<<

The new england journal of medicine

review article

mechanisms of disease Mitochondrial Respiratory-Chain Diseases

Salvatore DiMauro, M.D., and Eric A. Schon, Ph.D.

From the Departments of Neurology (S.D., ore than a billion years ago, aerobic colonized E.A.S.) and Genetics and Development primordial eukaryotic cells that lacked the ability to use metabolical- (E.A.S.), Columbia University College of m Physicians and Surgeons, New York. Ad- ly. A symbiotic relationship developed and became permanent. The bacteria dress reprint requests to Dr. DiMauro at evolved into mitochondria, thus endowing the host cells with aerobic , a 4-420 College of Physicians and Surgeons, much more efficient way to produce energy than anaerobic . Structurally, mito- 630 W. 168th St., New York, NY 10032, or at [email protected]. chondria have four compartments: the outer membrane, the inner membrane, the inter- membrane space, and the matrix (the region inside the inner membrane). They perform N Engl J Med 2003;348:2656-68. numerous tasks, such as pyruvate oxidation, the Krebs cycle, and metabolism of amino Copyright © 2003 Massachusetts Medical Society. acids, fatty acids, and , but the most crucial is probably the generation of energy as (ATP), by means of the electron-transport chain and the ox- idative-phosphorylation system (the “respiratory chain”) (Fig. 1). The respiratory chain, located in the inner mitochondrial membrane, consists of five multimeric complexes (Fig. 2B): reduced nicotinamide adenine dinucleotide (NADH) dehydrogenase–ubiquinone (complex I, approximately 46 sub- units), –ubiquinone oxidoreductase (complex II, 4 subunits), ubiquinone–cytochrome c oxidoreductase (complex III, 11 subunits), cytochrome c oxi- dase (complex IV, 13 subunits), and ATP (complex V, approximately 16 sub- units). The respiratory chain also requires two small electron carriers, ubiquinone (coen- zyme Q10) and cytochrome c. ATP synthesis entails two coordinated processes (Fig. 2B). First, electrons (actually hydrogen ions derived from NADH and reduced flavin adenine dinucleotide in inter- mediary metabolism) are transported along the complexes to molecular oxygen, thereby producing water. At the same time, protons are pumped across the mitochondrial inner membrane (i.e., from the matrix to the intermembrane space) by complexes I, III, and IV. ATP is generated by the influx of these protons back into the through complex V (ATP synthase), the world’s tiniest rotary motor.1,2 Mitochondria are the only of the cell besides the nucleus that contain their own DNA (called mtDNA) and their own machinery for synthesizing RNA and pro- teins. There are hundreds or thousands of mitochondria per cell, and each contains ap- proximately five mitochondrial genomes. Because mtDNA has only 37 , most of the approximately 900 products in the are encoded by nuclear DNA (nDNA) and are imported from the cytoplasm. Defects in any of the numerous mitochondrial pathways can cause mitochondrial diseases, but we will confine our discussion to disorders of the respiratory chain. Be- cause many of them involve and , these disorders are also known as mitochondrial encephalomyopathies. The fact that the respiratory chain is under dual genetic control makes these disorders particularly fascinating, because they involve both mendelian and mitochondrial genetics. Moreover, these diseases are not as rare as com- monly believed; their estimated prevalence of 10 to 15 cases per 100,000 persons is sim- ilar to that of better known neurologic diseases, such as amyotrophic lateral sclerosis and the muscular dystrophies.3 The genetic classification of the primary mitochondrial diseases distinguishes dis-

2656 n engl j med 348;26 www.nejm.org june 26, 2003

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

mechanisms of disease orders due to defects in mtDNA, which are inherit- mitochondrial genetics ed according to the rules of mitochondrial genetics, from those due to defects in nDNA, which are trans- The mtDNA is a 16,569-bp, double-strand- mitted by mendelian inheritance. Since the discov- ed, circular molecule containing 37 genes (Fig. 2A). ery of the first pathogenic in human Of these, 24 are needed for mtDNA (2 ri- mtDNA a mere 15 years ago,4,5 the increase in our bosomal RNAs [rRNAs] and 22 transfer RNAs knowledge of the role of mitochondria in disease [tRNAs]), and 13 encode subunits of the respirato- has exceeded all expectations. ry chain: seven subunits of complex I (ND1, 2, 3, 4,

Glucose Fatty acids

Glycolysis Fatty-acyl–CoA AlaninePyruvate Lactate

CPT-I

H+ Pyruvate ADP ATP Fatty-acyl–carnitine Carnitine

DIC ANT CACT CPT-II H+ Pyruvate ADP ATP Fatty-acyl–carnitine Carnitine Fatty-acyl–CoA

PDHC Acetyl-CoA b-oxidation

FAD Citrate Oxaloacetate TCA cycle Isocitrate FADH2 a-Ketoglutarate H+ ETF Malate

NADH Succinyl-CoA

ETF-DH Oxidative phosphorylation Fumarate FADH2 H+ Succinate H+ ADP ATP Electron-transport chain H+ H+

I II CoQ III IV V

H+ H+ Cytochrome c H+ H+

Figure 1. Selected Metabolic Pathways in Mitochondria. The spirals represent the spiral of reactions of the b-oxidation pathway, resulting in the liberation of acetyl–coenzyme A (CoA) and the reduction of . ADP denotes , ATP adenosine triphosphate, ANT adenine translocator, CACT carnitine–acyl- carnitine , CoQ coenzyme Q, CPT carnitine palmitoyltransferase, DIC dicarboxylate carrier, ETF electron-transfer flavoprotein, ETF- DH electron-transfer dehydrogenase, FAD flavin adenine dinucleotide, FADH2 reduced FAD, NADH reduced nicotinamide adenine dinucle- otide, PDHC complex, TCA tricarboxylic acid, I complex I, II complex II, III complex III, IV complex IV, and V complex V.

n engl j med 348;26 www.nejm.org june 26, 2003 2657

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

The new england journal of medicine

4L, 5, and 6 [ND stands for NADH dehydrogenase]), respiratory-chain disorders one subunit of complex III (cytochrome b), three due to defects in mtdna subunits of (COX I, II, and III), and two subunits of ATP synthase (A6 and A8). Mi- The small mitochondrial genome contains many tochondrial genetics differs from mendelian genet- mutations that cause a wide variety of clinical syn- ics in three major aspects: maternal inheritance, dromes, as shown in Figure 3.7 The genome is pep- heteroplasmy, and mitotic segregation. pered with mutations, although a few “hot spots” stand out. Relatively few mutations have been found maternal inheritance in rRNA genes, and all these are confined to the 12S As a general rule, all mitochondria (and all mtDNAs) rRNA. Not shown on the map are the hundreds of in the zygote derive from the ovum. Therefore, a different pathogenic giant deletions of 2 to 10 kb, mother carrying an mtDNA passes it on which invariably delete tRNA genes. to all her children, but only her daughters will trans- Although clinically distinct, most mtDNA-relat- mit it to their progeny. Recent evidence of paternal ed diseases share the features of transmission of mtDNA in skeletal muscle (but not and massive mitochondrial proliferation in muscle in other tissues) in a patient with a mitochondrial (resulting in ragged-red fibers). In muscle-biopsy myopathy6 serves as an important warning that ma- specimens, the mtDNAs accumulate prefer- ternal inheritance of mtDNA is not an absolute rule, entially in ragged-red fibers, and ragged-red fibers but it does not negate the primacy of maternal in- are typically negative for cytochrome c oxidase ac- heritance in mtDNA-related diseases. tivity8 (Fig. 4).

heteroplasmy and the threshold effect Figure 2 (facing page). Mitochondrial DNA (mtDNA) There are thousands of mtDNA molecules in each and the Mitochondrial Respiratory Chain. cell, and in general, pathogenic mutations of Panel A shows the map of the human mitochondrial ge- mtDNA are present in some but not all of these ge- nome. The protein-coding genes — seven subunits of nomes. As a result, cells and tissues harbor both complex I (ND), three subunits of cytochrome c oxidase normal (wild-type) and mutant mtDNA, a situation (COX), the cytochrome b subunit of complex III (Cyt b), known as heteroplasmy. Heteroplasmy can also ex- and two subunits of adenosine triphosphate (ATP) syn- thase (A6 and A8) — are shown in red. The protein-syn- ist at the organellar level: a single thesis genes — the 12S and 16S ribosomal RNAs and the can harbor both normal and mutant mtDNAs. In 22 transfer RNAs (three-letter symbols) — normal subjects, all mtDNAs are identical (ho- are shown in blue. The D-loop region controls the initia- moplasmy). Not surprisingly, a minimal number of tion of replication and transcription of mtDNA. Panel B mutant mtDNAs must be present before oxidative shows the subunits of the respiratory chain encoded by nuclear DNA (nDNA) in blue and the subunits encoded dysfunction occurs and clinical signs become appar- by mtDNA in red. As electrons (e–) flow along the elec- ent: this is the threshold effect. The threshold for tron-transport chain, protons (H+) are pumped from the disease is lower in tissues that are highly dependent matrix to the intermembrane space through complexes I, on oxidative metabolism, such as brain, , skel- III, and IV and then back into the matrix through complex etal muscle, retina, renal tubules, and endocrine V, to produce ATP. Coenzyme Q (CoQ) and cytochrome c (Cyt c) are electron-transfer carriers. Genes responsible glands. These tissues will therefore be especially for the indicated respiratory-chain disorders are also vulnerable to the effects of pathogenic mutations shown. ATPase 6 denotes ATP synthase 6; BCS1L cyto- in mtDNA. chrome b–c complex assembly protein(complex III); NDUF NADH dehydrogenase–ubiquinone oxidoreduc- mitotic segregation tase; SCO synthesis of cytochrome oxidase; SDHA, SDHB, SDHC, and SDHD succinate dehydrogenase sub- The random redistribution of organelles at the time units; SURF1 surfeit gene 1; FBSN familial bilateral stri- of cell division can change the proportion of mutant atal necrosis; LHON Leber’s hereditary optic mtDNAs received by daughter cells; if and when the neuropathy; MELAS mitochondrial encephalomyopathy, pathogenic threshold in a previously unaffected tis- lactic acidosis, and strokelike episodes; MILS maternally sue is surpassed, the can also change. inherited Leigh’s syndrome; NARP neuropathy, , and retinitis pigmentosa; GRACILE growth retardation, This explains the age-related, and even tissue-relat- , lactic acidosis, and early death; and ALS ed, variability of clinical features frequently observed amyotrophic lateral sclerosis. in mtDNA-related disorders.

2658 n engl j med 348;26 www.nejm.org june 26, 2003

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

mechanisms of disease

A D-loop region

Phe 12S Pro Val Thr Cyt b 16S

Leu(UUR) ND1 Glu ND6

Ile Gln Met

ND2 ND5

Trp Ala Asn Leu (CUN) Cys Ser (AGY) Tyr His

COXI Ser(UCN) ND4 Asp Arg ND4L Lys Gly COXII ND3 A8 A6 COXIII

B ND1, ND2, ND3, ND4, Cyt b COX1, COXII, ATPase 6 ND4L, ND5, ND6 Sporadic COXIII NARP LHON Encephalomyopathy Sporadic anemia MILS MELAS Septo-optic dysplasia Sporadic myopathy FBSN LHON and Cardiomyopathy Encephalomyopathy Sporadic myopathy ALS-like syndrome H+ H+ H+ H+

Succinate Fumarate Matrix O2 H2O ADP ATP Inner ND1 ND2 ND3 COXI Cyt b COXII A8 mitochondrial ND6 ND4L e¡ COXIII A6 membrane ND5 ND4 e¡ e¡ CoQ Intermembrane Cyt c space Complex I Complex II Complex III Complex IV Complex V NDUFS1, NDUFS2, SDHA, SDHB, BCS1L COX10, COX15, NDUFS4, NDUFS7, SDHC, SDHD Leigh’s syndrome SCO1, SCO2, SURF1 NDUFS8, NDUFV1 Leigh’s syndrome GRACILE syndrome Leigh’s syndrome Leigh’s syndrome Paraganglioma Hepatopathy Leukodystrophy Pheochromocytoma Cardioencephalomyopathy Leukodystrophy and tubulopathy No. of mtDNA- 7 01 3 2 encoded subunits No. of nDNA- ∼39 41010 ∼14 encoded subunits

n engl j med 348;26 www.nejm.org june 26, 2003 2659

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

The new england journal of medicine

Parkinsonism, aminoglycoside-induced deafness LS, MELAS, MELAS, multisystem disease myoglobinuria Myopathy, PEO Cardiomyopathy Cardiomyopathy, ECM PEO, LHON, MELAS, ECM, LHON, myopathy, myopathy, cardiomyopathy, F 12s P cardiomyopathy, MELAS V T 16S and parkinsonism LHON Cyt b L1 Cardiomyopathy, ECM Myopathy, cardiomyopathy, E PEO ND1 LHON, MELAS, diabetes, Myopathy, MELAS ND6 I LHON and dystonia Myopathy, lymphoma Q M Cardiomyopathy, LS, MELAS LHON ND2 ND5 LS, ataxia, chorea, myopathy Cardiomyopathy, ECM, W PEO A PEO, myopathy, N L2 Myopathy, C S2 PEO Y H Diabetes and deafness ECM COXI PEO ND4 LHON, myopathy, Myoglobinuria, LHON and dystonia motor neuron disease, S1 D ND4L sideroblastic anemia COXII ND3 R PPK, deafness, A8 LHON MERRF–MELAS Cardiomyopathy, K A6 COXIII G myoclonus Progressive myoclonus, Myopathy, epilepsy, and optic atrophy multisystem disease, NARP, MILS, encephalomyopathy Cardiomyopathy, FBSN SIDS, ECM Cardiomyopathy LS, ECM, PEO, MERRF, myoglobinuria MELAS, deafness

Figure 3. Mutations in the Human Mitochondrial Genome That Are Known to Cause Disease. Disorders that are frequently or prominently associated with mutations in a particular gene are shown in boldface. Diseases due to mutations that impair mitochondrial protein synthesis are shown in blue. Diseases due to mutations in protein-coding genes are shown in red. ECM de- notes encephalomyopathy; FBSN familial bilateral striatal necrosis; LHON Leber’s hereditary optic neuropathy; LS Leigh’s syndrome; MELAS mitochondrial encephalomyopathy, lactic acidosis, and strokelike episodes; MERRF myoclonic epilepsy with ragged-red fibers; MILS mater- nally inherited Leigh’s syndrome; NARP neuropathy, ataxia, and retinitis pigmentosa; PEO progressive external ophthalmoplegia; PPK pal- moplantar keratoderma; and SIDS sudden infant death syndrome.

Mutations in mtDNA can affect specific mitochondrial encephalomyopathy, lactic acidosis, of the respiratory chain or the synthesis of mito- and strokelike episodes (MELAS) syndrome, but chondrial proteins as a whole (mutations in tRNA they cause other syndromes as well. Conversely, mu- or rRNA genes, or giant deletions). There is no tations in different genes can cause the same syn- straightforward relation between the site of the mu- drome; MELAS, again, is a prime example (Fig. 3). tation and the clinical phenotype, even with a muta- There are exceptions: virtually all patients who have tion in a single gene. For example, mutations in the the myoclonus epilepsy with ragged-red fibers tRNALeu(UUR) gene are usually associated with the (MERRF) syndrome have mutations in the tRNALys

2660 n engl j med 348;26 www.nejm.org june 26, 2003

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

mechanisms of disease

* *

* * * *

A B

Figure 4. Serial Cross-Sections of Muscle from a Patient with the Kearns–Sayre Syndrome, Showing Increased Mitochondrial Activity in Ragged-Red Fibers on Staining with Succinate Dehydrogenase (Asterisks in Panel A; ¬120) and the Absence of Activity on Staining with Cytochrome c Oxidase (Asterisks in Panel B; ¬120). gene,9 all patients with Leber’s hereditary optic ovum and embryo allows only a minority of mater- neuropathy have mutations in ND genes,10 and nal mtDNAs to populate the fetus. On rare occa- most mutations in the cytochrome b gene cause ex- sions a partially deleted mtDNA (or its progeny, ercise intolerance.11 depending on when in oogenesis the oc- Because mitochondria are ubiquitous, every tis- curred) may slip through. A few mtDNAs with giant sue in the body can be affected by mtDNA muta- deletions in the blastocyst can then enter all three tions, which is why mitochondrial diseases are of- germ layers and result in the Kearns–Sayre syn- ten multisystemic. Table 1 lists the most common drome (a multisystem disorder), segregate to the mtDNA-related syndromes. Certain constellations hematopoietic lineage and cause Pearson’s syn- of symptoms and signs are characteristic of these drome, or segregate to muscle and cause progres- syndromes, and the diagnosis is relatively easy to es- sive external ophthalmoplegia (Table 1). In these tablish in patients with these typical features. How- three cases, all mutated mtDNAs in the patient are ever, as a result of heteroplasmy and the threshold identical, because they are a clonal expansion of effect, different tissues harboring the same mtDNA the original molecule. Mutations in protein-coding mutation may be affected to different degrees, thus genes of myogenic stem cells, presumably occur- explaining the frequent occurrence of oligosymp- ring after germ-layer differentiation, result in isolat- tomatic or asymptomatic carriers of the mutation ed myopathies11; 15 of the 17 known mutations in within a family. Selective organ involvement can also the cytochrome b gene fall into this category (however, occur, presumably as a result of skewed hetero- paternal inheritance of mtDNA in these cases must plasmy (i.e., disproportionately high levels of the be ruled out6). mutation in a given tissue), as in mitochondrial di- The pathogenesis of these disorders is unclear, abetes,12 mitochondrial cardiomyopathies,13 mi- although impaired production of ATP most likely tochondrial ,8 and mitochondrial deaf- has a central role. This concept has been borne out ness.14 by studies using cytoplasmic hybrid (“cybrid”) cell Although most mtDNA-related diseases are ma- cultures, which are established human cell lines ternally inherited, there are exceptions. The occur- that are depleted of their own mtDNA and then re- rence of giant deletions is almost always sporadic populated with the patient’s mitochondria contain- and probably takes place in oogenesis or early em- ing mutated genomes.16 bryogenesis. Oocytes from normal women contain The extraordinary variability of clinical presen- about 150,000 mtDNA molecules, some of which tations can largely be attributed to the peculiar rules may harbor deletions.15 A “bottleneck” between of mitochondrial genetics, especially heteroplasmy

n engl j med 348;26 www.nejm.org june 26, 2003 2661

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

The new england journal of medicine

Table 1. Clinical and Genetic Heterogeneity of Disorders Related to Mutations in Mitochondrial DNA (mtDNA).*

Mutation in Giant Deletions in Mutation in Ribosomal Mutation in Symptoms, Signs, and Findings mtDNA Transfer RNA RNA Messenger RNA

KSS PEO PS MERRF MELAS AID NARP MILS LHON Central – – – + + – – + – Ataxia + – – + + ¡ + ± – Myoclonus – – – + ± – – – – Psychomotor retardation – – – – – – – + – Psychomotor regression + – – ± + – – – – Hemiparesis and hemianopia – – – – + – – – – Cortical blindness – – – – + – – – – Migraine-like headaches – – – – + – – – – Dystonia – – – – + – – + ± Peripheral nervous system Peripheral neuropathy ± – – ± ± – + – – Muscle Weakness and exercise intolerance + + – + + – + + – Ophthalmoplegia + + ± – – – – – – Ptosis + + – – – – – – – Eye Pigmentary retinopathy + – – – – – + ± – Optic atrophy – – – – – – ± ± + Blood Sideroblastic anemia ± – + – – – – – – Endocrine system Diabetes mellitus ± – – – ± – – – – Short stature + – – + + – – – – Hypoparathyroidism ± – – – – – – – – Heart Conduction disorder + – – – ± – – – ± Cardiomyopathy ± – – – ± + – ± – Gastrointestinal system Exocrine pancreatic dysfunction ± – + – – – – – – Intestinal pseudo-obstruction – – – – + – – – – Ear, nose, and throat Sensorineural hearing loss ± – – + + + ± – – Kidney Fanconi’s syndrome ± – ± – ± – – – – Laboratory findings Lactic acidosis + ± + + + – – ± – Ragged-red fibers on muscle biopsy + + ± + + – – – – Mode of inheritance Maternal – – – + + – + + + Sporadic + + + – – – – – –

* Characteristic constellations of symptoms and signs are boxed. Plus signs indicate the presence of a symptom, sign, or finding; minus signs the absence of a symptom, sign, or finding; and plus–minus signs the possible presence of a symp- tom, sign, or finding. KSS denotes the Kearns–Sayre syndrome; PEO progressive external ophthalmoplegia; PS Pear- son’s syndrome; MERRF myoclonic epilepsy with ragged-red fibers; MELAS mitochondrial encephalomyopathy, lactic acidosis, and strokelike episodes; AID aminoglycoside-induced deafness; NARP neuropathy, ataxia, and retinitis pig- mentosa; MILS maternally inherited Leigh’s syndrome; and LHON Leber’s hereditary optic neuropathy.

2662 n engl j med 348;26 www.nejm.org june 26, 2003

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

mechanisms of disease and the threshold effect. For example, different mu- However, mutations in complex II have also been tational loads readily explain the different degrees associated with paragangliomas29,30 and pheochro- of severity between the neuropathy, ataxia, and reti- mocytomas.31 Although its genetic basis remains nitis pigmentosa syndrome and maternally inherit- unknown and it is likely to be heterogeneous, coen- ed Leigh’s syndrome, two encephalomyopathies zyme Q10 deficiency is emerging as an important caused by the same genetic defect in the ATPase 6 cause of autosomal recessive encephalomyopathies, gene.17 What is difficult to explain is the distinct ranging from predominantly myopathic forms “tissue proclivity” of seemingly similar mutations, (with recurrent myoglobinuria) to predominantly especially in the brain — for example, the strokelike encephalopathic forms (with ataxia and cerebellar episodes in MELAS, the myoclonus in MERRF, and atrophy).32 the pigmentary retinopathy in the Kearns–Sayre syn- drome. High concentrations of each mutation in mutations in ancillary proteins cerebral small vessels, in the dentate nucleus of the of the respiratory chain , and in the retinal pigment epithelium, No pathogenic mutations have been identified in respectively, do not explain why the mutation is in any nDNA-encoded subunits of complex III, IV, or that particular area of the brain. Conversely, many V, but defects of complexes III33,34 and IV35,36 have patients with mitochondrial diabetes have symp- been related to mutations in ancillary proteins re- toms even though they have a relatively small quired for the assembly or of cofactors. amount of mutation, suggesting that the pathoge- Cytochrome c oxidase deficiency is generalized in netic mechanism goes beyond a mere energy defi- disorders that are due to mutations in genes re- cit. For example, diabetes may be the result of sub- quired for the assembly of complex IV, but the en- tle interactions among oxidative energy levels and zyme defect and the symptoms are more severe in the glucose sensor,18 the NADH shuttle,19 and even certain tissues; for example, mutations in the sur- uncoupling proteins.20 Even more puzzling, muta- feit gene (SURF1) predominantly affect the brain and tions that are ubiquitous often have tissue-specific cause Leigh’s syndrome, mutations in a gene re- effects (e.g., deafness due to mutations in 12S rRNA, quired for the synthesis of cytochrome oxidase cardiopathy due to mutations in tRNAIle, and op- (SCO2) or in cytochrome c oxidase 15 (COX15)37 cause in- tic atrophy due to mutations in ND genes). Studies fantile cardiomyopathy in addition to brain disease, of animal models with mtDNA mutations (“mito- and mutations in cytochrome c oxidase 10 (COX10) and mice”)21,22 may provide some answers to these another gene for the synthesis of cytochrome oxi- riddles. dase (SCO1) affect kidney and tissues, respec- tively. Although pathogenic mutations may occur in respiratory-chain disorders structural subunits of complexes III, IV, and V, they due to defects in ndna may be lethal in utero because there is no metabolic compensation, and complex V is the sole site of ox- In recent years, interest has shifted toward mende- idative phosphorylation38 (Fig. 2B). This concept lian genetics in . This shift would apply only to the “all-or-none” type of men- is understandable, not only because most of the delian inheritance, since mutations in mtDNA- 75-plus respiratory-chain proteins are encoded by encoded components of complexes III, IV, and V do nDNA (Fig. 2B), but also because proper assembly indeed occur but are expressed incompletely be- and function of respiratory chain complexes require cause of heteroplasmy. approximately 60 additional (ancillary) nucleus- encoded proteins. Mutations in these genes can also defects in intergenomic signaling cause mitochondrial disease.23 affecting respiratory function In the course of , mitochondria lost their mutations in structural components independence, and mtDNA is now the slave of of the respiratory chain nDNA, depending on numerous nucleus-encoded Mutations in structural components of the respira- factors for its integrity and replication.39 Mutations tory chain have thus far been found only in complex- in these factors affect mtDNA directly, either quanti- es I24 and II25-27 and have generally been associat- tatively or qualitatively, and cause diseases that are ed with severe neurologic disorders of childhood, inherited as mendelian traits. such as Leigh’s syndrome and leukodystrophy.28 A quantitative alteration is exemplified by abnor-

n engl j med 348;26 www.nejm.org june 26, 2003 2663

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved. The new england journal of medicine

mal reductions in the number of mtDNA molecules may offer new approaches to therapeutic interven- (both per cell and per organelle) — mtDNA-deple- tion (e.g., lowering blood thymidine concentrations tion syndromes. A qualitative alteration is exempli- in patients with mitochondrial neurogastrointesti- fied by multiple deletions (in contrast to the single nal encephalomyopathy). mtDNA deletions in sporadic Kearns–Sayre syn- drome, progressive external ophthalmoplegia, and defects of the membrane milieu Pearson’s syndrome). Except for cytochrome c, which is located in the in- Ophthalmoplegia is the clinical hallmark of mul- termembrane space, all components of the respira- tiple mtDNA deletions,40 but patients with autoso- tory chain are embedded in the lipid milieu of the in- mal dominant progressive external ophthalmople- ner mitochondrial membrane, which is composed gia often have proximal limb weakness, peripheral predominantly of . Cardiolipin is not neuropathy, sensorineural hearing loss, cataracts, merely a scaffold but is an integral and indispen- endocrine dysfunction, and severe depression.41,42 sable part of some respiratory-chain components.53 Autosomal recessive progressive external ophthal- It therefore stands to reason that defects in cardio- moplegia has two main clinical presentations: one lipin would cause respiratory-chain dysfunction and consists of cardiomyopathy and ophthalmoplegia,43 mitochondrial disease. This concept is exemplified and the other of peripheral neuropathy, gastroin- by an X-linked disorder, Barth syndrome (mitochon- testinal dysmotility, and leukoencephalopathy (mi- drial myopathy, cardiomyopathy, growth retarda- tochondrial neurogastrointestinal encephalomy- tion, and leukopenia).54 The mutated gene in Barth opathy).44 syndrome, G4.5, encodes a family of acyl–coenzyme Both quantitative and qualitative defects may re- A synthetases (tafazzins) that must have an im- sult from impairment of the integrity of the mito- portant role in cardiolipin synthesis, because car- chondrial genome. Such impairment can be direct diolipin concentrations are markedly decreased in (e.g., affecting proteins required for the replication skeletal and cardiac muscle and in platelets from and maintenance of mtDNA) or indirect (e.g., af- affected patients.55 fecting proteins required to maintain nucleotide pools in mitochondria). For example, some families disorders with indirect with autosomal dominant progressive external involvement of the ophthalmoplegia have mutations in Twinkle, a mi- respiratory chain tochondrial protein similar to bacteriophage T7 /, whereas other families have mu- Indirect involvement of mitochondria has been tations in the mitochondrial adenine nucleotide documented or suggested in many conditions, in- translocator 1 (ANT1).45,46 Mutations in mitochon- cluding normal aging, late-onset neurodegenera- drial-specific DNA g have been associ- tive diseases, and cancer. However, the precise role ated with both dominant and recessive multiple- of mitochondrial dysfunction in these conditions deletion disorders.47 remains controversial. Mitochondrial neurogastrointestinal encephalo- myopathy is clearly due to the loss of function of thy- defects of mitochondrial protein midine , resulting in markedly in- importation creased concentrations of thymidine in the blood.48 Cytosolic proteins destined for mitochondria have Thymidine phosphorylase is not a mitochondrial mitochondrial targeting signals that enable them protein, yet it appears to have a selective effect on to be routed to the appropriate compartment within mitochondrial nucleotide pools required for main- the organelle, where they are then refolded into an taining the integrity and abundance of mtDNA. active configuration.56 Although a number of mu- The role of is bolstered by the pathoge- tations in mitochondrial targeting signals have been nicity of the ANT1 mutations and by recent findings found, few errors in the importation machinery it- that mutations in mitochondrial thymidine ki- self are known, perhaps because they may be le- nase49,50 and deoxyguanosine kinase51,52 are asso- thal.57 However, at least one such defect has been ciated with the myopathic and hepatocerebral forms identified, the deafness–dystonia syndrome (Mohr– of mtDNA depletion. Knowledge of these mutations Tranebjaerg syndrome), an X-linked recessive dis- makes prenatal diagnosis feasible for some families order characterized by progressive neurosensory with the infantile mtDNA-depletion syndromes and deafness, dystonia, cortical blindness, and psychiat-

2664 n engl j med 348;26 www.nejm.org june 26, 2003

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved. mechanisms of disease ric symptoms,58 features that are strikingly similar mon pathogenic mechanism in aging and in Alz- to those of the primary mitochondrial diseases. This heimer’s disease, amyotrophic lateral sclerosis, disorder is due to mutations in TIMM8A, encoding Huntington’s disease, progressive supranuclear pal- the deafness–dystonia protein (DDP1), a compo- sy, and Parkinson’s disease.66 Rearrangements of nent of the mitochondrial-protein–import machin- and point mutations in mtDNA may accumulate ery in the intermembrane space. According to a over time, eventually surpassing the pathogenic recent report, an autosomal dominant form of he- threshold in multiple tissues (aging) or in specific reditary spastic paraplegia is associated with mu- areas of the (neurodegener- tations in the mitochondrial import chaperonin ative disorders). Although mtDNA mutations do HSP60.59 increase in postmitotic tissues of healthy elderly persons, there is no convincing evidence that they defects in mitochondrial motility reach deleterious levels.67 The role of mtDNA mu- Mitochondria are not static organelles; they are pro- tations in neurodegenerative disorders, whether as pelled within the cell by energy-requiring the cumulative effect of generic mutations or as the along cytoskeletal microtubule “rails.”60 Mutations specific effect of putative pathogenic mutations, is in a gene encoding a mitochondrial -relat- even more controversial.68 ed guanosine triphosphatase (OPA1) are associat- ed with an autosomal dominant form of optic at- therapeutic approaches rophy, which together with Leber’s hereditary optic neuropathy, is a major cause of blindness in young Therapy for mitochondrial diseases is woefully in- adults.61,62 adequate. In the absence of a clear understanding of basic pathogenetic mechanisms, treatments have neurodegenerative diseases been palliative or have involved the indiscriminate A few neurodegenerative disorders are due to muta- administration of vitamins, cofactors, and oxygen- tions in proteins that target the mitochondria. These radical scavengers, with the aim of mitigating, post- include Friedreich’s ataxia, at least one form of he- poning, or circumventing the postulated damage to reditary spastic paraplegia, and Wilson’s disease. the respiratory chain.69 Friedreich’s ataxia is due to expansion of trinucleo- Rational therapies, on the whole, remain elusive, tide (GAA) repeats in the FRDA gene, which encodes but the broad outlines of such approaches are be- , a mitochondrion-targeted protein involved ginning to emerge. For the mtDNA-related disor- in iron .63 Excessive free iron resulting ders, the most promising approach is to reduce the from decreased concentrations of frataxin may ratio of mutated to wild-type genomes (“gene shift- damage proteins containing iron-sulfur groups, in- ing”).70 Such shifting might be accomplished by cluding complexes I, II, and III, and , a pharmacologic,71 physiological,70 or even surgi- Krebs-cycle . An autosomal recessive form cal72 approaches. Genetic approaches to treatment of hereditary spastic paraplegia is due to mutations are particularly daunting, since they will require tar- in the SPG7 gene, which encodes paraplegin, a mi- geting not only affected cells, but also the organelles tochondrial protein similar to metalloproteas- within them. Nevertheless, some headway is being es.64 Impairment of the respiratory chain is suggest- made here as well, although it is limited to in vitro ed by the presence of ragged-red fibers and fibers systems. These include the selective destruction of deficient in cytochrome c oxidase in muscle from af- mutant mtDNAs through importation of a restric- fected patients. In Wilson’s disease, an autosomal tion enzyme into mitochondria,73 the replacement recessive disease characterized by movement dis- of a mutant mtDNA-encoded protein with a geneti- order and liver failure, there are mutations of the cally engineered normal equivalent expressed from ATP7B gene, which encodes a -transporting the nucleus (allotopic expression),74-76 the replace- ATPase, one isoform of which is localized in mito- ment of a defective respiratory-chain complex with chondria.65 The pathogenesis of Wilson’s disease a cognate complex from another organism,77 and may involve either direct damage to copper-contain- the importation of a normal tRNA to compensate ing , such as cytochrome c oxidase, or more for a mutation in the corresponding organellar generic oxidative damage to the cell owing to the tRNA.78 accumulation of copper. As for nuclear mutations, the problems of gene Mitochondrial dysfunction may be a final com- therapy are the same as those encountered in oth-

n engl j med 348;26 www.nejm.org june 26, 2003 2665

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved. The new england journal of medicine

er mendelian disorders. However, pharmacologic dria.82 Only one other patient with Luft’s syndrome treatments might be of use in special cases, such as has been identified.83 Yet nowadays, the possibility lowering thymidine concentrations in patients with of mitochondrial dysfunction needs to be taken into mitochondrial neurogastrointestinal encephalomy- account by every medical subspecialty. Although opathy.79 Reports that cytochrome c oxidase defi- caution is required in discriminating primary from ciency can be reversed by the supplementation of secondary mitochondrial involvement, progress in copper in cultured cells with a mutation in SCO2, a this field has been striking enough to amply justify copper , suggest that copper supplemen- the term “mitochondrial medicine,”84 used by Luft tation may be useful in some patients.80,81 in the title of a review written 32 years after he first The concept of mitochondrial disease was intro- described “his” syndrome. duced 41 years ago, when Luft and coworkers de- Supported in part by grants from the National Institutes of Health scribed a young woman with severe, nonthyroidal (NS11766, NS28828, NS39854, and HD32062) and the Muscular Dystrophy Association. hypermetabolism due to loose coupling of oxida- We are indebted to Drs. Eduardo Bonilla, Michio Hirano, and tion and phosphorylation in muscle mitochon- Lewis P. Rowland for their collaboration and helpful suggestions.

references 1. Elston T, Wang H, Oster G. Energy trans- 13. Hirano M, Davidson MM, DiMauro S. JM, Smeitink JA. Respiratory chain complex I duction in ATP synthase. Nature 1998;391: Mitochondria and the heart. Curr Opin Car- deficiency. Am J Med Genet 2001;106:37-45. 510-3. diol 2001;16:201-10. 25. Bourgeron T, Rustin P, Chretien D, et al. 2. Noji H, Yoshida M. The rotary 14. Fischel-Ghodsian N. Mitochondrial mu- Mutation of a nuclear succinate dehydro- of the cell, ATP synthase. J Biol Chem 2001; tations and hearing loss: paradigm for mi- genase gene results in mitochondrial respi- 276:1665-8. tochondrial genetics. Am J Hum Genet ratory chain deficiency. Nat Genet 1995;11: 3. Chinnery PF, Turnbull DM. Epidemiolo- 1998;62:15-9. 144-9. gy and treatment of mitochondrial disorders. 15. Chen X, Prosser R, Simonetti S, Sadlock 26. Parfait B, Chretien D, Rotig A, Marsac C, Am J Med Genet 2001;106:94-101. J, Jagiello G, Schon EA. Rearranged mito- Munnich A, Rustin P. Compound heterozy- 4. Holt IJ, Harding AE, Morgan-Hughes JA. chondrial genomes are present in human gous mutations in the flavoprotein gene of Deletions of muscle mitochondrial DNA in oocytes. Am J Hum Genet 1995;57:239-47. the respiratory chain complex II in a patient patients with mitochondrial myopathies. Na- 16. King MP, Attardi G. Human cells lack- with . Hum Genet 2000; ture 1988;331:717-9. ing mtDNA: repopulation with exogenous 106:236-43. 5. Wallace DC, Singh G, Lott MT, et al. Mi- mitochondria by complementation. Science 27. Birch-Machin MA, Taylor RW, Cochran tochondrial DNA mutation associated with 1989;246:500-3. B, Ackrell BA, Turnbull DM. Late-onset op- Leber’s hereditary optic neuropathy. Science 17. Tatuch Y, Christodoulou J, Feigenbaum tic atrophy, ataxia, and myopathy associated 1988;242:1427-30. A, et al. Heteroplasmic mtDNA mutation with a mutation of a complex II gene. Ann 6. Schwartz M, Vissing J. Paternal inherit- (T→G) at 8993 can cause Leigh disease Neurol 2000;48:330-5. ance of mitochondrial DNA. N Engl J Med when the percentage of abnormal mtDNA is 28. Shoubridge EA. Nuclear genetic defects 2002;347:576-80. high. Am J Hum Genet 1992;50:852-8. of oxidative phosphorylation. Hum Mol Gen- 7. Servidei S. Mitochondrial encephalomy- 18. Matschinsky FM, Glaser B, Magnuson et 2001;10:2277-84. opathies: gene mutation. Neuromuscul Dis- MA. Pancreatic beta-cell : clos- 29. Baysal BE, Ferrell RE, Willett-Brozick ord 2002;12:101-10. ing the gap between theoretical concepts JE, et al. Mutations in SDHD, a mitochondri- 8. DiMauro S, Bonilla E. Mitochondrial and experimental realities. Diabetes 1998; al complex II gene, in hereditary paragan- encephalomyopathies. In: Rosenberg RN, 47:307-15. glioma. Science 2000;287:848-51. Prusiner S, DiMauro S, Barchi RL, eds. The 19. Eto K, Tsubamoto Y, Terauchi Y, et al. 30. Niemann S, Muller U. Mutations in molecular and genetic basis of neurological Role of NADH shuttle system in glucose- SDHC cause autosomal dominant paragan- disease. 2nd ed. Boston: Butterworth–Heine- induced activation of mitochondrial metab- glioma, type 3. Nat Genet 2000;26:268-70. mann, 1996:201-35. olism and insulin secretion. Science 1999; 31. Gimm O, Armanios M, Dziema H, Neu- 9. DiMauro S, Hirano M, Kaufmann P, et al. 283:981-5. mann HP, Eng C. Somatic and occult germ- Clinical features and genetics of myoclonic 20. Langin D. Diabetes, insulin secretion, line mutations in SDHD, a mitochondrial epilepsy with ragged red fibers. In: Fahn S, and the pancreatic beta-cell mitochondrion. complex II gene, in nonfamilial pheochro- Frucht SJ, eds. Myoclonus and paroxysmal N Engl J Med 2001;345:1772-4. [Erratum, mocytoma. Cancer Res 2000;60:6822-5. dyskinesia. Philadelphia: Lippincott Wil- N Engl J Med 2002;346:634.] 32. Musumeci O, Naini A, Slonim AE, et al. liams & Wilkins, 2002:217-29. 21. Inoue K, Nakada K, Ogura A, et al. Gen- Familial cerebellar ataxia with muscle coen- 10. Carelli V. Leber’s hereditary optic neu- eration of mice with mitochondrial dysfunc- zyme Q10 deficiency. Neurology 2001;56: ropathy. In: Schapira AHV, DiMauro S, eds. tion by introducing mouse mtDNA carrying 849-55. Mitochondrial disorders in neurology. 2nd a deletion in zygotes. Nat Genet 2000;26: 33. de Lonlay P, Valnot I, Barrientos A, et al. ed. Boston: Butterworth–Heinemann, 2002: 176-81. A mutant mitochondrial respiratory chain 115-42. 22. Sligh JE, Levy SE, Waymire KG, et al. assembly protein causes complex III defi- 11. Andreu AL, Hanna MG, Reichmann H, Maternal germ-line transmission of mutant ciency in patients with tubulopathy, enceph- et al. Exercise intolerance due to mutations mtDNAs from embryonic stem cell-derived alopathy and liver failure. Nat Genet 2001; in the cytochrome b gene of mitochondrial chimeric mice. Proc Natl Acad Sci U S A 29:57-60. DNA. N Engl J Med 1999;341:1037-44. 2000;97:14461-6. 34. Visapaa I, Fellman V, Vesa J, et al. 12. Kadowaki T, Kadowaki H, Mori Y, et al. 23. Shoubridge EA. Nuclear gene defects in GRACILE syndrome, a lethal metabolic A subtype of diabetes mellitus associated respiratory chain disorders. Semin Neurol disorder with , is caused by a with a mutation of mitochondrial DNA. 2001;21:261-7. in BCS1L. Am J Hum Genet N Engl J Med 1994;330:962-8. 24. Triepels RH, Van Den Heuvel LP, Trijbels 2002;71:863-76.

2666 n engl j med 348;26 www.nejm.org june 26, 2003

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved. mechanisms of disease

35. Shoubridge EA. Cytochrome c oxidase 49. Saada A, Shaag A, Mandel H, Nevo Y, al. Spastic paraplegia and OXPHOS impair- deficiency. Am J Med Genet 2001;106:46- Eriksson S, Elpeleg O. Mutant mitochondri- ment caused by mutations in paraplegin, a 52. al thymidine in mitochondrial DNA nuclear-encoded mitochondrial metallopro- 36. Zeviani M. The expanding spectrum of depletion myopathy. Nat Genet 2001;29: tease. Cell 1998;93:973-83. nuclear gene mutations in mitochondrial 342-4. 65. Lutsenko S, Cooper MJ. Localization of disorders. Semin Cell Dev Biol 2001;12:407- 50. Mancuso M, Salviati L, Sacconi S, et al. the Wilson’s disease protein to mi- 16. Mitochondrial DNA depletion: mutations in tochondria. Proc Natl Acad Sci U S A 1998; 37. Antonicka H, Mattman A, Carlson CG, gene with myopathy and 95:6004-9. et al. Mutations in COX15 produce a defect in SMA. Neuropathy 2002;59:1197-202. 66. Beal MF. Mitochondria in neurodegen- the mitochondrial heme biosynthetic path- 51. Mandel H, Szargel R, Labay V, et al. The eration. In: Desnuelle C, DiMauro S, eds. way, causing early-onset fatal hypertrophic gene is mutated in Mitochondrial disorders: from pathophysi- cardiomyopathy. Am J Hum Genet 2003;72: individuals with depleted hepatocerebral mi- ology to acquired defects. New York: Spring- 101-14. tochondrial DNA. Nat Genet 2001;29:337- er, 2002:17-35. 38. Sue CM, Schon EA. Mitochondrial res- 41. [Erratum, Nat Genet 2001;29:491.] 67. DiMauro S, Tanji K, Bonilla E, Pallotti F, piratory chain diseases and mutations in nu- 52. Salviati L, Sacconi S, Mancuso M, et al. Schon EA. Mitochondrial abnormalities in clear DNA: a promising start? Brain Pathol Mitochondrial DNA depletion and dGK gene muscle and other aging cells: classification, 2000;10:442-50. mutations. Ann Neurol 2002;52:311-7. causes, and effects. Muscle Nerve 2002;26: 39. Hirano M, Marti R, Ferreiro-Barros C, et 53. Schlame M, Rua D, Greenberg ML. The 597-607. al. Defects of intergenomic communication: biosynthesis and functional role of cardio- 68. Schon EA, Manfredi G. Neuronal de- autosomal disorders that cause multiple de- lipin. Prog Lipid Res 2000;39:257-88. generation and mitochondrial dysfunction. letions and depletion of mitochondrial DNA. 54. Barth PG, Wanders RJ, Vreken P, Jans- J Clin Invest 2003;111:303-12. Semin Cell Dev Biol 2001;12:417-27. sen EA, Lam J, Baas F. X-linked cardioskele- 69. DiMauro S, Hirano M, Schon EA. Mito- 40. Hirano M, DiMauro S. ANT1, Twinkle, tal myopathy and (Barth syn- chondrial encephalomyopathies: therapeutic POLG, and TP: new genes open our eyes to drome) (MIM 302060). J Inherit Metab Dis approaches. Neurol Sci 2000;21:S901-S908. ophthalmoplegia. Neurology 2001;57:2163- 1999;22:555-67. 70. Taivassalo T, Fu K, Johns T, Arnold D, 5. 55. Schlame M, Towbin JA, Heerdt PM, Jeh- Karpati G, Shoubridge EA. Gene shifting: 41. Servidei S, Zeviani M, Manfredi G, et al. le R, DiMauro S, Blanck TJ. Deficiency of te- a novel therapy for mitochondrial myopa- Dominantly inherited mitochondrial myopa- tralinoleoyl-cardiolipin in Barth syndrome. thy. Hum Mol Genet 1999;8:1047-52. thy with multiple deletions of mitochondrial Ann Neurol 2002;51:634-7. 71. Manfredi G, Gupta N, Vazquez-Memije DNA: clinical, morphologic, and biochemi- 56. Okamoto K, Brinker A, Paschen SA, et ME, et al. induces a decrease in cal studies. Neurology 1991;41:1053-9. al. The protein import motor of mitochon- the cellular content of a pathogenic muta- 42. Suomalainen A, Majander A, Wallin M, dria: a targeted molecular ratchet driving tion in the human mitochondrial ATPase 6 et al. Autosomal dominant progressive ex- unfolding and translocation. EMBO J 2002; gene. J Biol Chem 1999;274:9386-91. ternal ophthalmoplegia with multiple dele- 21:3659-71. 72. Clark KM, Bindoff LA, Lightowlers RN, tions of mtDNA: clinical, biochemical, and 57. Fenton WA. Mitochondrial protein trans- et al. Reversal of a mitochondrial DNA de- molecular genetic features of the 10q-linked port — a system in search of mutations. Am fect in human skeletal muscle. Nat Genet disease. Neurology 1997;48:1244-53. J Hum Genet 1995;57:235-8. 1997;16:222-4. 43. Bohlega S, Tanji K, Santorelli FM, Hira- 58. Roesch K, Curran SP, Tranebjaerg L, 73. Tanaka M, Borgeld HJ, Zhang J, et al. no M, al-Jishi A, DiMauro S. Multiple mito- Koehler CM. Human deafness dystonia syn- Gene therapy for mitochondrial disease by chondrial DNA deletions associated with drome is caused by a defect in assembly of delivering restriction endonuclease SmaI autosomal recessive ophthalmoplegia and the DDP1/TIMM8a-TIMM13 complex. Hum into mitochondria. J Biomed Sci 2002;9: severe cardiomyopathy. Neurology 1996;46: Mol Genet 2002;11:477-86. 534-41. 1329-34. 59. Hansen JJ, Durr A, Cournu-Rebeix I, et 74. Manfredi G, Fu J, Ojaimi J, et al. Rescue 44. Nishino I, Spinazzola A, Papadimitriou al. Hereditary spastic paraplegia SPG13 is of a deficiency in ATP synthesis by transfer A, et al. Mitochondrial neurogastrointesti- associated with a mutation in the gene en- of MTATP6, a mitochondrial DNA-encoded nal encephalomyopathy: an autosomal re- coding the mitochondrial chaperonin gene, to the nucleus. Nat Genet 2002;30: cessive disorder due to thymidine phos- Hsp60. Am J Hum Genet 2002;70:1328-32. 394-9. phorylase mutations. Ann Neurol 2000;47: 60. Boldogh IR, Yang HC, Nowakowski 75. Guy J, Qi X, Pallotti F, et al. Rescue of a 792-800. WD, et al. Arp2/3 complex and actin dynam- mitochondrial deficiency causing Leber he- 45. Kaukonen J, Juselius JK, Tiranti V, et al. ics are required for actin-based mitochon- reditary optic neuropathy. Ann Neurol 2002; Role of adenine nucleotide translocator 1 in drial motility in yeast. Proc Natl Acad Sci 52:534-42. mtDNA maintenance. Science 2000;289: U S A 2001;98:3162-7. 76. Ojaimi J, Pan J, Santra S, Snell WJ, Schon 782-5. 61. Alexander C, Votruba M, Pesch UE, et al. EA. An algal nucleus-encoded subunit of 46. Spelbrink JN, Li FY, Tiranti V, et al. Hu- OPA1, encoding a dynamin-related GTPase, mitochondrial ATP synthase rescues a de- man mitochondrial DNA deletions associat- is mutated in autosomal dominant optic at- fect in the analogous human mitochondrial- ed with mutations in the gene encoding rophy linked to 3q28. Nat encoded subunit. Mol Biol Cell 2002;13: Twinkle, a phage T7 gene 4-like protein lo- Genet 2000;26:211-5. 3836-44. calized in mitochondria. Nat Genet 2001;28: 62. Delettre C, Lenaers G, Griffoin JM, et al. 77. Bai Y, Hajek P, Chomyn A, et al. Lack of 223-31. [Erratum, Nat Genet 2001;29:100.] Nuclear gene OPA1, encoding a mitochon- complex I activity in human cells carrying a 47. Van Goethem G, Dermaut B, Lofgren A, drial dynamin-related protein, is mutated in mutation in MtDNA-encoded ND4 subunit Martin JJ, Van Broeckhoven C. Mutation of dominant optic atrophy. Nat Genet 2000;26: is corrected by the Saccharomyces cerevisi- POLG is associated with progressive external 207-10. ae NADH-quinone oxidoreductase (NDI1) ophthalmoplegia characterized by mtDNA 63. Cavadini P, O’Neill HA, Benada O, Isaya gene. J Biol Chem 2001;276:38808-13. deletions. Nat Genet 2001;28:211-2. G. Assembly and iron-binding properties of 78. Kolesnikova OA, Entelis NS, Mireau H, 48. Nishino I, Spinazzola A, Hirano M. Thy- human frataxin, the protein deficient in Fox TD, Martin RP, Tarassov IA. Suppres- midine phosphorylase gene mutations in Friedreich ataxia. Hum Mol Genet 2002;11: sion of mutations in mitochondrial DNA by MNGIE, a human mitochondrial disorder. 217-27. tRNAs imported from the cytoplasm. Sci- Science 1999;283:689-92. 64. Casari G, De Fusco M, Ciarmatori S, et ence 2000;289:1931-3.

n engl j med 348;26 www.nejm.org june 26, 2003 2667

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved. mechanisms of disease

79. Spinazzola A, Marti R, Nishino I, et al. 81. Salviati L, Hernandez-Rosa E, Walker and morphological study. J Clin Invest 1962; Altered thymidine metabolism due to de- WF, et al. Copper supplementation restores 41:1776-804. fects of thymidine phosphorylase. J Biol cytochrome c oxidase activity in cultured cells 83. DiMauro S, Bonilla E, Lee CP, et al. Chem 2002;277:4128-33. from patients with SCO2 mutations. Bio- Luft’s disease: further biochemical and ultra- 80. Jaksch M, Paret C, Stucka R, et al. Cyto- chem J 2002;363:321-7. structural studies of skeletal muscle in the chrome c oxidase deficiency due to mutations 82. Luft R, Ikkos D, Palmieri G, Ernster L, second case. J Neurol Sci 1976;27:212-32. in SCO2, encoding a mitochondrial copper- Afzelius B. A case of severe hypermetabo- 84. Luft R. The development of mitochon- binding protein, is rescued by copper in hu- lism of nonthyroid origin with a defect in the drial medicine. Proc Natl Acad Sci U S A man myoblasts. Hum Mol Genet 2001;10: maintenance of mitochondrial respiratory 1994;91:8731-8. 3025-35. control: a correlated clinical, biochemical, Copyright © 2003 Massachusetts Medical Society.

full text of all journal articles on the world wide web

Access to the complete text of the Journal on the Internet is free to all subscribers. To use this Web site, subscribers should go to the Journal’s home page (http://www.nejm.org) and register by entering their names and subscriber numbers as they appear on their mailing labels. After this one-time registration, subscribers can use their passwords to log on for electronic access to the entire Journal from any computer that is connected to the Internet. Features include a library of all issues since January 1993 and abstracts since January 1975, a full-text search capacity, and a personal archive for saving articles and search results of interest. All articles can be printed in a format that is virtually identical to that of the typeset pages. Beginning six months after publication the full text of all original articles and special articles is available free to nonsubscribers who have completed a brief registration.

2668 n engl j med 348;26 www.nejm.org june 26, 2003

Downloaded from www.nejm.org by GEOFFREY K. LIGHTHALL MD on September 11, 2003. Copyright © 2003 Massachusetts Medical Society. All rights reserved.