REVIEWS Disturbed mitochondrial dynamics and neurodegenerative disorders Florence Burté, Valerio Carelli, Patrick F. Chinnery and Patrick Yu-Wai-Man Abstract | Mitochondria form a highly interconnected tubular network throughout the cell via a dynamic process, with mitochondrial segments fusing and breaking apart continuously. Strong evidence has emerged to implicate disturbed mitochondrial fusion and fission as central pathological components underpinning a number of childhood and adult-onset neurodegenerative disorders. Several proteins that regulate the morphology of the mitochondrial network have been identified, the most widely studied of which are optic atrophy 1 and mitofusin 2. Pathogenic mutations that disrupt these two pro-fusion proteins cause autosomal dominant optic atrophy and axonal Charcot–Marie–Tooth disease type 2A, respectively. These disorders predominantly affect specialized neurons that require precise shuttling of mitochondria over long axonal distances. Considerable insight has also been gained by carefully dissecting the deleterious consequences of imbalances in mitochondrial fusion and fission on respiratory chain function, mitochondrial quality control (mitophagy), and programmed cell death. Interestingly, these cellular processes are also implicated in morecommon complex neurodegenerative disorders, such as Alzheimer disease and Parkinson disease, indicating a common pathological thread and a close relationship with mitochondrial structure, function and localization. Understanding how these fundamental processes become disrupted will prove crucial to the development of therapies for the growing number of neurodegenerative disorders linked to disturbed mitochondrial dynamics. Burté, F. et al. Nat. Rev. Neurol. advance online publication 9 December 2014; doi:10.1038/nrneurol.2014.228

Introduction

Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK (F.B., P.F.C., P.Y.-W.-M.). IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Via Altura 3, 40139 Bologna, Italy (V.C.). Correspondence to: P.Y.-W.-M. patrick.yu-wai-man@ ncl.ac.uk

Mitochondria do not exist as isolated organelles fixed at predetermined positions within the cell’s structure. Instead, they display a high degree of interconnec­tivity and plasticity, which are largely dictated by the cell’s metabolic demands under prevailing physiological conditions.1 The morphology of the mitochondrial network is in a constant state of flux, influenced by the delicate balance between opposing fusional and fissional forces. The main players in this intricate and tightly coordinated process were first identified in seminal experiments using yeast models. These mediators of mitochondrial dynamics have been highly conserved throughout evolution, which is in keeping with the critical regulatory roles of these proteins in both simple and complex organisms. Helped by the genomics revolution of the past decade, perturbation of mitochondrial dynamics has emerged as the central pathophysiological mechanism underpinning a broad range of human disorders, often with strikingly overlapping clinical features (Table 1). Unsurprisingly, pathogenic mutations have been identified in several pro-fusion and pro-fission nuclear genes, with disease phenotypes ranging from severe, early-onset and invariably lethal encephalomyopathies, through isolated optic atrophy and peripheral neuropathy, to more-complex late-onset multisystemic neuromuscular disorders. Competing interests The authors declare no competing interests.

Mutations in the pro-fusion genes optic atrophy 1 (OPA1) and mitofusin 2 (MFN2) were initially reported in families with autosomal dominant optic atrophy (DOA; OMIM #605290) and axonal Charcot–Marie–Tooth disease type 2A (CMT2A; OMIM #609260), respectively.2–4 By focusing on these two diseases, this Review will critically appraise the fundamental mechanisms linking disturbed mitochondrial dynamics with the development and progression of neurological deficits in both monogenic and complex neurodegenerative disorders.

Mitochondrial network dynamics Balancing mitochondrial fusion and fission OPA1 is an inner mitochondrial membrane protein, whereas MFN1 and MFN2 are embedded within the outer mitochondrial membrane.5,6 The two mitofusins are structurally and functionally complementary to OPA1 and, together, they choreograph a precise chain of mechanical events that sequentially trigger fusion of the outer, then the inner mitochondrial membranes (Figure 1).7,8 All three proteins belong to a large family of dynaminrelated mechanoenzymes characterized by a highly conserved dynamin GTPase domain, which is central to their multifaceted cellular roles and interactions.6 OPA1 is firmly anchored, via its transmembrane domain, within the narrow junctional regions that insulate the mitochondrial cristae in a zipper-like fashion from the rest of the intermembrane space.9,10 This specific

NATURE REVIEWS | NEUROLOGY

ADVANCE ONLINE PUBLICATION  |  1 © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS Key points ■■ Mitochondria do not exist as isolated organelles; instead, they form a highly interconnected tubular network throughout the cell ■■ The state of this dynamic mitochondrial network under both physiological and disease conditions is dictated by the balance between mitochondrial pro-fusion and pro-fission forces ■■ Optic atrophy protein 1 (OPA1) and mitofusin 2 (MFN2) are two important protein mediators of mitochondrial fusion ■■ Pathogenic OPA1 and MFN2 mutations cause autosomal dominant optic atrophy and axonal Charcot–Marie–Tooth disease type 2A, respectively ■■ Mounting evidence implicates disturbed mitochondrial dynamics in the pathogenesis of complex neurodegenerative disorders such as Alzheimer disease, Parkinson disease and Huntington disease ■■ Although treatment options for disorders of mitochondrial dynamics are currently limited, the future looks promising for the development of novel neuroprotective strategies and innovative gene therapy approaches

Table 1 | Human disorders associated with disturbed mitochondrial dynamics Gene

Locus

OMIM #

Phenotypes

1p36.22

609260

Charcot–Marie–Tooth disease axonal form type 2A (CMT2A)108 Hereditary motor and sensory neuropathy type 6 (HMSN6)111,132

Autosomal dominant Mitofusin 2 (MFN2)

601152 Optic atrophy 1 (OPA1)

3q28–q29

165500

Isolated optic atrophy and syndromic forms of dominant optic atrophy90,91,103

Ganglioside-induced differentiation associated protein 1 (GDAP1)

8q13.1–12.3

607831

Charcot–Marie–Tooth disease axonal form type 2K (CMT2K)199

Dynamin-related protein 1 (DRP1)

12p11.21

603850

Severe infantile neurodegenerative disease200,201

Optic atrophy 3 (OPA3)

19q13.2–q13.3

165300

Optic atrophy and premature cataracts202

GDAP1

8q13.1–12.3

214400

Charcot–Marie–Tooth disease demyelinating form type 4A (CMT4A)203

OPA3

19q13.2–q13.3

258501

3-methylglutaconic aciduria type III (Costeff syndrome)204

Autosomal recessive

Permission obtained from Springer © Yu-Wai-Man, P. et al. in Mitochondrial Disorders Caused by Nuclear Genes (ed. Wong, L.‑J. C.) 141–161 (2013).5

transmembrane protein domain is followed by a series of coiled-coil segments that allow OPA1 to homopolymerize into cylindrical tubular structures, which in turn facilitate the tethering of opposing membranes. A pathological reduction in OPA1 levels results in fragmentation of the mitochondrial network due to unopposed mitochondrial fission (Figure 2).11,12 Although much attention has focused on these main pro-fusion and pro-fission proteins (Table 2), a host of equally critical intermediaries are starting to emerge. These novel mitochondrial dynamic proteins include molecules that promote fission, such as endophilin‑B1, mitochondrial fission factor (MFF), mitochondrial dynamics proteins MID49 and MID51, mitochondrial protein 18 kDa (MTP18; also known as mitochondrial fission process protein 1), and mitochondrial fission regulator 1

(MTFR1).13–16 Endophilin‑B1 has also been linked with the regulation of autophagy.17

Proteolytic control Mitochondrial fusion is modulated by the proteolytic processing of two major mediators, OPA1 and MFN2. The degradation of MFN2 through ubiquitination is dependent on PTEN-induced putative kinase 1 (PINK1, also called mitochondrial serine/threonine-protein kinase PINK1), and this process is tightly linked to the loss of mitochondrial membrane potential and the autophagic process. OPA1 exists as eight isoforms that arise from alternative splicing of exons 4, 4b and 5b.18,19 Following its import into the mitochondrial inter­ membrane space, OPA1 is first processed by the matrix metalloproteases, which remove the mitochondrial targeting signal, and then by proteases of the mitochondrial AAA+ (ATPases associated with diverse cellular activities) superfamily, which leads to the formation of long and short forms of OPA1.20,21 The nature of the proteases involved in the maturation steps of OPA1 remains an area of intense investigation. The mitochondrial AAA+ proteases AFG3-like protein 2 (AFG3L2) and para­ plegin cleave OPA1 at site S1 within exon 5,20,22 whereas the ATP-dependent zinc metalloprotease YME1L constitutively cleaves OPA1 at S2 within exon 5b.23,24 Other important mitochondrial membrane proteases involved in OPA1 cleavage include the presenilins-­ associated rhomboid-like (PARL) protease and the ­metallo­endopeptidase OMA1.25–28 Mitochondrial morphology and bioenergetics The state of the mitochondrial network is a reflection of the cell’s metabolic needs under both physiological and stress conditions. Although the precise mechanisms still need to be elucidated, both OPA1 and MFN2 are involved in the regulation of mitochondrial respiratory chain coupling and oxidative phosphorylation.5,29 MFN2 is also thought to exert a direct influence on mitochondrial biogenesis by regulating the expression of nuclear-encoded respiratory chain subunits.30 Fibroblasts from patients harbouring pathogenic OPA1 or MFN2 mutations exhibit a mitochondrial coupling defect with reduced mitochondrial membrane potential and impaired ATP synthesis.11,12,30–32 There is now direct experimental evidence that OPA1 regulates the assembly and stability of the respiratory chain supercomplexes through its influence on remodelling of mitochondrial cristae.33 Recent data also suggest that proteolytic activation of OPA1 is sufficient to stimulate mitochondrial inner membrane fusion in a process that is sensitive to the oxidative phosphorylation output from the mitochondrial respiratory chain.29 These elegant tuning mechanisms ensure a close match between mitochondrial morphology and the energetic demands of the cell.29,33 Impaired oxidative phosphorylation has been found consistently with the use of in vivo phosphorus magnetic resonance spectroscopy in the calf muscle of patients harbouring a broad range of OPA1 mutations.34–36 Similarly, a serious biochemical defect in the visual cortex

2  |  ADVANCE ONLINE PUBLICATION

www.nature.com/nrneurol © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS Endoplasmic reticulum APP PS1 Secretase complex PS2

PS

PC Phospholipid transport

2

VDAC

Fission

3

IP3R

Grp75

DRP1

MAM

Ca2+

DRP1

MFN2 MFN1/2 MICU1

Fusion 1 MFN1/2

PE FIS1 DRP1 FIS1 OPA3 DRP1 FIS1 DRP1 FIS1 GDAP1

MCU MAM signalling

OPA1 OPA1

Mitochondrion

Figure 1 | Mitochondrial dynamics and interactions. OPA1, MFN1 and MFN2 are essential mediators of the sequential fusion of the outer and inner membranes of Nature Reviews | Neurology adjacent mitochondria (1). During fission, DRP1 is recruited from the cytosol to the outer mitochondrial membrane, where it assembles with FIS1 to constrict the mitochondrial tubule (2). Interaction between the endoplasmic reticulum and mitochondria at MAMs is essential for many processes, including Ca2+ signalling, γ-secretase activity, and phospholipid, cholesterol ester and fatty acid metabolism (3). MFN2 at MAM interfaces is involved in endoplasmic-reticulum–mitochondria tethering via interactions with both MFN1 and MFN2 on the outer mitochondrial membrane. Abbreviations: APP, amyloid precursor protein; DRP1, dynamin-related protein 1; FIS1, mitochondrial fission 1 protein; GDAP1, Ganglioside-induced differentiation-associated protein 1; Grp75, 75 kDa glucose-regulated protein; IP3R, inositol 1,4,5-trisphosphate receptor; MAM, mitochondria-associated membrane; MCU, mitochondrial calcium uniporter protein; MFN, mitofusin; MICU1, mitochondrial calcium uptake protein 1; OPA, optic atrophy protein; PC, phosphatidylcholine; PE, phosphatidylethalonamine; PS, phosphatidylserine; PS1, presenilin 1; PS2, presenilin 2; VDAC, voltage-dependent anion channel.

was observed in MFN2 mutation ­carriers with impaired vision secondary to optic atrophy.37

Endoplasmic reticulum interactions The crosstalk between the endoplasmic reticulum and mitochondrial compartments is currently a hot topic of research.38,39 Some fascinating insights have been uncovered over the past 5 years, with a number of seminal experiments gradually dissecting the intricate physical interactions and interorganellar signalling pathways operating at these interfaces (Figure 1). These socalled mitochondria-associated membranes (MAMs) create unique microenvironments that influence lipid ­biosynthesis and calcium homeostasis.40 Calcium regulates a broad range of functions that are directly related to cell survival, including intracellular signalling, the response to excitotoxic glutaminergic stimuli, and the initiation and propagation of programmed cell death.41,42 Intracellular calcium is predomi­nantly sequestered within the endoplasmic reticulum and the mitochondrial reticulum with the bidirectional flux between these two compartments being strictly controlled. However, spatial coupling alone cannot explain the highly

coordinated nature of calcium transfer between these two compartments. This riddle was clarified with the identification of the various proteins that constitute the mitochondrial calcium uniporter, in particular the mitochondrial calcium uniporter protein (MCU) and the mitochondrial calcium uptake protein 1 (MICU1).43 MCU is a transmembrane protein located within the mitochondrial inner membrane and it assembles to form the conducting pore of this highly selective ion channel.44 MICU1 is the calcium sensor that activates the mitochondrial calcium uniporter, and loss-of-­function mutations that disrupt this critical accessory protein result in a disease phenotype characterized by proximal myopathy, learning difficulties and a progressive extra­pyramidal movement disorder.45–47 Predictably, pathogenic OPA1 and MFN2 mutations cause significant disruption of all the homeostatic mechanisms that operate at MAM interfaces, clearly highlighting the important role of mitochondrial dynamics in the b ­ uffering of intracellular calcium concentrations.48–50 MFN2 is an outer mitochondrial membrane protein but, unlike MFN1, it is also enriched at the interface between the endoplasmic reticulum and the mitochondria.38,39 Ablation or silencing of MFN2 disrupts endoplasmic reticulum morphology and loosens the interactions with mitochondria, thereby reducing the efficiency of mitochondrial calcium uptake in response to stimuli that generate inositol‑1,4,5-trisphosphate.51 The current genetic and biochemical evidence supports a model in which MFN2 on the endoplasmic reticulum acts as a structural bridge by engaging in homotypic and heterotypic complexes with MFN1 or MFN2 located on the outer mitochondrial surface. The close physical interaction between the endoplasmic reticulum and the mitochondria is likely to be even more complex than is currently understood, and identi­ fication of the other mediators involved in the formation of MAMs is actively being pursued. Intriguingly, the areas of contact between these two organelles seem to mark future sites of mitochondrial division before the recruitment of dynamin-related protein 1 (DRP1, also called dynamin-1-like protein) to these sites.52 In this proposed model, which seems to be evolutionarily conserved between yeast and mammals, the endoplasmic reticulum tubules wrap around a mitochondrial segment, constricting this area and facilitating the recruitment of DRP1 to complete the fission process. An increasing number of neurodegenerative diseases, including Alzheimer disease (AD) and Parkinson disease (PD), are being linked to disturbed interactions between the endoplasmic reticulum and mitochondria, and this list is set to grow even further as the molecular bases for as yet undefined neuropathological entities are uncovered.53,54

Mitochondrial cell death pathways Programmed cell death involves several players operating along overlapping pathways (Figure 3). The actual sequence of events is still the subject of much debate but, regardless of the triggers and mediators involved, mitochondria remain central to the irreversible process

NATURE REVIEWS | NEUROLOGY

ADVANCE ONLINE PUBLICATION  |  3 © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS a

fusion,10,19,26,62,63 there is also evidence, albeit to a lesser extent, to support a contribution by MFN2.64,65

b

c

d

Figure 2 | Mitochondrial network fragmentation in OPA1-mutant fibroblasts. Nature Reviews | Neurology Staining was performed with MitoTrackerTM (Invitrogen, UK), a mitochondrionspecific fluorescent dye (green), and Hoechst 33342 (Invitrogen, UK) for the nucleus (blue). Images were captured as a series of Z‑stacks with 16 slices, each 0.2 μm apart, using an inverted epifluorescence microscope (Leica, UK). HuygensTM image processing software (Scientific Volume Imaging, Netherlands) generates a data file that includes information on the total number of mitochondrial fragments per cell, their individual lengths and their volumes. A false colour scheme aids visualization of network morphology. A control fibroblast, a | before and b | after image deconvolution and reconstruction, shows a highly interconnected mitochondrial network. c,d | Similarly processed images in an OPA1-mutant fibroblast reveal a fragmented mitochondrial network secondary to haploinsufficiency. Abbreviation: OPA1, optic atrophy 1. Permission obtained from Springer © Yu-Wai-Man, P. et al. in Mitochondrial Disorders Caused by Nuclear Genes (ed. Wong, L.‑J. C.) 141–161 (2013).5

that will ultimately result in the cell’s elimination.55,56 Proapoptotic and antiapoptotic signals are detected and balanced by a complex network of Bcl‑2 family proteins that includes both proapoptotic members— for example, apoptosis regulators BAK and BAX, and BAD (Bcl-2-associated agonist of cell death)—and anti­ apoptotic members such as Bcl‑2 (also known as Bcl-2like protein 1) and its isoform Bcl‑X(L).55–57 Once the point of no return is reached, BAX and BAD result in permeabilization of the mitochondrial outer membrane, and the release of a potent cocktail of proapoptotic molecules, including cytochrome c, from the inner ­membrane space.58,59 The nature of this ‘death squad’ remains controversial, and a number of the suggested mediators, such as mitochondrial apoptosis-inducing factor 1 (AIFM‑1) and apoptotic protease-activating factor 1 (APAF‑1), could eventually prove to be innocent bystanders that are simply released into the cytosol as the mitochondrial content implodes.60 The central role of cytochrome c in apoptosis remains undisputed and, because of its potent nature, it remains carefully sequestered within the mitochondrial cristae. OPA1 ensures the tightness of these cristae junctions and, crucially, it controls the structural remodelling that occurs before cytochrome c is released during apoptosis.10,61 In addition to the anti­ apoptotic properties of OPA1-mediated mitochondrial

Mitochondrial quality control In addition to its link with monogenic diseases, mitochondrial dysfunction has been associated with the normal ageing process and with cancer.66 During evolution, eukaryotic cells have acquired a number of defensive mechanisms to identify and eliminate damaged dysfunctional mitochondria that would otherwise be detrimental to their survival.67,68 Multiple proteins and pathways are implicated in mitochondrial quality control, known as mitophagy, and the emerging associ­ ation between this process and human diseases provides a tantalizing link to possible future treatment strategies.69 Proteins that regulate mitochondrial dynamics are closely involved in the autophagic process, especially the initiation and formation of autophagosomes.70 Two important protein mediators in this process are PINK1 and the E3 ubiquitin-protein ligase parkin (Figure 3).71 PINK1 selectively accumulates on the outer membrane of depolarized mitochondria, whereas parkin is a cytosolic component that ubiquitinates proteins targeted for degradation.72–74 The mechanism by which PINK1 recruits parkin from the cytosol to the outer mitochondrial membrane is still being hotly debated, but an intriguing role for MFN2 has recently emerged.75 When a mitochondrial segment loses its membrane potential, PINK1 accumulates on the outer mitochondrial membrane, leading to the phosphorylation of MFN2. The phosphorylated form of MFN2 is thought to be a highly effective receptor for parkin, which then undergoes a conformational change to its active form, in addition to being drawn into close juxtaposition with PINK1 on the outer mitochondrial membrane.76 This recruitment pathway is completed when PINK1 phosphorylates parkin, thereby activating its ubiquitin ligase activity and initiating the ubiquitination of several downstream proteins, including DRP1, voltage-­dependent anion-selective channel protein (VDAC1), Bcl‑2, parkin-interacting substrate (PARIS), NF‑κB essential modulator (NEMO), and mitochondrial Rho GTPase protein 1 (MIRO1).77–79 The proteosomal degradation of MIRO1 causes kinesin to detach from the mitochondrial surface, which represents a sophisticated way of putting damaged mitochondria into quarantine by severing their connection to the microtubule network before their eventual destruction.80,81 Parkin can also reduce cytochrome c release from mitochondria in response to proapoptotic stimuli; this stress-protective activity is mediated by OPA1 operating downstream of NEMO. The latter protein is essential for canonical NF‑κB signalling, and its ubiquitination by parkin results in activation of the NF‑κB pathway, ­followed by transcriptional upregulation of OPA1.79 Despite the considerable amount of in vitro evidence that links parkin to the regulation of mitochondrial dynamics and mitophagy, the situation in vivo might prove rather more complex. In one study, dopaminergic neurons were isolated from a ‘MitoPark’ mouse model that mirrors the key features of PD, namely, a slowly

4  |  ADVANCE ONLINE PUBLICATION

www.nature.com/nrneurol © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS Table 2 | Major mitochondrial pro-fusion and pro-fission mediators Protein

Location

Functions

OPA1

Inner mitochondrial membrane

Fusion of the inner mitochondrial membrane7,188 Maintenance of cristae junctions and regulation of cytochrome c release10,19,62 Assembly and stability of the mitochondrial respiratory chain complexes12 Mitochondrial DNA maintenance with a postulated role in anchoring nucleoids to the inner mitochondrial membrane136

MFN1/MFN2*

Outer mitochondrial membrane and endoplasmic reticulum

Fusion of the outer mitochondrial membrane7,8 Interactions between endoplasmic reticulum and mitochondria, and Ca2+ homeostasis (MFN2)51 Axonal transport of mitochondria via interaction with the MIRO1–Milton complex189,190 Regulation of mitochondrial respiratory chain coupling and oxidative phosphorylation30

DRP1

Cytoplasm

Permeabilization of the outer mitochondrial membrane mediated by the apoptosis regulator BAX191,192

FIS1

Outer mitochondrial membrane

Interacts with the endoplasmic reticulum to regulate apoptosis193

OPA3

Outer mitochondrial membrane

Control of lipid metabolism and thermogenesis194–196

GDAP1

Outer mitochondrial membrane

Regulation of oxidative phosphorylation and apoptosis197,198

Pro-fusion

Pro-fission

*MFN2 is located on both the outer mitochondrial membrane and the endoplasmic reticulum. Abbreviations: DRP1, dynamin-related protein 1; FIS1, mitochondrial fission 1 protein; GDAP1, ganglioside-induced differentiation associated protein 1; MFN, mitofusin; MIRO1, mitochondrial Rho GTPase protein 1; OPA, optic atrophy protein.

progressive degeneration of substantia nigra neurons, and impaired spontaneous locomotion in the adult animals.82 Although these dopaminergic neurons showed marked respiratory chain deficiency with mitochondrial network fragmentation, rather surprisingly, parkin was not recruited to dysfunctional mitochondria, and the loss of parkin did not inhibit mitophagy or modify the ­neuro­degenerative process in the MitoPark mice.

Mitochondrial dynamics and disease Autosomal dominant optic atrophy DOA is the most common inherited optic nerve disorder seen in clinical practice, and a population-based epidemiological study in northern England estimated the minimum prevalence at 1 in 25,000.83,84 The pathological hallmark of this disorder is preferential loss of the retinal ganglion cell (RGC) layer within the inner retina, which leads to optic nerve degeneration and subsequent visual failure.85 DOA has an insidious onset, and it typically presents in early childhood with bilateral, symmetrical central visual loss and dyschromatopsia.86,87 Visual loss is invariably progressive, and almost all affected indi­ viduals will eventually fulfil the legal requirement for blind registration.88,89 The majority (50–65%) of families with DOA harbour pathogenic mutations within the OPA1 gene, which consists of 30 coding exons spanning over 100 kb of genomic DNA.83,90–92 OPA1 codes for a 960-amino-acid, dynamin-related GTPase that localizes to the inner mitochondrial membrane. The gene is highly expressed within the RGC layer, although the protein is ubiquitous, and abundant levels have also been identified in photoreceptors and other nonocular tissues such as the inner ear and the brain.93–96 Over 200 disease-causing

variants have been reported so far in this highly polymorphic gene, with mutational hot spots in the catalytic GTPase domain (exons 8–15) and the dynamin central domain (exons 16–23).83,92 The majority of OPA1 mutations result in premature termination codons, and the resultant truncated mRNA species are highly un­stable, being rapidly degraded by protective surveillance mech­ anisms operating via nonsense-mediated mRNA decay. Haploinsufficiency, therefore, is a major disease mech­ an­ism in DOA, and the pathological consequences of a dramatic reduction in OPA1 protein levels is highlighted by those rare families who are heterozygous for mi­crodeletions spanning the entire OPA1 coding region.97 Progressive visual failure remains the defining feature of DOA but, with greater availability of genetic testing, a specific OPA1 mutation in exon 14 (c.1334G>A, p.Arg445His) has been found to have a particular predilection for causing sensorineural deafness.11,98,99 The phenotypes associated with OPA1-linked disease have expanded even further to encompass a wide range of prominent neuromuscular features such as ataxia, myo­ pathy, peripheral neuropathy, and classical chronic progressive external ophthalmoplegia (CPEO).100–102 These so-called DOA+ variants are mechanistically relevant, as they highlight the deleterious consequences of OPA1 mutations not only for RGCs, but also for other CNS ­populations, peripheral nerves, and skeletal muscle. Although DOA+ was only recently recognized as a distinct clinical entity, up to 20% of OPA1 mutation carriers are now thought to be at risk of developing DOA+ features (Figure 4), which has major implications for patient counselling.103 Furthermore, OPA1 screening is increasingly performed as part of diagnostic panels for patients with unexplained neurodegenerative disorders, and

NATURE REVIEWS | NEUROLOGY

ADVANCE ONLINE PUBLICATION  |  5 © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS

Mitochondrion

BAX Bcl-2

VDAC BAX

OPA1 OPA1OPA1 OPA1 OPA1

2 YME1L

OMA1 AFG3L2

OPA1

Respiratory chain complexes

Cristae remodelling

3 NEMO PARIS

Mitophagy

OPA1

Apoptosis

Cytochrome c

PINK1

Depolarized membrane

mtDNA

SPG7

MFN2 P

1

Bcl-2

MIRO1

Parkin Ubiquitin

Axonal transport

Kinesin/dynein/dynectin Destabilize from microtubule Microtubule

Figure 3 | Major pathways implicated in neurodegenerative disorders. Nature ReviewsUnder | Neurology stress conditions, AFG3L2 triggers OPA1 cleavage via an OMA1-dependent pathway. The resulting OPA1 isoform loses its fusion and antiapoptotic properties, resulting in a fragmented mitochondrial network, release of cytochrome c into the cytosol, and cell death (1). Cleavage of OPA1 by YME1L and SPG7 regulates several mitochondrial parameters, including activity of the respiratory chain complexes, mitochondrial cristae structure and mtDNA stability (2). Accumulation of PINK1 leads to phosphorylation of MFN2 (red circle) and recruitment of cytosolic parkin to the outer mitochondrial membrane. Activated parkin ubiquitinates a range of membrane proteins that inhibit axonal transport, leading to mitophagy (3). Abbreviations: AFG3L2, AFG3-like protein 2; MFN2, mitofusin 2; MIRO1, mitochondrial Rho GTPase protein 1; mtDNA, mitochondrial DNA; NEMO, NFκB essential modulator; PARIS, parkin-interacting substrate; PINK1, PTENinduced putative kinase 1; OMA1, metalloendopeptidase; OPA1, optic atrophy protein 1; SPG7, paraplegin; VDAC, voltage-dependent anion channel; YME1L, ATP-dependent zinc metalloprotease YME1L.

other hitherto unreported pathological manifestations are bound to emerge.104–107 Interestingly, two large Italian families with an extreme DOA+ phenotype have been described, with clinical features dominated by CPEO and neurodegenerative features—namely, parkinsonism and dementia—but with a distinct lack of marked optic atrophy among affected patients (V. Carelli, unpublished work). These two families were found to harbour closely located missense OPA1 mutations within the cata­lytic GTPase domain (p.Gly488Arg and p.Ala495Val), further reinforcing the close pathophysiological links between mitochondrial dynamics, mito­phagy and neuronal loss.

Charcot–Marie–Tooth disease CMT encompasses an important group of inherited peripheral neuropathies with an estimated prevalence of approximately 1 in 2,500 in the general population.3,4 CMT is characterized by progressive degeneration of the peripheral nerves, and the clinical classification depends on whether the underlying pathological process is predominantly axonal or demyelinative. Affected patients

develop distal muscle weakness and sensory loss but, depending on the actual causative gene, there can be quite marked variations in age of onset and rate of clinical progression.3,4 MFN2 mutations were initially identified in families with CMT2A, an autosomal domi­nant form of CMT.108 The MFN2 gene consists of 19 exons and codes for a 757-amino-acid, dynamin-related GTPase protein, which is anchored within the outer mito­chondrial membrane. The majority of patients with CMT2A develop a severe peripheral neuropathy that may be primarily motor or accompanied by prominent proprioceptive loss.109,110 However, there is a degree of phenotypic variability, and in some affected MFN2 mutation carriers, the peripheral neuropathy can be complicated by subacute visual failure and optic atrophy.111 This specific CMT subtype is also known as hereditary sensory motor neuropathy type 6 (HSMN6; OMIM #601152) and, although rare, it is mechanistically highly relevant because of the fascinating link it draws between disturbed MFN2 function, RGC axonal loss and optic nerve degeneration. Visual acuity is usually reduced to 20/200 or worse in patients with HSMN6, but despite the severity of the initial visual loss, some patients can experience significant visual recovery in later life.110 Recently, compound heterozygous MFN2 mutations have also been identified in patients with early-onset peripheral neuropathy and, overall, a more severe neurological prognosis. 112 The carrier parents were asymptomatic, which is consistent with an autosomal recessive mode of inheritance for some MFN2 mutations.

Pathological mechanisms of disease Insights from animal models Research into DOA and CMT2A has been severely limited by a lack of access to diseased human tissues. To circumvent these practical difficulties, several animal models have been used to investigate the developmental impact of OPA1 (Box 1) and MFN2 (Box 2) knockdown in tissues that are normally affected in human disease. An elegant Cre–loxP recombinase strategy was used to create a conditional Mfn2 knockout mouse model that allowed embryonic development to proceed to term by sparing placental tissues.113 These Mfn2loxP mutant mice exhibited striking histopathological changes, with marked Purkinje cell neurodegeneration in the cere­ bellum associated with increased levels of apoptosis throughout the cerebellar layers. A marked respiratory chain defect was detected in the surviving Purkinje cells, and the mitochondrial network was fragmented, with aberrant mitochondrial distribution and ultrastructure visible on electron microscopy. An even more fundamental observation in this conditional Mfn2 knockout mouse model was the direct correlation between a lack of mitochondrial fusion and loss of mitochondrial DNA (mtDNA) nucleoids within affected cells, resulting in severe mtDNA depletion.114 Therefore, the mitofusins are central to mtDNA stability and fidelity, as is OPA1 (see below). Although chronic inhibition of mitochondrial fusion clearly results in

6  |  ADVANCE ONLINE PUBLICATION

www.nature.com/nrneurol © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS a

b

c 9.9 kb

II:1

II:2 II:3 II:4 II:5 II:6 L

C

II:2

d III:1 III:2 III:3 III:4 III:5 III:6 III:7

IV:1 IV:2

TTAAGSCTTGC

IV:3

Nature Reviews | Neurology Figure 4 | DOA+ phenotype and mtDNA instability. a | A large British family with early-onset optic atrophy and progressive external ophthalmoplegia, and variable features including sensorineural deafness, ataxia and peripheral neuropathy. At their last follow-up, the affected children (IV:2 and IV:3) were A, p.Ser545Arg) segregates with disease status in this family. The arrow indicates the pathogenic variant on a sequence chromatogram. Abbreviations: C, control; DOA+, dominant optic atrophy plus; L, DNA ladder; mtDNA, mitochondrial DNA; OPA1, optic atrophy 1. © Hudson, G. et al. Mutation of OPA1 causes dominant optic atrophy with external ophthalmoplegia, ataxia, deafness and multiple mitochondrial DNA deletions: a novel disorder of mtDNA maintenance. Brain (2008), 131(2), 329–337, by permission of Oxford University Press.101

marked mtDNA depletion, recent experimental data suggest that the observed mitochondrial dysfunction is caused primarily by reduced stability of the respiratory chain supercomplexes.33 More-sophisticated transgenic models harbouring specific pathogenic OPA1 and MFN2 mutations have also been created by making use of available N‑ethyl‑Nnitrosourea (ENU) libraries or site-directed mutagenesis of embryonic stem cells. Mitochondrial genome instability Molecules of mtDNA are packaged within intricate replicative structures known as nucleoids.115–118 Several proteins with critical roles in mtDNA replication are also associated with mammalian nucleoids; these include mitochondrial polymerase‑γ (POLG, also called DNA polymerase subunit gamma-1), the Twinkle protein (which unwinds double-stranded mtDNA at the replication fork), the mitochondrial single-stranded binding protein, and the accessory topoisomerase and ligase proteins.119 Nuclear disorders of mtDNA maintenance represent an important group of human diseases.120,121 The common molecular hallmark is mitochondrial genome instability, which manifests itself either as a reduction in mtDNA copy number (depletion), or accumulation of high levels of somatic mtDNA mutations in affected tissues.122–124 These mutations are predominantly multiple mtDNA deletions, but mtDNA point mutations have also been reported in association with some specific nuclear genetic defects.125–127 There is mounting evidence that the accumulation of these secondary quantitative and qualitative mtDNA abnormalities disrupt oxidative

phosphorylation, triggering a biochemical defect at the cellular level, and eventually contributing to the onset of overt clinical disease.128–130 From a diagnostic perspective, the mitochondrial biochemical defect can be readily observed in skeletal muscle biopsies as cytochrome c oxidase (COX)-negative fibres (Figure 4).131 Crucially, both DOA and CMT2A have been linked with impaired mtDNA maintenance. 98,99,101,132 COXnegative fibres harbouring high levels of mtDNA deletions have been identified in skeletal muscle biopsies from patients harbouring pathogenic OPA1 or MFN2 mutations. This finding raises the distinct possibility that the accumulation of these somatic mtDNA defects is contributing, at least partly, to multisystemic cellular dysfunction.132–135 In support of this hypothesis, the level of COX-negative muscle fibres was found to be over four times higher in patients with DOA+ phenotypes than in those who only developed pure optic atrophy.101,128 Different mechanisms have been postulated to account for the mtDNA deletions, including an imbalance of the intramitochondrial nucleotide pool, and impaired ability of the N‑terminal domain of OPA1, encoded by exon 4b, to physically anchor nucleoids to the inner mitochondrial membrane. 133,134,136 Circumstantial evi­d ence obtained with ultra-deep next-generation sequencing suggests that OPA1 mutations lead to an increased rate of clonal expansion of somatic mtDNA mutations, triggering the emergence of COX-negative muscle fibres once a critical mutational threshold has been exceeded.137 Marked mitochondrial proliferation is frequently obser­ved in tissues from patients with DOA or CMT2A, presumably as a cellular compensatory mecha­nism in the face of impaired oxidative phosphorylation.138–140 Depletion of mtDNA has, so far, not been reported in the context of OPA1 mutations. However, a patient was recently described with optic atrophy and an earlyonset progressive multisystemic disorder secondary to a de novo missense mutation involving the MFN2 GTPase domain (c.628G>T, p.Asp210Tyr).141 A skeletal muscle biopsy revealed COX-negative fibres, lipid accumulation, dysmorphic mitochondria, and severe mtDNA depletion at 28% residual level. This rather unexpected finding is in sharp contrast with another MFN2 missense mutation altering the same amino acid (c.629A>T, p.Asp210Val), which had previously been identified in a large Tunisian family with a complicated neuro-­ophthalmological pheno­type.132 COX-negative fibres were present in skeletal muscle biopsies from several affected individuals in this family, and multiple mtDNA deletions were detected on long-range PCR analysis of DNA extracted from skeletal muscle homo­genates. However, the p.Asp210Val amino acid substitution, in contrast to p.Asp210Tyr, did not lead to mtDNA depletion. The divergent molecular consequences of these two MFN2 mutations are puzzling and remain unexplained.142 Disrupted mitochondrial axonal transport Mitochondrial transport and localization are central to the survival of highly specialized cells, especially for long

NATURE REVIEWS | NEUROLOGY

ADVANCE ONLINE PUBLICATION  |  7 © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS Box 1 | OPA1 disease models Drosophila melanogaster Homozygous Opa1 mutations are embryonically lethal. Eye-specific homozygous Opa1 mutants generated by somatic mutagenesis develop abnormal ommatidial phenotypes (rough and glossy eyes) secondary to increased apoptosis, increased levels of reactive oxygen species and mitochondrial fragmentation.205 Heterozygous Opa1 mutants do not show any gross ommatidial morphology, but there is evidence of disturbed visual function on electroretinography. The mutant heterozygotes also have a decreased heart rate, increased heart arrhythmia, and poor stress tolerance.206,207

Zebrafish Knockdown of the opa1 gene was achieved with translation-blocking antisense morpholinos. Eyes of opa1 morphants are small, but with no observable retinal ganglion cell loss or optic nerve degeneration. Embryos also exhibit abnormal blood circulation and heart defects, developmental delay, small pectoral fin buds, and impaired locomotor activity. Depletion of Opa1 protein levels causes bioenergetic defects without impairing mitochondrial efficiency.208 Mouse Homozygous mutant mice (Opa1–/–) die in utero during early embryogenesis, confirming a critical role for Opa1 in early development. Heterozygous mutant mice (Opa1+/–) develop a slowly progressive bilateral optic neuropathy and reduced visual function. Optic nerve degeneration has been documented as early as 6 months, but is much more striking by 2 years of age.209–213 Loss of dendritic arborization precedes the onset of retinal ganglion cell loss, and the surviving axons have abnormal morphologies, with segmental areas of demyelination and myelin aggregation. The mitochondrial network is fragmented in the tissues that have been examined. Some extraocular features are also noted in older mice, including decreased locomotor activity, abnormal body fat distribution, and cardiomyopathy. No cytochrome c oxidase-negative or ragged red muscle fibres, or multiple mitochondrial DNA deletions, have been detected.214–216 Abbreviation: Opa1, optic atrophy 1.

Box 2 | MFN2 disease models Drosophila melanogaster Mfn2 has been knocked down in the eye, heart tubes and the posterior compartment of the developing wing in different Drosophila models. Depletion of Mfn2 results in mitochondrial network fragmentation, disturbed calcium homeostasis, and induction of apoptosis. A number of developmental abnormalities have also been observed, including small dysmorphic eyes, dilated cardiomyopathy, and a striking loss of adult wing tissue.217–220 Zebrafish Loss of mfn2 results in severe alterations affecting both motor neurons and muscles fibres. Mutant zebrafish show progressive loss of swimming ability. At the cellular level, mitochondrial axonal transport is disrupted, suggesting that the latter is a key disease mechanism in Charcot–Marie–Tooth disease type 2A.221,222 Mouse Several knockout mouse models have been developed, some of which are tissue-specific (dopaminergic neurons, liver and skeletal muscle). Generalized depletion of Mfn1 and Mfn2 results in embryonic lethality secondary to placental insufficiency, further reinforcing the idea that these pro-fusion proteins are indespensable in early development. The mitochondrial network is severely fragmented in cultured embryonic fibroblasts from these knockout mice, and a dramatic loss of membrane potential has been observed in a subpopulation of these fragmented mitochondria.113,114,223–225 Transgenic mouse models of Charcot–Marie–Tooth disease type 2A harbouring pathogenic Mfn2 mutations have also been created that replicate the human disease phenotype. The following abnormalities have been reported in mutant Mfn2 mice: a combined respiratory chain defect involving mitochondrial complexes II and V; mitochondrial proliferation and impaired mitochondrial distribution along axons; and overrepresentation of smaller axons within myelinated peripheral nerves.226–228 Abbreviation: Mfn2, mitofusin 2.

neurons with axons that span considerable anatomical distances.143,144 This fundamental requirement is dramatically exemplified by a number of neurodegenerative disorders, which—though distinct—share a number of common molecular and phenotypic themes. A remarkable disease paradigm emerged with the identification of spastic paraplegia 7 (SPG7) and AFG3L2 mutations in patients with clinical manifestations ranging from isolated optic atrophy and spastic paraplegia, to morecomplex neurological phenotypes that were strikingly reminiscent of the ‘plus’ features reported with OPA1 and MFN2 mutations.21,145 Hereditary spastic paraplegia (HSP) is a slowly progressive neurological disorder marked by the development of lower limb spasticity and weakness.143 The classification into pure and complicated forms is based on the presence of additional clinical features besides spastic paraplegia, such as optic atrophy, ataxia, peripheral neuropathy, extrapyramidal deficits and cognitive decline.146 Genetically, HSP is highly heterogeneous with over 40 mapped loci reported to date.143,144 In a subgroup of patients with a type of autosomal recessive HSP, pathogenic mutations were identified in the SPG7 gene, which codes for paraplegin.147 Analogous to the human form of the disease, now called HSP7 (OMIM #607259), paraplegin-­deficient mice developed a distal axonopathy of spinal and peripheral axons secondary to impaired anterograde transport of organelles and neuro­filaments.148 Interestingly, axonal swelling and neuronal degeneration were preceded in the earliest stages by mitochondrial morphological abnormalities within synap­tic terminals and in the more distal nerve segments. The optic nerves of Spg7–/– mice were also abnormal, with swelling of RGC axons, but marked degeneration was not identified, indicating a later and milder degree of i­ nvolvement than in the spinal cord.148 Paraplegin is a major component of the mitochondrial AAA+ protease family, which is involved in the posttranslational processing of OPA1. Although the mol­ ecular link remains to be formally established, it is unlikely to be merely coincidental that SPG7 mutations can also result in an expanded neuro-ophthalmological phenotype that includes optic atrophy and CPEO, in addition to classic features of spastic paraplegia.149–151 Even more interestingly, SPG7 mutations are thought to cause CPEO through disordered mtDNA maintenance, similar to OPA1 and MFN2 mutations.152,153 Some SPG7 mutations can also behave dominantly, and in one large multi­generational family with DOA, a novel hetero­ zygous SPG7 mutation (c.1232A>C, p.Asp411Ala) was shown to segregate convincingly with pure optic atrophy and visual failure.149 AFG3L2 can homo-oligomerize or form functional hetero-oligomeric complexes with paraplegin. 21,67 AFG3L2 mutations cause autosomal dominant spino­ cerebellar ataxia type 28 (SCA28; OMIM #604581), a juvenile-onset syndrome marked by slowly progressive gait ataxia, dysarthria, limb hyperreflexia, nystagmus and ophthalmoplegia.154 Homozygous AFG3L2 missense mutations were also found, by whole-exome sequencing,

8  |  ADVANCE ONLINE PUBLICATION

www.nature.com/nrneurol © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS in a consangui­neous family with a novel severe spastic ataxia–­neuropathy syndrome marked by lower limb spasticity, peripheral neuropathy, ptosis, oculomotor apraxia, dystonia, cerebellar atrophy, and progressive myoclonic epilepsy.155 The loss of AFG3L2 led to mitochondrial respiratory chain dysfunction, decreased mitochondrial calcium uptake, and mitochondrial network fragmentation.156 A similar cascade of deleterious cellular events secondary to defective mitochondrial ribosomal protein synth­ esis was observed in a conditional Afg3l2 mouse model that allowed restricted deletion of the gene in cerebellar Purkinje cells.157 Depletion of AFG3L2 also impaired anterograde axonal transport of mitochondria in cortical neurons via a mechanism that is thought to involve reactive oxygen species signalling and hyperphosphorylation of the microtubule-associated protein tau.158 Disturbed axonal mitochondrial transport is not limited to disorders related to SPG7 and AFG3L2 mutations. In 2014, two elegant knockout mouse models highlighted the central importance of MIRO1 with regard to mammalian neuronal function and main­ tenance. MIRO1 anchors mitochondria to the motor proteins responsible for their anterograde and retrograde transport along neurons. In these two knockout mouse models, suppression of retrograde transport in MIRO1-deficient neurons led to mitochondrial depletion along corticospinal tract axons, and a progressive pattern of neurological deficits analogous to the human form of upper motor neuron disease.159 Mitochondrial dynamics and neurodegeneration The pathological spectrum associated with disturbed mitochondrial dynamics has expanded to encompass a more heterogeneous group of human disorders than initi­ ally envisaged. Pathogenic mutations in the leucine-rich repeat kinase 2 (LRRK2), huntingtin (HTT) and sacsin molecular chaperone (SACS) genes result in distinct neurodegenerative phenotypes: respectively, auto­somal dominant PD, 160 Huntington disease, 161 and auto­ somal recessive spastic ataxia of Charlevoix–Saguenay (ARSACS).162 All three protein products have been shown to potentiate the pro-fission activity of DRP1, both in vitro and in vivo, indicating a strong consistent link between disturbed mitochondrial dynamics and neurodegeneration.163–168 Impaired mitochondrial biogenesis, defective axonal transport of mitochondria, and increased DRP1mediated mitochondrial fission also seem to underlie the synaptic degeneration seen in neurons from patients with AD and from an established mouse model.169–171 One of the neuropathological hallmarks of AD is the accumulation of amyloid‑β (Aβ) plaques in the brain secondary to aberrant processing of amyloid precursor protein (APP) by the γ‑secretase complex. Intriguingly, presenilin‑1 (PS‑1) and presenilin‑2 (PS‑2), which are the key catalytic components of the γ‑secretase complex, are concentrated at MAM interfaces, and evidence is mounting that dysregulated crosstalk between the endoplasmic reticulum and mitochondrial compartments contributes to the development and progression

of the pathological process that underlies AD.38,172,173 Recent evidence also implicates disturbed mitochondrial dynamics in patients with frontotemporal dementia plus amyotrophic lateral sclerosis who harbour pathogenic mutations within the coiled-coil-helix-coiled-coil-helix domain containing 10 gene (CHCHD10), which encodes a mitochondrial intermembrane space protein enriched at cristae junctions.174,175

Management and treatment strategies Supportive measures Genetic counselling is central to the management of patients carrying pathogenic OPA1 or MFN2 mutations.176 Testing for these mutations raises sensitive ethical issues that are best addressed by specialist clinicians and trained genetic counsellors, who can provide patients and their families with the accurate information that would allow them to make an informed decision. Currently, diseasemodifying interventions for DOA and CMT2A are limited—a situation that places even higher importance on physical and occupational rehabilitation for affected patients.3,177 As OPA1 and MFN2 mutations can result in progressive multisystemic involvement, a multidisciplin­ ary team approach is essential to maximize the patient’s quality of life and minimize the long-term morbidity ­associated with chronic disease. Gene therapy A uniform treatment strategy for disorders related to OPA1 and MFN2 is not realistic given the genetic complexity involved, with some mutations resulting in haploinsufficiency and others thought to exert a d­ominant-negative effect. Furthermore, the group of tissues affected can be highly variable even between patients carrying the same pathogenic mutation.92,103 Gene replacement therapy is being explored for patients with DOA and visual failure, as the eye is an easily accessible organ that has the distinct advantage of providing robust outcome measures for judging treatment efficacy.178 The intraocular safety and the efficiency of RGC transfection after intravitreal injection of an adeno-associated virus vector (AAV2-pOPA1) are currently being investi­ gated in heterozygous mice harbouring a pathogenic OPA1 deletion (c.2708–2711delTTAG), which is commonly seen in patients with DOA (G. Lenaers, personal communication). Classic gene therapy paradigms will prove much more challenging for pathogenic mutations that result in gain of function, and for the correction of the underlying defects in tissues that are anatomically poorly defined or difficult to access, for example, the CNS. Modulating mitochondrial dynamics As disturbed mitochondrial dynamics are a prominent pathological feature in a number of late-onset neuro­ degenerative disorders, it is biologically plausible that restoration of physiological equilibrium between mitochondrial fusion and fission could help salvage the remaining neuronal populations, thereby halting further clinical deterioration.179 Tractable means of rectifying

NATURE REVIEWS | NEUROLOGY

ADVANCE ONLINE PUBLICATION  |  9 © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS this imbalance are currently rudimentary, and future approaches will require a high level of sophistication given the myriad of accessory factors that regulate mitochondrial shape, and the complex reciprocal inter­actions with other cellular processes such as mitochondrial transport and mitophagy.180–182

Neuroprotective drugs Mitochondrial antioxidants are a promising approach for the management of patients with Leber hereditary optic neuropathy (LHON), a classic primary mtDNA disorder, which, like DOA, is characterized by the preferen­tial destruction of the RGC layer, leading to cata­ strophic visual loss in at-risk mutation carriers.183 Two compounds have recently been investigated for LHON. The first is idebenone, a short-chain benzoquinone structurally related to coenzyme Q10, which promotes mitochondrial ATP synthesis in addition to having antioxidant properties.184–186 The second is EPI‑743, another quinone analogue, which is reported to be more potent than idebenone, at least in vitro.187 These two drugs are obvious candidates for patients with progressive visual failure secondary to optic nerve degeneration and, if properly conducted, future clinical trials could provide preliminary data on the possible benefits of idebenone and EPI‑743 for the neurological features associated with OPA1 and MFN2 mutations. Clearly, these are only the first tentative steps towards modulation of disease progression for this group of disorders intrinsically linked to mitochondrial dysfunction. Several groups are actively pursuing the identification of novel neuroprotective agents by using complementary approaches that combine the power of high-­throughput genomics with in vitro screening of small molecule libraries, and with further in vivo validation using existing animal models. This ambitious strategy will also be directly relevant to the broader field of mitochondrial dynamics, with implications for late-onset ­neurodegenerative disorders such as PD and AD.54,182 1.

2.

3.

4.

5.

6.

7.

Chan, D. C. Mitochondria: dynamic organelles in disease, aging, and development. Cell 125, 1241–1252 (2006). Yu‑Wai‑Man, P., Griffiths, P. G., Hudson, G. & Chinnery, P. F. Inherited mitochondrial optic neuropathies. J. Med. Genet. 46, 145–158 (2009). Reilly, M. M., Shy, M. E., Muntoni, F. & Pareyson, D. 168th ENMC International Workshop: outcome measures and clinical trials in Charcot–Marie–Tooth disease (CMT). Neuromuscul. Disord. 20, 839–846 (2010). Saporta, A. S. D. et al. Charcot–Marie–Tooth disease subtypes and genetic testing strategies. Ann. Neurol. 69, 22–33 (2011). Yu‑Wai‑Man, P., Lenaers, G. & Chinnery, P. F. in Mitochondrial Disorders Caused by Nuclear Genes (ed. Wong, L.‑J.C.) 141–161 (Springer, 2013). Belenguer, P. & Pellegrini, L. The dynamin GTPase OPA1: more than mitochondria? Biochim. Biophys. Acta 1833, 176–183 (2013). Chen, H. C. et al. Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J. Cell Biol. 160, 189–200 (2003).

8.

9.

10.

11.

12.

13.

14.

Conclusions

The genetic basis of several common neurodegenerative disorders has been clarified in recent years, and the pace of discovery will accelerate exponentially with the advent of next-generation whole-genome sequen­ cing and more-powerful bioinformatics technology. A remarkable element of the past decade of research has been the increasing realization that disturbed mitochondrial dynamics have a fundamental role in the pathophysiology of disease groups as diverse as DOA, CMT, HSP and the inherited spinocerebellar ataxias, reflecting to a large extent the broader phenotypic spectrum observed in classic mitochondrial syndromes. Treatment options for these progressive neurodegenerative dis­ orders remain limited, but the future looks promising given the availability of established disease models to test both the in vitro and the in vivo efficacy of potential new agents. Prospective deep-phenotyping studies that make use of existing national patient networks will also be essential to better define the natural history of these diseases, which in turn will reveal the clinical biomarkers that are most likely to predict disease progression, and the outcome measures that will prove most sensitive in detecting a treatment effect. Review criteria The articles included in this Review were primarily selected from the extensive personal bibliographies of the authors, and following discussions with experts in the field at national and international meetings. Additional studies were also identified on PubMed, MEDLINE and Web of Science using combinations of the following keywords: “AFG3L2”, “dominant optic atrophy”, “Charcot– Marie–Tooth disease”, “Drosophila model”, “gene therapy”, “mitochondrial disease”, “mitochondrial DNA instability”, “mitochondrial dynamics”, “mitochondriaassociated membranes”, “mitophagy”, “mouse model”, “neurodegenerative disease”, “neuroprotection”, “MFN2”, “OPA1”, “SPG7” and “zebrafish model”.

Eura, Y., Ishihara, N., Yokota, S. & Mihara, K. Two mitofusin proteins, mammalian homologues of FZO, with distinct functions are both required for mitochondrial fusion. J. Biochem. 134, 333–344 (2003). Lenaers, G. et al. OPA1 functions in mitochondria and dysfunctions in optic nerve. Int. J. Biochem. Cell Biol. 41, 1866–1874 (2009). Frezza, C. et al. OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion. Cell 126, 177–189 (2006). Amati-Bonneau, P. et al. OPA1 R445H mutation in optic atrophy associated with sensorineural deafness. Ann. Neurol. 58, 958–963 (2005). Zanna, C. et al. OPA1 mutations associated with dominant optic atrophy impair oxidative phosphorylation and mitochondrial fusion. Brain 131, 352–367 (2008). Loson, O. C., Song, Z., Chen, H. & Chan, D. C. Fis1, Mff, MiD49, and MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol. Biol. Cell 24, 659–667 (2013). Otera, H. et al. Mff is an essential factor for mitochondrial recruitment of Drp1 during

10  |  ADVANCE ONLINE PUBLICATION

15.

16.

17.

18.

19.

20.

mitochondrial fission in mammalian cells. J. Cell Biol. 191, 1141–1158 (2010). Rostovtseva, T. K. et al. Bax activates endophilin B1 oligomerization and lipid membrane vesiculation. J. Biol. Chem. 284, 34390–34399 (2009). Tondera, D. et al. The mitochondrial protein MTP18 contributes to mitochondrial fission in mammalian cells. J. Cell Sci. 118, 3049–3059 (2005). Takahashi, Y., Meyerkord, C. L. & Wang, H. G. Bif‑1/endophilin B1: a candidate for crescent driving force in autophagy. Cell Death Differ. 16, 947–955 (2009). Olichon, A. et al. The human dynamin-related protein OPA1 is anchored to the mitochondrial inner membrane facing the inter-membrane space. FEBS Lett. 523, 171–176 (2002). Olichon, A. et al. OPA1 alternate splicing uncouples an evolutionary conserved function in mitochondrial fusion from a vertebrate restricted function in apoptosis. Cell Death Differ. 14, 682–692 (2007). Duvezin-Caubet, S. et al. OPA1 processing reconstituted in yeast depends on the subunit

www.nature.com/nrneurol © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS

21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

composition of the m‑AAA protease in mitochondria. Mol. Biol. Cell 18, 3582–3590 (2007). Martinelli, P. & Rugarli, E. I. Emerging roles of mitochondrial proteases in neurodegeneration. Biochim. Biophys. Acta 1797, 1–10 (2010). Ishihara, N., Fujita, Y., Oka, T. & Mihara, K. Regulation of mitochondrial morphology through proteolytic cleavage of OPA1. EMBO J. 25, 2966–2977 (2006). Griparic, L., Kanazawa, T. & van der Bliek, A. M. Regulation of the mitochondrial dynamin-like protein Opa1 by proteolytic cleavage. J. Cell Biol. 178, 757–764 (2007). Song, Z., Chen, H., Fiket, M., Alexander, C. & Chan, D. C. OPA1 processing controls mitochondrial fusion and is regulated by mRNA splicing, membrane potential, and Yme1L. J. Cell Biol. 178, 749–755 (2007). McQuibban, G. A., Saurya, S. & Freeman, M. Mitochondrial membrane remodelling regulated by a conserved rhomboid protease. Nature 423, 537–541 (2003). Cipolat, S. et al. Mitochondrial rhomboid PARL regulates cytochrome c release during apoptosis via OPA1-dependent cristae remodeling. Cell 126, 163–175 (2006). Ehses, S. et al. Regulation of OPA1 processing and mitochondrial fusion by m‑AAA protease isoenzymes and OMA1. J. Cell Biol. 187, 1023–1036 (2009). Head, B., Griparic, L., Amiri, M., GandreBabbe, S. & van der Bliek, A. M. Inducible proteolytic inactivation of OPA1 mediated by the OMA1 protease in mammalian cells. J. Cell Biol. 187, 959–966 (2009). Mishra, P., Carelli, V., Manfredi, G. & Chan, D. C. Proteolytic cleavage of Opa1 stimulates mitochondrial inner membrane fusion and couples fusion to oxidative phosphorylation. Cell Metab. 19, 630–641 (2014). Pich, S. et al. The Charcot–Marie–Tooth type 2A gene product, Mfn2, up-regulates fuel oxidation through expression of OXPHOS system. Hum. Mol. Genet. 14, 1405–1415 (2005). Agier, V. et al. Defective mitochondrial fusion, altered respiratory function, and distorted cristae structure in skin fibroblasts with heterozygous OPA1 mutations. Biochim. Biophys. Acta 1822, 1570–1580 (2012). Chevrollier, A. et al. Hereditary optic neuropathies share a common mitochondrial coupling defect. Ann. Neurol. 63, 794–798 (2008). Cogliati, S. et al. Mitochondrial cristae shape determines respiratory chain supercomplexes assembly and respiratory efficiency. Cell 155, 160–171 (2013). Lodi, R. et al. Deficit of in vivo mitochondrial ATP production in OPA1-related dominant optic atrophy. Ann. Neurol. 56, 719–723 (2004). Lodi, R. et al. Defective mitochondrial adenosine triphosphate production in skeletal muscle from patients with dominant optic atrophy due to OPA1 mutations. Arch. Neurol. 68, 67–73 (2011). Yu‑Wai‑Man, P., Trenell, M. I., Hollingsworth, K. G., Griffiths, P. G. & Chinnery, P. F. OPA1 mutations impair mitochondrial function in both pure and complicated dominant optic atrophy. Brain 134, e164 (2011). Del Bo, R. et al. Mutated mitofusin 2 presents with intrafamilial variability and brain mitochondrial dysfunction. Neurology 71, 1959–1966 (2008). Area-Gomez, E. et al. Upregulated function of mitochondria-associated ER membranes in Alzheimer disease. EMBO J. 31, 4106–4123 (2012).

39. de Brito, O. M. & Scorrano, L. An intimate liaison: spatial organization of the endoplasmic reticulum-mitochondria relationship. EMBO J. 29, 2715–2723 (2010). 40. Kornmann, B. et al. An ER–mitochondria tethering complex revealed by a synthetic biology screen. Science 325, 477–481 (2009). 41. Merkwirth, C. & Langer, T. Mitofusin 2 builds a bridge between ER and mitochondria. Cell 135, 1165–1167 (2008). 42. Lam, A. K. M. & Galione, A. The endoplasmic reticulum and junctional membrane communication during calcium signaling. Biochim. Biophys. Acta 1833, 2542–2559 (2013). 43. Patron, M. et al. MICU1 and MICU2 finely tune the mitochondrial Ca2+ uniporter by exerting opposite effects on MCU activity. Mol. Cell 53, 726–737 (2014). 44. Baughman, J. M. et al. Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter. Nature 476, 341–345 (2011). 45. Perocchi, F. et al. MICU1 encodes a mitochondrial EF hand protein required for Ca2+ uptake. Nature 467, 291–296 (2010). 46. Csordas, G. et al. MICU1 controls both the threshold and cooperative activation of the mitochondrial Ca2+ uniporter. Cell Metab. 17, 976–987 (2013). 47. Logan, C. V. et al. Loss‑of‑function mutations in MICU1 cause a brain and muscle disorder linked to primary alterations in mitochondrial calcium signaling. Nat. Genet. 46, 188–193 (2014). 48. Dayanithi, G. et al. Characterization of Ca2+ signalling in postnatal mouse retinal ganglion cells: involvement of OPA1 in Ca2+ clearance. Ophthalmic Genet. 31, 53–65 (2010). 49. Kushnareva, Y. E. et al. Loss of OPA1 disturbs cellular calcium homeostasis and sensitizes for excitotoxicity. Cell Death Differ. 20, 353–365 (2013). 50. Singaravelu, K. et al. Mitofusin 2 regulates STIM1 migration from the Ca2+ store to the plasma membrane in cells with depolarized mitochondria. J. Biol. Chem. 286, 12189–12201 (2011). 51. de Brito, O. M. & Scorrano, L. Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature 456, 605–611 (2008). 52. Friedman, J. R. et al. ER tubules mark sites of mitochondrial division. Science 334, 358–362 (2011). 53. Hayashi, T., Rizzuto, R., Hajnoczky, G. & Su, T.‑P. MAM: more than just a housekeeper. Trends Cell Biol. 19, 81–88 (2009). 54. Schon, E. A. & Przedborski, S. Mitochondria: the next (neurode) generation. Neuron 70, 1033–1053 (2011). 55. Arnoult, D. Mitochondrial fragmentation in apoptosis. Trends Cell Biol. 17, 6–12 (2007). 56. Suen, D. F., Norris, K. L. & Youle, R. J. Mitochondrial dynamics and apoptosis. Genes Dev. 22, 1577–1590 (2008). 57. Levine, B., Sinha, S. & Kroemer, G. Bcl‑2 family members: dual regulators of apoptosis and autophagy. Autophagy 4, 600–606 (2008). 58. Renault, T. T. & Manon, S. Bax: addressed to kill. Biochimie 93, 1379–1391 (2011). 59. Walensky, L. D. & Gavathiotis, E. BAX unleashed: the biochemical transformation of an inactive cytosolic monomer into a toxic mitochondrial pore. Trends Biochem. Sci. 36, 642–652 (2011). 60. Ekert, P. G. & Vaux, D. L. The mitochondrial death squad: hardened killers or innocent bystanders? Curr. Opin. Cell Biol. 17, 626–630 (2005). 61. Scorrano, L. et al. A distinct pathway remodels mitochondrial cristae and mobilizes

NATURE REVIEWS | NEUROLOGY

62.

63.

64.

65.

66.

67.

68.

69.

70.

71.

72.

73.

74.

75.

76.

77.

78.

79.

80.

cytochrome c during apoptosis. Dev. Cell 2, 55–67 (2002). Olichon, A. et al. Loss of OPA1 perturbates the mitochondrial inner membrane structure and integrity, leading to cytochrome c release and apoptosis. J. Biol. Chem. 278, 7743–7746 (2003). Arnoult, D., Grodet, A., Lee, Y. J., Estaquier, J. & Blackstone, C. Release of OPA1 during apoptosis participates in the rapid and complete release of cytochrome c and subsequent mitochondrial fragmentation. J. Biol. Chem. 280, 35742–35750 (2005). Karbowski, M., Norris, K. L., Cleland, M. M., Jeong, S.‑Y. & Youle, R. J. Role of Bax and Bak in mitochondrial morphogenesis. Nature 443, 658–662 (2006). Neuspiel, M., Zunino, R., Gangaraju, S., Rippstein, P. & McBride, H. Activated mitofusin 2 signals mitochondrial fusion, interferes with Bax activation, and reduces susceptibility to radical induced depolarization. J. Biol. Chem. 280, 25060–25070 (2005). Chinnery, P. F., Samuels, D. C., Elson, J. & Turnbull, D. M. Accumulation of mitochondrial DNA mutations in ageing, cancer, and mitochondrial disease: is there a common mechanism? Lancet 360, 1323–1325 (2002). Rugarli, E. I. & Langer, T. Mitochondrial quality control: a matter of life and death for neurons. EMBO J. 31, 1336–1349 (2012). Twig, G. & Shirihai, O. S. The interplay between mitochondrial dynamics and mitophagy. Antioxid. Redox Signal. 14, 1939–1951 (2011). Twig, G., Hyde, B. & Shirihai, O. S. Mitochondrial fusion, fission and autophagy as a quality control axis: the bioenergetic view. Biochim. Biophys. Acta 1777, 1092–1097 (2008). Twig, G. et al. Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. EMBO J. 27, 433–446 (2008). Youle, R. J. & van der Bliek, A. M. Mitochondrial fission, fusion, and stress. Science 337, 1062–1065 (2012). Gegg, M. E. et al. Mitofusin 1 and mitofusin 2 are ubiquitinated in a PINK1/parkin-dependent manner upon induction of mitophagy. Hum. Mol. Genet. 19, 4861–4870 (2010). Matsuda, N. et al. PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J. Cell Biol. 189, 211–221 (2010). Narendra, D., Tanaka, A., Suen, D.‑F. & Youle, R. J. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J. Cell Biol. 183, 795–803 (2008). Pallanck, L. Mitophagy: mitofusin recruits a mitochondrial killer. Curr. Biol. 23, R570–R572 (2013). Chen, Y. & Dorn, G. W. 2nd. PINK1-phosphorylated mitofusin 2 is a Parkin receptor for culling damaged mitochondria. Science 340, 471–475 (2013). Kane, L. A. & Youle, R. J. PINK1 and Parkin flag Miro to direct mitochondrial traffic. Cell 147, 721–723 (2011). Shin, J. H. et al. PARIS (ZNF746) repression of PGC‑1α contributes to neurodegeneration in Parkinson’s disease. Cell 144, 689–702 (2011). Mueller-Rischart, A. K. et al. The E3 ligase Parkin maintains mitochondrial integrity by increasing linear ubiquitination of NEMO. Mol. Cell 49, 908–921 (2013). Liu, S. et al. Parkinson’s disease-associated kinase PINK1 regulates Miro protein level and axonal transport of mitochondria. PLoS Genet. 8, e1002537 (2012).

ADVANCE ONLINE PUBLICATION  |  11 © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS 81. Wang, X. et al. PINK1 and Parkin target Miro for phosphorylation and degradation to arrest mitochondrial motility. Cell 147, 893–906 (2011). 82. Sterky, F. H., Lee, S., Wibom, R., Olson, L. & Larsson, N.G. Impaired mitochondrial transport and Parkin-independent degeneration of respiratory chain-deficient dopamine neurons in vivo. Proc. Natl Acad. Sci. USA 108, 12937–12942 (2011). 83. Yu‑Wai‑Man, P. et al. The prevalence and natural history of dominant optic atrophy due to OPA1 mutations. Ophthalmology 117, 1538–1546 (2010). 84. Yu‑Wai‑Man, P. & Chinnery, P. F. Dominant optic atrophy: novel OPA1 mutations and revised prevalence estimates. Ophthalmology 120, 1712–1712.e1 (2013). 85. Yu‑Wai‑Man, P., Bailie, M., Atawan, A., Chinnery, P. F. & Griffiths, P. G. Pattern of retinal ganglion cell loss in dominant optic atrophy due to OPA1 mutations. Eye 25, 597–601 (2011). 86. Cohn, A. C. et al. The natural history of OPA1related autosomal dominant optic atrophy. Br. J. Ophthalmol. 92, 1333–1336 (2008). 87. Yu‑Wai‑Man, P. et al. Genetic screening for OPA1 and OPA3 mutations in patients with suspected inherited optic neuropathies. Ophthalmology 118, 558–563 (2011). 88. Votruba, M., Moore, A. T. & Bhattacharya, S. S. Clinical features, molecular genetics, and pathophysiology of dominant optic atrophy. J. Med. Genet. 35, 793–800 (1998). 89. Bailie, M., Votruba, M., Griffiths, P. G., Chinnery, P. F. & Yu‑Wai‑Man, P. Visual and psychological morbidity among patients with autosomal dominant optic atrophy. Acta Ophthalmol. 91, e413–e414 (2013). 90. Alexander, C. et al. OPA1, encoding a dynaminrelated GTPase, is mutated in autosomal dominant optic atrophy linked to chromosome 3q28. Nat. Genet. 26, 211–215 (2000). 91. Delettre, C. et al. Nuclear gene OPA1, encoding a mitochondrial dynamin-related protein, is mutated in dominant optic atrophy. Nat. Genet. 26, 207–210 (2000). 92. Ferre, M. et al. Molecular screening of 980 cases of suspected hereditary optic neuropathy with a report on 77 novel OPA1 mutations. Hum. Mutat. 30, E692–E705 (2009). 93. Aijaz, S., Erskine, L., Jeffery, G., Bhattacharya, S. S. & Votruba, M. Developmental expression profile of the optic atrophy gene product: OPA1 is not localized exclusively in the mammalian retinal ganglion cell layer. Invest. Ophthalmol. Vis. Sci. 45, 1667–1673 (2004). 94. Bette, S., Schlaszus, H., Wissinger, B., Meyermann, R. & Mittelbronn, M. OPA1, associated with autosomal dominant optic atrophy, is widely expressed in the human brain. Acta Neuropathol. 109, 393–399 (2005). 95. Pesch, U. E. et al. OPA1, the disease gene for autosomal dominant optic atrophy, is specifically expressed in ganglion cells and intrinsic neurons of the retina. Invest. Ophthalmol. Vis. Sci. 45, 4217–4225 (2004). 96. Wang, A. G., Fann, M. J., Yu, H. Y. & Yen, M. Y. OPA1 expression in the human retina and optic nerve. Exp. Eye Res. 83, 1171–1178 (2006). 97. Marchbank, N. J. et al. Deletion of the OPA1 gene in a dominant optic atrophy family: evidence that haploinsufficiency is the cause of disease. J. Med. Genet. 39, e47 (2002). 98. Amati-Bonneau, P. et al. The association of autosomal dominant optic atrophy and moderate deafness may be due to the R445H mutation in the OPA1 gene. Am. J. Ophthalmol. 136, 1170–1171 (2003).

99. Leruez, S. et al. Sensorineural hearing loss in OPA1-linked disorders. Brain 136, e236 (2013). 100. Amati-Bonneau, P. et al. OPA1 mutations induce mitochondrial DNA instability and optic atrophy plus phenotypes. Brain 131, 338–351 (2008). 101. Hudson, G. et al. Mutation of OPA1 causes dominant optic atrophy with external ophthalmoplegia, ataxia, deafness and multiple mitochondrial DNA deletions: a novel disorder of mtDNA maintenance. Brain 131, 329–337 (2008). 102. Spinazzi, M. et al. A novel deletion in the GTPase domain of OPA1 causes defects in mitochondrial morphology and distribution, but not in function. Hum. Mol. Genet. 17, 3291–3302 (2008). 103. Yu‑Wai‑Man, P. et al. Multi-system neurological disease is common in patients with OPA1 mutations. Brain 133, 771–786 (2010). 104. Pretegiani, E. et al. Spastic paraplegia in ‘dominant optic atrophy plus’ phenotype due to OPA1 mutation. Brain 134, e195 (2011). 105. Marelli, C. et al. Heterozygous OPA1 mutations in Behr syndrome. Brain 134, e169 (2011). 106. Liskova, P. et al. Novel OPA1 missense mutation in a family with optic atrophy and severe widespread neurological disorder. Acta Ophthalmol. 91, E225–E231 (2013). 107. Ranieri, M. et al. Optic atrophy plus phenotype due to mutations in the OPA1 gene: two more Italian families. J. Neurol. Sci. 315, 146–149 (2012). 108. Zuchner, S. et al. Mutations in the mitochondrial GTPase mitofusin 2 cause Charcot–Marie–Tooth neuropathy type 2A. Nat. Genet. 36, 449–451 (2004). 109. Feely, S. M. et al. MFN2 mutations cause severe phenotypes in most patients with CMT2A. Neurology 76, 1690–1696 (2011). 110. Verhoeven, K. et al. MFN2 mutation distribution and genotype/phenotype correlation in Charcot– Marie–Tooth type 2. Brain 129, 2093–2102 (2006). 111. Zuchner, S. et al. Axonal neuropathy with optic atrophy is caused by mutations in mitofusin 2. Ann. Neurol. 59, 276–281 (2006). 112. Polke, J. M. et al. Recessive axonal Charcot– Marie–Tooth disease due to compound heterozygous mitofusin 2 mutations. Neurology 77, 168–173 (2011). 113. Chen, H., McCaffery, J. M. & Chan, D. C. Mitochondrial fusion protects against neurodegeneration in the cerebellum. Cell 130, 548–562 (2007). 114. Chen, H. et al. Mitochondrial fusion is required for mtDNA stability in skeletal muscle and tolerance of mtDNA mutations. Cell 141, 280–289 (2010). 115. Larsson, N. G. et al. Mitochondrial transcription factor A is necessary for mtDNA maintenance and embryogenesis in mice. Nat. Genet. 18, 231–236 (1998). 116. Garrido, N. et al. Composition and dynamics of human mitochondrial nucleoids. Mol. Biol. Cell 14, 1583–1596 (2003). 117. Holt, I. J. et al. Mammalian mitochondrial nucleoids: organizing an independently minded genome. Mitochondrion 7, 311–321 (2007). 118. Kaufman, B. A. et al. The mitochondrial transcription factor TFAM coordinates the assembly of multiple DNA molecules into nucleoid-like structures. Mol. Biol. Cell 18, 3225–3236 (2007). 119. Hudson, G. & Chinnery, P. F. Mitochondrial DNA polymerase-γ and human disease. Hum. Mol. Genet. 15, R244–R252 (2006). 120. McFarland, R., Taylor, R. W. & Turnbull, D. M. A neurological perspective on mitochondrial disease. Lancet Neurol. 9, 829–840 (2010).

12  |  ADVANCE ONLINE PUBLICATION

121. Schapira, A. H. Mitochondrial diseases. Lancet 379, 1825–1834 (2012). 122. Alberio, S., Mineri, R., Tiranti, V. & Zeviani, M. Depletion of mtDNA: syndromes and genes. Mitochondrion 7, 6–12 (2007). 123. Chinnery, P. F. & Zeviani, M. 155th ENMC workshop: polymerase gamma and disorders of mitochondrial DNA synthesis, 21–23 September 2007, Naarden, The Netherlands. Neuromuscul. Disord. 18, 259–267 (2008). 124. Suomalainen, A. & Isohanni, P. Mitochondrial DNA depletion syndromes—many genes, common mechanisms. Neuromuscul. Disord. 20, 429–437 (2010). 125. Del Bo, R. et al. Remarkable infidelity of polymerase γA associated with mutations in POLG1 exonuclease domain. Neurology 61, 903–908 (2003). 126. Nishigaki, Y., Marti, R., Copeland, W. C. & Hirano, M. Site-specific somatic mitochondrial DNA point mutations in patients with thymidine phosphorylase deficiency. J. Clin. Invest. 111, 1913–1921 (2003). 127. Wanrooij, S. et al. Twinkle and POLG defects enhance age-dependent accumulation of mutations in the control region of mtDNA. Nucleic Acids Res. 32, 3053–3064 (2004). 128. Yu‑Wai‑Man, P. et al. OPA1 mutations cause cytochrome c oxidase deficiency due to loss of wild-type mtDNA molecules. Hum. Mol. Genet. 19, 3043–3052 (2010). 129. Durham, S. E., Samuels, D. C., Cree, L. M. & Chinnery, P. F. Normal levels of wild-type mitochondrial DNA maintain cytochrome c oxidase activity for two pathogenic mitochondrial DNA mutations but not for m.3243A→G. Am. J. Hum. Genet. 81, 189–195 (2007). 130. Koopman, W. J., Distelmaier, F., Smeitink, J. A. & Willems, P. H. OXPHOS mutations and neurodegeneration. EMBO J. 32, 9–29 (2013). 131. Taylor, R. W., Schaefer, A. M., Barron, M. J., McFarland, R. & Turnbull, D. M. The diagnosis of mitochondrial muscle disease. Neuromuscul. Disord. 14, 237–245 (2004). 132. Rouzier, C. et al. The MFN2 gene is responsible for mitochondrial DNA instability and optic atrophy ‘plus’ phenotype. Brain 135, 23–34 (2012). 133. Zeviani, M. OPA1 mutations and mitochondrial DNA damage: keeping the magic circle in shape. Brain 131, 314–317 (2008). 134. Vidoni, S., Zanna, C., Rugolo, M., Sarzi, E. & Lenaers, G. Why mitochondria must fuse to maintain their genome integrity. Antioxid. Redox Signal. 19, 379–388 (2013). 135. Stewart, J. D. et al. OPA1 in multiple mitochondrial DNA deletion disorders. Neurology 71, 1829–1831 (2008). 136. Elachouri, G. et al. OPA1 links human mitochondrial genome maintenance to mtDNA replication and distribution. Genome Res. 21, 12–20 (2011). 137. Payne, B. A. et al. Universal heteroplasmy of human mitochondrial DNA. Hum. Mol. Genet. 22, 384–390 (2013). 138. Sitarz, K. S. et al. OPA1 mutations induce mtDNA proliferation in leukocytes of patients with dominant optica atrophy. Neurology 79, 1515–1517 (2012). 139. Iommarini, L. et al. Revisiting the issue of mitochondrial DNA content in optic mitochondriopathies. Neurology 79, 1517–1519 (2012). 140. Sitarz, K. S. et al. MFN2 mutations cause compensatory mitochondrial DNA proliferation. Brain 135, e219 (2012). 141. Renaldo, F. et al. MFN2, a new gene responsible for mitochondrial DNA depletion. Brain 135, e223 (2012).

www.nature.com/nrneurol © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS 142. Yu‑Wai‑Man, P. & Chinnery, P. F. Dysfunctional mitochondrial maintenance: what breaks the circle of life? Brain 135, 9–11 (2012). 143. Salinas, S., Proukakis, C., Crosby, A. & Warner, T. T. Hereditary spastic paraplegia: clinical features and pathogenetic mechanisms. Lancet Neurol. 7, 1127–1138 (2008). 144. Blackstone, C., O’Kane, C. J. & Reid, E. Hereditary spastic paraplegias: membrane traffic and the motor pathway. Nat. Rev. Neurosci. 12, 31–42 (2011). 145. Timmerman, V., Clowes, V. E. & Reid, E. Overlapping molecular pathological themes link Charcot–Marie–Tooth neuropathies and hereditary spastic paraplegias. Exp. Neurol. 246, 14–25 (2013). 146. Harding, A. E. Classification of the hereditary ataxias and paraplegias. Lancet 1, 1151–1155 (1983). 147. Casari, G. et al. Spastic paraplegia and OXPHOS impairment caused by mutations in paraplegin, a nuclear-encoded mitochondrial metalloprotease. Cell 93, 973–983 (1998). 148. Ferreirinha, F. et al. Axonal degeneration in paraplegin-deficient mice is associated with abnormal mitochondria and impairment of axonal transport. J. Clin. Invest. 113, 231–242 (2004). 149. Klebe, S. et al. Spastic paraplegia gene 7 in patients with spasticity and/or optic neuropathy. Brain 135, 2980–2993 (2012). 150. van Gassen, K. L. et al. Genotype-phenotype correlations in spastic paraplegia type 7: a study in a large Dutch cohort. Brain 135, 2994–3004 (2012). 151. Greaves, L. C. et al. Mitochondrial DNA defects and selective extraocular muscle involvement in CPEO. Invest. Opthalmol. Vis. Sci. 51, 3340–3346 (2010). 152. Pfeffer, G. et al. Mutations in the SPG7 gene cause chronic progressive external ophthalmoplegia through disordered mitochondrial DNA maintenance. Brain 137, 1323–1336 (2014). 153. Wedding, I. M. et al. Spastic paraplegia type 7 is associated with multiple mitochondrial DNA deletions. PLoS ONE 9, e86340 (2014). 154. Di Bella, D. et al. Mutations in the mitochondrial protease gene AFG3L2 cause dominant hereditary ataxia SCA28. Nat. Genet. 42, 313–320 (2010). 155. Pierson, T. M. et al. Whole-exome sequencing identifies homozygous AFG3L2 mutations in a spastic ataxia-neuropathy syndrome linked to mitochondrial m‑AAA proteases. PLoS Genet. 7, e1002325 (2011). 156. Maltecca, F. et al. Respiratory dysfunction by AFG3L2 deficiency causes decreased mitochondrial calcium uptake via organellar network fragmentation. Hum. Mol. Genet. 21, 3858–3870 (2012). 157. Almajan, E. R. et al. AFG3L2 supports mitochondrial protein synthesis and Purkinje cell survival. J. Clin. Invest. 122, 4048–4058 (2012). 158. Kondadi, A. K. et al. Loss of the m‑AAA protease subunit AFG3L2 causes mitochondrial transport defects and tau hyperphosphorylation. EMBO J. 33, 1011–1026 (2014). 159. Nguyen, T. T. et al. Loss of Miro1-directed mitochondrial movement results in a novel murine model for neuron disease. Proc. Natl Acad. Sci. USA 111, E3631–E3640 (2014). 160. Paisan-Ruiz, C. et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron 44, 595–600 (2004). 161. Macdonald, M. E. et al. A novel gene containing a trinucleotide repeat that is expanded and

unstable on Huntington’s-disease chromosomes. Cell 72, 971–983 (1993). 162. Engert, J. C. et al. ARSACS, a spastic ataxia common in northeastern Quebec, is caused by mutations in a new gene encoding an 11.5‑kb ORF. Nat. Genet. 24, 120–125 (2000). 163. Costa, V. et al. Mitochondrial fission and cristae disruption increase the response of cell models of Huntington’s disease to apoptotic stimuli. EMBO Mol. Med. 2, 490–503 (2010). 164. Song, W. et al. Mutant huntingtin binds the mitochondrial fission GTPase dynamin-related protein‑1 and increases its enzymatic activity. Nat. Med. 17, 377–382 (2011). 165. Girard, M. et al. Mitochondrial dysfunction and Purkinje cell loss in autosomal recessive spastic ataxia of Charlevoix–Saguenay (ARSACS). Proc. Natl Acad. Sci. USA 109, 1661–1666 (2012). 166. Wang, X. et al. LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. Hum. Mol. Genet. 21, 1931–1944 (2012). 167. Shirendeb, U. et al. Abnormal mitochondrial dynamics, mitochondrial loss and mutant huntingtin oligomers in Huntington’s disease: implications for selective neuronal damage. Hum. Mol. Genet. 20, 1438–1455 (2011). 168. Shirendeb, U. P. et al. Mutant huntingtin’s interaction with mitochondrial protein Drp1 impairs mitochondrial biogenesis and causes defective axonal transport and synaptic degeneration in Huntington’s disease. Hum. Mol. Genet. 21, 406–420 (2012). 169. Manczak, M., Calkins, M. J. & Reddy, P. H. Impaired mitochondrial dynamics and abnormal interaction of amyloid beta with mitochondrial protein Drp1 in neurons from patients with Alzheimer’s disease: implications for neuronal damage. Hum. Mol. Genet. 20, 2495–2509 (2011). 170. Manczak, M. & Reddy, P. H. Abnormal interaction between the mitochondrial fission protein Drp1 and hyperphosphorylated tau in Alzheimer’s disease neurons: implications for mitochondrial dysfunction and neuronal damage. Hum. Mol. Genet. 21, 2538–2547 (2012). 171. Calkins, M. J., Manczak, M., Mao, P., Shirendeb, U. & Reddy, P. H. Impaired mitochondrial biogenesis, defective axonal transport of mitochondria, abnormal mitochondrial dynamics and synaptic degeneration in a mouse model of Alzheimer’s disease. Hum. Mol. Genet. 20, 4515–4529 (2011). 172. Schon, E. A. & Area-Gomez, E. Mitochondriaassociated ER membranes in Alzheimer disease. Mol. Cell. Neurosci. 55, 26–36 (2013). 173. Hedskog, L. et al. Modulation of the endoplasmic reticulum-mitochondria interface in Alzheimer’s disease and related models. Proc. Natl Acad. Sci. USA 110, 7916–7921 (2013). 174. Bannwarth, S. et al. A mitochondrial origin for frontotemporal dementia and amyotrophic lateral sclerosis through CHCHD10 involvement. Brain 137, 2329–2345 (2014). 175. Chaussenot, A. et al. Screening of CHCHD10 in a French cohort confirms the involvement of this gene in frontotemporal dementia with amyotrophic lateral sclerosis patients. Neurobiol. Aging 35, 2884.e1–2884.e4 (2014). 176. Yu‑Wai‑Man, P., Votruba, M., Moore, A. T. & Chinnery, P. F. Treatment strategies for inherited optic neuropathies: past, present and future. Eye (Lond.) 28, 527–537 (2014). 177. Yu‑Wai‑Man, P., Griffiths, P. G. & Chinnery, P. F. Mitochondrial optic neuropathies—disease mechanisms and therapeutic strategies. Prog. Retin. Eye Res. 30, 81–114 (2011).

NATURE REVIEWS | NEUROLOGY

178. Carelli, V., La Morgia, C. & Sadun, A. A. Mitochondrial dysfunction in optic neuropathies: animal models and therapeutic options. Curr. Opin. Neurol. 26, 52–58 (2013). 179. Chen, H. & Chan, D. C. Mitochondrial dynamicsfusion, fission, movement, and mitophagy-in neurodegenerative diseases. Hum. Mol. Genet. 18, R169–R176 (2009). 180. Sarkar, S. et al. Small molecules enhance autophagy and reduce toxicity in Huntington’s disease models. Nat. Chem. Biol. 3, 331–338 (2007). 181. Bove, J., Martinez-Vicente, M. & Vila, M. Fighting neurodegeneration with rapamycin: mechanistic insights. Nat. Rev. Neurosci. 12, 437–452 (2011). 182. Harris, H. & Rubinsztein, D. C. Control of autophagy as a therapy for neurodegenerative disease. Nat. Rev. Neurol. 8, 108–117 (2012). 183. Man, P. Y., Turnbull, D. M. & Chinnery, P. F. Leber hereditary optic neuropathy. J. Med. Genet. 39, 162–169 (2002). 184. Klopstock, T. et al. A randomized placebocontrolled trial of idebenone in Leber’s hereditary optic neuropathy. Brain 134, 2677–2686 (2011). 185. Carelli, V. et al. Idebenone treatment In Leber’s hereditary optic neuropathy. Brain 134, e188 (2011). 186. Klopstock, T. et al. Persistence of the treatment effect of idebenone in Leber’s hereditary optic neuropathy. Brain 136, e230 (2013). 187. Sadun, A. A. et al. Effect of EPI‑743 on the clinical course of the mitochondrial disease Leber hereditary optic neuropathy. Arch. Neurol. 69, 331–338 (2012). 188. Cipolat, S., de Brito, O. M., Dal Zilio, B. & Scorrano, L. OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc. Natl Acad. Sci. USA 101, 15927–15932 (2004). 189. MacAskill, A. F. et al. Miro1 is a calcium sensor for glutamate receptor-dependent localization of mitochondria at synapses. Neuron 61, 541–555 (2009). 190. Misko, A., Jiang, S., Wegorzewska, I., Milbrandt, J. & Baloh, R. H. Mitofusin 2 Is necessary for transport of axonal mitochondria and interacts with the Miro/Milton complex. J. Neurosci. 30, 4232–4240 (2010). 191. Cassidy-Stone, A. et al. Chemical inhibition of the mitochondrial division dynamin reveals its role in Bax/Bak-dependent mitochondrial outer membrane permeabilization. Dev. Cell 14, 193–204 (2008). 192. Cereghetti, G. M., Costa, V. & Scorrano, L. Inhibition of Drp1-dependent mitochondrial fragmentation and apoptosis by a polypeptide antagonist of calcineurin. Cell Death Differ. 17, 1785–1794 (2010). 193. Iwasawa, R., Mahul-Mellier, A.‑L., Datler, C., Pazarentzos, E. & Grimm, S. Fis1 and Bap31 bridge the mitochondria-ER interface to establish a platform for apoptosis induction. EMBO J. 30, 556–568 (2011). 194. Wells, T. et al. Opa3, a novel regulator of mitochondrial function, controls thermogenesis and abdominal fat mass in a mouse model for Costeff syndrome. Hum. Mol. Genet. 21, 4836–4844 (2012). 195. Huizing, M. et al. OPA3, mutated in 3‑methylglutaconic aciduria type III, encodes two transcripts targeted primarily to mitochondria. Mol. Genet. Metab. 100, 149–154 (2010). 196. Ryu, S.‑W., Jeong, H. J., Choi, M., Karbowski, M. & Choi, C. Optic atrophy 3 as a protein of the mitochondrial outer membrane induces mitochondrial fragmentation. Cell. Mol. Life Sci. 67, 2839–2850 (2010).

ADVANCE ONLINE PUBLICATION  |  13 © 2014 Macmillan Publishers Limited. All rights reserved

REVIEWS 197. Niemann, A., Ruegg, M., La Padula, V., Schenone, A. & Suter, U. Ganglioside-induced differentiation associated protein 1 is a regulator of the mitochondrial network: new implications for Charcot–Marie–Tooth disease. J. Cell Biol. 170, 1067–1078 (2005). 198. Niemann, A., Wagner, K. M., Ruegg, M. & Suter, U. GDAP1 mutations differ in their effects on mitochondrial dynamics and apoptosis depending on the mode of inheritance. Neurobiol. Dis. 36, 509–520 (2009). 199. Claramunt, R. et al. Genetics of Charcot–Marie– Tooth disease type 4A: mutations, inheritance, phenotypic variability, and founder effect. J. Med. Genet. 42, 358–365 (2005). 200. Waterham, H. R. et al. A lethal defect of mitochondrial and peroxisomal fission. N. Engl. J.Med. 356, 1736–1741 (2007). 201. Chang, C.‑R. et al. A lethal de novo mutation in the middle domain of the dynamin-related GTPase Drp1 impairs higher order assembly and mitochondrial division. J. Biol. Chem. 285, 32494–32503 (2010). 202. Reynier, P. et al. OPA3 gene mutations responsible for autosomal dominant optic atrophy and cataract. J. Med. Genet. 41, e110 (2004). 203. Baxter, R. V. et al. Ganglioside-induced differentiation-associated protein‑1 is mutant in Charcot‑Marie‑Tooth disease type 4A/8q21. Nat. Genet. 30, 21–22 (2002). 204. Anikster, Y., Kleta, R., Shaag, A., Gahl, W. A. & Elpeleg, O. Type III 3‑methylglutaconic aciduria (optic atrophy plus syndrome, or Costeff optic atrophy syndrome): identification of the OPA3 gene and its founder mutation in Iraqi Jews. Am. J. Hum. Genet. 69, 1218–1224 (2001). 205. Yarosh, W. et al. The molecular mechanisms of OPA1-mediated optic atrophy in Drosophila model and prospects for antioxidant treatment. PLoS Genet. 4, e6 (2008). 206. Tang, S., Le, P. K., Tse, S., Wallace, D. C. & Huang, T. Heterozygous mutation of Opa1 in Drosophila shortens lifespan mediated through increased reactive oxygen species production. PLoS ONE 4, e4492 (2009). 207. Shahrestani, P. et al. Heterozygous mutation of Drosophila Opa1 causes the development of multiple organ abnormalities in an agedependent and organ-specific manner. PLoS ONE 4, e6867 (2009). 208. Rahn, J. J., Stackley, K. D. & Chan, S. S. Opa1 is required for proper mitochondrial metabolism in early development. PLoS ONE 8, e59218 (2013).

209. Davies, V. J. et al. Opa1 deficiency in a mouse model of autosomal dominant optic atrophy impairs mitochondrial morphology, optic nerve structure and visual function. Hum. Mol. Genet. 16, 1307–1318 (2007). 210. Alavi, M. V. et al. A splice site mutation in the murine Opa1 gene features pathology of autosomal dominant optic atrophy. Brain 130, 1029–1042 (2007). 211. Alavi, M. V. et al. Subtle neurological and metabolic abnormalities in an Opa1 mouse model of autosomal dominant optic atrophy. Exp. Neurol. 220, 404–409 (2009). 212. Moore, B. A., Aviles, G. D., Larkins, C. E., Hillman, M. J. & Caspary, T. Mitochondrial retention of Opa1 is required for mouse embryogenesis. Mamm. Gen. 21, 350–360 (2010). 213. Sarzi, E. et al. The human OPA1delTTAG mutation induces premature age-related systemic neurodegeneration in mouse. Brain 135, 3599–3613 (2012). 214. Williams, P. A., Morgan, J. E. & Votruba, M. Opa1 deficiency in a mouse model of dominant optic atrophy leads to retinal ganglion cell dendropathy. Brain 133, 2942–2951 (2010). 215. Williams, P. A. et al. Opa1 is essential for retinal ganglion cell synaptic architecture and connectivity. Brain 135, 493–505 (2012). 216. Yu‑Wai‑Man, P. et al. Secondary mtDNA defects do not cause optic nerve dysfunction in a mouse model of dominant optic atropy. Invest. Opthalmol. Vis. Sci. 50, 4561–4566 (2009). 217. Chen, Y. et al. Mitofusin 2‑containing mitochondrial-reticular microdomains direct rapid cardiomyocyte bioenergetic responses via interorganelle Ca2+ crosstalk. Circ. Res. 111, 863–875 (2012). 218. Dorn, G. W. 2nd et al. MARF and Opa1 control mitochondrial and cardiac function in Drosophila. Circ. Res. 108, 12–17 (2011). 219. Eschenbacher, W. H. et al. Two rare human mitofusin 2 mutations alter mitochondrial dynamics and induce retinal and cardiac pathology in Drosophila. PLoS ONE 7, e44296 (2012). 220. Deng, H., Dodson, M. W., Huang, H. & Guo, M. The Parkinson’s disease genes pink1 and parkin promote mitochondrial fission and/or inhibit fusion in Drosophila. Proc. Natl Acad. Sci. USA 105, 14503–14508 (2008). 221. Chapman, A. L., Bennett, E. J., Ramesh, T. M., De Vos, K. J. & Grierson, A. J. Axonal transport defects in a mitofusin 2 loss of function model

14  |  ADVANCE ONLINE PUBLICATION

of Charcot–Marie–Tooth disease in zebrafish. PLoS ONE 8, e67276 (2013). 222. Vettori, A. et al. Developmental defects and neuromuscular alterations due to mitofusin 2 gene (MFN2) silencing in zebrafish: a new model for Charcot–Marie–Tooth type 2A neuropathy. Neuromuscul. Disord. 21, 58–67 (2011). 223. Chen, L. et al. OPA1 mutation and late-onset cardiomyopathy: mitochondrial dysfunction and mtDNA instability. J. Am. Heart Assoc. 1, e003012 (2012). 224. Lee, S. et al. Mitofusin 2 is necessary for striatal axonal projections of midbrain dopamine neurons. Hum. Mol. Genet. 21, 4827–4835 (2012). 225. Pham, A. H., Meng, S., Chu, Q. N. & Chan, D. C. Loss of Mfn2 results in progressive, retrograde degeneration of dopaminergic neurons in the nigrostriatal circuit. Hum. Mol. Genet. 21, 4817–4826 (2012). 226. Detmer, S. A., Velde, C. V., Cleveland, D. W. & Chan, D. C. Hindlimb gait defects due to motor axon loss and reduced distal muscles in a transgenic mouse model of Charcot–Marie– Tooth type 2A. Hum. Mol. Genet. 17, 367–375 (2008). 227. Cartoni, R. et al. Expression of mitofusin 2(R94Q) in a transgenic mouse leads to Charcot–Marie–Tooth neuropathy type 2A. Brain 133, 1460–1469 (2010). 228. Guillet, V. et al. Bioenergetic defect associated with mKATP channel opening in a mouse model carrying a mitofusin 2 mutation. FASEB J. 25, 1618–1627 (2011). Acknowledgements P.Y.-W.-M. is a Medical Research Council (MRC, UK) Clinician Scientist. He also receives funding from Fight for Sight (UK) and the UK National Institute of Health Research (NIHR) as part of the Rare Diseases Translational Research Collaboration. V.C. is supported by Telethon-Italy grants GPP1005, GGP06233 and GGP11182. P.F.C. is a Wellcome Trust Senior Fellow in Clinical Science and a UK NIHR Senior Investigator who also receives funding from the MRC and the NIHR Biomedical Research Centre for Ageing and AgeRelated Disease award to the Newcastle upon Tyne Hospitals NHS Foundation Trust. Author contributions F.B. and P.Y.-W.-M. researched data for and wrote the article. All the authors provided substantial contributions to the discussion of content and to editing the manuscript before submission.

www.nature.com/nrneurol © 2014 Macmillan Publishers Limited. All rights reserved

Disturbed mitochondrial dynamics and neurodegenerative disorders.

Mitochondria form a highly interconnected tubular network throughout the cell via a dynamic process, with mitochondrial segments fusing and breaking a...
1MB Sizes 0 Downloads 19 Views