[go: up one dir, main page]

0% found this document useful (0 votes)
24 views12 pages

Enfermedad Mitocondrial

1. Mitochondrial diseases are some of the most common inherited neurometabolic disorders. Understanding of their clinical presentations, genetic causes, diagnoses, and treatments has advanced significantly in recent years. 2. National disease cohorts and international collaborations have expanded knowledge of phenotypes and natural histories. High-throughput sequencing has improved genetic diagnoses. 3. While management focuses on surveillance and symptom treatment, new efforts explore drug repurposing, novel molecules, and gene therapies. Reproductive technologies also offer prevention options.

Uploaded by

Osvaldo Cortés
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
24 views12 pages

Enfermedad Mitocondrial

1. Mitochondrial diseases are some of the most common inherited neurometabolic disorders. Understanding of their clinical presentations, genetic causes, diagnoses, and treatments has advanced significantly in recent years. 2. National disease cohorts and international collaborations have expanded knowledge of phenotypes and natural histories. High-throughput sequencing has improved genetic diagnoses. 3. While management focuses on surveillance and symptom treatment, new efforts explore drug repurposing, novel molecules, and gene therapies. Reproductive technologies also offer prevention options.

Uploaded by

Osvaldo Cortés
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 12

Review

Mitochondrial disease in adults: recent advances and


future promise
Yi Shiau Ng, Laurence A Bindoff, Gráinne S Gorman, Thomas Klopstock, Cornelia Kornblum, Michelangelo Mancuso, Robert McFarland,
Carolyn M Sue, Anu Suomalainen, Robert W Taylor, David R Thorburn, Doug M Turnbull

Mitochondrial diseases are some of the most common inherited neurometabolic disorders, and major progress has Lancet Neurol 2021; 20: 573–84
been made in our understanding, diagnosis, and treatment of these conditions in the past 5 years. Development of Wellcome Centre for
national mitochondrial disease cohorts and international collaborations has changed our knowledge of the spectrum Mitochondrial Research,
Translational and Clinical
of clinical phenotypes and natural history of mitochondrial diseases. Advances in high-throughput sequencing
Research Institute,
technologies have altered the diagnostic algorithm for mitochondrial diseases by increasingly using a genetics-first Faculty of Medical Sciences,
approach, with more than 350 disease-causing genes identified to date. While the current management strategy for Newcastle University,
mitochondrial disease focuses on surveillance for multisystem involvement and effective symptomatic treatment, Newcastle upon Tyne, UK
(Y S Ng PhD,
new endeavours are underway to find better treatments, including repurposing current drugs, use of novel small
Prof G S Gorman PhD,
molecules, and gene therapies. Developments made in reproductive technology offer women the opportunity to Prof R McFarland PhD,
prevent transmission of DNA-related mitochondrial disease to their children. Prof R W Taylor PhD,
Prof D M Turnbull PhD); NHS
Introduction techniques, such as mitochondrial replacement therapy, Highly Specialised Service for
Rare Mitochondrial Disorders
Inherited mitochondrial diseases are complex neuro­ has provided hope for mothers with mtDNA-related (Y S Ng, Prof G S Gorman,
genetic conditions that present clinical, diagnostic, and disease. Prof R McFarland,
treat­
ment challenges for neurologists. The substantial Prof R W Taylor,

clinical variability and frequent involvement of other Clinical aspects of mitochondrial disease Prof D M Turnbull) and
Directorate of Neurosciences,
tissues means that recognition of potential mitochondrial Since mitochondria are essential organelles in all human Royal Victoria Infirmary
disease might be delayed. Establishing the genetic diag­ cells, mitochondrial disease can affect all organs; involve­ (Y S Ng, Prof G S Gorman),
nosis of mitochondrial disease can be difficult because ment of the nervous system, however, can be substantial. Newcastle upon Tyne Hospitals
NHS Foundation Trust,
mito­chondria are under dual genetic control by the Initially, many mitochondrial diseases were defined as a Newcastle upon Tyne, UK;
nuclear and mitochondrial genomes. For individuals who cluster of clinical syndromic features—eg, mitochondrial Department of Clinical
present with a recognised clinical syndrome or a positive myopathy, encephalopathy, lactic acidosis, and stroke- Medicine, University of Bergen,
family history, genetic diagnosis is straightforward, but for like episodes (MELAS) syndrome and Kearns-Sayre Bergen, Norway
(Prof L A Bindoff PhD);
many people, mitochondrial disease is just part of the syndrome.1 However, only a few individuals show the full Neuro-SysMed, Department of
differential diagnosis of complex neurogenetic disorders. spectrum of signs at presentation. Key to recognising Neurology, Haukeland
Nevertheless, establishing the genetic cause is crucial for patients with mitochondrial disease has been the use of University Hospital, Bergen,
both prog­nosis and genetic counselling, particularly in the deep phenotyping, full characterisation of clinical features, Norway (Prof L A Bindoff);
Department of Neurology,
case of mitochondrial DNA (mtDNA)-related disease, and identification of other organ involve­ment (appendix Friedrich-Baur-Institute, LMU
which presents unique challenges for affected women in pp 2–4). The initial clinical mani­festation of mitochondrial Hospital, Ludwig Maximilians
terms of reproductive options. In past years, even with a disease might be neuro­logical, or not, and can present University, Munich, Germany
diagnosis of mitochondrial disease, few treatment options acutely, subacutely, or evolve slowly. (Prof T Klopstock MD); German
Center for Neurodegenerative
were available and there were no care guidelines. Diseases , Munich, Germany
The aim of this Review is to highlight the substantial Acute and subacute neurological presentations (Prof T Klopstock); Munich
progress made in mitochondrial disease research in the Stroke-like episodes Cluster for Systems Neurology,
Munich, Germany
past few years, showcasing the important aspects for Stroke-like episodes in individuals with mitochondrial
(Prof T Klopstock); Department
neurologists to recognise, diagnose, and manage adult disease are characterised by headache, nausea and vomit­ of Neurology, Neuromuscular
patients with mitochondrial disease. Our understanding ing, encephalopathy, focal-onset seizures, or psychiatric Disease Section
of the clinical spectrum of mitochondrial disease has symptoms before development of a focal neurological (C Kornblum MD), and Centre
for Rare Diseases (C Kornblum),
progressed with development of large patient cohorts and deficit. Epilepsia partialis continua and, rarely, generalised
University Hospital Bonn,
recognition of genotype-specific symptoms. Advances in status epilepticus might also occur during the stroke-like Bonn, Germany; Department of
genetic sequencing technology have led to recognition episode. Headache and visual disturbance are often Clinical and Experimental
that defects in more than 350 genes of both mitochondrial experi­enced during the prodrome. A consensus statement Medicine, Neurological
Institute, University of Pisa,
and nuclear origin might cause mitochondrial disease, proposed that “a mitochondrial stroke-like episode is a
Italy (Prof M Mancuso PhD);
thereby transform­ ing diagnosis. Although no cure for subacute, evolving brain syndrome driven by seizure Department of Neurogenetics,
mito­chon­drial dis­ease currently exists, consensus-based activity in genetically determined mitochondrial disease”.2 Kolling Institute, Faculty of
clinical guide­lines are available to help manage patients Although stroke-like episodes were originally described Medicine and Health,
University of Sydney,
and, for some mitochondrial diseases, potential treatments in individuals younger than 40 years, late-onset presenta­
Sydney, NSW, Australia
are emerg­ing. An increasing number of clinical trials are tion is now recognised.3 Stroke-like episodes are the (Prof C M Sue PhD); Department
investi­
gating new or repurposed small molecules and defining feature of MELAS syndrome, with the pathogenic of Neurology, Royal North
gene therapy. Finally, the development of reproductive m.3243A→G MT-TL1 gene variant accounting for up Shore Hospital, Northern

www.thelancet.com/neurology Vol 20 July 2021 573


Review

Sydney Local Health District, Patients with the m.8344A→G variant can have both myo­
St Leonards, NSW, Australia A B C D clonic epilepsy and subcortical myoclonus without any
(Prof C M Sue); Research
Program in Stem Cells
EEG correlates.11,12
and Metabolism,
Faculty of Medicine Metabolic decompensation of brainstem function
(Prof A Suomalainen PhD) and Leigh syndrome is the most common presentation of
Neuroscience Centre, HiLife
(Prof A Suomalainen),
paediatric mitochondrial disease,13 although some children
E F G H present late and those who are mildly affected can survive
University of Helsinki, Helsinki,
Finland; Helsinki University into adulthood.14 Patients with Leigh syndrome can develop
Hospital, HUSlab, Helsinki, subacute brainstem dysfunction when metabolically chal­
Finland (Prof A Suomalainen);
Murdoch Children’s
lenged (eg, during intercurrent infections; figure 1).
Research Institute Features of Leigh syndrome include ophthalmoplegia,
(Prof D R Thorburn PhD), and Figure 1: Neuroimaging features of mitochondrial diseases ataxia, dysphagia, dysarthria, central hypoventilation,
Victorian Clinical Genetics Stroke-like lesions involving the right temporal, occipital (A) and parietal lobes hyper­­somnolence, and autonomic instability. Brainstem
Services (Prof D R Thorburn), (B) with restricted diffusion (not shown) are seen in a 20-year-old man with
Royal Children’s Hospital, m.3243A→G-related mitochondrial myopathy, encephalopathy, lactic acidosis,
syndrome has also been reported in adult patients who
Melbourne, VIC, Australia; and stroke-like episodes syndrome. Severe cerebral atrophy (C) is evident in a initially presented with neuropathy and ataxia related
Department of Paediatrics, 50-year-old man with the m.3243A→G variant who had experienced recurrent to MT-ATP6 pathogenic variants, without a pre-existing
University of Melbourne, stroke-like episodes since his mid-30s. Asymmetrical medullary lesions with diag­nosis of Leigh syndrome.15 Partial recovery of brain­
Melbourne, VIC, Australia restricted diffusion (D) are seen in a 25-year-old woman with the
(Prof D R Thorburn) m.9185T→C MT-ATP6 variant who presented with subacute onset dysphagia,
stem function occurs in some patients who survive the
Correspondence to: central hypoventilation, and type 2 respiratory failure; her initial clinical metabolic crisis.
Prof Doug M Turnbull, Wellcome phenotype was teenage-onset progressive ataxia and neuropathy. Symmetrical
Centre for Mitochondrial T2-hyperintensities in the basal ganglia (E) are shown in a woman in her Subacute visual loss and optic neuropathy
Research, Translational and mid-40s who had childhood-onset Leigh syndrome related to the
m.8993T→C MT-ATP6 variant. Symmetrical leukodystrophy (F) is identified in a
Subacute, painless, and progressive impairment of central
Clinical Research Institute,
Faculty of Medical Sciences, 13-year-old boy with Kearns-Sayre syndrome due to a single, large-scale vision is a typical presenting feature of the mitochon­
Newcastle University, mitochondrial DNA deletion. Asymmetrical reduction in dopamine uptake (G) is drial disease Leber hereditary optic neuropathy (LHON).
Newcastle upon Tyne seen on a dopamine transporter PET scan of a 45-year-old man with recessive Typically, onset of this disorder is unilateral, with the
NE2 4HH, UK POLG variants who presented with chronic progressive external ophthalmoplegia,
ataxia, and parkinsonism. Diffuse lipomatosis (H) is seen in the shoulder girdles
second eye becoming affected within weeks or months.
doug.turnbull@ncl.ac.uk
and cervical region in a 44-year-old woman with the m.8344A→G variant. Male carriers of one of the three primary pathogenic
See Online for appendix
variants (m.3460G→A MT-ND1, m.11778G→A MT-ND4,
to 80% of cases; pathogenic variants in other mtDNA and m.14484T→C MT-ND6) associated with LHON are
genes and in the nuclear gene POLG are also known to more likely to be affected by visual impairment than are
cause stroke-like episodes.2,4 women, and smoking and excess alcohol are trigger factors
On conventional MRI, stroke-like lesions associated for visual loss.16 Rapid deterioration of visual function can
with mitochondrial disease typically do not correspond to raise suspicion for inflammatory optic neuritis initially.
vascular territories and involve cortex and juxtacortical Therefore, early diagnosis of LHON could avoid the initia­
white matter (figure 1). MRI proton spectroscopy shows a tion of long-term immunotherapy, which is unnecessary,
lactate peak both in the affected regions and in ventricular ineffective, and potentially detrimental. Clinical features of
CSF or other apparently unaffected brain areas. Lesions LHON are young age (with peak age of onset in the second
might spread over days, weeks, and even months. Some and third decade of life), male sex, absence of pain with eye
signal abnormalities resolve completely; however, most movements, no relative afferent pupillary deficit, maternal
severe lesions develop into cortical laminar necrosis, family history of blindness, normal MRI and CSF results,
gliosis, and atrophy, potentially leading to progressive and no benefit from steroids. Typical ophthal­mological
physi­cal and cognitive decline. Based on the clinical and findings during the acute phase of LHON include hyper­
MRI features, clinicians should think about a mitochondrial aemia of the optic disc, oedema of the peripapillary retinal
stroke-like episode as a main differential diagnosis to nerve fibre layer, retinal telangiectasia, and absence of
atypical recurrent ischaemic stroke,5 encephalitis with a leakage on fluorescein angiography.17 A clinical study using
nega­tive infectious and autoimmune screen,6 and posterior optical coherence tomography showed that thicken­ing of
reversible encephalopathy syndrome.7 the retinal nerve fibre layer preceded angiopathic changes,
measured by an increase in choroidal thickness during the
Epilepsy acute phase of LHON.18
Epilepsy is a common feature of mitochondrial dis­ ­
ease8,9 with both generalised and focal seizures observed. Chronic neurological presentations
In individuals with m.3243A→G and POLG pathogenic Chronic progressive external ophthalmoplegia (CPEO)
variants, seizure activity leading to development of a CPEO is one of the most common adult clinical presenta­
stroke-like episode is typically refractory or super- tions of mitochondrial disease. Only a small proportion of
refractory to phar­maco­logical treatment, including general patients (<30%)19 report diplopia (which can affect reading
anaesthesia—particularly in POLG-related epilepsy.10 ability, balance, and movement; and an early onset of

574 www.thelancet.com/neurology Vol 20 July 2021


Review

which might result in chronic suppression and amblyopia) other mitochondrial diseases—eg, in m.8344A→G-related
due to the insidious nature of disease progression. or m.3243A→G-related disease.
Depend­ ing on the genetic defect, CPEO can occur in
isolation or with other neurological features and multi­ Other chronic neurological presentations
system involvement. Seronegative myasthenia gravis Cerebellar or sensory ataxia, peripheral neuropathy,
(without AchR and MuSK antibodies) is an important migraine, cognitive impairment, spasticity, and extra­pyra­
differential diagnosis, and patients with CPEO can midal movement disorders including tremor, parkinson­
have minor neuromuscular junction electro­physiological ism, dystonia, and chorea can also be presenting features
abnor­mali­ties.20 Other differ­
ential diagnoses21 include of mitochondrial disease. In some patients, the diagnosis
oculo­pharyngeal mus­ cular dys­
trophy, congenital myas­ of mitochondrial disease is straightforward because of
thenic syndrome, congenital myopathy, and myotonic other classic symptoms, such as a combination of CPEO
dystrophy. and parkinsonism in POLG-related disease, or maternally
inherited diabetes and deafness in m.3243A→G-related
Myopathy disease. In other patients, mitochondrial disease is just
Features that can alert the clinician to mitochondrial one of several diagnostic possibilities. Making a diagnosis
myopathies include fatiguability, myalgia (often exercise- in these patients can be challenging, particularly when
induced), exercise intolerance, and lactic acidaemia. disease onset is late and there is no family history.
Many patients with CPEO also develop myopathy, and Moreover, many aspects of mitochondrial disease are still
some mitochondrial diseases might have concomitant not understood, such as selective tissue involvement and
peri­pheral neuropathy that can be axonal or demyelinat­ the unique patterns of clinical manifestations that occur
ing, including POLG-related disease and mitochondrial with specific gene variants. In such cases, development of
neurogastrointestinal encephalomyopathy.22 Mild bulbar large cohorts and natural history studies should be help­
weakness is not uncommon;23 however, some patients ful, particularly when combined with mechanistic studies.
develop severe dysphagia and are at risk of recurrent
aspiration. Although an elevated creatinine kinase level Non-neurological features
(typically >1000 IU/L) is a recognised laboratory finding Management of patients with mitochondrial disease is
of mitochondrial disease, rhabdomyolysis manifesting complicated by the recognition that multiple organs can
with myoglobinuria is very rare.24 Severe neuromuscular be involved. Moreover, systemic features might be more
weakness (including diaphragmatic weakness) requiring prominent than neurological signs (appendix pp 2–4).
ventilatory support is infrequent in adult mitochondrial
diseases and is typically associated with specific genetic Recent advances in diagnosis and biomarkers
defects such as TK2 deficiency25 and the m.8344A→G Genomics and biomarker development are transforming
mutation. the diagnosis of mitochondrial disease.30,31 Securing a
precise genetic diagnosis is important since knowledge
Sensorineural hearing loss about genetic variants can facilitate the targeting of treat­
To be suggestive of a mitochondrial disorder, hearing ments, help with management of and surveillance for
loss is generally young-onset with other systemic fea­ specific complications, inform counselling for reproduc­
tures associated with a mitochondrial dis­ease,26 and a tive options, and enable patients to be enrolled into
maternal pattern of transmission. Some individuals with appropriate clinical trials. However, access to diagnostic
specific mtDNA pathogenic variants (eg, a homoplasmic tests remains inequitable in some countries.
m.1555A→G variant) develop deafness after exposure to
aminoglycoside antibiotics. The onset of hearing loss is Mitochondrial biomarkers
usually after postlingual develop­ment, indicating that Validated biomarkers now make it possible to avoid
cochlear implantation could be useful as a treatment in invasive muscle biopsies in some cases.32 Two novel
those patients with profound hearing loss.27 biomarkers of mitochondrial dysfunction have been
identified, FGF2133 and GDF15.34 In adults, increased
Optic neuropathy amounts of FGF21 and GDF15 are associated with muscle-
Around 60% of cases of autosomal-dominant optic manifesting mitochondrial translational defects (patho­
atrophy are due to OPA1 genetic defects, which have an genic mt-tRNA variants and mtDNA deletions) and levels
estimated prevalence of 1 in 25 000 people.28 Optic atrophy of the biomarkers correlate with the severity of the
is characterised by a slowly progressive, childhood-onset biochemical defect in muscle.35 Both FGF21 and GDF15
optic neuropathy with moderate-to-severe loss of visual have better sensitivity and specificity for adult mito­
acuity. At least a fifth of patients with OPA1 pathogenic chondrial disease than do other conventional serum
variants develop extra-optic nerve manifestations—eg, biomarkers, such as creatinine kinase and lactate.36,37
hearing impairment, CPEO, neuropathy, cerebellar symp­ Studies have shown that coupled measurement of serum
toms, or parkinsonism.29 Additionally, subclinical optic FGF21 and GDF15 was more sensitive in diagnosing
neuropathy can be an accompanying feature in many mitochondrial disease than were muscle histopathological

www.thelancet.com/neurology Vol 20 July 2021 575


Review

Cytosol low amounts of oxidised nicotinamide adenine dinucleo­


Outer membrane tide (NAD+) in patients with mitochondrial myopathy
Fusion and fission
Inner membrane space machinery were restored by the NAD-booster niacin.46 The potential
(eg, MFN2 and OPA1)
for using blood NAD+ concentrations as a mitochondrial
Matrix
disease biomarker requires further study.
mtDNA
Thymine replication
POLγ
TWNK Genomics
m-AAAs
Inner 22 tRNAs Mitochondrial diseases are the result of a defective oxida­
TYMP

Translation
membrane
13 mRNAs tive phosphorylation system (figure 2), which com­prises
dTTP mtDNA
dNTP 2 rRNAs subunits encoded by both the nuclear and mitochondrial
synthesis dCTP
and salvage
genomes. Mutations in genes encoding subunits of
dATP Transcription ADP + Pi H⁺ ATP
(eg, TK2) this system will lead to mitochondrial disease, as will
dGTP
V
defects in the many other nuclear mitochondrial proteins
NADH H2O
required for normal assembly and activity of the oxidative
NAD⁺ O2
FADH2 FAD
phosphorylation system.1 Nuclear mitochondrial proteins
are also essential for the replication, transcription, and
IV

III e
H⁺

I
II e
CoQ10 Cytochrome c maintenance of mtDNA and defects in these proteins can
H⁺ H⁺ lead to mtDNA depletion or multiple mtDNA deletions
(figure 2).
The genetic characteristics of mtDNA differ from those
Figure 2: Illustration of mitochondrial oxidative phosphorylation system and other pathways that are
commonly implicated in adult mitochondrial disease of nuclear DNA: mtDNA is maternally inherited and
The mitochondrial oxidative phosphorylation system comprises complexes I–V and two mobile electron carriers, present in multiple copies in individual cells. Pathogenic
coenzyme Q₁₀ and cytochrome c. As high-energy electrons are passed along the complexes, protons are pumped out variants can, therefore, affect all copies (homoplasmy) or
of the matrix space, creating an electrochemical membrane potential that is used by ATP synthase (complex V) to
only some (heteroplasmy). In the case of heteroplasmy,
generate ATP. The mitochondrial genome encodes 13 protein subunits, 22 tRNAs, and two rRNAs; there are multiple
copies of mtDNA per cell, ranging from hundreds to thousands depending on the cell type. The replication, the percentage of mtDNA copies affected by a pathogenic
maintenance, transcription, and translation of mtDNA and mtDNA-encoded proteins are dependent on many variant is an important factor in determining whether a
nuclear-encoded proteins. m-AAAs play an important role in the quality control of mitochondrial proteins. Genetic clinical phenotype will manifest (appendix pp 10).
defects in nucleotide synthesis and salvage (eg, TYMP and TK2), mitochondrial DNA replication and maintenance
(eg, catalytic subunit of POLγ and TWNK), fusion and fission machinery (eg, OPA1 and MFN2), and m-AAAs
If there is a high clinical suspicion of mitochondrial
(eg, SPG7) perturb mtDNA integrity and mtDNA copy number, leading to the formation of multiple deletions and disease, such as syndromic presentation and multi­
mtDNA depletion. black stars=amino acid. dA=deoxyadenosine. dC=deoxycytidine. dG=deoxyguanosine. system involvement, genetic testing should be prioritised
dT=thymidine. dATP=deoxyadenosine triphosphate. dCTP=deoxycytidine triphosphate. dGTP=deoxyguanosine (figure 3). Deep clinical phenotyping and an accurate
triphosphate. dNTP=deoxynucleoside triphosphate. dTTP=deoxythymidine triphosphate. m-AAAs=mitochondrial
ATP-dependent proteases. mtDNA=mitochondrial DNA. Pi=phosphate.
family history remain essential when considering if
genomic variants identified by techniques such as whole
abnormalities.38 A meta-analysis of data on the use of exome sequencing (WES) or whole genome sequencing
FGF21 and GDF15 for diagnosis (including five studies (WGS) can account for the clinical presentation. Systematic
of FGF21 and seven studies of GDF15) concluded that enquiries about the clinical histories of other family
FGF21 and GDF15 showed acceptable sensitivity (71% members should include asking about both neurological
for FDF21 and 83% for GDF15) and high specificity and extra-neurological signs and symptoms. Parental
(88% for FDF21 and 92% for GDF15).39 These findings consanguinity strongly sug­ gests an autosomal-recessive
highlight a supportive role for these two biomarkers in the disorder, but there are exceptions such as maternally
diagnosis of mito­ chondrial disease and potentially for inherited mtDNA disease in consanguineous families.48
monitoring of dis­eases such as TK2 deficiency.40 FGF21 is Conversely, an absence of family history should not reduce
of reduced value for non-muscle-related phenotypes of clinical suspicion since de-novo single, large-scale mtDNA
mitochon­drial disease.36 deletions, de-novo mtDNA pathogenic variants,49 and
New fluid biomarkers for mitochondrial disease are de-novo autosomal-dominant and recessive traits are all
emerging, including in amino acid and one-carbon known causes of adult mitochondrial disease.50,51 In clinical
metabolism. Circulating cell-free mtDNA was found to be practice, assessment of late-onset, non-syndromic neuro­
increased in m.3243A→G-related mitochondrial disease logical presentations in adults for a mitochondrial cause
and could be useful for monitoring disease severity,41 and remains a considerable challenge, even after extensive
the amount of serum neurofilament light chain seems to investigations to exclude other conditions.
reflect CNS involvement.42 Tissue-specific stress responses
in the affected organ involving one-carbon cycle, trans- mtDNA genomics
sulphuration, and mTORC1 remain to be established as Approximately two-thirds of cases of adult-onset disease are
specific biomarkers for diagnosis.43,44 Multibiomarkers, in caused by pathogenic mtDNA variants.52 The first step in
the form of metabolomic and proteomic approaches, diagnosing adults is typically mtDNA testing, unless there
show promise30,45 but have not yet advanced to clinical are specific indicators suggesting another gene or mode
implementation. A pilot study showed that systemically of inheritance. For the classic syndromic presentations

576 www.thelancet.com/neurology Vol 20 July 2021


Review

such as MELAS syndrome and LHON, screening for required, alternative approaches such as quantitative
common pathogenic mtDNA variants (eg, m.3243A→G in PCR should be used. Pathological mtDNA variants can
MELAS or three primary MT-ND gene variants in LHON), be detected robustly from WGS data and, with lower
will still have a moderate-to-high diagnostic yield (figure 3). sensitivity, from WES data.30,54
Testing sometimes proceeds straight to amplification of a Testing for pathogenic mtDNA variants in blood alone
full-length mtDNA amplicon, or overlapping amplicons, can yield false-negative diagnoses. Additional use of non-
for massively parallel sequencing.30,53 The high depth of invasively obtained samples (eg, urinary sediment, buccal
coverage with such sequencing provides more sensitive swab) can improve the sensitivity of diagnostic mtDNA
detection of single nucleotide mtDNA variants than with testing and familial screening, although the age-corrected
previous methods such as Sanger sequencing, and accurate level of mutant mtDNA hetero­plasmy in blood for the
quantitation of mutant load. Large-scale mtDNA rearrange­ common m.3243A→G variant has a stronger correla­
ments are also detected, but if accurate quantitation is tion with disease burden and progression than in urine.55

A Suspected acute presentations of mitochondrial disease in adults*

Stroke-like episodes or focal-onset status Brainstem syndrometh with or without basal Subacute optic neuropathy (LHON)
epilepticus (MELAS) ganglia changes (Leigh syndrome)

m.3243A>G and POLG† Common LHON variants (blood)‡

negative Full mtDNA sequencing (blood or urine) with or negative


without WES or WGS§

negative

Novel mtDNA variant or variant of undefined Discussion with mitochondrial specialist service
significance

Familial segregation studies (blood and urine)


and muscle biopsy

B Suspected slowly progressive neurological presentations of mitochondrial disease in adults¶||

Ataxia with Myopathy or exercise intolerance†† Neuropathy Progressive


other systemic optic
features** neuropathy

With other With CPEO Without With other Prominent OPA1§§


systemic CPEO‡‡ systemic gastrointes-
features features tinal symptoms

TYMP

negative
Full mtDNA (blood) or POLG gene Single mtDNA negative Muscle Full mtDNA
sequencing deletion biopsy|||| sequencing
negative
(urine)¶¶ (blood)
Mendellian inheritance

negative

Full mtDNA mtDNA


sequencing deletions OR
(blood) mtDNA
depletion
negative negative
negative
negative WES or
WGS***

www.thelancet.com/neurology Vol 20 July 2021 577


Review

If mtDNA screening is uninformative in blood, testing of advisable to expand the gene list for reanalysis of
muscle might be needed to fully exclude mtDNA-related undiagnosed cases. Early WES analyses were designed
disease.56 primarily to detect single nucleotide variants and short
insertions or deletions (indels), and the limitations of
Nuclear DNA genomics these analyses for identifying repeat-expansion disorders
Pathogenic variants in more than 300 nuclear genes such as Huntington’s disease, Friedreich’s ataxia, and
cause mitochondrial disease.50,57 New gene and pathway spinocerebellar ataxias have been highlighted. However,
discoveries and their link to mitochondrial disease will new bioinformatic approaches have been developed to
continue, as more than 1100 mitochondrial proteins have address such challenges.59
been identified to date.58 In many laboratories, unbiased Interpretation of genomic sequencing results has been
WES and WGS data have replaced the testing of single greatly improved by algorithms that prioritise the likeli­
genes. The improved detection of copy number variants hood of rare variants being pathogenic and by widespread
and mtDNA sequences, and the ability to investigate adoption of the American College of Medical Genetics
non-coding regions in the genome, means that the criteria for variant classification.60 Interpretation of data is
diagnostic yield of WGS is usually higher than with WES. usually straightforward when patients are found to have
A key consideration in WES or WGS is selection of the pathogenic or probably pathogenic variants, consistent
gene list to be analysed and whether to use validated and with their phenotype. However, variants of uncertain
curated gene panels (ie, a clinical exome). International impor­tance, without sufficient evidence to determine if
practices differ regarding selection of a narrow phenotype- they are pathogenic or benign, are common. Some of these
defined gene list or a mendeliome containing all genes variants require further functional testing or other studies
causing inherited disease. Standard­isation of gene lists to determine causality. Secondary findings of patho­genic
occurs through the use of curated consensus panels such variants in genes unrelated to the primary medical reason
For PanelApp see https:// as PanelApp. Phenotypic mimicry (eg, Charcot-Marie- for testing are found in up to 4% of patients.61,62 Finally,
panelapp.genomicsengland. Tooth neuro­pathy, genetic myopathy, or spinocerebellar up to 5% of patients with inherited conditions have
co.uk/panels/
ataxia)20 means that if a relatively narrow mitochondrial phenotypes result­ing from pathogenic variants in two or
gene list is used for initial variant analysis, it is usually more differ­ent genes.63 The quality of curation of genomic
variants is higher when ratified by a multi­ disciplinary
team com­ prising clinicians and clinical scientists with
expertise in the conditions being investi­gated. For adults
Figure 3: Algorithms for diagnosis of mitochondrial disease with ambiguous genetic results, or for whom no likely
(A) Diagnostic algorithm for suspected acute presentations of adult mitochondrial genetic diagnosis is achieved, func­tional testing is a follow-
disease. (B) Diagnostic algorithm for suspected slowly progressive neurological
presentations of adult mitochondrial disease. MELAS=mitochondrial myopathy,
up approach, most usually entailing muscle biopsy.
encephalopathy, lactic acidosis, and stroke-like episodes. LHON=Leber hereditary
optic neuropathy. mtDNA=mitochondrial DNA. WES=whole exome sequencing. Biochemical assessment of mitochondrial function
WGS=whole genome sequencing. CPEO=chronic progressive external Traditionally, investigation of suspected mitochondrial
ophthalmoplegia. *Evaluation for multisystem involvement such as sensorineural
hearing loss, diabetes, cardiac abnormality, gastrointestinal, renal, or hepatic
disease focused on detecting evidence of biochemical
involvement. †m.3243A→G and POLG pathogenic variants account for >80% of dysfunc­tion in muscle tissue, including the demonstration
cases of mitochondrial stroke-like episodes. ‡m.3460G→A, m.11778G→A, of COX-deficient fibres and measurements of oxidative
and m.14484T→C pathogenic variants account for >90% of cases of LHON. phosphorylation system enzyme activities. Some biopsy
§Consider other acquired or genetic mimics of mitochondrial disease. ¶Evaluation
for multisystem involvement. Ascertainment of family history. ||If myopathy findings, such as focal COX deficiency, have imperfect
forms part of the overall clinical phenotype, and plasma GDF15 and FGF21 levels sensitivity and specificity for mitochondrial disease since
are within the normal range, the probability of mitochondrial disease is low. defects in COX accumulate with age due to somatic
**If CPEO is also evident, follow the myopathy pathway. Isolated cerebellar mtDNA deletions. Muscle biopsy is typically used after
syndrome is uncommon in primary mitochondrial disease. ††Persistent creatinine
kinase concentration >1000 U/L is rare in mitochondrial disease. ‡‡Neurologists inconclusive genomic testing, and analysis of muscle
face considerable challenges when investigating late-onset, isolated, and tissue64 can provide invaluable information for interpreting
progressive myopathy in adults. Many differential diagnoses are possible, the pathogenicity of novel genetic variants; techniques
including both acquired and genetic causes, and muscle biopsy is important to
such as imaging mass cytometry offer the potential to study
identify the correct diagnosis. MRI of pelvis and lower limbs show a good
diagnostic value for specific genetic muscle diseases; however, no clear pattern of mitochondrial protein signatures at single cell resolution.65
muscle involvement has been identified in primary mitochondrial disease to date. Although many genetic variants can be studied in blood or
§§Account for at least 60% of dominant optic atrophy cases. ¶¶Accounting for non-invasive tissues, muscle-restricted mtDNA variants
30–50% of CPEO cases, the ability to detect a single, large-scale mtDNA deletion in
can only be diagnosed by analysis of muscle.32 Furthermore,
urothelial cells has been reported in patients who previously had muscle biopsy.47
||||Histopathological evidence of mitochondrial dysfunction can be observed in a in single, large-scale mtDNA rearrangements, disease
muscle biopsy, including COX-deficient and other oxidative phosphorylation onset and progression can be predicted by identifying
complex-deficient fibres, or mitochondrial subsarcolemmal accumulation the deletion size and mutant load within muscle tissue.66
(ragged-red fibres). However, these changes, usually occurring in only occasional
Finally, muscle biopsy helps confirm other causes (eg,
fibres, can be observed in non-primary mitochondrial disorders. ***Identification
of variants of undefined significance through WES or WGS might require further inflammatory myositis, inclusion body myositis, and myo­
functional characterisation by muscle biopsy to prove pathogenicity. fibrillar myopathies) and is a source of tissue for RNA

578 www.thelancet.com/neurology Vol 20 July 2021


Review

sequencing, which can lead to a genetic diagnosis in up led to idebenone becoming the first licensed drug in
to 40% of patients who are not diagnosed by WES.67 Europe for treatment of visual impairment in patients
with LHON. An inter­ national expert group reached
Natural history studies and patient registries consensus that idebenone was indicated in the subacute
In past years, sparse availability of longitudinal and natural and dynamic stages of LHON (ie, in the first year after
history data for mitochondrial disease created consider­ onset) but evidence was considered insufficient to approve
able barriers to establishing clear and comprehensive the drug for treatment in the chronic stage. In patients
guidance on medical management, and hampered treated with idebenone, clinically relevant recovery can
develop­­ment of clinical trials and patient-centred clinically occur after 24 months of treatment or, rarely, after an even
relevant outcome measures. Publications describing single longer time. Accord­ingly, idebenone treat­ment should be
cases of mitochondrial disease, or retrospective case series maintained for more than 24 months before judging a
of specific genotypes, often report patients with severe patient a non-responder.92 No evidence is available to
and so-called classic clinical manifestations and reflect, support the use of idebenone in asymptomatic carriers of
therefore, a bias towards severe disease. A greater under­ pathogenic LHON variants.
standing of the spectrum of clinical phenotypes of No randomised controlled trials have been done of
mitochondrial disease and disease severity has been treatments for stroke-like episodes. Use of L-arginine
provided by cohort-based, observational studies that used for this neurological presentation remains controver­
standardised methods of data collection.11,68,69 Substantial sial.2 A longitudinal open-label study in patients with
progress has been made in describing the nature and m.3243A→G-related MELAS syndrome did not attain the
frequency of clinical features of patient cohorts, including predefined primary outcome measure (“the improvement
m.3243A→G,69–71 m.8344A→G,11 MTA-TP6,15,72 single large- rates of headache and nausea or vomiting at 2 h after
scale mtDNA deletion,66,73 POLG,74 TK2,75 TYMP,76 and completion of the initial intravenous administration”),
Leigh syndrome.77 Moreover, establishing these cohorts and L-arginine did not prevent the development of
has facilitated investigation of a phenotype first approach, neurodegeneration.93 A prospective, multicentre, random­
furthering our understanding of the diverse genetic causes ised controlled trial would be required to evaluate the
of specific clinical features of mitochondrial diseases, efficacy of L-arginine to reduce the recurrence of stroke-
such as extrapyramidal movement disorders,78 epilepsy,8,9 like episodes. In an open-label study, taurine supple­
myoclonus,79 CPEO,80,81 pregnancy-related com­pli­cations,82 mentation reduced the frequency of stroke-like episodes
and gastrointestinal symptoms.83 Additionally, our under­ in ten patients with MELAS syndrome.94 However, the
standing of very rare but clinically important events, absence of a control arm in the study questions whether
such as sudden unexpected death in patients with the the change in the number of stroke-like episodes was due
m.3243A→G variant,84 has been augmented by systematic to the efficacy of taurine, a placebo effect, or simply
assessment and follow-up afforded by recruit­ment to these reflected the natural history of stroke-like episodes, which
cohort studies. These natural history studies have provided are paroxysmal and unpredictable. European consensus-
insight into different stages of disease and risk stratifica­ based guidance recommends an urgent administration
tion, and surveillance for complications is now routine of intravenous anti-epileptic drugs for all stroke-like
for common mtDNA-related diseases. Studies of patient episodes.2 Valproate should not be used in patients with
cohorts have also provided insights into new disease POLG-related disease, because this drug might precipitate
mechanisms caused by mitochondrial dysfunction—eg, liver failure.87 However, valproate can be useful for
the leukodystrophy observed on MRI in patients with myoclonic epilepsy in other mitochondrial diseases.
defects in several mt-tRNA synthetases (eg, AARS2, Young adults with pathogenic variants in mtDNA-
DARS2, and EARS2) that lead to impaired translation encoded complex I subunits95 or MTA-TP615 are at risk
of mitochondrial protein85 indicates a role for mito­ of developing subacute brainstem dysfunction. Prompt
chon­dria in myelin metabolism. Furthermore, establish­ recognition is crucial, and critical care support might be
ment of patient cohorts has been essential for development required in some cases.
of consensus-based international guidelines for the care of With few treatments specific for neurological presenta­
patients with mitochondrial disease,2,86–88 development tions of mitochondrial diseases, symptomatic manage­
of clinical phenotype and genomic resources dedicated to ment of chronic neurological symptoms is very important.
mitochondrial disease (including mtDNA variant data in For example, discussing management with an expert
gnomAD and GENOMIT) and selection of outcome oculo­plastic surgeon, who has experience in mitochondrial For gnomAD see https://
measures for clinical trials.89,90 disease, can result in substantial improve­ments in quality gnomad.broadinstitute.org/
of life for patients with CPEO. Exercise is encouraged For GENOMIT see
Treatment because of general health benefits (including cardiovascular https://genomit.eu/

Current treatment and surveillance for systemic fitness) and scant evidence of adverse effects.96,97
complications Some clinical problems, including sensorineural hearing
Findings of a randomised controlled study, and data sub­ loss, cardiac involvement, and gut dysmotility, are encoun­
se­quently derived from an expanded access programme,91,92 tered with multiple genetic defects; effective treatments are

www.thelancet.com/neurology Vol 20 July 2021 579


Review

available at least for sensorineural hearing loss and


cardiac involvement. By contrast, the risk of complications
A
such as diabetes, renal tubulopathy, hep­atic involvement,
Mitochondria Nucleus
Normal
and sideroblastic anaemia seem more genotype-specific
Mutated Primordial (appendix pp 2–4). Therefore, development of personal­
germ cell ised strategies based on knowledge of the natural histories
of different genetic defects is necessary. In view of the
multisystem involve­ment seen in many patients with mito­
chondrial disease, multidisciplinary clinics pro­vid­ing advice
about non-neurological involve­ment (eg, diabetes, bowel
dysfunction, and cardiac disease), as well as managing the
Primary oocyte neurological features, are crucial for optimal care.
No trial evidence supports routine dietary supple­
mentation in adult patients with mitochondrial dis­
ease.1 However, specific mitochondrial diseases—such as
primary coenzyme Q₁₀ deficiency,98 primary disorders
Mature oocyte
of vitamin cofactor metabolism (eg, thiamine, biotin,
and ribo­ flavin), and multiple acyl-CoA dehydrogenase
Fertilisation deficiency—are likely to benefit from supplementation.99
The long-term outcomes of these rare conditions after
appropriate supplementation are currently unknown.

Increase in disease severity Experimental treatments


Several experimental treatments are in development for
Environmental and other genetic factors mitochondrial diseases.100 The main approaches entail
small molecules (including repurposed drugs) to correct
B the biochemical abnormality, metabolic rewiring or bypass,
Fertilised oocyte or gene therapy to target the mitochondrial genome.
from woman with
mtDNA mutations Zygote Some small-molecule approaches have led to clinical trials
for the treatment of specific mitochondrial diseases.100,101
An area of increasing interest includes disorders
associated with abnormalities of mitochondrial nucleoside
metabolism leading to mtDNA depletion or multiple
Pronuclear
Oocyte Pronuclei replacement mtDNA deletions. For example, in mitochondrial neuro­
gastrointestinal encephalomyopathy due to thymidine
Fertilised oocyte phos­phorylase deficiency, therapeutic approaches include
from unaffected
woman
allogeneic stem-cell transplants, orthotopic liver trans­
planta­tion, and enzyme replacement therapy;102 in a
retrospec­tive study of thymidine kinase deficiency, deoxy­
nucleoside therapy led to clinical improvement.103
Other approaches to treatment of mitochondrial dis­
ease include augmentation of mitochondrial biogenesis,
Removal of
pronuclei
restoration of the cellular NAD+ to NADH ratio, increasing
mitophagy, metabolic reprogramming, or manipulation
Figure 4: Mitochondrial heteroplasmy and pronuclear transfer of oxidative stress.100 A pilot study using niacin, a form
(A) The mitochondrial bottleneck explains how there can be extreme divergence of vitamin B3, showed that it restored low NAD+ amounts
in the heteroplasmy between offspring. There is a genetic bottleneck
and improved muscle strength, performance, and metabo­
(the restriction in the number of mitochondrial genomes repopulating the
female germ line) during development that results in different levels of lome in a small group of patients with mitochondrial
heteroplasmy in each individual oocyte. This is a major problem when providing myopathy.46 These encouraging results need confirmation
genetic counselling for mothers with heteroplasmic variants since but suggest that targeting the NAD+ to NADH ratio might
the heteroplasmy level will affect the clinical outcome in the offspring.
be an important factor in some patients with mitochondrial
(B) Mitochondrial donation (mitochondrial replacement therapy) involves the
transfer of the nDNA from an oocyte or zygote from a woman with a pathogenic diseases.
mtDNA variant into an enucleated, recipient donor oocyte or zygote. Pronuclear Genetic manipulation techniques could be useful for
transfer is an in-vitro fertilisation technique that transfers the pronuclei treatment of mitochondrial diseases. Although techniques
(containing the nDNA) immediately after fertilisation from the patient zygote
to the donor zygote. The embryo has the nDNA of the woman with the mtDNA
such as CRISPR–Cas9 are already established for nuclear
variant (and that of the father) but the mtDNA of the donor woman. disorders, correction of defects in the mitochondrial
nDNA=nuclear DNA. mtDNA=mitochondrial DNA. genome is difficult due to the impermeability of mito­

580 www.thelancet.com/neurology Vol 20 July 2021


Review

chondrial membranes. One approach—called allotopic


expression—delivers wild-type copies of mtDNA genes Search strategy and selection criteria
using viral vectors that remain in the cytosol. Transport of We searched PubMed for articles published in English from
the protein into the mitochondria is facilitated by addition Jan 1, 2010, to Nov 1, 2020, using the search terms
of a mitochondrial targeting signal. An example of this “mitochondrial disease OR disorder”, “neurological feature”,
technique is allotopic AAV2–ND4 gene therapy for patients “MELAS”, “CPEO”, “myopathy”, “biomarker”, “genetic”,
with LHON with the m.11778G→A variant.104 Considerable “treatment”, “clinical trial”, AND “reproductive option”.
progress has been made with development of TALENs or We further examined the reference lists from relevant articles.
zinc finger nucleases that cross the mitochondrial mem­ The references were chosen based on their originality and
brane and directly target mutated mtDNA for degradation relevance to the theme of this Review.
in experimental models.105 Another advance is the discovery
of an enzyme that can precisely edit mtDNA, although the
success of edited bases was very low.106 For women with heteroplasmic mtDNA variants, the
situation with transmission is more complex due to a so-
Reproductive options called genetic bottleneck during development.111 Rapid
Although considerable progress has been made in the intergenerational shifts in the mtDNA heteroplasmy are
treatment of mitochondrial diseases, many of these evident in human pedigrees transmitting pathogenic
conditions remain severe and life-threatening, with high mtDNA variants due to the restriction in the number of
morbidity. Advances in genomics mean that most patients mitochondrial genomes repopulating the female germ
with mitochondrial disease now have a genetic diagnosis. line (the mtDNA bottleneck). Since the range of hetero­
Therefore, it is important that couples considering a plasmy is a major risk factor in developing symptoms,
family should be offered reproductive advice, which offspring of a mildly affected women can have either no
should be focused on the specific nuclear or mitochondrial signs or symptoms or very severe disease (figure 4).
genetic defect. For families with mtDNA pathogenic variants, various
For patients with pathogenic nuclear gene variants, reproductive options are available depending on the
reproductive advice will depend on the inheritance specific genetic abnormality and whether it is homoplasmic
pattern and the availability of IVF techniques such as or heteroplasmic.86 For heteroplasmic mtDNA variants,
PGD. Nuclear mitochondrial diseases typically present in options such as prenatal diagnosis or preimplantation
childhood, although several disorders present across the genetic diagnosis might be appropriate if available. How­
age spectrum (eg, POLG and other genes involved in ever, there could be difficult decisions about embryo
mtDNA maintenance). Reproductive options include, selection or continuation of a pregnancy in terms of a safe
where permitted, prenatal testing and preimplantation level of heteroplasmy. For women with high levels of
genetic diagnosis. For some diseases in which the mutant heteroplasmy or a high frequency of homoplasmic vari­
allele is present at high frequency within a population—eg, ants, mitochondrial donation (mitochondrial replacement
in consanguineous societies or geographically isolated therapy)112 is now an option in the UK in a highly regulated
communities—screening for common pathogenic alleles environment crucial for any new in-vitro fertilisation
might be justified. technique (figure 4).113
For patients with mtDNA pathogenic variants, provision
of reproductive advice is more complex than for other Conclusions and future directions
types of mutation. For example, even the most common Major advances in the past 5 years in our under­standing
sporadic mtDNA pathogenic variant—a single, large- of and ability to diagnose and prevent mito­ chondrial
scale mtDNA deletion—is transmitted on rare occasions. disease give rise to cautious optimism for improving
Therefore, reproductive options for women with mtDNA the lives of patients with mitochondrial disease. Robust
pathogenic variants should be discussed at centres that guidelines are in use for the management of patients,114 For more on guidelines see
specialise in mitochondrial genetics.107 Despite some and targeted treatments now exist for some patient https://www.newcastle-
mitochondria.com/wp-content/
reports suggesting that there is biparental transmission of popula­tions. Moreover, developments in in-vitro fertilisa­
cache/all/guidelines/index.html
mtDNA,108 other studies have shown that this is most tion mean new reproductive options are available for
likely not the case.109,110 All reported cases of transmission families with mitochondrial disease.
of mtDNA pathogenic variants have been maternally Nevertheless, this progress highlights the need to focus
inherited. For women carrying homo­ plasmic mtDNA our future research. Elucidation of disease mechanisms is
pathogenic variants, the advice might seem simple: they necessary to understand why specific patients have tissue-
will transmit the variant to their offspring. However, specific manifestations of mitochondrial disease. New or
transmission can be complicated by variable penetrance repurposed drugs are needed, and recognition is required
due to either nuclear genetic or environmental factors. For that some approaches—such a gene therapy—might be
example, in families that carry one of the three common genotype-specific. Development of new treatments will
pathogenic variants for LHON, men are more at risk of require not only cohorts of patients for clinical trials but
developing visual loss than are women. also validated outcome measures that are pertinent to

www.thelancet.com/neurology Vol 20 July 2021 581


Review

mitochondrial diseases. Provision of cascade family tracing Sigrid Jusélius Foundation, Jane and Aatos Erkko foundation, and
will be helpful, since early diagnosis is important for the University of Helsinki.
success of disease-modifying treatments and valuable if References
patients want to consider their reproductive options. 1 Gorman GS, Chinnery PF, DiMauro S, et al. Mitochondrial
diseases. Nat Rev Dis Primers 2016; 2: 16080.
A crucial factor driving research advancement is the 2 Ng YS, Bindoff LA, Gorman GS, et al. Consensus-based statements
close relationship between clinical and basic scientists, for the management of mitochondrial stroke-like episodes.
the pharmaceutical industry who are developing new Wellcome Open Res 2019; 4: 201.
3 Marques-Matos C, Reis J, Reis C, Castro L, Carvalho M.
treatments, and patient organisations. Mitochondrial Mitochondrial encephalomyopathy with lactic acidosis and
For more on mitochondrial disease research should factor in priorities set by patient strokelike episodes presenting before 50 years of age: when a stroke
disease research priorities organisations. is not just a stroke. JAMA Neurol 2016; 73: 604–06.
setting see https:// 4 Hikmat O, Naess K, Engvall M, et al. Simplifying the clinical
www.jla.nihr.ac.uk/priority- Contributors classification of polymerase gamma (POLG) disease based on age of
setting-partnerships/ YSN and DMT proposed the content of manuscript and did the initial onset; studies using a cohort of 155 cases. J Inherit Metab Dis 2020;
mitochondrial-disease/ literature search. All authors contributed equally to writing and 43: 726–36.
subsequent revision of this manuscript and approved the final version. 5 Sinnecker T, Andelova M, Mayr M, et al. Diagnosis of adult-onset
MELAS syndrome in a 63-year-old patient with suspected recurrent
Declaration of interests
strokes: a case report. BMC Neurol 2019; 19: 91.
GSG reports grants from the Wellcome Trust, the UK Medical Research
6 Yokota Y, Hara M, Akimoto T, et al. Late-onset MELAS syndrome
Council (MRC), and Lily Foundation, during the conduct of the study;
with mtDNA 14453G→A mutation masquerading as an acute
and grants from Stealth Therapeutics, Reneo, Julius Clinical (Khondrion), encephalitis: a case report. BMC Neurol 2020; 20: 247.
Neurovive, and AMO Pharma, outside the submitted work. TK reports
7 Ng YS, Gorman GS, Turnbull DM, Martikainen MH.
grants and personal fees from Santhera Pharmaceuticals and Chiesi The diagnosis of posterior reversible encephalopathy syndrome.
GmbH; and grants from Gensight Biologics and Stealth Biotherapeutics, Lancet Neurol 2015; 14: 1073.
outside the submitted work. CK reports personal fees from Santhera, 8 Whittaker RG, Devine HE, Gorman GS, et al. Epilepsy in adults
Sanofi Genzyme, Hormosan, Novartis, and Roche; and personal fees and with mitochondrial disease: a cohort study. Ann Neurol 2015;
other from Stealth Biotherapeutics, outside the submitted work. 78: 949–57.
RM reports grants from Wellcome and the MRC, during the conduct of 9 Ticci C, Sicca F, Ardissone A, et al. Mitochondrial epilepsy:
the study; consultancy fees from Minovia Therapeutics and Reneo to the a cross-sectional nationwide Italian survey. Neurogenetics 2020;
institution; and personal fees from Eisai, outside the submitted work. 21: 87–96.
AS reports grants from the Jane and Aatos Erkko Foundation and Business 10 Anagnostou ME, Ng YS, Taylor RW, McFarland R. Epilepsy due to
Finland; and personal fees from Khondrion, outside the submitted work; mutations in the mitochondrial polymerase gamma (POLG) gene:
AS also has a patent pending for NAD concentration measurement and a clinical and molecular genetic review. Epilepsia 2016; 57: 1531–45.
a patent pending for multibiomarkers for mitochondrial disease. 11 Altmann J, Büchner B, Nadaj-Pakleza A, et al. Expanded phenotypic
RWT reports grants from the Wellcome Trust, the MRC, and the Lily spectrum of the m.8344A→G “MERRF” mutation: data from the
Foundation, during the conduct of the study. DRT reports grants from the German mitoNET registry. J Neurol 2016; 263: 961–72.
Australian National Health and Medical Research Council (NHMRC) and 12 Mancuso M, Orsucci D, Angelini C, et al. Phenotypic heterogeneity
the US Department of Defense, during the conduct of the study. of the 8344A→G mtDNA “MERRF” mutation. Neurology 2013;
DMT reports grants from the Wellcome Trust, the MRC, and the National 80: 2049–54.
Institute for Health Research (NIHR); and personal fees from Khondrion, 13 Lake NJ, Compton AG, Rahman S, Thorburn DR. Leigh syndrome:
Imel, Pretzel therapeutics, and Nanna therapeutics, outside the submitted one disorder, more than 75 monogenic causes. Ann Neurol 2016;
work. All other authors declare no competing interests. 79: 190–203.
14 Sofou K, De Coo IFM, Isohanni P, et al. A multicenter study on
Acknowledgments Leigh syndrome: disease course and predictors of survival.
The work in Newcastle, UK, is supported by the Wellcome Centre for Orphanet J Rare Dis 2014; 9: 52.
Mitochondrial Research (203105/Z/16/Z), Newcastle University Centre for 15 Ng YS, Martikainen MH, Gorman GS, et al. Pathogenic variants in
Ageing and Vitality (supported by the Biotechnology and Biological MT-ATP6: a United Kingdom-based mitochondrial disease cohort
Sciences Research Council and Medical Research Council [G016354/1]), study. Ann Neurol 2019; 86: 310–15.
NIHR Biomedical Research Centre for Ageing and Age-Related Disease 16 Carelli V, d’Adamo P, Valentino ML, et al. Parsing the differences in
award to the Newcastle upon Tyne Hospitals National Health Service affected with LHON: genetic versus environmental triggers of
(NHS) Foundation Trust, the Lily Foundation, and the UK NHS Specialist disease conversion. Brain 2016; 139: e18.
Commissioners, which funds the Rare Mitochondrial Disorders of 17 Yu-Wai-Man P, Chinnery PF. Leber hereditary optic neuropathy.
Adults and Children Diagnostic Service in Newcastle upon Tyne. In: Adam MP, Ardinger HH, Pagon RA, et al, eds. GeneReviews.
TK acknowledges support by the German Federal Ministry of Education Seattle, WA: University of Washington, 2000: 1993–2021.
and Research (Bonn, Germany) through grants to the German Network 18 Borrelli E, Triolo G, Cascavilla ML, et al. Changes in choroidal
for Mitochondrial Disorders (01GM1906A) and to the E-Rare project thickness follow the RNFL changes in Leber’s hereditary optic
GENOMIT (01GM1920B). TK is a member of the European Reference neuropathy. Sci Rep 2016; 6: 37332.
Network for Rare Neurological Diseases and of the European Reference 19 Richardson C, Smith T, Schaefer A, et al. Ocular motility findings in
Network for neuromuscular diseases (EURO-NMD), co-funded by the chronic progressive external ophthalmoplegia. Eye 2005; 19: 258–63.
European Commission. YSN holds an NIHR Clinical Lectureship in 20 Braz LP, Ng YS, Gorman GS, et al. Neuromuscular junction
Neurology (CL-2016-01-003). CK is a member of EURO-NMD, co-funded abnormalities in mitochondrial disease: an observational cohort
by the European Commission. MM is partly supported by Telethon Grant study. Neurology Clin Prac 2021: 11: 97–104.
GUP09004 and Telethon-MITOCON grant GSP16001, RF-2016-02361495 21 Parikh S, Karaa A, Goldstein A, et al. Diagnosis of ‘possible’
and GENOMIT. CMS is a NHMRC practitioner fellow (APP 1136800) and mitochondrial disease: an existential crisis. J Med Genet 2019;
56: 123–30.
is supported by research grants from NHMRC, the Mito Foundation,
Aligning Science Across Parkinson’s Collaborative Research Network, and 22 Pareyson D, Piscosquito G, Moroni I, Salsano E, Zeviani M.
Peripheral neuropathy in mitochondrial disorders. Lancet Neurol
the Spastic Paraplegia Foundation. DRT is supported by a fellowship and
2013; 12: 1011–24.
grants from NHMRC and by research grants from the US Department of
23 de Laat P, Zweers HE, Knuijt S, Smeitink JA, Wanten GJ,
Defense Congressionally Directed Medical Research Programs, the Mito
Janssen MC. Dysphagia, malnutrition and gastrointestinal
Foundation, and the Victorian Government’s Operational Infrastructure problems in patients with mitochondrial disease caused by the
Support Program. AS has been funded by Academy of Finland, m3243A>G mutation. Neth J Med 2015; 73: 30–36.

582 www.thelancet.com/neurology Vol 20 July 2021


Review

24 Emmanuele V, López LC, Berardo A, et al. Heterogeneity of 47 Varhaug KN, Nido GS, de Coo I, et al. Using urine to diagnose
coenzyme Q10 deficiency: patient study and literature review. large-scale mtDNA deletions in adult patients.
Arch Neurol 2012; 69: 978–83. Ann Clin Transl Neurol 2020; 7: 1318–26.
25 Domínguez-González C, Hernández-Laín A, Rivas E, et al. 48 Alston CL, He L, Morris AA, et al. Maternally inherited
Late-onset thymidine kinase 2 deficiency: a review of 18 cases. mitochondrial DNA disease in consanguineous families.
Orphanet J Rare Dis 2019; 14: 100. Eur J Hum Genet 2011; 19: 1226–29.
26 Kullar PJ, Quail J, Lindsey P, et al. Both mitochondrial DNA and 49 Sallevelt SC, de Die-Smulders CE, Hendrickx AT, et al. De novo
mitonuclear gene mutations cause hearing loss through cochlear mtDNA point mutations are common and have a low recurrence
dysfunction. Brain 2016; 139: e33. risk. J Med Genet 2016; 54: 73–83.
27 Santarelli R, Rossi R, Scimemi P, et al. OPA1-related auditory 50 Frazier AE, Thorburn DR, Compton AG. Mitochondrial energy
neuropathy: site of lesion and outcome of cochlear implantation. generation disorders: genes, mechanisms, and clues to pathology.
Brain 2015; 138: 563–76. J Biol Chem 2019; 294: 5386–95.
28 Yu-Wai-Man P, Chinnery PF. Dominant optic atrophy: novel 51 Tan J, Wagner M, Stenton SL, et al. Lifetime risk of autosomal
OPA1 mutations and revised prevalence estimates. Ophthalmology recessive mitochondrial disorders calculated from genetic
2013; 120: 1712. databases. EBioMedicine 2020; 54: 102730.
29 Carelli V, Musumeci O, Caporali L, et al. Syndromic parkinsonism 52 Gorman GS, Schaefer AM, Ng Y, et al. Prevalence of nuclear and
and dementia associated with OPA1 missense mutations. mitochondrial DNA mutations related to adult mitochondrial
Ann Neurol 2015; 78: 21–38. disease. Ann Neurol 2015; 77: 753–59.
30 Stenton SL, Prokisch H. Genetics of mitochondrial diseases: 53 Palculict ME, Zhang VW, Wong LJ, Wang J. Comprehensive
identifying mutations to help diagnosis. EBioMedicine 2020; mitochondrial genome analysis by massively parallel sequencing.
56: 102784. Methods Mol Biol 2016; 1351: 3–17.
31 Watson E, Davis R, Sue CM. New diagnostic pathways for 54 Wagner M, Berutti R, Lorenz-Depiereux B, et al. Mitochondrial
mitochondrial disease. J Transl Gen Genom 2020; 4: 188–202. DNA mutation analysis from exome sequencing-A more holistic
32 Hardy SA, Blakely EL, Purvis AI, et al. Pathogenic mtDNA approach in diagnostics of suspected mitochondrial disease.
mutations causing mitochondrial myopathy: the need for muscle J Inherit Metab Dis 2019; 42: 909–17.
biopsy. Neurol Genet 2016; 2: e82. 55 Grady JP, Pickett SJ, Ng YS, et al. mtDNA heteroplasmy level and
33 Tyynismaa H, Carroll CJ, Raimundo N, et al. Mitochondrial copy number indicate disease burden in m.3243A→G
myopathy induces a starvation-like response. Hum Mol Genet 2010; mitochondrial disease. EMBO Mol Med 2018; 10: e8262.
19: 3948–58. 56 Ng YS, Thompson K, Loher D, et al. Novel MT-ND gene variants
34 Kalko SG, Paco S, Jou C, et al. Transcriptomic profiling of TK2 causing adult-onset mitochondrial disease and isolated complex I
deficient human skeletal muscle suggests a role for the p53 deficiency. Front Genet 2020; 11: 24.
signalling pathway and identifies growth and differentiation 57 Thompson K, Collier JJ, Glasgow RIC, et al. Recent advances in
factor-15 as a potential novel biomarker for mitochondrial understanding the molecular genetic basis of mitochondrial
myopathies. BMC Genomics 2014; 15: 91. disease. J Inherit Metab Dis 2020; 43: 36–50.
35 Formichi P, Cardone N, Taglia I, et al. Fibroblast growth factor 21 58 Rath S, Sharma R, Gupta R, et al. MitoCarta3.0: an updated
and grow differentiation factor 15 are sensitive biomarkers of mitochondrial proteome now with sub-organelle localization and
mitochondrial diseases due to mitochondrial transfer-RNA pathway annotations. Nucleic Acids Res 2021; 49: D1541–47.
mutations and mitochondrial DNA deletions. Neurol Sci2020; 59 Tankard RM, Bennett MF, Degorski P, Delatycki MB, Lockhart PJ,
41: 3653–62. Bahlo M. Detecting expansions of tandem repeats in cohorts
36 Suomalainen A, Elo JM, Pietiläinen KH, et al. FGF-21 as a sequenced with short-read sequencing data. Am J Hum Genet 2018;
biomarker for muscle-manifesting mitochondrial respiratory chain 103: 858–73.
deficiencies: a diagnostic study. Lancet Neurol 2011; 10: 806–18. 60 Richards S, Aziz N, Bale S, et al. Standards and guidelines for the
37 Yatsuga S, Fujita Y, Ishii A, et al. Growth differentiation factor 15 as interpretation of sequence variants: a joint consensus
a useful biomarker for mitochondrial disorders. Ann Neurol 2015; recommendation of the American College of Medical Genetics and
78: 814–23. Genomics and the Association for Molecular Pathology. Genet Med
38 Lehtonen JM, Auranen M, Darin N, et al. Diagnostic value of serum 2015; 17: 405–24.
biomarkers FGF21 and GDF15 compared to muscle sample in 61 Thauvin-Robinet C, Thevenon J, Nambot S, et al. Secondary
mitochondrial disease. J Inherit Metab Dis 2021; 44: 469–80. actionable findings identified by exome sequencing: expected
39 Lin Y, Ji K, Ma X, et al. Accuracy of FGF-21 and GDF-15 for the impact on the organisation of care from the study of
diagnosis of mitochondrial disorders: a meta-analysis. 700 consecutive tests. Eur J Hum Genet 2019; 27: 1197–214.
Ann Clin Transl Neurol 2020; 7: 1204–13. 62 Kalia S, Adelman K, Bale S, et al. Recommendations for reporting of
40 Dominguez-Gonzalez C, Badosa C, Madruga-Garrido M, et al. secondary findings in clinical exome and genome sequencing, 2016
Growth differentiation factor 15 is a potential biomarker of update (ACMG SF v2.0): a policy statement of the American College
therapeutic response for TK2 deficient myopathy. Sci Rep 2020; of Medical Genetics and Genomics. Genet Med 2017; 19: 249–55.
10: 10111. 63 Posey JE, Harel T, Liu P, et al. Resolution of disease phenotypes
41 Maresca A, Del Dotto V, Romagnoli M, et al. Expanding and resulting from multilocus genomic variation. N Engl J Med 2017;
validating the biomarkers for mitochondrial diseases. 376: 21–31.
J Mol Med (Berl) 2020; 98: 1467–78. 64 Frazier AE, Vincent AE, Turnbull DM, Thorburn DR, Taylor RW.
42 Varhaug KN, Hikmat O, Nakkestad HL, Vedeler CA, Bindoff LA. Assessment of mitochondrial respiratory chain enzymes in cells
Serum biomarkers in primary mitochondrial disorders. and tissues. Methods Cell Biol 2020; 155: 121–56.
Brain Commun 2021; 3: fcaa222. 65 Warren C, McDonald D, Capaldi R, et al. Decoding mitochondrial
43 Forsström S, Jackson CB, Carroll CJ, et al. Fibroblast growth factor heterogeneity in single muscle fibres by imaging mass cytometry.
21 drives dynamics of local and systemic stress responses in Sci Rep 2020; 10: 15336.
mitochondrial myopathy with mtDNA deletions. Cell Metab 2019; 66 Grady JP, Campbell G, Ratnaike T, et al. Disease progression in
30: 1040–54. patients with single, large-scale mitochondrial DNA deletions.
44 Nikkanen J, Forsström S, Euro L, et al. Mitochondrial DNA Brain 2014; 137: 323–34.
replication defects disturb cellular dNTP pools and remodel 67 Cummings BB, Marshall JL, Tukiainen T, et al. Improving genetic
one-carbon metabolism. Cell Metab 2016; 23: 635–48. diagnosis in Mendelian disease with transcriptome sequencing.
45 Buzkova J, Nikkanen J, Ahola S, et al. Metabolomes of Sci Transl Med 2017; 9: eaal5209. 66
mitochondrial diseases and inclusion body myositis patients: 68 Nesbitt V, Pitceathly RDS, Turnbull DM, et al. The UK MRC
treatment targets and biomarkers. EMBO Mol Med 2018; 10: e9091. Mitochondrial Disease Patient Cohort Study: clinical phenotypes
46 Pirinen E, Auranen M, Khan NA, et al. Niacin cures systemic NAD+ associated with the m.3243A→G mutation—implications for
deficiency and improves muscle performance in adult-onset diagnosis and management. J Neurol Neurosurg Psychiatry 2013;
mitochondrial myopathy. Cell Metab 2020; 31: 1078–90. 84: 936–38.

www.thelancet.com/neurology Vol 20 July 2021 583


Review

69 Mancuso M, Orsucci D, Angelini C, et al. The m.3243A→G 91 Klopstock T, Yu-Wai-Man P, Dimitriadis K, et al. A randomized
mitochondrial DNA mutation and related phenotypes: a matter of placebo-controlled trial of idebenone in Leber’s hereditary optic
gender? J Neurol 2014; 261: 504–10. neuropathy. Brain 2011; 134: 2677–86.
70 Pickett SJ, Grady JP, Ng YS, et al. Phenotypic heterogeneity in 92 Catarino CB, von Livonius B, Priglinger C, et al. Real-world clinical
m.3243A→G mitochondrial disease: the role of nuclear factors. experience with idebenone in the treatment of Leber hereditary
Ann Clin Transl Neurol 2018; 5: 333–45. optic neuropathy. J Neuroophthalmol 2020; 40: 55865.
71 de Laat P, Rodenburg RR, Roeleveld N, Koene S, Smeitink JA, 93 Koga Y, Povalko N, Inoue E, et al. Therapeutic regimen of
Janssen MC. Six-year prospective follow-up study in 151 carriers of L-arginine for MELAS: 9-year, prospective, multicenter, clinical
the mitochondrial DNA 3243 A>G variant. J Med Genet 2020; research. J Neurol 2018; 265: 2861–74.
58: 48–55. 94 Ohsawa Y, Hagiwara H, Nishimatsu SI, et al. Taurine
72 Stendel C, Neuhofer C, Floride E, et al. Delineating MT-ATP6- supplementation for prevention of stroke-like episodes in MELAS:
associated disease: from isolated neuropathy to early onset a multicentre, open-label, 52-week phase III trial.
neurodegeneration. Neurol Genet 2020; 6: e393. J Neurol Neurosurg Psychiatry 2019; 90: 529–36.
73 Mancuso M, Orsucci D, Angelini C, et al. Redefining phenotypes 95 Ng YS, Lax NZ, Maddison P, et al. MT-ND5 mutation exhibits
associated with mitochondrial DNA single deletion. J Neurol 2015; highly variable neurological manifestations at low mutant load.
262: 1301–09. EBioMedicine 2018; 30: 86–93.
74 Hikmat O, Naess K, Engvall M, et al. Simplifying the clinical 96 Stefanetti RJ, Blain A, Jimenez-Moreno C, et al. Measuring the
classification of polymerase gamma (POLG) disease based on age of effects of exercise in neuromuscular disorders: a systematic review
onset; studies using a cohort of 155 cases. J Inherit Metab Dis 2020; and meta-analyses. Wellcome Open Res 2020; 5: 84.
43: 726–36. 97 Jeppesen TD. Aerobic exercise training in patients with
75 Garone C, Taylor RW, Nascimento A, et al. Retrospective natural mtDNA-related mitochondrial myopathy. Front Physiol 2020; 11: 349.
history of thymidine kinase 2 deficiency. J Med Genet 2018; 98 Traschütz A, Schirinzi T, Laugwitz L, et al. Clinico-genetic, imaging
55: 515–21. and molecular delineation of COQ8A-ataxia: a multicenter study of
76 Corazza G, Pagan C, Hardy G, et al. MyoNeuroGastroIntestinal 59 patients. Ann Neurol 2020; 88: 251–63.
encephalopathy: natural history and means for early diagnosis. 99 Distelmaier F, Haack TB, Wortmann SB, Mayr JA, Prokisch H.
Gastroenterology 2019; 156: 1525–27. Treatable mitochondrial diseases: cofactor metabolism and beyond.
77 Sofou K, de Coo IFM, Ostergaard E, et al. Phenotype-genotype Brain 2017; 140: e11.
correlations in Leigh syndrome: new insights from a multicentre 100 Russell OM, Gorman GS, Lightowlers RN, Turnbull DM.
study of 96 patients. J Med Genet 2018; 55: 21–27. Mitochondrial diseases: hope for the future. Cell 2020; 181: 168–88.
78 Martikainen MH, Ng YS, Gorman GS, et al. Clinical, genetic, and 101 Viscomi C, Zeviani M. Strategies for fighting mitochondrial
radiological features of extrapyramidal movement disorders in diseases. J Intern Med 2020; 287: 665–84.
mitochondrial disease. JAMA Neurol 2016; 73: 668–74. 102 Bax BE. Mitochondrial neurogastrointestinal encephalomyopathy:
79 Mancuso M, Orsucci D, Angelini C, et al. Myoclonus in approaches to diagnosis and treatment. J Transl Genet Genom 2020;
mitochondrial disorders. Mov Disord 2014; 29: 722–28. 4: 1–16.
80 Heighton JN, Brady LI, Sadikovic B, Bulman DE, Tarnopolsky MA. 103 Domínguez-González C, Madruga-Garrido M, Mavillard F, et al.
Genotypes of chronic progressive external ophthalmoplegia in a Deoxynucleoside therapy for thymidine kinase 2-deficient
large adult-onset cohort. Mitochondrion 2019; 49: 227–31. myopathy. Ann Neurol 2019; 86: 293–303.
81 Orsucci D, Angelini C, Bertini E, et al. Revisiting mitochondrial 104 Yu-Wai-Man P, Newman NJ, Carelli V, et al. Bilateral visual
ocular myopathies: a study from the Italian Network. J Neurol 2017; improvement with unilateral gene therapy injection for Leber
264: 1777–84. hereditary optic neuropathy. Sci Transl Med 2020; 12: eaaz7423.
82 Feeney CL, Lim AZ, Fagan E, et al. A case-comparison study of 105 Jackson CB, Turnbull DM, Minczuk M, Gammage PA. Therapeutic
pregnant women with mitochondrial disease: what to expect? BJOG manipulation of mtDNA heteroplasmy: a shifting perspective.
2019; 126: 1380–89. Trends Mol Med 2020; 26: 698–709.
83 Ng YS, Feeney C, Schaefer AM, et al. Pseudo-obstruction, stroke, 106 Mok BY, de Moraes MH, Zeng J, et al. A bacterial cytidine
and mitochondrial dysfunction: a lethal combination. Ann Neurol deaminase toxin enables CRISPR-free mitochondrial base editing.
2016; 80: 686–92. Nature 2020; 583: 631–37.
84 Ng YS, Grady JP, Lax NZ, et al. Sudden adult death syndrome in 107 Gorman GS, McFarland R, Stewart J, Feeney C, Turnbull DM.
m.3243A→G-related mitochondrial disease: an unrecognized Mitochondrial donation: from test tube to clinic. Lancet 2018;
clinical entity in young, asymptomatic adults. Eur Heart J 2016; 392: 1191–92.
37: 2552–59. 108 Luo S, Valencia CA, Zhang J, et al. Biparental inheritance of
85 Sissler M, González-Serrano LE, Westhof E. Recent advances in mitochondrial DNA in humans. Proc Natl Acad Sci USA 2018;
mitochondrial aminoacyl-tRNA synthetases and disease. 115: 13039–44.
Trends Mol Med 2017; 23: 693–708. 109 Wei W, Pagnamenta AT, Gleadall N, et al. Nuclear-mitochondrial
86 Poulton J, Steffann J, Burgstaller J, McFarland R. 243rd ENMC DNA segments resemble paternally inherited mitochondrial DNA
international workshop: developing guidelines for management of in humans. Nat Commun 2020; 11: 1740.
reproductive options for families with maternally inherited mtDNA 110 Rius R, Cowley MJ, Riley L, Puttick C, Thorburn DR, Christodoulou J.
disease, Amsterdam, the Netherlands, 22–24 March 2019. Biparental inheritance of mitochondrial DNA in humans is not a
Neuromuscul Disord 2019; 29: 725–33. common phenomenon. Genet Med 2019; 21: 2823–26.
87 De Vries MC, Brown DA, Allen ME, et al. Safety of drug use in 111 Wilson IJ, Carling PJ, Alston CL, et al. Mitochondrial DNA
patients with a primary mitochondrial disease: an international sequence characteristics modulate the size of the genetic bottleneck.
Delphi-based consensus. J Inherit Metab Dis 2020; 43: 800–18. Hum Mol Genet 2016; 25: 1031–41.
88 Hirano M, Carelli V, De Giorgio R, et al. Mitochondrial 112 Greenfield A, Braude P, Flinter F, Lovell-Badge R, Ogilvie C,
neurogastrointestinal encephalomyopathy (MNGIE): position paper Perry ACF. Assisted reproductive technologies to prevent human
on diagnosis, prognosis, and treatment by the MNGIE International mitochondrial disease transmission. Nat Biotechnol 2017;
Network. J Inherit Metab Dis 2021; 44: 376–87. 35: 1059–68.
89 Shen L, Diroma MA, Gonzalez M, et al. MSeqDR: a centralized 113 National Academy of Medicine, National Academy of Sciences,
knowledge repository and bioinformatics web resource to facilitate Royal Society. Heritable human genome editing. Washington, DC:
genomic investigations in mitochondrial disease. Hum Mutat 2016; The National Academies Press, 2020.
37: 540–48.
114 Parikh S, Goldstein A, Karaa A, et al. Patient care standards for
90 Mancuso M, McFarland R, Klopstock T, Hirano M. International primary mitochondrial disease: a consensus statement from the
workshop: outcome measures and clinical trial readiness in primary Mitochondrial Medicine Society. Genet Med 2017; 19: 1380.
mitochondrial myopathies in children and adults—consensus
recommendations, 16–18 November 2016, Rome, Italy.
Neuromuscul Disord 2017; 27: 1126–37. © 2021 Elsevier Ltd. All rights reserved.

584 www.thelancet.com/neurology Vol 20 July 2021

You might also like