Gene therapy for the mitochondrial genome: Purging mutations, pacifying ailments

Gene therapy for the mitochondrial genome: Purging mutations, pacifying ailments

Accepted Manuscript Gene therapy for the mitochondrial genome: Purging mutations, pacifying ailments M. Aravintha Siva, R. Mahalakshmi, Dipita Bhakta...

2MB Sizes 0 Downloads 87 Views

Accepted Manuscript Gene therapy for the mitochondrial genome: Purging mutations, pacifying ailments

M. Aravintha Siva, R. Mahalakshmi, Dipita Bhakta-Guha, Gunjan Guha PII: DOI: Reference:

S1567-7249(18)30018-7 doi:10.1016/j.mito.2018.06.002 MITOCH 1292

To appear in:

Mitochondrion

Received date: Revised date: Accepted date:

27 January 2018 24 May 2018 7 June 2018

Please cite this article as: M. Aravintha Siva, R. Mahalakshmi, Dipita Bhakta-Guha, Gunjan Guha , Gene therapy for the mitochondrial genome: Purging mutations, pacifying ailments. Mitoch (2017), doi:10.1016/j.mito.2018.06.002

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Gene Therapy for the Mitochondrial Genome: Purging Mutations, Pacifying Ailments M. Aravintha Siva, R. Mahalakshmi, Dipita Bhakta-Guha* and Gunjan Guha* Cellular Dyshomeostasis Laboratory (CDHL), School of Chemical and Bio Technology, SASTRA University, Thanjavur 613 401. Tamil Nadu, India. *

PT

Corresponding authors:

Gunjan Guha

SC

RI

CDHL, SCBT, SASTRA University, Thanjavur 613 401. Tamil Nadu, India. Email: [email protected]; [email protected] Phone: +91 4362 264101-108 (ext: 3777); +91 4362 304000-010 (ext: 3777).

Dipita Bhakta-Guha

AC

CE

PT E

D

MA

NU

CDHL, SCBT, SASTRA University, Thanjavur 613 401. Tamil Nadu, India. Email: [email protected]; [email protected] Phone: +91 4362 264101-108 (ext: 3777); +91 4362 304000-010 (ext: 3777).

ACCEPTED MANUSCRIPT Abstract In the recent years, the reported cases of mitochondrial disorders have reached a colossal number. These disorders spawn a sundry of pathological conditions, which lead to pernicious symptoms and even fatality. Due to the unpredictable etiologies, mitochondrial diseases are putatively referred to as “mystondria” (mysterious diseases of mitochondria). Although present-day research has greatly improved our understanding of mitochondrial disorders,

PT

effective therapeutic interventions are still at the precursory stage. The conundrum becomes further complicated because these pathologies might occur due to either mitochondrial DNA

RI

(mtDNA) mutations or due to mutations in the nuclear DNA (nDNA), or both. While correcting nDNA mutations by using gene therapy (replacement of defective genes by delivering wild-

SC

type (WT) ones into the host cell, or silencing a dominant mutant allele that is pathogenic) has emerged as a promising strategy to address some mitochondrial diseases, the complications in

NU

correcting the defects of mtDNA in order to renovate mitochondrial functions have remained a steep challenge. In this review, we focus specifically on the selective gene therapy strategies

MA

that have demonstrated prospects in targeting the pathological mutations in the mitochondrial genome, thereby treating mitochondrial ailments.

AC

CE

PT E

D

Keywords: Mitochondrial diseases; mystondria; mtDNA mutations; gene therapy.

ACCEPTED MANUSCRIPT Abbreviations 3′-UTR

3′-untranslated region

AAV

adeno-associated virus

AAV2/2

AAV serotype 2

AIF apoptosis-inducing factor COX

cytochrome c oxidase

COI cytochrome c oxidase subunit I cytochrome c oxidase subunit II

COIII

cytochrome c oxidase subunit III

COIV

cytochrome c oxidase subunit IV

COVIII

cytochrome c oxidase subunit VIII

CO-X

cytochrome c oxidase subunit X

CPEO

chronic progressive external ophthalmoplegia

SC

RI

PT

COII

NU

CRISPR/Cas9 clustered regularly-interspaced short palindromic repeats/CRISPRassociated protein 9 DA dopaminergic displacement loop

DQA

dequalinium chloride

D

D-loop

MA

DF-MITO-porter dual function MITO-porter

DQAsomes DQA-based liposome-like nanovesicles enhanced green fluorescent protein

ETC

electron transport chain

PT E

eGFP

CE

FLAG epitope tag DYKDDDDK sequence (where D = aspartic acid, Y = tyrosine, K = lysine) familial bilateral striatal necrosis

GFP

green fluorescence protein

HSP KSS LHON

AC

FBSN

heat shock protein Kearns-Sayre syndrome Leber’s hereditary optic neuropathy

LS Leigh syndrome MELAS

mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes

MEMRI

manganese-enhanced magnetic resonance imaging

MEPR

myoclonic epilepsy and psychomotor regression

MERRF

myoclonic epilepsy with ragged-red fibers

MHCM

maternally inherited hypertrophic cardiomyopathy

MICM

maternally inherited cardiomyopathy

MID

maternally inherited deafness

ACCEPTED MANUSCRIPT MIDD

maternally inherited diabetes and deafness

mitoTALEN

mitochondrially targeted transcription activator-like effector nuclease

modRNA synthetic chemically modified mRNA mitochondrial transduction domain

mtDNA

mitochondrial DNA

MTS

mitochondria-targeting sequences

mtZFNs

mitochondrially targeted zinc-finger nucleases

NARP

neurogenic muscle weakness, ataxia and retinitis pigmentosa

NDI1

NADH-quinone oxidoreductase

nDNA

nuclear DNA

ORF

open reading frame

OriH

origin of replication for heavy strand of mtDNA

OriL

origin of replication for light strand of mtDNA

PD Parkinson’s disease

NU

OX-PHOS oxidative phosphorylation

SC

RI

PT

MTD

plasmid DNA

PICs

polyion complexes

RGCs

retinal ganglion cells

RNS

reactive nitrogen species

ROS

reactive oxygen species

SOD2

superoxide dismutase 2

TALE

transcription activator-like effector

TALEN

transcription activator-like effector nuclease

TFAM

mitochondrial transcription factor A

TFB1M

mitochondrial transcription factor B1

ZFP

zinc-finger protein

D

PT E

CE

AC

ΔΨm

MA

pDNA

mitochondrial membrane potential

ACCEPTED MANUSCRIPT 1. Mitochondria: The dynamics of functions and dysfunctions Mitochondria are eukaryotic cell organelles that arguably evolved from a symbiotic relationship between aerobic microbes and primordial eukaryotic cells (Chan, 2006; Sagan, 1967; Selosse, 2011). They putatively produce energy (ATP) through oxidative phosphorylation (OX-PHOS) by transporting electrons across multiple internal complexes (mitochondrial complexes I-V) of an electron transport chain (ETC) (Das and Guha 2010;

PT

Kaurola et al., 2016). These double-membrane organelles maintain their redox potential by pumping protons across their inner membrane (Chan, 2006). Mitochondria are involved in

RI

metabolism of glucose (Krebs cycle) and fatty acids (β-oxidation), and are also associated with calcium homeostasis (Schon and Przedborski, 2011; Palomo and Manfredi, 2015).

SC

Furthermore, they play a crucial role in apoptosis, which is triggered by the release of cytochrome c and apoptosis-inducing factor (AIF) from the mitochondria into the cytoplasm

NU

(Frank et al., 2001; Herold et al., 2002; Faridi et al., 2016; Zhao et al., 2016). Mitochondrial redox reactions produce reactive oxygen species (ROS) (such as superoxides, peroxides,

MA

hydroxides and singlet oxygen) and reactive nitrogen species (RNS) (peroxynitrites and dinitrogen trioxide), which might impair the ETC and cell cycle progression (Das and Guha

D

2010; Quijano et al., 2016).

PT E

Human mitochondria contain their own circular, double-stranded genome –– the mitochondrial DNA (mtDNA), which is 16,569 bp in length and constitutes 37 genes (Holt et al., 1989; Van Trappen et al., 2007). It is extremely susceptible to oxidative damage due to its close proximity

CE

to the mitochondrial ROS generation machinery and the limited availability of DNA repair enzymes (for mtDNA) (Das and Guha, 2010; Lee and Wei, 2007). Moreover, the absence of introns or intergenic sequences and histones in the mtDNA makes it prone to oxidative damage

AC

(Das and Guha, 2010; DiMauro et al., 2013). The mtDNA encodes for 2 rRNAs, 22 tRNAs and 13 polypeptides –– one subunit of cytochrome bc1 complex (complex III): cytochrome b; two subunits of ATP synthase (complex V): ATP6 and ATP8; three subunits of cytochrome c oxidase (COX; complex IV): COI, COII and COIII; and seven subunits of NADH-ubiquinone oxidoreductase (complex I): ND1, ND2, ND3, ND4, ND4L, ND5 and ND6; while complex II (succinate dehydrogenase), on the other hand, is entirely encoded by the nuclear DNA (nDNA) (Holt et al., 1989; Taanman, 1999; Mishmar et al., 2003; DiMauro et al., 2013). mtDNA also has a non-coding region called the displacement loop (D-loop), containing the origins of replication (OriH and OriL sites) and transcription promoters (Taanman, 1999; Singh, 2013; Kambe and Miyata, 2016). Bioenergetic dyshomeostasis of the mitochondria and consequent

ACCEPTED MANUSCRIPT overproduction of free radicals in the ETC lead to initiation and accumulation of considerable damage to the mtDNA, which can cause a plethora of pathological conditions (Quijano et al., 2016). Such dysfunctional conditions are extensively observed in tissues with higher energy demand like muscles, liver, heart, kidney and brain (Leslie et al., 2016). Since mitochondria are inherited only maternally (Camus et al., 2012; Sato and Sato, 2013), defective mitochondria containing mutant mtDNA are also inherited by the offspring from the mother (Thajeb et al.,

PT

2006; Tuppen et al., 2010). Mitochondrial diseases constitute a variegated conglomeration of clinical facets and symptoms involving a multitude of diverse tissues and organs, which cause significant morbidity and mortality (Wong, 2007; Moggio et al., 2014; Steele et al., 2017;

RI

Davis et al., 2018).

SC

The number of reported cases of mitochondrial disorders has increased in the recent years –– up to one in five thousand children harbor mitochondrial pathologies (Haas et al., 2007). In

NU

fact, up to four thousand newborns are diagnosed with mitochondrial diseases every year in the United States alone (Tachibana et al., 2013). Similar prevalence of mitochondrial pathologies

MA

has also been reported from other continents (Nesbitt et al., 2014; Ng and Turnbull 2016; Australian Mitochondrial Disease Foundation 2017). However, mitochondrial diseases are still referred to as “mystondria” (mysterious diseases of mitochondria) due to their unpredictable

D

genetic and mipigenetic (mitochondrial epigenetics) etiology (Singh, 2015). Impaired

PT E

respiratory subunits, production of dysfunctional enzymes and leakage of electrons from the ETC (mainly from complexes I and III) cause overproduction of ROS (Tretter and Adam-Vizi 2004; Görlach et al., 2015), which hinder mitochondrial biogenesis, and also lead to lipid

CE

peroxidation and protein carbonylation, along with significant damage to the mtDNA (Shokolenko et al., 2009; Suzuki et al., 2010; Das and Guha, 2010; Kwiecien et al., 2014; Yao

AC

et al., 2015; Birch-Machin and Bowman, 2016). Such mitochondrial and cellular dyshomeostases result in myriad pathological conditions. For example, type I and type II diabetes mellitus patients reportedly have defective insulin-producing β cells (in the islets of Langerhans in pancreas) (Lenzen, 2008) due to damaged mtDNA, as well as dysfunctional mitochondrial proteins and lipids (Verdile et al., 2015; Montgomery and Turner 2015). The role of mitochondria has also been established in obesity, where saturated fat in cardiomyocytes was observed to cause mitochondrial dysfunctions, such as decreased rate of OX-PHOS, augmented levels of mitochondrial ROS, and depolarization of the mitochondrial membrane (Joseph et al., 2016).

ACCEPTED MANUSCRIPT The colossal number of mitochondrial disorders has been attributed to a multitude of mutations in the mtDNA (Table 1). It has to be noted that mitochondrial diseases do not only occur due to mtDNA mutations, but also due to nDNA aberrations, since a considerable part of the mitochondrial proteome is encoded by the nucleus (Zhu et al., 2009; Gorman et al., 2015; Viscomi et al., 2015). In this review, however, we focus on the gene therapy strategies targeting

PT

solely the pathological mutations in the mitochondrial genome.

RI

2. Pathological phenotypes and mtDNA mutations

SC

Due to the inherent functional complexity of the mitochondria, it is often onerous to attribute a mitochondrial pathology to a particular mtDNA mutation, as well as to restrict a mutation to one single ailment (Doyle and Chan, 2008; Iyer, 2013). However, few principal types of

NU

mutations have been frequently reported in the majority of mitochondrial diseases (Yamada et al., 2007) (Fig. 1). For example, an m.11778G>A point mutation in the ND4 gene of complex

MA

I is observed in Leber’s hereditary optic neuropathy (LHON), a frequently occurring optic neuropathy (1 in 25,000 individuals), which is predominant (with 4-5 times greater probability) in males

(Newman, 1993; Yu-Wai-Man and Chinnery, 1993; Chinnery et al., 2001;

D

Chadderton et al., 2013). It is characterized by bilateral acute/subacute visual defects, with

PT E

deleterious effects on retinal ganglion cells (RGCs) (Yu-Wai-Man and Chinnery, 1993; Carelli et al., 2004; Chadderton et al., 2013). Most LHON incidences (~95%) occur via three substitution mutations in the genes encoding for complex I at positions m.3460G>A,

CE

m.3697G>A and m.4171C>A (ND1), m.11778G>A (ND4) and m.14484T>C (ND6) in the

AC

mtDNA (Chinnery et al., 2001; Kim et al., 2002; Spruijt et al., 2007). Substitution mutations in mitochondrial ATP6 gene (m.8993T>G; and m.8993T>C in a small percentage of patients) have been attributed to cause neurogenic muscle weakness, ataxia, and retinitis pigmentosa (NARP) (Thorburn and Rahman, 1993; De Meirleir et al., 2004; Kabunga et al., 2015), which is a neurological syndrome with an estimated prevalence rate of 0.81/100,000 individuals (Orphanet, 2017). These mutations lead to the replacement of amino acid leucine by arginine in F1FO-ATPase, thereby altering its function (Thorburn and Rahman, 1993). Similar to NARP, incidences of m.8993T>G and m.8993T>C mutations in the ATP6 gene in mtDNA are also observed in Leigh syndrome (LS), (Thorburn and Rahman, 1993; De Meirleir et al., 2004; Praeter et al., 2014). LS (also known as subacute necrotizing encephalomyelopathy) is a progressive neurodegenerative disorder of the central nervous

ACCEPTED MANUSCRIPT system that primarily affects the spinal cord, basal ganglia, pons, cerebellum and thalamus, as well as causes demyelination of optic nerves (Westarp et al., 1992; Duff et al., 2015; Gerards et al., 2016). Other mtDNA mutations, such as m.4296G>A (tRNAIle) (Martikainen et al., 2013), m.5523T>G and m.5559A>G (tRNATrp) (Mkaouar-Rebai et al., 2009), m.13513G>A (ND5) (Shanske et al., 2008), m.13514A>G (ND5) (Bannwarth et al., 2013), m.9191T>C (ATP6) (Moslemi et al., 2005) and m.9478T>C (COIII) (Mkaouar-Rebai et al., 2011), are also

PT

reportedly associated with LS (Fig. 1). The ailment has an incidence rate of one in 40,000 live births (Lake et al., 2015). While the maximal onset of this disorder takes place at about 3–12 months of age (with ~50% of the patients dying by the age of 3 years), late onset has also been

RI

observed in adults (Rahman and Thorburn, 1993). LS is caused by energy deficiency in cells

SC

due to defective enzymes viz. ETC complex I, complex IV and pyruvate dehydrogenase complex. The symptoms of this disease include hypotonia, ataxia, psychomotor regression,

NU

dystonia, cranial nerve palsies, spasticity, infantile spasms, sensory neural hearing loss, persistent vomiting, dysphagia and respiratory problems like hyper- and hypoventilation

MA

(Finsterer, 2008; Sofou et al., 2014; Gerards et al., 2016). The m.8993T>G/C mutation is associated with complex V deficiency too (De Meirleir et al., 2004). In fact, the disease has been associated with the etiologies of NARP and LS (De Vivo,

D

1993; National Organization for Rare Disorders, 2016; Rat Genome Database, 2017). Complex

PT E

V deficiency is a very rare metabolic disorder, which is also caused by other mutations in the ATP6 and ATP8 genes (Auré et al., 2013; Kytövuori et al., 2016), such as m.8561C>G (in the overlap region of ATP6 and ATP8) (Kytövuori et al., 2016), m.8969G>A (in ATP6) (Burrage

CE

et al., 2014) and m.8529G>A (in ATP8) (Jonckheere et al., 2008) (Fig. 1). Myriad mtDNA mutations have been reported in the substantia nigra (a nucleus in midbrain)

AC

of individuals with Parkinson's disease (PD) (Lin et al., 2012; Coxhead et al., 2016), which is the second most prevalent geriatric neurodegenerative disorder in humans, especially affecting people above 50 years of age (Gibson et al., 2012; Pringsheim et al., 2014). It is characterized by the deterioration of dopaminergic (DA) neurons in the substantia nigra pars compacta and by the accumulation of Lewy bodies (Gibson et al., 2012). PD-associated mutations have been reported in COI, COII, cytochrome b, ND5 and the D-Loop region of the mtDNA. These lead to mitochondrial alterations like decreased complex I activity, excessive ROS production, consequent mtDNA damage and mitophagy, which are associated with the etiology of PD (Winklhofer and Haass, 2010; Moon and Paek, 2015).

ACCEPTED MANUSCRIPT An m.8344A>G mutation in the gene encoding tRNALys is the most common cause of myoclonic epilepsy with ragged-red fibers (MERRF) syndrome (Shoffner et al., 1990; Jacobs, 2003), a disease that commonly affects muscles and the central nervous system, and is characterized by the presence of myoclonus, myopathy and spasticity. It has been estimated to affect one in every 5,000 individuals worldwide (US National Library of Medicine, Genetics Home Reference, 2017). MERRF can also occur due to point mutations in the mtDNA tRNAPhe gene at nucleotide position 616 and is observed in skeletal muscles (Zsurka et al., 2010).

PT

Mutations (Fig. 1) in the encoding region of tRNALeu (m.3243A>G and m.3271T>C) and tRNAVal (m.1642G>A) have been reported in mitochondrial encephalomyopathy, lactic

RI

acidosis, and stroke-like episodes (MELAS) (Goto et al., 1990), and maternally inherited

SC

cardiomyopathy (MICM) (Silvestri et al., 1994; Santorelli et al., 1996, 1999; Dipchand et al., 2001). MELAS is also associated with mutations in the COIII (m.9957T>C) and ND5

NU

(m.12770A>G, m.13045A>C, m.13513G>A and m.13514A>G) genes (Tuppen et al., 2010; Hashimoto et al., 2015).

MA

mtDNA deletions have been encountered in the chronic progressive external ophthalmoplegia (CPEO) (Van Goethem et al., 2003; Sundaram et al., 2011) and Kearns-Sayre syndrome (KSS) (Shanske et al., 1990). A number of other mutations are also associated with a host of other

D

mitochondrial disorders (Fig. 1), such as familial bilateral striatal necrosis (FBSN)

PT E

(Thyagarajan et al., 1995), maternally inherited diabetes and deafness (MIDD) (Mangiafico et al., 2004), myoclonic epilepsy and psychomotor regression (MEPR) (Shtilbans et al., 1999), maternally inherited hypertrophic cardiomyopathy (MHCM) (Thambisetty and Newman,

CE

2004; Tuppen et al., 2010; Hashimoto et al., 2015), and maternally inherited deafness (MID)

AC

(Yang et al., 2011; Zheng et al., 2012; Hoptasz et al., 2014).

3. Gene therapy: Confronting mitochondrial pathologies Although contemporary research has considerably enhanced our understanding of the genetic and biochemical etiology of mitochondrial disorders, therapeutic interventions for the same have been preliminary at best. Nevertheless, with recent advances in technology, gene therapy has emerged as a potential strategy to address mitochondrial diseases, either by manipulating the nuclear genome or by targeting the mitochondrial paraphernalia (Weissig and Torchilin, 2001). Gene therapy is the replacement of a defective gene by delivering a corrective allele into the host cell, or silencing a dominant mutant allele that is pathogenic to the host (Kaufmann

ACCEPTED MANUSCRIPT et al., 2013; Choong, Baba, and Mochizuki 2015). Since a large number of mitochondrial disorders are rooted to mutations in the mtDNA (Fig. 1), an intervention can be achieved by corrective gene therapy against the defective/missing facets of the mitochondrial genome. This can be materialized by delivery of functional therapeutic genes as substitutes for the endogenous counterparts (Yamada et al., 2007; Kyriakouli et al., 2008; Heller, Brockhoff, and Goepferich 2012). For most mitochondrial gene therapies, one of the paramount criteria is the

PT

construction of suitable vectors to accommodate the genes of interest (Yasuzaki et al., 2015). Among a plethora of vectors, adenoviruses have been most commonly used (accounting for 23.3% of the trials) because of their high transduction efficiency (leading to high gene

RI

expression levels) and larger DNA load carrying capacity in comparison to other viruses (Ginn

SC

et al., 2013). Adeno-associated virus (AAV) has also been extensively used due to its broad spectrum of hosts (Gao et al., 2002; Wang et al., 2016). Herpes simplex, pox and vaccinia

NU

viruses have also been successfully used in gene therapy (Ginn et al., 2013). Mitochondrial gene therapy has also been accomplished by administration of the naked plasmid DNA (pDNA) in vivo resulting in production of mRNA in the mitochondria (Yasuzaki et al., 2015).

MA

Variegated modules of mitochondrial gene therapy have demonstrated positive results in a host of neuromuscular and metabolic disorders related to mitochondrial dyshomeostasis. However,

D

although diverse in their characteristics, all applied strategies broadly follow one of the three major approaches: (1) allotopic expression (of mtDNA genes in the nucleus); (2) selective

CE

PT E

inhibition of mutant mtDNA; (3) delivery of WT mtDNA into the mitochondria.

3.1. Allotopic expression-mediated gene therapy

AC

Endosymbiotic gene transfer from the mitochondria to the nucleus is a ubiquitous ongoing evolutionary process that occurs at a substantial frequency (Timmis et al., 2004). This phenomenon is emulated for allotopic expression-mediated gene therapy for defective mtDNA genes for the integration of the corresponding corrective mitochondrial genes into the nucleus (Guy et al., 2002; Manfredi et al., 2002; Shokolenko et al., 2010; Cwerman-Thibault et al., 2015). Transport of the subsequently expressed protein into the mitochondria is facilitated by a mitochondria-targeting sequence (MTS; which helps in transporting a protein of interest into the mitochondria) (Shokolenko et al., 2010). nDNA-encoded proteins that are destined to be localized in the mitochondria contain an MTS at their N-terminal ends, which is recognized by mitochondrial protein import channels, thereby allowing protein import (Chin et al., 2018). Moreover, specific 3’- untranslated regions (3′-UTRs) are used to locate mRNAs on the

ACCEPTED MANUSCRIPT mitochondrial surface and help in co-translational import of these nDNA-encoded mitochondrial proteins (Kaltimbacher et al., 2006; Bonnet et al., 2007a). However, it has been recently reported that it is the choice of MTS that determines the efficiency of delivery of allotopically expressed proteins to the mitochondria, rather than the selected 3’-UTR (Chin et al., 2018). This method was first tested for its efficacy in mitochondrial ATP8 gene-deficient models of Saccharomyces cerevisiae, where the incorporation of the WT mtDNA gene in the

PT

nucleus resulted in successful restoration of function (Nagley et al., 1988). This strategy of integrating mtDNA genes in the nDNA has been applied to correct defects arising from several

RI

mtDNA mutations (Fig. 2A).

As a key strategy for treating LHON, allotopic expression-mediated gene therapy has been

SC

accomplished in vitro in cybrid cells (derived from homoplasmic osteosarcoma) harboring m.11778G>A mutation in the ND4 gene (Guy et al., 2002). Through this method, the WT ND4

NU

gene was integrated into the nucleus by recombinant AAV (rAAV) vector. The plasmid was constructed by fusing the ND4 gene with an MTS (from the P1 isoform of human ATP synthase

MA

subunit c), and a FLAG epitope tag. This sequence was then integrated with green fluorescence protein (GFP) via an internal ribosomal entry site (IRES) sequence, a chicken β-actin promoter and a cytomegalovirus enhancer. The final construct was cloned into pTR-UF12 plasmid vector

D

and packaged to generate rAAV particles. The cybrid cells were then stably transfected with

PT E

3×107 rAAV particles. The expression of the fused protein was driven by the chicken β-actin promoter and the enhancer. The transfected cells subsequently showed GFP and FLAG expression in the cytoplasm and mitochondria respectively. The FLAG localization to the

CE

mitochondria implied successful transport of the ND4 gene, which was further confirmed by the observed three-fold increase in ATP synthesis in the cybrid cells (Guy et al., 2002).

AC

In an in vivo LHON model (rats), the disease was developed using allotopic expression of mutant human ND4 gene (bearing m.11778G>A mutation) in the RGCs, thereby creating mitochondrial heteroplasmy (Ellouze et al., 2008), which is a condition where a single cell can harbor mitochondria with both WT and mutant genomes (Das and Guha, 2010; Wallace and Chalkia, 2013; Picard et al., 2014; Wernick et al., 2016). The WT ND4 gene was incorporated with pAAV-IRES-GFP and transformed pDNA was injected into the vitreous body of the eye through electroporation. The plasmid was constructed by fusion of CO-X (which is a nuclear gene) and ND4 into the pAAV-IRES-GFP vector. CO-X encodes for both MTS (present at the amino termini of pre-proteins) and the sequence for MTS cleavage (Omura, 1998; Ellouze et al., 2008). MTS helps to import ND4 proteins from cytoplasm into the mitochondria, while the

ACCEPTED MANUSCRIPT MTS cleavage sequence functions to generate the mature ND4 once the ND4-MTS complex is inside the mitochondria (Ellouze et al., 2008). GFP gene was used for transgene selection. The ND4-CO-X gene was integrated into the nuclear genome of RGC and the ND4 subunit protein was synthesized in the cytoplasm. With the help of MTS, ND4 was imported into the mitochondria, thus replacing the defective ND4, thereby restoring normal bioenergetics (Ellouze et al., 2008).

PT

In another approach, trans-kingdom mitochondrial gene therapy was accomplished by intraocular delivery of NDI1 (NADH-quinone oxidoreductase; a nuclear gene from S.

RI

cerevisiae that encodes for single subunit complex I) in mice. NDI1 gene contains endogenous MTS, which assists in efficient transport of the protein into the mitochondria (which is more

SC

effective than allotopic expression with introduced MTS) (Chadderton et al., 2013). AAV2 plasmid containing NDI1 (AAV2-NDI1) was constructed and delivered intravitreally into

NU

rotenone-treated (induces RGC loss and inhibits complex I) mice (Chadderton et al., 2013). Optokinetic assessment of the treated eye showed significant reduction in RGC death and optic

MA

nerve defects. Also, manganese-enhanced magnetic resonance imaging (MEMRI) exhibited improved functions of optic nerve by active transport of Mn+2 into the cells through voltagegated calcium channels (Chadderton et al., 2013; Marella et al., 2010). Observations up until

D

six months post-treatment showed no remarkable amount of anti-NDI1 antibody in the sera

PT E

(Marella et al., 2011), proving the potency of NDI1 for the replacement of defective complex I proteins. An added advantage of this approach is that it is not restricted to a particular mutation

CE

(Chadderton et al., 2013).

In an ongoing (2014–2019) clinical trial (NCT02161380) (Guy, 2018) at the Bascom Palmer Eye Institute (Miller School of Medicine, University of Miami, USA), AAV2-mediated gene

AC

therapy containing ND4 (AAV(scAAV)2(Y444,500,730F)-P1ND4v2) was done intravitreally in five patients with LHON (m.11778G>A mutation) (Feuer et al., 2016). The selfcomplementary genome improved the transgene expression, and the AAV2 capsid promoted efficient tissue-specific uptake (Koilkonda et al., 2010; Feuer et al., 2016). The patients were treated with differential doses of transformed vector genomes. The initial results of this clinical trial showed an improved vision in all the patients with none of them suffering from any severe side-effects. However, it has to be noted that the sampling size was very low and that the study is still recruiting patients (Feuer et al., 2016; Wan et al., 2016; Yang et al., 2016). Another set of recent prospective clinical trials (NCT02064569, NCT02652767, NCT02652780, NCT03406104 and NCT03293524) are testing the efficacy and safety of GS010 (rAAV2/2-

ACCEPTED MANUSCRIPT ND4) in patients affected by ND4 mutations, the results of which are currently unavailable (Newman, 2018; Subramanian, 2018; Vignal, 2018; Yu-Wai-Man, 2018a, 2018b). As discussed earlier (section 2), the m.8993T>G substitution mutation in the mitochondrial ATP6 gene is extremely common in NARP, LS and complex V deficiency (De Meirleir et al., 2004). To develop an allotopic expression-mediated gene therapy strategy to correct this mutation, skin biopsies from an 8-month old child with NARP syndrome (m.8993T>G

PT

mutation) were used for establishing fibroblast culture as the experimental model (Bonnet et al., 2007a). For effective localization of ATP6 mRNA into the mitochondria, the MTS sequence

RI

of superoxide dismutase 2 (SOD2) gene and its 3′-UTR were combined with the ATP6 gene (Bonnet et al., 2007a, 2007b). The transfection plasmid was constructed by inserting ATP6

SC

gene (with a FLAG epitope at the C terminus of the ATP6 ORF) into pCMV-Tag 4A vector, along with the MTS of the SOD2 gene and either the 3′-UTR of SOD2 or the 3′-UTR of SV40,

NU

thereby giving rise to the two following final constructs: SOD2MTSATP6-3′UTRSOD2 and SOD2MTSATP6-3′UTRSV40 (Kaltimbacher et al., 2006). It was observed that when both MTS

MA

and the 3′-UTR sequence were present (i.e., SOD2MTSATP6-3′UTRSOD2), ATP6 mRNA sorting to the mitochondrial surface was augmented, leading to 1.8 times higher ATP6 protein expression, than that with the MTS alone (i.e., SOD2MTSATP6-3′UTRSV40). SOD2-ATP6 co-

D

localization was observed by indirect immunofluorescence using antibodies against FLAG or

PT E

ATP synthase (-subunit). Complex V activity was evidently restored using this gene therapy approach, which was also verified by the increased production of ATP (Bonnet et al., 2007a).

CE

Another similar strategy was developed using a B6(B6SJLF1) (mice) model (Dunn and Pinkert, 2012). First, WT ATP6 genes were cloned in frame into the pEF/myc/mito plasmid, containing the promoter for human EF-1α, the MTS from the human COVIII gene (nuclear), and an in-

AC

frame 3’ myc epitope tag. This pEF/myc/mito/ATP6 plasmid was used to generate transgenic mice. Mitochondrial localization of allotopically expressed corrective ATP6 protein was confirmed by electron microscopy (Dunn and Pinkert, 2012). In a recent study on mtDNA complex V-deficient patient cells (null for the ATP8 protein, with significantly less ATP6 expression) harboring the m.8529G>A mutation (in the mitochondrial ATP8 gene) (Boominathan et al., 2016), researchers allotopically expressed ATP6 and ATP8 genes in the nucleus to correct the deficiency using a pCMV6 vector. Corrective DNA sequences for ATP6 and ATP8 were incorporated into the vector between the MluI and XhoI sites. A CO-X or ATP5G1- or ATP5G2-based MTS was also inserted into the pCMV6 at the 5′ end (between sites AsiSI and MluI) of the genes. Myc and FLAG or V5 epitope were also

ACCEPTED MANUSCRIPT introduced immediately downstream of the codon-corrected genes. This technique rescued expression of both the proteins and recovered their functions in the cell significantly (Boominathan et al., 2016). A strategy for allotopic expression of mitochondrial proteins without altering the nuclear genome uses synthetic chemically modified mRNA (modRNA) encoding for the corrective protein (Fig. 2B), whose uridine and cytosine have been substituted respectively by

PT

pseudouridine and 5-methyl-cytosine (Chin et al., 2018). These modifications allow the modRNA to avoid the Toll-like receptors of innate immune system (that would have otherwise

RI

degraded them) in vivo while facilitating efficient translation of the protein of interest (Karikó et al., 2005). A report published in 2018 has demonstrated allotopic expression of

SC

mitochondrial proteins by using modRNAs. In this study, the researchers constructed modRNAs for allotopic expression of MTS-ATP6 fusion protein in ATP6-mutant cybrids

NU

(harboring the m.8993T>G mutation), which consequently led to improved mitochondrial

MA

functions and cell survival (Chin et al., 2018).

3.2. Gene therapy by selective inhibition of mutant mtDNA

D

Mitochondrial heteroplasmy is implicated in various mitochondrial disorders including

PT E

mitochondrial myopathy, MERRF, and MELAS (Ye et al., 2014). In such a state, a certain threshold of mutant mtDNA gene(s) will be required to elicit the corresponding disease condition(s) (Taylor et al., 1997; Schon et al., 2012). A prudent strategy to alleviate such

CE

diseases is to selectively inhibit mutant mtDNA (Taylor et al., 1997; Russell and Turnbull, 2014), thereby mitigating the negative effects of heteroplasmy. This approach has shown

AC

promise in treating mitochondrial ailments that arise due to mutations in both protein- and RNA-coding genes (Tanaka et al., 2002; Hashimoto et al., 2015). In section 3.1, we have discussed how alloptopic expression-mediated gene therapy has found success in correcting the m.8993T>G mutation, which is involved with NARP, LS and complex V deficiency (De Meirleir et al., 2004) (as discussed in section 2). Gene therapy by selective inhibition of mutant mtDNA has also been used as a potent strategy to inhibit the effects of this substitution mutation in the mtDNA. One approach to reach this goal is to use mitochondriadirected restriction endonucleases specific for unique mutations in the mutant mtDNA (Fig. 3A). For this, a group of researchers (Tanaka et al., 2002) at the Gifu International Institute of Biotechnology in Japan transiently expressed the restriction endonuclease SmaI (fused with

ACCEPTED MANUSCRIPT MTS) in cells harboring the m.8993T>G mutation in a heteroplasmic state. SmaI selectively cleaved mutant mtDNA at the guanine residue (CCCGGG) of m.8993, while not affecting the thymine residue (CCCTGG) of m.8993 in the WT mtDNA. The same strategy was also applied by using EcoRI (another restriction endonuclease) instead of SmaI. The plasmid for transfection was constructed by fusing the cDNA encoding pCOIV (pre-sequence of cytochrome c oxidase subunit IV from S. cerevisiae) with pMACSKKII (mammalian expression vector) and either EcoRI or SmaI. The resultant gene therapy vectors were

PT

pMACSKKII-pCOIV-EcoRI and pMACSKKII-pCOIV-SmaI (Tanaka et al., 2002). pCOIV guided the delivery of restriction enzymes selectively into the mitochondria, hence avoiding

RI

cleavage of the nDNA. Interestingly, while SmaI required five transfection and selection cycles

SC

to successfully eliminate the mutant mtDNA, EcoRI needed only one to two cycles (probably due to differential numbers of recognition sites for the restriction enzymes) (Tanaka et al.,

NU

2002). Another restriction endonuclease, XmaI, has also been used to correct the m.8993T>G mutation by selectively targeting mutant mtDNA (Alexeyev et al., 2008).

MA

The research group of Prof. Carlos T. Moraes at the Miller School of Medicine (University of Miami, USA) has done some comprehensive work in using restriction endonucleases to obliterate mutant mtDNA. In an in vitro study (Srivastava and Moraes, 2001), they expressed

D

mitochondria-targeted PstI in murine cells harboring heteroplasmy of mtDNA (the mutant

PT E

containing two PstI-specific sites). In another study (Bayona-Bafaluy et al., 2005), performed both in vivo (brain and muscle of heteroplasmic mice) and in vitro (heteroplasmic mice hepatocytes), they transfected ApaLI fused with MTS of COVIII to selectively ablate the

CE

mutant mtDNA containing ApaLI-specific sites. The group later demonstrated that systemic delivery of viral vectors containing MTS-fused ApaLI to specific organs accomplished in

AC

treating mtDNA-associated disorders (Bacman et al., 2010, 2012). In yet another study, the same group has demonstrated application of mitochondria-targeted ScaI restriction endonuclease to scythe mutant mtDNA, thereby mitigating heteroplasmy-associated dyshomeostasis (Bacman et al., 2007). Although the restriction endonuclease approach has shown significant success, it is unfortunately limited to only a small number of mtDNA mutations with unique restriction sites, which are recognized by these enzymes (Choo and Klug, 1994). To avoid this disadvantage, mtDNA-binding zinc-finger nucleases (mtZFNs) have been engineered, which have preset selectivity for specific mtDNA sequence(s) (Kim et al., 1996; Isalan et al., 2001; Papworth et al., 2006; Minczuk et al., 2006; Gammage et al., 2016). mtZFNs are chimeric enzymes

ACCEPTED MANUSCRIPT containing a Cys2His2 zinc-finger protein (ZFP), which is conjugated with an MTS and the catalytic domain of restriction endonucleases (such as FokI), along with nuclear export signal (NES) peptides. This ensures their localization exclusively in the mitochondria (Minczuk et al., 2006; Gammage et al., 2016). This strategy demonstrated that mtZFNs can specifically localize to the mitochondria when combined with MTS, and can selectively cleave mutant mtDNA (Fig. 3B). The approach was successfully used in reducing the levels of mutant

PT

mtDNA harboring the m.8993T>G mutation (Gammage et al., 2016), which is associated with NARP, LS and complex V deficiency (De Meirleir et al., 2004). The mtZFN-based gene therapy also restored ATP homeostatsis and ΔΨm on selective removal of the mutant mtDNA

RI

(Gammage et al., 2016).

SC

Apart from restriction endonucleases and mtZFNs, there is another potent gene therapy strategy for selective elimination of mutant mtDNA in a heteroplasmic situation. This involves

NU

designing mitochondrially targeted transcription activator-like effector (TALE) nucleases (mitoTALENs), which are novel nucleases (containing FokI catalytic domains) that can

MA

selectively target and cleave mutant mtDNA (Hashimoto et al., 2015; Gammage et al., 2016; Gómez-Tatay et al., 2017) (Fig. 3C). TALENs work as heterodimers, which need two TALEN monomers to bind closely-spaced DNA sequences, allowing their FokI nuclease domains to

D

dimerize and cleave the flanked sequence of the bound DNA (Bacman et al., 2013). In addition

PT E

to the work done in restriction endonuclease-based gene therapy to obliterate mutant mtDNA, Prof. Moraes and his group have also worked on designing mitoTALENs, which demonstrated significant decline in the number of mutant mtDNA molecules in patient–derived cells

CE

(Bacman et al., 2013). This strategy was further applied by the group in reducing mtDNA with m.8344A>G (tRNALys) and m.13513G>A (ND5) mutations (in cybrids), which are respectively

AC

associated with MERRF and MELAS (Jacobs, 2003; Hashimoto et al., 2015). The researchers constructed plasmids coding for mitoTALEN monomers. They removed any nuclear localization signal from the construct, while incorporating (i) an SOD2- or COVIIIA/Su9derived MTS, (ii) a FLAG or hemagglutinin immuno-tag, (iii) a 3′-UTR, (iv) a fluorescent marker (eGFP or mCherry), and (v) a recoded picornaviral 2A-like sequence (T2A) between the mitoTALEN and the fluorescent marker. MitoTALENs showed significant mitigation in the levels of the mutant mtDNA (harboring m.8344A>G or m.13513G>A), along with considerable recovery in respiratory capacity and OX-PHOS efficiency (Hashimoto et al., 2015).

ACCEPTED MANUSCRIPT 3.3. Gene therapy via delivery of WT mtDNA Targeting WT mtDNA (containing the corrective genes) directly into the mitochondrion has emerged as a potent strategy to rectify pathogenic mtDNA mutations (Vaidya et al., 2009; Yu et al., 2012). Carrier-based delivery systems, such as mitochondrial transduction domain (MTD) conjugated to mitochondrial transcription factor A (MTD-TFAM), MITO-porters and DQAsomes, have shown great promise in this mitochondriotropic approach (Niazi et al., 2013).

PT

However, the paramount challenge still remains in understanding the mechanism of nucleic acid import into the mitochondria (Weber-Lotfi et al., 2015). Nevertheless, several mtDNA

RI

mutations, such as m.8993T>G and m.11778G>A, have been successfully tackled by

SC

employing this strategy (Iyer et al., 2012).

In an in vivo study (Yu et al., 2015), transgenic mice were generated as a disease model for

NU

LHON by introducing mutant human ND4 (mutND4) gene (m.11778G>A) into the mice mtDNA through rAAV (containing self-complementary sc-HSP-mutND4-FLAG+mCherry).

MA

The AAV vector was modified by appending a COVIII presequence that facilitated its localization directly to the mitochondria, rather than the nucleus. This strategy caused the mutant ND4 gene to assemble in the mitochondrial complex I, thereby causing visual loss in

D

the mice. The condition was found to be reversed when the animals were treated with

PT E

mitochondria-targeted self-complementary AAV serotype 2 (scAAV2) containing the WT human ND4 (scAAV2-HSP-ND4) (Yu et al., 2015). The m.11778G>A mutation has also been successfully rectified in SH-SY5Y cells using the

CE

protein transduction (protofection) technique (Iyer et al., 2009, 2012). In this approach, mtDNA (corrective, WT) cargo is delivered into the mitochondria using engineered protein

AC

vectors, such as recombinant TFAM conjugated with MTD (combination of an N-terminal protein transduction domain and the MTS of SOD2) (Khan and Bennett, 2004; Iyer et al., 2009; Keeney et al., 2009; Thomas et al., 2011; Iyer et al., 2012; Iyer, 2013). In the MTD-TFAM conjugate, TFAM carries the WT mtDNA cargo, the protein transduction domain facilitates conveyance through the cell membrane, and the MTS ensures mitochondriotropic targeting and uptake (Fig. 4A). This has reportedly corrected the mtDNA mutation and significantly enhanced mitochondrial function (Iyer et al., 2009). This strategy has also shown success in correcting the m.8993T>G mutation in the ATP6 gene, which is involved with the etiologies of NARP, LS and complex V deficiency (De Meirleir et al., 2004). MTD-TFAM conjugated with WT mtDNA improved mitochondrial functions of fibroblast-derived cybrid cells with 95% of mitochondrial genomes bearing the m.8993T>G mutation (Iyer et al., 2012).

ACCEPTED MANUSCRIPT Furthermore, in an in vitro model for PD (cybrids generated by fusing SH-SY5Y ρ0 cells with platelets obtained from PD patients) with reduced complex I activity, MTD-TFAM-mediated delivery of corrective mtDNA into the mitochondria showed increased mitochondrial gene expression with augmented mitochondrial functions (Keeney et al., 2009). Yet another evolving strategy for delivery of WT mtDNA into mitochondria (harboring mutant genomes) is by use of MITO-porters, which are liposome-based nanocarriers that can deliver

PT

nucleic acids, proteins or drugs selectively to the mitochondria via a membrane-fusion mechanism (Niazi et al., 2013; Yamada and Harashima, 2015; Ishikawa et al., 2018). These

RI

MITO-porters can be coated with two membrane-fusogenic outer envelopes to produce the dual function MITO-porters (DF-MITO-porters) (Yamada, 2014). After internalization into

SC

cells, the outer envelope of the DF-MITO-porters fuses with the endosomal membranes (the MITO-porter thus escaping from the endosome sans the outer envelope), while the inner one

NU

fuses with the mitochondrial membranes, thereby delivering the cargo exclusively into the mitochondria (Yamada et al., 2011; Niazi et al., 2013; Yamada, 2014; Yamada and Harashima,

MA

2015) (Fig. 4B). This approach has accomplished in delivering a variety of macromolecules into the mitochondria (Yamada et al., 2008; Yamada and Harashima, 2012; Yamada, 2014; Yamada and Harashima, 2015; Ishikawa et al., 2018). A recent study has reported successful

D

delivery of WT ND4 gene into the mitochondria in both in vitro (HeLa) and in vivo (C57BL/6

PT E

mice) systems (Ishikawa et al., 2018). The researchers designed a construct containing human ND4 gene (pCMV-mtLuc (CGG) [ND4]) and transfected it into the mitochondria by using a MITO-porter (Ishikawa et al., 2018; Yamada et al., 2017) that is conjugated with a DNA-

CE

binding a cationic amphipathic peptide called KALA, which causes membrane destabilization and facilitates DNA transfection (Wyman et al., 1997). These results make it prudent to explore

AC

further in the precincts of the MITO-porter systems to develop novel gene therapy strategies for delivery of WT mtDNA into the mitochondria. Another significant approach for delivery of nucleic acids into the mitochondria involves the application of dequalinium chloride (DQA) as a targeted mitochondrial vesicular delivery system. DQA forms cationic liposome-like nanovesicles (DQAsomes) at concentrations above the critical vesicular values (Weissig et al., 2000; Weissig, 2015). These mitochondriotropic nanocarriers directly transfer exogenous DNA, peptides, oligonucleotides, nucleic acids, pDNA and low molecular weight drugs into the mitochondria (Weissig, 2015) (Fig. 4C). Exogenous DNA along with an MTS can be combined with the DQAsome to form a DQAsome-DNA-MTS complex, which might then be injected into the host cell (D’Souza et

ACCEPTED MANUSCRIPT al., 2005; Lyrawati et al., 2011). Its contact with the mitochondrial membrane results in cleaving of the conjugate from the DQAsome and its selective accumulation on mitochondria. This selectivity is ascribed to the difference in the mitochondrial membrane potential (ΔΨm) and charge gradient across the mitochondrial matrix. The splitting is followed by its entry into the matrix with the help of protein import machinery (Weissig et al., 2000; Weissig and D’Souza, 2004). Hence, DQAsome-mediated transfection of functional mtDNA genes into the

RI

PT

mitochondria is an effective technique of gene therapy.

SC

4. Epilogue: The promises and pitfalls of mitochondrial gene therapy Gene therapy is currently the ‘closest to cure’ approach in the treatment of mitochondrial ailments (Doyle and Chan, 2008). Nevertheless, its strategies have their own share of

NU

impediments, such as concerns regarding the safety of different modes of therapies, stability of the introduced gene(s) over generations, actual in vivo efficacy and the extent to which it can

MA

be applied in practice (Kagawa and Hayashi, 1997; Doyle and Chan, 2008). The complexity in the genotype-phenotype relationship and the process of stochastic segregation of mitochondria make it difficult to diagnose the specific disorder, thereby rendering further complications to

D

design therapeutic approaches using gene therapy (Kagawa and Hayashi, 1997; Doyle and

PT E

Chan, 2008). Moreover, although about 67% of contemporary clinical trials use viral vectors, their immunogenicity and carcinogenicity pose a major challenge (Kamimura et al., 2015). To circumvent these plausible dire outcomes, application of liposome- or nanoparticle-based

CE

delivery systems are gaining significance. However, limited cargo-carrying capacity, low and transient gene expressions of these non-viral delivery systems, plague their ubiquitous use (Yu-

AC

Wai-Man, 2016). Also, for carrying out prospective clinical trials, extensive heterogeneity of mtDNA mutations in patients make it a challenging exercise to perform appropriate sampling of patient groups with significant homogeneity with respect to their genetic disorders, disease progression, biochemical facets and mtDNA mutation load (Kanabus et al., 2014; Rahman, 2015). Individual gene therapy strategies have also been marred with several problems. For example, allotopic expression-mediated gene therapy strategy, although proficient, often gets limited by the need of effective mitochondrial import and accurate incorporation of the allotopically expressed protein (Kyriakouli et al., 2008). Moreover, it has been recently shown that constructs used to deliver allotopically expressed proteins into the mitochondria often result in

ACCEPTED MANUSCRIPT mitochondrial fragmentation (Chin et al., 2018). The hurdles in transition of present gene therapy approaches from laboratory to clinic are, however, not limited to allotopic expressionmediated gene therapy only. For instance, in gene therapy via RE-mediated selective ablation of mutant mtDNA, a major issue is that only a handful of mutations harbor specific restriction sites for known REs (Nightingale et al., 2016). In the mtZFN approach (which moderately overcomes the above-mentioned problem by using ZFPs to bind to a specific DNA sequence),

PT

occurrence of cytotoxicity has been detrimental in molding this approach into a safe clinical intervention tactic (Nightingale et al., 2016). Also, the RE FokI, which is commonly used in this strategy, is known to dimerize, which in turn triggers off-target binding and subsequent

RI

cleavage (Ramalingam et al., 2011). In case of mitoTALENs, a pertinent problem lies in the

SC

fact that, owing to their large size and requirement of two monomers, it is often difficult to pack the large cargo into a suitable vector (Hashimoto et al., 2015). In case of carrier-based

NU

systems such as MITO-porters, selective delivery of the cargo into the mitochondria has been reported to be difficult, particularly during delivery of hydrophobic molecules or macromolecules (Yamada et al., 2011). Also, it has to be noted that most of these strategies

MA

have been predominantly tested in cell-based systems and/or animal models. Their individual efficacy in the clinical scenario still needs to be ascertained.

D

Despite the enormous promise that gene therapy offers, it is indispensible to explore further for

PT E

novel approaches to invoke safer and more efficient strategies to combat mitochondrial conundrums. Although more than five hundred mtDNA mutations are reportedly associated with mitochondrial diseases (Ye et al., 2014), the availability of gene therapy is restricted to

CE

only a handful of mutations, as discussed in section 3. For instance, although substitution mutations in the mitochondrial genome, such as m.4296G>A (tRNAIle) (Martikainen et al.,

AC

2013), m.5523T>G and m.5559A>G (tRNATrp) (Mkaouar-Rebai et al., 2009), m.9185T>C and m.9191T>C (ATP6) (Moslemi et al., 2005), m.9478T>C (COIII) (Mkaouar-Rebai et al., 2011), m.13513G>A and m.13514A>G (ND5) (Bannwarth et al., 2013) are associated with LS, gene therapy exists only for the commonly reported m.8993T>G (ATP6) mutation (Iyer et al., 2012). MELAS, which is caused by m.3243A>G (tRNALeu), m.1642G>A (tRNAVal), m.9957T>C (COIII), m.12770A>G (ND5), m.13045A>C (ND5), m.13514A>G (ND5), and m.3271T>C (tRNALeu) mutations (Tuppen et al., 2010), has so far only received gene therapy for the m.13513G>A (ND5) mutation (Hashimoto et al., 2015). In the case of LHON, gene therapy for mutations like m.3697G>A (ND1) (Spruijt et al., 2007), m.14484T>C (ND6), m.3460G>A (ND1) (Chinnery et al., 2001), and m.4171C>A (ND1) (Kim et al., 2002) is still speculative at best. Mutations implicated in complex V deficiency, such as m.8561C>G (in the overlap region

ACCEPTED MANUSCRIPT of ATP6 and ATP8) (Kytövuori et al., 2016) and m.8969G>A (in ATP6) (Burrage et al., 2014), are yet to have an efficient mode of gene therapy. In spite of the diverse hurdles, mitochondrial gene therapy has evolved exponentially in the recent years owing to the emergence of myriad novel approaches. For example, in polyion complexes (PICs), where methylated carrier molecules (such as poly-L-histidine) containing DNA for the effective transgene expression were used, higher gene expressions were reported

PT

(Asayama et al., 2015). Another emerging technique involves the use of DQAsomes for gene therapy, where augmented in vivo efficacy has been observed, which might be due to the fact

RI

that DQAsomes are not disintegrated by the plasmalemmal anionic lipids (i.e., they do not release the DNA during endocytosis) (Zupančič et al., 2014; Weissig, 2015). Furthermore, they

SC

strongly bind to the exogenous DNA and protect them from nuclease digestion (Zupančič et al., 2014; Weissig, 2015). Yet another advancement in tackling mitochondrial diseases has

NU

been observed in germline therapy, where the mutant mtDNA is replaced by normal mtDNA in the zygote (Kyriakouli et al., 2008). Contemporary breakthroughs in CRISPR/cas9

MA

technology have also shown significant promise in germline therapy of mitochondrial pathologies. This strategy overcomes the ethical concerns associated with embryonic gene therapy by avoiding the necessity of a WT mtDNA donor. The CRISPR/Cas9 technique is

D

easily programmable and facilitates targeting of single gene mutations in mtDNA (Fogleman

PT E

et al., 2016). Recent studies have further utilized a specially engineered Cas9 called mitoCas9, which have localization specificity for the mitochondrial matrix, thereby averting off-target delivery (Gómez-Tatay et al., 2017). In another approach, a 2015 study by Mitalipov and co-

CE

authors at Oregon Health and Science University (USA) used genetically rectified pluripotent stem cells in patients harboring mtDNA mutations (Ma et al., 2015). Among other strategies,

AC

the hydrodynamic injection approach might show potential in mitochondria-targeted gene therapy, where a momentary increase of hydrostatic pressure in the target tissue due to hydrodynamic injection facilitates easy passage of a corrective DNA into the mitochondria (Nagata et al., 2014). Using this approach, mitochondrial anomalies in skeletal muscles of rats have been targeted by hydrodynamically injecting pcDNA3.1(+)-luc plasmid into the limb veins, which resulted in specific delivery of the corrective gene into the mitochondria with minimum and reversible damage to the muscle cells (Yasuzaki et al., 2013, 2015). Therefore, this strategy might be adopted to treat neuromuscular mitochondrial disorders, such as MERRF and MELAS. Although a few vagaries plague the current strategies, the need of the hour is to plunge deeper

ACCEPTED MANUSCRIPT into the available approaches, and to improvise them further in order to bolster their efficacies against myriad mitochondrial diseases. The ‘powerhouse’ of the cell is an indispensable facet of human health, which necessitates further research. The staggering task of curbing mitochondrial pathologies via gene therapy might then be translated from bench to bedside.

Conflict of interests

AC

CE

PT E

D

MA

NU

SC

RI

PT

The authors declare that there is no conflict of interest.

ACCEPTED MANUSCRIPT References Alexeyev, M.F., Venediktova, N., Pastukh, V., Shokolenko, I., Bonilla, G., 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. https://doi.org/10.1038/sj.gt.2008.11

PT

Asayama, S., Kumagai, T., Kawakami, H., 2015. Screening for Methylated Poly(l-histidine) with Various Dimethylimidazolium/Methylimidazole/Imidazole Contents as DNA Carrier. Pharmaceutics 7, 224–232. https://doi.org/10.3390/pharmaceutics7030224

SC

RI

Auré, K., Dubourg, O., Jardel, C., Clarysse, L., Sternberg, D., Fournier, E., Laforêt, P., Streichenberger, N., Petiot, P., Gervais-Bernard, H., Vial, C., Bedat-Millet, A.-L., Drouin-Garraud, V., Bouillaud, F., Vandier, C., Fontaine, B., Lombès, A., 2013. Episodic weakness due to mitochondrial DNA MT-ATP6/8 mutations. Neurology 81, 1810–1818. https://doi.org/10.1212/01.wnl.0000436067.43384.0b

NU

Australian Mitochondrial Disease Foundation, 2017. Mito Info [WWW Document]. URL http://www.amdf.org.au/mito-info/ (accessed 5.2.17).

MA

Bacman, S., Williams, S., Hernandez, D., Moraes, C., 2007. Modulating mtDNA heteroplasmy by mitochondria-targeted restriction endonucleases in a ‘differential multiple cleavagesite’ model. Gene Ther. 14, 1309–1318. https://doi.org/10.1038/sj.gt.3302981

D

Bacman, S.R., Williams, S.L., Duan, D., 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. https://doi.org/10.1038/gt.2011.196

PT E

Bacman, S.R., Williams, S.L., Garcia, S., Moraes, C.T., 2010. Organ-specific shifts in mtDNA heteroplasmy following systemic delivery of a mitochondria-targeted restriction endonuclease. Gene Ther. 17, 713–720. https://doi.org/10.1038/gt.2010.25

CE

Bacman, S.R., Williams, S.L., Pinto, M., Peralta, S., Moraes, C.T., 2013. Specific elimination of mutant mitochondrial genomes in patient–derived cells by mitoTALENs. Nat. Med. 19, 1111–1113. https://doi.org/10.1038/nm.3261

AC

Bannwarth, S., Procaccio, V., Lebre, A.S., Jardel, C., Chaussenot, A., Hoarau, C., Maoulida, H., Charrier, N., Gai, X., Xie, H.M., Ferre, M., Fragaki, K., Hardy, G., Mousson de Camaret, B., Marlin, S., Dhaenens, C.M., Slama, A., Rocher, C., Paul Bonnefont, J., Rötig, A., Aoutil, N., Gilleron, M., Desquiret-Dumas, V., Reynier, P., Ceresuela, J., Jonard, L., Devos, A., Espil-Taris, C., Martinez, D., Gaignard, P., Le Quan Sang, K.H., Amati-Bonneau, P., Falk, M.J., Florentz, C., Chabrol, B., Durand-Zaleski, I., Paquis-Flucklinger, V., 2013. Prevalence of rare mitochondrial DNA mutations in mitochondrial disorders. J. Med. Genet. 50, 704–714. https://doi.org/10.1136/jmedgenet-2013-101604 Bayona-Bafaluy, M.P., Blits, B., Battersby, B.J., Shoubridge, E.A., 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. https://doi.org/10.1073/pnas.0502896102

ACCEPTED MANUSCRIPT Birch-Machin, M.A., Bowman, A., 2016. Oxidative stress and ageing. Br. J. Dermatol. 175, 26–29. https://doi.org/10.1111/bjd.14906 Bonnet, C., Kaltimbacher, V., Ellouze, S., Augustin, S., Bénit, P., Forster, V., Rustin, P., Sahel, J.-A., Corral-Debrinski, M., 2007a. 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. https://doi.org/10.1089/rej.2006.0526

PT

Bonnet, C., Kaltimbacher, V., Ellouze, S., Forster, V., Sahel, J.-A., Corral-Debrinski, M., 2007b. [mRNA localization to the mitochondrial surface: a tool to treat retinal pathologies due to mitochondrial DNA mutations]. J. Société Biol. 201, 69–74.

SC

RI

Boominathan, A., Vanhoozer, S., Basisty, N., Powers, K., Crampton, A.L., Wang, X., Friedricks, N., Schilling, B., Brand, M.D., O’Connor, M.S., 2016. Stable nuclear expression of ATP8 and ATP6 genes rescues a mtDNA Complex V null mutant. Nucleic Acids Res. 44, 9342–9357. https://doi.org/10.1093/nar/gkw756

MA

NU

Burrage, L.C., Tang, S., Wang, J., Donti, T.R., Walkiewicz, M., Luchak, J.M., Chen, L.-C., Schmitt, E.S., Niu, Z., Erana, R., Hunter, J.V., Graham, B.H., Wong, L.-J., Scaglia, F., 2014. Mitochondrial myopathy, lactic acidosis, and sideroblastic anemia (MLASA) plus associated with a novel de novo mutation (m.8969G>A) in the mitochondrial encoded ATP6 gene. Mol. Genet. Metab. 113, 207–212. https://doi.org/10.1016/j.ymgme.2014.06.004

D

Camus, M.F., Clancy, D.J., Dowling, D.K., 2012. Mitochondria, Maternal Inheritance, and Male Aging. Curr. Biol. 22, 1717–1721. https://doi.org/10.1016/j.cub.2012.07.018

PT E

Carelli, V., Ross-Cisneros, F.N., Sadun, A.A., 2004. Mitochondrial dysfunction as a cause of optic neuropathies. Prog. Retin. Eye Res. 23, 53–89. https://doi.org/10.1016/j.preteyeres.2003.10.003

AC

CE

Chadderton, N., Palfi, A., Millington-Ward, S., Gobbo, O., Overlack, N., Carrigan, M., O’Reilly, M., Campbell, M., Ehrhardt, C., Wolfrum, U., Humphries, P., Kenna, P.F., Farrar, G.J., 2013. Intravitreal delivery of AAV-NDI1 provides functional benefit in a murine model of Leber hereditary optic neuropathy. Eur. J. Hum. Genet. EJHG 21, 62– 68. https://doi.org/10.1038/ejhg.2012.112 Chan, D.C., 2006. Mitochondria: Dynamic Organelles in Disease, Aging, and Development. Cell 125, 1241–1252. https://doi.org/10.1016/j.cell.2006.06.010 Chin, R.M., Panavas, T., Brown, J.M., Johnson, K.K., 2018. Optimized Mitochondrial Targeting of Proteins Encoded by Modified mRNAs Rescues Cells Harboring Mutations in mtATP6. Cell Rep. 22, 2818–2826. https://doi.org/10.1016/j.celrep.2018.02.059 Chinnery, P.F., Brown, D.T., Andrews, R.M., Singh-Kler, R., Riordan-Eva, P., Lindley, J., Applegarth, D.A., Turnbull, D.M., Howell, N., 2001. The mitochondrial ND6 gene is a hot spot for mutations that cause Leber’s hereditary optic neuropathy. Brain J. Neurol. 124, 209–218.

ACCEPTED MANUSCRIPT Choo, Y., Klug, A., 1994. Selection of DNA binding sites for zinc fingers using rationally randomized DNA reveals coded interactions. Proc. Natl. Acad. Sci. U. S. A. 91, 11168– 11172. Choong, C.-J., Baba, K., Mochizuki, H., 2015. Gene therapy for neurological disorders. Expert Opin. Biol. Ther. 0, 1–17. https://doi.org/10.1517/14712598.2016.1114096

PT

Coxhead, J., Kurzawa-Akanbi, M., Hussain, R., Pyle, A., Chinnery, P., Hudson, G., 2016. Somatic mtDNA variation is an important component of Parkinson’s disease. Neurobiol. Aging 38, 217.e1-217.e6. https://doi.org/10.1016/j.neurobiolaging.2015.10.036

SC

RI

Cwerman-Thibault, H., Augustin, S., Lechauve, C., Ayache, J., Ellouze, S., Sahel, J.-A., Corral-Debrinski, M., 2015. Nuclear expression of mitochondrial ND4 leads to the protein assembling in complex I and prevents optic atrophy and visual loss. Mol. Ther. Methods Clin. Dev. 2, 15003. https://doi.org/10.1038/mtm.2015.3

NU

Das, P., Guha, G., 2010. Aging and Mitochondrial DNA. J. Sci. Res. 3, 177. https://doi.org/10.3329/jsr.v3i1.5078 Davis, R.L., Liang, C., Sue, C.M., 2018. Mitochondrial diseases. Handb. Clin. Neurol. 147, 125–141. https://doi.org/10.1016/B978-0-444-63233-3.00010-5

MA

De Meirleir, L., Seneca, S., Lissens, W., De Clercq, I., Eyskens, F., Gerlo, E., Smet, J., Van Coster, R., 2004. Respiratory chain complex V deficiency due to a mutation in the assembly gene ATP12. J. Med. Genet. 41, 120–124.

PT E

D

De Vivo, D.C., 1993. The expanding clinical spectrum of mitochondrial diseases. Brain Dev. 15, 1–22. DiMauro, S., Schon, E.A., Carelli, V., Hirano, M., 2013. The clinical maze of mitochondrial neurology. Nat. Rev. Neurol. 9, 429–444. https://doi.org/10.1038/nrneurol.2013.126

CE

Dipchand, A.I., Tein, I., Robinson, B., Benson, L.N., 2001. Maternally inherited hypertrophic cardiomyopathy: a manifestation of mitochondrial DNA mutations--clinical course in two families. Pediatr. Cardiol. 22, 14–22. https://doi.org/10.1007/s002460010145

AC

Doyle, S.R., Chan, C.K., 2008. Mitochondrial gene therapy: an evaluation of strategies for the treatment of mitochondrial DNA disorders. Hum. Gene Ther. 19, 1335–1348. https://doi.org/10.1089/hum.2008.090 D’Souza, G.G.M., Boddapati, S.V., Weissig, V., 2005. Mitochondrial leader sequence-plasmid DNA conjugates delivered into mammalian cells by DQAsomes co-localize with mitochondria. Mitochondrion 5, 352–358. https://doi.org/10.1016/j.mito.2005.07.001 Duff, R.M., Shearwood, A.-M.J., Ermer, J., Rossetti, G., Gooding, R., Richman, T.R., Balasubramaniam, S., Thorburn, D.R., Rackham, O., Lamont, P.J., Filipovska, A., 2015. A mutation in MT-TW causes a tRNA processing defect and reduced mitochondrial function in a family with Leigh syndrome. Mitochondrion 25, 113–119. https://doi.org/10.1016/j.mito.2015.10.008

ACCEPTED MANUSCRIPT Dunn, D.A., Pinkert, C.A., 2012. Nuclear Expression of a Mitochondrial DNA Gene: Mitochondrial Targeting of Allotopically Expressed Mutant ATP6 in Transgenic Mice. J. Biomed. Biotechnol. 2012. https://doi.org/10.1155/2012/541245 Ellouze, S., Augustin, S., Bouaita, A., Bonnet, C., Simonutti, M., Forster, V., Picaud, S., Sahel, J.-A., Corral-Debrinski, M., 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. https://doi.org/10.1016/j.ajhg.2008.08.013

RI

PT

Faridi, U., Dhawan, S.S., Pal, S., Gupta, S., Shukla, A.K., Darokar, M.P., Sharma, A., Shasany, A.K., 2016. Repurposing L-Menthol for Systems Medicine and Cancer Therapeutics? L-Menthol Induces Apoptosis through Caspase 10 and by Suppressing HSP90. OMICS J. Integr. Biol. 20, 53–64. https://doi.org/10.1089/omi.2015.0118

SC

Feuer, W.J., Schiffman, J.C., Davis, J.L., Porciatti, V., Gonzalez, P., Koilkonda, R.D., Yuan, H., Lalwani, A., Lam, B.L., Guy, J., 2016. Gene Therapy for Leber Hereditary Optic Neuropathy: Initial Results. Ophthalmology 123, 558–570. https://doi.org/10.1016/j.ophtha.2015.10.025

NU

Finsterer, J., 2008. Leigh and Leigh-Like Syndrome in Children and Adults. Pediatr. Neurol. 39, 223–235. https://doi.org/10.1016/j.pediatrneurol.2008.07.013

MA

Fogleman, S., Santana, C., Bishop, C., Miller, A., Capco, D.G., 2016. CRISPR/Cas9 and mitochondrial gene replacement therapy: promising techniques and ethical considerations. Am. J. Stem Cells 5, 39.

PT E

D

Frank, S., Gaume, B., Bergmann-Leitner, E.S., Leitner, W.W., Robert, E.G., Catez, F., Smith, C.L., Youle, R.J., 2001. The role of dynamin-related protein 1, a mediator of mitochondrial fission, in apoptosis. Dev. Cell 1, 515–525.

CE

Gammage, P.A., Gaude, E., Van Haute, L., Rebelo-Guiomar, P., Jackson, C.B., Rorbach, J., Pekalski, M.L., Robinson, A.J., Charpentier, M., Concordet, J.-P., Frezza, C., Minczuk, M., 2016. Near-complete elimination of mutant mtDNA by iterative or dynamic dosecontrolled treatment with mtZFNs. Nucleic Acids Res. 44, 7804–7816. https://doi.org/10.1093/nar/gkw676

AC

Gao, G.-P., Alvira, M.R., Wang, L., Calcedo, R., Johnston, J., Wilson, J.M., 2002. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc. Natl. Acad. Sci. U. S. A. 99, 11854–11859. https://doi.org/10.1073/pnas.182412299 Gerards, M., Sallevelt, S.C.E.H., Smeets, H.J.M., 2016. Leigh syndrome: Resolving the clinical and genetic heterogeneity paves the way for treatment options. Mol. Genet. Metab. 117, 300–312. https://doi.org/10.1016/j.ymgme.2015.12.004 Gibson, S.A., Gao, G.-D., McDonagh, K., Shen, S., 2012. Progress on stem cell research towards the treatment of Parkinson’s disease. Stem Cell Res. Ther. 3, 11. https://doi.org/10.1186/scrt102 Ginn, S.L., Alexander, I.E., Edelstein, M.L., Abedi, M.R., Wixon, J., 2013. Gene therapy clinical trials worldwide to 2012 – an update. J. Gene Med. 15, 65–77. https://doi.org/10.1002/jgm.2698

ACCEPTED MANUSCRIPT Gómez-Tatay, L., Hernández-Andreu, J.M., Aznar, J., 2017. Mitochondrial Modification Techniques and Ethical Issues. J. Clin. Med. 6. https://doi.org/10.3390/jcm6030025 Görlach, A., Dimova, E.Y., Petry, A., Martínez-Ruiz, A., Hernansanz-Agustín, P., Rolo, A.P., Palmeira, C.M., Kietzmann, T., 2015. Reactive oxygen species, nutrition, hypoxia and diseases: Problems solved? Redox Biol. 6, 372–385. https://doi.org/10.1016/j.redox.2015.08.016

RI

PT

Gorman, G.S., Schaefer, A.M., Ng, Y., Gomez, N., Blakely, E.L., Alston, C.L., Feeney, C., Horvath, R., Yu-Wai-Man, P., Chinnery, P.F., Taylor, R.W., Turnbull, D.M., McFarland, R., 2015. Prevalence of nuclear and mitochondrial DNA mutations related to adult mitochondrial disease. Ann. Neurol. 77, 753–759. https://doi.org/10.1002/ana.24362

SC

Goto, Y., Nonaka, I., Horai, S., 1990. A mutation in the tRNA(Leu)(UUR) gene associated with the MELAS subgroup of mitochondrial encephalomyopathies. Nature 348, 651– 653. https://doi.org/10.1038/348651a0

MA

NU

Guy, J., 2018. Clinical Trial (NCT02161380): An Open-label Dose Escalation Study of an Adeno-associated Virus Vector (scAAV2-P1ND4v2) for Gene Therapy of Leber’s Hereditary Optic Neuropathy (LHON) Caused by the G11778A Mutation in Mitochondrial DNA [WWW Document]. ClinicalTrials.gov. URL https://clinicaltrials.gov/ct2/show/NCT02161380 (accessed 5.8.18).

D

Guy, J., Qi, X., Pallotti, F., Schon, E.A., Manfredi, G., Carelli, V., Martinuzzi, A., Hauswirth, W.W., Lewin, A.S., 2002. Rescue of a mitochondrial deficiency causing Leber Hereditary Optic Neuropathy. Ann. Neurol. 52, 534–542. https://doi.org/10.1002/ana.10354

PT E

Haas, R.H., Parikh, S., Falk, M.J., Saneto, R.P., Wolf, N.I., Darin, N., Cohen, B.H., 2007. Mitochondrial Disease: A Practical Approach for Primary Care Physicians. Pediatrics 120, 1326–1333. https://doi.org/10.1542/peds.2007-0391

AC

CE

Hashimoto, M., Bacman, S.R., Peralta, S., Falk, M.J., Chomyn, A., Chan, D.C., Williams, S.L., Moraes, C.T., 2015. MitoTALEN: A General Approach to Reduce Mutant mtDNA Loads and Restore Oxidative Phosphorylation Function in Mitochondrial Diseases. Mol. Ther. J. Am. Soc. Gene Ther. 23, 1592–1599. https://doi.org/10.1038/mt.2015.126 Heller, A., Brockhoff, G., Goepferich, A., 2012. Targeting drugs to mitochondria. Eur. J. Pharm. Biopharm. Off. J. Arbeitsgemeinschaft Für Pharm. Verfahrenstechnik EV 82, 1–18. https://doi.org/10.1016/j.ejpb.2012.05.014 Herold, M.J., Kuss, A.W., Kraus, C., Berberich, I., 2002. Mitochondria-Dependent Caspase-9 Activation Is Necessary for Antigen Receptor-Mediated Effector Caspase Activation and Apoptosis in WEHI 231 Lymphoma Cells. J. Immunol. 168, 3902–3909. https://doi.org/10.4049/jimmunol.168.8.3902 Holt, I.J., Harding, A.E., Morgan-Hughes, J.A., 1989. Deletions of muscle mitochondrial DNA in mitochondrial myopathies: sequence analysis and possible mechanisms. Nucleic Acids Res. 17, 4465–4469.

ACCEPTED MANUSCRIPT Hoptasz, M., Szczuciński, A., Losy, J., 2014. Heterogeneous phenotypic manifestations of maternally inherited deafness associated with the mitochondrial A3243G mutation. Case report. Neurol. Neurochir. Pol. 48, 150–153. https://doi.org/10.1016/j.pjnns.2013.12.007 Isalan, M., Klug, A., Choo, Y., 2001. A rapid, generally applicable method to engineer zinc fingers illustrated by targeting the HIV-1 promoter. Nat. Biotechnol. 19, 656–660. https://doi.org/10.1038/90264

PT

Ishikawa, T., Somiya, K., Munechika, R., Harashima, H., Yamada, Y., 2018. Mitochondrial transgene expression via an artificial mitochondrial DNA vector in cells from a patient with a mitochondrial disease. J. Control. Release Off. J. Control. Release Soc. 274, 109–117. https://doi.org/10.1016/j.jconrel.2018.02.005

SC

RI

Iyer, S., 2013. Novel Therapeutic Approaches for Leber’s Hereditary Optic Neuropathy. Discov. Med. 15, 141–149.

NU

Iyer, S., Bergquist, K., Young, K., Gnaiger, E., Rao, R.R., Bennett, J.P., 2012. Mitochondrial gene therapy improves respiration, biogenesis, and transcription in G11778A Leber’s hereditary optic neuropathy and T8993G Leigh’s syndrome cells. Hum. Gene Ther. 23, 647–657. https://doi.org/10.1089/hum.2011.177

MA

Iyer, S., Thomas, R.R., Portell, F.R., Dunham, L.D., Quigley, C.K., Bennett, J.P., 2009. Recombinant Mitochondrial Transcription Factor A with N-terminal Mitochondrial Transduction Domain Increases Respiration and Mitochondrial Gene Expression. Mitochondrion 9, 196–203. https://doi.org/10.1016/j.mito.2009.01.012

PT E

D

Jacobs, H.T., 2003. Disorders of mitochondrial protein synthesis. Hum. Mol. Genet. 12 Spec No 2, R293-301. https://doi.org/10.1093/hmg/ddg285

CE

Jonckheere, A.I., Hogeveen, M., Nijtmans, L.G.J., van den Brand, M. a. M., Janssen, A.J.M., Diepstra, J.H.S., van den Brandt, F.C.A., van den Heuvel, L.P., Hol, F.A., Hofste, T.G.J., Kapusta, L., Dillmann, U., Shamdeen, M.G., Smeitink, J. a. M., Rodenburg, R.J.T., 2008. A novel mitochondrial ATP8 gene mutation in a patient with apical hypertrophic cardiomyopathy and neuropathy. J. Med. Genet. 45, 129–133. https://doi.org/10.1136/jmg.2007.052084

AC

Joseph, L.C., Barca, E., Subramanyam, P., Komrowski, M., Pajvani, U., Colecraft, H.M., Hirano, M., Morrow, J.P., 2016. Inhibition of NAPDH Oxidase 2 (NOX2) Prevents Oxidative Stress and Mitochondrial Abnormalities Caused by Saturated Fat in Cardiomyocytes. PLoS ONE 11, e0145750. https://doi.org/10.1371/journal.pone.0145750 Kabunga, P., Lau, A.K., Phan, K., Puranik, R., Liang, C., Davis, R.L., Sue, C.M., Sy, R.W., 2015. Systematic review of cardiac electrical disease in Kearns–Sayre syndrome and mitochondrial cytopathy. Int. J. Cardiol. 181, 303–310. https://doi.org/10.1016/j.ijcard.2014.12.038 Kagawa, Y., Hayashi, J.I., 1997. Gene therapy of mitochondrial diseases using human cytoplasts. Gene Ther. 4, 6–10. https://doi.org/10.1038/sj.gt.3300347 Kaltimbacher, V., Bonnet, C., Lecoeuvre, G., Forster, V., Sahel, J.-A., Corral-Debrinski, M., 2006. mRNA localization to the mitochondrial surface allows the efficient translocation

ACCEPTED MANUSCRIPT inside the organelle of a nuclear recoded ATP6 protein. RNA 12, 1408–1417. https://doi.org/10.1261/rna.18206 Kambe, Y., Miyata, A., 2016. Mitochondrial c-Fos May Increase the Vulnerability of Neuro2a Cells to Cellular Stressors. J. Mol. Neurosci. MN. https://doi.org/10.1007/s12031-0150710-7

PT

Kamimura, K., Yokoo, T., Abe, H., Kobayashi, Y., Ogawa, K., Shinagawa, Y., Inoue, R., Terai, S., 2015. Image-Guided Hydrodynamic Gene Delivery: Current Status and Future Directions. Pharmaceutics 7, 213–223. https://doi.org/10.3390/pharmaceutics7030213

RI

Kanabus, M., Heales, S.J., Rahman, S., 2014. Development of pharmacological strategies for mitochondrial disorders. Br. J. Pharmacol. 171, 1798–1817. https://doi.org/10.1111/bph.12456

SC

Karikó, K., Buckstein, M., Ni, H., Weissman, D., 2005. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity 23, 165–175. https://doi.org/10.1016/j.immuni.2005.06.008

NU

Kaufmann, K.B., Büning, H., Galy, A., Schambach, A., Grez, M., 2013. Gene therapy on the move. EMBO Mol. Med. 5, 1642–1661. https://doi.org/10.1002/emmm.201202287

MA

Kaurola, P., Sharma, V., Vonk, A., Vattulainen, I., Róg, T., 2016. Distribution and dynamics of quinones in the lipid bilayer mimicking the inner membrane of mitochondria. Biochim. Biophys. Acta BBA Biomembr. 1858, 2116–2122. https://doi.org/10.1016/j.bbamem.2016.06.016

PT E

D

Keeney, P.M., Quigley, C.K., Dunham, L.D., Papageorge, C.M., Iyer, S., Thomas, R.R., Schwarz, K.M., Trimmer, P.A., Khan, S.M., Portell, F.R., Bergquist, K.E., Bennett, J.P., 2009. Mitochondrial gene therapy augments mitochondrial physiology in a Parkinson’s disease cell model. Hum. Gene Ther. 20, 897–907. https://doi.org/10.1089/hum.2009.023

CE

Khan, S.M., Bennett, J.P., 2004. Development of mitochondrial gene replacement therapy. J. Bioenerg. Biomembr. 36, 387–393. https://doi.org/10.1023/B:JOBB.0000041773.20072.9e

AC

Kim, J.Y., Hwang, J.-M., Park, S.S., 2002. Mitochondrial DNA C4171A/ND1 is a novel primary causative mutation of Leber’s hereditary optic neuropathy with a good prognosis. Ann. Neurol. 51, 630–634. https://doi.org/10.1002/ana.10177 Kim, Y.G., Cha, J., Chandrasegaran, S., 1996. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc. Natl. Acad. Sci. U. S. A. 93, 1156–1160. Koilkonda, R.D., Chou, T.-H., Porciatti, V., Hauswirth, W.W., Guy, J., 2010. Induction of rapid and highly efficient expression of the human ND4 complex I subunit in the mouse visual system by self-complementary adeno-associated virus. Arch. Ophthalmol. Chic. Ill 1960 128, 876–883. https://doi.org/10.1001/archophthalmol.2010.135 Kwiecien, S., Jasnos, K., Magierowski, M., Sliwowski, Z., Pajdo, R., Brzozowski, B., Mach, T., Wojcik, D., Brzozowski, T., 2014. Lipid peroxidation, reactive oxygen species and antioxidative factors in the pathogenesis of gastric mucosal lesions and mechanism of

ACCEPTED MANUSCRIPT protection against oxidative stress - induced gastric injury. J. Physiol. Pharmacol. Off. J. Pol. Physiol. Soc. 65, 613–622. Kyriakouli, D.S., Boesch, P., Taylor, R.W., Lightowlers, R.N., 2008. Progress and prospects: gene therapy for mitochondrial DNA disease. Gene Ther. 15, 1017–1023. https://doi.org/10.1038/gt.2008.91

PT

Kytövuori, L., Lipponen, J., Rusanen, H., Komulainen, T., Martikainen, M.H., Majamaa, K., 2016. A novel mutation m.8561C>G in MT-ATP6/8 causing a mitochondrial syndrome with ataxia, peripheral neuropathy, diabetes mellitus, and hypergonadotropic hypogonadism. J. Neurol. https://doi.org/10.1007/s00415-016-8249-2

RI

Lake, N.J., Bird, M.J., Isohanni, P., Paetau, A., 2015. Leigh Syndrome. J. Neuropathol. Exp. Neurol. 74, 482–492. https://doi.org/10.1097/NEN.0000000000000195

SC

Lee, H.-C., Wei, Y.-H., 2007. Oxidative stress, mitochondrial DNA mutation, and apoptosis in aging. Exp. Biol. Med. Maywood NJ 232, 592–606.

NU

Lenzen, S., 2008. Oxidative stress: the vulnerable β-cell. Biochem. Soc. Trans. 36, 343–347. https://doi.org/10.1042/BST0360343

MA

Leslie, N., Wang, X., Peng, Y., Valencia, C.A., Khuchua, Z., Hata, J., Witte, D., Huang, T., Bove, K.E., 2016. Neonatal multiorgan failure due to ACAD9 mutation and complex I deficiency with mitochondrial hyperplasia in liver, cardiac myocytes, skeletal muscle, and renal tubules. Hum. Pathol. 49, 27–32. https://doi.org/10.1016/j.humpath.2015.09.039

PT E

D

Lin, M.T., Cantuti-Castelvetri, I., Zheng, K., Jackson, K.E., Tan, Y.B., Arzberger, T., Lees, A.J., Betensky, R.A., Beal, M.F., Simon, D.K., 2012. Somatic mitochondrial DNA mutations in early Parkinson and incidental Lewy body disease. Ann. Neurol. 71, 850– 854. https://doi.org/10.1002/ana.23568

CE

Lyrawati, D., Trounson, A., Cram, D., 2011. Expression of GFP in the mitochondrial compartment using DQAsome-mediated delivery of an artificial mini-mitochondrial genome. Pharm. Res. 28, 2848–2862. https://doi.org/10.1007/s11095-011-0544-0

AC

Ma, H., Folmes, C.D.L., Wu, J., Morey, R., Mora-Castilla, S., Ocampo, A., Ma, L., Poulton, J., Wang, X., Ahmed, R., Kang, E., Lee, Y., Hayama, T., Li, Y., Van Dyken, C., Gutierrez, N.M., Tippner-Hedges, R., Koski, A., Mitalipov, N., Amato, P., Wolf, D.P., Huang, T., Terzic, A., Laurent, L.C., Izpisua Belmonte, J.C., Mitalipov, S., 2015. Metabolic rescue in pluripotent cells from patients with mtDNA disease. Nature 524, 234–238. https://doi.org/10.1038/nature14546 Manfredi, G., Fu, J., Ojaimi, J., Sadlock, J.E., Kwong, J.Q., Guy, J., Schon, E.A., 2002. Rescue of a deficiency in ATP synthesis by transfer of MTATP6, a mitochondrial DNAencoded gene, to the nucleus. Nat. Genet. 30, 394–399. https://doi.org/10.1038/ng851 Mangiafico, R.A., Zeviani, M., Bartoloni, G., Fiore, C.E., 2004. Accelerated cardiomyopathy in maternally inherited diabetes and deafness. Int. J. Clin. Pharmacol. Res. 24, 15–21. Marella, M., Seo, B.B., Flotte, T.R., Matsuno-Yagi, A., Yagi, T., 2011. No Immune Responses by the Expression of the Yeast Ndi1 Protein in Rats. PLoS ONE 6. https://doi.org/10.1371/journal.pone.0025910

ACCEPTED MANUSCRIPT Marella, M., Seo, B.B., Thomas, B.B., Matsuno-Yagi, A., Yagi, T., 2010. Successful Amelioration of Mitochondrial Optic Neuropathy Using the Yeast NDI1 Gene in a Rat Animal Model. PLoS ONE 5. https://doi.org/10.1371/journal.pone.0011472 Martikainen, M.H., Kytövuori, L., Majamaa, K., 2013. Juvenile parkinsonism, hypogonadism and Leigh-like MRI changes in a patient with m.4296G>A mutation in mitochondrial DNA. Mitochondrion 13, 83–86. https://doi.org/10.1016/j.mito.2013.01.012

PT

Minczuk, M., Papworth, M.A., Kolasinska, P., Murphy, M.P., Klug, A., 2006. Sequencespecific modification of mitochondrial DNA using a chimeric zinc finger methylase. Proc. Natl. Acad. Sci. U. S. A. 103, 19689–19694. https://doi.org/10.1073/pnas.0609502103

SC

RI

Mishmar, D., Ruiz-Pesini, E., Golik, P., Macaulay, V., Clark, A.G., Hosseini, S., Brandon, M., Easley, K., Chen, E., Brown, M.D., Sukernik, R.I., Olckers, A., Wallace, D.C., 2003. Natural selection shaped regional mtDNA variation in humans. Proc. Natl. Acad. Sci. 100, 171–176. https://doi.org/10.1073/pnas.0136972100

MA

NU

Mkaouar-Rebai, E., Chamkha, I., Kammoun, F., Kammoun, T., Aloulou, H., Hachicha, M., Triki, C., Fakhfakh, F., 2009. Two new mutations in the MT-TW gene leading to the disruption of the secondary structure of the tRNA(Trp) in patients with Leigh syndrome. Mol. Genet. Metab. 97, 179–184. https://doi.org/10.1016/j.ymgme.2009.03.003

D

Mkaouar-Rebai, E., Ellouze, E., Chamkha, I., Kammoun, F., Triki, C., Fakhfakh, F., 2011. Molecular-clinical correlation in a family with a novel heteroplasmic Leigh syndrome missense mutation in the mitochondrial cytochrome c oxidase III gene. J. Child Neurol. 26, 12–20. https://doi.org/10.1177/0883073810371227

PT E

Moggio, M., Colombo, I., Peverelli, L., Villa, L., Xhani, R., Testolin, S., Di Mauro, S., Sciacco, M., 2014. Mitochondrial disease heterogeneity: a prognostic challenge. Acta Myol. Myopathies Cardiomyopathies Off. J. Mediterr. Soc. Myol. 33, 86–93.

CE

Montgomery, M.K., Turner, N., 2015. Mitochondrial dysfunction and insulin resistance: an update. Endocr. Connect. 4, R1–R15. https://doi.org/10.1530/EC-14-0092

AC

Moon, H.E., Paek, S.H., 2015. Mitochondrial Dysfunction in Parkinson’s Disease. Exp. Neurobiol. 24, 103–116. https://doi.org/10.5607/en.2015.24.2.103 Moslemi, A.-R., Darin, N., Tulinius, M., Oldfors, A., Holme, E., 2005. Two new mutations in the MTATP6 gene associated with Leigh syndrome. Neuropediatrics 36, 314–318. https://doi.org/10.1055/s-2005-872845 Nagata, K., Itaka, K., Baba, M., Uchida, S., Ishii, T., Kataoka, K., 2014. Muscle-targeted hydrodynamic gene introduction of insulin-like growth factor-1 using polyplex nanomicelle to treat peripheral nerve injury. J. Controlled Release 183, 27–34. https://doi.org/10.1016/j.jconrel.2014.03.021 Nagley, P., Farrell, L.B., Gearing, D.P., Nero, D., Meltzer, S., Devenish, R.J., 1988. Assembly of functional proton-translocating ATPase complex in yeast mitochondria with cytoplasmically synthesized subunit 8, a polypeptide normally encoded within the organelle. Proc. Natl. Acad. Sci. U. S. A. 85, 2091–2095.

ACCEPTED MANUSCRIPT National Organization for Rare Disorders, 2016. Leigh Syndrome [WWW Document]. NORD Natl. Organ. Rare Disord. URL https://rarediseases.org/rare-diseases/leigh-syndrome/ (accessed 1.31.17). Nesbitt, V., Alston, C.L., Blakely, E.L., Fratter, C., Feeney, C.L., Poulton, J., Brown, G.K., Turnbull, D.M., Taylor, R.W., McFarland, R., 2014. A national perspective on prenatal testing for mitochondrial disease. Eur. J. Hum. Genet. 22, 1255–1259. https://doi.org/10.1038/ejhg.2014.35

RI

PT

Newman, N.J., 2018. Clinical Trial (NCT02652767): A Randomized, Double-Masked, ShamControlled Clinical Trial to Evaluate the Efficacy of a Single Intravitreal Injection of GS010 in Subjects Affected for 6 Months or Less by LHON Due to the G11778A Mutation in the Mitochondrial ND4 Gene [WWW Document]. ClinicalTrials.gov. URL https://clinicaltrials.gov/ct2/show/NCT02652767 (accessed 5.8.18).

SC

Newman, N.J., 1993. Leber’s hereditary optic neuropathy. New genetic considerations. Arch. Neurol. 50, 540–548.

NU

Ng, Y.S., Turnbull, D.M., 2016. Mitochondrial disease: genetics and management. J. Neurol. 263, 179–191. https://doi.org/10.1007/s00415-015-7884-3

MA

Niazi, A.K., Mileshina, D., Cosset, A., Val, R., Weber-Lotfi, F., Dietrich, A., 2013. Targeting nucleic acids into mitochondria: progress and prospects. Mitochondrion 13, 548–558. https://doi.org/10.1016/j.mito.2012.05.004

D

Nightingale, H., Pfeffer, G., Bargiela, D., Horvath, R., Chinnery, P.F., 2016. Emerging therapies for mitochondrial disorders. Brain 139, 1633–1648. https://doi.org/10.1093/brain/aww081

PT E

Omura, T., 1998. Mitochondria-targeting sequence, a multi-role sorting sequence recognized at all steps of protein import into mitochondria. J. Biochem. (Tokyo) 123, 1010–1016.

CE

Orphanet, 2017. NARP syndrome [WWW Document]. URL http://www.orpha.net/consor/cgibin/OC_Exp.php?Lng=GB&Expert=644 (accessed 2.9.17).

AC

Palomo, G.M., Manfredi, G., 2015. Exploring new pathways of neurodegeneration in ALS: The role of mitochondria quality control. Brain Res., The multifaceted nature of ALS. Discoveries and Challenges of the last 5 years 1607, 36–46. https://doi.org/10.1016/j.brainres.2014.09.065 Papworth, M., Kolasinska, P., Minczuk, M., 2006. Designer zinc-finger proteins and their applications. Gene 366, 27–38. https://doi.org/10.1016/j.gene.2005.09.011 Picard, M., Zhang, J., Hancock, S., Derbeneva, O., Golhar, R., Golik, P., O’Hearn, S., Levy, S., Potluri, P., Lvova, M., Davila, A., Lin, C.S., Perin, J.C., Rappaport, E.F., Hakonarson, H., Trounce, I.A., Procaccio, V., Wallace, D.C., 2014. Progressive increase in mtDNA 3243A>G heteroplasmy causes abrupt transcriptional reprogramming. Proc. Natl. Acad. Sci. 111, E4033–E4042. https://doi.org/10.1073/pnas.1414028111 Praeter, C.D., Vanlander, A., Vanhaesebrouck, P., Smet, J., Seneca, S., Sutter, P.D., Coster, R.V., 2014. Extremely high mutation load of the mitochondrial 8993 T>G mutation in

ACCEPTED MANUSCRIPT a newborn: implications for prognosis and family planning decisions. Eur. J. Pediatr. 174, 267–270. https://doi.org/10.1007/s00431-014-2370-y Pringsheim, T., Jette, N., Frolkis, A., Steeves, T.D.L., 2014. The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Mov. Disord. Off. J. Mov. Disord. Soc. 29, 1583–1590. https://doi.org/10.1002/mds.25945

PT

Quijano, C., Trujillo, M., Castro, L., Trostchansky, A., 2016. Interplay between oxidant species and energy metabolism. Redox Biol. 8, 28–42. https://doi.org/10.1016/j.redox.2015.11.010 Rahman, S., 2015. Emerging aspects of treatment in mitochondrial disorders. J. Inherit. Metab. Dis. 38, 641–653. https://doi.org/10.1007/s10545-015-9855-3

SC

RI

Rahman, S., Thorburn, D., 1993. Nuclear Gene-Encoded Leigh Syndrome Overview, in: Pagon, R.A., Adam, M.P., Ardinger, H.H., Wallace, S.E., Amemiya, A., Bean, L.J., Bird, T.D., Fong, C.-T., Mefford, H.C., Smith, R.J., Stephens, K. (Eds.), GeneReviews(®). University of Washington, Seattle, Seattle (WA).

NU

Ramalingam, S., Kandavelou, K., Rajenderan, R., Chandrasegaran, S., 2011. Creating Designed Zinc Finger Nucleases with Minimal Cytotoxicity. J. Mol. Biol. 405, 630– 641. https://doi.org/10.1016/j.jmb.2010.10.043

D

MA

Rat Genome Database, 2017. Leigh Syndrome due to Mitochondrial Complex V Deficiency [WWW Document]. URL http://rgd.mcw.edu/rgdweb/ontology/view.html?acc_id=RDO:0013744&offset=153 (accessed 1.31.17).

PT E

Russell, O., Turnbull, D., 2014. Mitochondrial DNA disease—molecular insights and potential routes to a cure. Exp. Cell Res. 325, 38–43. https://doi.org/10.1016/j.yexcr.2014.03.012 Sagan, L., 1967. On the origin of mitosing cells. J. Theor. Biol. 14, 255–274.

AC

CE

Santorelli, F.M., Mak, S.C., El-Schahawi, M., Casali, C., Shanske, S., Baram, T.Z., Madrid, R.E., DiMauro, S., 1996. Maternally inherited cardiomyopathy and hearing loss associated with a novel mutation in the mitochondrial tRNA(Lys) gene (G8363A). Am. J. Hum. Genet. 58, 933–939. Santorelli, F.M., Tanji, K., Manta, P., Casali, C., Krishna, S., Hays, A.P., Mancini, D.M., DiMauro, S., Hirano, M., 1999. Maternally inherited cardiomyopathy: an atypical presentation of the mtDNA 12S rRNA gene A1555G mutation. Am. J. Hum. Genet. 64, 295–300. https://doi.org/10.1086/302188 Sato, M., Sato, K., 2013. Maternal inheritance of mitochondrial DNA by diverse mechanisms to eliminate paternal mitochondrial DNA. Biochim. Biophys. Acta BBA - Mol. Cell Res. 1833, 1979–1984. https://doi.org/10.1016/j.bbamcr.2013.03.010 Schon, E.A., DiMauro, S., Hirano, M., 2012. Human mitochondrial DNA: roles of inherited and somatic mutations. Nat. Rev. Genet. 13, 878–890. https://doi.org/10.1038/nrg3275 Schon, E.A., Przedborski, S., 2011. Mitochondria: the next (neurode)generation. Neuron 70, 1033–1053. https://doi.org/10.1016/j.neuron.2011.06.003

ACCEPTED MANUSCRIPT Selosse, M.-A., 2011. Morts d’amour: mitochondria are bacteria that sometimes become extinct through symbiosis. Trends Microbiol. 19, 255–256. https://doi.org/10.1016/j.tim.2011.04.001 Shanske, S., Coku, J., Lu, J., Ganesh, J., Krishna, S., Tanji, K., Bonilla, E., Naini, A.B., Hirano, M., DiMauro, S., 2008. The G13513A mutation in the ND5 gene of mitochondrial DNA as a common cause of MELAS or Leigh syndrome: evidence from 12 cases. Arch. Neurol. 65, 368–372. https://doi.org/10.1001/archneurol.2007.67

PT

Shanske, S., Moraes, C.T., Lombes, A., Miranda, A.F., Bonilla, E., Lewis, P., Whelan, M.A., Ellsworth, C.A., DiMauro, S., 1990. Widespread tissue distribution of mitochondrial DNA deletions in Kearns-Sayre syndrome. Neurology 40, 24–28.

SC

RI

Shoffner, J.M., Lott, M.T., Lezza, A.M., Seibel, P., Ballinger, S.W., Wallace, D.C., 1990. Myoclonic epilepsy and ragged-red fiber disease (MERRF) is associated with a mitochondrial DNA tRNA(Lys) mutation. Cell 61, 931–937.

NU

Shokolenko, I., Venediktova, N., Bochkareva, A., Wilson, G.L., Alexeyev, M.F., 2009. Oxidative stress induces degradation of mitochondrial DNA. Nucleic Acids Res. 37, 2539–2548. https://doi.org/10.1093/nar/gkp100

MA

Shokolenko, I.N., Alexeyev, M.F., LeDoux, S.P., Wilson, G.L., 2010. The approaches for manipulating mitochondrial proteome. Environ. Mol. Mutagen. 51, 451–461. https://doi.org/10.1002/em.20570

D

Shtilbans, A., El-Schahawi, M., Malkin, E., Shanske, S., Musumeci, O., DiMauro, S., 1999. A novel mutation in the mitochondrial DNA transfer ribonucleic acidAsp gene in a child with myoclonic epilepsy and psychomotor regression. J. Child Neurol. 14, 610–613.

PT E

Silvestri, G., Santorelli, F.M., Shanske, S., Whitley, C.B., Schimmenti, L.A., Smith, S.A., DiMauro, S., 1994. A new mtDNA mutation in the tRNA(Leu(UUR)) gene associated with maternally inherited cardiomyopathy. Hum. Mutat. 3, 37–43. https://doi.org/10.1002/humu.1380030107

AC

CE

Singh, K.K., 2015. Mipigenetics and Mipigenomics: Integrating mitochondria-induced mayhem contributing to mystondria. Mitochondrion, Supplement: Mitochondrial Medicine: 2015 24, Supplement, S6. https://doi.org/10.1016/j.mito.2015.07.024 Singh, K.K., 2013. Mitochondrial DNA Mutations in Aging, Disease and Cancer. Springer Science & Business Media. Sofou, K., De Coo, I.F.M., Isohanni, P., Ostergaard, E., Naess, K., De Meirleir, L., Tzoulis, C., Uusimaa, J., De Angst, I.B., Lönnqvist, T., Pihko, H., Mankinen, K., Bindoff, L.A., Tulinius, M., Darin, N., 2014. A multicenter study on Leigh syndrome: disease course and predictors of survival. Orphanet J. Rare Dis. 9, 52. https://doi.org/10.1186/17501172-9-52 Spruijt, L., Smeets, H.J., Hendrickx, A., Bettink-Remeijer, M.W., Maat-Kievit, A., Schoonderwoerd, K.C., Sluiter, W., de Coo, I.F., Hintzen, R.Q., 2007. A MELASassociated ND1 mutation causing leber hereditary optic neuropathy and spastic dystonia. Arch. Neurol. 64, 890–893. https://doi.org/10.1001/archneur.64.6.890

ACCEPTED MANUSCRIPT Srivastava, S., Moraes, C.T., 2001. Manipulating mitochondrial DNA heteroplasmy by a mitochondrially targeted restriction endonuclease. Hum. Mol. Genet. 10, 3093–3099. https://doi.org/10.1093/hmg/10.26.3093 Steele, H.E., Horvath, R., Lyon, J.J., Chinnery, P.F., 2017. Monitoring clinical progression with mitochondrial disease biomarkers. Brain J. Neurol. 140, 2530–2540. https://doi.org/10.1093/brain/awx168

RI

PT

Subramanian, P., 2018. Clinical Trial (NCT03293524): Efficacy and Safety of Bilateral Intravitreal Injection of GS010: A Randomized, Double-Masked, Placebo-Controlled Trial in Subjects Affected With G11778A ND4 Leber Hereditary Optic Neuropathy for Up to One Year [WWW Document]. ClinicalTrials.gov. URL https://clinicaltrials.gov/ct2/show/NCT03293524 (accessed 5.8.18).

NU

SC

Sundaram, C., Meena, A.K., Uppin, M.S., Govindaraj, P., Vanniarajan, A., Thangaraj, K., Kaul, S., Kekunnaya, R., Murthy, J.M.K., 2011. Contribution of muscle biopsy and genetics to the diagnosis of chronic progressive external opthalmoplegia of mitochondrial origin. J. Clin. Neurosci. Off. J. Neurosurg. Soc. Australas. 18, 535–538. https://doi.org/10.1016/j.jocn.2010.06.014 Suzuki, Y.J., Carini, M., Butterfield, D.A., 2010. Protein Carbonylation. Antioxid. Redox Signal. 12, 323–325. https://doi.org/10.1089/ars.2009.2887

MA

Taanman, J.W., 1999. The mitochondrial genome: structure, transcription, translation and replication. Biochim. Biophys. Acta 1410, 103–123.

PT E

D

Tachibana, M., Amato, P., Sparman, M., Woodward, J., Sanchis, D.M., Ma, H., Gutierrez, N.M., Tippner-Hedges, R., Kang, E., Lee, H.-S., Ramsey, C., Masterson, K., Battaglia, D., Lee, D., Wu, D., Jensen, J., Patton, P., Gokhale, S., Stouffer, R., Mitalipov, S., 2013. Towards germline gene therapy of inherited mitochondrial diseases. Nature 493, 627– 631. https://doi.org/10.1038/nature11647

AC

CE

Tanaka, M., Borgeld, H.-J., Zhang, J., Muramatsu, S., Gong, J.-S., Yoneda, M., Maruyama, W., Naoi, M., Ibi, T., Sahashi, K., Shamoto, M., Fuku, N., Kurata, M., Yamada, Y., Nishizawa, K., Akao, Y., Ohishi, N., Miyabayashi, S., Umemoto, H., Muramatsu, T., Furukawa, K., Kikuchi, A., Nakano, I., Ozawa, K., Yagi, K., 2002. Gene therapy for mitochondrial disease by delivering restriction endonuclease SmaI into mitochondria. J. Biomed. Sci. 9, 534–541. https://doi.org/64726 Taylor, R.W., Chinnery, P.F., Turnbull, D.M., Lightowlers, R.N., 1997. Selective inhibition of mutant human mitochondrial DNA replication in vitro by peptide nucleic acids. Nat. Genet. 15, 212–215. https://doi.org/10.1038/ng0297-212 Thajeb, P., Dai, D., Chiang, M.-F., Shyu, W.-C., 2006. Genotype-Phenotype Correlation of Maternally Inherited Disorders due to Mutations in Mitochondrial DNA. Taiwan. J. Obstet. Gynecol. 45, 201–207. https://doi.org/10.1016/S1028-4559(09)60225-4 Thambisetty, M., Newman, N.J., 2004. Diagnosis and management of MELAS. Expert Rev. Mol. Diagn. 4, 631–644. https://doi.org/10.1586/14737159.4.5.631 Thomas, R.R., Khan, S.M., Portell, F.R., Smigrodzki, R.M., Bennett Jr., J.P., 2011. Recombinant human mitochondrial transcription factor A stimulates mitochondrial biogenesis and ATP synthesis, improves motor function after MPTP, reduces oxidative

ACCEPTED MANUSCRIPT stress and increases survival after endotoxin. Mitochondrion 11, 108–118. https://doi.org/10.1016/j.mito.2010.08.004 Thorburn, D.R., Rahman, S., 1993. Mitochondrial DNA-Associated Leigh Syndrome and NARP, in: Pagon, R.A., Adam, M.P., Ardinger, H.H., Wallace, S.E., Amemiya, A., Bean, L.J., Bird, T.D., Ledbetter, N., Mefford, H.C., Smith, R.J., Stephens, K. (Eds.), GeneReviews(®). University of Washington, Seattle, Seattle (WA).

PT

Thyagarajan, D., Shanske, S., Vazquez-Memije, M., De Vivo, D., DiMauro, S., 1995. A novel mitochondrial ATPase 6 point mutation in familial bilateral striatal necrosis. Ann. Neurol. 38, 468–472. https://doi.org/10.1002/ana.410380321

RI

Timmis, J.N., Ayliffe, M.A., Huang, C.Y., Martin, W., 2004. Endosymbiotic gene transfer: organelle genomes forge eukaryotic chromosomes. Nat. Rev. Genet. 5, 123–135. https://doi.org/10.1038/nrg1271

NU

SC

Tretter, L., Adam-Vizi, V., 2004. Generation of Reactive Oxygen Species in the Reaction Catalyzed by α-Ketoglutarate Dehydrogenase. J. Neurosci. 24, 7771–7778. https://doi.org/10.1523/JNEUROSCI.1842-04.2004

MA

Tuppen, H.A.L., Blakely, E.L., Turnbull, D.M., Taylor, R.W., 2010. Mitochondrial DNA mutations and human disease. Biochim. Biophys. Acta BBA - Bioenerg. 1797, 113– 128. https://doi.org/10.1016/j.bbabio.2009.09.005

D

US National Library of Medicine, Genetics Home Reference, 2017. MERRF [WWW Document]. Genet. Home Ref. URL https://ghr.nlm.nih.gov/condition/myoclonicepilepsy-with-ragged-red-fibers (accessed 1.31.17).

PT E

Vaidya, B., Mishra, N., Dube, D., Tiwari, S., Vyas, S.P., 2009. Targeted nucleic acid delivery to mitochondria. Curr. Gene Ther. 9, 475–486.

CE

Van Goethem, G., Martin, J.-J., Van Broeckhoven, C., 2003. Progressive external ophthalmoplegia characterized by multiple deletions of mitochondrial DNA: unraveling the pathogenesis of human mitochondrial DNA instability and the initiation of a genetic classification. Neuromolecular Med. 3, 129–146. https://doi.org/10.1385/NMM:3:3:129

AC

Van Trappen, P.O., Cullup, T., Troke, R., Swann, D., Shepherd, J.H., Jacobs, I.J., Gayther, S.A., Mein, C.A., 2007. Somatic mitochondrial DNA mutations in primary and metastatic ovarian cancer. Gynecol. Oncol. 104, 129–133. https://doi.org/10.1016/j.ygyno.2006.07.010 Verdile, G., Keane, K.N., Cruzat, V.F., Medic, S., Sabale, M., Rowles, J., Wijesekara, N., Martins, R.N., Fraser, P.E., Newsholme, P., 2015. Inflammation and Oxidative Stress: The Molecular Connectivity between Insulin Resistance, Obesity, and Alzheimer’s Disease. Mediators Inflamm. 2015. https://doi.org/10.1155/2015/105828 Vignal, C., 2018. Clinical Trial (NCT02064569): An Open Label Dose Escalation Clinical Trial to Evaluate the Safety and the Tolerability of GS010 (rAAV2/2-ND4) in Patients With Leber Hereditary Optic Neuropathy Due to Mutations in the Mitochondrial NADH Dehydrogenase 4 Gene [WWW Document]. ClinicalTrials.gov. URL https://clinicaltrials.gov/ct2/show/NCT02064569 (accessed 5.8.18).

ACCEPTED MANUSCRIPT Viscomi, C., Bottani, E., Zeviani, M., 2015. Emerging concepts in the therapy of mitochondrial disease. Biochim Biophys Acta 1847, 544–557. https://doi.org/10.1016/j.bbabio.2015.03.001 Wallace, D.C., Chalkia, D., 2013. Mitochondrial DNA Genetics and the Heteroplasmy Conundrum in Evolution and Disease. Cold Spring Harb. Perspect. Biol. 5. https://doi.org/10.1101/cshperspect.a021220

PT

Wan, X., Pei, H., Zhao, M., Yang, S., Hu, W., He, H., Ma, S., Zhang, G., Dong, X., Chen, C., Wang, D., Li, B., 2016. Efficacy and Safety of rAAV2-ND4 Treatment for Leber’s Hereditary Optic Neuropathy. Sci. Rep. 6. https://doi.org/10.1038/srep21587

RI

Wang, Y.-G., Huang, P.-P., Zhang, R., Ma, B.-Y., Zhou, X.-M., Sun, Y.-F., 2016. Targeting adeno-associated virus and adenoviral gene therapy for hepatocellular carcinoma. World J. Gastroenterol. 22, 326–337. https://doi.org/10.3748/wjg.v22.i1.326

NU

SC

Weber-Lotfi, F., Koulintchenko, M.V., Ibrahim, N., Hammann, P., Mileshina, D.V., Konstantinov, Y.M., Dietrich, A., 2015. Nucleic acid import into mitochondria: New insights into the translocation pathways. Biochim. Biophys. Acta BBA - Mol. Cell Res. 1853, 3165–3181. https://doi.org/10.1016/j.bbamcr.2015.09.011

MA

Weissig, V., 2015. DQAsomes as the prototype of mitochondria-targeted pharmaceutical nanocarriers: preparation, characterization, and use. Methods Mol. Biol. Clifton NJ 1265, 1–11. https://doi.org/10.1007/978-1-4939-2288-8_1

D

Weissig, V., D’Souza, G.G.M., 2004. Cationic Mitochondriotropic Vesicles for DNA Delivery to Mitochondria. Mol. Ther. 9, S259–S259. https://doi.org/10.1016/j.ymthe.2004.06.574

PT E

Weissig, V., Lizano, C., Torchilin, V.P., 2000. Selective DNA release from DQAsome/DNA complexes at mitochondria-like membranes. Drug Deliv. 7, 1–5. https://doi.org/10.1080/107175400266722

CE

Weissig, V., Torchilin, V.P., 2001. Towards mitochondrial gene therapy: DQAsomes as a strategy. J. Drug Target. 9, 1–13.

AC

Wernick, R.I., Estes, S., Howe, D.K., Denver, D.R., 2016. Paths of Heritable Mitochondrial DNA Mutation and Heteroplasmy in Reference and gas-1 Strains of Caenorhabditis elegans. Front. Genet. 7. https://doi.org/10.3389/fgene.2016.00051 Westarp, M.E., Holzgraefe, M., Gallenkamp, U., Thomas, R., Bechinger, D., 1992. Diagnosis and treatment in a case of juvenile subacute necrotizing encephalopathy Leigh without cytochrome c oxidase deficiency. Eur. Neurol. 32, 206–211. Winklhofer, K.F., Haass, C., 2010. Mitochondrial dysfunction in Parkinson’s disease. Biochim. Biophys. Acta BBA - Mol. Basis Dis., Mitochondrial Dysfunction 1802, 29–44. https://doi.org/10.1016/j.bbadis.2009.08.013 Wong, L.-J.C., 2007. Diagnostic challenges of mitochondrial DNA disorders. Mitochondrion 7, 45–52. https://doi.org/10.1016/j.mito.2006.11.025 Wyman, T.B., Nicol, F., Zelphati, O., Scaria, P.V., Plank, C., Szoka, F.C., 1997. Design, synthesis, and characterization of a cationic peptide that binds to nucleic acids and

ACCEPTED MANUSCRIPT permeabilizes bilayers. Biochemistry https://doi.org/10.1021/bi9618474

(Mosc.)

36,

3008–3017.

Yamada, Y., 2014. [Development of the MITO-porter, a nano device for mitochondrial drug delivery via membrane fusion]. Yakugaku Zasshi 134, 1143–1155.

PT

Yamada, Y., Akita, H., Kamiya, H., Kogure, K., Yamamoto, T., Shinohara, Y., Yamashita, K., Kobayashi, H., Kikuchi, H., Harashima, H., 2008. MITO-Porter: A liposome-based carrier system for delivery of macromolecules into mitochondria via membrane fusion. Biochim. Biophys. Acta BBA Biomembr. 1778, 423–432. https://doi.org/10.1016/j.bbamem.2007.11.002

SC

RI

Yamada, Y., Akita, H., Kogure, K., Kamiya, H., Harashima, H., 2007. Mitochondrial drug delivery and mitochondrial disease therapy--an approach to liposome-based delivery targeted to mitochondria. Mitochondrion 7, 63–71. https://doi.org/10.1016/j.mito.2006.12.003

NU

Yamada, Y., Furukawa, R., Yasuzaki, Y., Harashima, H., 2011. Dual Function MITO-Porter, a Nano Carrier Integrating Both Efficient Cytoplasmic Delivery and Mitochondrial Macromolecule Delivery. Mol. Ther. 19, 1449–1456. https://doi.org/10.1038/mt.2011.99

MA

Yamada, Y., Harashima, H., 2015. Targeting the Mitochondrial Genome via a Dual Function MITO-Porter: Evaluation of mtDNA Levels and Mitochondrial Function, in: Mitochondrial Medicine, Methods in Molecular Biology. Humana Press, New York, NY, pp. 123–133. https://doi.org/10.1007/978-1-4939-2288-8_10

PT E

D

Yamada, Y., Harashima, H., 2012. Delivery of bioactive molecules to the mitochondrial genome using a membrane-fusing, liposome-based carrier, DF-MITO-Porter. Biomaterials 33, 1589–1595. https://doi.org/10.1016/j.biomaterials.2011.10.082

CE

Yamada, Y., Ishikawa, T., Harashima, H., 2017. Validation of the use of an artificial mitochondrial reporter DNA vector containing a Cytomegalovirus promoter for mitochondrial transgene expression. Biomaterials 136, 56–66. https://doi.org/10.1016/j.biomaterials.2017.05.016

AC

Yang, A., Zheng, J., Lv, J., Guan, M., 2011. [Modifier factors influencing the phenotypic manifestation of the deafness associated mitochondrial DNA mutations]. Zhonghua Yi Xue Yi Chuan Xue Za Zhi Zhonghua Yixue Yichuanxue Zazhi Chin. J. Med. Genet. 28, 165–171. https://doi.org/10.3760/cma.j.issn.1003-9406.2011.02.010 Yang, S., Ma, S., Wan, X., He, H., Pei, H., Zhao, M., Chen, C., Wang, D., Dong, X., Yuan, J., Li, B., 2016. Long-term outcomes of gene therapy for the treatment of Leber’s hereditary optic neuropathy. EBioMedicine 10, 258–268. https://doi.org/10.1016/j.ebiom.2016.07.002 Yao, X., Carlson, D., Sun, Y., Ma, L., Wolf, S.E., Minei, J.P., Zang, Q.S., 2015. Mitochondrial ROS Induces Cardiac Inflammation via a Pathway through mtDNA Damage in a Pneumonia-Related Sepsis Model. PLoS ONE 10. https://doi.org/10.1371/journal.pone.0139416 Yasuzaki, Y., Yamada, Y., Ishikawa, T., Harashima, H., 2015. Validation of Mitochondrial Gene Delivery in Liver and Skeletal Muscle via Hydrodynamic Injection Using an

ACCEPTED MANUSCRIPT Artificial Mitochondrial Reporter DNA Vector. Mol. Pharm. 12, 4311–4320. https://doi.org/10.1021/acs.molpharmaceut.5b00511 Yasuzaki, Y., Yamada, Y., Kanefuji, T., Harashima, H., 2013. Localization of exogenous DNA to mitochondria in skeletal muscle following hydrodynamic limb vein injection. J. Control. Release Off. J. Control. Release Soc. 172, 805–811. https://doi.org/10.1016/j.jconrel.2013.09.029

PT

Ye, K., Lu, J., Ma, F., Keinan, A., Gu, Z., 2014. Extensive pathogenicity of mitochondrial heteroplasmy in healthy human individuals. Proc. Natl. Acad. Sci. U. S. A. 111, 10654– 10659. https://doi.org/10.1073/pnas.1403521111

SC

RI

Yu, H., Koilkonda, R.D., Chou, T.-H., Porciatti, V., Mehta, A., Hentall, I.D., Chiodo, V.A., Boye, S.L., Hauswirth, W.W., Lewin, A.S., Guy, J., 2015. Consequences of zygote injection and germline transfer of mutant human mitochondrial DNA in mice. Proc. Natl. Acad. Sci. U. S. A. 112, E5689-5698. https://doi.org/10.1073/pnas.1506129112

NU

Yu, H., Koilkonda, R.D., Chou, T.-H., Porciatti, V., Ozdemir, S.S., Chiodo, V., Boye, S.L., Boye, S.E., Hauswirth, W.W., Lewin, A.S., Guy, J., 2012. Gene delivery to mitochondria by targeting modified adenoassociated virus suppresses Leber’s hereditary optic neuropathy in a mouse model. Proc. Natl. Acad. Sci. U. S. A. 109, E1238–E1247. https://doi.org/10.1073/pnas.1119577109

D

MA

Yu-Wai-Man, P., 2018a. Clinical Trial (NCT02652780): Randomized, Double-Masked, ShamControlled Clinical Trial to Evaluate the Efficacy of a Single Intravitreal Injection of GS010 in Subjects Affected for More Than 6 Months and To 12 Months by LHON Due to the G11778A Mutation in the ND4 Gene [WWW Document]. ClinicalTrials.gov. URL https://clinicaltrials.gov/ct2/show/NCT02652780 (accessed 5.8.18).

PT E

Yu-Wai-Man, P., 2018b. Clinical Trial (NCT03406104): Long-term Follow-up of ND4 LHON Subjects Treated With GS010 Ocular Gene Therapy in the RESCUE or REVERSE Phase III Clinical Trials [WWW Document]. ClinicalTrials.gov. URL https://clinicaltrials.gov/ct2/show/NCT03406104 (accessed 5.8.18).

AC

CE

Yu-Wai-Man, P., 2016. Genetic manipulation for inherited neurodegenerative diseases: myth or reality? Br. J. Ophthalmol. 100, 1322–1331. https://doi.org/10.1136/bjophthalmol2015-308329 Yu-Wai-Man, P., Chinnery, P.F., 1993. Leber Hereditary Optic Neuropathy, in: Pagon, R.A., Adam, M.P., Ardinger, H.H., Wallace, S.E., Amemiya, A., Bean, L.J., Bird, T.D., Ledbetter, N., Mefford, H.C., Smith, R.J., Stephens, K. (Eds.), GeneReviews(®). University of Washington, Seattle, Seattle (WA). Zhao, L., Wen, Q., Yang, G., Huang, Z., Shen, T., Li, H., Ren, D., 2016. Apoptosis induction of dehydrobruceine B on two kinds of human lung cancer cell lines through mitochondrial-dependent pathway. Phytomedicine 23, 114–122. https://doi.org/10.1016/j.phymed.2015.12.019 Zheng, J., Ji, Y., Guan, M.-X., 2012. Mitochondrial tRNA mutations associated with deafness. Mitochondrion 12, 406–413. https://doi.org/10.1016/j.mito.2012.04.001 Zhu, X., Peng, X., Guan, M.-X., Yan, Q., 2009. Pathogenic mutations of nuclear genes associated with mitochondrial disorders. Acta Biochim. Biophys. Sin. 41, 179–187.

ACCEPTED MANUSCRIPT Zsurka, G., Hampel, K.G., Nelson, I., Jardel, C., Mirandola, S.R., Sassen, R., Kornblum, C., Marcorelles, P., Lavoué, S., Lombès, A., Kunz, W.S., 2010. Severe epilepsy as the major symptom of new mutations in the mitochondrial tRNA(Phe) gene. Neurology 74, 507–512. https://doi.org/10.1212/WNL.0b013e3181cef7ab

AC

CE

PT E

D

MA

NU

SC

RI

PT

Zupančič, Š., Kocbek, P., Zariwala, M.G., Renshaw, D., Gul, M.O., Elsaid, Z., Taylor, K.M.G., Somavarapu, S., 2014. Design and development of novel mitochondrial targeted nanocarriers, DQAsomes for curcumin inhalation. Mol. Pharm. 11, 2334–2345. https://doi.org/10.1021/mp500003q

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

AC

CE

PT E

D

Fig. 1. mtDNA loci map for mitochondrial pathologies. Respective mutations are denoted in parentheses. CPEO: chronic progressive external ophthalmoplegia; FBSN: familial bilateral striatal necrosis; KSS: Kearns-Sayre syndrome; LHON: Leber’s hereditary optic neuropathy; LS: Leigh syndrome; MEPR: myoclonic epilepsy and psychomotor regression; MELAS: mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes; MERRF: myoclonic epilepsy with ragged-red fibers; MHCM: maternally inherited hypertrophic cardiomyopathy; MICM: maternally inherited cardiomyopathy; MID: maternally inherited deafness; MIDD: maternally inherited diabetes and deafness, NARP: neurogenic muscle weakness, ataxia, and retinitis pigmentosa.

PT E

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

AC

CE

Fig. 2. Allotopic expression-mediated gene therapy. (A) Stable transfection of corrective gene in the nDNA: A recombinant DNA construct comprising of a promoter, a mitochondriatargeting sequence (MTS), the gene of interest (GOI) (encoding the corrective protein), a tag and a 3’-untranslated region (3’-UTR) is stably transfected into the nuclear DNA using a suitable vector. Post transcription, the mRNA is exported into the cytoplasm where it gets translated to yield the protein of interest (POI) fused with MTS, which directs the POI specifically into the mitochondria, thereby subsequently restoring mitochondrial functions. (B) Chemically modified mRNA (modRNA)-mediated therapy: A modRNA (containing the open reading frames of an MTS, a GOI, a tag and a 3’-UTR) is inserted into the cytoplasm. The modRNA, being an mRNA, is translated by the ribosome machinery of the cell to form a fused protein (MTS-POI). The translated MTS-conjugated POI enters specifically into the mitochondria, thereby functionally restoring them.

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

AC

CE

PT E

Fig. 3. Gene therapy by selective inhibition of mutant mtDNA. (A) Restriction endonucleases (REs): A construct, which contains a suitable promoter, the gene for a mitochondria-targeting sequence (MTS) and a specific RE-encoding gene, is transiently transfected into the nucleus. On translation, the MTS-conjugated RE localizes to mitochondria and cuts (obliterates) the mutant (MT) mtDNA. (B) mtDNA-binding zinc-finger nucleases (mtZFNs): The construct contains a promoter, an MTS, a tag, a nuclear export signal (NES) and genes for a specific zinc-finger protein (ZFP) and a RE. It is transiently transfected into the nucleus. The translated MTS-ZFP-RE fusion protein localizes to the mitochondria, where the ZFP recognizes specific sequence in the MT mtDNA, while the fused RE subsequently cuts the mtDNA at the recognized site. (C) Mitochondrially targeted trancription activator-like effector (TALE) nucleases (mitoTALENs): Two monomeric constructs, each containing a promoter, an MTS, a tag, and genes encoding for TALE (left or right) and FokI nuclease, are transiently transfected into the nucleus. On translation, the fused proteins are delivered to the mitochondria by the MTS. Here, TALE (left) and TALE (right) from the corresponding fused proteins bind to specific sequences of the MT mtDNA, thereby flanking a specific site. The two FokI nuclease proteins dimerize and cut the flanked site, thereby destroying the MT mtDNA.

PT E

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

AC

CE

Fig. 4. Gene therapy by delivery of wild type (WT) mtDNA. (A) Mitochondrial transcription factor A (TFAM): This carrier-based delivery system comprises of a mitchondrial transduction domain (MTD), which is a combination of a mitochondria-targeting sequence (MTS) and a protein transduction domain (PTD), conjugated with TFAM (MTDTFAM). While TFAM carries the WT mtDNA, PTD facilitates transport of the MTD-TFAM conjugate across the cell membrane. This is then followed by MTS-mediated delivery of the WT gene into the mitochondria harboring the mutant (MT) mtDNA. (B) Dual function MITOporters (DF-MITO-porters): These liposome-based nanocarriers deliver WT mtDNA to the mitochondria with the MT mtDNA. The DF-MITO-porters are two membrane-fusogenic systems comprising of outer and inner envelopes capable of fusing specifically with the endosomal and mitochondrial membranes respectively. DF-MITO-porters containing the WT mtDNA are endocytosed within the cell and are captured in the endosome. The outer envelope fuses with the endosomal membrane, subsequently triggering the escape of the WT mtDNA still packed within the inner envelope. Selective fusion of the inner envelope with the mitochondrial membrane releases the WT mtDNA inside the mitochondria. (C) DQAsomes: Liposome-like nanovesicles made of dequalinium chloride (DQA) carry WT mtDNA tagged with MTS (DQAsome-DNA-MTS complex) inside the cell. The MTS delivers the DQAsome to the mitochondria. On contact with the mitochondrial membrane, the DQA-coated membrane of the nanovesicle disintegrates and releases the WT mtDNA on to the mitochondria from where it is taken into the matrix via the protein import machinery.

ACCEPTED MANUSCRIPT Table 1: Major mitochondrial diseases arising from mutations in the mtDNA

# Mitochondrial disease

5 6

PT

RI

4

tRNAAla ATP6, ATP8 ATP6 COII, COIII

ND1, ND4, ND6 tRNALeu, tRNAVal rRNA12s, tRNAThr, Auditory defects tRNALeu, tRNASer tRNALeu, Diabetes and hearing loss tRNALys, tRNAGlu tRNALeu, Neuromuscular disorder of the cardiac tRNAIle, tissue tRNALys, ND5

SC

3

Chronic progressive external Visual defects due to neurological ophthalmoplegia (CPEO) degeneration Metabolic disorder due defective ETC Complex V deficiency complex V Familial bilateral striatal necrosis Striatonigral degeneration in brain (FBSN) Neuromuscular disorder causing Kearns-Sayre syndrome (KSS) retinopathy, cardiomyopathy, deafness, ataxia and other defects Leber’s hereditary optic Visual defects due to mutant retinal neuropathy (LHON) ganglion cells (RGCs) Maternally inherited Neuromuscular disorder of the cardiac cardiomyopathy (MICM) tissue

7

Maternally inherited deafness (MID)

8

Maternally inherited diabetes and deafness (MIDD)

D

Maternally inherited 9 hypertrophic cardiomyopathy (MHCM) Mitochondrial 1 encephalomyopathy, lactic 0 acidosis, and stroke-like episodes (MELAS) 1 Myoclonic epilepsy and 1 psychomotor regression (MEPR) 1 Myoclonic epilepsy with ragged2 red fibers (MERRF) Neurogenic muscle weakness, 1 ataxia, and retinitis pigmentosa 3 (NARP)

NU

2

Characteristics

MA

1

Mutant mtDNA gene(s)

Neuromuscular disorder

tRNAAsp

Neuromuscular disorder

tRNALys, tRNAPhe

AC

CE

PT E

Neurodegenerative disorder

tRNALeu, tRNAVal, ND5, COIII

Neurodegenerative disorder arising from ATP6 defective F1FO-ATPase activity

COI, COII, Neuronal defects in the substantia nigra of cytochrome b, brain ND5, D-Loop Subacute necrotizing Neurodegenerative disorder due to defects tRNAIle, 1 encephalomyelopathy / Leigh in ETC complex I, complex IV and tRNATrp, ATP6, 5 syndrome (LS) pyruvate dehydrogenase complex ND5, COIII 1 Parkinson's disease (PD) 4

ACCEPTED MANUSCRIPT Highlights 

Variegated mutations in the nuclear and/or mitochondrial DNA (mtDNA) give rise to a plethora of mitochondrial diseases.



Limited gene therapy options exist for mtDNA mutation-associated disorders in comparison to conditions arising from mutations in the nuclear DNA. Contemporary gene therapy strategies for correcting mtDNA mutations include

PT



allotopic expression of corrective proteins, site-specific ablation of mtDNA and carrier-

SC

There is a need for further exploration into novel strategies for improved efficacy and

CE

PT E

D

MA

NU

safety in gene therapy against mtDNA mutation-associated ailments.

AC



RI

based delivery of wild-type mtDNA.