. The rst pathogenic mutations of POLG1

    were identied in families with autosomal dominant chronic

    progressive external ophthalmoplegia (adPEO) and an accu-

    mulation of multiple mtDNA deletions was demonstrated in

    aected tissues. Since then, both compound heterozygous and

    homozygous POLG1 mutations have been described in

    recessive PEO, adult-onset spinocerebellar ataxia with multi-

    ple mtDNA deletions and Alpers syndrome due to

    quantitative mtDNA depletion. POLG1 mutations have also

    been linked to Parkinsonism and premature ovarian failure

    (1), whilst mice expressing a mutator (proofreading-decient)

    POLG1 enzyme accumulate mtDNA mutations at a higher

    Received 14 February 2006; accepted 2 March 2006Address correspondence to: Dr R. W. Taylor, Mitochondrial

    Research Group, School of Neurology, Neurobiology and Psychiatry,University of Newcastle upon Tyne, Newcastle upon Tyne, NE24HH, UK. Tel: 44 (0)191 2223685. Fax: 44 (0)191 2228553.E-mail: [email protected]

    IUBMBLife, 58(3): 143 151, March 2006

    ISSN 1521-6543 print/ISSN 1521-6551 online 2006 IUBMBDOI: 10.1080/15216540600686888

  • rate than normal and display features of accelerated aging

    (2, 3).

    BASIC MITOCHONDRIAL GENETICS

    Mitochondrial DNA has dierent genetic rules that set it

    apart from Mendelian genetics and which strongly dictate the

    functional consequences of pathogenic mtDNA mutations (4).

    These include heteroplasmy and the threshold eect, a strict

    maternal pattern of inheritance and mitotic segregation.

    The mitochondrial genome is polyploid, with several

    hundreds or thousands of copies present in a single cell which

    distribute randomly to daughter cells during cell division.

    Homoplasmy describes the existence of identical mtDNA

    copies within a cell and heteroplasmy is when there is a

    mixture of two or more mitochondrial genotypes. The

    majority of deleterious mtDNA mutations are heteroplasmic

    and exhibit a threshold level of mutation load which is

    required to cause a biochemical defect, and hence clinical

    expression of the disease. This critical proportion of wild type:

    mutated mtDNA molecules is dierent for specic mutations,

    varies amongst tissues even in an individual and is demon-

    strated for a number of mtDNA mutations by the

    histocytochemical assessment of cytochrome c oxidase

    (complex IV) activity in individual cells. Although there are

    pathogenic mutations that are truly homoplasmic, the concept

    Figure 1.Map of the human mitochondrial genome depicting common genotype:phenotype correlations. Human mitochondrial

    DNA is a 16,569 bp circle of double stranded DNA that encodes 13 essential respiratory chain subunits: ND1-ND6, ND4L are

    seven subunits of complex I (NADH:ubiquione oxidoreductase), CYT b of complex III (ubiqionol:cytochrome c

    oxidoreductase), COX I COX III are the three catalytic subunits of complex IV (cytochrome c oxidase), and the ATPase6

    and ATPase8 subunits of complex V (ATP synthase). Also shown are the two ribosomal RNA (12S rRNA and 16S rRNA) genes

    and the twenty-two transfer RNA genes (depicted by their single letter abbreviation) required for intramitochondrial protein

    synthesis. The only major non-coding regions are the 1.1 kb displacement (D-) loop that contains the origins of heavy- and light-

    strand transcription and heavy-strand replication (shown as OH), and the origin of light-strand replication (shown as OL). In

    addition, the sites of the common mtDNA mutations are indicated, together with their associated clinical presentations. CPEO,

    chronic progressive external ophthalmoplegia; LHON, Leber hereditary optic neuropathy; MELAS, Mitochondrial myopathy,

    encephalopathy, lactic acidosis and stroke-like episodes; MERRF, Myoclonic epilepsy and ragged-red bres; MIDD,

    Maternally-inherited diabetes and deafness; MILS, Maternally-inherited Leigh Syndrome; NARP, Neurogenic weakness, ataxia

    and retinitis pigmentosa.

    144 GREAVES AND TAYLOR

  • of homoplasmy is rather more apparent than real as mtDNA

    is constantly undergoing mutation with the clonal expansion

    or loss of either point mutations or deletions; in some tissues,

    such as the human colon, this occurs at a high frequency (5).

    Since these mutational events occur stochastically, all acquired

    mutations are present at very low levels and will only be

    detectable in single cell experiments and not by the analysis of

    whole tissue homogenates or blood sample.

    The inheritance of the mitochondrial genome, organized

    in vivo as discrete DNA-protein structures termed nucleoids,

    is strictly maternal. Every mtDNA copy present in the

    fertilized egg is derived from the oocyte, with no apparent

    contribution from paternal mtDNA these organelles are

    ubiquitinated and are selectively destroyed in the fertilized

    embryo prior to the breakdown of mitochondrial structures

    (6). There are challenges to this paradigm, with evidence of

    low levels of paternal transmission of mtDNA in crosses

    between, but not within, species, though data regarding

    mtDNA recombination within the human population re-

    mains contentious. A recent case of paternal mtDNA

    inheritance in a patient with a sporadic mtDNA mutation

    (2-bp microdeletion in the MTND2 gene) has documented

    mtDNA recombination at the cellular level (7) although

    studies of larger cohorts of patients with sporadic mtDNA

    mutations have shown this to be a rare event (8). The genetic

    advice to patients harbouring pathogenic, germline point

    mutations remains rmly based on the strict maternal

    transmission of mtDNA.

    Mitotic segregation describes the random distribution of

    mtDNA genotypes to daughter cells during cell division, and

    accounts for the remarkable change in mutation load

    between generations that has been documented in some

    families harbouring pathogenic, heteroplasmic mtDNA

    mutations. Such a phenomenon explains how patients may

    develop specic clinical phenotypes over time, and together

    with a number of other factors (the specic gene that is

    mutated, the mutation distribution within a tissue and the

    reliance of that tissue on mitochondrial-generated ATP as

    energy source) gives rise to the astonishing range of diverse

    clinical presentations of mtDNA disorders presenting in both

    childhood and adulthood (9). Relatively little is known

    about this process which is controlled by nuclear genetic

    factors (10).

    CLINICAL FEATURES OF mtDNA DISEASE

    Disease-related mutations of mtDNA can be divided into

    two groups maternally-inherited point mutations which may

    be heteroplasmic or homoplasmic and aect protein, tRNA or

    rRNA genes, and large-scale rearrangements (mainly single

    deletions) which span several genes (Fig. 1). More than 250

    pathogenic mutations have now been described (see also

    MITOMAP Database, http://www.mitomap.org) associated

    with an impressive array of clinical features ensuring patients

    present to every clinical speciality (summarized in Table 1);

    whilst muscle and brain involvement remain prominent,

    patients with mtDNA disease frequently present outside of

    neurology. Some classical clinical syndromes are widely

    recognized and associated with specic genetic defects making

    diagnosis straightforward; the most common of these are

    highlighted in Fig. 1. For many patients with suspected

    mtDNA disease, the clinical symptoms might be relatively

    non-specic, whilst in others there is a mtDNA mutation but

    aecting another nucleotide or gene, giving rise to the

    widespread genetic heterogeneity that characterizes these

    disorders; perhaps the best example of this is the syndrome

    of MELAS (mitochondrial myopathy, encephalopathy, lactic

    acidosis and stroke-like episodes) which in 480% of cases iscaused by a specic tRNA gene mutation (3243A4G intRNALeu(UUR)) yielding a range of biochemical defects. Many

    other point mutations, however, in this gene, other tRNA

    genes or even protein-encoding genes such as MTND5 and

    MTND1 can also cause MELAS. In cases where mtDNA

    disease is suspected, a diagnosis is often only made following

    an approach that integrates several lines of investigation

    including clinical, histochemical, biochemical and ultimately

    molecular genetic tests. Such investigations continue to rely

    heavily on the study of a clinically-aected tissue, often

    skeletal muscle, in which characteristic, cytochemical features

    such as ragged-red bres and cytochrome c oxidase decient

    Table 1Common clinical features of mtDNA disease

    Adult presentations

    Neurological migraine, strokes, epilepsy, myopathy,

    peripheral neuropathy, ataxia, sensorineural deafness

    Gastrointestinal irritable bowel, dysphagia

    Cardiac heart failure, heart block, cardiomyopathy

    Respiratory respiratory failure, nocturnal hypoventilation

    Endocrine diabetes, thyroid and parathyroid disease,

    ovarian failure

    Ophthalmology ophthalmoplegia, ptosis, optic atrophy,

    cataracts

    Paediatric presentations

    Neurological epilepsy, myopathy, psychomotor retardation,

    ataxia, spasticity, dystonia, sensorineural deafness

    Gastrointestinal vomiting, failure to thrive, dysphagia

    Cardiac hypertrophic cardiomyopathy, rhythm

    abnormalities

    Respiratory central hypoventilation/apnoea

    Endocrine diabetes, adrenal failure

    Ophthalmology optic atrophy

    Haematological anaemia, pancytopenia

    Renal renal tubular defects

    Liver hepatic failure

    MITOCHONDRIAL DNA MUTATIONS IN HUMAN DISEASE 145

  • bres are apparent (Fig. 2). Whole genome sequencing is now

    commonplace in many diagnostic centres, with novel mtDNA

    mutations still regularly being discovered, ever-widening the

    clinical spectrum of disease phenotypes (11). Such an

    increasing recognition of clinical presentations, together with

    the development of methods (e.g., screening urinary sedi-

    ments) for the non-invasive testing of patients and their

    asymptomatic relatives has helped to conrm mtDNA

    disorders as common genetic diseases with a minimum

    prevalence of at least 1 in 5000 aected individuals in some

    populations (12).

    The following sections of this review consider recent

    progress in understanding the role of mtDNA mutations in

    all aspects of human disease including ageing and cancer.

    CLASSICAL AND NON-CLASSICAL PRESENTATIONSASSOCIATED WITH mtDNA MUTATIONS

    The historical classication of mitochondrial disease was a

    cause of much debate between splitters and lumpers those

    who predicted a tight correlation between genotype and

    phenotype versus a belief that considerable overlap exists

    leading to genetic and phenotypic heterogeneity. Although

    there is evidence for and against both arguments, the diversity

    of presentations has made it dicult to dene the impact of

    mtDNA mutations on human health and as such it is helpful

    to stratify mtDNA mutations into one of the following

    groups: classic mtDNA syndromes (e.g., MELAS, MERRF,

    CPEO, NARP and LHON as indicated in Fig. 1), clinical

    syndromes with a high risk of mtDNAmutations, involvement

    in common disease phenotypes and mtDNA as a predisposi-

    tion for common disease (4). Well-recognized clinical

    syndromes in which mtDNA mutations have recently been

    demonstrated by whole mitochondrial genome sequencing

    include Leigh or Leigh-like encephalopathy (recurrent MTND

    and MTATP gene mutations) and exercise intolerance and

    rhabdomyolysis (several dierent MTCYB or tRNA muta-

    tions). Dening pathogenicity of specic base substitutions

    identied by sequencing may be dicult however due to the

    polymorphic nature of mtDNA and should be based on a

    number of functional and structural canonical criteria (13, 14).

    Misattribution of pathogenicity to a neutral mtDNA sequence

    variant clearly has important ramications for families, but

    this is not straightforward, especially as several homoplasmic

    mutations in mitochondrial tRNA genes are now recognized

    as causative (15).

    The role of mtDNA mutations in common disease

    phenotypes has been recognized for some time. The classic

    Figure 2. Cytochrome c oxidase decient cells a marker of mitochondrial disease and ageing. The rst three panels show

    transversely-orientated sections of skeletal muscle from a patient with an mtDNA-tRNA point mutation reacted for: (a)

    cytochrome c oxidase cytochemical activity alone, (b) succinate dehydrogenase (entirely encoded by nuclear DNA) activity, and

    (c) cytochrome c oxidase and succinate dehydrogenase activities sequentially. The latter activity stain exentuates the cytochrome

    c oxidase decient (blue) bres; the asterisked bre shows clear evidence of subsarcolemmal mitochondrial accumulation, a

    typical ragged-red bre. (d) transverse section of human colonic epithelium showing marked cytochrome c oxidase deciency

    due to the accumulation of somatic mtDNA mutations in the stem cells. The presence of a single mtDNA mutation (7275T4CinMTCO1) in the cytochrome c oxidase decient crypts (panel e) that is absent in surrounding positive staining crypts (panel f)

    is strong evidence that crypts expand throughout the colon by the process of crypt ssion (see ref. 42 for details).

    146 GREAVES AND TAYLOR

  • example is diabetes mellitus which often presents in association

    with other phenotypes including deafness and myopathy (Fig.

    1). Well-characterized, causative mtDNA mutations have only

    been described however in a small (0.1 3%) proportion of

    diabetic populations, although this does seem to be more

    prevalent in Japanese diabetics, suggesting a role for nuclear

    or other mtDNA genetic inuences. Polymorphic length

    variation in a poly-C tract of the mtDNA non-coding region

    has been demonstrated in several, late-onset multifactorial

    diseases including diabetes and was postulated to inuence

    pathophysiology through subtle eects on mtDNA replication

    (16) but a recent meta-analysis failed to conrm this

    association (17). Further interest in this area has been sparked

    by the report of a large pedigree with metabolic syndrome,

    characterized by hypertension, hypomagnesaemia, hypercho-

    lesterolaemia and often insulin resistance, in which the authors

    documented a marked association between aected family

    members and a homoplasmic mitochondrial tRNAIle muta-

    tion (18).

    Finally, considerable interest remains in the possibility that

    mtDNA mutation as a result of increased oxidative stress

    and free radical damage may either contribute to the

    neuronal cell damage or genetically predispose to common

    neurodegenerative diseases such as Alzheimers disease (AD),

    Parkinsons disease (PD) and amyotrophic lateral sclerosis

    (ALS). Whilst case-control studies have identied genuine,

    heteroplasmic pathogenic mtDNA mutations causing neuro-

    degenerative disease phenotypes (19), much interest has

    focussed on the phylogenetic analysis of mtDNA haplotypes.

    Ethnic populations can be divided into several mtDNA

    haplogroups dened by specic, single nucleotide polymorph-

    isms in both coding and non-coding mtDNA regions

    which have become xed by discrete maternal lineages,

    and it has been suggested that accumulations of these

    natural genetic variants might have subtle eects on respira-

    tory chain function, thus contributing to disease expression.

    Recent studies of two European populations have suggested

    that haplogroups J and K (20) and the haplogroup cluster

    UKJT (21) were associated with a decreased risk of PD, yet a

    similar association was not apparent in AD. Further

    considerations for a role of mtDNA mutations in both AD

    and PD are discussed in a recent review by Howell and

    colleagues (22).

    PATHOGENICITY: MODIFICATIONS ANDMECHANISMS

    Understanding the phenotypic and genotypic diversity and

    dissecting the molecular mechanisms by which mtDNA

    mutations result in disease remains a signicant challenge.

    For some mutations, particularly those that occur in

    mitochondrial tRNA genes and impair mitochondrial protein

    synthesis, the cybrid transfer of mutant mitochondria to the

    control nuclear background of either osteosarcoma or lung

    carcinoma r0 cells has yielded important information aboutthe precise molecular mechanisms (impairment of tRNA

    stability, aminoacylation,maturationor evenpost-transcriptional

    modication (23)) by which they impair cellular function.

    Transmitochondrial cybrid cells have proved to be a particu-

    larly useful system for the study of mtDNA disorders, not

    least because the generation of mice harbouring pathogenic,

    heteroplasmic mtDNA mutations has proved frustratingly

    problematic.

    Many dierent molecular defects result in the same

    endpoint a compromised respiratory chain activity leading

    to a reduction in ATP synthesis yet how can a biochemical

    defect involving a single enzyme complex, say complex I, lead

    to very dierent clinical phenotypes including Leigh syn-

    drome, LHON and MELAS and exercise intolerance? The

    tissue segregation of mutant mtDNA is an important factor,

    but not relevant to diseases caused by homoplasmic mtDNA

    mutations where matrilineal relatives with an identical

    mtDNA mutation can exhibit marked inconsistency in disease

    severity and progression. Two such examples are the

    1555A4Gmutation in the small (12S) rRNA subunit causingnonsyndromic deafness and the primary MTND1, MTND4

    and MTND6 LHON mutations which are usually homo-

    plasmic. Both disorders show incomplete penetrance and

    variable expressivity of either hearing loss (in the case of the

    1555A4G mutation) or visual loss in patients with LHONassociated with the specic mtDNA mutation, which is

    thought to be due to the interaction between other genetic

    factors and environmental factors. For example, the pheno-

    typic expression of the 11778G4A LHON mutation can beinuenced by both mtDNA haplogroup (24) and a homo-

    plasmic mitochondrial tRNAMet mutation (25) whilst a

    collaborative study of a large number of LHON pedigrees

    to search for nuclear genetic modiers has identied an

    X-chromosomal haplotype which may explain variable pene-

    trance and male sex bias (26). The inuence of environmental

    factors is best illustrated by patients with the 1555A4Gmutation who exhibit sensitivity to aminoglycoside antibiotics,

    an interaction enhanced by a conformational change in the

    secondary structure of the 12S rRNA molecule of the

    ribosome.

    ATP production aside, the role of mitochondria in the

    generation of reactive oxygen species (ROS) and regulation

    of the apoptotic cascade has prompted a closer inspection of

    these pathways in patient cell lines and tissues harbouring

    mtDNA mutations. Neuronal NT2 cybrid cells harbouring

    pathogenic LHON mutations show increased ROS production

    (27), whilst the ROS-mediated oxidative phosphorylation

    defect in cybrids carrying the 8993T4G NARP mutationcan be rescued by administering various antioxidants (28). The

    data concerning the eect of mtDNA mutations on apoptosis

    is a little more ambiguous; there is evidence from patient

    muscle biopsies and cell culture studies that certain mutations

    lead to an increase in programmed cell death, but data

    MITOCHONDRIAL DNA MUTATIONS IN HUMAN DISEASE 147

  • from the POLG1 mutator mice which accumulate mtDNA

    mutations sucient to cause severe respiratory chain dysfunc-

    tion showed normal levels of ROS and no increased sensitivity

    to oxidative stress-induced apoptotic cell death (29).

    mtDNA MUTATIONS IN AGEING

    Ageing is dened as a multi-factorial decline in an

    organisms physiological function, accompanied by a decline

    in fertility and an increasing risk of death. DNA damage

    including damge to mtDNA has been proposed to be a major

    contributing factor in the ageing process as protein, RNA and

    other macromolecules are rapidly turned over and therefore

    are unlikely candidates for a lifelong accumulation of damage.

    Given its essential role for the normal physiological function

    of the cell, the mitochondrial theory of ageing predicts that the

    progressive accumulation of ROS-induced, somatic mtDNA

    mutations during a lifetime, leads to an inevitable decline in

    mitochondrial function. ROS are produced at very low levels

    during the normal process of oxidative phosphorylation, and

    due to the close proximity of the respiratory chain and its lack

    of protective histone protein, mtDNA is a prime target for

    ROS-mediated damage. Mutations induced by ROS are

    thought to impair cellular OXPHOS leading to enhanced

    ROS production and further mtDNA mutation, and an

    exponentially increasing load of oxidative damage and

    mitochondrial dysfunction is produced (30). This results in

    eventual loss of cellular and tissue function through a

    combination of mechanisms including diminished energy

    supplies, signalling defects and apoptosis.

    An accumulation of mtDNA mutations with age was rst

    shown in the early 1990s with the 4,977-bp common mtDNA

    deletion being shown to accumulate to high levels in cells from

    a number of post-mitotic tissues (31). This followed the

    observation of cells in post mitotic tissues reecting an age-

    related deciency in cytochrome c oxidase (32). More recently

    studies have been carried out to see if this phenomenon occurs

    in dividing cells. Colonic epithelium (5), buccal epithelium (33)

    and CD34 cells from bone marrow (34) have all been shownto harbour an age-related accumulation of mtDNA mutations

    and the colonic epithelium has been shown to have an

    exponential age-related increase in the incidence of cyto-

    chrome c oxidase decient crypts (5). These data indicate that

    the mutational events are occurring in the stem cells which give

    rise to these tissues. It is interesting that large-scale mtDNA

    deletions have not been observed in any of the dividing cell

    populations investigated so far, only mtDNA point mutations

    have been reported. This suggests that perhaps there is either a

    dierent mechanism of mutation accumulation in mitotic and

    post-mitotic cells, or that there are dierent mechanisms of

    DNA damage detection and/or repair. One thing both cell

    types have in common is that there are not multiple dierent

    mutations within a cell, but clonal expansion of a single

    mutated genotype to high levels with in the cell. The

    mechanism by which clonal expansion occurs is unknown

    though mathematical modelling studies favour the theory of

    random genetic drift (35).

    The above data provide a correlative link between mtDNA

    mutations and ageing, but in order to investigate whether there

    is a causative link between mtDNA mutations and ageing mice

    have been developed which have a defect in the proof-reading

    portion of POLG1, the nuclear encoded catalytic subunit of

    mtDNA polymerase. This knock-in mutation causes a pro-

    found reduction in the exonuclease activity of the polymerase

    which is necessary for proof-reading of newly synthesized

    mtDNA. The mice have a greatly increased incidence of

    mtDNA point mutations and deletions, and show many of the

    classical features of premature ageing including osteoporosis,

    reduced subcutaneous fat, reduced muscle mass and alopecia

    (2, 3). Cytochrome c oxidase-decient cells were also observed

    in a number of tissues (2). ROS play a central role in the

    mitochondrial theory of ageing, however when ROS levels and

    oxidative stress levels were measured in the mutator mice, ROS

    production was found to be normal, and levels of oxidative

    stress to be only slightly elevated (3, 29). This has added to the

    intense debate about the exact role of ROS in ageing.

    Transgenic mice have been developed which over-express

    human catalase targeted to mitochondria, peroxisomes and

    the nucleus (36). Mice expressing mitochondrially-targeted

    catalase were found to have extended lifespan, delayed cardiac

    pathology and cataract development, associated with a decrease

    in the frequency of mtDNA deletions. Targeting catalase to the

    nucleus or peroxisomes had no eect on extending lifespan.

    Further studies on levels of ROS production and mtDNA

    mutations are clearly required to attempt to elucidate the role of

    these processes in ageing.

    mtDNA MUTATIONS IN TUMOURS

    Following the publication in 1998 of a paper by Polyak

    et al. which reported that 70% of colorectal cancers have

    somatic mtDNA mutations (37), there has been immense

    interest in the association of mtDNA mutations with cancer.

    This debate remains highly contentious with recent data

    supporting both sides of the argument. A recent paper

    investigating the incidence of mtDNA mutations in prostate

    cancer described an increase in the number of mutations in the

    MTCO1 gene in prostate cancer patients compared to controls

    (12% of patients to 2% of controls harbouring mutations)

    (38). These authors also introduced a pathogenic MTATP6

    mtDNA mutation (8993T4G) into a prostate cancer cell lineand found that when this was injected into nude mice, tumours

    were seven times larger than those without the pathogenic

    mutation. A similar study constructed cybrids using a HeLa

    nucleus and mtDNA containing either the 8993T4G NARPmutation or a second pathogenic MTATP6 mutation,

    9176T4C (39). Similarly, it was reported that the mutantsconferred a growth advantage in the early stages of the

    148 GREAVES AND TAYLOR

  • tumour, the authors documenting that apoptosis occurred less

    frequently in mutant cybrid cultures compared to wild-type

    cybrids. The decreased levels of apoptosis may be the

    mechanism by which tumour growth is promoted in the

    mutant cybrids.

    Although there are many reports describing the association

    with mtDNA mutations and tumours, there have been

    criticisms of the way in which the analyses have been carried

    out. A recent paper published by Bandelt and colleagues in

    PLoS Medicine reassessed many of these studies (40). They

    concluded that many of the authors appear to have scant

    knowledge of human phylogenies and that when numerous

    mtDNA sequence changes between the tumour and control

    sequence are documented, often these variants are present on a

    haplogroup other than that of the patient, and are therefore

    indicative of sample contamination. A second point is that

    although somatic mtDNAmutations do exist in many dierent

    tumours, there is little evidence to suggest that they are a cause

    of tumour formation; perhaps they are present in the normal

    tissue prior to tumourigenesis. In epithelial tissues such as the

    colonic (5) and buccal (33) epithelium, there are high levels of

    somatic mtDNA mutations in normal aged human tissue,

    suggesting that in those cancers the mutations are likely to be

    passive bystanders carried along during the formation of the

    tumour. However this might prove to be useful in deciphering

    the mechanism by which tumours form. In the development of

    colorectal cancer, which is considered to be a disease of colonic

    stem cells, there is controversy concerning the mechanism by

    which the tumour spreads following the initial transformation

    event. Crypt ssion is a process by which one crypt divides to

    form 2 daughter crypts and as such, the colon is thought to be

    composed of patches of genotypically-related crypts. This

    process has been proposed to be an important part of the

    mechanism by which tumours spread (41). Greaves et al. used

    cytochrome c oxidase deciency to identify crypts which might

    be related based on their mitochondrial phenotype. They

    sequenced the mitochondrial genome of two small patches (2

    and 3 crypts) of cytochrome c oxidase decient crypts and

    adjacent cytochrome c oxidase positive crypts, showing that

    each patch of decient crypts contained a somatic mtDNA

    mutation which was not present in adjacent positive crypts or

    in bulk homogenate DNA from that patient (see Fig. 2).

    Subsequent sequencing of a pair of cytochrome c oxidase

    decient crypts in the process of ssion revealed these to have a

    somatic mtDNA mutation not present in adjacent cytochrome

    c oxidase positive crypts. These data predict that an mtDNA

    mutation originating in the stem cell of a colonic crypt can go

    on to occupy the whole crypt, and that mutated clones expand

    by crypt ssion (42).

    CONCLUSION

    Although pathogenic mtDNA mutations were rst

    described nearly 20 years ago, their rightful impact on human

    health is only now becoming apparent. Despite the availability

    of cell culture and some animal models of disease, surprisingly

    little is known of the pathophysiological mechanisms by which

    mtDNA mutations, through respiratory chain deciency,

    cause cell dysfunction and death. Sadly, eective treatment

    strategies for these disorders remain lacking despite many

    novel experimental approaches to the problems associated

    with providing cures (43). Current clinical practice for patients

    and their families is based upon supportive measures and

    genetic counselling, yet given the inherent diculties in

    treating these disorders, a dierent line of attack, the

    prevention of mtDNA disease transmission, is being actively

    pursued. Preimplantation genetic diagnosis (44) and pro-

    nuclear transfer between single cell embryos (45) are two such

    avenues, and whilst there are considerable ethical and moral

    issues to be considered, clearly developing techniques to

    improve the chances of a mother with mtDNA disease having

    a healthy child is a major priority for families, scientists and

    clinicians alike (46).

    ACKNOWLEDGEMENTS

    We thank the Wellcome Trust, Muscular Dystrophy Cam-

    paign and Newcastle upon Tyne NHS Hospitals Trust for

    their continuing nancial support of this work.

    REFERENCES1. Luoma, P., Melberg, A., Rinne, J. O., Kaukonen, J. A., Nupponen,

    N. N., Chalmers, R. M., Oldfors, A., Rautakorpi, I., Peltonen, L.,

    Majamaa, K., Somer, H., and Suomalainen, A. (2004) Parkinsonism,

    premature menopause, and mitochondrial DNA polymerase gamma

    mutations: clinical and molecular genetic study. Lancet 364, 875 882.

    2. Trifunovic, A., Wredenberg, A., Falkenberg, M., Spelbrink, J. N.,

    Rovio, A. T., Bruder, C. E., Bohlooly-Y, M., Gidlof, S., Oldfors, A.,

    Wibom, R., Tornell, J., Jacobs, H. T., and Larsson, N. G. (2004)

    Premature ageing in mice expressing defective mitochondrial DNA

    polymerase. Nature 429, 417 423.

    3. Kujoth, G. C., Hiona, A., Pugh, T. D., Someya, S., Panzer, K.,

    Wohlgemuth, S. E., Hofer, T., Seo, A. Y., Sullivan, R., Jobling, W.

    A., Morrow, J. D., Van Remmen, H., Sedivy, J. M., Yamasoba, T.,

    Tanokura, M., Weindruch, R., Leeuwenburgh, C., and Prolla, T. A.

    (2005) Mitochondrial DNA mutations, oxidative stress, and apoptosis

    in mammalian aging. Science 309, 481 484.

    4. Taylor, R. W., and Turnbull, D. M. (2005) Mitochondrial DNA

    mutations in human disease. Nat. Rev. Genet. 6, 389 402.

    5. Taylor, R. W., Barron, M. J., Borthwick, G. M., Gospel, A.,

    Chinnery, P. F., Samuels, D. C., Taylor, G. A., Plusa, S. M.,

    Needham, S. J., Greaves, L. C., Kirkwood, T. B., and Turnbull, D. M.

    (2003) Mitochondrial DNA mutations in human colonic crypt stem

    cells. J. Clin. Invest. 112, 1351 1360.

    6. Nishimura, Y., Yoshinari, T., Naruse, K., Yamada, T., Sumi, K.,

    Mitani, H., Higashiyama, T., and Kuroiwa, T. (2006) Active digestion

    of sperm mitochondrial DNA in single living sperm revealed by

    optical tweezers. Proc. Natl. Acad. Sci. USA 103, 1382 1387.

    7. Kraytsberg, Y., Schwartz, M., Brown, T. A., Ebralidse, K., Kunz,

    W. S., Clayton, D. A., Vissing, J., and Khrapko, K. (2004)

    Recombination of human mitochondrial DNA. Science 304, 981.

    MITOCHONDRIAL DNA MUTATIONS IN HUMAN DISEASE 149

  • 8. Taylor, R. W., McDonnell, M. T., Blakely, E. L., Chinnery, P. F.,

    Taylor, G. A., Howell, N., Zeviani, M., Briem, E., Carrara, F., and

    Turnbull, D. M. (2003) Genotypes from patients indicate no paternal

    mitochondrial DNA contribution. Ann. Neurol. 54, 521 524.

    9. DiMauro, S., and Schon, E. A. (2003) Mitochondrial respiratory-

    chain diseases. N. Engl. J. Med. 348, 2656 2668.

    10. Battersby, B. J., Loredo-Osti, J. C., and Shoubridge, E. A. (2003)

    Nuclear genetic control of mitochondrial DNA segregation. Nat.

    Genet. 33, 183 186.

    11. Swalwell, H., Deschauer, M., Hartl, H., Strauss, M., Turnbull, D. M.,

    Zierz, S., and Taylor, R. W. (2006) Pure myopathy associated with a

    novel mitochondrial tRNA gene mutation. Neurology 66, 447 449.

    12. Schaefer, A. M., Taylor, R. W., Turnbull, D. M., and Chinnery, P. F.

    (2004) The epidemiology of mitochondrial disorders past, present

    and future. Biochim. Biophys. Acta 1659, 115 120.

    13. McFarland, R., Elson, J. L., Taylor, R. W., Howell, N., and Turnbull,

    D. M. (2004) Assigning pathogenicity to mitochondrial tRNA

    mutations: when denitely maybe is not good enough. Trends

    Genet. 20, 591 596.

    14. Mitchell, A. L., Elson, J. L., Howell, N., Taylor, R. W., and Turnbull,

    D. M. (2006) Sequence variation in mitochondrial complex I genes:

    mutation or polymorphism? J. Med. Genet. 43, 175 179.

    15. Taylor, R. W., McDonnell, M. T., Blakely, E. L., Chinnery, P. F.,

    Taylor, G. A., Howell, N., Zeviani, M., Briem, E., Carrara, F., and

    Turnbull, D. M. (2003) Genotypes from patients indicate no paternal

    mitochondrial DNA contribution. Ann. Neurol. 54, 521 524.

    16. Poulton, J., Luan, J., Macaulay, V., Hennings, S., Mitchell, J., and

    Wareham, N. J. (2002) Type 2 diabetes is associated with a common

    mitochondrial variant: evidence from a population-based case-control

    study. Hum. Mol. Genet. 11, 1581 1583.

    17. Chinnery, P. F., Elliott, H. R., Patel, S., Lambert, C., Keers, S. M.,

    Durham, S. E., McCarthy, M. I., Hitman, G. A., Hattersley, A. T.,

    and Walker, M. (2005) Role of the mitochondrial DNA 16184-16193

    poly-C tract in type 2 diabetes. Lancet 366, 1650 1651.

    18. Wilson, F. H., Hariri, A., Farhi, A., Zhao, H., Petersen, K. F., Toka,

    H. R., Nelson-Williams, C., Raja, K. M., Kashgarian, M., Shulman,

    G. I., Scheinman, S. J., and Lifton, R. P. (2004) A cluster of metabolic

    defects caused by mutation in a mitochondrial tRNA. Science 306,

    1190 1194.

    19. Borthwick, G. M., Taylor, R. W., Walls, T. J., Tonska, K., Taylor, G.

    A., Shaw, P. J., Ince, P. G., and Turnbull, D. M. (2006) Motor neuron

    disease in a patient with a mitochondrial tRNAIle mutation. Ann.

    Neurol., 59, 570 574.

    20. van der Walt, J. M., Nicodemus, K. K., Martin, E. R., Scott, W. K.,

    Nance, M. A., Watts, R. L., Hubble, J. P., Haines, J. L., Koller,

    W. C., Lyons, K., Pahwa, R., Stern, M. B., Colcher, A., Hiner, B. C.,

    Jankovic, J., Ondo, W. G., Allen, F. H. Jr., Goetz, C. G., Small,

    G. W., Mastaglia, F., Stajich, J. M., McLaurin, A. C., Middleton,

    L. T., Scott, B. L., Schmechel, D. E., Pericak-Vance, M. A., and

    Vance, J. M. (2003) Mitochondrial polymorphisms signicantly

    reduce the risk of Parkinson disease. Am. J. Hum. Genet. 72, 804 811.

    21. Pyle, A., Foltynie, T., Tiangyou, W., Lambert, C., Keers, S. M.,

    Allcock, L. M., Davison, J., Lewis, S. J., Perry, R. H., Barker, R.,

    Burn, D. J., and Chinnery, P. F. (2005) Mitochondrial DNA

    haplogroup cluster UKJT reduces the risk of PD. Ann. Neurol. 57,

    564 567.

    22. Howell, N., Elson, J. L., Chinnery, P. F., and Turnbull, D. M. (2005)

    mtDNA mutations and common neurodegenerative disorders. Trends

    Genet. 21, 583 586.

    23. Kirino, Y., Yasukawa, T., Marjavaara, S. K., Jacobs, H. T., Holt,

    I. J., Watanabe, K., and Suzuki, T. (2006) Acquisition of the wobble

    modication in mitochondrial tRNALeu(CUN) bearing the G12300A

    mutation suppresses the MELAS molecular defect. Hum. Mol. Genet.

    15, 897 904.

    24. Torroni, A., Petrozzi, M., DUrbano, L., Sellitto, D., Zeviani, M.,

    Carrara, F., Carducci, C., Leuzzi, V., Carelli, V., Barboni, P., De

    Negri, A., and Scozzari, R. (1997) Haplotype and phylogenetic

    analyses suggest that one European-specic mtDNA background

    plays a role in the expression of Leber hereditary optic neuropathy by

    increasing the penetrance of the primary mutations 11778 and 14484.

    Am. J. Hum. Genet. 60, 1107 1121.

    25. Qu, J., Li, R., Zhou, X., Tong, Y., Lu, F., Qian, Y., Hu, Y., Mo, J. Q.,

    West, C. E., and Guan, M. X. (2006) The novel A4435G mutation in

    the mitochondrial tRNAMet may modulate the phenotypic expression

    of the LHON-associated ND4 G11778A mutation. Invest. Ophthal-

    mol. Vis. Sci. 47, 475 483.

    26. Hudson, G., Keers, S., Yu Wai Man, P., Griths, P., Huoponen, K.,

    Savontaus, M. L., Nikoskelainen, E., Zeviani, M., Carrara, F.,

    Horvath, R., Karcagi, V., Spruijt, L., de Coo, I. F., Smeets, H. J., and

    Chinnery, P. F. (2005) Identication of an X-chromosomal locus and

    haplotype modulating the phenotype of a mitochondrial DNA

    disorder. Am. J. Hum. Genet. 77, 1086 1091.

    27. Wong, A., Cavelier, L., Collins-Schramm, H. E., Seldin, M. F.,

    McGrogan, M., Savontaus, M. L., and Cortopassi, G. A. (2002)

    Dierentiation-specic eects of LHON mutations introduced into

    neuronal NT2 cells. Hum. Mol. Genet. 11, 431 438.

    28. Mattiazzi, M., Vijayvergiya, C., Gajewski, C. D., DeVivo, D. C.,

    Lenaz, G., Wiedmann, M., and Manfredi, G. (2004) The mtDNA

    T8993G (NARP) mutation results in an impairment of oxidative

    phosphorylation that can be improved by antioxidants. Hum. Mol.

    Genet. 13, 869 879.

    29. Trifunovic, A., Hansson, A., Wredenberg, A., Rovio, A. T., Dufour,

    E., Khvorostov, I., Spelbrink, J. N., Wibom, R., Jacobs, H. T., and

    Larsson, N. G. (2005). Somatic mtDNA mutations cause aging

    phenotypes without aecting reactive oxygen species production.

    Proc. Natl. Acad. Sci. USA 102, 17993 17998.

    30. Miquel, J., Economos, A. C., Fleming, J., and Johnson, J. E. Jr.

    (1980) Mitochondrial role in cell aging. Exp. Gerontol. 15, 575 591.

    31. Cortopassi, G. A., and Arnheim, N. (1990). Detection of a specic

    mitochondrial DNA deletion in tissues of older humans. Nucleic Acids

    Res. 18, 6927 6933.

    32. Muller-Hocker, J. (1989) Cytochrome c oxidase decient cardiomyo-

    cytes in the human heart an age-related phenomenon. A

    histochemical ultracytochemical study. Am. J. Pathol. 134, 11671173.

    33. Nekhaeva, E., Bodyak, N. D., Kraytsberg, Y., McGrath, S. B., Van

    Orsouw,N. J., Pluzhnikov,A.,Wei, J.Y.,Vijg, J., andKhrapko,K. (2002)

    Clonally expanded mtDNA point mutations are abundant in individual

    cells of human tissues. Proc. Natl. Acad. Sci. USA 99, 55215526.

    34. Shin, M. G., Kijigaya, S., Tarnowka, M., McCoy, J. P. Jr., Levin, B.

    C., and Young, N. S. (2004) Mitochondrial DNA sequence hetero-

    geneity in circulating normal human CD34 cells and granulocytes.

    Blood 103, 4466 4477.

    35. Elson, J. L., Samuels, D. C., Turnbull, D. M., and Chinnery, P. F.

    (2001) Random intracellular drift explains the clonal expansion of mito-

    chondrial DNAmutations with age. Am. J. Hum. Genet. 68, 802 806.

    36. Schriner, S. E., Linford, N. J., Martin, G. M., Treuting, P., Ogburn,

    C. E., Emond, M., Coskun, P. E., Ladiges, W., Wolf, N., Van

    Remmen, H., Wallace, D. C., and Rabinovitch, P. S. (2005) Extension

    of murine life span by overexpression of catalase targeted to

    mitochondria. Science 308, 1909 1911.

    37. Polyak, K., Li, Y., Zhu, H., Lengauer, C., Willson, J. K., Markowitz,

    S. D., Trush, M. A., Kinzler, K. W., and Vogelstein, B. (1998)

    Somatic mutations of the mitochondrial genome in human colorectal

    tumours. Nat. Genet. 20, 291 293.

    38. Petros, J. A., Baumann, A. K., Ruiz-Pesini, E., Amin, M. B., Sun, C. Q.,

    Hall, J., Lim, S., Issa, M.M., Flanders, W. D., Hosseini, S. H., Marshall,

    F. F., and Wallace, D. C. (2005) mtDNA mutations increase tumor-

    igenicity in prostate cancer. Proc. Natl. Acad. Sci. USA 102, 719 724.

    150 GREAVES AND TAYLOR

  • 39. Shidara, Y., Yamagata, K., Kanamori, T., Nakano, K., Kwong, J. Q.,

    Manfredi, G., Oda, H., and Ohta, S. (2005) Positive contribution of

    pathogenic mutations in the mitochondrial genome to the promotion

    of cancer by prevention from apoptosis. Cancer Res. 65, 1655 1663.

    40. Salas, A., Yao, Y. G., Macaulay, V., Vega, A., Carracedo, A., and

    Bandelt, H. J. (2005) A critical reassessment of the role of

    mitochondria in tumorigenesis. PLoS Med. 2, e296.

    41. Preston, S. L., Wong, W. M., Chan, A. O., Poulsom, R., Jeery, R.,

    Goodlad, R. A., Mandir, N., Elia, G., Novelli, M., Bodmer, W. F.,

    Tomlinson, I. P., and Wright, N. A. (2003) Bottom-up histogenesis of

    colorectal adenomas: origin in the monocryptal adenoma and initial

    expansion by crypt ssion. Cancer Res. 63, 3819 3825.

    42. Greaves, L. C., Preston, S. L., Tadrous, P. J., Taylor, R. W., Barron,

    M. J., Oukrif, D., Leedham, S. J., Deheragoda, M., Sasieni, P.,

    Novelli, M. R., Jankowski, J. A., Turnbull, D. M., Wright, N. A., and

    McDonald, S. A. (2006) Mitochondrial DNA mutations are

    established in human colonic stem cells, and mutated clones expand

    by crypt ssion. Proc. Natl. Acad. Sci. USA 103, 714 719.

    43. Taylor, R. W. (2005) Gene therapy for the treatment of mitochondrial

    DNA disorders. Expert Opin. Biol. Ther. 5, 183 194.

    44. Steann, J., Frydman, N., Gigarel, N., Burlet, P., Ray, P. F., Fanchin,

    R., Feyereisen, E., Kerbrat, V., Tachdjian, G., Bonnefont, J. P.,

    Frydman, R., and Munnich, A. (2005) Analysis of mtDNA variant

    segregation during early human embryonic development: a tool for

    successful NARP preimplantation diagnosis. J. Med. Genet. 43, 244

    247.

    45. Sato, A., Kono, T., Nakada, K., Ishikawa, K., Inoue, S., Yonekawa,

    H., and Hayashi, J. (2005) Gene therapy for progeny of mito-mice

    carrying pathogenic mtDNA by nuclear transplantation. Proc. Natl.

    Acad. Sci. USA 102, 16765 16770.

    46. Brown, D. T., Herbert, M., Lamb, V. K., Chinnery, P. F., Taylor, R.

    W., Lightowlers, R. N., Craven, L., Cree, L., Gardner, J. L., and

    Turnbull, D. M. (2006) Transmission of mitochondrial DNA

    disorders: possibilities for the future. Lancet, in press.

    MITOCHONDRIAL DNA MUTATIONS IN HUMAN DISEASE 151