Next Article in Journal
A Self-Standing Binder-Free Biomimetic Cathode Based on LMO/CNT Enhanced with Graphene and PANI for Aqueous Rechargeable Batteries
Next Article in Special Issue
Efficient TGF-β1 Delivery to Articular Chondrocytes In Vitro Using Agro-Based Liposomes
Previous Article in Journal
Simultaneous Improvement of Grain Yield and Quality through Manipulating Two Type C G Protein Gamma Subunits in Rice
Previous Article in Special Issue
Development of a Gene-Activated Scaffold Incorporating Multifunctional Cell-Penetrating Peptides for pSDF-1α Delivery for Enhanced Angiogenesis in Tissue Engineering Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mitochondrial Genome Editing to Treat Human Osteoarthritis—A Narrative Review

Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Saar, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(3), 1467; https://doi.org/10.3390/ijms23031467
Submission received: 13 January 2022 / Revised: 24 January 2022 / Accepted: 26 January 2022 / Published: 27 January 2022

Abstract

:
Osteoarthritis (OA) is a severe, common chronic orthopaedic disorder characterised by a degradation of the articular cartilage with an incidence that increases over years. Despite the availability of various clinical options, none can stop the irreversible progression of the disease to definitely cure OA. Various mutations have been evidenced in the mitochondrial DNA (mtDNA) of cartilage cells (chondrocytes) in OA, leading to a dysfunction of the mitochondrial oxidative phosphorylation processes that significantly contributes to OA cartilage degeneration. The mitochondrial genome, therefore, represents a central, attractive target for therapy in OA, especially using genome editing procedures. In this narrative review article, we present and discuss the current advances and breakthroughs in mitochondrial genome editing as a potential, novel treatment to overcome mtDNA-related disorders such as OA. While still in its infancy and despite a number of challenges that need to be addressed (barriers to effective and site-specific mtDNA editing and repair), such a strategy has strong value to treat human OA in the future, especially using the groundbreaking clustered regularly interspaced short palindromic repeats (CRIPSR)/CRISPR-associated 9 (CRISPR/Cas9) technology and mitochondrial transplantation approaches.

1. Introduction

Osteoarthritis (OA) is the most prevalent chronic joint disorder that affects millions of adult individuals worldwide [1,2]. The progressive, irreversible degradation of the extracellular cartilage matrix (ECM) with the persistence of chronic inflammation are all hallmarks of OA [3,4,5,6,7]. Current treatments against OA mostly target pain and inflammation or address cartilage degeneration via surgical interventions for severe and end-stage cases of the disease, but none of them is capable of fully reproducing a natural cartilage surface and joint integrity [8,9], showing the critical need for improved therapeutic approaches to cure OA.
OA has also been described as a mitochondrial disorder in light of findings showing a deterioration of the functions of these central cell organelles in OA chondrocytes during the development and progression of the disease [10]. Such mitochondrial dysfunction is associated with the presence of mutations in the peculiar DNA of the mitochondria (mtDNA) [10,11] that codes for proteases supporting mitochondrial oxidative phosphorylation processes [12], leading to increased oxidative stress and contributing to alterations in the chondrocyte phenotype and, ultimately, to cartilage degeneration [13,14].
Exploring novel ways to address the pathology of OA based on the perception that OA is also a mitochondrial disease may help define improved treatments to cure this severe disorder. The emergence of genome editing technologies that may be applied to target the mitochondrial genome provides hope to precisely modulate mtDNA genes involved in the development of OA as a means to reliably manage this disease in patients. Such innovative mitochondrial treatment options for OA are being discussed in this narrative article with the goal of feasible and safe translational applications in the near future.

2. Osteoarthritis

Osteoarthritis (OA) is the most common chronic disease of large joints, such as the knee, hip and hand joints, that mostly occurs in elderly individuals, with an incidence of greater than 60% in individuals over 65 years of age and that may further rise within the next 30 years [1,2]. OA does not only shatter joint functions and physical abilities but also brings long-term, considerable pain in patients [8]. The risk factors for OA include primary (gender, genetic inheritance, obesity, hormonal changes) and secondary (trauma, mechanical stress, metabolic changes) components [7]. The features of OA include the degeneration of the articular cartilage, synovial inflammation, subchondral bone sclerosis and osteophyte formation [3,4,5,6,7].
The healthy articular cartilage consists of a layer of hyaline cartilage devoid of vascularisation, the lymphatic system and innervation, covering the end of bones in the joint to resist shocks and withstand mechanical loading and impact [15]. Under pathological conditions in OA, the chondrocytes undergo phenotypic changes and alterations in their homeostasis in response to an accumulation of detrimental pro-inflammatory mediators (interleukin-1 beta—IL-1β, tumour necrosis factor alpha—TNF-α, nitric oxide—NO, prostaglandin E2—PGE2, reactive oxygen species—ROS, etc.) and matrix-degrading enzymes (matrix metalloproteinases—MMPs, A disintegrin-like and metalloproteinase with thrombospondin motifs—ADAMTS, etc.), leading to the loss of ECM elements (proteoglycans, type-II collagen) [3,4,5,6,7].
The current drugs against OA mainly focus on analgesia and inflammation (celecoxib, rofecoxib), providing temporary relief symptoms, but they are not capable of preventing the progression of the disease [8,9] and may exert serious side effects in the gastrointestinal and blood systems [16]. Other treatments include the use of joint lubricants (hyaluronic acid, glucosamine sulphate, chondroitin sulphate) to reduce frictions on damaged cartilage surfaces [8], but their efficacy remains controversial [17]. For severe and end-stage cases of OA, surgical procedures may be attempted (fusion, debridement, osteotomy, joint replacement), but they lead to a considerable loss of quality of life, aside from the economic burden [9].
These findings show the crucial need to identify novel pathomechanisms of the disease that may lead to the generation of improved, more effective therapeutic options for OA.

3. Implication of the Mitochondria in Osteoarthritis

Accumulated evidence indicates that the nutrient supply of the articular cartilage is provided by the synovial fluid [18]. Independently from the oxygen blood supply, the chondrocytes have been traditionally perceived as highly glycolytic cells, obtaining energy from anaerobic respiration of glucose, a notion explaining that, until recently, the mitochondria were long neglected in the pathophysiology of OA [19,20]. However, up to 25% of articular cartilage energy needs to be met by high-energy electron transfer across the inner mitochondrial membrane during oxidative phosphorylation, a percentage pulled up under the high energy demand of the tissue [21,22]. In fact, nutrients and oxygen in the synovial fluid supply chondrocytes mainly through differential penetration in the ECM, depending on their molecular size, charge and depth of penetration [10]. As a result, this minimal oxygen partial pressure is maintained in the deep layer of the cartilage, where the glycolysis of chondrocytes and even of mesenchymal stem cells (MSCs) contributes much more to energy metabolism than to aerobic respiration. Chondrocytes and MSCs maintain an oxidative phosphorylation-independent survival after long-term evolutionary adaptation, and MSCs exhibit an environment-adaptive chondrogenic differentiation potential [23]. The surface layer of the cartilage can receive 5–7% oxygen, which is enough for the chondrocytes to carry out sufficient oxidative phosphorylation undertaken by the mitochondria [18].
Mitochondria are organelles with a double-layer membrane, converting various forms of energy into intracellular energy in the form of adenosine triphosphate (ATP) via ATPases and with the participation of high-energy electrons in the electron transport chain of the inner mitochondrial membrane through the process of oxidative phosphorylation [24,25]. ATPase binding proteins are responsible for material exchange and signal transmission with other cellular compartments, especially the nucleus, branching out the metabolism and biological behaviour of the mitochondria [26].
Mitochondria have an independent, small circular double-stranded DNA (mtDNA) encoding 13 proteins, 22 transfer RNAs (tRNAs) and 2 ribosomal RNAs (rRNAs) [27] that contribute to oxidative phosphorylation processes [28]. The complexity of the mitochondrial genome is reflected by the multiple copies of mtDNA where wild-type and variant mtDNAs coexist in heteroplasmy [29,30].
mtDNA is susceptible to exogenous stimuli (ultraviolet radiation, radioactive substances) and endogenous factors (by-products of aerobic respiration, ROS, RNS) that may lead to high-rate mutations. Mitochondria have nuclear-like DNA repair mechanisms to withstand mtDNA damage, including base excision repair (BER), direct reversal (DR), mismatch repair (MMR), cross-damage repair synthesis (TLS) and double bond break repair (DSBR) [31,32]. Nevertheless, the repair effects are fragile and balancing mitochondrial homeostasis in extreme environments remains challenging [31]. Accumulated DNA damage will, thus, cause lasting mitochondrial dysfunction as well as a pathological shift in the mitochondrial phenotype when the accumulation of damaged mitochondria reaches a threshold [33].
During the pathogenesis of OA, the mitochondria display important abnormal features in diseased chondrocytes compared with healthy cells, showing an increase in mitochondrial mass, decreased activities of the respiratory complexes II and III, reduced capacities of antioxidant enzymes (superoxide dismutase—SOD, glutathione peroxidase—GSH) and incomplete mitochondrial electron transfer resulting in excessive ROS and RNS production consuming high-energy electrons and greatly reducing the production of ATP [10,34,35]. The accumulation of ROS and RNS subsequently damages the mtDNA and mitochondrial membrane proteins, challenging the metabolism of the nucleus and of the entire chondrocytes [13].
Mutations in the mtDNA have been reported during the pathology of the disease [10,11] (Figure 1), with a minimum prevalence rate of 20 per 100,000 [36], generally accompanied by reduced mtDNA repair capacity, decreased cell viability and increased apoptosis in OA chondrocytes [37,38]. Moreover, conducting systematic genetic screening of OA cases, Chang et al. [39] evidenced age-dependent gene deletions in the 4.977-bp mtDNA related to the process of idiopathic knee joint OA.
In light of such findings, the manipulation of the mtDNA, especially using potent genome editing strategies, may be the basis for novel, highly effective options to adeptly and stably treat this incurable joint disorder.

4. Mitochondrial Genome Editing and Potential Treatments for Osteoarthritis

Translationally adapted technologies and systems have been developed to edit the mitochondrial genome to manage mtDNA mutations and mitochondrial dysfunction, including antigenomic mtDNA therapy, restriction endonucleases, zinc-finger nucleases (ZFNs), transcription activator-like effectors nucleases (TALENs) and clustered regularly interspaced short palindromic repeats/CRISPR-associated 9 (CRISPR/Cas9) (Table 1 and Figure 2) [40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84].
The potential of antigenomic mtDNA therapy [40] was reported based on the use of peptide nucleic acids (PNAs) complementary to human mtDNA templates containing pathogenic deletion breakpoints and single-base mutations using mitochondria-targeting sequences (MTS) and triphenylphosphine to specifically inhibit the replication of the mutant sequences [41,42]. This therapy was also tested to reduce mtDNA ND5 mutant genes in human cells using a 20-nucleotide sequence paired with a variant mtDNA region for mitochondrial point mutations [43].
Mitochondria-targeted restriction (bacterial) endonucleases have been employed to shift mtDNA heterogeneity and reduce the burden of mitochondrial genome mutations [44,45,46,47]. The PstI endonuclease was manipulated to shift mtDNA heterogeneity in rodent cells via accumulation of mtDNA phenotypes lacking PstI sites [48]. Xmal and SmaI were used to digest fragments in the mitochondrial T-to-G position 8993 (T8993G) mutation sequence affecting the ATP 6 gene in models of Leigh syndrome (lacticacidemia, hypotonia, neurodegeneration), normalising the production of ATP as well as the mitochondrial membrane potential [30,49,50,51]. This strategy, however, is limited by the low number of mutations that can be targeted by these endonucleases and by the fact that it may affect the entire mtDNA, potentially causing its entire degradation [44].
ZFNs, composed of a specific DNA recognition domain with tandems of Cys2-His2 zinc-finger proteins and a sequence-independent endonuclease, cut specific sites in the form of dimers [52]. ZFNs were applied to target the T8993G mutation site in the mtDNA with an MTS by inserting the marker hDNMT3a methylase sequence as a tracer of specific mtDNA modification [53] and by shifting mtDNA heterogeneity to reduce or eliminate this mutation and to restore the mitochondrial respiratory functions in human and mammalian cells [54,55,56]. ZFNs recognising the C5024T mutation of mtDNA with an MTS were also employed to eliminate mutant mtDNA in the heart of a mouse model and to alleviate mitochondrial dysfunction in cardiac tissue [57]. Yet, while ZFNs can be used for almost all mtDNA predetermined sequences [54,56,58], their presence in the targets may raise cytotoxic responses, hindering their safe application in vivo [58,59].
TALENs, naturally secreted by plant pathogens of the Xanthomonas species, exhibit a DNA sequence recognition function by their amino acid combination with repeat variable two-residue (RVD) modules assigned with amino acids at positions 12 and 13 [60]. Four different RVD modules with customisable arrays of polymorphic amino acids (Asn-Asn, Asn-Ile, His-Asp and Asn-Gly) are the most widely used to identify guanine and adenine, cytosine and thymine, respectively [60]. When two TALENs monomers bind to DNA, initiating the formation of dimers, the functional FokI unit of the system breaks the double bond [61]. TALENS have been broadly employed since their DNA-binding specificity is easier to engineer than that of ZFNs [62,63]. TALENS fused to an MTS were employed to shift mitochondrial heterogeneity by eliminating the point mutations G14459A [64], 8483_13459del4977 [65], A8344G and G13513A [66,67,68] and A3243G [69], increasing complex VI activity and the mitochondrial oxidative phosphorylation capacity. Recent work also reported the efficacy of TALENs fused with split-DddA halves from the DddA interbacterial toxin and with a uracil glycosylase inhibitor to accurately manipulate mtDNA at the level of a single base, catalysing C•G to T•A conversions [70] instead of eliminating mtDNA copy numbers that may be potentially toxic in vivo. Such an approach may help better understand mtDNA mutations in human mtDNA-related diseases and conditions such as ageing and to treat or prevent primary mtDNA mutation diseases [71,72].
The discovery of the groundbreaking CRISPR/Cas9 technology by Emmanuelle Charpentier and Jennifer Doudna [73], recipients of the 2020 Nobel Prize in Chemistry, opens new avenues of research that may be adapted to treat mitochondrial diseases. CRISPR/Cas9, a bacterial immunoprotective system that affords resistance to foreign DNA, is composed of a sequence-dependent single guide RNA (sgRNA) and a Cas9 endonuclease. The sgRNA (a CRISPR RNA-crRNA—and trans-activating crRNA-tracrRNA) specifically recognises the base sequence of a target gene and instructs Cas9 to generate DNA double-strand breaks (DSBs) at protospacer adjacent motifs (PAMs) [74], allowing to develop therapeutic strategies for cancer (lung metastasis), blood disorders (haemoglobinopathies) or viral infections (human immunodeficiency virus—HIV) [75,76,77,78,79]. CRISPR/Cas9 has been employed to target the Cox1 and Cox3 loci in mtDNA, leading to mitochondrial membrane potential disruption and cell growth inhibition [80]. Yet, the use of this system for mtDNA editing remains challenging. First, the double-layer membrane of the mitochondria is a natural barrier to the sgRNA [81]. This issue might be addressed by using a system based on the Leishmania RNA import complex (RIC) capable of translocating tRNA in the mitochondria to normalise mitochondrial dysfunction caused by the mt-tRNALys (MT-TK) mutation [82,83,84]. Next, the problems of the absence of DNA repair mechanisms in the mitochondria [85,86] and potential off-target effects need to be considered for mtDNA editing, although the heterogeneity and multi-copy characteristics of the mitochondria reserve fault-tolerant space for the biological therapy of mitochondrial gene editing.
As mitochondrial gene mutations are associated with the occurrence and progression of OA, manipulating or restoring the normal state of the mtDNA via genome editing procedures may provide potentially effective treatments to cure this disorder. While little is still known on the potential of mitochondrial genome editing in OA, strategies targeting mutated mitochondria genes contributing to OA-associated oxidation, or even genes involved in inflammation and/or ageing, may be envisaged by correcting the altered phenotypes or by transferring adapted anti-inflammatory, differentiation and/or regenerative gene sequences. Such edited mitochondria may further be used as immunopriviledged organelle transplants [87] for administration in OA targets as a means to enhance the efficacy of mtDNA editing in light of evidence showing that increasing the total amount of mtDNA improves the function of mitochondria and alleviates the symptoms of mitochondrial diseases [88]. Transplantation of edited mitochondria may be performed directly or in conjunction with clinically used, biocompatible materials to promote a sustained, controlled release of the organelles in a precise spatial and temporal manner in the OA joints [89,90].

5. Conclusions

Editing the mitochondrial genome is an emerging, attractive strategy to manage mutations in the mtDNA in OA and, subsequently, the dysfunction of the mitochondrial oxidative phosphorylation processes in OA chondrocytes that substantially contribute to the pathogenesis of this severe, highly prevalent human disorder. Mitochondrial genome editing applied to OA is still in its infancy, but such an innovative concept may be envisaged using all available editing tools, especially the CRISPR/Cas9 system. It may further be combined with the transplantation of edited (autologous or even allogeneic) mitochondria with inherent (and further improved) antioxidative abilities [91] to shift mitochondrial heterogeneity, to correct mtDNA mutations and to restore the oxidative functions to natural levels in OA cells (and patients) upon direct administration or used as cargos via controlled release (scaffold-guided) approaches. While a number of challenges remain to be addressed for the effective and safe application of genome editing procedures to target the mtDNA in OA, including the selection of an optimal editing system, the presence of barriers to effective editing in the mitochondria (double-layer membrane), the absence of mtDNA repair mechanisms and potential off-target effects, this strategy holds strong potential and value in translational applications to treat human OA in the future.

Author Contributions

Conceptualisation, G.Z. and M.C.; validation, G.Z., H.M. and M.C.; resources, M.C.; data curation, G.Z. and M.C.; writing—original draft preparation, G.Z. and M.C.; writing—review and editing, G.Z., H.M. and M.C.; visualisation, G.Z. and M.C.; supervision, M.C.; funding acquisition, H.M. and M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the World Arthrosis Foundation (grant P229-A15-Madry-EP1-madr7-allgemein-pr-I-226k-2010-14 to M.C. and H.M.) and by the National Natural Science Foundation of China (NSFC) (grant 82002355 to G.Z.). The APC was funded by the Saarland University within the funding programme Open Access Publishing.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study, in the collection, analyses, or interpretation of data, in the writing of the manuscript or in the decision to publish the results.

References

  1. Hunter, D.J.; March, L.; Chew, M. Osteoarthritis in 2020 and beyond: A Lancet Commission. Lancet 2021, 396, 1711–1712. [Google Scholar] [CrossRef]
  2. Quicke, J.G.; Conaghan, P.G.; Corp, N.; Peat, G. Osteoarthritis year in review 2021: Epidemiology and therapy. Osteoarthr. Cartil. 2022, 30, 196–206. [Google Scholar] [CrossRef] [PubMed]
  3. Liu-Bryan, R.; Terkeltaub, R. Emerging regulators of the inflammatory process in osteoarthritis. Nat. Rev. Rheumatol. 2015, 11, 35–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kapoor, M.; Martel-Pelletier, J.; Lajeunesse, D.; Pelletier, J.-P.; Fahmi, H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat. Rev. Rheumatol. 2011, 7, 33–42. [Google Scholar] [CrossRef]
  5. Loeser, R.F.; Collins, J.A.; Diekman, B.O. Ageing and the pathogenesis of osteoarthritis. Nat. Rev. Rheumatol. 2016, 12, 412–420. [Google Scholar] [CrossRef] [Green Version]
  6. Sharma, L. Osteoarthritis of the knee. N. Engl. J. Med. 2021, 284, 51–59. [Google Scholar] [CrossRef]
  7. Martel-Pelletier, J.; Barr, A.J.; Cicuttini, F.M.; Conaghan, P.G.; Cooper, C.; Goldring, M.B.; Goldring, S.R.; Jones, G.; Teichtal, A.J.; Pelletier, J.-P. Osteoarthritis. Nat. Rev. Dis. Primers 2016, 2, 16072. [Google Scholar] [CrossRef] [Green Version]
  8. Jones, I.A.; Togashi, R.; Wilson, M.L.; Heckmann, N.; Vansgness, C.T., Jr. Intra-articular treatment options for knee osteoarthritis. Nat. Rev. Rheumatol. 2019, 15, 77–90. [Google Scholar] [CrossRef]
  9. Hunter, D.J.; Bierma-Zeinstra, S. Osteoarthritis. Lancet 2019, 393, 1745–1759. [Google Scholar] [CrossRef]
  10. Blanco, F.J.; Rego, I.; Ruiz-Romero, C. The role of mitochondria in osteoarthritis. Nat. Rev. Rheumatol. 2011, 7, 161–169. [Google Scholar] [CrossRef]
  11. Blanco, F.J.; Valdes, A.M.; Rego-Perez, I. Mitochondrial variations and the pathogenesis of osteoarthritis phenotypes. Nat. Rev. Rheumatol. 2018, 14, 327–340. [Google Scholar] [CrossRef] [PubMed]
  12. Taanman, J.W. The mitochondrial genome: Structure, transcription, translation and replication. Biochim. Biophys. Acta 1999, 1410, 103–123. [Google Scholar] [CrossRef] [Green Version]
  13. Bolduc, J.A.; Collins, J.A.; Loeser, R.F. Reactive oxygen species, aging and articular cartilage homeostasis. Free Radic. Biol. Med. 2019, 132, 73–82. [Google Scholar] [CrossRef]
  14. Henrotin, Y.; Bruckner, P.; Pujol, J.-P.L. The role of reactive oxygen species in homeostasis and degradation of cartilage. Osteoarthr. Cartil. 2003, 11, 747–755. [Google Scholar] [CrossRef] [Green Version]
  15. Hunter, D.J.; Eckstein, F. From joint anatomy to clinical outcomes in osteoarthritis and cartilage repair: Summary of the fifth annual osteoarthritis imaging workshop. Osteoarthr. Cartil. 2011, 19, 1263–1269. [Google Scholar] [CrossRef] [Green Version]
  16. Chan, F.K.; Lanas, A.; Scheiman, J.; Berger, M.F.; Nguyen, H.; Goldstein, J.L. Celecoxib versus omeprazole and diclofenac in patients with osteoarthritis and rheumatoid arthritis (CONDOR): A randomised trial. Lancet 2010, 376, 173–179. [Google Scholar] [CrossRef]
  17. Bijlsma, J.W.; Berenbaum, F.; Lafeber, F.P. Osteoarthritis: An ujpdate with relevance for clinical practice. Lancet 2011, 377, 2115–2126. [Google Scholar] [CrossRef]
  18. Zhou, S.; Cui, Z.; Urban, J.P. Factors influencing the oxygen concentration gradient from the synovial surface of articular cartilage to the cartilage-bone interface: A modeling study. Arthritis Rheum. 2004, 50, 3915–3924. [Google Scholar] [CrossRef]
  19. Lee, R.B.; Urban, J.P. Functional replacement of oxygen by other oxidants in articular cartilage. Arthritis Rheum. 2002, 46, 3190–3200. [Google Scholar] [CrossRef]
  20. Gavriilidis, C.; Miwa, S.; von Zglinicki, T.; Taylor, R.W.; Young, D.A. Mitochondrial dysfunction in osteoarthritis is associated with down-regulation of superoxide dismutase 2. Arthritis Rheum. 2013, 65, 378–387. [Google Scholar] [CrossRef]
  21. Heer, C.D.; Brenner, C. Letting off electrons to cope with metabolic stress. Nat. Metab. 2020, 2, 485–486. [Google Scholar] [CrossRef] [PubMed]
  22. Lee, R.B.; Urban, J.P. Evidence for a negative Pasteur effect in articular cartilage. Biochem. J. 1997, 321, 95–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mylotte, L.A.; Duffy, A.M.; Murphy, M.; O’Brien, T.; Samali, A.; Barry, F.; Szegezdi, A. Metabolic flexibility permits mesenchymal stem cell survival in an ischemic environment. Stem Cells 2008, 26, 1325–1336. [Google Scholar] [CrossRef] [PubMed]
  24. Giacomello, M.; Pyakurel, A.; Glytsou, C.; Scorrano, L. The cell biology of mitochondrial membrane dynamics. Nat. Rev. Mol. Cell. Biol. 2020, 21, 204–224. [Google Scholar] [CrossRef] [PubMed]
  25. Chan, D.C. Mitochondrial dynamics and its involvement in disease. Annu. Rev. Pathol. 2020, 15, 235–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Szabo, I.; Zoratti, M. Mitochondrial channels: Ion fluxes and more. Physiol. Rev. 2014, 94, 519–608. [Google Scholar] [CrossRef]
  27. Andrews, R.M.; Kubacka, I.; Chinnery, P.F.; Lightowlers, R.N.; Turnbull, D.M.; Howell, N. Reanalysis and revision of the Cambridge reference sequence for human mitochondrial DNA. Nat. Genet. 1999, 23, 147. [Google Scholar] [CrossRef]
  28. Anderson, S.; Bankier, A.T.; Barrell, B.G.; de Bruijn, M.H.; Coulson, A.R.; Drouin, J.; Eperon, I.C.; Nierlich, D.P.; Roe, B.A.; Sanger, F.; et al. Sequence and organization of the human mitochondrial genome. Nature 1981, 290, 457–465. [Google Scholar] [CrossRef]
  29. Holt, I.J.; Harding, A.E.; Petty, R.K.; Morgan-Hughes, J.A. A new mitochondrial disease associated with mitochondrial DNA heteroplasmy. Am. J. Hum. Genet. 1990, 46, 428–433. [Google Scholar]
  30. Tatuch, Y.; Christodoulou, J.; Feigenbaum, A.; Clarke, J.T.; Wherret, J.; Smith, C.S.; Rudd, N.; Petrova-Benedict, R.; Robinson, B.H. Heteroplasmic mtDNA mutation (T----G) at 8993 can cause Leigh disease when the percentage of abnormal mtDNA is high. Am. J. Hum. Genet. 1992, 50, 852–858. [Google Scholar]
  31. Larsen, N.B.; Rasmussen, M.; Rasmussen, L.J. Nuclear and mitochondrial DNA repair: Similar pathways? Mitochondrion 2005, 5, 89–108. [Google Scholar] [CrossRef] [PubMed]
  32. Fontana, G.A.; Gahlon, H.L. Mechanisms of replication and repair in mitochondrial DNA deletion formation. Nucleic Acids Res. 2020, 48, 11244–11258. [Google Scholar] [CrossRef]
  33. Gorman, G.S.; Chinnery, P.F.; DiMauro, S.; Hirano, M.; Koga, Y.; McFarland, R.; Suomalainen, A.; Thorburn, D.R.; Zeviani, M.; Turnbull, D.M. Mitochondrial diseases. Nat. Rev. Dis. Primers 2016, 2, 16080. [Google Scholar] [CrossRef] [PubMed]
  34. Maneiro, E.; Martín, M.A.; de Andres, M.C.; López-Armada, M.J.; Fernández-Sueiro, J.L.; del Hoyo, P.; Galdo, F.; Arenas, J.; Blanco, F.J. Mitochondrial respiratory activity is altered in osteoarthritic human articular chondrocytes. Arthritis Rheum. 2003, 48, 700–708. [Google Scholar] [CrossRef] [PubMed]
  35. Ruiz-Romero, C.; Calamia, V.; Mateos, J.; Carreira, V.; Martínez-Gomariz, M.; Fernández, M.; Blanco, F.J. Mitochondrial dysregulation of osteoarthritic human articular chondrocytes analyzed by proteomics: A decrease in mitochondrial superoxide dismutase points to a redox imbalance. Mol. Cell Proteom. 2009, 8, 172–189. [Google Scholar] [CrossRef] [Green Version]
  36. 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.; et al. Prevalence of nuclear and mitochondrial DNA mutations related to adult mitochondrial disease. Ann. Neurol. 2015, 77, 753–759. [Google Scholar] [CrossRef] [Green Version]
  37. Grishko, V.I.; Ho, R.; Wilson, G.L.; Pearsall, A.W., IV. Diminished mitochondrial DNA integrity and repair capacity in OA chondrocytes. Osteoarthr. Cartil. 2009, 17, 107–113. [Google Scholar] [CrossRef] [Green Version]
  38. Kim, J.; Xu, M.; Xo, R.; Mates, A.; Wilson, G.L.; Pearsall, A.W., IV. Mitochondrial DNA damage is involved in apoptosis caused by pro-inflammatory cytokines in human OA chondrocytes. Osteoarthr. Cartil. 2010, 18, 424–432. [Google Scholar] [CrossRef] [Green Version]
  39. Chang, M.C.; Hung, S.C.; Chen, W.Y.; Chen, T.L.; Lee, C.F.; Lee, H.C.; Wang, K.L.; Chiou, C.C.; Wei, Y.H. Accumulation of mitochondrial DNA with 4977-bp deletion in knee cartilage—an association with idiopathic osteoarthritis. Osteoarthr. Cartil. 2005, 13, 1004–1011. [Google Scholar] [CrossRef] [Green Version]
  40. Taylor, R.W.; Wardell, T.M.; Smith, P.M.; Muratovska, M.P.; Turnbull, D.M.; Lightowlers, R.N. An antigenomic strategy for treating heteroplasmic mtDNA disorders. Adv. Drug Deliv. Rev. 2001, 49, 121–125. [Google Scholar] [CrossRef]
  41. Chinnery, P.F.; Taylor, R.W.; Diekert, K.; Lill, R.; Turnbull, D.M.; Lighowlers, R.N. Peptide nucleic acid delivery to human mitochondria. Gene Ther. 1999, 6, 1919–1928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Muratovska, A.; Lightowlers, R.N.; Taylor, R.W.; Turnbull, D.M.; Smith, R.A.; Wilce, J.A.; Martin, S.W.; Murphy, M.P. Targeting peptide nucleic acid (PNA) oligomers to mitochondria withing cells by conjugation to lipophilic cations: Implications for mitochondrial DNA replication, expression and disease. Nucleic Acids Res. 2001, 29, 1852–1863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Tonin, Y.; Heckel, A.M.; Vysokikh, M.; Dovydenko, I.; Meschaninova, M.; Rötig, A.; Munnich, A.; Venyaminova, A.; Tarassov, I.; Entelis, N. Modeling of antigenomic therapy of mitochondrial diseases by mitochondrially addressed RNA targeting a pathogenic point mutation in mitochondrial DNA. J. Biol. Chem. 2014, 289, 13323–13334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Costa, N.D.; Thacker, J. The efficiency of restriction endonuclease digests determind by PCR. Biotechniques 1992, 13, 190. [Google Scholar] [PubMed]
  45. Bayona-Bafaluy, M.P.; Blits, B.; Battersby, B.J.; Shoubridge, E.A.; Moraes, C.T. Rapid directional shift of mitochondrial DNA heteroplasmy in animal tissues by a mitochondrially targeted restriction endonuclease. Proc. Natl. Acad. Sci. USA 2005, 102, 14392–14397. [Google Scholar] [CrossRef] [Green Version]
  46. Bacman, S.R.; Williams, S.L.; Moraes, C.T. Manipulation of mtDNA heteroplasmy in all striated muscles of newborn mice by AAV9-mediated delivery of a mitochondria-targeted restriction endonuclease. Gene Ther. 2012, 19, 1101–1106. [Google Scholar] [CrossRef] [Green Version]
  47. Bacman, S.R.; Williams, S.L.; Garcia, S.; Moraes, C.T. Organ-specific shifts in mtDNA heteroplasmy following systemic delivery of a mitochondria-targeted restriction endonuclease. Gene Ther. 2010, 17, 713–720. [Google Scholar] [CrossRef]
  48. Srivastava, S.; Moraes, C.T. Manipulating mitochondrial DNA heteroplasmy by a mitochondrially targeted restriction endonuclease. Hum. Mol. Genet. 2001, 10, 3093–3099. [Google Scholar] [CrossRef] [Green Version]
  49. Baracca, A.; Barogi, S.; Carelli, V.; Lenaz, G.; Solaini, G. Catalytic activities of mitochondrial ATP synthase in patients with mitochondrial DNA T8993G mutation in the ATPase 6 gene encoding subunit a. J. Biol. Chem. 2000, 275, 4177–4182. [Google Scholar] [CrossRef] [Green Version]
  50. Sgarbi, G.; Baracca, A.; Lenaz, G.; Valentino, L.M.; Carelli, V.; Solaini, G. Inefficient coupling between proton transport and ATP synthesis may be the pathogenic mechanism for NARP and Leigh syndrome resulting from the T8993G mutation in mtDNA. Biochem. J. 2006, 395, 493–500. [Google Scholar] [CrossRef] [Green Version]
  51. Tanaka, M.; Borgeld, H.J.; Zhang, J.; Muramatsu, S.; Gong, J.S.; Yoneda, M.; Maruyama, W.; Naoi, M.; Ibi, T.; Sahashi, K.; et al. Gene therapy for mitochondrial disease by delivering restriction endonuclease SmaI into mitochondria. J. Biomed. Sci. 2002, 9, 534–541. [Google Scholar] [PubMed]
  52. Urnov, F.D.; Rebar, E.J.; Holmes, M.C.; Zhang, H.S.; Gregory, P.D. Genome editing with engineered zinc finger nucleases. Nat. Rev. Genet. 2010, 11, 636–646. [Google Scholar] [CrossRef] [PubMed]
  53. Minczuk, M.; Papworth, M.A.; Kolasinska, P.; Murphy, M.P.; Klug, A. Sequence-specific modification of mitochondrial DNA using a chimeric zinc finger methylase. Proc. Natl. Acad. Sci. USA 2006, 103, 19689–19694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Minczuk, M.; Papworth, M.A.; Miller, J.C.; Murphy, M.P.; Klug, A. Development of a single-chain, quasi-dimeric zinc-finger nuclease for the selective degradation of mutated human mitochondrial DNA. Nucleic Acids Res. 2008, 36, 3926–3938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. 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.; et al. Near-complete elimination of mutant mtDNA by iterative or dynamic dose-controlled treatment with mtZFNs. Nucleic Acids Res. 2016, 44, 7804–7816. [Google Scholar] [CrossRef]
  56. Gammage, P.A.; Rorbach, J.; Vincent, A.I.; Rebar, E.J.; Minczuk, M. Mitochondrially targeted ZFNs for selective degradation of pathogenic mitochondrial genomes bearing large-scale deletions or point mutations. EMBO Mol. Med. 2014, 6, 458–466. [Google Scholar] [CrossRef]
  57. Gammage, P.A.; Viscomi, C.; Simard, M.L.; Costa, A.S.H.; Gaude, E.; Powell, C.A.; Van Haute, L.; McCann, B.J.; Rebelo-Guiomar, P.; Cerutti, R.; et al. Genome editing in mitochondria corrects a pathogenic mtDNA mutation in vivo. Nat. Med. 2018, 24, 1691–1695. [Google Scholar] [CrossRef]
  58. Szczepek, M.; Brondani, V.; Büchel, J.; Serrano, L.; Segal, D.J.; Cathomen, T. Structure-based redesign of the dimerization interface reduces the toxicity of zinc-finger nucleases. Nat. Biotechnol. 2007, 25, 786–793. [Google Scholar] [CrossRef]
  59. Pruett-Miller, S.M.; Connelly, J.P.; Maeder, M.L.; Joung, J.K.; Porteus, M.H. Comparison of zinc finger nucleases for use in gene targeting in mammalian cells. Mol. Ther. 2008, 16, 707–717. [Google Scholar] [CrossRef]
  60. Pereira, C.V.; Moraes, C.T. Current strategies towards therapeutic manipulation of mtDNA heteroplasmy. Front. Biosci. (Landmark Ed.) 2017, 22, 991–1010. [Google Scholar]
  61. Deng, D.; Yan, C.; Pan, X.; Mahfouz, M.; Wang, J.; Zhu, J.K.; Shi, Y.; Yan, N. Structural basis for sequence-specific recognition of DNA by TAL effectors. Science 2012, 335, 720–723. [Google Scholar] [CrossRef] [Green Version]
  62. Qasim, W.; Zhan, H.; Samarasinghe, S.; Adams, S.; Amrolia, P.; Stafford, S.; Butler, K.; Rivat, C.; Wright, G.; Somana, K.; et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci. Transl. Med. 2017, 9, 1–8. [Google Scholar] [CrossRef] [PubMed]
  63. Miller, J.C.; Tan, S.; Qiao, G.; Barlow, K.A.; Wang, J.; Xia, D.F.; Meng, X.; Paschon, D.E.; Leung, E.; Hinkley, S.J.; et al. A TALE nuclease architecture for efficient genome editing. Nat. Biotechnol. 2011, 29, 143–148. [Google Scholar] [CrossRef] [PubMed]
  64. Jun, A.S.; Trounce, I.A.; Brown, M.D.; Shoffner, J.M.; Wallace, D.C. Use of transmitochondrial cybrids to assign a complex I defect to the mitochondrial DNA-encoded NADH dehydrogenase subunit 6 gene mutation at nucleotide pair 14459 that causes Leber hereditary optic neuropathy and dystonia. Mol. Cell Biol. 1996, 16, 771–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Bacman, S.R.; Williams, S.L.; Pinto, M.; Peralta, S.; Moraes, C.T. Specific elimination of mutant mitochondrial genomes in patient-derived cells by mitoTALENs. Nat. Med. 2013, 19, 1111–1113. [Google Scholar] [CrossRef] [PubMed]
  66. Hashimoto, M.; Bacman, S.R.; Peralta, S.; Falk, M.J.; Chomyn, A.; Chan, D.C.; Williams, S.L.; Moraes, C. MitoTALEN: A general approach to reduce mutant mtDNA loads and restore oxidative phosphorylation function in mitochondrial diseases. Mol. Ther. 2015, 23, 1592–1599. [Google Scholar] [CrossRef] [Green Version]
  67. Pereira, C.V.; Bacman, S.R.; Arguello, T.; Zekonyte, U.; Williams, S.L.; Edgell, D.R.; Moraes, C.T. mitoTev-TALE: A monomeric DNA editing enzyme to reduce mutant mitochondrial DNA levels. EMBO Mol. Med. 2018, 10, e8084. [Google Scholar] [CrossRef]
  68. Yahata, N.; Matsumoto, Y.; Omi, M.; Yamamoto, N.; Hata, R. TALEN-mediated shift of mitochondrial DNA heteroplasmy in MELAS-iPSCs with m.13513G>A mutation. Sci. Rep. 2017, 7, 15557. [Google Scholar] [CrossRef] [Green Version]
  69. Yang, Y.; Wu, H.; Kang, X.; Liang, Y.; Lan, T.; Li, T.; Tan, T.; Peng, J.; Zhang, Q.; An, G.; et al. Targeted elimination of mutant mitochondrial DNA in MELAS-iPSCs by mitoTALENs. Protein. Cell 2018, 9, 283–297. [Google Scholar] [CrossRef] [Green Version]
  70. Mok, B.Y.; de Moraes, M.H.; Zeng, J.; Bosch, D.E.; Kotrys, A.V.; Raguram, A.; Hsu, F.; Radey, M.C.; Peterson, S.B.; Mootha, V.K.; et al. A bacterial cytidine deaminase toxin enables CRISPR-free mitochondrial base editing. Nature 2020, 583, 631–637. [Google Scholar] [CrossRef]
  71. Falkenberg, M.; Hirano, M. Editing the mitochondrial genome. N. Engl. J. Med. 2020, 383, 1489–1491. [Google Scholar] [CrossRef] [PubMed]
  72. Lee, H.; Lee, S.; Baek, G.; Kim, A.; Kang, B.C.; Seo, H.; Kim, J.S. Mitochondrial DNA editing in mice with DddA-TALE fusion deaminases. Nat. Commun. 2021, 12, 1190–1195. [Google Scholar] [CrossRef] [PubMed]
  73. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef] [PubMed]
  74. Chylinski, K.; Makarova, K.S.; Charpentier, E.; Koonin, E.V. Classification and evolution of type II CRISPR-Cas systems. Nucleic Acids Res. 2014, 42, 6091–6105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Porteus, M.H. A new class of medicines through DNA editing. N. Engl. J. Med. 2019, 380, 947–959. [Google Scholar] [CrossRef] [PubMed]
  76. Cyranoski, D. CRISPR gene-editing tested in a person for the first time. Nature 2016, 539, 479. [Google Scholar] [CrossRef]
  77. Stadtmauer, E.A.; Fraietta, J.A.; Davis, M.M.; Cohen, A.D.; Weber, K.L.; Lancaster, E.; Mangan, P.A.; Kulikovskaya, I.; Gupta, M.; Chen, F.; et al. CRISPR-engineered T cells in patients with refractory cancer. Science 2020, 367, 1–13. [Google Scholar] [CrossRef]
  78. The Lancet Haematology. CRISPR-Cas9 gene editing for patients with haemoglobinopathies. Lancet Haematol. 2019, 6, e438. [Google Scholar] [CrossRef]
  79. Xu, L.; Wang, J.; Liu, Y.; Xie, L.; Su, B.; Mou, D.; Wang, L.; Liu, T.; Wang, X.; Zhang, B.; et al. CRISPR-edited stem cells in a patient with HIV and acute lymphocytic leukemia. N. Engl. J. Med. 2019, 381, 1240–1247. [Google Scholar] [CrossRef]
  80. Jo, A.; Ham, S.; Lee, G.H.; Lee, Y.I.; Kim, S.; Lee, Y.S.; Shin, J.H.; Lee, Y. Efficient mitochondrial genome editing by CRISPR/Cas9. Biomed. Res. Int. 2015, 2015, 305716. [Google Scholar] [CrossRef] [Green Version]
  81. Gammage, P.A.; Moraes, C.T.; Minczuk, M. Mitochondrial genome engineering: The revolution may not be CRISPR-Ized. Trends Genet. 2018, 34, 101–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Mahata, B.; Mukherjee, S.; Mishra, S.; Bandyopadhyay, A.; Adhya, S. Functional delivery of a cytosolic tRNA into mutant mitochondria of human cells. Science 2006, 314, 471–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Goswami, S.; Dhar, G.; Mukherjee, S.; Mahata, B.; Chatterjee, S.; Home, P.; Adhya, S. A bifunctional tRNA import receptor from Leishmania mitochondria. Proc. Natl. Acad. Sci. USA 2006, 103, 8354–8359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Entelis, N.; Brandina, I.; Kamenski, P.; Krasheninnikov, I.A.; Martin, R.P.; Tarassov, I. A glycolytic enzyme, enolase, is recruited as a cofactor of tRNA targeting toward mitochondria in Saccharomyces cerevisiae. Genes Dev. 2006, 20, 1609–1620. [Google Scholar] [CrossRef] [Green Version]
  85. Moretton, A.; Morel, F.; Macao, B.; Lachaume, P.; Ishak, L.; Lefebvre, M.; Garreau-Balandier, I.; Vernet, P. Selective mitochondrial DNA degradation following double-strand breaks. PLoS ONE 2017, 12, e0176795. [Google Scholar] [CrossRef] [Green Version]
  86. Hagstrom, E.; Freyer, C.; Battersby, B.J.; Stewart, J.B.; Larsson, N.G. No recombination of mtDNA after heteroplasmy for 50 generations in the mouse maternal germline. Nucleic Acids Res. 2014, 42, 1111–1116. [Google Scholar] [CrossRef] [Green Version]
  87. Weinberg, S.E.; Sena, L.A.; Chandel, N.S. Mitochondria in the regulation of innate and adaptive immunity. Immunity 2015, 42, 406–417. [Google Scholar] [CrossRef] [Green Version]
  88. Filograna, R.; Koolmeister, C.; Upadhyay, M.; Pajak, A.; Clemente, P.; Wibom, R.; Simard, M.L.; Wredenberg, A.; Freyer, C.; Stewart, J.B.; et al. Modulation of mtDNA copy number ameliorates the pathological consequences of a heteroplasmic mtDNA mutation in the mouse. Sci. Adv. 2019, 5, eaav9824. [Google Scholar] [CrossRef] [Green Version]
  89. Rey-Rico, A.; Cucchiarini, M. Controlled release strategies for rAAV-mediated gene delivery. Acta Biomater. 2016, 29, 1–10. [Google Scholar] [CrossRef]
  90. Cucchiarini, M.; Madry, H. Biomaterial-guided delivery of gene vectors for targeted articular cartilage repair. Nat. Rev. Rheumatol. 2019, 15, 18–29. [Google Scholar] [CrossRef]
  91. Jiang, D.; Gao, F.; Zhang, Y.; Wong, D.S.; Li, Q.; Tse, H.F.; Xu, G.; Yu, Z.; Lian, Q. Mitochondrial transfer of mesenchymal stem cells effectively protects corneal epithelial cells from mitochondrial damage. Cell Death Dis. 2016, 7, e2467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Influence of mtDNA heterogeneity and mitochondrial functions in OA. Under the influence of various risk factors, mtDNA mutations are observed in OA, affecting the synthesis of oxidative phosphorylation-related enzymes. Reduced synthesis of these enzymes leads to a poor transfer of high-energy electrons, which, in turn, reduces the production of ATP, increases the production of ROS and reduces the mitochondrial membrane potential. Increased ROS can subsequently aggravate the oxidative damage of chondrocytes, leading to cartilage degradation. Abbreviations: OA, osteoarthritis; mtDNA, mitochondrial DNA; ATP, adenosine triphosphate; ROS, reactive oxygen species; NAD, nicotinamide adenine dinucleotide; FAD, flavin adenine dinucleotide; Cyt c, cytochrome c; ADP, adenosine diphosphate (created with BioRender).
Figure 1. Influence of mtDNA heterogeneity and mitochondrial functions in OA. Under the influence of various risk factors, mtDNA mutations are observed in OA, affecting the synthesis of oxidative phosphorylation-related enzymes. Reduced synthesis of these enzymes leads to a poor transfer of high-energy electrons, which, in turn, reduces the production of ATP, increases the production of ROS and reduces the mitochondrial membrane potential. Increased ROS can subsequently aggravate the oxidative damage of chondrocytes, leading to cartilage degradation. Abbreviations: OA, osteoarthritis; mtDNA, mitochondrial DNA; ATP, adenosine triphosphate; ROS, reactive oxygen species; NAD, nicotinamide adenine dinucleotide; FAD, flavin adenine dinucleotide; Cyt c, cytochrome c; ADP, adenosine diphosphate (created with BioRender).
Ijms 23 01467 g001
Figure 2. Mitochondrial genome editing strategies to target the mtDNA. The approaches include the use of antigenomic therapy, restriction endonucleases, ZFNs, TALENs and CRISPR/Cas9. Abbreviations: mtDNA, mitochondrial DNA; ZFNs, zinc-finger nucleases; TALENS, transcription activator-like effectors nucleases; CRISPR/Cas9, clustered regularly interspaced short palindromic repeats/CRISPR-associated 9; MTS, mitochondria-targeting sequence; PNA, peptide nucleic acid; sgRNA, single guide RNA; WT, wild-type; DSBs, double-strand breaks (created with BioRender).
Figure 2. Mitochondrial genome editing strategies to target the mtDNA. The approaches include the use of antigenomic therapy, restriction endonucleases, ZFNs, TALENs and CRISPR/Cas9. Abbreviations: mtDNA, mitochondrial DNA; ZFNs, zinc-finger nucleases; TALENS, transcription activator-like effectors nucleases; CRISPR/Cas9, clustered regularly interspaced short palindromic repeats/CRISPR-associated 9; MTS, mitochondria-targeting sequence; PNA, peptide nucleic acid; sgRNA, single guide RNA; WT, wild-type; DSBs, double-strand breaks (created with BioRender).
Ijms 23 01467 g002
Table 1. Genome editing technologies.
Table 1. Genome editing technologies.
TypesRecognition SitesAdvantagesLimitationsRefs.
antigenomic mtDNAantigenomic-BSeasy to designlarge, limited target sites, poor specificity[40,41,42,43]
REsRE-BSeasy to designlarge, limited target sites, poor specificity[44,45,46,47,48,49,50,51]
ZFNsZFN-BSsmall, highly specificdifficult to design, limited target sites[52,53,54,55,56,57,58]
TALENsTALEN-BSeasy to design, highly specificlarge, limited target sites[60,61,62,63,64,65,66,67,68,69,70,71,72]
CRISPR/Cas9CRISPR/Cas9-BSeasy to designgRNA import, limited target sites[80,82,83,84]
Abbreviations: mtDNA, mitochondrial DNA; REs, restriction endonucleases; ZFNs, zinc-finger nucleases; TALENS, transcription activator-like effectors nucleases; CRISPR/Cas9, clustered regularly interspaced short palindromic repeats/CRISPR-associated 9; BS, binding sites.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhong, G.; Madry, H.; Cucchiarini, M. Mitochondrial Genome Editing to Treat Human Osteoarthritis—A Narrative Review. Int. J. Mol. Sci. 2022, 23, 1467. https://doi.org/10.3390/ijms23031467

AMA Style

Zhong G, Madry H, Cucchiarini M. Mitochondrial Genome Editing to Treat Human Osteoarthritis—A Narrative Review. International Journal of Molecular Sciences. 2022; 23(3):1467. https://doi.org/10.3390/ijms23031467

Chicago/Turabian Style

Zhong, Gang, Henning Madry, and Magali Cucchiarini. 2022. "Mitochondrial Genome Editing to Treat Human Osteoarthritis—A Narrative Review" International Journal of Molecular Sciences 23, no. 3: 1467. https://doi.org/10.3390/ijms23031467

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop