BMB Reports 2023; 56(8): 463-468  https://doi.org/10.5483/BMBRep.2022-0204
Selection of iPSCs without mtDNA deletion for autologous cell therapy in a patient with Pearson syndrome
Yeonmi Lee1,2,#, Jongsuk Han1,#, Sae-Byeok Hwang1,2, Soon-Suk Kang2, Hyeoung-Bin Son1, Chaeyeon Jin1, Jae Eun Kim2, Beom Hee Lee3,* & Eunju Kang1,2,*
1Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, 2Cell Therapy 3 Center, CHA Advanced Research Institute, CHA University, Seongnam 13488, 3Medical Genetics Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
Correspondence to: Beom Hee Lee, Tel: +82-2-3010-5950; Fax: +82-2-3010-6630; E-mail: bhlee@amc.seoul.kr; Eunju Kang, Tel: +82-31-881-7846; Fax: +82-31-881-7958; E-mail: ekang@cha.ac.kr
#These authors contributed equally to this work.
Received: December 9, 2022; Revised: January 2, 2023; Accepted: May 2, 2023; Published online: May 23, 2023.
© Korean Society for Biochemistry and Molecular Biology. All rights reserved.

cc This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
ABSTRACT
Screening for genetic defects in the cells should be examined for clinical application. The Pearson syndrome (PS) patient harbored nuclear mutations in the POLG and SSBP1 genes, which could induce systemic large-scale mitochondrial genome (mtDNA) deletion. We investigated iPSCs with mtDNA deletions in PS patient and whether deletion levels could be maintained during differentiation. The iPSC clones derived from skin fibroblasts (9% deletion) and blood mononuclear cells (24% deletion) were measured for mtDNA deletion levels. Of the 13 skin-derived iPSC clones, only 3 were found to be free of mtDNA deletions, whereas all blood-derived iPSC clones were found to be free of deletions. The iPSC clones with (27%) and without mtDNA deletion (0%) were selected and performed in vitro and in vivo differentiation, such as embryonic body (EB) and teratoma formation. After differentiation, the level of deletion was retained or increased in EBs (24%) or teratoma (45%) from deletion iPSC clone, while, the absence of deletions showed in all EBs and teratomas from deletion-free iPSC clones. These results demonstrated that non-deletion in iPSCs was maintained during in vitro and in vivo differentiation, even in the presence of nuclear mutations, suggesting that deletion-free iPSC clones could be candidates for autologous cell therapy in patients.
Keywords: EB formation, iPSCs, mtDNA deletion, PS patient, Teratoma formation
INTRODUCTION

Mitochondria are essential organelles for eukaryotes that perform vital functions, such as ATP-induced energy production, the synthesis of nucleotides, calcium homeostasis, inflammation, and apoptosis (1, 2). Human mtDNA is double-stranded and encodes 13 proteins, two rRNAs, and 22 tRNAs (3). Defects in mtDNA are associated with inherited and acquired degenerative diseases in humans (4-6). Kearns-Sayre syndrome, chronic progressive external ophthalmoplegia, and Pearson syndrome (PS) are known to be caused by large-scale mtDNA deletions (7-9).

Nuclear genes, such as POLG and SSBP1, are essential in mtDNA replication and proofreading (10, 11). The defect in mtDNA replication could lead to single or multiple mutations in mtDNA and result in mtDNA deletions (12). Co-deficient expression of POLG and SSBP1 genes, caused by nuclear mutations and hypermethylation, could induce large mtDNA deletions (13).

Screening for genetic defects in the cells for clinical application should be involved in selecting intact cells. Mutations in mtDNA could occur randomly in individual cells (14), and the heteroplasmic level of the same mutation could be different in each cell (15, 16). Therefore, cells with a low or absent number of mtDNA mutations could be selected among pooled cells for therapeutic application. Furthermore, previous studies have demonstrated that the heteroplasmic level of mtDNA deletion in iPSCs could be reduced during expanded culture or differentiation, even the original tissue of the cells harbored high heteroplasmy in humans (17, 18). However, the presence of nuclear mutations was not analyzed in those studies.

In our previous study, the PS patient harbored nuclear mutations in POLG and SSBP1 genes, which could induce deficient gene expression. Finally, large mtDNA deletions occurred in the multiple organs of the patient (13). Based on these results, we attempted to examine whether iPSC clones in this patient could maintain the deletion level during cell differentiation. Therefore, we generated single-cell derived iPSC clones in PS patient, selected iPSC clones with or without mtDNA deletion, and analyzed the deletion levels during in vitro and in vivo differentiation to examine the potential for autologous cell therapy (Fig. 1A).

RESULTS

Screening of mtDNA deletions in iPSCs derived from blood and skin fibroblasts in PS patient

In a previous study, we identified large mtDNA deletion in PS patient, 4,395 base pairs (bp) located between the ATPase6 (mt8921) and ND5 (mt13316) genes, in skin tissue with 9% heteroplasmy and 24% with blood (Fig. 1B) (13). We generated iPSC clones from skin fibroblasts (e.g., skin-derived iPSCs, SiPSCs) and peripheral blood mononuclear cells (PBMCs) of blood cells (blood-derived iPSCs, BiPSCs). SiPSCs and BiPSCs showed typical morphology of human iPSCs (19) (Fig. 1C).

Thirteen SiPS clones were randomly selected and analyzed for patient-specific mtDNA deletion (4,395 bp deletion). Three SiPS clones (SiPS1, 9, and 12) did not exhibit mtDNA deletion, whereas the levels in the other 10 SiPS clones ranged from 7 to 53% (Fig. 2A). Furthermore, 10 BiPS clones were analyzed in patient-specific mtDNA deletion, resulting in no mtDNA deletion in all BiPS clones (Fig. 2B). The average heteroplasmy of mtDNA deletion in 13 SiPS clones was 19% in SiPSCs, whereas there was no mtDNA deletion in any of the BiPS clones. (Fig. 2C).

We also analyzed the presence of other mtDNA deletions, including the common deletion of 4,977 bp in BiPS clones (20). Among the 10 BiPS clones, five (BiPS1, 3, 4, 8, and 9) exhibited no deletion, while the other five clones showed 1-7% heteroplasmy (Fig. 2D).

In the expansion culture of iPS clones, the high heteroplasmy of mtDNA deletion, such as 53% heteroplasmy in SiPS2, did not survive until passage 5, while SiPS10 clones with 27% heteroplasmy survived (Supplementary Fig. 1A).

In summary, some BiPS clones had no patient-specific and other deletions, including common mtDNA deletion. These clones could have an advantage for clinical treatment regarding the safety of the deletion-free iPSC clones. For further study, we used the deletion-free BiPS clones (BiPS1, 3, and 8) and SiPS10 with 27% heteroplasmic deletion (red boxes in Fig. 2A, B) to induce differentiation in vitro and in vivo.

Pluripotency of the deletion-free iPSC clones

Before differentiation in vitro and in vivo, the deletion-free iPSC clones (BiPS1, 3, and 8) were selected and analyzed for pluripotent status. Pluripotent stem cell markers, including OCT4, SOX2, and NANOG (21), were analyzed by qRT-PCR, resulting in the gene expression levels being significantly increased in iPSC clones compared to original cells (Supplementary Fig. 2A). Furthermore, OCT4, SSEA4, and TRA-1–60 proteins, as markers of pluripotent stem cells (22), were detected in all BiPSC clones (Supplementary Fig. 2B). Based on the results, pluripotency was confirmed in the deletion-free BiPS clones.

Maintenance of deletion-free during in vitro and in vivo differentiation

The deletion-free iPSC clones, BiPS1, 3, and 8, were performed the differentiation in vitro. BiPS clones were aggregated in 3D culture dishes and differentiated to EB, which were expressed as three germ layer markers (Fig. 3A, B and Supplementary Fig. 3A) (22, 23). PS patient-specific mtDNA deletion was examined in differentiated EBs, and no deletion was found in any of the EBs (Supplementary Fig. 1B). Furthermore, different deletions, including 4,977 bp common deletion, were not detected in all EBs (Supplementary Fig. 1B).

These deletion-free BiPSC clones (BiPS1, 3, and 8) were injected into immunosuppressed mice to analyze the maintenance of the deletion-free status during in vivo differentiation. Subsequently, teratomas were formed, and three germ layers were observed (Fig. 3C, D and Supplementary Fig. 3B) (22, 24). All teratomas derived from BiPS clones were neither detected with PS patient-specific mtDNA deletion nor other deletions (Supplementary Fig. 1C).

The SiPS10 with 27% heteroplasmy was examined for the formation of EB and teratoma. The EB was formed, and three germ layer markers were expressed significantly higher in EB than in the original iPS clone (Fig. 3E, F). However, the relative mRNA expressions of three germ layer markers in EB were lower than those in BiPS clones (Fig. 3B). Nevertheless, the heteroplasmy of EB was 25% similar to the original iPS clone (Supplementary Fig. 1B). Teratomas were formed and observed three germ layers with 45% heteroplasmy, which was increased than original iPS clone (Fig. 3G, H and Supplementary Fig. 1C). These results demonstrated that mtDNA deletion was not regenerated during in vitro and in vivo differentiation in the deletion-free iPS clones, while the deletion level was retained or increased after differentiation when original iPSCs harbored deletion.

DISCUSSION

The PS patient with nuclear mutations in the POLG and SSBP1 genes harbored patient-specific mtDNA deletion (mt8921– 13316 deletion) in skin tissue and blood, and multiple iPSC clones were generated from the skin fibroblasts and PBMCs. The iPSC clones were screened for mtDNA deletion level, and the deletion-free iPSC clones were selected for differentiation. The deletion-free iPSC clones induced EB formation in vitro and teratoma formation in vivo, resulting in non-patient-specific deletion status after differentiation in vitro and in vivo. We further analyzed other mtDNA deletions, including the 4,977 bp common deletion, which was not detected after in vitro and in vivo differentiation. These results demonstrated that mtDNA deletion in iPSCs was not newly generated during differentiation, suggesting that deletion-free iPSC clones could be a valuable source for autologous cell therapy in patients.

Autologous cell therapy in patients has the advantage of eliminating the risk of immune rejection and reducing the need for immunosuppressant treatment (25-27). However, the application of patient-derived cells in autologous cell therapy may be limited in the presence of potential gene mutations that could induce cellular malfunction and systemic disease (28). The PS patient in our study presented with a nuclear mutation in both the POLG and SSBP1 genes, which could affect the induction of large mtDNA deletion in multiple organs (13). Further, mtDNA deletion is associated with mitochondrial dysfunction. The previous reports demonstrated that the fibroblasts and iPSCs of PS patients with mtDNA deletion displayed lower mitochondrial metabolism than cells with no deletion (13, 17). In addition, the SiPS clones with the highest heteroplasmic mtDNA mutations could not survive after freezing and thawing, which could be also associated with mitochondrial dysfunction (17).

Thus, it was imperative to investigate whether mtDNA deletions could re-appeared during differentiation for clinical applications. To address this concern, the deletion-free iPSC clones of the PS patient should be confirmed to not re-appear deletion status following in vitro and in vivo differentiation. Further, the nuclear mutation in PS patients could produce other deletions including the 4,977 bp common deletion, which could be also investigated. Our results demonstrated that mtDNA deletions were not generated during in vitro and in vivo differentiation in iPSCs, suggesting that deletion-free iPSC clones of PS patients could be a safe candidate for cell therapy.

In this study, all BiPS clones had no identified mtDNA deletion even though the original PBMC was 27% heteroplasmy. The previous study reported that heteroplasmy levels of the 4,977 bp common mtDNA deletion were various among the type of blood cells (29). Of them, monocytes and B lymphocytes had fewer deletion levels than T lymphocytes and granulocytes. Probably, our BiPS clones could be established from monocytes (30). Several studies reported that the level of mtDNA deletion in iPSCs could be reduced during in vitro expanded culture or differentiation (17, 18). On the other hand, the deletion levels were increased after teratoma formation in vivo (31).

The present study demonstrated that strategies, such as the screening of mtDNA deletion in iPSCs and the selection of the deletion-free iPSC clones, could be suitable to select the cell sources for clinical application in patients harboring systemic mtDNA deletion induced by nuclear mutation. Further, an opportunity for autologous transplantation for patients with nuclear mutations may be provided by these strategies.

MATERIALS AND METHODS

Ethics

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board of the Asan Medical Center (IRB number 2017-0260). Informed consent was obtained from the subject involved in the study.

PBMC isolation

PBMC isolation was performed as previously described (14). PBMCs were isolated from whole blood using Lymphoprep (STEMCELL Technologies) according to the manufacturer’s protocol. Briefly, the diluted blood with an equal volume of PBS supplemented with 2% fetal bovine serum (FBS) was centrifuged in a density gradient medium at 1,200 x g for 15 min at room temperature. Then, a white buffy coat layer containing PBMC was collected.

Skin fibroblast culture

Isolated fibroblasts from skin tissues were cultured as previously described (13). Fibroblasts were cultured using Dulbecco’s Modified Eagle Medium/Nutrient Mixture F-12 (DMEM/F12 medium, Life Technologies), supplemented with 10% FBS (Life Technologies) and 4 ng/ml of basic fibroblast growth factor (bFGF; Sigma).

Generation of iPSCs

PBMCs or skin fibroblasts were transduced with a CytoTune-iPS Reprogramming Kit (Life Technologies) according to the manufacturer’s protocol. One day before transduction, 1 × 105 cells per well were plated in 6-well culture dishes containing DMEM/F12 with 10% FBS. The cells were transduced with reprogramming vectors (at a multiplicity of infection of 5) diluted with 1 ml of medium per well. One week after transduction, cells were replated onto 100 mm dishes containing feeder layers of mitomycin C-inactivated mouse embryonic fibroblasts. iPSC colonies appeared 3–4 weeks after transduction. Colonies were randomly picked, manually propagated and plated onto 6-well culture dishes coated with 5 μg/ml vitronectin (Life Technologies) in StemMACS medium (Miltenyi Biotec).

Quantitative PCR for the detection of patient-specific mtDNA deletion

Real-time PCR to detect patient-specific mtDNA deletion was performed, as previously described (13). DNA was extracted using the PicoPure DNA Extraction Kit (Qiagen). Regions of the mtDNA, including patient-specific mtDNA deletion (mt8921–13316), were amplified by PCR using the following primer set: F-8822 CTATAAAC-CTAGCCATGGCC and R-13436 GGTATGGTTTTGAGTAGTCC. Wild mtDNA was amplified using PCR with the following primer set: F-16483 GTGAACTG-TATCCGACATCTG and R-100 CAGCGTCTCGCAATGCTATC. PCR reactions were performed under the following conditions: 1 cycle at 95°C for 10 min, then 40 cycles at 95°C for 15 s, 60°C for 30 s and 72°C for 30 s, and finally one cycle at 72°C for 5 min. Heteroplasmy for each deletion was calculated as follows: heteroplasmy of deletion (%) = quantity of deletion mtDNA/quantity of wild mtDNA × 100.

Quantitative PCR for the detection of mtDNA deletions, including common deletion

Real-time PCR to detect mtDNA deletions, including common deletion, was performed as previously described with minor modifications (32). Briefly, real-time PCR was performed using a probe-based, single-tube multiplex qPCR assay on a QuantStudio 6 Flex Real-Time PCR System (Life Technologies). PCR reactions were performed under the following conditions: 95°C for 10 min, 40 cycles of 95°C for 15 s, 55°C for 15 s, and 60°C for 1 min.

Immunocytochemistry

Immunocytochemistry was performed as previously described (22). Fixed iPSCs with 4% formaldehyde were permeabilized with 0.1% Triton X-100 in 0.1% PBS/Tween for 30 min at room temperature and blocked using a blocking solution consisting of 0.1% PBS/Tween with 3% BSA for 30 min at room temperature. After blocking, the cells were incubated with primary antibody (TRA-1-60, SSEA4, and OCT4, Stemgent) solution diluted with 0.1% PBS/Tween (1:100) in 4°C overnight. Cells were washed three times with 0.1% PBS/Tween and incubated for 1 hour at room temperature with a secondary antibody solution (1:1,000). Samples were imaged and captured using a Carl Zeiss inverted microscope.

EB formation

EB formation was performed as described previously, with minor modifications (22). Dissociated iPSCs were seeded onto a StemFIT 3D culture dish (MicroFIT) and cultured in Essential 6 (Gibco) with 100 U/ml penicillin, 100 mg/ml streptomycin (GE Life Science), and 10 μM fasudil (Adooq) for two days. Aggregated cells were transferred to Petri dishes and cultured for six days using the same medium.

Quantitative RT-PCR (RT-qPCR)

Total RNAs were isolated using RNeasy Mini Kits (Qiagen, Valencia, CA, USA), and reverse transcription was performed with cDNA synthesis kits (PCR Biosystems). qRT-PCR was performed according to the previously described (13) with minor modifications using the following gene-specific primers: OCT4 F-GACAGGGGGAGGGGAGGAGCTAGG, R-CTTCCCTCCAACCAGTTGCCCCAAA; SOX2 F-AGCTACAGCATGATGCAGGA, R-GGTCATGGAGTTGTACTGCA; NANOG F-TGAACCTCAGCTACAAACAG, R-TGGTGGTAGGAAGAGTAAAG; PAX6 F-TCCGTTGGAACTGATGGAGT, R-GTTGGTATCCGGGGACTTC; TBXT F-ACCCAGTTCATAGCGGTGAC, R-CCATTGGGAGTACCCAGGTT; SOX17 CAAGGGCGAGTCCCGTAT, R-CGACTTGCCCAGCATCTT; GAPDH F-CCCATGTTCGTCATGGGTGT, R-TGGTCATGAGTCCTTCCACGATA. The expression levels for each gene were measured with qRT-PCR using Power SYBR Green PCR Master Mix (Applied Biosystems). The reaction of qRT-PCR was as follows: one cycle at 95°C for 10 min; followed by 40 cycles at 95°C for 30 s, 56°C for 30 s, and 72°C for 30 s; and a final elongation step at 72°C for 5 min. Gene expression levels were expressed as relative 2−∆∆Ct values.

Teratoma formation

Teratoma formation was performed as previously described (22). iPSCs were injected into the femoral region of 7-week-old NOD-SCID Gamma mice (Jackson Laboratory) using a 1 ml syringe (Korea Vaccine Co.). Mice with tumors were euthanized, and isolated teratomas were sectioned and stained with hematoxylin and eosin. Further, mtDNA deletion in teratomas was also analyzed.

Statistical analysis

Statistical comparisons between the two groups were made using the Student’s two-tailed t-test. Statistical analyses were performed using Prism 8.0.1 software (GraphPad). Data are reported as the mean ± standard error of the mean (SEM).

ACKNOWLEDGEMENTS
This work was supported by the Ministry of Health and Welfare of Korea (HR16C0002(HI16C1559)), and intramural grants from the Asan Institute for Life Sciences, AMC (No. 2019–756).
CONFLICTS OF INTEREST

The authors have no conflicting interests.

FIGURES
Fig. 1. Generation of iPSCs derived from skin and blood in PS patient. (A) The schema for analyzing mtDNA deletion in iPSCs and iPSC-derived EB and teratoma. (B) Heteroplasmic level of patient-specific mtDNA deletion (mt8921-13316) in skin tissue and blood cells. Skin tissue and blood cells harbored 9% and 24% deletion, respectively. This result refers to Lee et al. (13). (C) Morphology of SiPSCs and BiPSCs. SiPSCs and BiPSCs displayed the typical morphology of human iPSCs. Scale bar: 100 nm.
Fig. 2. The level of mtDNA deletions in the iPSC clones of PS patient. (A) Heteroplasmic level of patient-specific deletion in individual SiPSC clones. Three SiPSC clones (SiPS1, 9, and 12) had no deletions. (B) Heteroplasmic level of patient-specific deletion in individual BiPSC clones. All clones showed no deletion. (C) Average heteroplasmy of patient-specific deletion in SiPSCs and BiPSCs. The average heteroplasmy in the SiPSCs was 19%, while there was no mtDNA deletion in all the BiPSCs. (D) The heteroplasmy of other deletions, including the 4,977 bp common deletion in individual BiPSC clones. Five clones had no deletion. SiPSC: skin-derived iPSCs; BiPSCs: blood-derived iPSCs. Red boxes indicate the selected iPS clones for further studies.
Fig. 3. Generation of EB and teratoma from iPSC clones in the PS patient. (A) EB formation is derived from BiPS1 without mtDNA deletion. Scale bar: 100 μm. Undetectable means that mtDNA deletion was not identified. (B) Quantitative RT-PCR analysis of the expression of markers of the three germ layers, PAX6, TBXT, and SOX17, relative to GAPDH in BiPSC clones (BiPS1, 3, 8). Three germ layer markers were significantly higher expressed in EBs than in BiPSC clones. **, and *** mean P < 0.01, and 0.001, respectively. Mean ± SEM. (C) Teratoma formation derived from BiPS1. The black arrow indicates teratoma formation in the femoral region. (D) Three germ layer formations of BiPS1 by teratoma assay. Scale bars: 100 μm. (E) EB formation is derived from a SiPS10 clone with 27% heteroplasmy of mtDNA deletion. Scale bar: 100 μm. 25% deletion means the heteroplasmy of mtDNA deletion in EB. (F) Quantitative RT-PCR analysis of the expression of markers of the three germ layers in SiPS10. Three germ layer markers were significantly higher expressed in EBs than SiPS10. *** mean P < 0.001. Mean ± SEM. (G) Teratoma formation derived from SiPS10. The black arrow indicates teratoma formation. (H) Three germ layer formations of SiPS10 by teratoma assay. Scale bars: 100 μm. SiPSC: skin-derived iPSCs; BiPSCs: blood-derived iPSCs; EB: embryonic body.
REFERENCES
  1. Brand M, Orr A, Perevoshchikova I and Quinlan C (2013) The role of mitochondrial function and cellular bioenergetics in ageing and disease. Br J Dermatol 169, 1-8
    Pubmed KoreaMed CrossRef
  2. Missiroli S, Genovese I, Perrone M, Vezzani B, Vitto VA and Giorgi C (2020) The role of mitochondria in inflammation: from cancer to neurodegenerative disorders. J Clin Med 9, 740
    Pubmed KoreaMed CrossRef
  3. Taanman JW (1999) The mitochondrial genome: structure, transcription, translation and replication. Biochim Biophys Acta 1410, 103-123
    Pubmed CrossRef
  4. Taylor RW and Turnbull DM (2005) Mitochondrial DNA mutations in human disease. Nat Rev Genet 6, 389-402
    Pubmed KoreaMed CrossRef
  5. Ryzhkova AI, Sazonova MA and Sinyov VV et al (2018) Mitochondrial diseases caused by mtDNA mutations: a mini-review. Ther Clin Risk Manag 14, 1933
    Pubmed KoreaMed CrossRef
  6. Greaves LC, Reeve AK, Taylor RW and Turnbull DM (2012) Mitochondrial DNA and disease. J Pathol 226, 274-286
    Pubmed CrossRef
  7. DiMauro S and Schon EA (2003) Mitochondrial respiratory-chain diseases. N Engl J Med 348, 2656-2668
    Pubmed CrossRef
  8. Ferreira M, Evangelista T and Almeida LS et al (2011) Relative frequency of known causes of multiple mtDNA deletions: two novel POLG mutations. Neuromuscul Disord 21, 483-488
    Pubmed CrossRef
  9. Fontana GA and Gahlon HL (2020) Mechanisms of replication and repair in mitochondrial DNA deletion formation. Nucleic Acids Res 48, 11244-11258
    Pubmed KoreaMed CrossRef
  10. Rusecka J, Kaliszewska M, Bartnik E and Tońska K (2018) Nuclear genes involved in mitochondrial diseases caused by instability of mitochondrial DNA. J Appl Genet 59, 43-57
    Pubmed KoreaMed CrossRef
  11. Korhonen JA, Pham XH, Pellegrini M and Falkenberg M (2004) Reconstitution of a minimal mtDNA replisome in vitro. EMBO J 23, 2423-2429
    Pubmed KoreaMed CrossRef
  12. Hudson G, Amati-Bonneau P and Blakely EL et al (2008) Mutation of OPA1 causes dominant optic atrophy with external ophthalmoplegia, ataxia, deafness and multiple mitochondrial DNA deletions: a novel disorder of mtDNA maintenance. Brain 131, 329-337
    Pubmed CrossRef
  13. Lee Y, Kim T and Lee M et al (2021) De novo development of mtDNA deletion due to decreased POLG and SSBP1 expression in humans. Genes 12, 284
    Pubmed KoreaMed CrossRef
  14. Kang E, Wang X and Tippner-Hedges R et al (2016) Age-related accumulation of somatic mitochondrial DNA mutations in adult-derived human iPSCs. Cell Stem Cell 18, 625-636
    Pubmed CrossRef
  15. Ma H, Folmes CD and Wu J et al (2015) Metabolic rescue in pluripotent cells from patients with mtDNA disease. Nature 524, 234-238
    Pubmed CrossRef
  16. Park J, Lee Y and Shin J et al (2019) Mitochondrial genome mutations in mesenchymal stem cells derived from human dental induced pluripotent stem cells. BMB Rep 52, 689
    Pubmed KoreaMed CrossRef
  17. Cherry AB, Gagne KE and Mcloughlin EM et al (2013) Induced pluripotent stem cells with a mitochondrial DNA deletion. Stem Cells 31, 1287-1297
    Pubmed KoreaMed CrossRef
  18. Van Tienen F, Zelissen R and Timmer E et al (2019) Healthy, mtDNA-mutation free mesoangioblasts from mtDNA patients qualify for autologous therapy. Stem Cell Res Ther 10, 1-10
    Pubmed KoreaMed CrossRef
  19. Bang JS, Choi NY, Lee M, Ko K, Park YS and Ko K (2019) Reprogramming of cancer cells into induced pluripotent stem cells questioned. Int J Stem Cells 12, 430-439
    Pubmed KoreaMed CrossRef
  20. Chen T, He J and Shen L et al (2011) The mitochondrial DNA 4,977-bp deletion and its implication in copy number alteration in colorectal cancer. BMC Med Genet 12, 1-9
    Pubmed KoreaMed CrossRef
  21. Lee HJ, Choi NY and Lee SW et al (2019) Alteration of genomic imprinting status of human parthenogenetic induced pluripotent stem cells during neural lineage differentiation. Int J Stem Cells 12, 31-42
    Pubmed KoreaMed CrossRef
  22. So S, Lee Y and Choi J et al (2020) The Rho-associated kinase inhibitor fasudil can replace Y-27632 for use in human pluripotent stem cell research. PLoS One 15, e0233057
    Pubmed KoreaMed CrossRef
  23. Kwon J, Lee N and Jeon I et al (2012) Neuronal differentiation of a human induced pluripotent stem cell line (FS-1) derived from newborn foreskin fibroblasts. Int J Stem Cells 5, 140
    Pubmed KoreaMed CrossRef
  24. Malhotra N (2016) Induced pluripotent stem (iPS) cells in dentistry: a review. Int J Stem Cells 9, 176-185
    Pubmed KoreaMed CrossRef
  25. Madrid M, Sumen C, Aivio S and Saklayen N (2021) Autologous induced pluripotent stem cell-based cell therapies: promise, progress, and challenges. Curr Protoc 1, e88
    Pubmed CrossRef
  26. Abi Chahine N, Wehbe T, Rashed J, Hilal R and Elias N (2016) Autologous bone marrow derived stem cells for the treatment of multiple sclerosis. Int J Stem Cells 9, 207-212
    Pubmed KoreaMed CrossRef
  27. Kazmi B, Inglefield CJ and Lewis MP (2009) Autologous cell therapy: current treatments and future prospects. Wounds 21, 234-242
  28. Li C, Zhao H, Cheng L and Wang B (2021) Allogeneic vs. autologous mesenchymal stem/stromal cells in their medication practice. Cell Biosci 11, 1-21
    Pubmed KoreaMed CrossRef
  29. Mohamed SA, Wesch D and Blumenthal A (2004) Detection of the 4977 bp deletion of mitochondrial DNA in different human blood cells. Exp Gerontol 39, 181-188
    Pubmed CrossRef
  30. Isogai S, Yamamoto N and Hiramatsu N et al (2018) Preparation of induced pluripotent stem cells using human peripheral blood monocytes. Cell Reprogram 20, 347-355
    Pubmed KoreaMed CrossRef
  31. Russell OM, Fruh I and Rai PK et al (2018) Preferential amplification of a human mitochondrial DNA deletion in vitro and in vivo. Sci Rep 8, 1-10
    Pubmed KoreaMed CrossRef
  32. Phillips NR, Sprouse ML and Roby RK (2014) Simultaneous quantification of mitochondrial DNA copy number and deletion ratio: a multiplex real-time PCR assay. Sci Rep 4, 1-7
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles

Author ORCID Information

Funding Information

Services
Social Network Service

e-submission

Archives