For the vast majority of patients with mitochondrial diseases, only supportive and symptomatic therapies are available. However, in the last decade, due to extraordinary advances in defining the causes and pathomechanisms of these diverse disorders, new therapies are being developed in the laboratory and are entering human clinical trials. In this review, we highlight the current use of dietary supplement and exercise therapies as well as emerging therapies that may be broadly applicable across multiple mitochondrial diseases or tailored for specific disorders. Examples of non-tailored therapeutic targets include: activation of mitochondrial biogenesis, regulation of mitophagy and mitochondrial dynamics, bypass of biochemical defects, mitochondrial replacement therapy, and hypoxia. In contrast, tailored therapies are: scavenging of toxic compounds, deoxynucleoside and deoxynucleotide treatments, cell replacement therapies, gene therapy, shifting mitochondrial DNA mutation heteroplasmy, and stabilization of mutant mitochondrial transfer RNAs.

There are disappointingly few therapies for mitochondrial diseases [1,2]. Over the last decade, for most mitochondrial disease patients, treatment has been largely restricted to exercise, dietary supplements of uncertain benefit. Only a handful of diseases respond to specific supplements such as coenzyme Q10 (CoQ10) for primary and secondary forms of CoQ10 deficiency [3–5]. Obstacles to the discovery of treatments for mitochondrial disorders include the disease: rarity, clinical diversity, pathogenic complexity, etiological heterogeneity, and insufficient clinical trials [6]. In contrast, there has been remarkable progress in our understanding of the molecular genetic causes, pathomechanisms, and clinical presentations of mitochondrial diseases [2,7]. Due to these advances and emerging clinical trials, new treatment modalities are on the horizon.

Pharmacological approaches

Multiple vitamins and cofactors are often used in patients with mitochondrial disorders, although these therapies are not yet standardized or definitively proven to be effective. The dietary supplements are used with different purposes, such as (1) increase respiratory chain flux (CoQ10, riboflavin), (2) serve as antioxidants (e.g. CoQ10, idebenone, α-lipoic acid, vitamin C and E), and/or act as cofactors (e.g. riboflavin, thiamine), or (3) function as mitochondrial substrates (L-carnitine).

In many mitochondrial diseases due to respiratory chain dysfunction, excessive toxic reactive oxygen species (ROS) may lead to pathogenic cellular damage. Moreover, a transgenic murine model overexpressing a catalase targeted to mitochondria extended life span [8]. Based on this rationale, antioxidants are frequently used in the treatment of mitochondrial patients. CoQ10 is the most commonly utilized and many clinical trials have been investigating its efficacy and that of its analogs, like idebenone and EPI-743, in multiple mitochondrial diseases (see “clinical trials”). Other antioxidants like vitamins C and E might also be beneficial in patients with mitochondrial diseases. An example is an analog of vitamin E, trolox ornithylamide hydrochloride, when applied to fibroblasts from patients with Leigh syndrome reduced ROS levels and increased activities of mitochondrial complexes I, IV, and citrate synthase [9]. Nevertheless, efficacy of antioxidants in patients with mitochondrial diseases remains controversial. A Cochrane review of mitochondrial therapies has found little evidence supporting the use of any vitamin or cofactor [1]. However, benefits of various agents (riboflavin, α-lipoic acid etc.) have been anecdotally reported.

Consensus recommendations from the Mitochondrial Medicine Society [5] aimed to standardize treatment options for mitochondrial patients. According to these recommendations, patients with primary mitochondrial disorders should be offered CoQ10 in its reduced form (ubiquinol), and plasma or leukocyte levels should be monitored to assess adherence to treatment. In addition, α-lipoic acid (ALA) and riboflavin are frequently offered to mitochondrial patients. L-carnitine should be administered when deficient. Folinic acid should be given to mitochondrial patients when deficient and the central nervous system is involved. Moreover, supplements should be given starting with one supplement at the time, avoiding “cocktails” initially.

In contrast with the recommendations of the Mitochondrial Medicine Society, a survey on the use of dietary supplements conducted by the North American Mitochondrial Disease Consortium (NAMDC) [10] revealed that in practice, the majority of patients take a cocktail of at least four dietary supplements. Patients reported no or minor side effects with overall subjective improvements; however, the economic burden to the families was considerable; 90% of patients purchased the supplements out-of-pocket. The authors conclude that this burden and the potential side-effects are not justifiable, considering the lack of evidence for using these “cocktails”. Importantly, this survey underscored the importance of considering the patients’ perception of their care and quality of life; this approach is critical in selecting reliable outcome measures for future randomized placebo-controlled double-blind clinical trials, which is still the unattained gold-standard for assessing dietary supplements as therapy for mitochondrial disease.

Effective treatment of acute stroke-like episodes or their prevention has not been established. Open-label studies suggest that treatment of acute mitochondrial stroke-like episodes with intravenous (IV) arginine hydrochloride, a precursor of nitric oxide, is beneficial for patient with the m.3243A>G mutation in MTTL1 [11]. Open-label studies also suggest that daily oral arginine to prevent strokes should be considered in patients with mitochondrial encephalomyopathy lactic acidosis and stroke-like episodes (MELAS) due to the m.3243A>G mutation [5]. Placebo-controlled randomized clinical trials are necessary before L-arginine can be definitively recommended to ameliorate or treat stroke-like episodes in MELAS.

Exercise

Exercise has been proven beneficial in some patients with mitochondrial diseases [12,13]. In particular, aerobic endurance training can increase mitochondrial mass, by stimulating mitochondrial biogenesis, and increase muscle mitochondrial enzyme activities and muscle strength. Endurance training has been proven beneficial and safe in trials of patients with mitochondrial DNA mutations [14,15]. A combination of progressive endurance with or without resistance exercise should be recommended to mitochondrial patients [5].

Few clinical trials have been conducted in mitochondrial diseases. In 2012, a comprehensive Cochrane review evaluated 1,335 studies comparing pharmacological and nonpharmacological treatments for mitochondrial diseases [1]. Only 12 studies were selected for inclusion in the review, with the most common reason for exclusion being lack of randomization/blinding and presence of methodological biases. The primary outcome measures included any change in muscle strength or neurological features. Secondary outcome measures included quality life evaluation, biochemical biomarkers (e.g. lactic acidosis), and negative outcomes. The 12 studies investigated the effects of CoQ10, dichloroacetate (DCA), creatinine, dimethylglycine, whey-based cysteine and combination therapy of creatine, α-lipoic acid and CoQ10. Dramatic effects were not observed in any of these studies, and one trial assessing the effects of DCA in MELAS patients had to be terminated because of toxicity (NCT00068913). Several clinical trials are currently underway or have been recently completed, but the results were not published for most of them and the outcomes are still unclear. The majority of the studies focused on patients with MELAS and LHON, which could be studied in relatively large cohorts. Many other trials analyzed less homogeneous cohort of patients, including, for instance, patients with similar phenotype (i.e. mitochondrial myopathy), but different genetic cause. A summary of the studies is reported in Table 1.

Table 1
Recent clinical trials in mitochondrial disorders
TreatmentDiseaseDesignMechanismStatusOutcomeTrial number
Active studies 
EPI-743 Mitochondrial diseases Phase 2, Emergency use protocol in acutely ill patients (90 days endo-of-life care) Antioxidant Active/not recruiting na NCT01370447 
EPI-743 Children (2–11 years of age) with MDs or metabolic diseases Phase 2, randomized, double blind, placebo-controlled, crossover Antioxidant Active/not recruiting na NCT01642056 
KH176 MELAS, MIDD, mitochondrial myopathies, mitochondrial diseases Phase 2, randomized, double blind, placebo-controlled, crossover Antioxidant Recruiting na NCT02909400 
RTA 408 Mitochondrial myopathy Phase 2 randomized, double blind, placebo-controlled, dose-escalating Antioxidant, NRF2 activator, NFκB inhibitor Completed na NCT02255422 
MTP-131 (Elamipretide) Mitochondrial myopathy Phase 2, randomized, double blind, placebo-controlled, crossover Cardiolipin stabilization Active/not recruiting na NCT02805790 
MTP-131 (Elamipretide) Mitochondrial myopathy Phase 2, open label Cardiolipin stabilization Enrolling by invitation na NCT02976038 
MTP-131 (Elamipretide) ophthalmic LHON Phase 2, prospective, randomized, double blind, vehicle-controlled Cardiolipin stabilization Active/not recruiting na NCT02693119 
DCA (Dichloroacetate) PDC deficiency Phase 3, randomized, placebo-controlled, crossover Lowering lactate levels Recruiting na NCT02616484 
Resistance exercise Barth syndrome Phase 2, open label Increase glycolytic type 1 muscle fibers Recruiting na NCT01629459 
scAAV2-P1ND4v2 LHON m.11778G>A Phase 1, open label, dose-escalating Gene therapy Recruiting na NCT02161380 
rAAV2/2-ND4 (GS010) LHON Phase1/2 safety, open label, dose escalating Gene therapy Active/not recruiting na NCT02064569 
Allogenic HSCT MNGIE Phase 1 safety study Production of TP Recruiting na NCT02427178 
Completed studies 
CoQ10 Children with mitochondrial diseases with mtDNA mutations or specific OXPHOS complexes defects Phase 3, randomized, double blind OXPHOS/ROS Completed na NCT00432744 
Idebenone LHON Phase 2, randomized, double blind, placebo-controlled Antioxidant Completed Primary endpoint did not reach statistical significance; secondary outcomes significantly differ in a subgroup ot patients with discordant acuity at baseline NCT00747487 
Idebenone MELAS Phase 2, randomized, double blind, placebo-controlled, dose-finding Antioxidant Completed Primary endpoint did not reach statistical significance NCT00887562 
EPI-743 Leigh syndrome Phase 2b, randomized, double blind, placebo-controlled Antioxidant Completed na NCT01721733 
EPI-743 Pearson syndrome Phase 2, open label Antioxidant Terminated Results from other studies did not support continuation of this trial NCT02104336 
KH176 MELAS, LHON, Leigh, and other mitochondrial diseases Phase 1, randomized, double blind, placebo-controlled, crossover Antioxidant Completed Well tolerated with promising pharmacokinetic profile NCT02544217 
Bezafibrate Mitochondrial myopathy (m.3243A>G) Phase 2, open label Mitochondrial biogenesis Completed na NCT02398201 
Curcumin LHON Phase 3, randomized, double blind, placebo-controlled Antioxidant Completed na NCT00528151 
MTP-131 Mitochondrial myopahty Phase 1/2 randomized, double blind, placebo-controlled, dose-escalating Cardiolipin stabilization Completed na NCT02367014 
RP103 (Cysteamine bitartrate delayed-release) Childhood mitochondrial diseases including Leigh syndrome Phase 2 open-label, dose-escalating Cystine-depleting agent Completed Primary endpoint did not reach statistical significance; high percentage of serious adverse events (30.6%) NCT02023866 
RP103 (Cysteamine bitartrate delayed-release) Childhood MDs including Leigh syndrome Phase 2, long term open-label extension study Cystine-depleting agent Completed na NCT02473445 
Medium chain triglycerides MELAS Phase 1, open label Shift heteroplasmy Completed na NCT01252979L 
L-Arginine MELAS Phase 2, open label NO precursor Completed Improvement in aerobic capacity and muscle metabolism NCT01603446 
L-Arginine (IV) MELAS Phase 3, open label NO precursor Completed Improvement of stroke-like symptoms JMA-IIA00023 
L-Arginine (PO) MELAS Phase 3, open-label NO precursor Completed Improved endothelial dysfunction JMA-IIA00025 
Arginine and citrulline MELAS Phase 1, open-label NO precursor Completed na NCT01339494 
Lipoic acid Mitochondrial myopathy Pilot compassionate use study – Completed na NCT00004770 
RG2133 (2′,3,5′-tri-o-acetyluridine) Mitochondrial diseases Phase 1, open label, dose escalating – Completed na NCT00060515 
SPP-004 (5-Ala and SFC) Mitochondrial diseases, mainly cranial nerve symptoms Phase 2, randomized, placebo-controlled – Completed na JMA-IIA00200 
Taurine MELAS Phase 2/3 open-label Taurine modification Completed na UMIN000011908 
Pyruvate MELAS Phase 2, randomized, placebo-controlled NAD donor Unknown na JMA-IIA00093 
DCA (Dichloroacetate) MELAS Phase 2, randomized, double blind, crossover Lowering lactate levels Terminated Terminated because of peripheral nerve toxicity NCT00068913 
Cyclosporine LHON acute phase Phase 2, open label Inhibition of mitochondrial PTP Unknown na NCT02176733 
rAAV2-ND4 LHON m.11778G>A Open label Gene therapy Completed Improvement of visual acuity and enlargement of visual field NCT01267422 
TreatmentDiseaseDesignMechanismStatusOutcomeTrial number
Active studies 
EPI-743 Mitochondrial diseases Phase 2, Emergency use protocol in acutely ill patients (90 days endo-of-life care) Antioxidant Active/not recruiting na NCT01370447 
EPI-743 Children (2–11 years of age) with MDs or metabolic diseases Phase 2, randomized, double blind, placebo-controlled, crossover Antioxidant Active/not recruiting na NCT01642056 
KH176 MELAS, MIDD, mitochondrial myopathies, mitochondrial diseases Phase 2, randomized, double blind, placebo-controlled, crossover Antioxidant Recruiting na NCT02909400 
RTA 408 Mitochondrial myopathy Phase 2 randomized, double blind, placebo-controlled, dose-escalating Antioxidant, NRF2 activator, NFκB inhibitor Completed na NCT02255422 
MTP-131 (Elamipretide) Mitochondrial myopathy Phase 2, randomized, double blind, placebo-controlled, crossover Cardiolipin stabilization Active/not recruiting na NCT02805790 
MTP-131 (Elamipretide) Mitochondrial myopathy Phase 2, open label Cardiolipin stabilization Enrolling by invitation na NCT02976038 
MTP-131 (Elamipretide) ophthalmic LHON Phase 2, prospective, randomized, double blind, vehicle-controlled Cardiolipin stabilization Active/not recruiting na NCT02693119 
DCA (Dichloroacetate) PDC deficiency Phase 3, randomized, placebo-controlled, crossover Lowering lactate levels Recruiting na NCT02616484 
Resistance exercise Barth syndrome Phase 2, open label Increase glycolytic type 1 muscle fibers Recruiting na NCT01629459 
scAAV2-P1ND4v2 LHON m.11778G>A Phase 1, open label, dose-escalating Gene therapy Recruiting na NCT02161380 
rAAV2/2-ND4 (GS010) LHON Phase1/2 safety, open label, dose escalating Gene therapy Active/not recruiting na NCT02064569 
Allogenic HSCT MNGIE Phase 1 safety study Production of TP Recruiting na NCT02427178 
Completed studies 
CoQ10 Children with mitochondrial diseases with mtDNA mutations or specific OXPHOS complexes defects Phase 3, randomized, double blind OXPHOS/ROS Completed na NCT00432744 
Idebenone LHON Phase 2, randomized, double blind, placebo-controlled Antioxidant Completed Primary endpoint did not reach statistical significance; secondary outcomes significantly differ in a subgroup ot patients with discordant acuity at baseline NCT00747487 
Idebenone MELAS Phase 2, randomized, double blind, placebo-controlled, dose-finding Antioxidant Completed Primary endpoint did not reach statistical significance NCT00887562 
EPI-743 Leigh syndrome Phase 2b, randomized, double blind, placebo-controlled Antioxidant Completed na NCT01721733 
EPI-743 Pearson syndrome Phase 2, open label Antioxidant Terminated Results from other studies did not support continuation of this trial NCT02104336 
KH176 MELAS, LHON, Leigh, and other mitochondrial diseases Phase 1, randomized, double blind, placebo-controlled, crossover Antioxidant Completed Well tolerated with promising pharmacokinetic profile NCT02544217 
Bezafibrate Mitochondrial myopathy (m.3243A>G) Phase 2, open label Mitochondrial biogenesis Completed na NCT02398201 
Curcumin LHON Phase 3, randomized, double blind, placebo-controlled Antioxidant Completed na NCT00528151 
MTP-131 Mitochondrial myopahty Phase 1/2 randomized, double blind, placebo-controlled, dose-escalating Cardiolipin stabilization Completed na NCT02367014 
RP103 (Cysteamine bitartrate delayed-release) Childhood mitochondrial diseases including Leigh syndrome Phase 2 open-label, dose-escalating Cystine-depleting agent Completed Primary endpoint did not reach statistical significance; high percentage of serious adverse events (30.6%) NCT02023866 
RP103 (Cysteamine bitartrate delayed-release) Childhood MDs including Leigh syndrome Phase 2, long term open-label extension study Cystine-depleting agent Completed na NCT02473445 
Medium chain triglycerides MELAS Phase 1, open label Shift heteroplasmy Completed na NCT01252979L 
L-Arginine MELAS Phase 2, open label NO precursor Completed Improvement in aerobic capacity and muscle metabolism NCT01603446 
L-Arginine (IV) MELAS Phase 3, open label NO precursor Completed Improvement of stroke-like symptoms JMA-IIA00023 
L-Arginine (PO) MELAS Phase 3, open-label NO precursor Completed Improved endothelial dysfunction JMA-IIA00025 
Arginine and citrulline MELAS Phase 1, open-label NO precursor Completed na NCT01339494 
Lipoic acid Mitochondrial myopathy Pilot compassionate use study – Completed na NCT00004770 
RG2133 (2′,3,5′-tri-o-acetyluridine) Mitochondrial diseases Phase 1, open label, dose escalating – Completed na NCT00060515 
SPP-004 (5-Ala and SFC) Mitochondrial diseases, mainly cranial nerve symptoms Phase 2, randomized, placebo-controlled – Completed na JMA-IIA00200 
Taurine MELAS Phase 2/3 open-label Taurine modification Completed na UMIN000011908 
Pyruvate MELAS Phase 2, randomized, placebo-controlled NAD donor Unknown na JMA-IIA00093 
DCA (Dichloroacetate) MELAS Phase 2, randomized, double blind, crossover Lowering lactate levels Terminated Terminated because of peripheral nerve toxicity NCT00068913 
Cyclosporine LHON acute phase Phase 2, open label Inhibition of mitochondrial PTP Unknown na NCT02176733 
rAAV2-ND4 LHON m.11778G>A Open label Gene therapy Completed Improvement of visual acuity and enlargement of visual field NCT01267422 

Abbreviations: AHSCT, allogeneic hematopoietic stem cell transplantation; IV, intravenous; LHON, Leber hereditary optic neuropathy; MELAS, mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes; MIDD, maternally inherited diabetes-deafness syndrome; MNGIE, mitochondrial neurogastrointestinal encephalomyopathy; na, not available; PDC, pyruvate dehydrogenase complex; PO, per oral; PTP, permeability transition pore.

MELAS

MELAS is one of the most frequent maternally inherited mitochondrial disorders. The pathogenesis of this disorder is not completely understood and results from different factor. Energy failure due to faulty mitochondria is a common feature of mitochondrial diseases as well as the overproduction of ROS. Different approaches have been studied in order to reduce the oxidative stress in mitochondrial diseases and in particular in MELAS patients. Idebenone is a short-tail ubiquinone synthetic analog, which is more water-soluble compared with CoQ10 and acts as and antioxidant. Several studies have been conducted to assess the efficacy and safety of idebenone in Friedreich ataxia (FA), and proven that it is in fact well-tolerated but not clinically effective [16–20]. A phase 2a, randomized, double blind, placebo-control, dose-finding study in patients with MELAS syndrome has recently been completed and showed that the primary endpoint did not reach statistical significance (NCT00887562). KH176 is a small molecule vitamin E derivative and a potent ROS scavenger that enhances the antioxidant thioredoxin/peroxiredoxin system [21]. After the completion of a dose escalating clinical trial with KH176 in healthy individual that has demonstrated good tolerability and a promising pharmacokinetic profile [22], a double-blind randomized, placebo-controlled two-way crossover phase II study of mildly affected patients with the m.3243A>G mutation revealed no significant improvement in the primary outcome, gait analysis, but showed positive effects on attention performance and measures of depression and anxiety [21]. Moreover, bezafibrate is an activator of the transcription factor peroxisomal proliferator-activated receptors (PPARs), which in turn when activated promote transcription of nuclear-encoded mitochondrial genes. Its efficacy is being evaluated in patients with m.3243A>G mutation (NCT02398201) and evidence of myopathy, but results are not available.

In addition to energy failure and ROS accumulation, there has been growing evidence that nitric oxide (NO) deficiency plays a central role in the pathogenesis of the stroke-like episodes [23]. Arginine is the substrate of nitric oxide synthase, which produces NO, therefore arginine is a promising treatment for MELAS patients. Multiple open-label trials have been conducted (NCT01603446, JMA-IIA00023, and JMA-IIA00025) and have suggested efficacy of chronic oral administration [24–26] and acute intravenous administration [24,27] of arginine in patients with MELAS syndrome; however, a placebo-controlled randomized clinical trial is required to demonstrate convincing evidence of efficacy. Further preliminary evidence has suggested more potent effect of citrulline than L-arginine in stroke-like episodes in MELAS patients [23]. A Phase 1, open-label dose-escalation study is being conducted (NCT01339494). Other molecules investigated in clinical trials for patients with MELAS include pyruvate (JMA-IIA00093), taurine (UMIN000011908), and supplemental medium chain triglycerides (NCT01252979); however, those results of trials are still pending.

Leber hereditary optic neuropathy (LHON)

LHON is a mitochondrial disorder characterized by painless, subacute visual loss affecting the central visual field in one eye, followed by similar symptoms in the other eye typically with 2 or 3 months of delay. Three mtDNA mutations are commonly associated with LHON: m.3460G>A in MT-ND1, m.11778G>A in MT-ND4, or m.14484T>C in MT-ND6 [28]. Many therapeutic approaches have been tested in patients with LHON, the majority of which are focused on the use of antioxidants. In particular, idebenone has been evaluated in a clinical trial of LHON; although primary endpoints did not reach statistical significance, overall, almost one in three patients achieved clinically relevant recovery (30.2% versus 10.3% of placebo patients, P=0.056) and a possible beneficial effect was observed in a subgroup of patients with discordant visual acuity at baseline [29,30] (NCT00747487). Idebenone has been recently approved for the treatment of LHON in Europe.

MTP-131 (elamipretide) is a small molecule targeting inner mitochondrial membrane that has been proven able to correct excessive ROS and increase ATP synthesis in preclinical studies [31]; this molecule has entered clinical trials for LHON (NCT02693119), as well as for mitochondrial myopathies and mitochondrial diseases (NCT02367014, NCT02976038, NCT02805790) and has shown promising preliminary results. Curcumin, a derivate of the spice turmeric (Curcuma longa), has also displayed antioxidant properties and a trial has been completed in patients with LHON, but no results are available (NCT00528151). At last, aiming at mitochondrial delivery of a normal mtDNA-encoded ND4 protein, allotopic expression has been attempted in patients with LHON, despite controversial preclinical results. One trial (NCT01267422) has been recently completed and has shown significant improvement of visual acuity and enlargement of visual field in the treated group [32]. Two additional trials are underway (NCT02161380; NCT02064569).

Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is an autosomal recessive mitochondrial disorder characterized by severe gastrointestinal dysmotility, cachexia, progressive external ophthalmoplegia, myopathy, and peripheral demyelinating neuropathy [33]. It is caused by autosomal recessive mutations in TYMP gene, encoding thymidine phosphorylase (TP). TP is a cytosolic enzyme that catalyzes the first step of thymidine and deoxyuridine catabolism. When the enzyme is deficient, thymidine and deoxyuridine accumulate and become toxic, leading to mtDNA instability. A retrospective analysis of 24 MNGIE patients who underwent allogeneic hematopoietic stem cell transplant (AHSCT) as a mean to replace TP enzyme, for MNGIE patients, revealed that only 9 (37.5%) were alive at last follow-up with an unacceptably high mortality attributed to the transplant (9 patients 37.5%); however, survival was associated with human leukocyte antigen match (10/10) and absence of liver disease and gastrointestinal pseudo-obstruction [34]. After successful AHSCT, patients showed normalization of buffy coat TP activity and plasma thymidine and deoxyuridine levels as well as clinical improvements more than 2 years after transplant. Two trials at Columbia University, USA, are currently recruiting to define natural history of the disease (NCT01694953) and to assess the safety of AHSCT (NCT01694953). Liver transplantation has been proposed as a safer alternative to AHSCT due to absence of a stressful conditioning regiment prior to this procedure. One patient who underwent a successful hepatic transplant showed normalization of plasma thymidine and deoxyuridine with clinical stabilization for over 1 year; however, data from additional patients are needed before this therapy can be recommended. Preclinical studies indicate that gene therapy is also a potentially effective means to restore TP activity in MNGIE [35–37]. Erythrocyte-encapsulated thymidine phosphorylase has also been demonstrated to be effective at delivering TP to MNGIE patients and may be useful as a bridge therapy to more permanent treatment [38].

Heterogeneous cohort of patients with mitochondrial diseases

Many of the therapeutic interventions acting on common pathogenic pathways of mitochondrial diseases have been tested in nonhomogeneous cohort of patients or in different disorders. One example is EPI-743, a para-benzoquinone analog modified to exert a higher antioxidant effect compared with CoQ10 and idebenone [39]. This molecule is supposed to enhance the biosynthesis of glutathione (GSH), which is an important cellular antioxidant. Two open label studies conducted independently in North America with patients with various mitochondrial diseases and in Italy with a cohort of Leigh syndrome patients showed promising results [40,41]. As a result, a randomized clinical trial has started in patients with Leigh syndrome (NCT01721733) as well as in patients with FA (NCT01728064), and in acutely ill patients (90 days of end of life care) (NCT01370447). A clinical trial of Pearson syndrome, a disorder caused by single large-scale mtDNA deletions, has been terminated because results of other studies have not supported continuation (NCT02104336). EPI-743 has also been studied in an open-label trial of patients with LHON with favorable outcome [42].

In the past few years, many potential treatments have been proposed for mitochondrial disorders. These approaches act on different disease mechanisms and can be broadly divided in “non-tailored strategies”, acting on common pathways thus in theory relevant to different mitochondrial diseases, and “disease-tailored” strategies [43]. Examples of these strategies are summarized in Table 2. The first group includes strategies aimed at: (1) activation of mitochondrial biogenesis; (2) regulation of mitophagy and mitochondrial dynamics; (3) bypass of OXPHOS defects; (4) mitochondrial replacement therapy (MRT); and (5) chronic hypoxia. The second group includes: (1) scavenging of specific toxic compounds; (2) supplementation of nucleosides and nucleotides; (3) cell replacement therapies; (4) gene therapy; (5) shifting mutant mtDNA heteroplasmy; and (6) stabilizing mutant tRNAs. Some of these approaches have been proven effective only in preclinical models while others have already been successfully applied anecdotally to patients with mitochondrial diseases (Table 2).

Table 2
Examples of preclinical therapies in mitochondrial disorders
StrategyMethodModel
Nontailored 
Activation of mitochondrial biogenesis • Nicotinamide riboside and PARP1 [47,48]
• AICAR [49]
• Bezafibrate [49–54]
• Resveratrol [55–57]
• Retinoic acid [58]
• Endurance exercise [59–62
Sco2 knockout/knockin mouse, Deletor mouse model
• Patients fibroblasts and mouse models of COX deficiency
• Patients fibroblasts, cybrids, mouse models of COX deficiency, Deletor mouse model
In vitro models, Drosophila models, human fibroblasts
• Cybrids (m.3243A>G)
• Mouse models of COX deficiency, mtDNA mutator mice, patients with MDs 
Regulating mitophagy and mitochondrial dynamics • Rapamycin [65,66In vitro models and Ndufs4−/− mouse model of Leigh syndrome; Tk2H126N/ H126N mouse model 
Bypassing OXPHOS blocks • Ndi1 (Complex I defect) [67,68] and AOX (Complex III and IV defects) [69,70In vitro models and Drosophila models 
Mitochondrial replacement therapy • Oocyte nuclear genetic material transfer [71• Nonhuman primates; healthy subjects and patients with mitochondrial diseases 
Chronic hypoxia • Genetic or small molecule activation of the hypoxia response [72]
• Moderate environmental hypoxia [72
• Cultured cells and zebrafish models
Ndufs4−/− mouse model of Leigh syndrome 
Disease-tailored 
Scavenging of specific toxic compounds • N-acetyl cysteine and metronidazole [73]
• Hemodialysis [74
Ethe1−/− mouse model of EE; patients with EE
• Patients with MNGIE 
Supplementation of nucleotides/nucleosides • Deoxycytidine and deoxythymidine monophosphates [75]; deoxycytidine and deoxythymidine [76]
• Deoxycytidine or tetrahydrouridine [77]
• Deoxyguanosine [77
Tk2H126N/ H126N mouse model; patients with TK2 deficiency
• thymidine-induced mtDNA depleted cells and Tymp/Upp1 knockout murine model of MNGIE
• dGK deficient human fibroblasts 
Cell replacement therapies • Erythrocyte-encapsulated thymidine phsphorylase [38]
• Allogenic HSCT [34]
• Liver transplantation [78
• Patients with MNGIE
• Patients with MNGIE
• One patient with EE 
Gene therapy • AAV-mediated gene therapy [35,79–82]
• CRIPR/Cas9 [84]
• Allotopic expression of mtDNA encoded proteins [85–88
Ant1−/- mouse model; Harlequin mouse; mouse model of EE; mouse model of MNGIE; mpv17 knockout mouse model
• iPS cell model of CoQ10 deficiency (c.483G>C in COQ4 gene)
• Human fibroblasts (mutations in ND1, ND4, and ATP6 genes) and a mouse model of LHON 
Shifting heteroplasmy • Mitochondrial-targeted restriction endonucleases [89,92–97,99]
• mZFNs [90,98]
• TALENs [91,99
• Cybrids (m.8993T>C)
• Artificial mammalian oocytes and the NZB/BALB heteroplasmic mouse model
• Cybrids (m.8993T>C, mtDNA common deletion)
• Cybrids (mtDNA common deletion, m. 14459G>A)
• Artificial mammalian oocytes and the NZB/BALB heteroplasmic mouse model 
Stabilizing mutant tRNAs • Overexpressing cognate and noncognate aminoacyl mt-tRNA synthetase [100–105In vitro yeast and human cell line models 
StrategyMethodModel
Nontailored 
Activation of mitochondrial biogenesis • Nicotinamide riboside and PARP1 [47,48]
• AICAR [49]
• Bezafibrate [49–54]
• Resveratrol [55–57]
• Retinoic acid [58]
• Endurance exercise [59–62
Sco2 knockout/knockin mouse, Deletor mouse model
• Patients fibroblasts and mouse models of COX deficiency
• Patients fibroblasts, cybrids, mouse models of COX deficiency, Deletor mouse model
In vitro models, Drosophila models, human fibroblasts
• Cybrids (m.3243A>G)
• Mouse models of COX deficiency, mtDNA mutator mice, patients with MDs 
Regulating mitophagy and mitochondrial dynamics • Rapamycin [65,66In vitro models and Ndufs4−/− mouse model of Leigh syndrome; Tk2H126N/ H126N mouse model 
Bypassing OXPHOS blocks • Ndi1 (Complex I defect) [67,68] and AOX (Complex III and IV defects) [69,70In vitro models and Drosophila models 
Mitochondrial replacement therapy • Oocyte nuclear genetic material transfer [71• Nonhuman primates; healthy subjects and patients with mitochondrial diseases 
Chronic hypoxia • Genetic or small molecule activation of the hypoxia response [72]
• Moderate environmental hypoxia [72
• Cultured cells and zebrafish models
Ndufs4−/− mouse model of Leigh syndrome 
Disease-tailored 
Scavenging of specific toxic compounds • N-acetyl cysteine and metronidazole [73]
• Hemodialysis [74
Ethe1−/− mouse model of EE; patients with EE
• Patients with MNGIE 
Supplementation of nucleotides/nucleosides • Deoxycytidine and deoxythymidine monophosphates [75]; deoxycytidine and deoxythymidine [76]
• Deoxycytidine or tetrahydrouridine [77]
• Deoxyguanosine [77
Tk2H126N/ H126N mouse model; patients with TK2 deficiency
• thymidine-induced mtDNA depleted cells and Tymp/Upp1 knockout murine model of MNGIE
• dGK deficient human fibroblasts 
Cell replacement therapies • Erythrocyte-encapsulated thymidine phsphorylase [38]
• Allogenic HSCT [34]
• Liver transplantation [78
• Patients with MNGIE
• Patients with MNGIE
• One patient with EE 
Gene therapy • AAV-mediated gene therapy [35,79–82]
• CRIPR/Cas9 [84]
• Allotopic expression of mtDNA encoded proteins [85–88
Ant1−/- mouse model; Harlequin mouse; mouse model of EE; mouse model of MNGIE; mpv17 knockout mouse model
• iPS cell model of CoQ10 deficiency (c.483G>C in COQ4 gene)
• Human fibroblasts (mutations in ND1, ND4, and ATP6 genes) and a mouse model of LHON 
Shifting heteroplasmy • Mitochondrial-targeted restriction endonucleases [89,92–97,99]
• mZFNs [90,98]
• TALENs [91,99
• Cybrids (m.8993T>C)
• Artificial mammalian oocytes and the NZB/BALB heteroplasmic mouse model
• Cybrids (m.8993T>C, mtDNA common deletion)
• Cybrids (mtDNA common deletion, m. 14459G>A)
• Artificial mammalian oocytes and the NZB/BALB heteroplasmic mouse model 
Stabilizing mutant tRNAs • Overexpressing cognate and noncognate aminoacyl mt-tRNA synthetase [100–105In vitro yeast and human cell line models 

Abbreviations: CRISPR, clustered regularly interspaced palindromic repeat; EE, ethylmalonic encephalopathy; HSCT, hematopoietic stem cell transplantation; iPS, induced pluripotent stem; MNGIE, mitochondrial neurogastrointestinal encephalomyopathy; TALEN, transcription activator-like effectors nucleases; ZFN, zinc finger endonuclease.

Activation of mitochondrial biogenesis

Energy failure is a hallmark of mitochondrial diseases and various therapeutic interventions have been used to stimulate mitochondrial biogenesis. Although these interventions do not rectify the underlying cause of the disease, increasing mitochondrial mass may increase energy production, thus ameliorating the phenotype. There is increasing evidence in in vitro studies and animal models that increased mitochondrial biogenesis might be beneficial in many mitochondrial diseases. Interestingly, a recent observation suggested that increased mitochondrial content influences incomplete penetrance in LHON patients and that mitochondrial biogenesis could be used as a therapeutic strategy in this group of patients [44]. The biological pathway that controls mitochondrial biogenesis is complex and relies mostly on the PPARγ coactivator 1α (PGC1α). PGC1α interacts with several transcription factors, including nuclear respiratory factors 1 and 2 (NRF1 and NRF2) and PPAR α, β, and γ. Once activated, NRFs increases the transcription of OXPHOS genes and PPARs increase the expression of genes related to fatty acid oxidation (FAO) [45]. Besides, PGC1α activity is increased by deacetylation and phosphorylation. Importantly, two enzymes responsible for these modifications, deacetylation by Sirt1 and phosphorylation by AMPK, can be modulated by drugs [46] and have been tested in preclinical models. Different agents used with this purpose are listed below.

Sirt1 is a nuclear deacetylase that utilizes oxidized nicotinamide adenine dinucleotide (NAD+) to deacetylate residues of acetyl-lysine in proteins. Notably, Sirt1 is activated by increased cellular levels of NAD+. This increase can be achieved by providing NAD precursor, such as nicotinamide riboside, or inhibiting NAD consuming enzymes, such as poly(ADP) ribosylpolymerase 1 (PARP1). These approaches have shown beneficial effects in animal models of mitochondrial myopathies [47,48].

5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an adenosine monophosphate analog, is an agonist of adenosine monophosphate-activated protein kinase (AMPK) that has been used to increase the respiratory chain complex activities in three mouse models of cytochrome c oxidase (COX) deficiency (Surf−/−, Sco2KOKI, and ACTA-Cox15−/−) with striking improvement of motor performances only in the Sco2 model [49].

Bezafibrate, a pan-PPAR activator, was tested in fibroblasts of patients with heterogeneous mitochondrial diseases and stimulated PGC1α and improved mitochondrial respiratory chain defects [50]. These findings were subsequently buttressed by in vivo studies in mouse models of COX-deficiency [51,52]. However, studies on other mouse models did not show induction of mitochondrial biogenesis or increased mitochondrial respiratory chain enzyme activities [49,53,54].

Resveratrol has also been described as an activator of mitochondrial biogenesis in animal models and in human fibroblasts [55,56], although its mechanism of action is still unknown and it did not appear to increase OXPHOS activities in another study on human fibroblasts [57].

Retinoic acid has been used to stimulate the retinoid X receptor-α (RXRα) in cybrid cells containing the m.3243A>G mutation, ameliorating the respiratory chain defect [58].

Endurance training has also been used as an activator of mitochondrial biogenesis and has been reported to be beneficial and safe in the mitochondrial DNA (mtDNA) mutator mice [59], and in patients with mitochondrial diseases [60,61]. Endurance training seems able to regulate not only PGC1α but also PGC1β, AMPK, and the hypoxia inducible factors (HIFs) [62].

Regulating mitophagy and mitochondrial dynamics

Mitophagy is the selective elimination of dysfunctional mitochondria, a physiological process fundamental for maintaining normal mitochondrial function [63,64]. This process is under the regulation of various pathways. One way of targeting mitophagy is via mTOR inhibition, which can be achieved by rapamycin. This approach has been investigated in a mouse model of Leigh syndrome (Ndufs4−/−) [65] and in a knockin mouse model of mtDNA depletion syndrome [66] and appeared to ameliorate the clinical phenotype and life span of the treated mice, although the oxidative phosphorylation (OXPHOS) defects were not rescued and mechanism of action remains uncertain.

Bypassing OXPHOS defects

The use of single-peptide enzymes derived from yeast or low eukaryotes to bypass mitochondrial respiratory chain defects has been tested in in vitro and in vivo models. In particular, Ndi1 substitutes complex I in yeast and transfers electron to CoQ, without pumping protons across the membrane. AOX is present in lower eukaryotes and bypasses complex III and IV by accepting electrons from CoQ. Expression of these enzymes has been used to bypass complex I deficiency [67,68] and complex III–IV deficiencies in human cells and Drosophila [69,70], but not in mammals in vivo.

Mitochondrial replacement therapy

As discussed in the accompanying Essays in Biochemistry article “Advances in methods for reducing mitochondrial DNA disease by replacing or manipulating the mitochondrial genome” by Rai et al. [71], replacement of mutant mtDNA in oocytes or single-cell embryos by mitochondrial replacement therapy is a novel method to prevent maternal transmission of mtDNA mutations (for further details, please the Rai et al. review [71]).

Hypoxia

An innovative approach to treatment of mitochondrial diseases is activation of the hypoxic response pathway [72]. Through a genome-wide Cas9-mediated screen of pharmacologically induced complex III deficiency, inhibition of the Von Hippel–Lindau (VHL) factor was identified as the most effector suppressor of the mitochondrial dysfunction. Because VHL negatively regulates HIFs, down-regulation of VHL activates the HIF transcriptional response, which, in part, shifts cellular bioenergetic reliance on mitochondrial OXPHOS. Genetic and pharmacological activation of the HIF pathway in zebrafish models as well as hypoxic treatment in the Ndusf4 knockout mouse model of Leigh syndrome provided further in vivo support for this therapeutic strategy.

Scavenging of specific toxic compounds

Ethylmalonic encephalopathy (EE) is a devastating disorder of infancy due to ETHE1 mutations. ETHE1 encodes a mitochondrial sulfur dioxygenase (SDO) involved in the elimination of H2S. Accumulation of H2S, produced by the catabolism of amino acids and by the anaerobic flora of the intestine, is toxic and leads to inhibition of COX activity and to endothelial damage. N-acetyl cysteine (NAC) is a precursor of glutathione and can be used to buffer intracellular H2S. Metronidazole is an intestinal antibiotic active against anaerobic bacteria that produce H2S. The use of metronidazole and NAC in a mouse model of ethylmalonic encephalopathy (Ethe1−/−) prolonged the life span and ameliorated the clinical phenotype of this model. Moreover, the administration of these compounds in a cohort of patients with EE was able to improve some of the clinical features of the disease [73]. This treatment has not been tested in clinical trials yet.

In MNGIE, hemodialysis has been used to attempt to remove toxic deoxynucleosides but was not effective in decreasing thymidine or deoxyuridine levels [74].

Nucleoside substrate enhancement and nucleotide bypass therapies

Supplementation of deoxyribonucleotides and deoxyribonucleosides has been exploited in in vitro and in vivo models of mitochondrial deoxynucleotide triphosphate (dNTP) pool unbalance. Mitochondrial dNTP pool unbalance causes mtDNA instability and consequent mtDNA depletion, multiple deletions, and point mutations. Different enzymes are involved in the maintenance of dNTP pools, such as thymidine kinase 2 (TK2), deoxyguanosine kinase (dGK), and thymidine phosphorylase (TP).

TK2 is a mitochondrial matrix protein that phosphorylates thymidine and deoxycytidine nucleosides to generate deoxythymidine and deoxycytidine monophosphate (dTMP, dCMP), which are then converted into dNTPs, fundamental for mtDNA synthesis. Recessive mutations in TK2 gene cause dNTP pool unbalance and mtDNA instability. The consequent clinical phenotypes range from a severe infantile neuromuscular form to adult-onset chronic progressive external ophthalmoplegia. Promising results were obtained in a Tk2 knockin mouse model (Tk2H126N/H126N) with oral administration of deoxycytidine and deoxythymidine monophosphates and subsequently deoxycytidine and deoxythymidine; both treatments increased mtDNA levels and mitochondrial respiratory chain enzyme activities, and prolonged the life span of the homozygous mutant mice [75,76].

Depletion of mtDNA has been corrected in vivo in a TppUpp double knockout mouse model of MNGIE disease by administrating deoxycytidine or tetrahydrouridine [77]. In the same study, the addition of deoxycytidine and tetrahydrouridine to a cell model of MNGIE disease (dThd-induced mtDNA depleted fibroblasts) was also able to prevent mtDNA depletion. Depletion of mtDNA was also corrected in dGK deficient human fibroblasts by adding deoxyguanosine [77].

Cell replacement therapies

Cell replacement therapies such as erythrocyte-encapsulated thymidine phosphorylase [38] as well as AHSCT and liver transplantation to restore TP activity in MNGIE are described above.

Liver transplantation has been recently performed in an infant with EE due to ETHE1 mutations [78]. The patient showed progressive improvement of the neurological function and normalization of the biochemical abnormalities. Liver transplantation can replace the deficient enzyme and clear the toxic compounds that accumulate in this disorder, constituting a feasible therapeutic option in patients with EE.

Gene therapy

Nuclear DNA defects

Adeno-associated viruses (AAVs) are good candidates as viral vectors for gene therapy, given their low risk of insertional mutagenesis, and a long-term persistence in cells. AAVs are currently the most widely used in preclinical approaches. AAV-mediated gene therapy has been performed in different mouse models of nuclear-encoded mitochondrial diseases. The first animal model treated with muscle injections of AAV2 was an Ant1−/- mouse [79]. In another approach, AAV2 vector targeted to retina was used to express AIF1 in the eye of the Harlequin mouse and restore complex I deficiency [80]. A liver specific AAV2/8 serotype with ETHE1 was applied to a mouse model of EE and dramatically improve the clinical course and the biochemical abnormalities of mutant mice [81]. This study demonstrated that restoring ETHE1 activity selectively in the liver was sufficient to correct the enzymatic defect and led to the hypothesis that liver transplant could be used in patients with EE. Similarly, the same hepatotropic vector was used in a mouse model of MNGIE and was proven successful as noted previously [77]. AAV2,8 vector was also used to express the wild-type MPV17 protein in a Mpv17 knockout mouse model of mtDNA depletion and hepatocerebral syndrome [82]. The vector was able to rescue the mtDNA depletion and prevent liver steatosis induced by ketogenic diet in this model.

Another approach using the CRISPR/Cas9 system, an endonuclease-based system, has been reported to rescue mitochondrial and skeletal muscle impairment in an iPS cell model of CoQ10 deficiency due to a mutation in the COQ4 gene [83,84].

mtDNA defects

Even more challenging is delivering gene therapy into mitochondria. One attempted approach is to allotopically express recombinant mtDNA encoded proteins containing a mitochondrial targeting sequence (MTS) in the nucleus. This approach has been tried in fibroblasts carrying mutations in ND1, ND4, and ATP6 genes [85–87] and in an animal model of LHON [88]. Despite the controversial preclinical results, clinical trials have started in patients with LHON (NCT01267422, NCT02064569, NCT02161380) (see “clinical trials”).

Shifting mtDNA mutation heteroplasmy

Pathogenic mtDNA mutations are usually heteroplasmic, requiring a minimum critical mutation load to cause mitochondrial dysfunction. Shifting heteroplasmic levels in order to reduce the amount of mutated DNA below this threshold, therefore, has been used as a therapeutic approach. This can be achieved with different techniques: mitochondrial-targeted restriction endonucleases [89], zinc finger endonucleases (ZFNs) [90], transcription activator-like effectors nucleases (TALENs) [91], and CRISPR (clustered regularly interspaced palindromic repeat)/Cas9. Restriction endonuclease SmaI has been used in cybrids carrying the m.8399T>G mutation and was able to reduce the mutation load and increase ATP levels [92,93]. Restriction endonucleases have been exploited also in heteroplasmic mouse models, using AAV vectors, with promising results [94–97]. The major limitation of this approach is the uncommon generation of a suitable restriction site by the mtDNA mutation. The introduction of programmable nucleases such as ZFNs and TALENs overcomes this limitation. ZFNs are engineered mitochondrially targeted heterodimeric zinc finger peptides conjugated to the nucleolytic domain of the type IIs restriction enzyme FokI. Each zinc finger domain recognizes three nucleotides, so arranging zinc finger modules appropriately allow for recognition of virtually any DNA sequence. Expression of mtZFNs in cybrids was able to reduce the mutant mtDNA and restore mitochondrial function [98]. TALENs also work as heterodimers, requiring two monomers to bind close DNA sequence in order to allow the FokI nuclease to dimerize and cleave DNA, as for the mtZFNs. Reengineered TALENs targeted to different mtDNA point mutations and deletion (MitoTALENs) were able to permanently reduce the mutation load in patient-derived cells [91]. A possible limitation to the use of MitoTALENs in clinical practice is the small packaging capacity (generally <5 kb) of AAV vectors. Moreover, the risk of a rapid reduction in mtDNA copy numbers of inducing a potential mtDNA depletion syndrome remains a limitation for the potential clinical application of these approaches. Mitochondria-targeted restriction endonucleases and TALENs have also been used in the selective elimination of mtDNA mutations in the germline of the heteroplasmic mouse model and artificial mammalian oocytes as a potential approach for preventing transmission of mtDNA mutations [99].

Stabilizing mutant mitochondrial tRNAs

The majority of the mtDNA mutations are localized to tRNA genes. It is not surprising therefore that several therapies have been targeting mt-tRNAs. In particular, tRNA synthetases are enzymes that catalyze the attachment of amino acids to their cognate tRNA during protein synthesis. Many studies indicated that overexpressing cognate and noncognate aminoacyl mt-tRNA synthetases, or their fragments, could stabilize mt-tRNAs and attenuate the detrimental effect of the mutation in yeast and human cell lines [100–105].

Remarkable progress has been achieved in the three decades of mitochondrial medicine since the identification of the first mtDNA mutations. Many potential therapeutic approaches for mitochondrial diseases have been proposed and are now at different stages of development. Translating preclinical studies to bedside remains challenging and well-controlled clinical trials of high quality are necessary to define the efficacy of potential therapies already in use and to develop novel drugs [6]. Based on the knowledge acquired with the previous studies, these future trials may overcome the challenges posed by this heterogeneous group of disorders in the context of multicenter collaborations, by selecting numerous subgroups of homogeneous patients and by selecting outcome measures that are objective and relevant to patient care and quality of life. Clearly, there are important unmet needs for evidence-based guidelines in the treatment of mitochondrial patients and the development of more effective therapies. The emerging therapies provide exciting promise for clinically meaningful treatments for mitochondrial diseases.

  • Although dietary supplements are frequently used by patients with mitochondrial disease, efficacy in most individuals is uncertain.

  • Exercise therapy has been demonstrated to be beneficial for mitochondrial diseases.

  • Emerging therapies for mitochondrial disease include nontailored therapies that can be applied across multiple mitochondrial diseases: (1) activation of mitochondrial biogenesis; (2) regulation of mitophagy and mitochondrial dynamics; (3) bypass of OXPHOS defects; (4) mitochondrial replacement therapy (MRT); and (5) hypoxia.

  • New tailored therapeutic strategies for mitochondrial diseases include: (1) scavenging of specific toxic compounds; (2) supplementation of deoxynucleosides and deoxynucleotides; (3) cell replacement therapies; (4) gene therapy; (5) shifting mtDNA mutation heteroplasmy; and (6) stabilizing mutant mitochondrial tRNAs.

The authors thank the patients and their families who have collaborated with the clinical therapy studies on mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) and thymidine kinase 2 (TK2) deficiency. With special thanks to JJMP and AE and their families for their contributions to TK2 deficiency research.

C.M.Q and M.H. are supported by NIH grant [P01 HD080642]. M.H. is also supported by U54 NS NS078059 North American Mitochondrial Diseases Consortium (NAMDC), the Marriott Mitochondrial Disorders Clinical Research Network; the Nicholas Nunno Fund; the Mileti Fund; the Bernard and Anne Spitzer Fund; and the Finn Foundation. V.E. is supported by the NAMDC fellowship program and has also been supported by L`Oréal- UNESCO for Women in Science 2016, Italy. NAMDC is part of the Rare Diseases Clinical Research Network (RDCRN), an initiative of the Office of Rare Diseases Research (ORDR), National Center for Advanced Translational Sciences (NCATS), National Institutes of Health (NIH). NAMDC is jointly funded through an NIH U54 grant mechanism by NCATS, the National Institute of Neurological Disorders and Stroke (NINDS), the Eunice Kennedy Shriver National Institute of Child Health and Development (NICHD), and the Office of Dietary Supplements (ODS). CMQ is also funded by the Department of Defense and the National Ataxia Foundation.

M.H. is a co-inventor on patent applications and holds Orphan Drug Designation (ODD) from the Food and Drug Administration and Rare Pediatric Disease Designation for deoxynucleoside therapy for mitochondrial DNA depletion syndrome including TK2 deficiency. The patent, RDD, and OPDs have been licensed via the Columbia Technology Ventures office to Modis Pharmaceuticals. M.H. and Columbia University Medical Center (CUMC) have filed patent applications covering the potential use of deoxynucleoside treatment for TK2 deficiency in humans. CUMC has licensed pending patent applications related to the technology to Modis Pharmaceuticals, Inc. and CUMC may be eligible to receive payments related to the development and commercialization of the technology. Any potential licensing fees earned will be paid to CUMC and are shared with MH through CUMC distribution policy. M.H. is a paid consultant to Modis Pharmaceutical, Inc. The other authors declare no conflicts of interest.

AAV

adeno-associated virus

AHSCT

allogeneic hematopoietic stem cell transplantation

AICAR

5-aminoimidazole-4-carboxamide ribonucleotide

AMPK

adenosine monophosphate-activated protein kinase

CoQ10

coenzyme Q10

COX

cytochrome c oxidase

CRISPR

clustered regularly interspaced palindromic repeat

DCA

dichloroacetate

dCMP

deoxycytidine monophosphate

DGK

deoxyguanosine kinase

dNTP

deoxynucleotide triphosphate

dTMP

deoxythymidine monophosphate

EE

ethylmalonic encephalopathy

FA

friedreich ataxia

HIF

hypoxia inducible factor

IV

intravenous

LHON

Leber hereditary optic neuropathy

MELAS

mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes

MIDD

maternally inherited diabetes-deafness syndrome

MNGIE

mitochondrial neurogastrointestinal encephalomyopathy

mtDNA

mitochondrial DNA

MTS

mitochondrial targeting sequence

NAC

N-acetyl cysteine

NAD

nicotinamide adenine dinucleotide

NAMDC

North American Mitochondrial Disease Consortium

NO

nitric oxide

NRF1

nuclear respiratory factors 1

NRF2

nuclear respiratory factors 2

OXPHOS

oxidative phosphorylation

PARP1

poly(ADP) ribosylpolymerase 1

PDC

pyruvate dehydrogenase complex

PO

per oral

PPAR

peroxisomal proliferator-activated receptors

PTP

permeability transition pore

ROS

reactive oxygen species

SDO

sulfur dioxygenase

TALEN

transcription activator-like effectors nuclease

TK2

thymidine kinase 2

TP

thymidine phosphorylase

VHL

Von Hippel–Lindau

ZFN

zinc finger endonuclease

1.
Pfeffer
G.
,
Majamaa
K.
,
Turnbull
D.M.
,
Thorburn
D.
and
Chinnery
P.F.
(
2012
)
Treatment for mitochondrial disorders
.
Cochrane Database Syst. Rev.
CD004426
[PubMed]
2.
Gorman
G.S.
,
Chinnery
P.F.
,
DiMauro
S.
,
Hirano
M.
,
Koga
Y.
,
McFarland
R.
et al.
(
2016
)
Mitochondrial diseases
.
Nat. Rev. Dis. Primers
2
,
16080
[PubMed]
3.
Quinzii
C.M.
,
Emmanuele
V.
and
Hirano
M.
(
2014
)
Clinical presentations of coenzyme q10 deficiency syndrome
.
Mol. Syndromol.
5
,
141
146
[PubMed]
4.
Emmanuele
V.
,
Lopez
L.C.
,
Berardo
A.
,
Naini
A.
,
Tadesse
S.
,
Wen
B.
et al.
(
2012
)
Heterogeneity of coenzyme Q10 deficiency: patient study and literature review
.
Arch. Neurol.
69
,
978
983
[PubMed]
5.
Parikh
S.
,
Goldstein
A.
,
Koenig
M.K.
,
Scaglia
F.
,
Enns
G.M.
,
Saneto
R.
et al.
(
2015
)
Diagnosis and management of mitochondrial disease: a consensus statement from the Mitochondrial Medicine Society
.
Genet. Med.
17
,
689
701
[PubMed]
6.
Pfeffer
G.
,
Horvath
R.
,
Klopstock
T.
,
Mootha
V.K.
,
Suomalainen
A.
,
Koene
S.
et al.
(
2013
)
New treatments for mitochondrial disease-no time to drop our standards
.
Nat. Rev. Neurol.
9
,
474
481
[PubMed]
7.
Schon
E.A.
,
DiMauro
S.
,
Hirano
M.
and
Gilkerson
R.W.
(
2010
)
Therapeutic prospects for mitochondrial disease
.
Trends Mol. Med.
16
,
268
276
[PubMed]
8.
Schriner
S.E.
,
Linford
N.J.
,
Martin
G.M.
,
Treuting
P.
,
Ogburn
C.E.
,
Emond
M.
et al.
(
2005
)
Extension of murine life span by overexpression of catalase targeted to mitochondria
.
Science
308
,
1909
1911
[PubMed]
9.
Blanchet
L.
,
Smeitink
J.A.
,
van Emst-de Vries
S.E.
,
Vogels
C.
,
Pellegrini
M.
,
Jonckheere
A.I.
et al.
(
2015
)
Quantifying small molecule phenotypic effects using mitochondrial morpho-functional fingerprinting and machine learning
.
Sci. Rep.
5
,
8035
[PubMed]
10.
Karaa
A.
,
Kriger
J.
,
Grier
J.
,
Holbert
A.
,
Thompson
J.L.
,
Parikh
S.
et al.
(
2016
)
Mitochondrial disease patients’ perception of dietary supplements’ use
.
Mol. Genet. Metab.
119
,
100
108
[PubMed]
11.
Koenig
M.K.
,
Emrick
L.
,
Karaa
A.
,
Korson
M.
,
Scaglia
F.
,
Parikh
S.
et al.
(
2016
)
Recommendations for the management of strokelike episodes in patients with mitochondrial encephalomyopathy, lactic acidosis, and strokelike episodes
.
JAMA Neurol.
73
,
591
594
[PubMed]
12.
Voet
N.B.
,
van der Kooi
E.L.
,
Riphagen
I.I.
,
Lindeman
E.
,
van Engelen
B.G.
and
Geurts
A.C.
(
2013
)
Strength training and aerobic exercise training for muscle disease
.
Cochrane Database Syst. Rev.
CD003907
[PubMed]
13.
Tarnopolsky
M.A.
(
2014
)
Exercise as a therapeutic strategy for primary mitochondrial cytopathies
.
J. Child Neurol.
29
,
1225
1234
[PubMed]
14.
Taivassalo
T.
,
Gardner
J.L.
,
Taylor
R.W.
,
Schaefer
A.M.
,
Newman
J.
,
Barron
M.J.
et al.
(
2006
)
Endurance training and detraining in mitochondrial myopathies due to single large-scale mtDNA deletions
.
Brain
129
,
3391
3401
[PubMed]
15.
Taivassalo
T.
,
Shoubridge
E.A.
,
Chen
J.
,
Kennaway
N.G.
,
DiMauro
S.
,
Arnold
D.L.
et al.
(
2001
)
Aerobic conditioning in patients with mitochondrial myopathies: physiological, biochemical, and genetic effects
.
Ann. Neurol.
50
,
133
141
[PubMed]
16.
Mariotti
C.
,
Solari
A.
,
Torta
D.
,
Marano
L.
,
Fiorentini
C.
and
Di Donato
S.
(
2003
)
Idebenone treatment in Friedreich patients: one-year-long randomized placebo-controlled trial
.
Neurology
60
,
1676
1679
[PubMed]
17.
Di Prospero
N.A.
,
Sumner
C.J.
,
Penzak
S.R.
,
Ravina
B.
,
Fischbeck
K.H.
and
Taylor
J.P.
(
2007
)
Safety, tolerability, and pharmacokinetics of high-dose idebenone in patients with Friedreich ataxia
.
Arch. Neurol.
64
,
803
808
[PubMed]
18.
Di Prospero
N.A.
,
Baker
A.
,
Jeffries
N.
and
Fischbeck
K.H.
(
2007
)
Neurological effects of high-dose idebenone in patients with Friedreich’s ataxia: a randomised, placebo-controlled trial
.
Lancet Neurol.
6
,
878
886
[PubMed]
19.
Meier
T.
,
Perlman
S.L.
,
Rummey
C.
,
Coppard
N.J.
and
Lynch
D.R.
(
2012
)
Assessment of neurological efficacy of idebenone in pediatric patients with Friedreich’s ataxia: data from a 6-month controlled study followed by a 12-month open-label extension study
.
J. Neurol.
259
,
284
291
[PubMed]
20.
Lynch
D.R.
,
Perlman
S.L.
and
Meier
T.
(
2010
)
A phase 3, double-blind, placebo-controlled trial of idebenone in friedreich ataxia
.
Arch. Neurol.
67
,
941
947
[PubMed]
21.
Beyrath
J.
,
Pellegrini
M.
,
Renkema
H.
,
Houben
L.
,
Pecheritsyna
S.
,
van Zandvoort
P.
et al.
(
2018
)
KH176 safeguards mitochondrial diseased cells from redox stress-induced cell death by interacting with the thioredoxin system/peroxiredoxin enzyme machinery
.
Sci. Rep.
8
,
6577
[PubMed]
22.
Koene
S.
,
Spaans
E.
,
Van Bortel
L.
,
Van Lancker
G.
,
Delafontaine
B.
,
Badilini
F.
et al.
(
2017
)
KH176 under development for rare mitochondrial disease: a first in man randomized controlled clinical trial in healthy male volunteers
.
Orphanet. J. Rare Dis.
12
,
163
[PubMed]
23.
El-Hattab
A.W.
,
Emrick
L.T.
,
Hsu
J.W.
,
Chanprasert
S.
,
Almannai
M.
,
Craigen
W.J.
et al.
(
2016
)
Impaired nitric oxide production in children with MELAS syndrome and the effect of arginine and citrulline supplementation
.
Mol. Genet. Metab.
117
,
407
412
[PubMed]
24.
Koga
Y.
,
Akita
Y.
,
Junko
N.
,
Yatsuga
S.
,
Povalko
N.
,
Fukiyama
R.
et al.
(
2006
)
Endothelial dysfunction in MELAS improved by L-arginine supplementation
.
Neurology
66
,
1766
1769
[PubMed]
25.
Koga
Y.
,
Akita
Y.
,
Nishioka
J.
,
Yatsuga
S.
,
Povalko
N.
,
Katayama
K.
et al.
(
2007
)
MELAS and L-arginine therapy
.
Mitochondrion
7
,
133
139
[PubMed]
26.
Rodan
L.H.
,
Wells
G.D.
,
Banks
L.
,
Thompson
S.
,
Schneiderman
J.E.
and
Tein
I.
(
2015
)
L-arginine affects aerobic capacity and muscle metabolism in MELAS (mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes) syndrome
.
PLoS One
10
,
e0127066
[PubMed]
27.
Koga
Y.
,
Akita
Y.
,
Nishioka
J.
,
Yatsuga
S.
,
Povalko
N.
,
Tanabe
Y.
et al.
(
2005
)
L-arginine improves the symptoms of strokelike episodes in MELAS
.
Neurology
64
,
710
712
[PubMed]
28.
Yu-Wai-Man
P.
and
Chinnery
P.F.
(
1993
)
Leber hereditary optic neuropathy
. In
GeneReviews
(
Pagon
R.A.
,
Adam
M.P.
,
Ardinger
H.H.
,
Wallace
S.E.
,
Amemiya
A.
,
Bean
L.J.H.
et al.
, eds),
GeneReviews
®,
Seattle (WA)
29.
Klopstock
T.
,
Yu-Wai-Man
P.
,
Dimitriadis
K.
,
Rouleau
J.
,
Heck
S.
,
Bailie
M.
et al.
(
2011
)
A randomized placebo-controlled trial of idebenone in Leber’s hereditary optic neuropathy
.
Brain
134
,
2677
2686
[PubMed]
30.
Klopstock
T.
,
Metz
G.
,
Yu-Wai-Man
P.
,
Buchner
B.
,
Gallenmuller
C.
,
Bailie
M.
et al.
(
2013
)
Persistence of the treatment effect of idebenone in Leber’s hereditary optic neuropathy
.
Brain
136
,
e230
[PubMed]
31.
Szeto
H.H.
(
2014
)
First-in-class cardiolipin-protective compound as a therapeutic agent to restore mitochondrial bioenergetics
.
Br. J. Pharmacol.
171
,
2029
2050
[PubMed]
32.
Wan
X.
,
Pei
H.
,
Zhao
M.J.
,
Yang
S.
,
Hu
W.K.
,
He
H.
et al.
(
2016
)
Efficacy and Safety of rAAV2-ND4 Treatment for Leber’s Hereditary Optic Neuropathy
.
Sci. Rep.
6
,
21587
[PubMed]
33.
Hirano
M.
(
1993
)
Mitochondrial neurogastrointestinal encephalopathy disease
. In (
Pagon
R.A.
,
Adam
M.P.
,
Ardinger
H.H.
,
Wallace
S.E.
,
Amemiya
A.
,
Bean
LJH
et al.
, eds),
GeneReviews®
,
Seattle (WA)
34.
Halter
J.P.
,
Michael
W.
,
Schupbach
M.
,
Mandel
H.
,
Casali
C.
,
Orchard
K.
et al.
(
2015
)
Allogeneic haematopoietic stem cell transplantation for mitochondrial neurogastrointestinal encephalomyopathy
.
Brain
138
,
2847
2858
[PubMed]
35.
Torres-Torronteras
J.
,
Viscomi
C.
,
Cabrera-Perez
R.
,
Camara
Y.
,
Di Meo
I.
,
Barquinero
J.
et al.
(
2014
)
Gene therapy using a liver-targeted AAV vector restores nucleoside and nucleotide homeostasis in a murine model of MNGIE
.
Mol. Ther.
22
,
901
907
[PubMed]
36.
Torres-Torronteras
J.
,
Gomez
A.
,
Eixarch
H.
,
Palenzuela
L.
,
Pizzorno
G.
,
Hirano
M.
et al.
(
2011
)
Hematopoietic gene therapy restores thymidine phosphorylase activity in a cell culture and a murine model of MNGIE
.
Gene Ther.
18
,
795
806
[PubMed]
37.
Yadak
R.
,
Cabrera-Perez
R.
,
Torres-Torronteras
J.
,
Bugiani
M.
,
Haeck
J.C.
,
Huston
M.W.
et al.
(
2018
)
Preclinical efficacy and safety evaluation of hematopoietic stem cell gene therapy in a mouse model of MNGIE
.
Mol. Ther. Methods Clin. Dev.
8
,
152
165
[PubMed]
38.
Bax
B.E.
,
Bain
M.D.
,
Scarpelli
M.
,
Filosto
M.
,
Tonin
P.
and
Moran
N.
(
2013
)
Clinical and biochemical improvements in a patient with MNGIE following enzyme replacement
.
Neurology
81
,
1269
1271
[PubMed]
39.
Enns
G.M.
,
Kinsman
S.L.
,
Perlman
S.L.
,
Spicer
K.M.
,
Abdenur
J.E.
,
Cohen
B.H.
et al.
(
2012
)
Initial experience in the treatment of inherited mitochondrial disease with EPI-743
.
Mol. Genet. Metab.
105
,
91
102
[PubMed]
40.
Blankenberg
F.G.
,
Kinsman
S.L.
,
Cohen
B.H.
,
Goris
M.L.
,
Spicer
K.M.
,
Perlman
S.L.
et al.
(
2012
)
Brain uptake of Tc99m-HMPAO correlates with clinical response to the novel redox modulating agent EPI-743 in patients with mitochondrial disease
.
Mol. Genet. Metab.
107
,
690
699
[PubMed]
41.
Martinelli
D.
,
Catteruccia
M.
,
Piemonte
F.
,
Pastore
A.
,
Tozzi
G.
,
Dionisi-Vici
C.
et al.
(
2012
)
EPI-743 reverses the progression of the pediatric mitochondrial disease–genetically defined Leigh Syndrome
.
Mol. Genet. Metab.
107
,
383
388
[PubMed]
42.
Sadun
A.A.
,
Chicani
C.F.
,
Ross-Cisneros
F.N.
,
Barboni
P.
,
Thoolen
M.
,
Shrader
W.D.
et al.
(
2012
)
Effect of EPI-743 on the clinical course of the mitochondrial disease Leber hereditary optic neuropathy
.
Arch. Neurol.
69
,
331
338
[PubMed]
43.
Viscomi
C.
(
2016
)
Toward a therapy for mitochondrial disease
.
Biochem. Soc. Trans.
44
,
1483
1490
[PubMed]
44.
Giordano
C.
,
Iommarini
L.
,
Giordano
L.
,
Maresca
A.
,
Pisano
A.
,
Valentino
M.L.
et al.
(
2014
)
Efficient mitochondrial biogenesis drives incomplete penetrance in Leber’s hereditary optic neuropathy
.
Brain
137
,
335
353
[PubMed]
45.
Scarpulla
R.C.
(
2008
)
Transcriptional paradigms in mammalian mitochondrial biogenesis and function
.
Physiol. Rev.
88
,
611
638
[PubMed]
46.
Puigserver
P.
and
Spiegelman
B.M.
(
2003
)
Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1 alpha): transcriptional coactivator and metabolic regulator
.
Endocr. Rev.
24
,
78
90
[PubMed]
47.
Cerutti
R.
,
Pirinen
E.
,
Lamperti
C.
,
Marchet
S.
,
Sauve
A.A.
,
Li
W.
et al.
(
2014
)
NAD(+)-dependent activation of Sirt1 corrects the phenotype in a mouse model of mitochondrial disease
.
Cell Metab.
19
,
1042
1049
[PubMed]
48.
Khan
N.A.
,
Auranen
M.
,
Paetau
I.
,
Pirinen
E.
,
Euro
L.
,
Forsstrom
S.
et al.
(
2014
)
Effective treatment of mitochondrial myopathy by nicotinamide riboside, a vitamin B3
.
EMBO Mol. Med.
6
,
721
731
[PubMed]
49.
Viscomi
C.
,
Bottani
E.
,
Civiletto
G.
,
Cerutti
R.
,
Moggio
M.
,
Fagiolari
G.
et al.
(
2011
)
In vivo correction of COX deficiency by activation of the AMPK/PGC-1alpha axis
.
Cell Metab.
14
,
80
90
[PubMed]
50.
Bastin
J.
,
Aubey
F.
,
Rotig
A.
,
Munnich
A.
and
Djouadi
F.
(
2008
)
Activation of peroxisome proliferator-activated receptor pathway stimulates the mitochondrial respiratory chain and can correct deficiencies in patients’ cells lacking its components
.
J. Clin. Endocrinol. Metab.
93
,
1433
1441
[PubMed]
51.
Noe
N.
,
Dillon
L.
,
Lellek
V.
,
Diaz
F.
,
Hida
A.
,
Moraes
C.T.
et al.
(
2013
)
Bezafibrate improves mitochondrial function in the CNS of a mouse model of mitochondrial encephalopathy
.
Mitochondrion
13
,
417
426
[PubMed]
52.
Hofer
A.
,
Noe
N.
,
Tischner
C.
,
Kladt
N.
,
Lellek
V.
,
Schauss
A.
et al.
(
2014
)
Defining the action spectrum of potential PGC-1alpha activators on a mitochondrial and cellular level in vivo
.
Hum. Mol. Genet.
23
,
2400
2415
[PubMed]
53.
Yatsuga
S.
and
Suomalainen
A.
(
2012
)
Effect of bezafibrate treatment on late-onset mitochondrial myopathy in mice
.
Hum. Mol. Genet.
21
,
526
535
[PubMed]
54.
Dillon
L.M.
,
Hida
A.
,
Garcia
S.
,
Prolla
T.A.
and
Moraes
C.T.
(
2012
)
Long-term bezafibrate treatment improves skin and spleen phenotypes of the mtDNA mutator mouse
.
PLoS One
7
,
e44335
[PubMed]
55.
Lopes Costa
A.
,
Le Bachelier
C.
,
Mathieu
L.
,
Rotig
A.
,
Boneh
A.
,
De Lonlay
P.
et al.
(
2014
)
Beneficial effects of resveratrol on respiratory chain defects in patients’ fibroblasts involve estrogen receptor and estrogen-related receptor alpha signaling
.
Hum. Mol. Genet.
23
,
2106
2119
[PubMed]
56.
Mizuguchi
Y.
,
Hatakeyama
H.
,
Sueoka
K.
,
Tanaka
M.
and
Goto
Y.I.
(
2017
)
Low dose resveratrol ameliorates mitochondrial respiratory dysfunction and enhances cellular reprogramming
.
Mitochondrion
[PubMed]
57.
De Paepe
B.
,
Vandemeulebroecke
K.
,
Smet
J.
,
Vanlander
A.
,
Seneca
S.
,
Lissens
W.
et al.
(
2014
)
Effect of resveratrol on cultured skin fibroblasts from patients with oxidative phosphorylation defects
.
Phytother. Res.
28
,
312
316
[PubMed]
58.
Chae
S.
,
Ahn
B.Y.
,
Byun
K.
,
Cho
Y.M.
,
Yu
M.H.
,
Lee
B.
et al.
(
2013
)
A systems approach for decoding mitochondrial retrograde signaling pathways
.
Sci. Signal
6
,
rs4
[PubMed]
59.
Safdar
A.
,
Bourgeois
J.M.
,
Ogborn
D.I.
,
Little
J.P.
,
Hettinga
B.P.
,
Akhtar
M.
et al.
(
2011
)
Endurance exercise rescues progeroid aging and induces systemic mitochondrial rejuvenation in mtDNA mutator mice
.
Proc. Natl. Acad. Sci. U.S.A.
108
,
4135
4140
60.
Jeppesen
T.D.
,
Duno
M.
,
Schwartz
M.
,
Krag
T.
,
Rafiq
J.
,
Wibrand
F.
et al.
(
2009
)
Short- and long-term effects of endurance training in patients with mitochondrial myopathy
.
Eur. J. Neurol.
16
,
1336
1339
[PubMed]
61.
Zeviani
M.
(
2008
)
Train, train, train! No pain, just gain
.
Brain
131
,
2809
2811
[PubMed]
62.
Rowe
G.C.
,
El-Khoury
R.
,
Patten
I.S.
,
Rustin
P.
and
Arany
Z.
(
2012
)
PGC-1alpha is dispensable for exercise-induced mitochondrial biogenesis in skeletal muscle
.
PLoS One
7
,
e41817
[PubMed]
63.
Kim
I.
,
Rodriguez-Enriquez
S.
and
Lemasters
J.J.
(
2007
)
Selective degradation of mitochondria by mitophagy
.
Arch. Biochem. Biophys.
462
,
245
253
[PubMed]
64.
Ashrafi
G.
and
Schwarz
T.L.
(
2013
)
The pathways of mitophagy for quality control and clearance of mitochondria
.
Cell Death Differ.
20
,
31
42
[PubMed]
65.
Johnson
S.C.
,
Yanos
M.E.
,
Kayser
E.B.
,
Quintana
A.
,
Sangesland
M.
,
Castanza
A.
et al.
(
2013
)
mTOR inhibition alleviates mitochondrial disease in a mouse model of Leigh syndrome
.
Science
342
,
1524
1528
[PubMed]
66.
Siegmund
S.E.
,
Yang
H.
,
Sharma
R.
,
Javors
M.
,
Skinner
O.
,
Mootha
V.
et al.
(
2017
)
Low-dose rapamycin extends lifespan in a mouse model of mtDNA depletion syndrome
.
Hum. Mol. Genet.
26
,
4588
4605
[PubMed]
67.
Perales-Clemente
E.
,
Bayona-Bafaluy
M.P.
,
Perez-Martos
A.
,
Barrientos
A.
,
Fernandez-Silva
P.
and
Enriquez
J.A.
(
2008
)
Restoration of electron transport without proton pumping in mammalian mitochondria
.
Proc. Natl. Acad. Sci. U.S.A.
105
,
18735
18739
68.
Sanz
A.
,
Soikkeli
M.
,
Portero-Otin
M.
,
Wilson
A.
,
Kemppainen
E.
,
McIlroy
G.
et al.
(
2010
)
Expression of the yeast NADH dehydrogenase Ndi1 in Drosophila confers increased lifespan independently of dietary restriction
.
Proc. Natl. Acad. Sci. U.S.A.
107
,
9105
9110
69.
Dassa
E.P.
,
Dufour
E.
,
Goncalves
S.
,
Paupe
V.
,
Hakkaart
G.A.
,
Jacobs
H.T.
et al.
(
2009
)
Expression of the alternative oxidase complements cytochrome c oxidase deficiency in human cells
.
EMBO Mol. Med.
1
,
30
36
[PubMed]
70.
Fernandez-Ayala
D.J.
,
Sanz
A.
,
Vartiainen
S.
,
Kemppainen
K.K.
,
Babusiak
M.
,
Mustalahti
E.
et al.
(
2009
)
Expression of the Ciona intestinalis alternative oxidase (AOX) in Drosophila complements defects in mitochondrial oxidative phosphorylation
.
Cell Metab.
9
,
449
460
[PubMed]
71.
Rai
K.P.
,
Craven
L.
,
Hoogewijs
K.
and
Russel
O.M.
,
Lightowlers
R.N
(
2018
)
Advances in methods for reducing mitochondrial DNA disease by replacing or manipulating the mitochondrial genome
.
Essays Biochem.
,
62
,
455
465
72.
Jain
I.H.
,
Zazzeron
L.
,
Goli
R.
,
Alexa
K.
,
Schatzman-Bone
S.
,
Dhillon
H.
et al.
(
2016
)
Hypoxia as a therapy for mitochondrial disease
.
Science
352
,
54
61
[PubMed]
73.
Viscomi
C.
,
Burlina
A.B.
,
Dweikat
I.
,
Savoiardo
M.
,
Lamperti
C.
,
Hildebrandt
T.
et al.
(
2010
)
Combined treatment with oral metronidazole and N-acetylcysteine is effective in ethylmalonic encephalopathy
.
Nat. Med.
16
,
869
871
[PubMed]
74.
Spinazzola
A.
,
Marti
R.
,
Nishino
I.
,
Andreu
A.L.
,
Naini
A.
,
Tadesse
S.
et al.
(
2002
)
Altered thymidine metabolism due to defects of thymidine phosphorylase
.
J. Biol. Chem.
277
,
4128
4133
[PubMed]
75.
Garone
C.
,
Garcia-Diaz
B.
,
Emmanuele
V.
,
Lopez
L.C.
,
Tadesse
S.
,
Akman
H.O.
et al.
(
2014
)
Deoxypyrimidine monophosphate bypass therapy for thymidine kinase 2 deficiency
.
EMBO Mol. Med.
6
,
1016
1027
[PubMed]
76.
Lopez-Gomez
C.
,
Levy
R.J.
,
Sanchez-Quintero
M.J.
,
Juanola-Falgarona
M.
,
Barca
E.
,
Garcia-Diaz
B.
et al.
(
2017
)
Deoxycytidine and deoxythymidine treatment for thymidine kinase 2 deficiency
.
Ann. Neurol.
81
,
641
652
[PubMed]
77.
Camara
Y.
,
Gonzalez-Vioque
E.
,
Scarpelli
M.
,
Torres-Torronteras
J.
,
Caballero
A.
,
Hirano
M.
et al.
(
2014
)
Administration of deoxyribonucleosides or inhibition of their catabolism as a pharmacological approach for mitochondrial DNA depletion syndrome
.
Hum. Mol. Genet.
23
,
2459
2467
[PubMed]
78.
Dionisi-Vici
C.
,
Diodato
D.
,
Torre
G.
,
Picca
S.
,
Pariante
R.
,
Giuseppe Picardo
S.
et al.
(
2016
)
Liver transplant in ethylmalonic encephalopathy: a new treatment for an otherwise fatal disease
.
Brain
139
,
1045
1051
[PubMed]
79.
Flierl
A.
,
Chen
Y.
,
Coskun
P.E.
,
Samulski
R.J.
and
Wallace
D.C.
(
2005
)
Adeno-associated virus-mediated gene transfer of the heart/muscle adenine nucleotide translocator (ANT) in mouse
.
Gene Ther.
12
,
570
578
[PubMed]
80.
Bouaita
A.
,
Augustin
S.
,
Lechauve
C.
,
Cwerman-Thibault
H.
,
Benit
P.
,
Simonutti
M.
et al.
(
2012
)
Downregulation of apoptosis-inducing factor in Harlequin mice induces progressive and severe optic atrophy which is durably prevented by AAV2-AIF1 gene therapy
.
Brain
135
,
35
52
[PubMed]
81.
Di Meo
I.
,
Auricchio
A.
,
Lamperti
C.
,
Burlina
A.
,
Viscomi
C.
and
Zeviani
M.
(
2012
)
Effective AAV-mediated gene therapy in a mouse model of ethylmalonic encephalopathy
.
EMBO Mol. Med.
4
,
1008
1014
[PubMed]
82.
Bottani
E.
,
Giordano
C.
,
Civiletto
G.
,
Di Meo
I.
,
Auricchio
A.
,
Ciusani
E.
et al.
(
2014
)
AAV-mediated liver-specific MPV17 expression restores mtDNA levels and prevents diet-induced liver failure
.
Mol. Ther.
22
,
10
17
[PubMed]
83.
Doudna
J.A.
and
Charpentier
E.
(
2014
)
Genome editing. The new frontier of genome engineering with CRISPR-Cas9
.
Science
346
,
1258096
[PubMed]
84.
Romero-Moya
D.
,
Castano
J.
,
Santos-Ocana
C.
,
Navas
P.
and
Menendez
P.
(
2017
)
Generation, genome edition and characterization of iPSC lines from a patient with coenzyme Q10 deficiency harboring a heterozygous mutation in COQ4 gene
.
Stem Cell Res.
24
,
144
147
[PubMed]
85.
Bonnet
C.
,
Kaltimbacher
V.
,
Ellouze
S.
,
Augustin
S.
,
Benit
P.
,
Forster
V.
et al.
(
2007
)
Allotopic mRNA localization to the mitochondrial surface rescues respiratory chain defects in fibroblasts harboring mitochondrial DNA mutations affecting complex I or v subunits
.
Rejuvenation Res.
10
,
127
144
[PubMed]
86.
Kaltimbacher
V.
,
Bonnet
C.
,
Lecoeuvre
G.
,
Forster
V.
,
Sahel
J.A.
and
Corral-Debrinski
M.
(
2006
)
mRNA localization to the mitochondrial surface allows the efficient translocation inside the organelle of a nuclear recoded ATP6 protein
.
RNA
12
,
1408
1417
[PubMed]
87.
Bonnet
C.
,
Augustin
S.
,
Ellouze
S.
,
Benit
P.
,
Bouaita
A.
,
Rustin
P.
et al.
(
2008
)
The optimized allotopic expression of ND1 or ND4 genes restores respiratory chain complex I activity in fibroblasts harboring mutations in these genes
.
Biochim. Biophys. Acta
1783
,
1707
1717
[PubMed]
88.
Ellouze
S.
,
Augustin
S.
,
Bouaita
A.
,
Bonnet
C.
,
Simonutti
M.
,
Forster
V.
et al.
(
2008
)
Optimized allotopic expression of the human mitochondrial ND4 prevents blindness in a rat model of mitochondrial dysfunction
.
Am. J. Hum. Genet.
83
,
373
387
[PubMed]
89.
Srivastava
S.
and
Moraes
C.T.
(
2001
)
Manipulating mitochondrial DNA heteroplasmy by a mitochondrially targeted restriction endonuclease
.
Hum. Mol. Genet.
10
,
3093
3099
[PubMed]
90.
Minczuk
M.
,
Papworth
M.A.
,
Miller
J.C.
,
Murphy
M.P.
and
Klug
A.
(
2008
)
Development of a single-chain, quasi-dimeric zinc-finger nuclease for the selective degradation of mutated human mitochondrial DNA
.
Nucleic Acids Res.
36
,
3926
3938
[PubMed]
91.
Bacman
S.R.
,
Williams
S.L.
,
Pinto
M.
,
Peralta
S.
and
Moraes
C.T.
(
2013
)
Specific elimination of mutant mitochondrial genomes in patient-derived cells by mitoTALENs
.
Nat. Med.
19
,
1111
1113
[PubMed]
92.
Tanaka
M.
,
Borgeld
H.J.
,
Zhang
J.
,
Muramatsu
S.
,
Gong
J.S.
,
Yoneda
M.
et al.
(
2002
)
Gene therapy for mitochondrial disease by delivering restriction endonuclease SmaI into mitochondria
.
J. Biomed. Sci.
9
,
534
541
[PubMed]
93.
Alexeyev
M.F.
,
Venediktova
N.
,
Pastukh
V.
,
Shokolenko
I.
,
Bonilla
G.
and
Wilson
G.L.
(
2008
)
Selective elimination of mutant mitochondrial genomes as therapeutic strategy for the treatment of NARP and MILS syndromes
.
Gene Ther.
15
,
516
523
[PubMed]
94.
Bayona-Bafaluy
M.P.
,
Blits
B.
,
Battersby
B.J.
,
Shoubridge
E.A.
and
Moraes
C.T.
(
2005
)
Rapid directional shift of mitochondrial DNA heteroplasmy in animal tissues by a mitochondrially targeted restriction endonuclease
.
Proc. Natl. Acad. Sci. U.S.A.
102
,
14392
14397
95.
Bacman
S.R.
,
Williams
S.L.
,
Hernandez
D.
and
Moraes
C.T.
(
2007
)
Modulating mtDNA heteroplasmy by mitochondria-targeted restriction endonucleases in a ‘differential multiple cleavage-site’ model
.
Gene Ther.
14
,
1309
1318
[PubMed]
96.
Bacman
S.R.
,
Williams
S.L.
,
Garcia
S.
and
Moraes
C.T.
(
2010
)
Organ-specific shifts in mtDNA heteroplasmy following systemic delivery of a mitochondria-targeted restriction endonuclease
.
Gene Ther.
17
,
713
720
[PubMed]
97.
Bacman
S.R.
,
Williams
S.L.
,
Duan
D.
and
Moraes
C.T.
(
2012
)
Manipulation of mtDNA heteroplasmy in all striated muscles of newborn mice by AAV9-mediated delivery of a mitochondria-targeted restriction endonuclease
.
Gene Ther.
19
,
1101
1106
[PubMed]
98.
Gammage
P.A.
,
Rorbach
J.
,
Vincent
A.I.
,
Rebar
E.J.
and
Minczuk
M.
(
2014
)
Mitochondrially targeted ZFNs for selective degradation of pathogenic mitochondrial genomes bearing large-scale deletions or point mutations
.
EMBO Mol. Med.
6
,
458
466
[PubMed]
99.
Reddy
P.
,
Ocampo
A.
,
Suzuki
K.
,
Luo
J.
,
Bacman
S.R.
,
Williams
S.L.
et al.
(
2015
)
Selective elimination of mitochondrial mutations in the germline by genome editing
.
Cell
161
,
459
469
[PubMed]
100.
De Luca
C.
,
Besagni
C.
,
Frontali
L.
,
Bolotin-Fukuhara
M.
and
Francisci
S.
(
2006
)
Mutations in yeast mt tRNAs: specific and general suppression by nuclear encoded tRNA interactors
.
Gene
377
,
169
176
[PubMed]
101.
De Luca
C.
,
Zhou
Y.
,
Montanari
A.
,
Morea
V.
,
Oliva
R.
,
Besagni
C.
et al.
(
2009
)
Can yeast be used to study mitochondrial diseases? Biolistic tRNA mutants for the analysis of mechanisms and suppressors
Mitochondrion
9
,
408
417
[PubMed]
102.
Li
R.
and
Guan
M.X.
(
2010
)
Human mitochondrial leucyl-tRNA synthetase corrects mitochondrial dysfunctions due to the tRNALeu(UUR) A3243G mutation, associated with mitochondrial encephalomyopathy, lactic acidosis, and stroke-like symptoms and diabetes
.
Mol. Cell. Biol.
30
,
2147
2154
[PubMed]
103.
Rorbach
J.
,
Yusoff
A.A.
,
Tuppen
H.
,
Abg-Kamaludin
D.P.
,
Chrzanowska-Lightowlers
Z.M.
,
Taylor
R.W.
et al.
(
2008
)
Overexpression of human mitochondrial valyl tRNA synthetase can partially restore levels of cognate mt-tRNAVal carrying the pathogenic C25U mutation
.
Nucleic Acids Res.
36
,
3065
3074
[PubMed]
104.
Perli
E.
,
Giordano
C.
,
Pisano
A.
,
Montanari
A.
,
Campese
A.F.
,
Reyes
A.
et al.
(
2014
)
The isolated carboxy-terminal domain of human mitochondrial leucyl-tRNA synthetase rescues the pathological phenotype of mitochondrial tRNA mutations in human cells
.
EMBO Mol. Med.
6
,
169
182
[PubMed]
105.
Hornig-Do
H.T.
,
Montanari
A.
,
Rozanska
A.
,
Tuppen
H.A.
,
Almalki
A.A.
,
Abg-Kamaludin
D.P.
et al.
(
2014
)
Human mitochondrial leucyl tRNA synthetase can suppress non cognate pathogenic mt-tRNA mutations
.
EMBO Mol. Med.
6
,
183
193
[PubMed]
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).