Nuclear Gene-Encoded Leigh Syndrome Spectrum Overview

Rahman S, Thorburn D.

Publication Details

Estimated reading time: 33 minutes

Summary

The purpose of this overview is to increase the awareness of clinicians regarding genetic causes of nuclear gene-encoded Leigh syndrome spectrum (LSS), management, especially treatable disorders, and relevant genetic counseling issues. The following are the goals of this overview.

Goal 1.

Describe the clinical characteristics of nuclear gene-encoded LSS.

Goal 2.

Review the genetic causes of nuclear gene-encoded LSS.

Goal 3.

Provide an evaluation strategy to identify the genetic cause of nuclear gene-encoded LSS in a proband (when possible).

Goal 4.

Review management of nuclear gene-encoded LSS, especially treatable disorders.

Goal 5.

Inform genetic counseling of family members of an individual with nuclear gene-encoded LSS.

1. Clinical Characteristics of Nuclear Gene-Encoded Leigh Syndrome Spectrum

Clinical manifestations of nuclear gene-encoded Leigh syndrome spectrum. The term Leigh syndrome spectrum comprises both Leigh syndrome and Leigh-like syndrome.

Leigh syndrome (or subacute necrotizing encephalomyelopathy) is characterized by decompensation (often with elevated lactate levels in blood and/or cerebrospinal fluid [CSF]) during an intercurrent illness. It is typically associated with psychomotor retardation or regression, often followed by transient or prolonged stabilization or even improvement, but inevitably resulting in eventual progressive neurologic decline, typically occurring in stepwise decrements.

Neurologic manifestations include hypotonia, spasticity, movement disorders (including chorea), cerebellar ataxia, and peripheral neuropathy.

Extraneurologic manifestations may include hypertrophic cardiomyopathy, hypertrichosis, anemia, renal tubulopathy, liver involvement, ptosis, and muscle weakness.

Onset is typically between ages three and 12 months, frequently following a viral infection. About 50% of affected individuals die by age three years, most often as a result of respiratory or cardiac failure.

Later onset (including in adulthood) and long-term survival may occasionally occur.

Life expectancy and extraneurologic manifestations appear to be related, at least in part, to the underlying genetic defect [Wedatilake et al 2013, Rahman et al 2017].

"Leigh-like syndrome" is often used when clinical and other features strongly suggest Leigh syndrome but do not fulfil the stringent diagnostic criteria because of atypical or normal neuroimaging, lack of evidence of abnormal energy metabolism, atypical neuropathology (variation in the distribution or character of lesions or with the additional presence of unusual features such as extensive cortical destruction), and/or incomplete evaluation. The term Leigh syndrome spectrum comprises both Leigh syndrome and Leigh-like syndrome.

Neuropathologic features of Leigh syndrome. Leigh syndrome was originally defined in 1951 by characteristic neuropathologic features including multiple focal symmetric necrotic lesions in the basal ganglia, thalamus, brain stem, dentate nuclei, and optic nerves. Histologically, lesions have a spongiform appearance and are characterized by demyelination, gliosis, and vascular proliferation. Although neuronal loss can occur, typically the neurons are relatively spared. The advent of magnetic resonance imaging (MRI) has made it possible to establish the diagnosis by neuroimaging, and thus postmortem examination is now rarely performed outside of a research context.

Establishing the diagnosis of nuclear gene-encoded Leigh syndrome spectrum requires the following [Rahman et al 1996, Lake et al 2016]:

  • Characteristic clinical presentation
  • Bilateral symmetric T2-weighted hyperintensities in the basal ganglia and/or brain stem on brain MRI
  • Evidence of abnormal energy metabolism based on one or more of the following:
    • Elevated lactate in blood and/or CSF
    • Other evidence of disturbed oxidative phosphorylation or pyruvate dehydrogenase activity
  • Identification of pathogenic variant(s) in a specific nuclear gene (See Section 2.)

Differential diagnosis of nuclear gene-encoded Leigh syndrome spectrum includes mitochondrial DNA-associated Leigh syndrome (see Mitochondrial DNA-Associated Leigh Syndrome and NARP and Mitochondrial Disorders Overview), nonmitochondrial genetic causes of bilateral striatal necrosis (e.g., heterozygous pathogenic variant in ADAR, biallelic pathogenic variants in NUP62, or heterozygous pathogenic variant in RANBP2), and acquired nongenetic causes such as viral encephalopathy.

2. Causes of Nuclear Gene-Encoded Leigh Syndrome Spectrum

Pathogenic variants in more than 80 nuclear genes have been associated with autosomal recessive, autosomal dominant, and X-linked nuclear gene-encoded Leigh syndrome spectrum (LSS), as summarized below [Rahman et al 2017].

Autosomal Recessive Leigh Syndrome Spectrum

Autosomal recessive nuclear gene-encoded LSS (Table 1) includes:

  • Pathogenic variants in genes encoding proteins needed for:
  • Organic acidemias with accumulation of metabolites leading to secondary OXPHOS dysfunction.
Table 1.

Table 1.

Autosomal Recessive Leigh Syndrome Spectrum

Autosomal Dominant Leigh Syndrome Spectrum

DNM1L. To date, DNM1L is the only gene known to be associated with autosomal dominant LSS [Zaha et al 2016]; heterozygous de novo variants in this gene account for <1% of LSS. DNM1L-LSS is associated with infantile spasms with burst suppression on EEG; laboratory findings include multiple respiratory chain enzyme deficiencies and elongated mitochondria and peroxisomes on electron microscopy. Autosomal recessive DNM1L-related disease has been reported but – to date – not in association with LSS.

X-Linked Leigh Syndrome Spectrum

Causes of X-linked nuclear gene-encoded LSS are summarized in Table 2.

Table 2.

Table 2.

X-Linked Leigh Syndrome Spectrum

3. Evaluation Strategies to Identify the Genetic Cause of Nuclear Gene-Encoded Leigh Syndrome in a Proband

Once Leigh syndrome spectrum (LSS) is considered in an individual, determining the specific cause (see Table 1 and Table 2) aids in discussions of prognosis and treatment (see Management) and in genetic counseling.

The following information can be used to establish the specific cause of nuclear gene-encoded LSS for a given individual: clinical findings, family history, specialized testing, and molecular genetic/genomic testing.

Clinical Findings

Clinical manifestations of LSS are described in Tables 1 and 2.

Retrospective review of the currently known genetic causes of nuclear gene-encoded Leigh syndrome (see Tables 1 and 2) suggests some differences in phenotype, but clinical findings in individuals with variants in different genes typically overlap [Rahman et al 2017]. Hence, it would now be unusual for specific clinical and/or imaging findings to guide testing of a subset of genes, but these differences may be useful in guiding variant curation. For example:

  • More than 40 individuals with pathogenic variants in SURF1 or LRPPRC have been reported [Debray et al 2011, Wedatilake et al 2013] (see Table 1). Mean survival is longer in those with SURF1 deficiency (5.4 years) than in those with LRPPRC deficiency (1.8 years), apparently due to the occurrence of more frequent and severe metabolic crises in the latter. SURF1 deficiency also appears to have a high incidence of hypertrichosis and peripheral neuropathy [Wedatilake et al 2013].
  • Brain malformations are typically seen in males with a hemizygous variant in PDHA1 and some females with a heterozygous variant [Patel et al 2012] (see Table 2). Specific brain tracts may be involved in some subgroups of complex I deficiency; for example, brain stem lesions are seen within the mamillothalamic tracts, substantia nigra, medial lemniscus, medial longitudinal fasciculus, and spinothalamic tracts on T2-weighted MRI in individuals with mutation of NDUFAF2 [Fassone & Rahman 2012].

Family History

A three-generation family history should be obtained with attention to other relatives with neurologic manifestations, or other clinical features compatible with a mitochondrial disorder. Documentation of relevant findings in relatives can be accomplished either through direct examination of those individuals or by review of their medical records including the results of molecular genetic testing, neuroimaging studies, and autopsy examinations.

Specific findings such as a family history in which affected individuals are related to each other through females (i.e., no male-to-male transmission) may prompt initial investigation of X-linked genes, or consanguinity may prompt initial investigation of autosomal recessive genes. Of course, such features are sometimes a chance occurrence and the possibility of an underlying mitochondrial DNA (mtDNA) variant should be followed up with more comprehensive testing if no pathogenic variants are identified in nuclear genes [Alston et al 2011].

Biochemical Testing

Elevated lactate levels in blood and/or cerebrospinal fluid can:

  • Suggest LSS as opposed to other disorders with similar clinical findings;
  • Implicate mutation of one of the genes causing PDH deficiency when the ratio of lactate to pyruvate is normal to low [Debray et al 2007].

Measurement of enzyme activity. Activity of enzymes, such as PDH, are typically measured in cultured skin fibroblasts (fbs; see Tables 1 and 2), and respiratory chain enzymes are typically measured in a skeletal muscle biopsy (mb; see Tables 1 and 2).

  • Although identifying an enzyme defect can help prioritize molecular genetic testing, this approach can still leave a large number of genes to be tested (e.g., respiratory chain complex I-deficient Leigh syndrome has to date been shown to be caused by pathogenic variants in at least 15 autosomal genes (see Table 1), one X-linked gene (see Table 2), and six genes encoded by mtDNA.
  • Moreover, although muscle biopsy was traditionally used as a first-line diagnostic test in the investigation of Leigh syndrome and other mitochondrial disorders, the widespread availability of multigene panels and comprehensive genomic testing has obviated the need for muscle biopsy in many instances. However, in a small minority of individuals enzymatic testing of a muscle or skin biopsy may be necessary to confirm pathogenicity of variants of uncertain significance identified by multigene panel, exome or genome sequencing.

Molecular Genetic Testing

Testing approaches can include use of a multigene panel, or more comprehensive genomic testing of an exome or genome.

A mitochondrial disorders multigene panel that includes some or all of the genes listed in Tables 1 and 2 is most likely to identify the genetic cause of the condition while limiting identification of variants of uncertain significance and pathogenic variants in genes that do not explain the underlying phenotype. Note: (1) The genes included in the panel and the diagnostic sensitivity of the testing used for each gene vary by laboratory and are likely to change over time. (2) Some multigene panels may include genes not associated with the condition discussed in this GeneReview. Of note, given the rarity of some of the genes associated with nuclear gene-encoded LSS, some panels may not include all the genes mentioned in this overview. (3) In some laboratories, panel options may include a custom laboratory-designed panel and/or custom phenotype-focused exome analysis that includes genes specified by the clinician. (4) Methods used in a panel may include sequence analysis, deletion/duplication analysis, and/or other non-sequencing-based tests. (5) While the vast majority of individuals with classic Leigh syndrome have nuclear or mtDNA defects related to mitochondrial energy generation, this may not be true for individuals with Leigh-like syndrome; thus, use of a multigene panel with a more extensive gene list may be considered.

For an introduction to multigene panels click here. More detailed information for clinicians ordering genetic tests can be found here.

Comprehensive genomic testing does not require the clinician to determine which gene(s) are likely involved. Exome sequencing is most commonly used; genome sequencing is also possible.

If exome or genome sequencing is not diagnostic, analysis for copy number variants, typically by algorithmic analysis for read depth and other parameters, (often called exome array) may be considered to detect (multi)exon deletions or duplications that cannot be detected by sequence analysis.

For an introduction to comprehensive genomic testing click here. More detailed information for clinicians ordering genomic testing can be found here.

Testing for Treatable Disorders

Treatable disorders should be rapidly tested for biochemically or genetically as indicated; or if this is not possible, trials of the relevant vitamins/cofactors should be instituted as soon as the diagnosis is considered. Ideally, therapy should continue until these disorders have been excluded by biochemical and/or genetic testing, and continued for life if the diagnosis is confirmed.

Treatable causes of nuclear gene-encoded LSS (see Treatment of Manifestations):

4. Management of Nuclear Gene-Encoded Leigh Syndrome

Treatment of Manifestations

Specific treatment is possible for the following three nuclear gene-encoded Leigh-like syndromes:

  • Biotin-thiamine-responsive basal ganglia disease (also known as thiamine transporter-2 deficiency) (mutation of SLC19A3). Biotin (5-10 mg/kg/day) and thiamine (in doses ranging from 300-900 mg) should be given orally as early in the disease course as possible and continued lifelong. Symptoms typically resolve within days.
  • Biotinidase deficiency (BTD). All symptomatic children with profound biotinidase deficiency improve when treated with 5-10 mg of oral biotin per day. Biotin treatment should be continued lifelong in all individuals with profound biotinidase deficiency.
  • Coenzyme Q10 biosynthesis deficiency (PDSS2, COQ9). Supplementation with oral coenzyme Q10 (10-30 mg/kg/day in children and 1200-3000 mg/day in adults) should be commenced as early in the disease course as possible and continued lifelong [Rahman et al 2012].

Supportive management for any of the causes of nuclear gene-encoded LSS includes the following:

  • Acidosis. Sodium bicarbonate or sodium citrate is appropriate for acute exacerbations of acidosis.
  • Seizures. Appropriate anti-seizure medications tailored to the type of seizure should be administered under the supervision of a neurologist. Sodium valproate and barbiturates should be avoided because of their inhibitory effects on the mitochondrial respiratory chain [Melegh & Trombitas 1997, Anderson et al 2002].
  • Dystonia
    • Benzhexol, baclofen, tetrabenezine, and gabapentin may be useful, alone or in various combinations; an initial low dose should be started and gradually increased until symptom control is achieved or intolerable side effects occur.
    • Botulinum toxin injection has also been used in individuals with Leigh syndrome and severe intractable dystonia.
  • Cardiomyopathy. Medical therapy may be required and should be supervised by a cardiologist.
  • Nutrition. Regular assessment of daily caloric intake and adequacy of dietary structure including micronutrients and feeding management is indicated. While the ketogenic diet may be indicated for individuals with pyruvate dehydrogenase (PDH) deficiency or drug-resistant epilepsy, there is no evidence for efficacy of the ketogenic diet in other forms of LSS.
  • Psychological support for the affected individual and family is essential.

Prevention of Secondary Complications

Anesthesia can potentially aggravate respiratory symptoms and precipitate respiratory failure; thus, careful consideration should be given to its use and to monitoring the individual prior to, during, and after its use [Shear & Tobias 2004, Niezgoda & Morgan 2013].

Surveillance

Affected individuals should be followed at regular intervals (typically every 6-12 months) to monitor progression and the appearance of new manifestations. Neurologic, ophthalmologic, audiologic, and cardiologic evaluations are recommended. Surveillance may be directed by knowledge of phenotypes known to be associated with specific gene defects [Rahman et al 2017].

Agents/Circumstances to Avoid

A recent Delphi review has examined drug safety in mitochondrial disorders [De Vries et al 2020].

5. Genetic Counseling of Family Members of an Individual with Nuclear Gene-Encoded Leigh Syndrome

Genetic counseling is the process of providing individuals and families with information on the nature, mode(s) of inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members; it is not meant to address all personal, cultural, or ethical issues that may arise or to substitute for consultation with a genetics professional. —ED.

Mode of Inheritance

Nuclear gene-encoded Leigh syndrome spectrum (LSS) can be inherited in an autosomal recessive, X-linked, or autosomal dominant manner.

  • Of the more than 80 nuclear gene-encoded LSS-related genes identified to date, pathogenic variants in all but four genes are associated with autosomal recessive inheritance.
  • LSS caused by a heterozygous or hemizygous pathogenic variant in AIFM1, NDUFA1, or PDHA1 is inherited in an X-linked manner. Note: Almost equal numbers of males and females affected with PDHA1-LSS have been reported [Lissens et al 2000, Imbard et al 2011]. Although relatively few affected individuals with NDUFA1-LSS and AIFM1-LSS have been reported, it is expected that the same sex ratio would be seen in all three X-linked disorders.
  • LSS caused by a heterozygous pathogenic variant in DNM1L is an autosomal dominant disorder; to date, all individuals with DNM1L-LSS have had the disorder as the result of a de novo pathogenic variant.

Genetic counseling regarding risk to family members depends on accurate diagnosis, determination of the mode of inheritance in each family, and results of molecular genetic testing.

Autosomal Recessive LSS – Risk to Family Members

Parents of a proband

  • The parents of an affected child are obligate heterozygotes (i.e., presumed to be carriers of one LSS-causing pathogenic variant based on family history).
  • If the causative pathogenic variants have been identified in the proband, molecular genetic testing is recommended for the parents of a proband to confirm that both parents are carriers and to allow reliable recurrence risk assessment. (De novo variants are known to occur at a low but appreciable rate in autosomal recessive disorders [Jónsson et al 2017].)
  • Heterozygotes (carriers) are asymptomatic and are not at risk of developing the disorder.

Sibs of a proband

  • If both parents are known to be heterozygous for an LSS-causing pathogenic variant, each sib of an affected individual has at conception a 25% chance of being affected, a 50% chance of being an asymptomatic carrier, and a 25% chance of being unaffected and not a carrier.
  • Heterozygotes (carriers) are asymptomatic and are not at risk of developing the disorder.

Offspring of a proband. Individuals with autosomal recessive LSS are not known to reproduce.

Other family members. Each sib of the proband's parents is at a 50% risk of being a carrier of a pathogenic variant.

Carrier detection. Carrier testing for at-risk relatives requires prior identification of the pathogenic variants in the family.

X-Linked LSS – Risk to Family Members

Parents of a male proband

  • The father of an affected male will not have X-linked LSS nor will he be hemizygous for an AIFM1, NDUFA1, or PDHA1 pathogenic variant; therefore, he does not require further evaluation/testing.
  • In a family with more than one affected individual, the mother of an affected male is an obligate heterozygote. Note: The 11 heterozygous mothers reported by Imbard et al [2011] were said to be asymptomatic. In the authors' experience, however, some learning difficulties or other features are often present.
  • If a woman has more than one affected child and no other affected relatives and if the pathogenic variant identified in the proband cannot be detected in her leukocyte DNA, she most likely has germline mosaicism. (Note: If the mother has both somatic and germline mosaicism for the variant, she may be mildly/minimally affected.)
  • If a male is the only affected family member (i.e., a simplex case):
    • The mother may be a heterozygote (approximately 25% of mothers of children with PDHA1 pathogenic variants were found to be heterozygous for the pathogenic variant [Lissens et al 2000, Imbard et al 2011]); or
    • The proband may have a de novo pathogenic variant, in which case the mother is not a heterozygote. Data reported by Lissens et al [2000] and Imbard et al [2011] suggest that up to 75% of affected individuals have a de novo pathogenic variant. In at least seven affected individuals, somatic mosaicism has been demonstrated with the de novo variant presumably occurring in early or mid-embryogenesis [Imbard et al 2011].

Parents of a female proband. A female proband may have inherited the AIFM1, NDUFA1, or PDHA1 pathogenic variant from either her mother or (theoretically) her father (if he has germline mosaicism for the pathogenic variant), or the pathogenic variant may be de novo.

Sibs of a male proband. The risk to sibs of a male proband depends on the genetic status of the mother:

  • If the mother of the proband has an AIFM1, NDUFA1, or PDHA1 pathogenic variant, the chance of transmitting it in each pregnancy is 50%. Males who inherit the pathogenic variant will be affected; females who inherit the pathogenic variant are likely to be affected with manifestations ranging from mild learning difficulty to LSS depending on the X-chromosome inactivation ratio.
  • If the proband represents a simplex case (i.e., a single occurrence in a family) and if the pathogenic variant cannot be detected in the leukocyte DNA of the mother, the risk to sibs is greater than that of the general population because of the possibility of maternal germline mosaicism.

Sibs of a female proband. The risk to sibs of a female proband depends on the genetic status of the mother (see above) and the father. Theoretically, the father of an affected female may have germline mosaicism for a pathogenic variant, in which case all of his daughters (and none of his sons) would be at risk of inheriting a pathogenic variant.

Offspring of a male proband. Males with X-linked LSS are not known to reproduce.

Offspring of a female proband. Each child of a female with X-linked LSS has a 50% chance of inheriting the pathogenic variant; males who inherit the pathogenic variant will be affected; females who inherit the pathogenic variant are likely to be affected with manifestations ranging from mild learning difficulty to LSS depending on the X-chromosome inactivation ratio.

Other family members. The proband's maternal aunts may be at risk of being heterozygous and the aunts' offspring may be at risk of inheriting a pathogenic variant and being affected.

Autosomal Dominant LSS – Risk to Family Members

Parents of a proband

  • All probands reported to date with DNM1L-LSS whose parents have undergone molecular genetic testing have the disorder as the result of a de novo pathogenic variant.
  • Molecular genetic testing is recommended for the parents of the proband to confirm their genetic status and to allow reliable recurrence risk counseling.
  • If the DNM1L pathogenic variant found in the proband cannot be detected in parental DNA, the pathogenic variant most likely occurred de novo in the proband. Another possible explanation is that the proband inherited a pathogenic variant from a parent with germline mosaicism. Although theoretically possible, no instances of parental germline mosaicism have been reported to date.

Sibs of a proband. The risk to the sibs of the proband depends on the genetic status of the proband's parents:

  • If a parent of the proband has the DNM1L pathogenic variant, the risk to the sibs of inheriting the variant is 50%.
  • If the DNM1L pathogenic variant found in the proband cannot be detected in the leukocyte DNA of either parent, the recurrence risk to sibs is estimated to be 1% because of the theoretic possibility of parental germline mosaicism [Rahbari et al 2016].

Offspring of a proband. Individuals with DNM1L-LSS are not known to reproduce.

Other family members. Given that all probands with autosomal dominant DNM1L-LSS reported to date have the disorder as a result of a de novo pathogenic variant, the risk to other family members is presumed to be low.

Prenatal Testing and Preimplantation Genetic Testing

Once the pathogenic variant(s) have been identified in an affected family member, prenatal testing for a pregnancy at increased risk and preimplantation genetic testing are possible.

Note: For families with X-linked LSS, molecular genetic prenatal test results cannot be used to predict the risk of an affected outcome for a female conceptus heterozygous for the familial pathogenic variant.

Differences in perspective may exist among medical professionals and within families regarding the use of prenatal testing. While most centers would consider use of prenatal testing to be a personal decision, discussion of these issues may be helpful.

Resources

GeneReviews staff has selected the following disease-specific and/or umbrella support organizations and/or registries for the benefit of individuals with this disorder and their families. GeneReviews is not responsible for the information provided by other organizations. For information on selection criteria, click here.

  • United Mitochondrial Disease Foundation
    Phone: 888-317-UMDF (8633)
    Email: info@umdf.org
  • Association Contre Les Maladies Mitochondriales
    France
    Phone: 33 6 30 84 58 27
    Email: assoammi@gmail.com
  • Deutsche Gesellschaft für Muskelkranke e.V.
    Germany
    Email: info@dgm.org
  • International Mito Patients
  • Mito Foundation
    Australia
    Phone: 61-1-300-977-180
    Email: info@mito.org.au
  • MitoAction
    Phone: 888-648-6228
    Email: support@mitoaction.org
  • MitoCanada Foundation
    Canada
    Phone: 289-807-2929; 877-708-6486 (MITO)
    Email: info@mitocanada.org
  • Mitocon – Insieme per lo studio e la cura delle malattie mitocondriali Onlus
    Mitocon is the reference association in Italy for patients suffering from mitochondrial diseases and their families and is the main link between patients and the scientific community.
    Italy
    Phone: 06 66991333/4
    Email: info@mitocon.it
  • People Against Leigh Syndrome (PALS)
    Phone: 713-248-8782
  • The Freya Foundation
    The aim of The Freya Foundation is to raise awareness for the condition called PDH, or pyruvate dehydrogenase deficiency complex.
    United Kingdom
  • The Lily Foundation
    United Kingdom
    Email: liz@thelilyfoundation.org.uk
  • RDCRN Patient Contact Registry: North American Mitochondrial Disease Consortium

Chapter Notes

Author Notes

Professor Rahman's web page

Professor Thorburn's web page

Professor Rahman's research interests include identification of novel nuclear genes causing mitochondrial disease using a combination of approaches including homozygosity mapping and exome and genome next-generation sequencing. Her group has identified a number of nuclear genes causing childhood-onset mitochondrial disorders, including genes involved in mitochondrial DNA maintenance and expression, complex I and complex IV function, and biosynthesis of coenzyme Q10. Other research interests aim to identify biomarkers and novel therapies for childhood mitochondrial disorders.

David Thorburn's research focuses on improving diagnosis, prevention, and treatment of mitochondrial energy generation disorders. This has included translating knowledge of mitochondrial DNA genetics into reproductive options for families, defining diagnostic criteria and epidemiology, and discovery of new "disease" genes through next-generation DNA sequencing. His group also uses cellular and mouse models to understand pathogenic mechanisms and trial new treatment approaches.

Revision History

  • 16 July 2020 (bp) Comprehensive update posted live
  • 1 October 2015 (me) Review posted live
  • 17 February 2015 (sr) Original submission

References

Literature Cited

  • Abrams AJ, Hufnagel RB, Rebelo A, Zanna C, Patel N, Gonzalez MA, Campeanu IJ, Griffin LB, Groenewald S, Strickland AV, Tao F, Speziani F, Abreu L, Schüle R, Caporali L, La Morgia C, Maresca A, Liguori R, Lodi R, Ahmed ZM, Sund KL, Wang X, Krueger LA, Peng Y, Prada CE, Prows CA, Schorry EK, Antonellis A, Zimmerman HH, Abdul-Rahman OA, Yang Y, Downes SM, Prince J, Fontanesi F, Barrientos A, Németh AH, Carelli V, Huang T, Zuchner S, Dallman JE. Mutations in SLC25A46, encoding a UGO1-like protein, cause an optic atrophy spectrum disorder. Nat Genet. 2015;47:926–32. [PMC free article: PMC4520737] [PubMed: 26168012]

  • Ahola S, Isohanni P, Euro L, Brilhante V, Palotie A, Pihko H, Lönnqvist T, Lehtonen T, Laine J, Tyynismaa H, Suomalainen A. Mitochondrial EFTs defects in juvenile-onset Leigh disease, ataxia, neuropathy, and optic atrophy. Neurology. 2014;83:743–51. [PMC free article: PMC4150129] [PubMed: 25037205]

  • Alston CL, He L, Morris AA, Hughes I, de Goede C, Turnbull DM, McFarland R, Taylor RW. Maternally inherited mitochondrial DNA disease in consanguineous families. Eur J Hum Genet. 2011;19:1226–9. [PMC free article: PMC3230363] [PubMed: 21712854]

  • Alston CL, Veling MT, Heidler J, Taylor LS, Alaimo JT, Sung AY, He L, Hopton S, Broomfield A, Pavaine J, Diaz J, Leon E, Wolf P, McFarland R, Prokisch H, Wortmann SB, Bonnen PE, Wittig I, Pagliarini DJ, Taylor RW. Pathogenic bi-allelic mutations in NDUFAF8 cause Leigh syndrome with an isolated complex I deficiency. Am J Hum Genet. 2020;106:92–101. [PMC free article: PMC7042492] [PubMed: 31866046]

  • Anderson CM, Norquist BA, Vesce S, Nicholls DG, Soine WH, Duan S, Swanson RA. Barbiturates induce mitochondrial depolarization and potentiate excitotoxic neuronal death. J Neurosci. 2002;22:9203–9. [PMC free article: PMC6758030] [PubMed: 12417645]

  • Antonicka H, Leary SC, Guercin GH, Agar JN, Horvath R, Kennaway NG, Harding CO, Jaksch M, Shoubridge EA. Mutations in COX10 result in a defect in mitochondrial heme A biosynthesis and account for multiple, early onset clinical phenotypes associated with isolated COX deficiency. Hum Mol Genet. 2003;12:2693–702. [PubMed: 12928484]

  • Antonicka H, Ostergaard E, Sasarman F, Weraarpachai W, Wibrand F, Pedersen AM, Rodenburg RJ, van der Knaap MS, Smeitink JA, Chrzanowska-Lightowlers ZM, Shoubridge EA. Mutations in C12orf65 in patients with encephalomyopathy and a mitochondrial translation defect. Am J Hum Genet. 2010;87:115–22. [PMC free article: PMC2896764] [PubMed: 20598281]

  • Baertling F, Sánchez-Caballero L, van den Brand MAM, Wintjes LT, Brink M, van den Brandt FA, Wilson C, Rodenburg RJT, Nijtmans LGJ. NDUFAF4 variants are associated with Leigh syndrome and cause a specific mitochondrial complex I assembly defect. Eur J Hum Genet. 2017;25:1273–7. [PMC free article: PMC5643967] [PubMed: 28853723]

  • Baker PR 2nd, Friederich MW, Swanson MA, Shaikh T, Bhattacharya K, Scharer GH, Aicher J, Creadon-Swindell G, Geiger E, Maclean KN, Lee WT, Deshpande C, Freckmann ML, Shih LY, Wasserstein M, Rasmussen MB, Lund AM, Procopis P, Cameron JM, Robinson BH, Brown GK, Brown RM, Compton AG, Dieckmann CL, Collard R, Coughlin CR 2nd, Spector E, Wempe MF, Van Hove JL. Variant nonketotic hyperglycinemia is caused by mutations in LIAS, BOLA3 and the novel gene GLRX5. Brain. 2014;137:366–79. [PMC free article: PMC3914472] [PubMed: 24334290]

  • Barca E, Ganetzky RD, Potluri P, Juanola-Falgarona M, Gai X, Li D, Jalas C, Hirsch Y, Emmanuele V, Tadesse S, Ziosi M, Akman HO, Chung WK, Tanji K, McCormick EM, Place E, Consugar M, Pierce EA, Hakonarson H, Wallace DC, Hirano M, Falk MJ. USMG5 Ashkenazi Jewish founder mutation impairs mitochondrial complex V dimerization and ATP synthesis. Hum Mol Genet. 2018;27:3305–12. [PMC free article: PMC6140788] [PubMed: 29917077]

  • Barel O, Shorer Z, Flusser H, Ofir R, Narkis G, Finer G, Shalev H, Nasasra A, Saada A, Birk OS. Mitochondrial complex III deficiency associated with a homozygous mutation in UQCRQ. Am J Hum Genet. 2008;82:1211–6. [PMC free article: PMC2427202] [PubMed: 18439546]

  • Barghuti F, Elian K, Gomori JM, Shaag A, Edvardson S, Saada A, Elpeleg O. The unique neuroradiology of complex I deficiency due to NDUFA12L defect. Mol Genet Metab. 2008;94:78–82. [PubMed: 18180188]

  • Bénit P, Chretien D, Kadhom N, de Lonlay-Debeney P, Cormier-Daire V, Cabral A, Peudenier S, Rustin P, Munnich A, Rotig A. Large-scale deletion and point mutations of the nuclear NDUFV1 and NDUFS1 genes in mitochondrial complex I deficiency. Am J Hum Genet. 2001;68:1344–52. [PMC free article: PMC1226121] [PubMed: 11349233]

  • Bénit P, Slama A, Cartault F, Giurgea I, Chretien D, Lebon S, Marsac C, Munnich A, Rotig A, Rustin P. Mutant NDUFS3 subunit of mitochondrial complex I causes Leigh syndrome. J Med Genet. 2004;41:14–7. [PMC free article: PMC1757256] [PubMed: 14729820]

  • Borna NN, Kishita Y, Kohda M, Lim SC, Shimura M, Wu Y, Mogushi K, Yatsuka Y, Harashima H, Hisatomi Y, Fushimi T, Ichimoto K, Murayama K, Ohtake A, Okazaki Y. Mitochondrial ribosomal protein PTCD3 mutations cause oxidative phosphorylation defects with Leigh syndrome. Neurogenetics. 2019;20:9–25. [PubMed: 30607703]

  • Bourgeron T, Rustin P, Chretien D, Birch-Machin M, Bourgeois M, Viegas-Pequignot E, Munnich A, Rotig A. Mutation of a nuclear succinate dehydrogenase gene results in mitochondrial respiratory chain deficiency. Nat Genet. 1995;11:144–9. [PubMed: 7550341]

  • Budde SM, van den Heuvel LP, Janssen AJ, Smeets RJ, Buskens CA, DeMeirleir L, Van Coster R, Baethmann M, Voit T, Trijbels JM, Smeitink JA. Combined enzymatic complex I and III deficiency associated with mutations in the nuclear encoded NDUFS4 gene. Biochem Biophys Res Commun. 2000;275:63–8. [PubMed: 10944442]

  • Calvo SE, Tucker EJ, Compton AG, Kirby DM, Crawford G, Burtt NP, Rivas M, Guiducci C, Bruno DL, Goldberger OA, Redman MC, Wiltshire E, Wilson CJ, Altshuler D, Gabriel SB, Daly MJ, Thorburn DR, Mootha VK. High-throughput, pooled sequencing identifies mutations in NUBPL and FOXRED1 in human complex I deficiency. Nat Genet. 2010;42:851–8. [PMC free article: PMC2977978] [PubMed: 20818383]

  • Cameron JM, MacKay N, Feigenbaum A, Tarnopolsky M, Blaser S, Robinson BH, Schulze A. Exome sequencing identifies complex I NDUFV2 mutations as a novel cause of Leigh syndrome. Eur J Paediatr Neurol. 2015;19:525–32. [PubMed: 26008862]

  • Debray FG, Mitchell GA, Allard P, Robinson BH, Hanley JA, Lambert M. Diagnostic accuracy of blood lactate-to-pyruvate molar ratio in the differential diagnosis of congenital lactic acidosis. Clin Chem. 2007;53:916–21. [PubMed: 17384007]

  • Debray FG, Morin C, Janvier A, Villeneuve J, Maranda B, Laframboise R, Lacroix J, Decarie JC, Robitaille Y, Lambert M, Robinson BH, Mitchell GA. LRPPRC mutations cause a phenotypically distinct form of Leigh syndrome with cytochrome c oxidase deficiency. J Med Genet. 2011;48:183–9. [PubMed: 21266382]

  • de Lonlay P, Valnot I, Barrientos A, Gorbatyuk M, Tzagoloff A, Taanman JW, Benayoun E, Chrétien D, Kadhom N, Lombès A, de Baulny HO, Niaudet P, Munnich A, Rustin P, Rötig A. A mutant mitochondrial respiratory chain assembly protein causes complex III deficiency in patients with tubulopathy, encephalopathy and liver failure. Nat Genet. 2001;29:57–60. [PubMed: 11528392]

  • De Vries MC, Brown DA, Allen ME, Bindoff L, Gorman GS, Karaa A, Keshavan N, Lamperti C, McFarland R, Ng YS, O'Callaghan M, Pitceathly RDS, Rahman S, Russel FGM, Varhaug KN, Schirris TJJ, Mancuso M. Safety of drug use in patients with a primary mitochondrial disease: An international Delphi-based consensus. J Inher Metab Dis. 2020;43:800–18. [PMC free article: PMC7383489] [PubMed: 32030781]

  • Elpeleg O, Miller C, Hershkovitz E, Bitner-Glindzicz M, Bondi-Rubinstein G, Rahman S, Pagnamenta A, Eshhar S, Saada A. Deficiency of the ADP-forming succinyl-CoA synthase activity is associated with encephalomyopathy and mitochondrial DNA depletion. Am J Hum Genet. 2005;76:1081–6. [PMC free article: PMC1196446] [PubMed: 15877282]

  • Fassone E, Duncan AJ, Taanman JW, Pagnamenta AT, Sadowski MI, Holand T, Qasim W, Rutland P, Calvo SE, Mootha VK, Bitner-Glindzicz M, Rahman S. FOXRED1, encoding an FAD-dependent oxidoreductase complex-I-specific molecular chaperone, is mutated in infantile-onset mitochondrial encephalopathy. Hum Mol Genet. 2010;19:4837–47. [PMC free article: PMC4560042] [PubMed: 20858599]

  • Fassone E, Rahman S. Complex I deficiency: clinical features, biochemistry and molecular genetics. J Med Genet. 2012;49:578–90. [PubMed: 22972949]

  • Fassone E, Wedatilake Y, DeVile CJ, Chong WK, Carr LJ, Rahman S. Treatable Leigh-like encephalopathy presenting in adolescence. BMJ Case Rep. 2013;2013:200838. [PMC free article: PMC3822156] [PubMed: 24099834]

  • Ferdinandusse S, Waterham HR, Heales SJ, Brown GK, Hargreaves IP, Taanman JW, Gunny R, Abulhoul L, Wanders RJ, Clayton PT, Leonard JV, Rahman S. HIBCH mutations can cause Leigh-like disease with combined deficiency of multiple mitochondrial respiratory chain enzymes and pyruvate dehydrogenase. Orphanet J Rare Dis. 2013;8:188. [PMC free article: PMC4222069] [PubMed: 24299452]

  • Fernandez-Moreira D, Ugalde C, Smeets R, Rodenburg RJ, Lopez-Laso E, Ruiz-Falco ML, Briones P, Martin MA, Smeitink JA, Arenas J. X-linked NDUFA1 gene mutations associated with mitochondrial encephalomyopathy. Ann Neurol. 2007;61:73–83. [PubMed: 17262856]

  • Floyd BJ, Wilkerson EM, Veling MT, Minogue CE, Xia C, Beebe ET, Wrobel RL, Cho H, Kremer LS, Alston CL, Gromek KA, Dolan BK, Ulbrich A, Stefely JA, Bohl SL, Werner KM, Jochem A, Westphall MS, Rensvold JW, Taylor RW, Prokisch H, Kim JJ, Coon JJ, Pagliarini DJ. Mitochondrial protein interaction mapping identifies regulators of respiratory chain function. Mol Cell. 2016;63:621–32. [PMC free article: PMC4992456] [PubMed: 27499296]

  • Friederich MW, Elias AF, Kuster A, Laugwitz L, Larson AA, Landry AP, Ellwood-Digel L, Mirsky DM, Dimmock D, Haven J, Jiang H, MacLean KN, Styren K, Schoof J, Goujon L, Lefrancois T, Friederich M, Coughlin CR 2nd, Banerjee R, Haack TB, Van Hove JLK. Pathogenic variants in SQOR encoding sulfide: quinone oxidoreductase are a potentially treatable cause of Leigh disease. J Inherit Metab Dis. 2020;43:1024–36. [PMC free article: PMC7484123] [PubMed: 32160317]

  • Fukumura S, Ohba C, Watanabe T, Minagawa K, Shimura M, Murayama K, Ohtake A, Saitsu H, Matsumoto N, Tsutsumi H. Compound heterozygous GFM2 mutations with Leigh syndrome complicated by arthrogryposis multiplex congenita. J Hum Genet. 2015;60:509–13. [PubMed: 26016410]

  • Gerards M, Kamps R, van Oevelen J, Boesten I, Jongen E, de Koning B, Scholte HR, de Angst I, Schoonderwoerd K, Sefiani A, Ratbi I, Coppieters W, Karim L, de Coo R, van den Bosch B, Smeets H. Exome sequencing reveals a novel Moroccan founder mutation in SLC19A3 as a new cause of early-childhood fatal Leigh syndrome. Brain. 2013;136:882–90. [PubMed: 23423671]

  • Gerards M, Sluiter W, van den Bosch BJ, de Wit LE, Calis CM, Frentzen M, Akbari H, Schoonderwoerd K, Scholte HR, Jongbloed RJ, Hendrickx AT, de Coo IF, Smeets HJ. Defective complex I assembly due to C20orf7 mutations as a new cause of Leigh syndrome. J Med Genet. 2010;47:507–12. [PMC free article: PMC2921275] [PubMed: 19542079]

  • Ghezzi D, Arzuffi P, Zordan M, Da Re C, Lamperti C, Benna C, D'Adamo P, Diodato D, Costa R, Mariotti C, Uziel G, Smiderle C, Zeviani M. Mutations in TTC19 cause mitochondrial complex III deficiency and neurological impairment in humans and flies. Nat Genet. 2011;43:259–63. [PubMed: 21278747]

  • Ghezzi D, Sevrioukova I, Invernizzi F, Lamperti C, Mora M, D'Adamo P, Novara F, Zuffardi O, Uziel G, Zeviani M. Severe X-linked mitochondrial encephalomyopathy associated with a mutation in apoptosis-inducing factor. Am J Hum Genet. 2010;86:639–49. [PMC free article: PMC2850437] [PubMed: 20362274]

  • Grafakou O, Oexle K, van den Heuvel L, Smeets R, Trijbels F, Goebel HH, Bosshard N, Superti-Furga A, Steinmann B, Smeitink J. Leigh syndrome due to compound heterozygosity of dihydrolipoamide dehydrogenase gene mutations. Description of the first E3 splice site mutation. Eur J Pediatr. 2003;162:714–8. [PubMed: 12925875]

  • Hallmann K, Kudin AP, Zsurka G, Kornblum C, Reimann J, Stüve B, Waltz S, Hattingen E, Thiele H, Nürnberg P, Rüb C, Voos W, Kopatz J, Neumann H, Kunz WS. Loss of the smallest subunit of cytochrome c oxidase, COX8A, causes Leigh-like syndrome and epilepsy. Brain. 2016;139:338–45. [PubMed: 26685157]

  • Hayhurst H, de Coo IFM, Piekutowska-Abramczuk D, Alston CL, Sharma S, Thompson K, Rius R, He L, Hopton S, Ploski R, Ciara E, Lake NJ, Compton AG, Delatycki MB, Verrips A, Bonnen PE, Jones SA, Morris AA, Shakespeare D, Christodoulou J, Wesol-Kucharska D, Rokicki D, Smeets HJM, Pronicka E, Thorburn DR, Gorman GS, McFarland R, Taylor RW, Ng YS. Leigh syndrome caused by mutations in MTFMT is associated with a better prognosis. Ann Clin Transl Neurol. 2019;6:515–24. [PMC free article: PMC6414492] [PubMed: 30911575]

  • Head RA, Brown RM, Zolkipli Z, Shahdadpuri R, King MD, Clayton PT, Brown GK. Clinical and genetic spectrum of pyruvate dehydrogenase deficiency: dihydrolipoamide acetyltransferase (E2) deficiency. Ann Neurol. 2005;58:234–41. [PubMed: 16049940]

  • Hoefs SJ, Dieteren CE, Distelmaier F, Janssen RJ, Epplen A, Swarts HG, Forkink M, Rodenburg RJ, Nijtmans LG, Willems PH, Smeitink JA, van den Heuvel LP. NDUFA2 complex I mutation leads to Leigh disease. Am J Hum Genet. 2008;82:1306–15. [PMC free article: PMC2427319] [PubMed: 18513682]

  • Hoefs SJ, van Spronsen FJ, Lenssen EW, Nijtmans LG, Rodenburg RJ, Smeitink JA, van den Heuvel LP. NDUFA10 mutations cause complex I deficiency in a patient with Leigh disease. Eur J Hum Genet. 2011;19:270–4. [PMC free article: PMC3061993] [PubMed: 21150889]

  • Imbard A, Boutron A, Vequaud C, Zater M, de Lonlay P, de Baulny HO, Barnerias C, Miné M, Marsac C, Saudubray JM, Brivet M. Molecular characterization of 82 patients with pyruvate dehydrogenase complex deficiency. Structural implications of novel amino acid substitutions in E1 protein. Mol Genet Metab. 2011;104:507–16. [PubMed: 21914562]

  • Janer A, Prudent J, Paupe V, Fahiminiya S, Majewski J, Sgarioto N, Des Rosiers C, Forest A, Lin ZY, Gingras AC, Mitchell G, McBride HM, Shoubridge EA. SLC25A46 is required for mitochondrial lipid homeostasis and cristae maintenance and is responsible for Leigh syndrome. EMBO Mol Med. 2016;8:1019–38. [PMC free article: PMC5009808] [PubMed: 27390132]

  • Jónsson H, Sulem P, Kehr B, Kristmundsdottir S, Zink F, Hjartarson E, Hardarson MT, Hjorleifsson KE, Eggertsson HP, Gudjonsson SA, Ward LD, Arnadottir GA, Helgason EA, Helgason H, Gylfason A, Jonasdottir A, Jonasdottir A, Rafnar T, Frigge M, Stacey SN, Th Magnusson O, Thorsteinsdottir U, Masson G, Kong A, Halldorsson BV, Helgason A, Gudbjartsson DF, Stefansson K. Parental influence on human germline de novo mutations in 1,548 trios from Iceland. Nature. 2017;549:519–22. [PubMed: 28959963]

  • Joost K, Rodenburg R, Piirsoo A, van den Heuvel B, Zordania R, Ounap K. A novel mutation in the SCO2 gene in a neonate with early-onset cardioencephalomyopathy. Pediatr Neurol. 2010;42:227–30. [PubMed: 20159436]

  • Koch J, Feichtinger RG, Freisinger P, Pies M, Schrödl F, Iuso A, Sperl W, Mayr JA, Prokisch H, Haack TB. Disturbed mitochondrial and peroxisomal dynamics due to loss of MFF causes Leigh-like encephalopathy, optic atrophy and peripheral neuropathy. J Med Genet. 2016;53:270–8. [PubMed: 26783368]

  • Kopajtich R, Nicholls TJ, Rorbach J, Metodiev MD, Freisinger P, Mandel H, Vanlander A, Ghezzi D, Carrozzo R, Taylor RW, Marquard K, Murayama K, Wieland T, Schwarzmayr T, Mayr JA, Pearce SF, Powell CA, Saada A, Ohtake A, Invernizzi F, Lamantea E, Sommerville EW, Pyle A, Chinnery PF, Crushell E, Okazaki Y, Kohda M, Kishita Y, Tokuzawa Y, Assouline Z, Rio M, Feillet F, Mousson de Camaret B, Chretien D, Munnich A, Menten B, Sante T, Smet J, Régal L, Lorber A, Khoury A, Zeviani M, Strom TM, Meitinger T, Bertini ES, Van Coster R, Klopstock T, Rötig A, Haack TB, Minczuk M, Prokisch H. Mutations in GTPBP3 cause a mitochondrial translation defect associated with hypertrophic cardiomyopathy, lactic acidosis, and encephalopathy. Am J Hum Genet. 2014;95:708–20. [PMC free article: PMC4259976] [PubMed: 25434004]

  • Kremer LS, Bader DM, Mertes C, Kopajtich R, Pichler G, Iuso A, Haack TB, Graf E, Schwarzmayr T, Terrile C, Konarikova E, Repp B, Kastenmuller G, Adamski J, Lichtner P, Leonhardt C, Funalot B, Donati A, Tiranti V, Lombes A, Jardel C, Glaser D, Taylor RW, Ghezzi D, Mayr JA, Rotig A, Freisinger P, Distelmaier F, Strom TM, Meitinger T, Gagneur J, Prokisch H. Genetic diagnosis of Mendelian disorders via RNA sequencing. Nat Commun. 2017;8:15824. [PMC free article: PMC5499207] [PubMed: 28604674]

  • Lake NJ, Compton AG, Rahman S, Thorburn DR. Leigh syndrome: one disorder, more than 75 monogenic causes. Ann Neurol. 2016;79:190–203. [PubMed: 26506407]

  • Lake NJ, Webb BD, Stroud DA, Richman TR, Ruzzenente B, Compton AG, Mountford HS, Pulman J, Zangarelli C, Rio M, Bodaert N, Assouline Z, Sherpa M, Schadt EE, Houten SM, Byrnes J, McCormick EM, Zolkipli-Cunningham Z, Haude K, Zhang Z, Retterer K, Bai R, Calvo SE, Mootha VK, Christodoulou J, Rotig A, Filipovska A, Cristian I, Falk MJ, Metodiev MD, Thorburn DR. Biallelic mutations in MRPS34 lead to instability of the small mitoribosomal subunit and Leigh syndrome. Am J Hum Genet. 2017;101:239–54. [PMC free article: PMC5544391] [PubMed: 28777931]

  • Lim SC, Smith KR, Stroud DA, Compton AG, Tucker EJ, Dasvarma A, Gandolfo LC, Marum JE, McKenzie M, Peters HL, Mowat D, Procopis PG, Wilcken B, Christodoulou J, Brown GK, Ryan MT, Bahlo M, Thorburn DR. A founder mutation in PET100 causes isolated complex IV deficiency in Lebanese individuals with Leigh syndrome. Am J Hum Genet. 2014;94:209–22. [PMC free article: PMC3928654] [PubMed: 24462369]

  • Lissens W, De Meirleir L, Seneca S, Liebaers I, Brown GK, Brown RM, Ito M, Naito E, Kuroda Y, Kerr DS, Wexler ID, Patel MS, Robinson BH, Seyda A. Mutations in the X-linked pyruvate dehydrogenase (E1) alpha subunit gene (PDHA1) in patients with a pyruvate dehydrogenase complex deficiency. Hum Mutat. 2000;15:209–19. [PubMed: 10679936]

  • Loeffen J, Elpeleg O, Smeitink J, Smeets R, Stöckler-Ipsiroglu S, Mandel H, Sengers R, Trijbels F, van den Heuvel L. Mutations in the complex I NDUFS2 gene of patients with cardiomyopathy and encephalomyopathy. Ann Neurol. 2001;49:195–201. [PubMed: 11220739]

  • Loeffen J, Smeitink J, Triepels R, Smeets R, Schuelke M, Sengers R, Trijbels F, Hamel B, Mullaart R, van den Heuvel L. The first nuclear-encoded complex I mutation in a patient with Leigh syndrome. Am J Hum Genet. 1998;63:1598–608. [PMC free article: PMC1377631] [PubMed: 9837812]

  • López LC, Schuelke M, Quinzii CM, Kanki T, Rodenburg RJ, Naini A, Dimauro S, Hirano M. Leigh syndrome with nephropathy and CoQ10 deficiency due to decaprenyl diphosphate synthase subunit 2 (PDSS2) mutations. Am J Hum Genet. 2006;79:1125–9. [PMC free article: PMC1698707] [PubMed: 17186472]

  • Maas RR, Iwanicka-Pronicka K, Kalkan Ucar S, Alhaddad B, AlSayed M, Al-Owain MA, Al-Zaidan HI, Balasubramaniam S, Barić I, Bubshait DK, Burlina A, Christodoulou J, Chung WK, Colombo R, Darin N, Freisinger P, Garcia Silva MT, Grunewald S, Haack TB, van Hasselt PM, Hikmat O, Hörster F, Isohanni P, Ramzan K, Kovacs-Nagy R, Krumina Z, Martin-Hernandez E, Mayr JA, McClean P, De Meirleir L, Naess K, Ngu LH, Pajdowska M, Rahman S, Riordan G, Riley L, Roeben B, Rutsch F, Santer R, Schiff M, Seders M, Sequeira S, Sperl W, Staufner C, Synofzik M, Taylor RW, Trubicka J, Tsiakas K, Unal O, Wassmer E, Wedatilake Y, Wolff T, Prokisch H, Morava E, Pronicka E, Wevers RA, de Brouwer AP, Wortmann SB. Progressive deafness-dystonia due to SERAC1 mutations: A study of 67 cases. Ann Neurol. 2017;82:1004–15. [PMC free article: PMC5847115] [PubMed: 29205472]

  • Martinelli D, Catteruccia M, Piemonte F, Pastore A, Tozzi G, Dionisi-Vici C, Pontrelli G, Corsetti T, Livadiotti S, Kheifets V, Hinman A, Shrader WD, Thoolen M, Klein MB, Bertini E, Miller G. EPI-743 reverses the progression of the pediatric mitochondrial disease--genetically defined Leigh Syndrome. Mol Genet Metab. 2012;107:383–8. [PubMed: 23010433]

  • Mayr JA, Freisinger P, Schlachter K, Rolinski B, Zimmermann FA, Scheffner T, Haack TB, Koch J, Ahting U, Prokisch H, Sperl W. Thiamine pyrophosphokinase deficiency in encephalopathic children with defects in the pyruvate oxidation pathway. Am J Hum Genet. 2011;89:806–12. [PMC free article: PMC3234371] [PubMed: 22152682]

  • Melegh B, Trombitas K. Valproate treatment induces lipid globule accumulation with ultrastructural abnormalities of mitochondria in skeletal muscle. Neuropediatrics. 1997;28:257–61. [PubMed: 9413004]

  • Mineri R, Rimoldi M, Burlina AB, Koskull S, Perletti C, Heese B, von Döbeln U, Mereghetti P, Di Meo I, Invernizzi F, Zeviani M, Uziel G, Tiranti V. Identification of new mutations in the ETHE1 gene in a cohort of 14 patients presenting with ethylmalonic encephalopathy. J Med Genet. 2008;45:473–8. [PubMed: 18593870]

  • Mitchell G, Ogier H, Munnich A, Saudubray JM, Shirrer J, Charpentier C, Rocchiccioli F. Neurological deterioration and lactic acidemia in biotinidase deficiency. A treatable condition mimicking Leigh's disease. Neuropediatrics. 1986;17:129–31. [PubMed: 3762868]

  • Mootha VK, Lepage P, Miller K, Bunkenborg J, Reich M, Hjerrild M, Delmonte T, Villeneuve A, Sladek R, Xu F, Mitchell GA, Morin C, Mann M, Hudson TJ, Robinson B, Rioux JD, Lander ES. Identification of a gene causing human cytochrome c oxidase deficiency by integrative genomics. Proc Natl Acad Sci U S A. 2003;100:605–10. [PMC free article: PMC141043] [PubMed: 12529507]

  • Niezgoda J, Morgan PG. Anesthetic considerations in patients with mitochondrial defects. Paediatr Anaesth. 2013;23:785–93. [PMC free article: PMC3711963] [PubMed: 23534340]

  • Ohlenbusch A, Edvardson S, Skorpen J, Bjornstad A, Saada A, Elpeleg O, Gärtner J, Brockmann K. Leukoencephalopathy with accumulated succinate is indicative of SDHAF1 related complex II deficiency. Orphanet J Rare Dis. 2012;7:69. [PMC free article: PMC3492161] [PubMed: 22995659]

  • Oktay Y, Güngör S, Zeltner L, Wiethoff S, Schöls L, Sonmezler E, Yilmaz E, Munro B, Bender B, Kernstock C, Kaemereit S, Liepelt I, Töpf A, Yis U, Laurie S, Yaramis A, Zuchner S, Hiz S, Lochmüller H, Schüle R, Horvath R. Confirmation of TACO1 as a Leigh syndrome disease gene in two additional families. J Neuromuscul Dis. 2020;7:301–8. [PMC free article: PMC7458500] [PubMed: 32444556]

  • Oquendo CE, Antonicka H, Shoubridge EA, Reardon W, Brown GK. Functional and genetic studies demonstrate that mutation in the COX15 gene can cause Leigh syndrome. J Med Genet. 2004;41:540–4. [PMC free article: PMC1735852] [PubMed: 15235026]

  • Ostergaard E, Hansen FJ, Sorensen N, Duno M, Vissing J, Larsen PL, Faeroe O, Thorgrimsson S, Wibrand F, Christensen E, Schwartz M. Mitochondrial encephalomyopathy with elevated methylmalonic acid is caused by SUCLA2 mutations. Brain. 2007;130:853–61. [PubMed: 17287286]

  • Ostergaard E, Rodenburg RJ, van den Brand M, Thomsen LL, Duno M, Batbayli M, Wibrand F, Nijtmans L. Respiratory chain complex I deficiency due to NDUFA12 mutations as a new cause of Leigh syndrome. J Med Genet. 2011;48:737–40. [PubMed: 21617257]

  • Pagliarini DJ, Calvo SE, Chang B, Sheth SA, Vafai SB, Ong SE, Walford GA, Sugiana C, Boneh A, Chen WK, Hill DE, Vidal M, Evans JG, Thorburn DR, Carr SA, Mootha VK. A mitochondrial protein compendium elucidates complex I disease biology. Cell. 2008;134:112–23. [PMC free article: PMC2778844] [PubMed: 18614015]

  • Pagnamenta AT, Hargreaves IP, Duncan AJ, Taanman JW, Heales SJ, Land JM, Bitner-Glindzicz M, Leonard JV, Rahman S. Phenotypic variability of mitochondrial disease caused by a nuclear mutation in complex II. Mol Genet Metab. 2006;89:214–21. [PubMed: 16798039]

  • Patel KP, O'Brien TW, Subramony SH, Shuster J, Stacpoole PW. The spectrum of pyruvate dehydrogenase complex deficiency: clinical, biochemical and genetic features in 371 patients. Mol Genet Metab. 2012;106:385–94. [PMC free article: PMC4003492] [PubMed: 22896851]

  • Peters H, Buck N, Wanders R, Ruiter J, Waterham H, Koster J, Yaplito-Lee J, Ferdinandusse S, Pitt J. ECHS1 mutations in Leigh disease: a new inborn error of metabolism affecting valine metabolism. Brain. 2014;137:2903–8. [PubMed: 25125611]

  • Pitceathly RD, Rahman S, Wedatilake Y, Polke JM, Cirak S, Foley AR, Sailer A, Hurles ME, Stalker J, Hargreaves I, Woodward CE, Sweeney MG, Muntoni F, Houlden H, Taanman JW, Hanna MG. UK10K Consortium. NDUFA4 mutations underlie dysfunction of a cytochrome c oxidase subunit linked to human neurological disease. Cell Rep. 2013;3:1795–805. [PMC free article: PMC3701321] [PubMed: 23746447]

  • Quinonez SC, Leber SM, Martin DM, Thoene JG, Bedoyan JK. Leigh syndrome in a girl with a novel DLD mutation causing E3 deficiency. Pediatr Neurol. 2013;48:67–72. [PMC free article: PMC4535688] [PubMed: 23290025]

  • Quintana E, Mayr JA, García Silva MT, Font A, Tortoledo MA, Moliner S, Ozaez L, Lluch M, Cabello A, Ricoy JR, Koch J, Ribes A, Sperl W, Briones P. PDH E(1)beta deficiency with novel mutations in two patients with Leigh syndrome. J Inherit Metab Dis. 2009;32 Suppl 1:S339–43. [PubMed: 19924563]

  • Rahbari R, Wuster A, Lindsay SJ, Hardwick RJ, Alexandrov LB, Turki SA, Dominiczak A, Morris A, Porteous D, Smith B, Stratton MR, Hurles ME, et al. Timing, rates and spectra of human germline mutation. Nat Genet. 2016;48:126–33. [PMC free article: PMC4731925] [PubMed: 26656846]

  • Rahman J, Noronha A, Thiele I, Rahman S. Leigh map: A novel computational diagnostic resource for mitochondrial disease. Ann Neurol. 2017;81:9–16. [PMC free article: PMC5347854] [PubMed: 27977873]

  • Rahman S, Blok RB, Dahl HH, Danks DM, Kirby DM, Chow CW, Christodoulou J, Thorburn DR. Leigh syndrome: clinical features and biochemical and DNA abnormalities. Ann Neurol. 1996;39:343–51. [PubMed: 8602753]

  • Rahman S, Clarke CF, Hirano M. 176th ENMC International Workshop: diagnosis and treatment of coenzyme Q10 deficiency. Neuromuscul Disord. 2012;22:76–86. [PMC free article: PMC3222743] [PubMed: 21723727]

  • Renkema GH, Visser G, Baertling F, Wintjes LT, Wolters VM, van Montfrans J, de Kort GAP, Nikkels PGJ, van Hasselt PM, van der Crabben SN, Rodenburg RJT. Mutated PET117 causes complex IV deficiency and is associated with neurodevelopmental regression and medulla oblongata lesions. Hum Genet. 2017;136:759–69. [PMC free article: PMC5429353] [PubMed: 28386624]

  • Riley LG, Cowley MJ, Gayevskiy V, Roscioli T, Thorburn DR, Prelog K, Bahlo M, Sue CM, Balasubramaniam S, Christodoulou J. A. SLC39A8 variant causes manganese deficiency, and glycosylation and mitochondrial disorders. J Inher Metab Dis. 2017;40:261–9. [PubMed: 27995398]

  • Sakai C, Yamaguchi S, Sasaki M, Miyamoto Y, Matsushima Y, Goto Y. ECHS1 mutations cause combined respiratory chain deficiency resulting in Leigh syndrome. Hum Mutat. 2015;36:232–9. [PubMed: 25393721]

  • Saunders C, Smith L, Wibrand F, Ravn K, Bross P, Thiffault I, Christensen M, Atherton A, Farrow E, Miller N, Kingsmore SF, Ostergaard E. CLPB variants associated with autosomal-recessive mitochondrial disorder with cataract, neutropenia, epilepsy, and methylglutaconic aciduria. Am J Hum Genet. 2015;96:258–65. [PMC free article: PMC4320254] [PubMed: 25597511]

  • Schiff M, Miné M, Brivet M, Marsac C, Elmaleh-Bergés M, Evrard P, Ogier de Baulny H. Leigh's disease due to a new mutation in the PDHX gene. Ann Neurol. 2006;59:709–14. [PubMed: 16566017]

  • Schwartzentruber J, Buhas D, Majewski J, Sasarman F, Papillon-Cavanagh S, Thiffault I, Sheldon KM, Massicotte C, Patry L, Simon M, Zare AS, McKernan KJ., FORGE Canada Consortium. Michaud J, Boles RG, Deal CL, Desilets V, Shoubridge EA, Samuels ME. Mutation in the nuclear-encoded mitochondrial isoleucyl-tRNA synthetase IARS2 in patients with cataracts, growth hormone deficiency with short stature, partial sensorineural deafness, and peripheral neuropathy or with Leigh syndrome. Hum Mutat. 2014;35:1285–9. [PubMed: 25130867]

  • Shamseldin HE, Alshammari M, Al-Sheddi T, Salih MA, Alkhalidi H, Kentab A, Repetto GM, Hashem M, Alkuraya FS. Genomic analysis of mitochondrial diseases in a consanguineous population reveals novel candidate disease genes. J Med Genet. 2012;49:234–41. [PubMed: 22499341]

  • Shear T, Tobias JD. Anesthetic implications of Leigh's syndrome. Paediatr Anaesth. 2004;14:792–7. [PubMed: 15330965]

  • Simon M, Richard EM, Wang X, Shahzad M, Huang VH, Qaiser TA, Potluri P, Mahl SE, Davila A, Nazli S, Hancock S, Yu M, Gargus J, Chang R, Al-Sheqaih N, Newman WG, Abdenur J, Starr A, Hegde R, Dorn T, Busch A, Park E, Wu J, Schwenzer H, Flierl A, Florentz C, Sissler M, Khan SN, Li R, Guan MX, Friedman TB, Wu DK, Procaccio V, Riazuddin S, Wallace DC, Ahmed ZM, Huang T, Riazuddin S. Mutations of human NARS2, encoding the mitochondrial asparaginyl-tRNA synthetase, cause nonsyndromic deafness and Leigh syndrome. PLoS Genet. 2015;11:e1005097. [PMC free article: PMC4373692] [PubMed: 25807530]

  • Smith AC, Ito Y, Ahmed A, Schwartzentruber JA, Beaulieu CL, Aberg E, Majewski J, Bulman DE, Horsting-Wethly K, Koning DV, Rodenburg RJ, Boycott KM, Penney LS. A family segregating lethal neonatal coenzyme Q(10) deficiency caused by mutations in COQ9. J Inher Metab Dis. 2018;41:719–29. [PubMed: 29560582]

  • Soreze Y, Boutron A, Habarou F, Barnerias C, Nonnenmacher L, Delpech H, Mamoune A, Chrétien D, Hubert L, Bole-Feysot C, Nitschke P, Correia I, Sardet C, Boddaert N, Hamel Y, Delahodde A, Ottolenghi C, de Lonlay P. Mutations in human lipoyltransferase gene LIPT1 cause a Leigh disease with secondary deficiency for pyruvate and alpha-ketoglutarate dehydrogenase. Orphanet J Rare Dis. 2013;8:192. [PMC free article: PMC3905285] [PubMed: 24341803]

  • Spiegel R, Shaag A, Edvardson S, Mandel H, Stepensky P, Shalev SA, Horovitz Y, Pines O, Elpeleg O. SLC25A19 mutation as a cause of neuropathy and bilateral striatal necrosis. Ann Neurol. 2009;66:419–24. [PubMed: 19798730]

  • Sugiana C, Pagliarini DJ, McKenzie M, Kirby DM, Salemi R, Abu-Amero KK, Dahl HH, Hutchison WM, Vascotto KA, Smith SM, Newbold RF, Christodoulou J, Calvo S, Mootha VK, Ryan MT, Thorburn DR. Mutation of C20orf7 disrupts complex I assembly and causes lethal neonatal mitochondrial disease. Am J Hum Genet. 2008;83:468–78. [PMC free article: PMC2561934] [PubMed: 18940309]

  • Taanman JW, Rahman S, Pagnamenta AT, Morris AA, Bitner-Glindzicz M, Wolf NI, Leonard JV, Clayton PT, Schapira AH. Analysis of mutant DNA polymerase gamma in patients with mitochondrial DNA depletion. Hum Mutat. 2009;30:248–54. [PubMed: 18828154]

  • Taylor RW, Pyle A, Griffin H, Blakely EL, Duff J, He L, Smertenko T, Alston CL, Neeve VC, Best A, Yarham JW, Kirschner J, Schara U, Talim B, Topaloglu H, Baric I, Holinski-Feder E, Abicht A, Czermin B, Kleinle S, Morris AA, Vassallo G, Gorman GS, Ramesh V, Turnbull DM, Santibanez-Koref M, McFarland R, Horvath R, Chinnery PF. Use of whole-exome sequencing to determine the genetic basis of multiple mitochondrial respiratory chain complex deficiencies. JAMA. 2014;312:68–77. [PMC free article: PMC6558267] [PubMed: 25058219]

  • Tort F, Ferrer-Cortès X, Thió M, Navarro-Sastre A, Matalonga L, Quintana E, Bujan N, Arias A, García-Villoria J, Acquaviva C, Vianey-Saban C, Artuch R, García-Cazorla À, Briones P, Ribes A. Mutations in the lipoyltransferase LIPT1 gene cause a fatal disease associated with a specific lipoylation defect of the 2-ketoacid dehydrogenase complexes. Hum Mol Genet. 2014;23:1907–15. [PubMed: 24256811]

  • Triepels RH, van den Heuvel LP, Loeffen JL, Buskens CA, Smeets RJ, Rubio Gozalbo ME, Budde SM, Mariman EC, Wijburg FA, Barth PG, Trijbels JM, Smeitink JA. Leigh syndrome associated with a mutation in the NDUFS7 (PSST) nuclear encoded subunit of complex I. Ann Neurol. 1999;45:787–90. [PubMed: 10360771]

  • Tucker EJ, Hershman SG, Köhrer C, Belcher-Timme CA, Patel J, Goldberger OA, Christodoulou J, Silberstein JM, McKenzie M, Ryan MT, Compton AG, Jaffe JD, Carr SA, Calvo SE. RajBhandary UL, Thorburn DR, Mootha VK. Mutations in MTFMT underlie a human disorder of formylation causing impaired mitochondrial translation. Cell Metab. 2011;14:428–34. [PMC free article: PMC3486727] [PubMed: 21907147]

  • Valente L, Tiranti V, Marsano RM, Malfatti E, Fernandez-Vizarra E, Donnini C, Mereghetti P, De Gioia L, Burlina A, Castellan C, Comi GP, Savasta S, Ferrero I, Zeviani M. Infantile encephalopathy and defective mitochondrial DNA translation in patients with mutations of mitochondrial elongation factors EFG1 and EFTu. Am J Hum Genet. 2007;80:44–58. [PMC free article: PMC1785320] [PubMed: 17160893]

  • van den Bosch BJ, Gerards M, Sluiter W, Stegmann AP, Jongen EL, Hellebrekers DM, Oegema R, Lambrichs EH, Prokisch H, Danhauser K, Schoonderwoerd K, de Coo IF, Smeets HJ. Defective NDUFA9 as a novel cause of neonatally fatal complex I disease. J Med Genet. 2012;49:10–5. [PubMed: 22114105]

  • Van Hove JL, Saenz MS, Thomas JA, Gallagher RC, Lovell MA, Fenton LZ, Shanske S, Myers SM, Wanders RJ, Ruiter J, Turkenburg M, Waterham HR. Succinyl-CoA ligase deficiency: a mitochondrial hepatoencephalomyopathy. Pediatr Res. 2010;68:159–64. [PMC free article: PMC2928220] [PubMed: 20453710]

  • Vedrenne V, Gowher A, de Lonlay P, Nitschke P, Serre V, Boddaert N, Altuzarra C, Mager-Heckel AM, Chretien F, Entelis N, Munnich A, Tarassov I, Rötig A. Mutation in PNPT1, which encodes a polyribonucleotide nucleotidyltransferase, impairs RNA import into mitochondria and causes respiratory-chain deficiency. Am J Hum Genet. 2012;91:912–8. [PMC free article: PMC3487136] [PubMed: 23084291]

  • Wedatilake Y, Brown RM, McFarland R, Yaplito-Lee J, Morris AA, Champion M, Jardine PE, Clarke A, Thorburn DR, Taylor RW, Land JM, Forrest K, Dobbie A, Simmons L, Aasheim ET, Ketteridge D, Hanrahan D, Chakrapani A, Brown GK, Rahman S. SURF1 deficiency: a multi-centre natural history study. Orphanet J Rare Dis. 2013;8:96. [PMC free article: PMC3706230] [PubMed: 23829769]

  • Weraarpachai W, Antonicka H, Sasarman F, Seeger J, Schrank B, Kolesar JE, Lochmüller H, Chevrette M, Kaufman BA, Horvath R, Shoubridge EA. Mutation in TACO1, encoding a translational activator of COX I, results in cytochrome c oxidase deficiency and late-onset Leigh syndrome. Nat Genet. 2009;41:833–7. [PubMed: 19503089]

  • Wortmann SB, Vaz FM, Gardeitchik T, Vissers LE, Renkema GH, Schuurs-Hoeijmakers JH, Kulik W, Lammens M, Christin C, Kluijtmans LA, Rodenburg RJ, Nijtmans LG, Grünewald A, Klein C, Gerhold JM, Kozicz T, van Hasselt PM, Harakalova M, Kloosterman W, Barić I, Pronicka E, Ucar SK, Naess K, Singhal KK, Krumina Z, Gilissen C, van Bokhoven H, Veltman JA, Smeitink JA, Lefeber DJ, Spelbrink JN, Wevers RA, Morava E, de Brouwer AP. Mutations in the phospholipid remodeling gene SERAC1 impair mitochondrial function and intracellular cholesterol trafficking and cause dystonia and deafness. Nat Genet. 2012;44:797–802. [PubMed: 22683713]

  • Zaha K, Matsumoto H, Itoh M, Saitsu H, Kato K, Kato M, Ogata S, Murayama K, Kishita Y, Mizuno Y, Kohda M, Nishino I, Ohtake A, Okazaki Y, Matsumoto N, Nonoyama S. DNM1L-related encephalopathy in infancy with Leigh syndrome-like phenotype and suppression-burst. Clin Genet. 2016;90:472–4. [PubMed: 27301544]