Abstract
Mitochondrion is a semi-autonomous organelle, important for cell energy metabolism, apoptosis, the production of reactive oxygen species (ROS), and Ca2+ homeostasis. Mitochondrial DNA (mtDNA) mutation is one of the primary factors in mitochondrial disorders. Though much progress has been made, there remain many difficulties in constructing cell models for mitochondrial diseases. This seriously restricts studies related to targeted drug discovery and the mechanism and therapy for such diseases. Here we summarize the characteristics of patient-specific immortalized lymphoblastoid cells, fibroblastoid cells, cytoplasmic hybrid (cybrid) cell lines, and induced pluripotent stem cells (iPSCs)-derived differentiation cells in the study of mitochondrial disorders, as well as offering discussion of roles and advances of these cell models, particularly in the screening of drugs.
Keywords: Mitochondrial diseases, Mitochondrial DNA, Cell model, Drug discovery
1. Introduction
In many mitochondrial diseases, mitochondrial dysfunction triggers profound clinical manifestations, especially those relating to defects in oxidative phosphorylation. Incidences of mitochondrial diseases are conservatively estimated to occur in one in 6500 people (Yan and Guan, 2008). Primary mutations in the mitochondrial DNA (mtDNA) could disrupt mitochondrial function and contribute to mitochondrial diseases. These mainly affect tissues and organs that have high energy requirements, including those of the heart, brain, and skeletal muscle (Wallace, 2013). Mitochondria are semi-autonomous organelles, which play central roles in cell energy production, regulation of apoptosis, generation of reactive oxygen species (ROS), and calcium homeostasis. Mitochondrial functions are cooperatively controlled by nuclear DNA (nDNA) and mtDNA. While nDNA encodes approximately 1500 mitochondrial proteins, mtDNA encodes only 13 polypeptide components of the electron transport chain complexes (ETCs), as well as the 22 transfer RNAs (tRNAs) and 2 ribosomal RNAs (rRNAs) in the mitochondrion. Since the mtDNA mutation and deletion were first reported to be associated with mitochondrial diseases in 1988 (Holt et al., 1988; Wallace et al., 1988), hundreds of variants in mtDNA related to mitochondrial diseases have now been identified.
Drug discovery is the preliminary process of detecting and studying the pharmacological activity of potential therapeutic compounds. Traditionally, drug discovery is based on the search for target molecules, which possibly represents the primary focus of the pharmaceutical industry. However, identifying disease-related drug targets is rarely a straight forward or simple process. One of the reasons is the existence of compensation mechanisms and feedback pathways in the complex cellular environment, which often causes the targeting and therapeutic efficacy of drugs to be greatly reduced, even when the drug has been previously demonstrated to be effective at the molecular level in vitro (Inak et al., 2017). In this way, attempts at target-based drug discovery not only cost time and money, but also have less success. Currently, phenotypic screening has regained momentum and is receiving extensive attention. The discovery of phenotypic drugs initially requires the identification of disease-specific features. Although cellular or animal models of nuclear gene mutation-associated diseases can be generated by techniques such as gene knockout, knockdown, or editing, there are still many difficulties in constructing cell or animal models for mitochondrial diseases. Every mammalian cell contains hundreds or even thousands of mitochondria with lipid bilayer membranes. Each mitochondrion contains multiple mtDNA copies. Specific and efficient genetic operating systems have not yet been established for site-directed mutagenesis or modification of mtDNA (Wallace and Fan, 2009; Farrar et al., 2013). Currently, somatic cell nuclear transfer (SCNT) technology has been shown to be capable of replacing mtDNA of fibroblasts from Leigh syndrome patients harboring the m.8993T>C mutation (Ma et al., 2015). The pathogenic mitochondrial mutations of the oocyte have been exchanged using mitochondrial replacement therapy (Kang E et al., 2016), and “three-parent babies” have been born based on mitochondrial donation linked with in vitro fertilization (IVF) (Reardon, 2016). Nevertheless, for mitochondrial replacement therapy, ethical and practical questions remain. The possibility that phenotypic changes may result due to the mismatch between nDNA and mtDNA is of concern and overall, the whole process is still hampered by technical difficulties and high costs (Inak et al., 2017). Up to the present, peripheral blood lymphocytes or fibroblasts from patients, and their derived cytoplasmic hybrid (cybrid) cells have been the main focus for conventional strategies of the study of mitochondrial diseases (Wu et al., 2016). However, in recent years, patient-specific induced pluripotent stem cells (iPSCs) reprogrammed from somatic cells and iPSC-derived differentiation cells have been widely recognized as providing new opportunities for novel cell models of disease (Yamanaka, 2007).
Here we summarize the generation and characteristics of patient-specific immortalized lymphoblastoid cells, fibroblastoid cells, cybrid cells, and iPSC-derived cells in mitochondrial disorders. We assess the roles and advances of these cell models in the screening of mitochondrial drugs, and in the light of these, analyze and discuss the existing problems and potential developmental directions.
2. Lymphoblastoid cells and fibroblasts for mitochondrial drug discovery
Immortalized lymphoblastoid cell lines are produced from the transformation of peripheral blood B lymphocytes by the Epstein Barr virus (EBV) (Amoli et al., 2008). There are several advantages of using immortalized lymphoblastoid cells as a drug screening model for mitochondrial diseases. Firstly, large quantities of lymphocytes are available and can be easily obtained from human peripheral blood. Secondly, lymphocytes can be immortalized effectively to proliferate indefinitely (Sun et al., 2016). Finally, cells carrying mtDNA mutations can perpetually maintain their biological properties at a cellular level. A large proportion of the mitochondrial drugs that have been approved or are currently being developed are small molecule compounds. The efficiency and accuracy of mitochondrial drug discovery can be improved greatly using such cell models of disease. To evaluate the use of patient-derived lymphoblastoid cell lines for small molecule drug discovery in mitochondrial disease, Chin et al. (2018) constructed five immortalized lymphoblastoid cell lines harboring mtDNA point mutations. Idebenone, an analog of coenzyme Q (CoQ), increased the basal respiration of Leber’s hereditary optic neuropathy (LHON) lymphoblastoid cell lines harboring the m.3460G>A variant in MT-ND1 or the m.11778G>A variant in MT-ND4, but not in Leigh disease cells harboring the m.8993T>G variant in MT-ATP6 (Chin et al., 2018). GNE-7915, a leucine-rich repeat kinase 2 (LRRK2) inhibitor, could repair the mtDNA damage caused by the LRRK2 p.G2019S mutation associated with Parkinson’s disease (Howlett et al., 2017). These results indicate that patient-specific immortalized lymphoblastoid cells carrying mtDNA variants can exhibit different sensitivities to the same drug. This reveals the potential value of lymphoblastoid as a mitochondrial drug screening cell model.
Fibroblasts are much enriched and easily obtained during a muscle biopsy. Fibroblast cultures are highly proliferative and provide a renewable source of cells in vitro. Patient’s fibroblasts harboring an mtDNA variant may provide a suitable platform for the search of small molecules for the treatment of mitochondrial disease (Saada, 2011). The deficiency of the mitochondrial ETC complex I (nicotinamide adenine dinucleotide (NADH) ubiquinone oxidoreductase) is the most common form of oxidative phosphorylation (OXPHOS) defect. It has no known effective cure and results in decreased ATP, increased ROS, and imbalanced NAD+/NADH ratio due to NADH accumulation (Pfeffer et al., 2013). Mitochondria-targeted antioxidants, succinate or short chain quinones, could bypass the defective complex I and directly transfer electrons to the next complexes. This is the main treatment strategy of complex I defects (Pfeffer et al., 2013; Koopman et al., 2016). Saada (2011) evaluated the effectiveness of the drug by measuring the ATP content of a fibroblast cell line with complex I deficiency. However, in clinical trials, different responses to the same drug have been found from different patients, suggesting that it is necessary to test the therapeutic effect of drugs on individual patients, such that patient-specific cell models may be necessary in order to deliver personalized therapy. The m.3243A>G (MT-TL1) variant is one of the major pathogenic mutations in mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS). In this condition, a decrease in mitochondrial membrane potential and mitochondrial respiratory chain enzyme activity was found in fibroblasts, together with the activation of autophagy. All of these abnormalities were significantly restored after CoQ treatment (Cotán et al., 2011). Antioxidants such as N-acetylcysteine (NAC) and dihydrolipoic acid have been applied to treat patients with neuropathy, ataxia, and retinitis pigmentosa (NARP) fibroblasts harboring the m.8993T>G variant. In this treatment, mitochondrial function was recovered with a notable increase in mitochondrial oxygen consumption rate and ATP synthesis (Mattiazzi et al., 2004). Overall, patient-derived fibroblasts are suitable cell models for drug discovery. More information about immortalized lymphocytes and fibroblasts as models of drug discovery is available in Table 1.
Table 1.
Lymphoblastoid cells and fibroblasts for mitochondrial drug discovery
Cell model | Disease | Variant site | Drug | Reference |
Lymphoblastoid cells | LHON | m.3460G>A | Idebenone | Chin et al., 2018 |
LHON | m.11778G>A | Idebenone | Chin et al., 2018 | |
Fibroblastoid cells | MELAS | m.3243A>G | CoQ10 | Cotán et al., 2011 |
MELAS | m.3243A>G | Riboflavin or CoQ10 | Garrido-Maraver et al., 2012 | |
NARP and MILS | m.8993T>G | N-acetylcysteine; dihydrolipoic acid | Mattiazzi et al., 2004 | |
MERRF | m.8344A>G | CoQ10 | de la Mata et al., 2012 |
LHON: Leber’s hereditary optic neuropathy; MELAS: mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes; NARP: neuropathy, ataxia, and retinitis pigmentosa; MILS: maternally inherited Leigh syndrome; MERRF: myoclonic epilepsy with ragged red fibers; CoQ: coenzyme Q
3. Cytoplasmic hybrid cells for mitochondrial drug discovery
Mitochondrial function is controlled by both nuclear and mitochondrial genes. Immortalized lymphocytes or fibroblasts harboring the same pathogenic mtDNA variants derived from the same pedigree may exhibit different mitochondrial function resulting from their different nuclear genes. In order to eliminate the interference of this nuclear background, the nucleus was removed from patient-derived cells such as immortalized lymphocytes or fibroblasts, and the cytoplasts were fused with ρ0 cells lacking mtDNA. After screening, cybrid cell lines with a consistent nuclear background were constructed. This provided an important and effective cell model for mitochondrial disorders reflecting the effects of mtDNA mutations on mitochondrial function (Wilkins et al., 2014).
This cybrid cell model is of great value for mitochondrial drug discovery. Barrow et al. (2016) designed and developed a high-throughput in-cell enzyme-linked immunoassay based on the mitochondrial cell model and identified the I-BET 525762A, a bromodomain inhibitor, from 10 015 small molecule compound libraries. This could lead to the dramatic accumulation of the complex IV subunit COX5A expression levels, and protect the cybrids with the oxidative respiratory chain complex I mutation (m.3796A>G, found in adult onset dystonia) from cell death. Since cells are forced to produce ATP by oxidative phosphorylation in media with galactose, they found that m.3796A>G mutant cybrids died within 72 h while I-BET 525762A-treated cells could still survive. After performing genome-wide editing clustered regularly interspaced short palindromic repeat (CRISPR) screening, the researchers confirmed that I-BET 525762A could remodel the mitochondrial proteome and increase the expression and activity of OXPHOS protein complexes, which rescued from the bioenergetic defects and cell death caused by complex I variants (Barrow et al., 2016). NARP and maternally inherited Leigh syndrome (MILS), which are both mitochondrial disorders, could be attributed to the m.8993T>G mutation. Fibroblasts from patients with NARP tend to undergo apoptosis and abnormal OXPHOS with decreased ATP synthesis, owing to a large amount of generated free radicals. Mattiazzi et al. (2004) found that antioxidants such as NAC and dihydrolipoic acid (DHLPA) could rescue mitochondrial functions, including aerobic respiration and ATP synthesis in transferring mitochondria cells with the m.8993T>G variant. Cybrid cells carrying the same mutation also present CoQ10 deficiency and elevated autophagy activity. All these abnormal phenotypes were ameliorated after CoQ treatment. This was in accordance with m.3243A>G variant fibroblasts (Cotán et al., 2011). β-Lapachone (β-lap) is a substrate of NAD(P)H:quinone oxidoreductase 1 (NQO1), which can increase significantly energy production, reduce ROS and lactate generation, and restore mitochondrial content in MELAS cybrid cells (Jeong et al., 2014). In general, patient-derived mitochondrial cybrids are promising cell models in drug screening and may be applied equally to other mitochondrial diseases. More information about cybrid cells as models of mitochondrial disease drug screening is shown in Table 2.
Table 2.
Cytoplasmic hybrid cells for mitochondrial drug discovery
Disease | Variant site | Drug | Reference |
MELAS | m.3243A>G | CoQ10 | Cotán et al., 2011 |
m.3243A>G | Riboflavin or CoQ10 | Garrido-Maraver et al., 2012 | |
m.3243A>G | β-Lapachone | Jeong et al., 2014 | |
MERRF | m.8344A>G | CoQ10 | de la Mata et al., 2012 |
Adult onset dystonia | m.3796A>G | I-BET 525762A | Barrow et al., 2016 |
NARP and MILS | m.8993T>G | N-acetylcysteine or dihydrolipoic acid | Mattiazzi et al., 2004 |
NARP | m.8993T>G | Sodium pyrithione | Aiyar et al., 2014 |
m.8993T>G | Chlorhexidine or oleic acid | Couplan et al., 2011 |
MELAS: mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes; MERRF: myoclonic epilepsy with ragged red fibers; NARP: neuropathy, ataxia, and retinitis pigmentosa; MILS: maternally inherited Leigh syndrome; CoQ: coenzyme Q
4. iPSCs and iPSC-derived cells for mitochondrial drug discovery
The phenotypes that result from mtDNA mutations often exhibit tissue specificity where lymphocytes or fibroblasts, as common models, have significant differences in morphology, structure, and function from the cells of affected tissues. Whether such models can reflect the real functions and characteristics of the such tissues remains an area of debate. iPSCs are a class of cells reprogrammed from terminally differentiated cells. They are identical to embryonic stem cells in self-renewal and multi-directional differentiation potential. iPSCs have attracted great attention as a new disease cell model due to their abundant cell sources, unlimited powers of proliferation, and ability to differentiate into various functional cells. During the process of reprogramming and directed differentiation, disease-specific iPSCs derived from somatic cells of patients retain the disease-causing genes.
Recently, we generated patient-specific iPSCs and iPSC-derived cardiomyocytes (iPSC-CMs) from a maternally inherited hypertrophic cardiomyopathy (HCM) family who carry the m.2336T>C variant in the mitochondrial 16S rRNA gene (MT-RNR2) (Li et al., 2018). Results showed that m.2336T>C variant led to enlarged cell size, increased action potential delay after depolarizations (DADs), and prolonged action potential duration (APD), all of which were similar to that observed in HCM cardiomyocytes. Functional research on such cells has shown that the ATP/ADP ratio and mitochondrial membrane potential (Δψ m) are also reduced, thereby elevating the intracellular Ca2+ concentration and decreasing in L-type Ca2+ current (I CaL) (Li et al., 2018). Our findings therefore provide an ideal model for both the study of the pathogenesis of maternally inherited HCM and for drug discovery. Several other patient-specific iPSCs carrying mtDNA mutations and iPSC-derived cell models have also been generated. These include a 2.5-kb deletion of mtDNA associated with Pearson bone marrow pancreatic syndrome (PS) (Cherry et al., 2013), the m.3243A>G variant in mitochondrial causing MELAS (Ma et al., 2015) or diabetes (Fujikura et al., 2012; Matsubara et al., 2018), the m.8344A>G variant correlated with myoclonic epilepsy with ragged red fibers (MERRF) syndrome (Chou et al., 2016, 2018), the m.11778G>A variant implicated in LHON (Lu et al., 2018; Wu et al., 2018), and the m.1555A>G variant related to sensorineural hearing loss (SNHL) (Hsu et al., 2017).
The iPSC-derived cells from patients were able to model the specific related pathology (Ebert et al., 2009; Kang JF et al., 2016), which could be rescued after drug therapy. Lorenz et al. (2017) generated iPSCs and their differentiated neural progenitor cells (NPCs) carrying the m.9185T>C variant in MT-ATP6 related to Leigh syndrome. Using high-throughput screening, it was found that avanafil as a phosphodiesterase type 5 (PDE5) inhibitor could dramatically improve the mitochondrial membrane potential and adjust mitochondrial calcium homeostasis, which in turn rescued the function of patient’s NPCs and differentiated neurons (Lorenz et al., 2017). mtDNA depletion syndrome 3 (MTDPS3) is a kind of genetic disorder characterized by mtDNA depletion and comprised ATP production, while deoxyguanosine kinase (DGUOK) deficiency is among the most common causes of hepatic mtDNA depletion syndrome (Cámara et al., 2014). DGUOK−/− iPSCs were generated using CRISPR/CRISPR-associated protein 9 (CAS9) and iPSC-derived hepatocyte-like cells to identify drugs that could elevate ATP synthesis in DGUOK-deficient hepatocytes by phenotypic screening. It was found that NAD treatment resulted in an improvement in intracellular NAD+ levels, promoting Sirtuin 1 (SIRT1)-mediated peroxisome proliferator-activated receptor-γ coactivator-1-α (PGC-1α) activation. Addition of NAD also increased the levels of messenger RNAs (mRNAs) of fatty acid oxidation (FAO) and tricarboxylic acid (TCA) cycling, indicating a boost in fatty acid and glucose metabolism, which further strengthened mitochondrial activity and ATP synthesis (Jing et al., 2018).
iPSCs, along with iPSC-derived cells, still have a long way to go for drug discovery. Differentiating iPSCs into specific cells not only takes time, but also tends to lead to heteroplasmy when cells differentiated from different batches of iPSCs are compared (Cherry et al., 2013). In addition, iPSC-derived cells as a drug screening model have certain limitations. Since human beings represent an integrated and complex system, different types of cells can interact with each other. Even though iPSCs can differentiate into specific cells, it is difficult to totally imitate the pathophysiological condition of the human body (Cámara et al., 2014). Improving the differentiation efficiency of iPSCs and studying how to effectively simulate complex human physiological states are key areas for future research.
5. Conclusions
It should also be noted that there are some limitations of the above cell models of mitochondrial diseases for drug screening. Mitochondrial disorders usually affect high energy demand tissues. Lymphoblasts and fibroblasts are not considered as high energy demand. Cybrid cells disrupt the patient-specific interaction between nDNA and mtDNA, as all nDNA-encoded mitochondrial proteins in cybrids come from ρ0 cells (Inak et al., 2017). Human iPSC-derived cells exhibit high phenotypic immaturity and variability, which presents some differences compared with adult terminally differentiated cells (Boulting et al., 2011; Sala et al., 2017). On the other hand, with the continuous improvement of high throughput drug screening technology, intelligent information collection and analysis, and a small molecule compound library and natural product library, it will be possible to miniaturize, scale, automate, and intellectualize phenotype drug screening. Appropriate cell models thus contribute to providing insights into the pathogenesis of mitochondrial disorders with mtDNA mutations, and shortening the period of drug discovery for mitochondrial diseases.
Acknowledgments
We thank Chris WOOD of the College of Life Sciences Zhejiang University (Hangzhou, China) for checking the English of this manuscript.
Footnotes
Project supported by the National Basic Research Program of China (No. 2014CB943001), the National Natural Science Foundation of China (Nos. 31771398 and 31571299), the Fundamental Research Funds for the Central Universities (No. 2019QNA6001), and the Zhejiang Provincial Natural Science Foundation of China (Nos. LZ19C060001 and LY14C060004)
Contributors: Qing-feng YAN conceived and designed the review. Shuang-yi HU, Qian-qian ZHUANG, Yue QIU, and Xu-fen ZHU collected the references and wrote the manuscript. Xu-fen ZHU and Qing-feng YAN revised the manuscript. All authors read and approved the final manuscript.
Compliance with ethics guidelines: Shuang-yi HU, Qian-qian ZHUANG, Yue QIU, Xu-fen ZHU, and Qing-feng YAN declare that they have no conflict of interest.
This article does not contain any studies with human or animal subjects performed by any of the authors.
References
- 1.Aiyar RS, Bohnert M, Duvezin-Caubet S, et al. Mitochondrial protein sorting as a therapeutic target for ATP synthase disorders. Nat Commun, 5:5585. 2014 doi: 10.1038/ncomms6585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Amoli MM, Carthy D, Platt H, et al. EBV Immortalization of human B lymphocytes separated from small volumes of cryo-preserved whole blood. Int J Epidemiol. 2008;37(Suppl 1):i41–i45 . doi: 10.1093/ije/dym285. [DOI] [PubMed] [Google Scholar]
- 3.Barrow JJ, Balsa E, Verdeguer F, et al. Bromodomain inhibitors correct bioenergetic deficiency caused by mitochondrial disease complex I mutations. Mol Cell. 2016;64(1):163–175. doi: 10.1016/j.molcel.2016.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Boulting GL, Kiskinis E, Croft GF, et al. A functionally characterized test set of human induced pluripotent stem cells. Nat Biotechnol. 2011;29(3):279–286. doi: 10.1038/nbt.1783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Cámara Y, González-Vioque E, Scarpelli M, et al. Administration of deoxyribonucleosides or inhibition of their catabolism as a pharmacological approach for mitochondrial DNA depletion syndrome. Hum Mol Genet. 2014;23(9):2459–2467. doi: 10.1093/hmg/ddt641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Cherry ABC, Gagne KE, Mcloughlin EM, et al. Induced pluripotent stem cells with a mitochondrial DNA deletion. Stem Cells. 2013;31(7):1287–1297. doi: 10.1002/stem.1354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chin RM, Panavas T, Brown JM, et al. Patient-derived lymphoblastoid cell lines harboring mitochondrial DNA mutations as tool for small molecule drug discovery. BMC Res Notes, 11:205. 2018 doi: 10.1186/s13104-018-3297-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Chou SJ, Tseng WL, Chen CT, et al. Impaired ROS scavenging system in human induced pluripotent stem cells generated from patients with MERRF syndrome. Sci Rep, 6:23661. 2016 doi: 10.1038/srep23661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chou SJ, Ko YL, Yang YH, et al. Generation of two isogenic human induced pluripotent stem cell lines from a 15 year-old female patient with MERRF syndrome and A8344G mutation of mitochondrial DNA. Stem Cell Res. 2018;30:201–205. doi: 10.1016/j.scr.2018.05.011. [DOI] [PubMed] [Google Scholar]
- 10.Cotán D, Cordero MD, Garrido-Maraver J, et al. Secondary coenzyme Q10 deficiency triggers mitochondria degradation by mitophagy in MELAS fibroblasts. FASEB J. 2011;25(8):2669–2687. doi: 10.1096/fj.10-165340. [DOI] [PubMed] [Google Scholar]
- 11.Couplan E, Aiyar RS, Kucharczyk R, et al. A yeast-based assay identifies drugs active against human mitochondrial disorders. Proc Natl Acad Sci USA. 2011;108(29):11989–11994. doi: 10.1073/pnas.1101478108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.de la Mata M, Garrido-Maraver J, Cotán D, et al. Recovery of MERRF fibroblasts and cybrids pathophysiology by coenzyme Q10 . Neurotherapeutics. 2012;9(2):446–463. doi: 10.1007/s13311-012-0103-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ebert AD, Yu JY, Rose FF, Jr, et al. Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature. 2009;457(7227):277–280. doi: 10.1038/nature07677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Farrar GJ, Chadderton N, Kenna PF, et al. Mitochondrial disorders: aetiologies, models systems, and candidate therapies. Trends Genet. 2013;29(8):488–497. doi: 10.1016/j.tig.2013.05.005. [DOI] [PubMed] [Google Scholar]
- 15.Fujikura J, Nakao K, Sone M, et al. Induced pluripotent stem cells generated from diabetic patients with mitochondrial DNA A3243G mutation. Diabetologia. 2012;55(6):1689–1698. doi: 10.1007/s00125-012-2508-2. [DOI] [PubMed] [Google Scholar]
- 16.Garrido-Maraver J, Cordero MD, Moñino ID, et al. Screening of effective pharmacological treatments for MELAS syndrome using yeasts, fibroblasts and cybrid models of the disease. Br J Pharmacol. 2012;167(6):1311–1328. doi: 10.1111/j.1476-5381.2012.02086.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Holt IJ, Harding AE, Morgan-Hughes JA. Deletions of muscle mitochondrial DNA in patients with mitochondrial myopathies. Nature. 1988;331(6158):717–719. doi: 10.1038/331717a0. [DOI] [PubMed] [Google Scholar]
- 18.Howlett EH, Jensen N, Belmonte F, et al. LRRK2 G2019S-induced mitochondrial DNA damage is LRRK2 kinase dependent and inhibition restores mtDNA integrity in Parkinson’s disease. Hum Mol Genet. 2017;26(22):4340–4351. doi: 10.1093/hmg/ddx320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hsu YH, Wu YT, Huang CY, et al. Generation of an induced pluripotent stem cell line from a 39-year-old female patient with severe-to-profound non-syndromic sensorineural hearing loss and a A1555G mutation in the mitochondrial MTRNR1 gene. Stem Cell Res. 2017;25:245–249. doi: 10.1016/j.scr.2017.10.024. [DOI] [PubMed] [Google Scholar]
- 20.Inak G, Lorenz C, Lisowski P, et al. Concise review: induced pluripotent stem cell-based drug discovery for mitochondrial disease. Stem Cells. 2017;35(7):1655–1662. doi: 10.1002/stem.2637. [DOI] [PubMed] [Google Scholar]
- 21.Jeong MH, Kim JH, Seo KS, et al. β-Lapachone attenuates mitochondrial dysfunction in MELAS cybrid cells. Biochem Biophys Res Commun. 2014;454(3):417–422. doi: 10.1016/j.bbrc.2014.10.093. [DOI] [PubMed] [Google Scholar]
- 22.Jing R, Corbett JL, Cai J, et al. A screen using iPSC-derived hepatocytes reveals NAD+ as a potential treatment for mtDNA depletion syndrome. Cell Rep. 2018;25(6):1469–1484e5. doi: 10.1016/j.celrep.2018.10.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kang E, Wu J, Gutierrez NM, et al. Mitochondrial replacement in human oocytes carrying pathogenic mitochondrial DNA mutations. Nature. 2016;540(7632):270–275. doi: 10.1038/nature20592. [DOI] [PubMed] [Google Scholar]
- 24.Kang JF, Tang BS, Guo JF. The progress of induced pluripotent stem cells as models of Parkinson’s disease. Stem Cells Int, 2016:4126214. 2016 doi: 10.1155/2016/4126214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Koopman WJH, Beyrath J, Fung CW, et al. Mitochondrial disorders in children: toward development of small-molecule treatment strategies. EMBO Mol Med. 2016;8(4):311–327. doi: 10.15252/emmm.201506131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Li SS, Pan HY, Tan C, et al. Mitochondrial dysfunctions contribute to hypertrophic cardiomyopathy in patient iPSC-derived cardiomyocytes with MT-RNR2 mutation. Stem Cell Reports. 2018;10(3):808–821. doi: 10.1016/j.stemcr.2018.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lorenz C, Lesimple P, Bukowiecki R, et al. Human iPSC-derived neural progenitors are an effective drug discovery model for neurological mtDNA disorders. Cell Stem Cell. 2017;20(5):659–674e9. doi: 10.1016/j.stem.2016.12.013. [DOI] [PubMed] [Google Scholar]
- 28.Lu HE, Yang YP, Chen YT, et al. Generation of patient-specific induced pluripotent stem cells from Leber’s hereditary optic neuropathy. Stem Cell Res. 2018;28:56–60. doi: 10.1016/j.scr.2018.01.029. [DOI] [PubMed] [Google Scholar]
- 29.Ma H, Folmes CDL, Wu J, et al. Metabolic rescue in pluripotent cells from patients with mtDNA disease. Nature. 2015;524(7564):234–238. doi: 10.1038/nature14546. [DOI] [PubMed] [Google Scholar]
- 30.Matsubara M, Kanda H, Imamura H, et al. Analysis of mitochondrial function in human induced pluripotent stem cells from patients with mitochondrial diabetes due to the A3243G mutation. Sci Rep. 2018;8(1):949. doi: 10.1038/s41598-018-19264-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Mattiazzi M, Vijayvergiya C, Gajewski CD, et al. The mtDNA T8993G (NARP) mutation results in an impairment of oxidative phosphorylation that can be improved by antioxidants. Hum Mol Genet. 2004;13(8):869–879. doi: 10.1093/hmg/ddh103. [DOI] [PubMed] [Google Scholar]
- 32.Pfeffer G, Horvath R, Klopstock T, et al. New treatments for mitochondrial disease-no time to drop our standards. Nat Rev Neurol. 2013;9(8):474–481. doi: 10.1038/nrneurol.2013.129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Reardon S. Reports of ‘three-parent babies’ multiply. Nature News [Accessed on Apr. 11, 2019]; 2016. [DOI] [Google Scholar]
- 34.Saada A. The use of individual patient’s fibroblasts in the search for personalized treatment of nuclear encoded OXPHOS diseases. Mol Genet Metab. 2011;104(1-2):39–47. doi: 10.1016/j.ymgme.2011.07.016. [DOI] [PubMed] [Google Scholar]
- 35.Sala L, Bellin M, Mummery CL. Integrating cardiomyocytes from human pluripotent stem cells in safety pharmacology: has the time come? Br J Pharmacol. 2017;174(21):3749–3765. doi: 10.1111/bph.13577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sun JJ, Zhao XX, Qiao LH, et al. Cellular models for mitochondrial DNA-based diseases: lymphoblastoid cell lines and transmitochondrial cybrids. Hereditas (Beijing) 2016;38(7):666–673. doi: 10.16288/j.yczz.16-085. (in Chinese) [DOI] [PubMed] [Google Scholar]
- 37.Wallace DC. A mitochondrial bioenergetic etiology of disease. J Clin Invest. 2013;123(4):1405–1412. doi: 10.1172/JCI61398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wallace DC, Fan WW. The pathophysiology of mitochondrial disease as modeled in the mouse. Genes Dev. 2009;23(15):1714–1736. doi: 10.1101/gad.1784909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wallace DC, Singh G, Lott MT, et al. Mitochondrial DNA mutation associated with Leber’s hereditary optic neuropathy. Science. 1988;242(4884):1427–1430. doi: 10.1126/science.3201231. [DOI] [PubMed] [Google Scholar]
- 40.Wilkins HM, Carl SM, Swerdlow RH. Cytoplasmic hybrid (cybrid) cell lines as a practical model for mitochondriopathies. Redox Biol. 2014;2:619–631. doi: 10.1016/j.redox.2014.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wu J, Ocampo A, Belmonte JCI. Cellular metabolism and induced pluripotency. Cell. 2016;166(6):1371–1385. doi: 10.1016/j.cell.2016.08.008. [DOI] [PubMed] [Google Scholar]
- 42.Wu YR, Wang AG, Chen YT, et al. Bioactivity and gene expression profiles of hiPSC-generated retinal ganglion cells in MT-ND4 mutated Leber’s hereditary optic neuropathy. Exp Cell Res. 2018;363(2):299–309. doi: 10.1016/j.yexcr.2018.01.020. [DOI] [PubMed] [Google Scholar]
- 43.Yamanaka S. Strategies and new developments in the generation of patient-specific pluripotent stem cells. Cell Stem Cell. 2007;1(1):39–49. doi: 10.1016/j.stem.2007.05.012. [DOI] [PubMed] [Google Scholar]
- 44.Yan QF, Guan MX. Nuclear genes and mitochondrial genes associated with mitochondrial diseases. Chin Bull Life Sci. 2008;20(4):497–505. doi: 10.3969/j.issn.1004-0374.2008.04.002. (in Chinese) [DOI] [Google Scholar]