Abstract
Human pluripotent stem cells (hPSCs) are sensitive to DNA damage and undergo rapid apoptosis compared to their differentiated progeny cells. Here, we explore the underlying mechanism(s) for the increased apoptotic sensitivity of hPSCs that helps to determine pluripotent stem cell fate. Apoptosis was induced by exposure to actinomycin D, etoposide, or tunicamycin, with each agent triggering a distinct apoptotic pathway. We show that hPSCs are more sensitive to all three types of apoptosis induction than are lineage non-specific, retinoic acid (RA) differentiated hPSCs. Also, Bax activation and pro-apoptotic mitochondrial intermembrane space protein release, which are required to initiate the mitochondrial mediated apoptosis pathway, is more rapid in hPSCs than RA-differentiated hPSCs. Surprisingly, Bak and not Bax is essential for actinomycin D induced apoptosis in human embryonic stem cells (hESCs). Finally, P53 is degraded rapidly in an ubiquitin proteasome-dependent pathway in hPSCs at steady-state, but quickly accumulates and induces apoptosis when Mdm2 function is impaired. Rapid degradation of P53 ensures the survival of healthy hPSCs, but avails these cells for immediate apoptosis upon cellular damage by P53 stabilization. Altogether, we provide an underlying, interconnected molecular mechanism that primes hPSCs for quick clearance by apoptosis to eliminate hPSCs with unrepaired genome alterations and preserves organismal genomic integrity during the early critical stages of human embryonic development.
Keywords: mitochondria, Bak/Bax, pluripotency, differentiation, cell fate
Graphical abstract

Introduction
Human pluripotent stem cells (hPSCs) have both the ability to self-renew and the ability to differentiate into all cell types in the human body [1, 2]. Genomic integrity of hPSCs in utero and in tissue culture is essential for cell lineage fidelity during further development and differentiation and therefore a rapid response to cellular damage is required. Human embryonic stem cells (hESCs) have been shown to undergo rapid apoptosis in preference to repair in response to DNA damage when compared to differentiated cells [3–8]. The mechanism(s), however, that potentiates the rapid response of DNA damaged hESCs toward apoptosis instead of repair is still not fully understood. The tumor suppressor protein P53 is a known regulator of apoptosis in response to DNA damage in hESCs [3–5, 7, 8]. In somatic cells, following DNA damage P53 contributes to apoptosis induction and execution through transcriptional activation of pro-apoptotic genes, through sequestration of anti-apoptotic proteins, or by interacting with and positively influencing pro-apoptotic proteins, Bax and Bak, in the cytosol [9–15]. Knockdown of P53 in hESCs eliminates the apoptotic response to DNA damage. hESCs expressing P53 lacking a nuclear localization signal can activate apoptosis in response to DNA damage, indicating that cytosol localized P53, in addition to nuclear P53, contributes to apoptosis in hESCs [5]. Inhibition of cyclin-dependent kinase 1 (CDK1) can selectively induce the DNA damage response and P53-dependent apoptosis in hESCs, in contrast to only causing transient cell cycle arrest during DNA repair in differentiated cells [16]. Expression of P53 target genes is rapidly induced in response to DNA damage in ESCs, but this rapid response is also seen in differentiated cells [5]. Whereas P53 plays a large and potentially distinct role in the DNA damage responses of hESCs and somatic cells, no actual differences in the apoptosis inducing behavior of P53 or its regulation have yet been identified between hPSCs and differentiated cells. Instead, what has been reported is that the mitochondria in hPSCs are primed for apoptosis due to a difference in the balance between pro-apoptotic and anti-apoptotic proteins, leading to a higher sensitivity and lower apoptotic threshold for hESCs compared to differentiated cells [5, 17].
Activation of apoptosis by cell intrinsic stimuli, such as DNA damage, occurs through mitochondrial outer membrane permeabilization (MOMP), which requires the activation of pro-apoptotic BCL-2 family member proteins Bax or Bak [11, 18, 19]. Some hESC lines show constitutively activated pro-apoptotic Bax localized to the Golgi apparatus during S phase, where it is unable to activate apoptosis until DNA damage induces its translocation to the mitochondria to induce MOMP [20]. Knockdown of Bax in hESCs decreases apoptosis in response to DNA damage. Additionally, P53 is required for the translocation of Bax from the Golgi apparatus to the mitochondria with DNA damage in hESCs [20]. In other cell types and hESC lines, Bax is localized to the cytosol in an inactive state. Once activated by BH3-only proteins, Bax undergoes a conformational change and insertion into the mitochondrial outer membrane [18, 21]. Nonetheless, since activated Bax is not detectable in the Golgi apparatus of most hESC lines, this potential sensitizing mechanism cannot be exclusively responsible for the rapid activation of apoptosis in response to DNA damage in these hESC lines [20].
Here, we further investigate apoptotic mechanisms in hPSCs and discover that differential regulation of P53 stability sensitizes hPSCs to apoptosis. Initially, we evaluated the similarities and differences in the apoptotic machinery between hPSCs and differentiated cells to elucidate the pathways underlying the rapid activation of apoptosis in hPSCs. We discovered that hPSCs activate apoptosis rapidly not only in response to DNA damage, but also in response to transcriptional inhibition and the induction of endoplasmic reticulum (ER) stress. In addition, we identified important roles for the mitochondrial fission protein Drp1 and pro-apoptotic BCL-2 family member protein Bak in hESC apoptotic hypersensitivity. Finally we report that P53 is rapidly degraded at steady-state in hPSCs, but inhibition of ubiquitin proteasome-dependent degradation by Mdm2 causes prompt stabilization of P53 and the induction of apoptosis in hESCs.
Results
hESCs are hypersensitive to diverse mitochondria mediated apoptotic stimuli
In addition to rapid apoptosis in response to DNA damage, hPSCs also undergo mitochondria-dependent apoptosis upon dissociation into single cells, which can be suppressed by Rho-dependent protein kinase (ROCK) inhibitors [22, 23]. Therefore, we considered whether hPSCs are more generally hypersensitive to mitochondria-mediated apoptosis by evaluating whether hPSCs are more sensitive to multiple intrinsic apoptotic stimuli than their differentiated counterparts. We used retinoic acid (RA) induced differentiation of hPSCs as a differentiated comparative cell derivative of hPSCs [24–26]. RA is an important morphogen during development and is commonly used for neuronal differentiation [27–29]. RA treatment of monolayer culture of hESCs causes preferential differentiation into ectodermal and mesodermal lineages [30–32]. Undifferentiated hPSCs and RA-differentiated cells were evaluated for apoptosis by incubation with various apoptosis stimulating agents. Exposure of H9 hESCs to RA reduced the expression of Nanog and Oct4, key transcription factors for maintaining hPSC self-renewal, to undetectable levels by days 3 and 5, respectively (Fig. 1a). Actinomycin D, a potent inducer of apoptosis through transcriptional repression, was used to induce apoptosis in H9 and H1 hESCs as well as a human induced pluripotent stem cell (hiPSC) line, HIPS2, on days 0, 1, 3, and 5 of RA induced differentiation (Fig. 1b and Fig. S1b,c). The fragmentation of cells into apoptotic bodies is carried out by a family of intracellular cysteine-dependent aspartate-directed proteases (caspases). Upon activation of mitochondrial mediated apoptosis and MOMP, procaspase-3 is cleaved and in turn cleaved caspase-3 executes apoptosis through digestion of other intracellular proteins including the DNA repair protein poly(ADP-ribose) polymerase (PARP) [33–35]. Therefore, apoptosis was evaluated by western blot analysis of cleaved caspase-3 and cleaved PARP. Undifferentiated hPSCs showed a robust time dependent cleavage of caspase-3 and PARP after actinomycin D exposure (Fig. 1b and Fig. S1b,c,). Although uncleaved caspase-3 showed only slightly decrease during differentiation, hPSCs that were differentiated for 5 days, however, had only low levels of caspase-3 and PARP cleavage after actinomycin D treatment (Fig. 1b and S1a,b,c). Next, DNA damage was induced in undifferentiated and differentiated hPSCs by treatment with etoposide, which causes DNA double-strand breaks through the inhibition of DNA topoisomerase II [36]. Undifferentiated H9 and HIPS2 cells again show robust, time-dependent cleavage of caspase-3 and PARP, but hPSCs differentiated for 5 days have no detectable cleavage of caspase-3 or PARP (Fig. 1c and Fig. S1d).
Fig. 1.

Rapid apoptosis induction in hESCs. (a) Immunoblot of Oct4 and Nanog in H9 hESCs differentiated with 1 μM RA for 0, 1, 3, and 5 days. TOMM40 was included as a loading control. (b–d) H9 hESCs differentiated with 1 μM RA for 0, 1, 3, and 5 days were treated with multiple intrinsic apoptosis inducers: 1 μM actinomycin D (b), 1μM etoposide (c) and 1 μg/ml tunicamycin (d) for the indicated times. Whole cell extracts were analyzed by SDS-PAGE and immunoblotted with antibodies for cleaved caspase-3 (c-caspase-3) and cleaved PARP (c-PARP). TOMM40 was included as a loading control. (e–f) H1 hESCs were treated with actinomycin D in the presence of pan-caspase inhibitor Q-VD-OPH. Cells were fixed and IMS protein release of cytochrome c (e) and Smac (f) were detected by immunofluorescence confocal microscopy. Data was obtained from 100 cells in each of three independent experiments. Error bars represent standard deviation (SD). (g) Mitochondria-targeted Bax was detected by immunofluorescence confocal microscopy using anti-Bax and anti-Mortalin (mitochondrial chaperone) antibodies. Cells were treated as in (e–f) and Bax and TOMM20 co-localization were counted in each of three independent experiments. Error bars represent SD.
Apoptosis induced by ER stress is also mediated though the mitochondria [37–40]. To determine whether hPSCs are sensitive to ER stress-induced apoptosis, undifferentiated and differentiated hESCs were treated with tunicamycin, which induces ER stress through the inhibition of N-linked glycosylation [41]. Whereas high levels of cleaved caspase-3 and cleaved PARP were detected in undifferentiated hESCs, there was an absence of these cleaved apoptotic signature proteins in cells differentiated for 5 days (Fig. 1d).
Because cleavage of caspase-3 and PARP is a late event in apoptosis, we next evaluated the release of pro-apoptotic intermembrane space (IMS) proteins cytochrome c and Smac from the mitochondrial IMS upon MOMP [42–44]. Cytochrome c and Smac release was detected by imaging colocalization with the mitochondrial marker TOMM20 using confocal immunofluorescence microscopy following timed actinomycin D exposure. Cytochrome c and Smac were released into the cytosol from a greater percentage of undifferentiated hESCs than from differentiated cells (Fig. 1e,f and Fig. S2a,b). Bax recruitment to the mitochondria can lead to MOMP [11, 18], and precedes the release of IMS proteins from the mitochondria into the cytosol and caspase activation. Therefore, Bax localization to the mitochondria was also measured by immunofluorescence microscopy. Timed actinomycin D exposure induced the mitochondrial localization of Bax in a greater percentage of undifferentiated hESCs than in differentiated cells (Fig. 1g and Fig. S3a,b).
Mitochondrial elongation reduces the sensitivity of hPSCs to apoptosis
Mitochondria are fused, interlacing network structures that extend peripherally in the cytosol of most differentiated cell types, in contrast to the fragmented, punctate, perinuclear appearance of mitochondria in hPSCs [24, 45]. Mitochondrial fragmentation occurs during apoptosis [46–48], and therefore cells with a more fragmented network structure may be more sensitive to apoptotic stimuli. The dynamin-related GTPase Drp1 mediates fission of the mitochondrial network and localizes to the mitochondrial outer membrane at sites of fission [46, 49, 50]. In somatic cells, Drp1 depletion retards the usual rate of cytochrome c release and caspase activation during apoptosis [46]. To determine whether the fragmented mitochondrial morphology seen in hESCs influences their hypersensitivity to intrinsic apoptotic stimuli, mitochondrial fission was inhibited by shRNA knockdown of Drp1 (Fig. 2a). Immunofluorescent confocal imaging of TOMM20, a mitochondrial outer membrane translocon protein, in Drp1 knockdown hESCs revealed mitochondria that were more elongated when compared to scramble control hESCs (Fig. 2b). Therefore, Drp1 depletion induces mitochondrial network fusion in hESCs. To determine whether an elongated mitochondrial morphology decreases the apoptotic hypersensitivity of hESCs, apoptosis was induced by actinomycin D treatment in Drp1 knockdown and scramble control hESCs. Timed actinomycin D exposure resulted in less cleavage of caspase-3 and PARP in Drp1 knockdown hESCs when compared to scramble control (Fig. 2c). Thus, the fragmented mitochondrial structure in hESCs helps to promote their sensitivity to apoptosis induction.
Fig. 2.

Knockdown of Drp1 induces mitochondria elongation and suppressed apoptosis sensitivity in hESCs. (a) Knockdown efficiency of Drp1 in H9 hESCs was confirmed by immunoblot. (b) Mitochondrial morphology in H9 hESCs transduced with Drp1 or control shRNAs were analyzed by immunofluorescence confocal microscopy using anti-TOMM20 antibody. (c) H9 hESCs transduced with Drp1 or scramble shRNAs were exposed to actinomycin D for the indicated times. Whole cell extracts were analyzed by SDS-PAGE and immunoblotted with antibodies for cleaved caspase-3 (c-caspase-3) and cleaved PARP (c-PARP). TOMM0 was included as a loading control.
Bak has a crucial role in hPSC hypersensitivity to apoptosis
Activation of BCL-2 family member proteins Bak and Bax causes their oligomerization and initiates MOMP, which releases IMS proteins into the cytosol and activates the caspase cascade [19, 51, 52]. Bak and Bax have overlapping roles during development. Bak-deficient mice develop normally, and Bax-deficient mice have limited abnormalities. Double knockouts for both Bax and Bak, however, die perinatally with multiple developmental defects related to defective apoptosis induction, such as persistent interdigital webs and excessive cells in the central nervous and hematopoietic systems [53]. In addition, Bax has been shown to be important for the apoptotic response to etoposide in hESCs, but the role of Bax and Bak have not been analyzed in response to other apoptotic stimuli [20]. Therefore because Bax or Bak are essential for mitochondrial mediated apoptosis and their contribution to apoptosis in hESCs is controversial, we next investigated whether either protein had a more dominant role in mediating hESC hypersensitivity to apoptosis. Bak, Bax and Bcl-XL protein levels are not markedly changed during RA-induced differentiation (Fig. 3a). To further assess the role of Bax and Bak in hESC sensitivity to apoptosis, the expression of Bax and Bak was separately knocked down with shRNA in H1 and H9 hESCs (Fig. 3b and Fig. S4a). The expression of pluripotency biomarker proteins SSEA4, Lin28, Nanog, and Oct4 was not affected by knockdown of either Bax or Bak (Fig. 3c). Activation of apoptosis by exposure to actinomycin D was almost completely inhibited in Bak knockdown cells (Fig. 3d and Fig. S4b,c). Therefore, Bak is an important component contributing to the hypersensitivity of hESCs to mitochondrial mediated apoptosis. In contrast, knockdown of Bax only slightly decreased the induction of apoptosis in hESCs (Fig. 3d,e and Fig. S4b,c). With etoposide treatment, however, Bax played a more crucial role as previously reported (Fig. S4d) [20]. Hence while Bax has an important role in hESC response to etoposide, Bak is important for the response to actinomycin D. This suggests that both Bax and Bak play important roles in hPSC apoptosis.
Fig. 3.

Bak has a dominant role in actinomycin D induced apoptosis of hESCs. (a) Immunoblots of BCL-2 family proteins in H1 hESCs differentiated with 1 μM RA for 0, 3, and 5 days. (b) Immunoblots showing Bak and Bax protein levels in scramble, Bak, and Bax knockdown H1 hESCs. Tubulin was included as a loading control. (c) Immunofluorescent detection of pluripotency biomarker proteins from scramble, Bak, and Bax knockdown H1 hESCs. Scale Bar is 200 μm. (d) Scramble, Bak, and Bax knockdown H1 hESCs were exposed to actinomycin D for the indicated times. Whole cell extracts were analyzed by SDS-PAGE and immunoblotted with antibodies for cleaved caspase-3 (c-caspase-3) and cleaved PARP (c-PARP). TOMM40 was included as a loading control. (e) H1 hESCs were exposed to actinomycin D in the presence of the pan-caspase inhibitor Q-VD-OPH for the indicated times. Cells were fixed and analyzed by immunofluorescence confocal microscopy using antibodies against cytochrome c. Data were obtained from three independent experiments. Error bars represent SD.
P53 turnover is rapid in hPSCs
Tumor suppressor P53 is a central regulator of growth arrest, senescence, and apoptosis in response to a broad array of cellular damage [10, 13, 54–56]. Regulation of P53 is mainly accomplished through ubiquitin-dependent proteasome degradation [57]. Surprisingly, while P53 has been carefully studied in hPSCs, differences in P53 protein stability have not been examined as the potential cause of hPSC hypersensitivity. Mdm2 is the primary E3 ubiquitin ligase that negatively regulates P53 by degradation or by masking the transactivation domain of P53 [58–60]. Mdm2, in turn, is degraded following DNA damage which stabilizes P53 [61, 62].
In H1 hESCs, Mdm2 protein levels decrease after RA-induced differentiation, whereas P53 protein levels do not change (Fig. 4a). To determine whether the decrease in Mdm2 was regulated transcriptionally, qRT-PCR was performed to measure Mdm2 mRNA levels. No change in Mdm2 expression was detected between hESCs and RA-differentiated cells (Fig. S5a). P53 expression was 2-fold higher in H9 hESCs than RA-differentiated H9s, but no change was seen in H1 cells (Fig. S5b). To determine whether the p53 protein that we detected was wild type p53, we compared the molecular weight of p53 in H1 hESC cell lysate with recombinant p53 and observed equal molecular weights (Fig. S5c).
Fig. 4.

P53 turnover is rapid in hESCs. (a) Immunoblot of Mdm2 and P53 in H1 hESCs and day 5 RA-induced H1 differentiated cells. (b) Immunoblot of P53, cleaved PARP (c-PARP), and cleaved caspase-3 (c-caspse-3) in H1 hESCss and RA-differentiated H1 cells after 1 μM etoposide or 4 μM nutlin-3a for the indicated times. (c,e) P53 protein stability analyzed by immunoblot in undifferentiated H1 hESCs (c) and H1 cells differentiated with RA for 5 days (e) were examined by cycloheximide (CHX) exposure for in the presence of DMSO or MG132. (d,f) Band intensities corresponding to P53 protein levels in undifferentiated H1 hESCs (d) and H1 cells differentiated with RA for 5 days (f) were quantified by Image J and shown as relative amounts. Protein levels at 0 hours was set as 1.0.
To determine whether p53 protein stability is differentially regulated in hESCs and their differentiated counterparts, cells were treated with an Mdm2 inhibitor, nutlin-3a [63]. Timed exposure of hESCs to nutlin-3a rapidly stabilizes high levels of P53 (Fig. 4b). Treatment of differentiated cells with nutlin-3a, however, stabilizes only a small amount of P53 (Fig. 4b). Furthermore, treatment of hPSCs with nutlin-3a activates apoptosis, but apoptosis is not detectible after nutlin-3a treatment of RA differentiated cells (Fig. 4b). Therefore p53 is rapidly degraded in hPSCs through ubiquitination by Mdm2. To further probe the difference in P53 stability, H1, H9, and HIPS2 hPSCs were treated with the protein translation inhibitor cycloheximide (CHX). After 2 hours of CHX exposure, P53 levels were diminished (Fig. 4c,d and Fig. S5d,f). In contrast, P53 slowly decreases after CHX exposure in RA-differentiated hPSCs (Fig. 4e,f and Fig. S5e,g). Additional treatment with a proteasome inhibitor MG132 slowed the decrease in levels of P53 in both hPSCs and RA-differentiated hPSCs, indicating that P53 degradation is ubiquitin proteasome-dependent (Fig. 4c,f, Fig. S5d–g). This indicates that P53 undergoes Mdm2 initiated ubiquitin-dependent proteasome degradation at a higher rate in hPSCs than hPSCs induced to undergo differentiation.
Discussion
hESCs and hiPSCs have immense potential in regenerative medicine because of their ability to self-renew and to differentiate into every cell type in the human body. Therefore, maintenance of genomic integrity is essential to avoid defects that may cause aberrant development or cancer. Apoptosis is one available mechanism for eliminating damaged cells following DNA or other forms of cellular damage [64]. Recent studies have shown that hESCs undergo rapid apoptosis; however, the mechanism(s) underlying this enhanced apoptosis sensitivity still remains unclear [3–5, 20].
Here, we show that hPSCs undergo rapid mitochondria-dependent apoptosis in response to three independent apoptotic stimuli. Furthermore we investigated each step of the cell death cascade and found hPSCs to be hypersensitive all stages. We further showed the importance of mitochondria in rapid apoptosis by inducing elongated mitochondria by Drp1 depletion. The inhibition of rapid apoptosis in hESCs by Drp1 depletion emphasizes the involvement of mitochondria in this unique event. We further examined the contribution of Bax and Bak, two main pro-apoptotic members of the BCL-2 protein family that directly mediate the mitochondrial apoptosis pathway [39]. Although a recent study showed that etoposide-induced hESC apoptosis is Bax-dependent and Bak-independent, our data showed a crucial role for Bak in actinomycin D induced apoptosis, suggesting that both Bax and Bak have an important role in hESC apoptosis that is regulated by many apoptotic factors in a complex manner.
Mdm2, a negative regulator of P53, is inactivated following DNA damage, leading to P53 stabilization, which can initiate apoptosis [61, 62]. We discovered that a rapid accumulation of P53 caused by Mdm2 inhibitor nutlin-3a is sufficient to induce apoptosis in hESCs, but only has a modest effect in differentiated cells. Yet, the P53 protein level in steady-state cells is similar in both hESCs and RA-differentiated cells. Therefore, hESCs rapidly turn over p53 in an ubiquitin proteasome-dependent manner that correlates with high expression of Mdm2. A previous study reported that P53 exhibits no difference in apoptosis-inducing behavior in both differentiated and undifferentiated hESCs and, therefore, the rapid apoptosis in hESCs is due to their highly mitochondrial primed state [5]. However, because P53 interacts with mitochondrial proteins [7], P53 could be a key factor that primes the mitochondria for rapid induction of apoptosis in hESCs. Given that suppression of P53 increases the efficiency of hiPSC generation up to 10–20 fold [65], inactivation of P53 by Mdm2 in healthy hPSCs may be crucial for their survival and maintenance. Our study identifies a unique regulation of P53 in hESCs which prepares them to induce rapid apoptosis to maintain genome integrity, while assuring survival at steady state.
Materials and Methods
Cell culture
H1, H9 [1], and HIPS2 [66] hPSCs were grown on matrigel and fed daily with mTeSR-1 medium (Stemcell Technologies), maintained at 37°C and 5% CO2. The medium was changed daily and passaged with Dispase (Stemcell Technologies) before confluent. Differentiation of hPSCs was induced by 1μM RA treatment, with a fresh medium change every other day.
Antibodies and reagents
Antibodies were used against Oct4 (Santa Cruz), Nanog (Milliprore), SSEA4 (Millipore), Lin28 (Cell Signaling), Tubulin (Sigma), cytochrome c (BD Biosciences), Smac/Diablo (BD Biosciences), Bax-NT (Milliprore), Bak-NT (Milliprore), Drp1 (BD Biosciences), Mdm2 (Santa Cruz), TOMM20 (Santa Cruz), Bcl-XL (Cell Signaling), c-caspase3 (Cell Signaling), full length caspase-3 (Cell Signaling), c-PARP (Cell Signaling), and P53 (Cell Signaling) proteins. A polyclonal antibody against recombinant TOMM40 (Pacific Immunology) was described previously [67]. Cycloheximide, MG132, actinomycin D and etoposide were obtained from Sigma, Q-VD-OPH from SM Biochemicals, recombinant human p53 protein from BD Pharmingen, and nutlin-3a from Cayman Chemical.
Western blotting
Cultured cells were harvested into SDS buffer [40 mM tris-HCl (pH6.8), 3% glycerol, 1% SDS]. Protein concentrations were quantified using the BCA assay (Pierce). Immunodetection was performed with ECL reagent (GE Healthcare). For protein turn over analyses, cells were incubated with 50 μg/ml cycloheximide with or without 10 μM MG132 for the times indicated in each figure legend.
Lentiviral-mediated shRNA knockdown in hESCs
To generate hESCs that are knocked down for Drp1, Bak, and Bax, shRNAs that specifically target Drp1 (TRCN0000001097), Bak (TRCN0000033466) and Bax (TRCN0000033472) were obtained from Sigma. On Day1, hESC colonies on matrigel were transduced with 25 μl of 1×107 TU/ml of viral particles and 7 μg/ml polybrene for 8h. On days 2 and 3, 75 μl of 1×107 TU/ml of viral particles was also added. Selection with 1 μg/ml puromycin was started on days 5–7 post-infection. Cell culture medium supplemented with puromycin was replaced daily.
Immunofluorescence confocal microscopy
Cells were grown on matrigel coated chambered cover glass (Thermo Scientific). For cytochrome c/Smac release assays, and Bax targeting analysis, cells were exposed to apoptosis stimuli with 25 μM in the presence of the broad caspase inhibitor, Q-VD-OPH. Cells were fixed for 15 min at room temperature with pre-warmed 4% PFA and then peremeabilized for 5 min with 1% Triton X-100 in PBS followed by incubation with primary antibodies. After incubation with Alexa Fluor-conjugated secondary antibodies (Invitrogen), images were acquired with either LSM 5 PASCAL or LSM 780 confocal microscopes (Carl Zeiss).
qRT-PCR
Cultured cells were harvested with TRIzol reagent (Life Technologies). RNA was extracted according to manufacturer instructions. RNA was converted to cDNA with iScript (Bio-Rad). qRT-PCR was performed on a LightCycler480 (Roche) using SYBR green (Roche). Primers used to detect P53 were agg cct tgg aac tca agg at and ccc ttt ttg gac ttc agg tg. Primers used to detect Mdm2 were gac tcc aag cgc gaa aac and ggt ggt tac agc acc atc agt. Primers used to detect Eifb2 were ttg agg cga tta atg agc tg and tgg agt gaa tgt gct cca ga.
Supplementary Material
Highlights.
hPSCs are hypersensitive to diverse apoptotic stimuli
Fragmented mitochondrial morphology in hESCs sensitizes them to apoptosis
Apoptosis induced by transcriptional repression is mainly mediated by Bak in hPSCs
Mdm2 mediates rapid P53 degradation in hPSCs, but not in differentiated cells
Acknowledgments
K. S. is supported by California Institute for Regenerative Medicine (CIRM) Basic Biology-I Award RB1-01397. T. T. is supported by a Ruth L. Kirschstein National Research Service Award GM007185. M. A. T. and C. M. K. are supported by California Institute for Regenerative Medicine (CIRM) Basic Biology 1 Award RB1-01397 and Tools & Technologies 3 Award RT3-07678, by UC Discovery/NantWorks Biotechnology Awards Bio07-10663 and 178517, and by National Institutes of Health (NIH) Awards GM073981, GM061721, P01GM081621, CA156674, CA90571, GM114188, and CA185189.
Abbreviations
- CHX
cycloheximide
- hESC
human embryonic stem cell
- hiPSC
human induced pluripotent stem cell
- hPSC
human pluripotent stem cell
- IMS
intermembrane space
- MOMP
mitochondrial outer membrane permeabilization
- PARP
poly(ADP-ribose) polymerase
- RA
retinoic acid
- ROCK
rho-dependent protein kinase
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of interest
The authors declare no conflicts of interest.
References
- 1.Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, et al. Embryonic Stem Cell Lines Derived from Human Blastocysts. Science. 1998;282:1145–7. doi: 10.1126/science.282.5391.1145. [DOI] [PubMed] [Google Scholar]
- 2.Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors. Cell. 2007;131:861–72. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
- 3.Wilson KD, Sun N, Huang M, Zhang WY, Lee AS, Li Z, et al. Effects of Ionizing Radiation on Self-Renewal and Pluripotency of Human Embryonic Stem Cells. Cancer Research. 2010;70:5539–48. doi: 10.1158/0008-5472.CAN-09-4238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Momcilovic O, Knobloch L, Fornsaglio J, Varum S, Easley C, Schatten G. DNA Damage Responses in Human Induced Pluripotent Stem Cells and Embryonic Stem Cells. PLoS ONE. 2010;5:e13410. doi: 10.1371/journal.pone.0013410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Liu Julia C, Guan X, Ryan Jeremy A, Rivera Ana G, Mock C, Agarwal V, et al. High Mitochondrial Priming Sensitizes hESCs to DNA-Damage-Induced Apoptosis. Cell Stem Cell. 2013;13:483–91. doi: 10.1016/j.stem.2013.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Filion TM, Qiao M, Ghule PN, Mandeville M, van Wijnen AJ, Stein JL, et al. Survival responses of human embryonic stem cells to DNA damage. Journal of Cellular Physiology. 2009;220:586–92. doi: 10.1002/jcp.21735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Qin H, Yu T, Qing T, Liu Y, Zhao Y, Cai J, et al. Regulation of Apoptosis and Differentiation by p53 in Human Embryonic Stem Cells. Journal of Biological Chemistry. 2007;282:5842–52. doi: 10.1074/jbc.M610464200. [DOI] [PubMed] [Google Scholar]
- 8.Grandela C, Pera MF, Wolvetang EJ. p53 is required for etoposide-induced apoptosis of human embryonic stem cells. Stem Cell Research. 2008;1:116–28. doi: 10.1016/j.scr.2007.10.003. [DOI] [PubMed] [Google Scholar]
- 9.Leu JIJ, Dumont P, Hafey M, Murphy ME, George DL. Mitochondrial p53 activates Bak and causes disruption of a Bak-Mcl1 complex. Nat Cell Biol. 2004;6:443–50. doi: 10.1038/ncb1123. [DOI] [PubMed] [Google Scholar]
- 10.Mihara M, Erster S, Zaika A, Petrenko O, Chittenden T, Pancoska P, et al. p53 Has a Direct Apoptogenic Role at the Mitochondria. Molecular Cell. 2003;11:577–90. doi: 10.1016/s1097-2765(03)00050-9. [DOI] [PubMed] [Google Scholar]
- 11.Chipuk JE, Kuwana T, Bouchier-Hayes L, Droin NM, Newmeyer DD, Schuler M, et al. Direct Activation of Bax by p53 Mediates Mitochondrial Membrane Permeabilization and Apoptosis. Science. 2004;303:1010–4. doi: 10.1126/science.1092734. [DOI] [PubMed] [Google Scholar]
- 12.Pietsch EC, Perchiniak E, Canutescu AA, Wang G, Dunbrack RL, Murphy ME. Oligomerization of BAK by p53 Utilizes Conserved Residues of the p53 DNA Binding Domain. Journal of Biological Chemistry. 2008;283:21294–304. doi: 10.1074/jbc.M710539200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Vousden KH, Lu X. Live or let die: the cell’s response to p53. Nat Rev Cancer. 2002;2:594–604. doi: 10.1038/nrc864. [DOI] [PubMed] [Google Scholar]
- 14.Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B. A model for p53-induced apoptosis. Nature. 1997;389:300–5. doi: 10.1038/38525. [DOI] [PubMed] [Google Scholar]
- 15.Toshiyuki M, Reed JC. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell. 1995;80:293–9. doi: 10.1016/0092-8674(95)90412-3. [DOI] [PubMed] [Google Scholar]
- 16.Huskey Noelle E, Guo T, Evason Kimberley J, Momcilovic O, Pardo D, Creasman Katelyn J, et al. CDK1 Inhibition Targets the p53-NOXA-MCL1 Axis, Selectively Kills Embryonic Stem Cells, and Prevents Teratoma Formation. Stem Cell Reports. 2015 doi: 10.1016/j.stemcr.2015.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ardehali R, Inlay MA, Ali SR, Tang C, Drukker M, Weissman IL. Overexpression of BCL2 enhances survival of human embryonic stem cells during stress and obviates the requirement for serum factors. Proceedings of the National Academy of Sciences. 2011;108:3282–7. doi: 10.1073/pnas.1019047108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kuwana T, Bouchier-Hayes L, Chipuk JE, Bonzon C, Sullivan BA, Green DR, et al. BH3 Domains of BH3-Only Proteins Differentially Regulate Bax-Mediated Mitochondrial Membrane Permeabilization Both Directly and Indirectly. Molecular Cell. 2005;17:525–35. doi: 10.1016/j.molcel.2005.02.003. [DOI] [PubMed] [Google Scholar]
- 19.Dewson G, Kratina T, Sim HW, Puthalakath H, Adams JM, Colman PM, et al. To Trigger Apoptosis, Bak Exposes Its BH3 Domain and Homodimerizes via BH3:Groove Interactions. Molecular Cell. 2008;30:369–80. doi: 10.1016/j.molcel.2008.04.005. [DOI] [PubMed] [Google Scholar]
- 20.Dumitru R, Gama V, Fagan BM, Bower Jacquelyn J, Swahari V, Pevny Larysa H, et al. Human Embryonic Stem Cells Have Constitutively Active Bax at the Golgi and Are Primed to Undergo Rapid Apoptosis. Molecular Cell. 2012;46:573–83. doi: 10.1016/j.molcel.2012.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wolter KG, Hsu Y-T, Smith CL, Nechushtan A, Xi X-G, Youle RJ. Movement of Bax from the Cytosol to Mitochondria during Apoptosis. The Journal of Cell Biology. 1997;139:1281–92. doi: 10.1083/jcb.139.5.1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ohgushi M, Matsumura M, Eiraku M, Murakami K, Aramaki T, Nishiyama A, et al. Molecular Pathway and Cell State Responsible for Dissociation-Induced Apoptosis in Human Pluripotent Stem Cells. Cell Stem Cell. 2010;7:225–39. doi: 10.1016/j.stem.2010.06.018. [DOI] [PubMed] [Google Scholar]
- 23.Watanabe K, Ueno M, Kamiya D, Nishiyama A, Matsumura M, Wataya T, et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotech. 2007;25:681–6. doi: 10.1038/nbt1310. [DOI] [PubMed] [Google Scholar]
- 24.Zhang J, Khvorostov I, Hong JS, Oktay Y, Vergnes L, Nuebel E, et al. UCP2 regulates energy metabolism and differentiation potential of human pluripotent stem cells. The EMBO Journal. 2011;30:4860–73. doi: 10.1038/emboj.2011.401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gu P, LeMenuet D, Chung AC-K, Mancini M, Wheeler DA, Cooney AJ. Orphan Nuclear Receptor GCNF Is Required for the Repression of Pluripotency Genes during Retinoic Acid-Induced Embryonic Stem Cell Differentiation. Molecular and Cellular Biology. 2005;25:8507–19. doi: 10.1128/MCB.25.19.8507-8519.2005. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
- 26.Kang H-B, Kim Y-E, Kwon H-J, Sok D-E, Lee Y. Enhancement of NF-κB Expression and Activity Upon Differentiation of Human Embryonic Stem Cell Line SNUhES3. Stem Cells and Development. 2007;16:615–24. doi: 10.1089/scd.2007.0014. [DOI] [PubMed] [Google Scholar]
- 27.Soprano DR, Teets BW, Soprano KJ. Role of Retinoic Acid in the Differentiation of Embryonal Carcinoma and Embryonic Stem Cells. In: Gerald L, editor. Vitamins & Hormones. Academic Press; 2007. pp. 69–95. [DOI] [PubMed] [Google Scholar]
- 28.Durston AJ, Timmermans JPM, Hage WJ, Hendriks HFJ, de Vries NJ, Heideveld M, et al. Retinoic acid causes an anteroposterior transformation in the developing central nervous system. Nature. 1989;340:140–4. doi: 10.1038/340140a0. [DOI] [PubMed] [Google Scholar]
- 29.Niederreither K, Subbarayan V, Dollé P, Chambon P. Embryonic retinoic acid synthesis is essential for early mouse post-implantation development. Nature genetics. 1999;21:444–8. doi: 10.1038/7788. [DOI] [PubMed] [Google Scholar]
- 30.Inanç B, Elçin AE, Elçin YM. Human Embryonic Stem Cell Differentiation on Tissue Engineering Scaffolds: Effects of NGF and Retinoic Acid Induction. Tissue Engineering Part A. 2008;14:955–64. doi: 10.1089/ten.tea.2007.0213. [DOI] [PubMed] [Google Scholar]
- 31.Okada Y, Shimazaki T, Sobue G, Okano H. Retinoic-acid-concentration-dependent acquisition of neural cell identity during in vitro differentiation of mouse embryonic stem cells. Developmental Biology. 2004;275:124–42. doi: 10.1016/j.ydbio.2004.07.038. [DOI] [PubMed] [Google Scholar]
- 32.Schuldiner M, Yanuka O, Itskovitz-Eldor J, Melton DA, Benvenisty N. Effects of eight growth factors on the differentiation of cells derived from human embryonic stem cells. Proceedings of the National Academy of Sciences. 2000;97:11307–12. doi: 10.1073/pnas.97.21.11307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kaufmann SH, Desnoyers S, Ottaviano Y, Davidson NE, Poirier GG. Specific Proteolytic Cleavage of Poly(ADP-ribose) Polymerase: An Early Marker of Chemotherapy-induced Apoptosis. Cancer Research. 1993;53:3976–85. [PubMed] [Google Scholar]
- 34.Lazebnik YA, Kaufmann SH, Desnoyers S, Poirier GG, Earnshaw WC. Cleavage of poly(ADP-ribose) polymerase by a proteinase with properties like ICE. Nature. 1994;371:346–7. doi: 10.1038/371346a0. [DOI] [PubMed] [Google Scholar]
- 35.Faleiro L, Kobayashi R, Fearnhead H, Lazebnik Y. Multiple species of CPP32 and Mch2 are the major active caspases present in apoptotic cells. 1997;16:2271–81. doi: 10.1093/emboj/16.9.2271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hande KR. Etoposide: four decades of development of a topoisomerase II inhibitor. European Journal of Cancer. 1998;34:1514–21. doi: 10.1016/s0959-8049(98)00228-7. [DOI] [PubMed] [Google Scholar]
- 37.Tabas I, Ron D. Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress. Nat Cell Biol. 2011;13:184–90. doi: 10.1038/ncb0311-184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hetz C, Bernasconi P, Fisher J, Lee A-H, Bassik MC, Antonsson B, et al. Proapoptotic BAX and BAK Modulate the Unfolded Protein Response by a Direct Interaction with IRE1α. Science. 2006;312:572–6. doi: 10.1126/science.1123480. [DOI] [PubMed] [Google Scholar]
- 39.Wei MC, Zong W-X, Cheng EH-Y, Lindsten T, Panoutsakopoulou V, Ross AJ, et al. Proapoptotic BAX and BAK: A Requisite Gateway to Mitochondrial Dysfunction and Death. Science. 2001;292:727–30. doi: 10.1126/science.1059108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Puthalakath H, O’Reilly LA, Gunn P, Lee L, Kelly PN, Huntington ND, et al. ER Stress Triggers Apoptosis by Activating BH3-Only Protein Bim. Cell. 2007;129:1337–49. doi: 10.1016/j.cell.2007.04.027. [DOI] [PubMed] [Google Scholar]
- 41.Larsson O, Carlberg M, Zetterberg A. Selective killing induced by an inhibitor of N-linked glycosylation. Journal of Cell Science. 1993;106:299–307. doi: 10.1242/jcs.106.1.299. [DOI] [PubMed] [Google Scholar]
- 42.Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, et al. Cytochrome c and dATP-Dependent Formation of Apaf-1/Caspase-9 Complex Initiates an Apoptotic Protease Cascade. Cell. 1997;91:479–89. doi: 10.1016/s0092-8674(00)80434-1. [DOI] [PubMed] [Google Scholar]
- 43.Liu X, Kim CN, Yang J, Jemmerson R, Wang X. Induction of Apoptotic Program in Cell-Free Extracts: Requirement for dATP and Cytochrome c. Cell. 1996;86:147–57. doi: 10.1016/s0092-8674(00)80085-9. [DOI] [PubMed] [Google Scholar]
- 44.Du C, Fang M, Li Y, Li L, Wang X. Smac, a Mitochondrial Protein that Promotes Cytochrome c–Dependent Caspase Activation by Eliminating IAP Inhibition. Cell. 2000;102:33–42. doi: 10.1016/s0092-8674(00)00008-8. [DOI] [PubMed] [Google Scholar]
- 45.Youle RJ, van der Bliek AM. Mitochondrial Fission, Fusion, and Stress. Science. 2012;337:1062–5. doi: 10.1126/science.1219855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Frank S, Gaume B, Bergmann-Leitner ES, Leitner WW, Robert EG, Catez F, et al. The Role of Dynamin-Related Protein 1, a Mediator of Mitochondrial Fission. Apoptosis Developmental Cell. 2001;1:515–25. doi: 10.1016/s1534-5807(01)00055-7. [DOI] [PubMed] [Google Scholar]
- 47.Youle RJ, Karbowski M. Mitochondrial fission in apoptosis. Nat Rev Mol Cell Biol. 2005;6:657–63. doi: 10.1038/nrm1697. [DOI] [PubMed] [Google Scholar]
- 48.Brooks C, Wei Q, Feng L, Dong G, Tao Y, Mei L, et al. Bak regulates mitochondrial morphology and pathology during apoptosis by interacting with mitofusins. Proceedings of the National Academy of Sciences. 2007;104:11649–54. doi: 10.1073/pnas.0703976104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Imoto M, Tachibana I, Urrutia R. Identification and functional characterization of a novel human protein highly related to the yeast dynamin-like GTPase Vps1p. Journal of Cell Science. 1998;111:1341–9. doi: 10.1242/jcs.111.10.1341. [DOI] [PubMed] [Google Scholar]
- 50.Otera H, Wang C, Cleland MM, Setoguchi K, Yokota S, Youle RJ, et al. Mff is an essential factor for mitochondrial recruitment of Drp1 during mitochondrial fission in mammalian cells. The Journal of Cell Biology. 2010;191:1141–58. doi: 10.1083/jcb.201007152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Dai H, Smith A, Meng XW, Schneider PA, Pang Y-P, Kaufmann SH. Transient binding of an activator BH3 domain to the Bak BH3-binding groove initiates Bak oligomerization. The Journal of Cell Biology. 2011;194:39–48. doi: 10.1083/jcb.201102027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Czabotar Peter E, Westphal D, Dewson G, Ma S, Hockings C, Fairlie WD, et al. Bax Crystal Structures Reveal How BH3 Domains Activate Bax and Nucleate Its Oligomerization to Induce Apoptosis. Cell. 2013;152:519–31. doi: 10.1016/j.cell.2012.12.031. [DOI] [PubMed] [Google Scholar]
- 53.Lindsten T, Ross AJ, King A, Zong W-X, Rathmell JC, Shiels HA, et al. The Combined Functions of Proapoptotic Bcl-2 Family Members Bak and Bax Are Essential for Normal Development of Multiple Tissues. Molecular Cell. 2000;6:1389–99. doi: 10.1016/s1097-2765(00)00136-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16 INK4a. Cell. 1997;88:593–602. doi: 10.1016/s0092-8674(00)81902-9. [DOI] [PubMed] [Google Scholar]
- 55.Chen Z, Trotman LC, Shaffer D, Lin H-K, Dotan ZA, Niki M, et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature. 2005;436:725–30. doi: 10.1038/nature03918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Bunz F, Dutriaux A, Lengauer C, Waldman T, Zhou S, Brown J, et al. Requirement for p53 and p21 to sustain G2 arrest after DNA damage. Science. 1998;282:1497–501. doi: 10.1126/science.282.5393.1497. [DOI] [PubMed] [Google Scholar]
- 57.Kruse J-P, Gu W. Modes of p53 Regulation. Cell. 2009;137:609–22. doi: 10.1016/j.cell.2009.04.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kubbutat MHG, Jones SN, Vousden KH. Regulation of p53 stability by Mdm2. Nature. 1997;387:299–303. doi: 10.1038/387299a0. [DOI] [PubMed] [Google Scholar]
- 59.Momand J, Zambetti GP, Olson DC, George D, Levine AJ. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell. 1992;69:1237–45. doi: 10.1016/0092-8674(92)90644-r. [DOI] [PubMed] [Google Scholar]
- 60.Haupt Y, Maya R, Kazaz A, Oren M. Mdm2 promotes the rapid degradation of p53. Nature. 1997;387:296–9. doi: 10.1038/387296a0. [DOI] [PubMed] [Google Scholar]
- 61.Zhang Y, Xiong Y, Yarbrough WG. ARF Promotes MDM2 Degradation and Stabilizes p53: ARF-INK4a Locus Deletion Impairs Both the Rb and p53 Tumor Suppression Pathways. Cell. 1998;92:725–34. doi: 10.1016/s0092-8674(00)81401-4. [DOI] [PubMed] [Google Scholar]
- 62.Li L, Liao J, Ruland J, Mak TW, Cohen SN. A TSG101/MDM2 regulatory loop modulates MDM2 degradation and MDM2/p53 feedback control. Proceedings of the National Academy of Sciences. 2001;98:1619–24. doi: 10.1073/pnas.98.4.1619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kojima K, Konopleva M, McQueen T, O’Brien S, Plunkett W, Andreeff M. Mdm2 inhibitor Nutlin-3a induces p53-mediated apoptosis by transcription-dependent and transcription-independent mechanisms and may overcome Atm-mediated resistance to fludarabine in chronic lymphocytic leukemia. Blood. 2006;108:993–1000. doi: 10.1182/blood-2005-12-5148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Marión RM, Strati K, Li H, Murga M, Blanco R, Ortega S, et al. A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature. 2009;460:1149–53. doi: 10.1038/nature08287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Hong H, Takahashi K, Ichisaka T, Aoi T, Kanagawa O, Nakagawa M, et al. Suppression of induced pluripotent stem cell generation by the p53–p21 pathway. Nature. 2009;460:1132–5. doi: 10.1038/nature08235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lowry WE, Richter L, Yachechko R, Pyle AD, Tchieu J, Sridharan R, et al. Generation of human induced pluripotent stem cells from dermal fibroblasts. Proceedings of the National Academy of Sciences. 2008;105:2883–8. doi: 10.1073/pnas.0711983105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wang G, Shimada E, Zhang J, Hong JS, Smith GM, Teitell MA, et al. Correcting human mitochondrial mutations with targeted RNA import. Proceedings of the National Academy of Sciences. 2012;109:4840–5. doi: 10.1073/pnas.1116792109. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
