152
Views
16
CrossRef citations to date
0
Altmetric
Review

Mitochondrial diseases: advances and issues

, , , &
Pages 21-26 | Published online: 15 Feb 2017

Abstract

Mitochondrial diseases (MDs) are a clinically heterogeneous group of disorders caused by a dysfunction of the mitochondrial respiratory chain. They can be related to mutation of genes encoded using either nuclear DNA or mitochondrial DNA. The advent of next generation sequencing and whole exome sequencing in studying the molecular bases of MDs will bring about a revolution in the field of mitochondrial medicine, also opening the possibility of better defining pathogenic mechanisms and developing novel therapeutic approaches for these devastating disorders. The canonical rules of mitochondrial medicine remain milestones, but novel issues have been raised following the use of advanced diagnostic technologies. Rigorous validation of the novel mutations detected using deep sequencing in patients with suspected MD, and a clear definition of the natural history, outcome measures, and biomarkers that could be usefully adopted in clinical trials, are mandatory goals for the scientific community. Today, therapy is often inadequate and mostly palliative. However, important advances have been made in treating some clinical entities, eg, mitochondrial neuro-gastrointestinal encephalomyopathy, for which approaches using allogeneic hematopoietic stem cell transplantation, orthotopic liver transplantation, and carrier erythrocyte entrapped thymidine phosphorylase enzyme therapy have recently been developed. Promising new treatment methods are being identified so that researchers, clinicians, and patients can join forces to change the history of these untreatable disorders.

Introduction

Mitochondrial diseases (MDs) encompass a broad group of disorders that may affect children and adults and are among the most common inherited neuromuscular disorders with a minimum prevalence of around 1:5,000 live births.Citation1 They may be manifested in a tissue-specific or a multisystem manner.Citation2 Even though tissues with high energy demand, such as brain, muscle, and eye, are more frequently involved, patients’ phenotype could be extremely variegated and heterogeneous, largely but not solely depending on the classical “rules” of mitochondrial medicine: dual genetic control (nuclear DNA [nDNA] and mitochondrial DNA [mtDNA]), level of heteroplasmy (percentage of mutated DNA in single cells and tissues), tissue energy demand, maternal inheritance, and mitotic segregation.Citation2

MDs are usually strictly considered diseases caused by a biochemical defect of the respiratory chain (RC).Citation3 This definition has been progressively expanded during recent years in parallel with the growth of knowledge on the role of mitochondrial- and nuclear-derived proteins in mitochondrial functions and the role of nuclear genes in regulating mitochondrial homeostasis and functioning, now also including defects in the lipid milieu, in mitochondrial translation, and in mitochondrial fission and fusion.Citation3,Citation4

Last but not least, mitochondrial impairment plays an important role in the pathogenesis of many neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis, and a growing number of studies are being developed in this field.Citation5 Next generation sequencing/whole exome sequencing (NGS/WES) approaches will provide valuable information on the genes implicated in these disorders and could better define the impact of mitochondrial dysfunction on their pathogenesis.

A deeper knowledge of the mechanisms regulating mitochondrial function and interactions between mtDNA and nDNA in single cellular systems and different cell subtypes is very important for revealing the role of mitochondria in neurodegenerative processes, also with the aim of identifying novel and more specific therapies.

Diagnosing mitochondrial diseases

The mitochondrial genome remains of crucial importance in the pathogenesis of many MD, and it should be analyzed in every suspected MD in which there is no clear clinical and biochemical evidence of a nuclear origin of the disease.Citation6

Due to the specific “rules” of mitochondrial medicine, if pathogenic mtDNA variants are suspected, the tissue in which mtDNA is studied is decisive in achieving a correct diagnosis.Citation2 Although slightly invasive, muscle biopsy is the gold standard for the diagnostic workup in both children and adults because it enables us to obtain valuable morphological, biochemical, and molecular data.Citation3 Moreover, muscle is sometimes the only tissue in which the molecular defect is present, especially in adult patients with a pure myopathic phenotype.Citation7 On the one hand, muscle tissue could reveal the presence of single mtDNA deletions, classically found in sporadic cases of chronic progressive external ophthalmoplegia, or point mutation in transfer RNAs or in gene-encoding structural subunits of the RC complexes; on the other hand, a Southern blot analysis of mtDNA extracted from muscle tissue could show multiple deletions and/or depletion of mtDNA, referring to a mutation of the nDNA.

As alternatives to muscle, more available tissues for molecular analysis are urinary epithelium and skin fibroblasts that could also be easily used for experimental studies.Citation8

Diagnostic and therapeutic advances

There is currently a large availability of powerful diagnostic technologies, especially in the field of genetics. Studying thousands of patients’ genomes through NGS and WES approaches is going to revolutionize the diagnostic process in mitochondrial medicine, allowing us to obtain molecular diagnosis in a growing number of previously unresolved cases and revealing novel molecular changes and previously unknown genetic and pathogenic mechanisms.Citation9 For instance, mutations with variable penetrance, large-scale DNA rearrangements, and mutations in known genes with previously unreported “mitochondrial” phenotypes (ie, genes universally known to cause neurodegenerative disorders as SPG7 mutations, usually related to spastic paraplegia, can cause chronic external ophthalmoplegia) have been identified, as well as variants in genes encoding proteins the role of which in the mitochondrial function has been unknown for a long time (ie, MPV17, which has only recently been demonstrated to serve as a nonselective channel with a pore diameter of 1.8 nm that contributes to mitochondrial homeostasis under different conditions).Citation10Citation12

On the other hand, patients with RC enzyme deficiency suggesting MD may harbor pathogenic mutations in genes apparently not related to mitochondrial function, and therefore they ultimately could be affected by other types of disorder in which RC abnormalities may be secondary to various cellular processes, including neuromuscular transmission, calcium metabolism, and abnormal messenger RNA degradation.Citation13

Therapies for MD are still limited, and in most cases only targeted at symptoms. Traditionally, patients have been treated with different vitamins, cofactors, and nutritional supplements.Citation14 However, these “mitochondrial cocktails” do not have a major therapeutic impact on most MD.Citation15

The large advances made in recent years, through the development of molecular genetic techniques for understanding the molecular bases and pathophysiological processes underlying MDs, have given a new momentum to research on effective treatments, and a big effort in developing new therapeutic approaches is offering patients hope for changing the natural history of these devastating disorders.

Although extremely promising, therapies directed toward mtDNA-related disorders are very much at the preclinical stage, and they require the adoption of rigorous clinical trials in the clinical setting.Citation15

A promising therapeutic strategy is to force a shift in heteroplasmy, thereby reducing the ratio of mutant to wild-type genomes (heteroplasmic shifting), which can be obtained by selectively inhibiting the replication of mutant genomes.Citation16 Several research groups worldwide are particularly interested in techniques aimed at preventing the transmission of maternally inherited MD. Reproductive technologies designed to uncouple the inheritance of mtDNA from nDNA may enable affected women to have a genetically related child with a greatly reduced risk of mtDNA-related disease, although they do not guarantee prevention.Citation17

From a clinical perspective, therapies for nuclear gene-related enzyme defects are at a more advanced stage. In some cases, etiological therapies are focused on restoring or bypassing specific enzyme or cofactor defects.

MD caused by defects in Coenzyme Q10 biosynthesis could be effectively treated with oral coenzyme supplementation, and riboflavin supplementation has been found to be beneficial in adults with riboflavin transporter disorders.Citation18,Citation19

Stem cell therapy is being used in specific contexts, and gene therapy trials have been successfully evaluated in mouse models.Citation14,Citation20

To date, the most important advances have been made in treating mitochondrial neuro-gastrointestinal encephalomyopathy (MNGIE), one of the rarest MDs that may benefit from available therapies, potentially improving life expectancy in the affected patients.

Certainly, MNGIE can be considered a prototype of how it may be possible to treat a MD related to a specific enzyme deficiency.

This is a devastating mitochondrial disorder caused by deficiency of the thymidine phosphorylase (TP) enzyme with consequent accumulation of nucleosides and an imbalance in the mitochondrial nucleotides pool.Citation21 The clinical picture includes progressive gastrointestinal dysmotility, cachexia, ptosis and ophthalmoparesis, peripheral neuropathy, and diffuse leukoencephalopathy, which usually lead to death in early adulthood.Citation21,Citation22

Excesses of nucleosides should be cleared from MNGIE patients through a method that continuously removes or catabolizes the nucleosides, as they are also continuously produced. Attempts of this treatment strategy include: 1) enzyme replacement, through infusion of a stabilized and active form of TP protein; 2) cell replacement, either hematopoietic stem cells from adult donors and cord blood or organ transplantation; and 3) gene therapy.Citation23

Carrier erythrocyte entrapped TP enzyme therapy has been associated with a marked reduction of plasma and urine thymidine and deoxyuridine, with the advantage that encapsulation prevents the formation of neutralizing antibodies and maintains enzyme activity for the erythrocyte life span.Citation24 Although periodical infusions are needed, it should be considered a rescue or maintenance therapy for MNGIE patients.Citation24

Allogeneic hematopoietic stem cell transplantation is a well-defined treatment option for this condition.Citation25Citation27 A consensus conference has proposed a common approach to allogeneic hematopoietic stem cell transplantation in MNGIE based on standardization of the transplant protocol and the clinical and biochemical assessments useful in evaluating its safety and efficacy.Citation27 This approach, however, has serious limitations including the difficulty in obtaining suitable donors, the toxicity of the conditioning regimen, and the risk of graft failure and graft-vs-host disease. In addition, MNGIE patients are generally in poor medical condition at the time of the diagnosis.Citation28 As a consequence, this treatment is associated with high morbidity and mortality rates in this particular disorder.Citation26,Citation27

The use of autologous hematopoietic stem cells, genetically engineered to produce normal TYMP, would eliminate those risks, and also has the intrinsic advantage of inducing immune tolerance to the recombinant therapeutic transgene product.Citation29 Third generation lentiviral vectors were developed to express a codon-optimized TYMP transgene using the phosphoglycerate kinase promoter to provide high TYMP levels in cell lines, and proof-of-principle has been obtained in the MNGIE mouse model.Citation29

More recently, it has been reported that AAV2/8-mediated transfer of the human TYMP coding sequence under the control of a liver-specific promoter prevents the biochemical imbalances in a murine model of MNGIE, demonstrating that the use of AAV to direct TYMP expression in liver is feasible as a potentially safe gene therapy strategy for MNGIE.Citation30

Organ transplantation may represent a suitable alternative option for treating patients with MNGIE. Since the liver is the main organ for protein biosynthesis and transplantation success is estimated at 90% of cases, it has been proposed as a good source of TP, and MNGIE patients may likely benefit from orthotopic liver transplantation.Citation31,Citation32

MNGIE belongs to a subgroup of mtDNA depletion syndromes characterized by a reduction in the mtDNA copy number and consequent mitochondrial dysfunction in the affected tissues. These diseases are caused by mutations in specific genes that disrupt deoxy ribonucleotide metabolism, which ultimately leads to limited availability of one or more deoxy ribonucleoside triphosphates, and subsequent mtDNA depletion.Citation33 Experience and results in treating MNGIE patients could be extended to other diseases affecting similar pathways, in particular mtDNA depletion syndromes caused by defective nucleotide metabolism in which supplementation of appropriate precursors and/or administration of substances that inhibit their catabolism are good candidates for treatment strategies.Citation33

Current issues

Although our knowledge about mitochondrial medicine has greatly increased, many questions still remain that have no definite answers, and several difficulties in diagnostic and pathogenic interpretation are experienced daily by mitochondriologists all over the world, for both the mtDNA-related and nDNA-related diseases.

It is generally accepted that heteroplasmy is one of the markers of pathogenicity of an mtDNA mutation. However, homoplasmic mtDNA mutations may have been inappropriately considered as benign polymorphisms in the past, whereas it has been clearly demonstrated that they could cause both tissue-specific (ie, Leber’s inherited optic neuropathy) and multisystem disorders.Citation34

The widespread availability of complete mtDNA sequencing has revealed that mtDNA mutations are much more common in the general population than was previously thought and are found in between 0.5% and 1% of the population at >1% heteroplasmy levels.Citation35

These findings raised the question of the pathogenic threshold of heteroplasmy and stress the need to further elucidate the other possible factors that contribute to the pathogenesis of the disease.

Classification of MD remains difficult. Although genetic classification by nDNA and mtDNA mutations is obviously the most reliable way to classify these disorders, this approach raises several issues including the large number of cases in which the pathogenic variants remain to be identified, and the locus and allelic heterogeneity for which mutations in different gene loci may have the same phenotypic effect and, conversely, the same pathogenic variant may cause many different clinical phenotypes (ie, the classical m.3243A>G single nucleotide variant).Citation3

Nowadays, modern NGS and WES technologies permit researchers to extensively study nuclear genome, giving them the possibility to reach the correct diagnosis in many cases that were previously unsolved.Citation9 Yet, on the other hand, a massive parallel sequencing of the nDNA unmasks a large number of variations in different genes, and it could be hard to assign a pathogenic role to each variation in a specific context. Multiple in vitro studies on isolated mutant and wild-type proteins, mutant cell characterization, modeling in yeast or other in vivo systems, including different animal species, are necessary to validate novel mutations both in genes directly linked to mitochondrial function and in genes encoding proteins not targeted toward mitochondria and therefore with roles not obviously related to mitochondrial physiology.Citation9

Conclusion

The field of MD is highly dynamic and new scenarios arise every day. The improvement and the large availability of highly sensitive diagnostic technologies are increasing the number of patients with a confirmed molecular diagnosis, advancing understanding of the pathogenic mechanisms, and offering the possibility to design targeted and more specific treatments for MD.Citation36Citation43

International efforts to share data will facilitate this work, enabling different groups to confirm or refute putative novel pathogenic mutations as early as possible.

It remains crucially important for clinicians and researchers to have a close relationship in order to validate the results of both diagnostic and therapeutic studies.

The discovery of gene mutations linked to MD is providing clues as to how target therapies and detailed studies of genetically homogeneous clinical cohorts are required in order to best define the natural histories and to identify reliable biomarkers and features that change over a long observation period that could be used as clinical trial endpoints.

MNGIE is a prototypical example of how translational studies can reveal scientifically rational therapies. Certainly, overall survival and restoration of TP activity are main indicators of good clinical outcome during therapy. However, it is still difficult to define whether or not stabilization rather than an improvement of clinical symptoms is satisfactory.

The future looks brighter as the revolution in the diagnostic approach could prelude the revolution in the treatment options for MD.

Disclosure

All authors have read and agreed upon the contents of the paper. The authors report no conflicts of interest in this work.

References

  • SchaeferAMMcFarlandRBlakelyELPrevalence of mitochondrial DNA disease in adultsAnn Neurol200863353917886296
  • DiMauroSSchonEAMitochondrial respiratory-chain diseasesN Engl J Med20033482656266812826641
  • FilostoMMancusoMMitochondrial diseases: a nosological updateActa Neurol Scand200711521122117376118
  • VafaiSBMoothaVKMitochondrial disorders as windows into an ancient organelleNature201249137438323151580
  • FilostoMScarpelliMCotelliMSThe role of mitochondria in neurodegenerative diseasesJ Neurol20112581763177421604203
  • CuiHLiFChenDComprehensive next-generation sequence analyses of the entire mitochondrial genome reveal new insights into the molecular diagnosis of mitochondrial DNA disordersGenet Med20131538839423288206
  • JeppesenTDDunoMRisomLA novel de novo mutation of the mitochondrial tRNAlys gene mt.8340G>A associated with pure myopathyNeuromuscul Disord20142416216624161205
  • WhittakerRGBlackwoodJKAlstonCLUrine heteroplasmy is the best predictor of clinical outcome in the m.3243A>G mtDNA mutationNeurology20097256856919204268
  • LegatiAReyesANascaANew genes and pathomechanisms in mitochondrial disorders unraveled by NGS technologiesBiochim Biophys Acta201618571326133526968897
  • PfefferGGormanGSGriffinHMutations in the SPG7 gene cause chronic progressive external ophthalmoplegia through disordered mitochondrial DNA maintenanceBrain20141371323133624727571
  • MendelsohnBAMehtaNHameedBPekmezciMPackmanSRalphJAdult-onset fatal neurohepatopathy in a woman caused by MPV17 mutationJIMD Rep201413374124190800
  • AntonenkovVDIsomursuAMennerichDThe human mitochondrial DNA depletion syndrome gene MPV17 encodes a non-selective channel that modulates membrane potentialJ Biol Chem2015290138401386125861990
  • PyleANightingaleHJGriffinHRespiratory chain deficiency in non mitochondrial diseaseNeurol Genet20151e627066545
  • ScarpelliMTodeschiniARinaldiFRotaSPadovaniAFilostoMStrategies for treating mitochondrial disorders: an updateMol Genet Metab201411325326025458518
  • PfefferGHorvathRKlopstockTNew treatments for mitochondrial disease—no time to drop our standardsNat Rev Neurol2013947448123817350
  • ScarpelliMCotelliMSMancusoMCurrent options in the treatment of mitochondrial diseasesRecent Pat CNS Drug Discov2010520320920722626
  • HyslopLABlakeleyPCravenLTowards clinical application of pronuclear transfer to prevent mitochondrial DNA diseaseNature201653438338627281217
  • MontiniGMalaventuraCSalviatiLEarly coenzyme Q10 supplementation in primaryoenzyme Q10 deficiencyN Engl J Med20083582849285018579827
  • FoleyARMenezesMPPandraudATreatable childhood neuronopathy caused by mutations in riboflavin transporter RFVT2Brain2014137445624253200
  • BottaniEGiordanoCCivilettoGAAV-mediated liver-specific MPV17 expression restores mtDNA levels and prevents diet-induced liver failureMol Ther201422101724247928
  • FilostoMScarpelliMToninPPitfalls in diagnosing mitochondrial neurogastrointestinal encephalomyopathyJ Inherit Metab Dis2011341199120321503690
  • GaroneCTadesseSHiranoMClinical and genetic spectrum of mitochondrial neurogastrointestinal encephalomyopathyBrain20111343326333221933806
  • HiranoMGaroneCQuinziiCMCoQ(10) deficiencies and MNGIE: two treatable mitochondrial disordersBiochim Biophys Acta2012182062563122274133
  • BaxBEBainMDScarpelliMFilostoMToninPMoranNClinical and biochemical improvements in a patient with MNGIE following enzyme replacementNeurology2013811269127123966250
  • HiranoMMartíRCasaliCAllogeneic stem cell transplantation corrects biochemical derangements in MNGIENeurology2006671458146016971696
  • FilostoMScarpelliMToninPCourse and management of allogeneic stem cell transplantation in patients with mitochondrial neurogastrointestinal encephalomyopathyJ Neurol20122592699270622711161
  • HalterJPMichaelWSchüpbachMAllogeneic haematopoietic stem cell transplantation for mitochondrial neurogastrointestinal encephalomyopathyBrain20151382847285826264513
  • ScarpelliMRussignanAZomborMPoor outcome in a mitochondrial neurogastrointestinal encephalomyopathy patient with a novel TYMP mutation: the need for early diagnosisCase Rep Neurol2012424825323341816
  • Torres-TorronterasJGómezAEixarchHHematopoietic gene therapy restores thymidine phosphorylase activity in a cell culture and a murine model of MNGIEGene Ther20111879580621451581
  • Torres-TorronterasJViscomiCCabrera-PérezRGene therapy using a liver-targeted AAV vector restores nucleoside and nucleotide homeostasis in a murine model of MNGIEMol Ther20142290190724448160
  • O’MahonyCAGossJAThe future of liver transplantationTex Heart Inst J20123987487523304042
  • De GiorgioRPironiLRinaldiRLiver transplantation for mitochondrial neurogastrointestinal encephalomyopathyAnn Neurol20168044845527421916
  • CámaraYGonzález-VioqueEScarpelliMAdministration of deoxyribonucleosides or inhibition of their catabolism as a pharmacological approach for mitochondrial DNA depletion syndromeHum Mol Genet2014232459246724362886
  • McFarlandRClarkKMMorrisAAMultiple neonatal deaths due to a homoplasmic mitochondrial DNA mutationNat Genet20023014514611799391
  • ElliottHRSamuelsDCEdenJAReltonCLChinneryPFPathogenic mitochondrial DNA mutations are common in the general populationAm J Hum Genet20088325426018674747
  • KohrogiKImagawaEMutoYBiotin-responsive basal ganglia disease: a case diagnosed by whole exome sequencingJ Hum Genet20156038138525876998
  • GaroneCGurgel-GiannettiJSanna-CherchiSA novel SUCLA2 mutation presenting as a complex childhood movement disorderJ Child Neurol2016
  • GaroneCDonatiMASacchiniMMitochondrial encephalomyopathy due to a novel mutation in ACAD9JAMA Neurol2013701177117923836383
  • ChungWKMartinKJalasCMutations in COQ4, an essential component of coenzyme Q biosynthesis, cause lethal neonatal mitochondrial encephalomyopathyJ Med Genet20155262763526185144
  • XuBLiXDuMZhouCFangHLyuJYangYNovel mutation of ND4 gene identified by targeted next-generation sequencing in patient with Leigh syndromeJ Hum Genet2016
  • DaiHZhangVWEl-HattabAWcprFBXL4 defects are common in patients with congenital lactic acidemia and encephalomyopathic mitochondrial DNA depletion syndromeClin Genet2016
  • ThompsonKMajdHDallabonaCRecurrent de novo dominant mutations in SLC25A4 cause severe early-onset mitochondrial disease and loss of mitochondrial DNA copy numberAm J Hum Genet20169986087627693233
  • NgYSAlstonCLDiodatoDThe clinical, biochemical and genetic features associated with RMND1-related mitochondrial diseaseJ Med Genet2016