Abstract
Purpose of Review
Primary Mitochondrial Disease (PMD) are a genetically and phenotypically diverse group of inherited energy deficiency disorders caused by impaired mitochondrial oxidative phosphorylation (OXPHOS) capacity. Mutations in more than 350 genes in both mitochondrial and nuclear genomes are now recognized to cause primary mitochondrial disease following every inheritance pattern. Next-generation sequencing technologies have dramatically accelerated mitochondrial disease gene discovery and diagnostic yield. Here, we provide an up-to-date review of recently-identified, novel mitochondrial disease genes and/or pathogenic variants that directly impair mitochondrial structure, dynamics, and/or function.
Recent Findings
A review of PubMed publications was performed from the past 12 months that identified 16 new PMD genes and/or pathogenic variants, as well as expanded phenotype recognition for a wide variety of mitochondrial disease genes.
Summary
Broad-based exome sequencing has become the standard first-line diagnostic approach for PMD. This has facilitated more rapid and accurate disease identification, and greatly expanded understanding of the wide spectrum of distinct clinical phenotypes. A comprehensive dual-genome sequencing approach to PMD diagnosis continues to improve diagnostic yield, advance understanding of mitochondrial physiology, and provide strong potential to develop precision therapeutics targeted to diverse aspects of mitochondrial disease pathophysiology.
Keywords: Primary mitochondrial disease, nuclear genome, mitochondrial genome, phenotype expansion, novel gene discovery
INTRODUCTION
Mitochondria are ubiquitous intracellular organelles in which oxidative metabolism occurs to generate the majority of cellular energy in the chemical form of adenosine triphosphate (ATP)(1). Approximately 1,500 proteins exist within mitochondria, with origins from two distinct cellular genomes. The vast majority of mitochondrial proteins are encoded by the nuclear DNA (nDNA) genome, while only 13 mitochondrial proteins that are all core structural subunits of complexes I, III, IV and V in the oxidative phosphorylation (OXPHOS) pathway are encoded in the mitochondrial genome (mtDNA) (2). Primary mitochondrial disease (PMD) is a highly heterogeneous collection of inherited genetic-based disorders that share in common disrupted energy metabolism due to impaired oxidative phosphorylation (OXPHOS) capacity. The broad and highly variable but commonly progressive phenotypic spectrum ranges from adult-onset, isolated organ system involvement to infantile-onset, multi-systemic, lethal disease (2). Systemic involvement that may affect nearly every organ has been described, leading to immense variation of clinical phenotype that can pose a diagnostic challenge (3). In a recent survey of self-reported PMD individuals (n=270), both pediatric and adult PMD subjects reported a mean of 16 clinical symptoms, highlighting the substantial burden of this disease (4). While PMDs are individually rare, they have a collective minimal prevalence of 1 in 4,300 (5). To date, pathogenic variants in more than 350 genes have been associated with PMD (6, 7).
Since the early characterization of cellular energy production within mitochondria in the 1950s and the discovery of pathogenic mitochondrial DNA (mtDNA) variants only three decades ago (8–10), knowledge of the diverse molecular etiologies of PMD and deleterious effects of pathogenic variants has dramatically increased. This, in turn, has led to the elucidation of the underlying gene defects in individual patients with increasing potential to develop specific therapeutic strategies (2). Further, recent approval in the United Kingdom of novel reproductive technologies such as mitochondrial replacement technologies (MRT) holds potential to prevent the transmission of maternally inherited mtDNA disorders in families with known pathogenic mutations (11). Here, we summarize recent advances in PMD molecular etiologies, diagnostic approach, and emerging treatment approaches.
MOLECULAR ETIOLOGIES OF PRIMARY MITOCHONDRIAL DISEASE
Since the pioneering discoveries of pathogenic variants in mtDNA in 1988 (8, 9) and pathogenic variants in nuclear genes encoding proteins necessary for mitochondrial function in 2000–2001(12–14), pathogenic variants in more than 350 genes across both genomes have been recognized to cause PMD (6, 7, 10).
Mitochondrial DNA
The mitochondrial genome is comprised of 16,569 basepairs that encodes 37 genes, including 13 proteins, 22 transfer RNAs (tRNA), and 2 ribosomal RNAs (rRNA). mtDNA is exclusively maternally inherited through the oocyte, and replicates independently of the cell cycle. There are multiple copies of mtDNA in each mitochondrion, and multiple mitochondria per cell. mtDNA genome variants may exist in a state of homoplasmy, meaning in all mitochondrial genomes within a mitochondrion or cell, or in a state of heteroplasmy, meaning the mutation is only present in a portion of the mitochondrial genomes. The portion of mitochondrial genomes with a specific variant may differ greatly between a given individual’s tissues and among members of the same family. In general, the proportion of mitochondrial genomes that harbor a pathogenic variant directly correlates with disease severity (1). Many healthy individuals have low levels of pathogenic variants accumulate in some tissues with age, although if a pathogenic variant reaches a certain threshold heteroplasmy level that may vary greatly by tissue energy demand, phenotypic symptoms may develop over time.
While the basic concepts of heteroplasmy and threshold effect hold true for many pathogenic mtDNA variants, the correlation of precise heteroplasmy levels among various tissues with disease onset and tissue-specific manifestations remains poorly understood. Grady et al, 2018, recently characterized heteroplasmy levels in several tissues (blood, n=231; urine, n=235; skeletal muscle, n=77) over time in a cohort of individuals with the well-characterized pathogenic mtDNA variant m.3243A>G that may cause a range of clinical syndromes from Mitochondrial Encephalomyopathy Lactic Acidosis and Stroke-Like Episodes (MELAS) to Leigh Syndrome or Maternally Inherited Diabetes and Deafness (MIDD) (15). Many of the m.3243A>G carriers were clinically affected, with assessment made to correlate heteroplasmy level, disease burden, and disease progression (15). Results showed greatest correlation between heteroplasmy levels in blood and urine > muscle and blood > muscle and urine. Muscle heteroplasmy levels did not correlate with age, whereas both blood and urine heteroplasmy levels declined with age. Overall, blood heteroplasmy levels decreased at a rate of ~2.3% per year when adjusted for age. Males had 19.2% higher urine heteroplasmy levels than females. Urine heteroplasmy levels showed the greatest variability, up to 55%, compared to 15% in blood. Age-adjusted blood heteroplasmy levels showed greatest correlation with disease burden and progression. Muscle mtDNA copy number was associated with a decreased disease burden, providing valuable information about the role of mitochondrial biogenesis in m.3243A>G clinical phenotype penetrance.
Given that individuals who carry low heteroplasmy levels of pathogenic mtDNA variants may be asymptomatic, heteroplasmy shift therapy is a potential therapeutic strategy for PMD, with the goal to decrease the heteroplasmy level of a given pathogenic mtDNA mutation in affected individuals. Different approaches have been considered in the research setting, including use of mitoTALENS (mitochondrial targeted transcription activator-like effector nucleases) that utilize engineered nucleases to selectively degrade specific target mtDNA genomes(16, 17). Yang et al., 2018, showed that m.3243A>G pathogenic variant heteroplasmy levels in induced pluripotent stem cells (iPSCs) could be eliminated by mitoTALENs, with ‘rescued’ cells having normal energy production (18). When tested in porcine oocytes, mutant heteroplasmy levels were similarly decreased. Yahata et al., 2017, similarly showed that mitoTALENs could be used to decrease mutation heteroplasmy levels in iPSCs generated from individuals with the known pathogenic complex I ND5 subunit m.13513G>A mutation (19).
As mtDNA is maternally inherited through the oocyte, affected individuals with mtDNA-related disease may either inherit the variant from their mother or it may occur de novo in the oocyte or embryo that forms the affected individual. Whether the mother of an affected individual is symptomatic depends on whether she carries the same mutation in her somatic cells and if so, at what level of heteroplasmy(20). Until recently, the only reliable reproductive options for women with mtDNA pathogenic variants to assure their offspring did not inherit a disease-causing mtDNA mutation were either adoption or utilizing an egg donor, with prenatal heteroplasmy testing largely considered unreliable. Vachin et al, 2018, sought to evaluate how informative placental heteroplasmy level testing was when compared to testing heteroplasmy levels in chorionic villi, amniotic fluid, cord blood, and fetal tissues (21). Enrolling individuals who carried several different pathogenic mtDNA variants, heteroplasmy levels were similar among all tissue samples tested when pathogenic variants were present at the extremes, either above 80% or below 20%. When heteroplasmy levels fell in an intermediate range, much greater variability between tissue heteroplasmy levels was seen. Significantly more variance was also seen in tissues obtained from full-term fetuses as compared to tissues obtained from pregnancies terminated between 12–18 weeks’ gestation.
Preimplantation genetic diagnosis, or PGD, of embryos in the setting of in vitro fertilization (IVF) for mtDNA mutations has been a challenge due to both biological and technologic considerations (22). This option has been further refined in the past year, as Sallevelt et al., 2017 showed that assessing mtDNA heteroplasmy level in a single blastomere from an embryo carries a low diagnostic error rate and has improved outcomes on live birth delivery as compared to assessing two blastomeres (23).
MRT involves transplantation of the nuclear genome from the egg of an affected woman to an enucleated egg from an unaffected donor and has now been approved in the United Kingdom on a case-by-case basis (11, 24), however governmental and legal hurdles remain in place in many countries including the United States (25).
Nuclear DNA
Primary mitochondrial disease is commonly caused by pathogenic variants in nuclear genes, which can occur in any Mendelian pattern including autosomal dominant, autosomal recessive, or X-linked inheritance. Reproductive options for individuals with pathogenic variant(s) in a nuclear DNA gene exist to the same extent as for other Mendelian conditions. Nuclear DNA pathogenic variants are most commonly inherited in an autosomal recessive manner, particularly for childhood-onset diseases. mtDNA pathogenic variants are more commonly causative of adult-onset primary mitochondrial disease. However, improved diagnostic testing has demonstrated that mutations in both genomes can variably cause pediatric and adult onset disease.
Nuclear DNA genes in which mutations cause PMD encode proteins that have an array of roles within mitochondria, including as assembly factors or structural subunits of the electron transport chain (ETC) complexes; mtDNA maintenance; mtDNA replication, transcription, or translation; mitochondrial import and export; mitochondrial dynamics; and others.
NOVEL GENES AND EXPANDING PHENOTYPES
The advent of molecular technologies has accelerated the identification of PMD genes. Over the last 12 months alone, several novel PMD genes and/or pathogenic variant(s) have been discovered (Table 1). In addition, the delineation of previously well-recognized clinical syndromes has expanded in light of improved genomic diagnosis (Table 2). Several excellent PMD review articles have been written in the past year (Table 3).
Table 1:
Component | Gene | Product | Phenotype | Relevance | Inheritance | Reference |
---|---|---|---|---|---|---|
Electron transport chain complex subunits | COX4I1 | Complex IV subunit | Suspected Fanconi anemia (failure to thrive, short stature, macrocytosis, increased chromosome breakage) | One of only several nuclear-encoded complex IV subunits associated with disease | AR | (26) |
USMG5 | Complex V subunit | Leigh syndrome | Identified Ashkenazi Jewish founder mutation with allele frequency of 0.57%. | AR | (27) | |
Iron sulfur cluster biogenesis | FDXR | Involved in iron-sulfur cluster biosynthesis and initiating electron transport chain (ETC) function by transferring electrons to mitochondrial cytochrome P450 from NADPH |
Optic atrophy, neuropathy, hearing loss, ataxia | Expands understanding of iron sulfter cluster-related mitochondrial diseases | AR | (28, 29) |
mtDNA translation | WARS2 | Tryptophanyl-tRNA synthetase | Ranges from neonatal lactic acidosis to global developmental delay and intellectual disability, optic atrophy, and ataxia | Last mitochondrial aminoacyl tRNA synthetase to be associated with disease; identified severity range of phenotype, and lack of phenotype specificity | AR | (30, 31) |
MRPS34 | A small mitoribosome subunit | Leigh syndrome | Additional insight into small mitoribosome subunit stability | AR | (32) | |
MRPS2 | A small mitoribosome subunit | Sensorineural hearing loss, developmental delay, hypoglycemia, lactic acidemia | Increased understanding of mtDNA translation defects | AR | (33) | |
m.15927G>A (tRNA-Thr) | Mitochondrial tRNA for threonine | Leber hereditary optic neuropathy (LHON) | Identified first mitochondrial tRNA variant associated with LHON | Maternal | (34) | |
C1QBP | Suspected to be involved in mitochondrial protein synthesis and mtDNA maintenance | Ranges from from infantile lactic acidosis and cardiomyopathy to adult onset progressive external ophthalmoplegia (PEO) and neuropathy | Associated with cardiomyopathy independent of age of onset, lack of central nervous system involvement | AR | (35) | |
MRM2 | 2-O-ribose methyl-transferase in mitochondrial 16S rRNA, also functions in assembly and stability of large mitoribosome | MELAS-like syndrome | First report of defective mitochondrial ribosomal RNA nucleotide modification causing ETC deficiencies | AR | (36) | |
Mitochondrial membrane function and import | MIEF2 | Regulates outer mitochondrial membrane fission | Proximal muscle weakness, exercise intolerance, elevated creatine kinase (CK), ragged red fibers and COX negative fibers on muscle biopsy | First mitochondrial fission/fusion defect resulting in myopathy | AR | (37) |
MSTO1 | Regulates mitochondrial fusion |
Myopathy, mood disorder | Localized to cytoplasm and interacts with mitochondrial fusion machinery | AD | (38, 39) | |
SLC25A21 | Mitochondrial inner membrane oxodicarboxylate carrier | Spinal muscular atrophy-like features, myopathy | Identified novel mitochondrial carrier defect leading to targeted therapy | AR | (40) | |
SLC25A24 | Mitochondrial inner membrane ATP-Mg/Pi carrier | Craniosynostosis, progeroid appearance, microphthalmia, increased sensitivity to oxidative stress | Increased sensitivity to oxidative stress leading to progeroid-like phenotype | AD | (41) | |
MICU2 | Mitochondrial calcium uniporter subunit | Intellectual disability, white matter involvement | Second gene encoding a subunit of the mitochondrial calcium uniporter, with distinct phenotype from first MCU subunit gene reported (MICU1) | AR | (42) | |
Mitochondrial proteostasis | PITRM1 | Mitochondrial peptidase | Progressive spinocerebellar ataxia, cerebellar atrophy, intellectual disability, psychotic episodes | Expanded understanding of mitochondrial proteostasis defects | AR | (43) |
Nuclear-encoded mitochondrial protein processing | PMPCB | Mitochondrial processing protease (MPP) catalytic subunit | Leigh-like syndrome, developmental regression, epilepsy | Expanded understanding of MPP defects | AR | (44) |
Key: AR = autosomal recessive, AD = autosomal dominant
Table 2:
Component | Gene | Product/Function | Relevance | Reference |
---|---|---|---|---|
Electron transport chain enzyme subunits and assembly factors | NDUFA2 | Complex I accessory subunit | Second and third reports of pathogenic variants in NDUFA2; first report of leukoencephalopathy with NDUFA2 pathogenic variants | (45) |
NDUFA9 | Complex I structural accessory subunit | Milder phenotype and correlation with effects on complex I assembly | (46) | |
m.13094T>C (mt-ND5) | Complex I subunit | Summary of 20 affected and 4 asymptomatic individuals; range of heteroplasmy levels including undetectable in certain tissues even in those severely affected | (47) | |
NDUFAF4 | Complex I assembly factor | Report of second family with NDUFAF4 pathogenic variant, expands phenotype to include Leigh syndrome, expands understanding of complex I assembly | (48) | |
SCO2 | Cytochrome c oxidase (Complex IV) assembly protein | First report of milder phenotype with neuropathy without cardiomyopathy | (49) | |
m.8969G>A (mt-ATP6) | Complex V subunit | First report of stable phenotype with intellectual disability and short stature | (50) | |
LYRM7 | Chaperone protein for Rieske iron-sulfur protein important for Complex III assembly | Three additional probands reported (total now 11) | (51) | |
Iron sulfur cluster assembly | ISCA1 | Iron-sulfur cluster (ISC) assembly machinery | Third proband reported, milder phenotype; further characterization of disease mechanism | (52) |
ISCA2 | Iron-sulfur cluster (ISC) assembly machinery | Additional case reports, biochemical characterization, mitochondrial dysfunction characterization | (53–55) | |
mtDNA translation | FARS2 | Phenylalanyl-tRNA synthetase | Review of cases reported to date, delineation of phenotypic spectrum (epileptic vs spastic paraplegia) | (56) |
GARS | Glycyl-tRNA synthetase | AR mode of inheritance, most prior cases were AD; implication of mitochondrial GARS rather than cytosolic GARS in prior case reports; loss of function in AR cases and gain of function in AD cases | (57) | |
Mitochondrial membrane function and import | DNM1L | Mitochondrial and peroxisomal fission | First report of milder phenotype (optic atrophy) and delineation of genotype-phenotype correlation | (58) |
MICU1 | Mitochondrial calcium uniporter subunit | Review and summary of additional 13 cases, all reported cases with elevated CK and elevated liver transaminases, increased appreciation of intellectual disability and learning disability as phenotypic feature | (59) | |
Cofactor import | SLC25A42 | Imports Coenzyme A into mitochondrial matrix | Thirteen additional probands (total 14) reported, further describing phenotypic spectrum (lactic acidosis to epilepsy and severe movement disorder) | (60, 61) |
Other | AIFM1 | NADH oxidoreductase, regulates apoptosis | Expands genotype-phenotype correlation; showed response to riboflavin in those with ataxia | (62, 63) |
FBXL4 | mtDNA maintenance | Summary of all 87 affected individuals and pathogenic variants reported to date | (64) | |
PNPLA8 | Regulates oxidative stress | Second and third reports of pathogenic variants in PNPLA8 providing further evidence of progressive neurodegenerative phenotype | (65) | |
SERAC1 | Phosphatidylglycerol remodeling essential for mitochondrial function and intracellular cholesterol trafficking | Summary of 67 affected individuals with 41 different pathogenic variants | (66) | |
TK2 | Maintains mitochondrial deoxynucleotide pool | Summary of all 92 affected individuals reported to date | (67) |
Table 3:
Topic | Highlights | Reference |
---|---|---|
mtDNA mechanisms | ||
mtDNA replication | Summary of mtDNA replication process | (68) |
mtDNA replication defects | Overview of mtDNA maintenance and disorders arising from impaired mtDNA maintenance | (69) |
mtDNA transcription and translation defects | Overview of genes important for mtDNA transcription and translation with pathogenic variants reported, organized by place in transcription or translation process in which gene product is involved, provides overview figure and summary tables organized by genetic mutations | (70) |
mtDNA bottleneck | Summary of the mitochondrial genome bottleneck hypothesis | (1) |
Biomarkers | ||
Biomarkers for primary mitochondrial disease | Overview of currently utilized biomarkers and summary of novel approaches | (71) |
Natural History of PMD | ||
Review of PMD natural history studies | Summary of PMD natural history studies highlighting trends in phenotype, biochemical markers, genotype-phenotype correlation, and prognosis | (3) |
Genomic Diagnosis | ||
Genomic diagnosis of PMD | Summarizes state of PMD diagnosis, proposes genomic sequencing as the first step | (72) |
Approaches to diagnosis of PMD | Summary of different approaches to genomic sequencing for diagnosis of PMD and utility of databases and “–omics” to aid in variant interpretation | (7) |
Treatment | ||
PMD treatment | Summary of existing therapies (mitochondrial supplemental medicines, exercise) and emerging therapies (both specific to the gene defect and universal) | (73) |
Recent advances in understanding of mitochondrial pathophysiology and therapies | Overview of mitochondrial function, genomics, transcriptomics, proteomics, metabolomics, mitochondrial disease pathogenesis, and therapies | (6) |
Genomic-based therapies for mtDNA-related PMD | Summary of technology for prevention of mtDNA transmission, mtDNA heteroplasmy shifting, and CRISPR | (74) |
DIAGNOSIS OF PRIMARY MITOCHONDRIAL DISEASE
While summarizing the diagnostic approach to PMD is outside the scope of this review, tremendous improvements in the diagnostic approach have been achieved in the molecular era of PMD(20, 75). Due to the challenges presented by significant clinical heterogeneity and lack of reliable biomarkers for all possible PMD, the genomic approach now tends to be the first-line investigation coupled to a specialist clinical assessment and biochemical screen. The genomic approach has several additional advantages, allowing the genetic diagnosis of other rare treatable diseases, more than one genetic disorder in a given patient, mitochondrial disease phenocopies and, even more importantly, ensuring that mitochondrial disorders are diagnosed when they may have been overlooked clinically (72).
Rapidly reducing costs have facilitated more wide-spread implementation of whole exome sequencing (WES) and whole genome sequencing (WGS) in large cohorts of PMD patients. The increasing application of next generation sequencing (NGS) in extended gene panels, WES, and WGS of both nuclear and mtDNA genomes to routine mitochondrial disease diagnostics has led to a dramatic increase in the PMD diagnostic yield (6). More invasive diagnostic tests such as the muscle biopsy are increasingly reserved for cases that cannot be solved by a first-line genomic approach in a more readily accessible tissue. Despite these substantial technologic advances, the diagnostic odyssey of patients with mitochondrial disease is often complex and burdensome. Grier et al., 2018 recently described the diagnostic odyssey in patients with self-reported mitochondrial disease and found that, on average, individuals saw eight different physicians and underwent many tests before receiving a diagnosis of PMD(76). More than half of the individuals surveyed received an incorrect clinical diagnosis prior to receiving their PMD diagnosis.
TOOLS AND RESOURCES FOR ASSESSING GENOMIC VARIANTS RELATED TO PRIMARY MITOCHONDRIAL DISEASE
With improving genomic sequencing technologies and increased global utilization of massively parallel diagnostic tests, the need for robust bioinformatics tools and data resources to accurately curate novel disease genes and variants for PMD has been well-recognized.
The Mitochondrial Disease Sequence Data Resource, MSeqDR, is an online, centralized Web portal that organizes both nuclear and mtDNA variants in all known and candidate genes for PMD(77–79). Available through this resource are tools both to provide comprehensive information on variants and genes, as well as tools to assist clinicians and researchers in efficiently mining genomic data. MSeqDR has partnered with the Clinical Genome Resource, ClinGen, to form a mitochondrial disease expert panel to enable expert curation of variants and novel disease genes associated with PMD(80). MSeqDR also now provides mtDNA variant manually curated assertions from MITOMAP, the gold-standard resource for mtDNA variant curation(81). In addition, MSeqDR and MITOMAP incorporate data from an informatics tool recently developed to predict effects of novel mitochondrial tRNA variants (mitoTIP)(82), as well as HmtDB and HmtVAR, databases of mtDNA sequences, variants, and their predicted effects(83). MSeqDR custom tools are available to support PMD gene and variant submission by users or expert panels that are linked to ClinVar, as well as MSeqDR mvTool(77), which is a recently launched Web and API resource for comprehensive variant annotation, universal nomenclature collation, and reference genome conversion for mtDNA.
Lastly, there exists two tools that are regularly updated for variants in a common nuclear gene cause of PMD with a wide variety of phenotypes, POLG(84). POLG variants are expertly catalogued in the Human DNA Polymerase Gamma Mutation Database, https://tools.niehs.nih.gov/polg/. In addition, given recent recognition that there are distinct biochemical clusters within the POLG protein, variants falling within these clusters can have characteristic phenotypic effects that can be evaluated in an easily accessible online tool, the POLG variant server (85).
CLINICAL TRIALS FOR PRIMARY MITOCHONDRIAL DISEASES
While there are no cures or FDA approved drugs for PMD, several therapeutic PMD strategies are now being evaluated in clinical trials. The lack of validated biomarkers and outcome measures of PMD disease progression, limited natural history data, and inherent variable and fluctuations in PMD course impose intrinsic challenges to conducting PMD clinical trials. Several clinical trials are underway for treating PMD(75). In effort to improve trial enrollment in the mitochondrial disease patient community, a recent publication reported survey results of affected individuals and their family members performed to ascertain PMD patient motivations and barriers to clinical trial participation(4). Both adults and children with PMD prioritized weakness, exercise intolerance, fatigue, imbalance, and gastrointestinal involvement, along with developmental delay in children, as their predominant concerns they would most like to be evaluated in clinical trials.
CONCLUSION
Since the first discovery of a molecular etiology for primary mitochondrial disease 30 years ago (86), the field of mitochondrial genetics has advanced rapidly in the molecular era with at least 20 PMD genes discovered each year for the past decade(87). Genomic diagnosis has become the frontline testing modality, enabling increasingly broader spectrum of clinical phenotypes to be identified including atypical and less severe presentations. Nevertheless, many individuals with features highly concerning for PMD remain undiagnosed, highlighting that gaps still remain in our knowledge of the genetic basis of mitochondrial disease. Identifying the genetic basis of disease in each patient is particularly important, as the development of effective, precision therapies requires improved understanding of underlying PMD etiology and mitochondrial pathophysiology.
KEY POINTS:
Primary mitochondrial disease is a heterogeneous group of energy deficiency disorders caused by pathogenic variants in nuclear or mitochondrial DNA, with more than 20 new genes discovered each year for the past decade.
Bioinformatic tools and data resources for mitochondrial disease gene and variant curation are increasingly available and user-friendly, curated through a central portal at https://mseqdr.org.
Precision therapies are emerging based on improved understanding of mitochondrial disease etiology and pathophysiology in PMD individuals.
Acknowledgments
FINANCIAL SUPPORT AND SPONSORSHIP
This work was funded in part by the National Institutes of Health (U24-HD093483; U41-HG006834; T32-GM008638). MSeqDR is funded in part by ongoing support from the United Mitochondrial Disease Foundation (UMDF) and North American Mitochondrial Disease Consortium (NAMDC, funded in part through U54-NS078059). The content is solely the responsibility of the authors and does not necessarily represent the official views of the funding agencies.
Footnotes
CONFLICTS OF INTEREST
The authors have no relevant conflicts of interest to declare.
REFERENCES
- 1.Zhang H, Burr SP, Chinnery PF. The mitochondrial DNA genetic bottleneck: inheritance and beyond. Essays Biochem 2018. [DOI] [PubMed]
- 2.Gorman GS, Chinnery PF, DiMauro S, Hirano M, Koga Y, McFarland R, et al. Mitochondrial diseases. Nat Rev Dis Primers 2016;2:16080.** This review describes advances in next-generation sequencing techniques leading to substantially improved diagnosis and advances in in vitro fertilization techniques, including mitochondrial donation.
- 3.Keshavan N, Rahman S. Natural history of mitochondrial disorders: a systematic review. Essays Biochem 2018. [DOI] [PubMed]
- 4.Zolkipli-Cunningham Z, Xiao R, Stoddart A, McCormick EM, Holberts A, Burrill N, et al. Mitochondrial disease patient motivations and barriers to participate in clinical trials. PLoS One 2018;13(5):e0197513.* This study conveyed clear PMD subject preferences and priorities to enable improved clinical treatment trial design, and that PMD subjects report a mean of 16 clinical symptoms.
- 5.Gorman GS, Schaefer AM, Ng Y, Gomez N, Blakely EL, Alston CL, et al. Prevalence of nuclear and mitochondrial DNA mutations related to adult mitochondrial disease. Ann Neurol 2015;77(5):753–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Rahman J, Rahman S. Mitochondrial medicine in the omics era. Lancet 2018;391(10139):2560–74.** This review discusses recent advances in mitochondrial biology and medicine arising from widespread use of high-throughput omics technologies, and a broad discussion of emerging therapies for mitochondrial disease.
- 7.Stenton SL, Prokisch H. Advancing genomic approaches to the molecular diagnosis of mitochondrial disease. Essays Biochem 2018. [DOI] [PubMed]
- 8.Holt IJ, Harding AE, Morgan-Hughes JA. Deletions of muscle mitochondrial DNA in patients with mitochondrial myopathies. Nature 1988;331(6158):717–9. [DOI] [PubMed] [Google Scholar]
- 9.Wallace DC, Singh G, Lott MT, Hodge JA, Schurr TG, Lezza AM, et al. Mitochondrial DNA mutation associated with Leber’s hereditary optic neuropathy. Science 1988;242(4884):1427–30. [DOI] [PubMed] [Google Scholar]
- 10.Dimauro S A history of mitochondrial diseases. J Inherit Metab Dis 2011;34(2):261–76. [DOI] [PubMed] [Google Scholar]
- 11.Herbert M, Turnbull D. Progress in mitochondrial replacement therapies. Nat Rev Mol Cell Biol 2018;19(2):71–2. [DOI] [PubMed] [Google Scholar]
- 12.Kaukonen J, Juselius JK, Tiranti V, Kyttala A, Zeviani M, Comi GP, et al. Role of adenine nucleotide translocator 1 in mtDNA maintenance. Science 2000;289(5480):782–5. [DOI] [PubMed] [Google Scholar]
- 13.Spelbrink JN, Li FY, Tiranti V, Nikali K, Yuan QP, Tariq M, et al. Human mitochondrial DNA deletions associated with mutations in the gene encoding Twinkle, a phage T7 gene 4-like protein localized in mitochondria. Nat Genet 2001;28(3):223–31. [DOI] [PubMed] [Google Scholar]
- 14.Van Goethem G, Dermaut B, Lofgren A, Martin JJ, Van Broeckhoven C. Mutation of POLG is associated with progressive external ophthalmoplegia characterized by mtDNA deletions. Nat Genet 2001;28(3):211–2. [DOI] [PubMed] [Google Scholar]
- 15.Grady JP, Pickett SJ, Ng YS, Alston CL, Blakely EL, Hardy SA, et al. mtDNA heteroplasmy level and copy number indicate disease burden in m.3243A>G mitochondrial disease. EMBO Mol Med 2018;10(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bacman SR, Williams SL, Pinto M, Peralta S, Moraes CT. Specific elimination of mutant mitochondrial genomes in patient-derived cells by mitoTALENs. Nat Med 2013;19(9):1111–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hashimoto M, Bacman SR, Peralta S, Falk MJ, Chomyn A, Chan DC, et al. MitoTALEN: A General Approach to Reduce Mutant mtDNA Loads and Restore Oxidative Phosphorylation Function in Mitochondrial Diseases. Mol Ther 2015;23(10):1592–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yang Y, Wu H, Kang X, Liang Y, Lan T, Li T, et al. Targeted elimination of mutant mitochondrial DNA in MELAS-iPSCs by mitoTALENs. Protein Cell 2018;9(3):283–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Yahata N, Matsumoto Y, Omi M, Yamamoto N, Hata R. TALEN-mediated shift of mitochondrial DNA heteroplasmy in MELAS-iPSCs with m.13513G>A mutation. Sci Rep 2017;7(1):15557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.McCormick EM, Muraresku CC, Falk MJ. Mitochondrial Genomics: A Complex Field Now Coming of Age. Current Genetic Medicine Reports 2018. [DOI] [PMC free article] [PubMed]
- 21.Vachin P, Adda-Herzog E, Chalouhi G, Elie C, Rio M, Rondeau S, et al. Segregation of mitochondrial DNA mutations in the human placenta: implication for prenatal diagnosis of mtDNA disorders. J Med Genet 2018;55(2):131–6. [DOI] [PubMed] [Google Scholar]
- 22.Mitalipov S, Amato P, Parry S, Falk MJ. Limitations of preimplantation genetic diagnosis for mitochondrial DNA diseases. Cell Rep 2014;7(4):935–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sallevelt S, Dreesen J, Coonen E, Paulussen ADC, Hellebrekers D, de Die-Smulders CEM, et al. Preimplantation genetic diagnosis for mitochondrial DNA mutations: analysis of one blastomere suffices. J Med Genet 2017;54(10):693–7. [DOI] [PubMed] [Google Scholar]
- 24.Herbert M, Turnbull D. Mitochondrial Donation - Clearing the Final Regulatory Hurdle in the United Kingdom. N Engl J Med 2017;376(2):171–3. [DOI] [PubMed] [Google Scholar]
- 25.Falk MJ, Decherney A, Kahn JP. Mitochondrial Replacement Techniques--Implications for the Clinical Community. N Engl J Med 2016;374(12):1103–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Abu-Libdeh B, Douiev L, Amro S, Shahrour M, Ta-Shma A, Miller C, et al. Mutation in the COX4I1 gene is associated with short stature, poor weight gain and increased chromosomal breaks, simulating Fanconi anemia. Eur J Hum Genet 2017;25(10):1142–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Barca E, Ganetzky RD, Potluri P, Juanola-Falgarona M, Gai X, Li D, et al. USMG5 Ashkenazi Jewish founder mutation impairs mitochondrial complex V dimerization and ATP synthesis. Hum Mol Genet 2018. [DOI] [PMC free article] [PubMed]
- 28.Peng Y, Shinde DN, Valencia CA, Mo JS, Rosenfeld J, Truitt Cho M, et al. Biallelic mutations in the ferredoxin reductase gene cause novel mitochondriopathy with optic atrophy. Hum Mol Genet 2017;26(24):4937–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Paul A, Drecourt A, Petit F, Deguine DD, Vasnier C, Oufadem M, et al. FDXR Mutations Cause Sensorial Neuropathies and Expand the Spectrum of Mitochondrial Fe-S-Synthesis Diseases. Am J Hum Genet 2017;101(4):630–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wortmann SB, Timal S, Venselaar H, Wintjes LT, Kopajtich R, Feichtinger RG, et al. Biallelic variants in WARS2 encoding mitochondrial tryptophanyl-tRNA synthase in six individuals with mitochondrial encephalopathy. Hum Mutat 2017;38(12):1786–95. [DOI] [PubMed] [Google Scholar]
- 31.Theisen BE, Rumyantseva A, Cohen JS, Alcaraz WA, Shinde DN, Tang S, et al. Deficiency of WARS2, encoding mitochondrial tryptophanyl tRNA synthetase, causes severe infantile onset leukoencephalopathy. Am J Med Genet A 2017;173(9):2505–10. [DOI] [PubMed] [Google Scholar]
- 32.Lake NJ, Webb BD, Stroud DA, Richman TR, Ruzzenente B, Compton AG, et al. Biallelic Mutations in MRPS34 Lead to Instability of the Small Mitoribosomal Subunit and Leigh Syndrome. Am J Hum Genet 2017;101(2):239–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Gardeitchik T, Mohamed M, Ruzzenente B, Karall D, Guerrero-Castillo S, Dalloyaux D, et al. Bi-allelic Mutations in the Mitochondrial Ribosomal Protein MRPS2 Cause Sensorineural Hearing Loss, Hypoglycemia, and Multiple OXPHOS Complex Deficiencies. Am J Hum Genet 2018;102(4):685–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhang J, Ji Y, Liu X, Chen J, Wang B, Zhang M, et al. Leber’s hereditary optic neuropathy caused by a mutation in mitochondrial tRNA(Thr) in eight Chinese pedigrees. Mitochondrion 2017. [DOI] [PubMed]
- 35.Feichtinger RG, Olahova M, Kishita Y, Garone C, Kremer LS, Yagi M, et al. Biallelic C1QBP Mutations Cause Severe Neonatal-, Childhood-, or Later-Onset Cardiomyopathy Associated with Combined Respiratory-Chain Deficiencies. Am J Hum Genet 2017;101(4):525–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Garone C, D’Souza AR, Dallabona C, Lodi T, Rebelo-Guiomar P, Rorbach J, et al. Defective mitochondrial rRNA methyltransferase MRM2 causes MELAS-like clinical syndrome. Hum Mol Genet 2017;26(21):4257–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bartsakoulia M, Pyle A, Troncoso-Chandia D, Vial-Brizzi J, Paz-Fiblas MV, Duff J, et al. A novel mechanism causing imbalance of mitochondrial fusion and fission in human myopathies. Hum Mol Genet 2018;27(7):1186–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Gal A, Balicza P, Weaver D, Naghdi S, Joseph SK, Varnai P, et al. MSTO1 is a cytoplasmic pro-mitochondrial fusion protein, whose mutation induces myopathy and ataxia in humans. EMBO Mol Med 2017;9(7):967–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Nasca A, Scotton C, Zaharieva I, Neri M, Selvatici R, Magnusson OT, et al. Recessive mutations in MSTO1 cause mitochondrial dynamics impairment, leading to myopathy and ataxia. Hum Mutat 2017;38(8):970–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Boczonadi V, King MS, Smith AC, Olahova M, Bansagi B, Roos A, et al. Mitochondrial oxodicarboxylate carrier deficiency is associated with mitochondrial DNA depletion and spinal muscular atrophy-like disease. Genet Med 2018. [DOI] [PMC free article] [PubMed]
- 41.Ehmke N, Graul-Neumann L, Smorag L, Koenig R, Segebrecht L, Magoulas P, et al. De Novo Mutations in SLC25A24 Cause a Craniosynostosis Syndrome with Hypertrichosis, Progeroid Appearance, and Mitochondrial Dysfunction. Am J Hum Genet 2017;101(5):833–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Shamseldin HE, Alasmari A, Salih MA, Samman MM, Mian SA, Alshidi T, et al. A null mutation in MICU2 causes abnormal mitochondrial calcium homeostasis and a severe neurodevelopmental disorder. Brain 2017;140(11):2806–13. [DOI] [PubMed] [Google Scholar]
- 43.Langer Y, Aran A, Gulsuner S, Abu Libdeh B, Renbaum P, Brunetti D, et al. Mitochondrial PITRM1 peptidase loss-of-function in childhood cerebellar atrophy. J Med Genet 2018. [DOI] [PubMed]
- 44.Vogtle FN, Brandl B, Larson A, Pendziwiat M, Friederich MW, White SM, et al. Mutations in PMPCB Encoding the Catalytic Subunit of the Mitochondrial Presequence Protease Cause Neurodegeneration in Early Childhood. Am J Hum Genet 2018;102(4):557–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Perrier S, Gauquelin L, Tetreault M, Tran LT, Webb N, Srour M, et al. Recessive mutations in NDUFA2 cause mitochondrial leukoencephalopathy. Clin Genet 2018;93(2):396–400. [DOI] [PubMed] [Google Scholar]
- 46.Baertling F, Sanchez-Caballero L, van den Brand MAM, Fung CW, Chan SH, Wong VC, et al. NDUFA9 point mutations cause a variable mitochondrial complex I assembly defect. Clin Genet 2018;93(1):111–8. [DOI] [PubMed] [Google Scholar]
- 47.Ng YS, Lax NZ, Maddison P, Alston CL, Blakely EL, Hepplewhite PD, et al. MT-ND5 Mutation Exhibits Highly Variable Neurological Manifestations at Low Mutant Load. EBioMedicine 2018;30:86–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Baertling F, Sanchez-Caballero L, van den Brand MAM, Wintjes LT, Brink M, van den Brandt FA, et al. NDUFAF4 variants are associated with Leigh syndrome and cause a specific mitochondrial complex I assembly defect. Eur J Hum Genet 2017;25(11):1273–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Rebelo AP, Saade D, Pereira CV, Farooq A, Huff TC, Abreu L, et al. SCO2 mutations cause early-onset axonal Charcot-Marie-Tooth disease associated with cellular copper deficiency. Brain 2018;141(3):662–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Isohanni P, Carroll CJ, Jackson CB, Pohjanpelto M, Lonnqvist T, Suomalainen A. Defective mitochondrial ATPase due to rare mtDNA m.8969G>A mutation-causing lactic acidosis, intellectual disability, and poor growth. Neurogenetics 2018;19(1):49–53. [DOI] [PubMed] [Google Scholar]
- 51.Hempel M, Kremer LS, Tsiakas K, Alhaddad B, Haack TB, Lobel U, et al. LYRM7 - associated complex III deficiency: A clinical, molecular genetic, MR tomographic, and biochemical study. Mitochondrion 2017;37:55–61. [DOI] [PubMed] [Google Scholar]
- 52.Torraco A, Stehling O, Stumpfig C, Rosser R, De Rasmo D, Fiermonte G, et al. ISCA1 Mutation In A Patient With Infantile-Onset Leukodystrophy Causes Defects In Mitochondrial [4Fe-4S] Proteins. Hum Mol Genet 2018. [DOI] [PubMed]
- 53.Toldo I, Nosadini M, Boscardin C, Talenti G, Manara R, Lamantea E, et al. Neonatal mitochondrial leukoencephalopathy with brain and spinal involvement and high lactate: expanding the phenotype of ISCA2 gene mutations. Metab Brain Dis 2018;33(3):805–12. [DOI] [PubMed] [Google Scholar]
- 54.Alaimo JT, Besse A, Alston CL, Pang K, Appadurai V, Samanta M, et al. Loss-of-function mutations in ISCA2 disrupt 4Fe-4S cluster machinery and cause a fatal leukodystrophy with hyperglycinemia and mtDNA depletion. Hum Mutat 2018;39(4):537–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Alfadhel M, Nashabat M, Alrifai MT, Alshaalan H, Al Mutairi F, Al-Shahrani SA, et al. Further delineation of the phenotypic spectrum of ISCA2 defect: A report of ten new cases. Eur J Paediatr Neurol 2018;22(1):46–55. [DOI] [PubMed] [Google Scholar]
- 56.Vantroys E, Larson A, Friederich M, Knight K, Swanson MA, Powell CA, et al. New insights into the phenotype of FARS2 deficiency. Mol Genet Metab 2017;122(4):172–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Nafisinia M, Riley LG, Gold WA, Bhattacharya K, Broderick CR, Thorburn DR, et al. Compound heterozygous mutations in glycyl-tRNA synthetase (GARS) cause mitochondrial respiratory chain dysfunction. PLoS One 2017;12(6):e0178125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Gerber S, Charif M, Chevrollier A, Chaumette T, Angebault C, Kane MS, et al. Mutations in DNM1L, as in OPA1, result indominant optic atrophy despite opposite effectson mitochondrial fusion and fission. Brain 2017;140(10):2586–96. [DOI] [PubMed] [Google Scholar]
- 59.Musa S, Eyaid W, Kamer K, Ali R, Al-Mureikhi M, Shahbeck N, et al. A Middle Eastern Founder Mutation Expands the Genotypic and Phenotypic Spectrum of Mitochondrial MICU1 Deficiency: A Report of 13 Patients. JIMD Rep 2018. [DOI] [PMC free article] [PubMed]
- 60.Iuso A, Alhaddad B, Weigel C, Kotzaeridou U, Mastantuono E, Schwarzmayr T, et al. A Homozygous Splice Site Mutation in SLC25A42, Encoding the Mitochondrial Transporter of Coenzyme A, Causes Metabolic Crises and Epileptic Encephalopathy. JIMD Rep 2018. [DOI] [PMC free article] [PubMed]
- 61.Almannai M, Alasmari A, Alqasmi A, Faqeih E, Al Mutairi F, Alotaibi M, et al. Expanding the phenotype of SLC25A42-associated mitochondrial encephalomyopathy. Clin Genet 2018;93(5):1097–102. [DOI] [PubMed] [Google Scholar]
- 62.Heimer G, Eyal E, Zhu X, Ruzzo EK, Marek-Yagel D, Sagiv D, et al. Mutations in AIFM1 cause an X-linked childhood cerebellar ataxia partially responsive to riboflavin. Eur J Paediatr Neurol 2018;22(1):93–101. [DOI] [PubMed] [Google Scholar]
- 63.Miyake N, Wolf NI, Cayami FK, Crawford J, Bley A, Bulas D, et al. X-linked hypomyelination with spondylometaphyseal dysplasia (H-SMD) associated with mutations in AIFM1. Neurogenetics 2017;18(4):185–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.El-Hattab AW, Dai H, Almannai M, Wang J, Faqeih EA, Al Asmari A, et al. Molecular and clinical spectra of FBXL4 deficiency. Hum Mutat 2017;38(12):1649–59. [DOI] [PubMed] [Google Scholar]
- 65.Shukla A, Saneto RP, Hebbar M, Mirzaa G, Girisha KM. A neurodegenerative mitochondrial disease phenotype due to biallelic loss-of-function variants in PNPLA8 encoding calcium-independent phospholipase A2gamma. Am J Med Genet A 2018;176(5):1232–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Maas RR, Iwanicka-Pronicka K, Kalkan Ucar S, Alhaddad B, AlSayed M, Al-Owain MA, et al. Progressive deafness-dystonia due to SERAC1 mutations: A study of 67 cases. Ann Neurol 2017;82(6):1004–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Garone C, Taylor RW, Nascimento A, Poulton J, Fratter C, Dominguez-Gonzalez C, et al. Retrospective natural history of thymidine kinase 2 deficiency. J Med Genet 2018. [DOI] [PMC free article] [PubMed]
- 68.Falkenberg M Mitochondrial DNA replication in mammalian cells: overview of the pathway. Essays Biochem 2018. [DOI] [PMC free article] [PubMed]
- 69.Almannai M, El-Hattab AW, Scaglia F. Mitochondrial DNA replication: clinical syndromes. Essays Biochem 2018. [DOI] [PubMed]
- 70.Boczonadi V, Ricci G, Horvath R. Mitochondrial DNA transcription and translation: clinical syndromes. Essays Biochem 2018. [DOI] [PMC free article] [PubMed]
- 71.Boenzi S, Diodato D. Biomarkers for mitochondrial energy metabolism diseases. Essays Biochem 2018. [DOI] [PubMed]
- 72.Raymond FL, Horvath R, Chinnery PF. First-line genomic diagnosis of mitochondrial disorders. Nat Rev Genet 2018;19(7):399–400. [DOI] [PubMed] [Google Scholar]
- 73.Hirano M, Emmanuele V, Quinzii CM. Emerging therapies for mitochondrial diseases. Essays Biochem 2018. [DOI] [PMC free article] [PubMed]
- 74.Rai PK, Craven L, Hoogewijs K, Russell OM, Lightowlers RN. Advances in methods for reducing mitochondrial DNA disease by replacing or manipulating the mitochondrial genome. Essays Biochem 2018. [DOI] [PMC free article] [PubMed]
- 75.Muraresku CMMEM, Falk MJ Mitochondrial Disease: Advances in Clinical Diagnosis, Management, Therapeutic Development, and Preventative Strategies. Current Genetic Medicine Reports 2018;6:62–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Grier J, Hirano M, Karaa A, Shepard E, Thompson JLP. Diagnostic odyssey of patients with mitochondrial disease: Results of a survey. Neurol Genet 2018;4(2):e230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Shen L, Attimonelli M, Bai R, Lott MT, Wallace DC, Falk MJ, et al. MSeqDR mvTool: A mitochondrial DNA Web and API resource for comprehensive variant annotation, universal nomenclature collation, and reference genome conversion. Hum Mutat 2018. [DOI] [PMC free article] [PubMed]
- 78.Falk MJ, Shen L, Gai X. From case studies to community knowledge base: MSeqDR provides a platform for the curation and genomic analysis of mitochondrial diseases. Cold Spring Harb Mol Case Stud 2016;2(3):a001065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Shen L, Diroma MA, Gonzalez M, Navarro-Gomez D, Leipzig J, Lott MT, et al. MSeqDR: A Centralized Knowledge Repository and Bioinformatics Web Resource to Facilitate Genomic Investigations in Mitochondrial Disease. Hum Mutat 2016;37(6):540–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Rehm HL, Berg JS, Brooks LD, Bustamante CD, Evans JP, Landrum MJ, et al. ClinGen--the Clinical Genome Resource. N Engl J Med 2015;372(23):2235–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lott MT, Leipzig JN, Derbeneva O, Xie HM, Chalkia D, Sarmady M, et al. mtDNA Variation and Analysis Using Mitomap and Mitomaster. Curr Protoc Bioinformatics 2013;44:1 23 1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sonney S, Leipzig J, Lott MT, Zhang S, Procaccio V, Wallace DC, et al. Predicting the pathogenicity of novel variants in mitochondrial tRNA with MitoTIP. PLoS Comput Biol 2017;13(12):e1005867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Clima R, Preste R, Calabrese C, Diroma MA, Santorsola M, Scioscia G, et al. HmtDB 2016: data update, a better performing query system and human mitochondrial DNA haplogroup predictor. Nucleic Acids Res 2017;45(D1):D698–D706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Longley MJ, Graziewicz MA, Bienstock RJ, Copeland WC. Consequences of mutations in human DNA polymerase gamma. Gene 2005;354:125–31. [DOI] [PubMed] [Google Scholar]
- 85.Nurminen A, Farnum GA, Kaguni LS. Pathogenicity in POLG syndromes: DNA polymerase gamma pathogenicity prediction server and database. BBA Clin 2017;7:147–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Holt IJ, Harding AE, Morgan-Hughes JA. Mitochondrial DNA polymorphism in mitochondrial myopathy. Hum Genet 1988;79(1):53–7. [DOI] [PubMed] [Google Scholar]
- 87.Frazier AE, Thorburn DR, Compton AG. Mitochondrial energy generation disorders: genes, mechanisms and clues to pathology. J Biol Chem 2017. [DOI] [PMC free article] [PubMed]