Abstract Although pluripotent stem cell (PSC) properties, such as differentiation and infinite proliferation, have been well documented within the frameworks of transcription factor networks, epigenomes, and signal transduction, they remain unclear and fragmented. Directing attention toward translational regulation as a bridge between these events can yield additional insights into previously unexplained mechanisms. Our functional CRISPR interference screen–based approach revealed that EIF3D, a translation initiation factor, is crucial for maintaining primed pluripotency. Loss of EIF3D disrupted the balance of pluripotency-associated signaling pathways, thereby compromising primed pluripotency. Moreover, EIF3D ensured robust proliferation by controlling the translation of various p53 regulators, which maintain low p53 activity in the undifferentiated state. In this way, EIF3D-mediated translation contributes to tuning the homeostasis of the primed pluripotency networks, ensuring the maintenance of an undifferentiated state with high proliferative potential. This study provides further insights into the translation network in maintaining pluripotency. __________________________________________________________________ Translation controls the homeostasis of key signaling pathways that define primed pluripotent stem cell identity. INTRODUCTION Pluripotent stem cells (PSCs) have the capacity for self-renewal under appropriate conditions while maintaining their distinct attributes, including differentiation potential and unlimited proliferation ([38]1–[39]5). Pluripotency is categorized into two types: naïve, resembling the preimplantation inner cell mass, and primed, akin to the postimplantation epiblast ([40]6). These categories differ in their specific requirements for self-renewal, differentiation capacity, and epigenetic status. Previous studies have shown that inhibiting multiple kinases helps sustain naïve pluripotency in both rodents and humans, suggesting a conserved, kinase-independent strategy across species ([41]7–[42]10). Conversely, shifting from kinase inhibition to specific growth factor stimuli enables naïve PSCs to transition into primed pluripotency, a state poised for differentiation into various somatic lineages ([43]7, [44]11). Unlike the naïve state, the fate of primed pluripotency depends on a range of signaling inputs, including fibroblast growth factor (FGF), insulin-like growth factor, and transforming growth factor–β (TGFβ) ([45]12–[46]14). Thus, kinase signaling dynamics are crucial for the transition between these states and their ongoing maintenance. Paradoxically, the same growth factors that support primed pluripotency also initiate lineage-specific differentiation programs ([47]15, [48]16). Maintaining a delicate balance between strong and weak kinase signaling is key to preserving the equilibrium between self-renewal and differentiation induction ([49]17, [50]18). Although primed pluripotency, maintained by finely tuned signaling, is an important research area in stem cell and developmental biology, the complex mechanisms governing this balance remain elusive. Translation, the process by which RNA is converted to protein, is critical for cellular homeostasis and the study of primed pluripotency. In this context, it is notable that differentiation cues actively enhance protein synthesis ([51]19). This shift in translation dynamics highlights the importance of translational control in dictating pluripotency and differentiation. Gene analysis showing discrepancies between mRNA and protein levels has unveiled the critical role of context-dependent posttranscriptional regulation in maintaining primed pluripotency ([52]20). This suggests that translational modulation substantially influences the primed pluripotent state, independent of transcriptional regulation. Several translational factors associated with primed pluripotency have been identified ([53]21–[54]24). For example, one of them, eukaryotic translation initiation factor gamma 2 (EIF4G2) (also known as NAT1 and DAP5) has been reported to interact with the eIF3 complex and regulate the translation of specific mRNAs in a noncanonical manner; however, a comprehensive understanding of the role of translational regulation in stem cell homeostasis and fate determination remains elusive ([55]25, [56]26). Here, we show that translational regulation by EIF3 subunit D (EIF3D) is critical for maintaining the identity of human primed PSCs. Loss of EIF3D resulted in impaired self-renewal of primed PSCs and their differentiation potential into three germ layer lineages. As a mechanism to explain these phenotypes, we revealed that EIF3D regulates the activities of key signaling pathways for the self-renewal of primed PSCs. In addition, EIF3D also controls the translation of a set of p53 regulators that render p53 activity low, resulting in robust proliferation of undifferentiated PSCs. Collectively, we conclude that the homeostasis of primed pluripotency is maintained through translational regulation by EIF3D. RESULTS EIF3D is essential for the maintenance of human primed pluripotency To identify the genes essential for maintaining primed pluripotency, we conducted genome-wide functional screening using the CRISPR interference (CRISPRi) platform ([57]27, [58]28). A single guide RNA (sgRNA) library targeting genome-wide mRNAs was delivered into a human PSC line carrying a doxycycline (Dox)–inducible KRAB-dCas9 transgene ([59]28). Self-renewing cells positive for stage-specific embryonic antigen 5 (SSEA-5) ([60]29), a surface marker specifically expressed on undifferentiated primed PSCs, were purified before (day 0) and 8 and 16 days after the induction of knockdown (KD). The essential genes for the self-renewal of primed PSCs were identified as targets of significantly depleted sgRNAs in purified SSEA-5–positive cells. MAGeCK, the method for model-based normalization and robust ranking aggregation, identified 1255 genes on day 8 and 1686 genes on day 16 that positively influence the self-renewal of primed human PSCs (data S1 and S2) ([61]30, [62]31). A comparison with the previously reported CRISPR screens in human primed PSCs confirmed the reliability of our screening results (fig. S1, A and B) ([63]32–[64]34). Gene ontology (GO) analysis of these genes on day 16, critical for maintaining primed PSC identity, highlighted translation-related terms as significantly enriched (fig. S1C). We focused on the eIF3 family, which interacts with EIF4G2, a translational regulator that is critical for maintaining primed pluripotency ([65]22). Our CRISPRi screen revealed that EIF4G2 was essential for the self-renewal of primed human PSCs on days 8 and 16 (fig. S1D and data S1 and S2). The screening results suggested that EIF3D, a cap-binding protein ([66]35), had a top-ranked notable role in both early (day 8) and late (day 16) phases of KD among the eIF3 family genes ([67]Fig. 1A and fig. S1E). Therefore, we focused on the role of EIF3D in primed pluripotency. EIF3D is abundantly expressed in undifferentiated induced PSCs (iPSCs) compared with that in human dermal fibroblasts (HDFs), and its levels markedly decreased upon differentiation ([68]Fig. 1B). This expression pattern is similar to pluripotency factors, such as Octamer-binding transcription factor 3/4 (OCT3/4), SRY-box transcription factor 2 (SOX2), and Nanog homeobox (NANOG), suggesting the potential role of EIF3D in primed PSCs. Fig. 1. EIF3D is essential for maintaining primed pluripotency. [69]Fig. 1. [70]Open in a new tab (A) Rank plot from CRISPRi screening. Red and blue dots represent genes significantly increased or decreased (±1 SD) following 8 and 16 days of KD, respectively. n = 3. The full list is provided in data S1 and S2. (B) Protein expression in undifferentiated and differentiating PSCs (10 and 20 days post-FGF withdrawal) and HDFs. Vinculin (VCL) was used as a loading control. (C) Representative images of control and EIF3D KD iPSCs, 5 days post-KD induction. Scale bars, 100 μm. (D) Cell counts on days 3 (P = 4.70 × 10^−4) and 5 (P = 3.53 × 10^−7) post-KD induction (mean ± SD, n = 6). P values determined via unpaired t test. (E) Cell cycle phase distribution (mean ± SD, n = 3). G[1]: P = 2.14 × 10^−3; S: P = 4.43 × 10^−7; G[2]/M: P = 1.99 × 10^−4, calculated by unpaired t test. (F) RNA expression of pluripotency and p53-related genes during EIF3D KD. n = 3. (G) Expression of pluripotency and p53-associated proteins during EIF3D KD. d, days; FC, fold change. To explore its function, we generated Dox-inducible EIF3D KD iPSC lines using a CRISPRi platform (fig. S2A). Control cell lines expressing a sgRNA with nontarget spacer sequence were used for comparison in all subsequent analyses. Following EIF3D KD, we observed morphological changes characterized by flattened colonies with indistinct edges and notably enlarged nuclei, evoking differentiation ([71]Fig. 1C and fig. S2, B and C). In addition, EIF3D KD resulted in a marked reduction in cell numbers between days 3 and 5 postinduction ([72]Fig. 1D). Investigating this phenotype, we measured DNA synthesis via 5-ethynyl-2′-deoxyuridine (EdU) incorporation, revealing a significant decrease in the S phase cell population and a corresponding increase in cells in the G[1] and G[2]/M phases following EIF3D KD ([73]Fig. 1E and fig. S2, D to F). These results collectively suggest that EIF3D KD imposes growth arrest on primed human PSCs. Moreover, EIF3D KD reduced the expression of transcripts encoding core pluripotency transcription factors, which are indicative markers of PSCs ([74]Fig. 1F). The decrease in NANOG expression was more rapid and pronounced than that of POU5F1 (encoding OCT3/4) and SOX2. Protein expression analyses exhibited a similar trend ([75]Fig. 1G and fig. S2G). Notably, transcriptional targets of p53, such as CDKN1A and MDM2 mRNAs, and their translation products were significantly up-regulated in EIF3D KD cells, akin to the changes seen in differentiated iPSCs induced by FGF withdrawal ([76]Fig. 1, B, F, and G). Other hallmarks of senescence (INK4A and ARF) and cell death (FAS) markers were also elevated ([77]Fig. 1F). Similar to iPSC differentiation, the absence of EIF3D led to increased p53 protein expression, while TP53 mRNA showed only modest changes ([78]Fig. 1, B, F, and G, and fig. S2G). The p53 protein up-regulated by EIF3D KD increased in its size over time. This observation can be explained by the fact that the short form lacking the N terminus (known as dN40) is expressed in undifferentiated PSCs, whereas the p53 protein induced by EIF3D KD is the full-length form (fig. S2H) ([79]36). In correlation with the increase in p53 protein, the levels of p21 and MDM2 proteins increased in EIF3D KD PSCs ([80]Fig. 1, B and G, and fig. S2G). In addition, we detected remarkable elevation of the phosphorylated Ser^51 of EIF2α on day 5 of EIF3D KD, suggesting the increase of cellular stress ([81]Fig. 1G). Overall, the KD phenotypes suggested that EIF3D loss diminishes the primed PSC identity along with several intriguing changes. On the basis of the EIF3D KD phenotype in primed PSCs, we investigated the effects of EIF3D KD on cell morphology and proliferation in other cell types. Short hairpin RNA (shRNA) against EIF3D efficiently suppressed EIF3D expression in 293T/17 cells, HepG2 cells, and HDFs (fig. S3A). EIF3D KD did not alter the morphology of these three cell types (fig. S3B). One of the two shRNAs against EIF3D caused a significant decrease in the proliferation of 293T/17 cells. At the same time, there was no significant change in the proliferative capacity of HepG2 cells or HDFs (fig. S3C). These data suggest that the effects of EIF3D KD on cell morphology and proliferation are limited in the cell types tested, and the phenotype of primed PSC is remarkable. To confirm that the phenotypes observed in the primed PSCs were due to EIF3D KD, we introduced EIF3D and its mutants into the EIF3D KD iPSCs to perform the rescue experiments. Although the mCherry transgene introduced as a control could not prevent the phenotypes observed in the EIF3D KD iPSCs, the EIF3D transgene prevented the emergence of the phenotypes, including morphological changes, reduction of PSC marker expression, activation of the p53 pathway, and proliferation defect (fig. S4, A to D). Furthermore, either phosphomimetic (S528D/S529D) or phosphoinhibitory (S528N/S529N) mutation ([82]37) in EIF3D, known to inhibit or promote Cap binding, respectively, did not affect the prevention of the EIF3D KD phenotypes by EIF3D transgene (fig. S4, A to D). These data demonstrate that the EIF3D KD-induced disruption of self-renewal in human primed PSCs shown above is independent of the phosphorylation status of EIF3D. Given these results, we explored the potential roles of increased p53 levels and reduced NANOG expression in the phenotypes arising from EIF3D KD. Introducing exogenous NANOG did not restore pluripotency marker expression or resolve the proliferative impairment (fig. S5, A to D). In addition, the concurrent KD of TP53 with EIF3D did not fully restore pluripotency marker expression (fig. S5, E to H). However, these experiments showed partial recovery of cellular proliferation and a decrease in elevated p21 expression (fig. S5, G and H). These findings suggest that analyzing hallmark genes associated with undifferentiated or differentiated states alone is insufficient to fully understand the role of EIF3D in pluripotency. Nonetheless, the data indicate the involvement of the p53-p21 pathway in regulating the proliferation of primed PSCs, although other pathways likely contribute to EIF3D-mediated self-renewal. In summary, the collective findings highlight the multifaceted role of EIF3D in maintaining primed pluripotency through complex molecular interactions. Loss of EIF3D diminishes primed pluripotency with limited impact on three germ layer specifications We then conducted a comprehensive genome-wide transcriptome analysis to further investigate the underlying mechanisms from a broader perspective. To elucidate the cell fate changes in primed PSCs induced by EIF3D KD, we compared the global gene expression profiles of EIF3D KD cells with those of iPSCs differentiated through suppression of core transcription factors (fig. S6, A to C), and iPSC-derived cells directed toward endoderm (EN), mesoderm (ME), and neuroectoderm (NE) lineages (fig. S6D). EIF3D KD resulted in an incremental increase in differentially expressed genes (DEGs) over successive days compared with that in the controls ([83]Fig. 2A and fig. S6E). Notably, despite notable changes in gene expression, the EIF3D KD profile showed less similarity to the corresponding comparatives such as trilineage differentiated cells and core transcription factor KD cells ([84]Fig. 2A and fig. S6F). Instead, it seemed to enter an independent state, marked by a lack of clear lineage commitment to any of the three germ layers. This was accompanied by down-regulating key pluripotency and primed PSC markers, including ZIC2, CD24, and SFRP2 ([85]Fig. 2, A and B, and table S1) ([86]32, [87]38–[88]42). By contrast, the increase or decrease in transcription factors and housekeeping genes ([89]43–[90]45) due to EIF3D KD was limited (fig. S6G). GO analysis revealed that EIF3D KD up-regulated genes associated with inconsistent differentiation terms, while genes related to cell cycle and division were down-regulated (fig. S6H). These results are consistent with the observed EIF3D KD phenotypes, including growth retardation and loss of pluripotency, with minimal contribution to specific lineages. The evidence indicates that EIF3D maintains primed pluripotency through mechanisms distinct from those of core transcription factors or the inhibition of specific lineage commitments. Fig. 2. Loss of EIF3D impairs trilineage specifications. [91]Fig. 2. [92]Open in a new tab (A) Principal components (PC) analysis of RNA-seq data. Each dot represents the average value of replicates. n = 3. (B) Volcano plots displaying DEGs between control and EIF3D KD iPSCs, 3 and 5 days after Dox addition. Understated-colored dots represent DEGs. Highlighted-colored dots denote key pluripotency markers. Red and blue dots indicate genes significantly up-regulated and down-regulated, respectively [|log[2] fold change (FC)| > 1, adjusted P < 0.05]. n = 3. See full gene list and FC in table S1. See also fig. S6. (C) The scheme of trilineage differentiation. Three days before differentiation induction, Dox was added to the control and sgEIF3D cell lines. On day 3, the cells were exposed to the specific culture conditions of each lineage and simultaneously maintained under the PSC condition for comparison. Dox continued to be added until day 8. (D) Phase contrast and immunocytochemistry images of control (top) and sgEIF3D (bottom) cells after differentiation, showing specified proteins (red). Nuclei visualized with Hoechst 33342 (blue). Scale bars, 100 μm. (E) RNA expression of pluripotency and lineage marker genes after differentiation (mean ± SD, n = 9). *P < 0.0001 for control versus sgEIF3D in the same condition determined by one-way analysis of variance (ANOVA). Loss of EIF3D impairs trilineage differentiation The impairment of self-renewal by EIF3D KD, including the down-regulation of pluripotency genes, suggests that EIF3D contributes to the capacity of primed PSCs. Therefore, we next tested whether EIF3D KD primed PSCs could differentiate into three germ layers. After 3 days of KD induction, a time when EIF3D proteins have almost disappeared, the cells were exposed to culture conditions for differentiation into each differentiated lineage ([93]Fig. 2C). Following a 5-day period of differentiation, the expression of distinct lineage markers, including SOX17 and FOXA2 for EN, HAND1 and FOXF1 for ME, and PAX6 and SOX1 for NE, was assessed. It was shown that the control cells exhibited successful differentiation, while the EIF3D KD cells displayed limited expression of these specific markers, indicating a failure to differentiate ([94]Fig. 2, D and E). The expression of the pluripotency marker NANOG was completely silenced in well-differentiated control cells, whereas EIF3D KD cells retained its partial expression ([95]Fig. 2E). EIF3D KD cells not only lacked lineage marker activation but also exhibited a morphology characterized by enlarged nuclei in all culture conditions, including the PSC condition ([96]Fig. 2D). In addition, cells could survive even after a total of 8 days of EIF3D KD regardless of culture conditions. These results indicate that primed PSCs lacking EIF3D not only fail to maintain self-renewal but also exhibit deficiencies in their ability to differentiate into the trilineage, a hallmark of pluripotency. Loss of EIF3D decreases naïve pluripotency slower than in primed PSC To further investigate the effects of EIF3D KD in primed PSCs, beyond the typical three germ layers derived from these cells, our study expanded to naïve PSCs which resemble an earlier developmental fate. Therefore, we examined the differentiation of naïve PSCs into a primed state to further evaluate the role of EIF3D in maintaining primed pluripotency ([97]Fig. 3A). Before the transition to the primed state, we induced EIF3D KD in naïve PSCs for 3 days, which did not result in any observable abnormalities ([98]Fig. 3B). However, when we changed the culture conditions from the kinase-inhibiting naïve state to the growth factor-rich primed state, the EIF3D KD naïve PSCs showed an inability to differentiate into the primed state. This was marked by massive cell death within 4 days ([99]Fig. 3B). Given this evidence of the inability to self-renew primed PSCs following EIF3D KD, we conclude that EIF3D is essential for the maintenance of primed pluripotency. Fig. 3. The comparison of EIF3D’s role in primed and naïve PSCs. [100]Fig. 3. [101]Open in a new tab (A) Scheme of differentiation of naïve PSCs into primed state. (B) Differentiation of naïve PSCs to primed state. Induction of differentiation from naïve to primed PSCs, 3 days post-Dox addition, by altering culture conditions. Representative images of specified cell lines and days are shown. Scale bars, 100 μm. (C) Scheme of the comparison between naïve and primed PSCs with EIF3D KD. (D) Representative images of control and sgEIF3D naïve PSCs, 5 days post-Dox addition. Scale bars, 100 μm. (E) Protein expression in control and EIF3D KD naïve (N) and primed (P) PSCs, 5 days post-Dox addition. (F) Hierarchical clustering of the averaged values of RNA-seq data. (G) Scatterplots of log[2]FC (control and EIF3D KD) from the Wald tests of primed 5 and 3 days (left) and primed 5 days and naïve 5 days (right) post-Dox addition. Colors show up (red) and down (blue) from the result of primed control and EIF3D KD 5 days after Dox addition. Pearson’s correlation values and P values of t test were shown on top. (H) The DEG numbers between control versus EIF3D KD. (I) RNA expression of EIF3D and pluripotency marker genes after 7 days of Dox addition (mean ± SD, n = 3). *P < 0.05 determined by unpaired t test. To assess the EIF3D KD phenotype in naïve PSCs, we compared primed and naïve PSCs after 5 days of EIF3D KD ([102]Fig. 3C). EIF3D KD attenuated naïve pluripotent signatures, as evidenced by decreased expression of a naïve PSC marker KLF17, although without noticeable morphological changes in naïve PSC colonies ([103]Fig. 3, D and E). In addition, the comparison with the samples shown in [104]Fig. 2A such as control and EIF3D KD primed PSCs, trilineage differentiated cells, and core transcription factor KD cells revealed that the impact of EIF3D KD on gene expression changes in naïve PSCs was less pronounced than those in the primed state ([105]Fig. 3F). The variance of fold changes between control versus EIF3D KD and the number of DEGs also supported the significant alterations of gene expression by EIF3D KD day 5 post-KD induction in primed PSCs rather than naïve state ([106]Fig. 3, G and H). However, when the culture of EIF3D KD naïve PSCs reached day 7, it could not be maintained due to a notable decrease in cell number. The quantitative reverse transcription polymerase chain reaction (qRT-PCR) results showed that the expression of PSC markers was reduced by EIF3D KD ([107]Fig. 3I). Although the phenotype emergence of naïve PSCs was slower than that of primed PSCs, EIF3D was required for self-renewal of naïve PSCs. While EIF3D plays critical roles in naïve and primed pluripotency, we further focused on its role in primed PSCs. EIF3D orchestrates translation of key signaling pathways in primed pluripotency On the basis of global transcriptome data, we observed a lesser degree of change induced by EIF3D KD on day 3 compared to day 5 ([108]Fig. 2, A and B, and fig. S6, E and F). Therefore, to understand the initial response to EIF3D loss, we analyzed translation status on day 3 post-KD induction. Puromycin incorporation showed that EIF3D KD reduced de novo protein synthesis to 45% relative to control cells ([109]Fig. 4A and fig. S7). Polysome profile analysis indicated a significant accumulation of the 80S ribosomal subunit with EIF3D KD, suggesting decreased translation initiation ([110]Fig. 4, B and C). These results indicated that the absence of EIF3D in primed PSCs led to noticeable alterations in translation. Fig. 4. EIF3D-mediated translation regulates multiple signaling pathways. [111]Fig. 4. [112]Open in a new tab (A) Quantification of de novo protein synthesis by detecting incorporated puromycin. n = 3. P = 6.75 × 10^−3 determined by unpaired t test. a.u., arbitrary units. (B) Representative polysome profiles of sgEIF3D primed PSCs compared to control lines on day 3 post-Dox addition. (C) Area under the curve (AUC) quantification for specified ribosomal fractions (mean ± SD, n = 3). Ratios 60S/40S: P = 0.011; 80S/40S: P = 0.012; polysomes (PS)/40S: P = 0.094, calculated using unpaired t test. (D) Categorization of ORFs with varying translation efficiency (TE) during EIF3D KD. Displayed are all ORFs translated in human iPSCs (all) and those down-regulated (down) or up-regulated (up) by EIF3D KD over 3 days. (E) Volcano plot showing up-regulated DTEGs (uDTEG; red) and down-regulated DTEGs (dDTEGs; blue) (|log[2]FC| > 1, adjusted P < 0.05). See full gene list, FC, and adjusted P value in data S3 and S4. (F) Pathway enrichment analysis of dDTEGs using WikiPathways. (G) Volcano plots displaying log[2] FC of transcripts in specified pathways according to WikiPathways (WP437, WP399, WP481, WP382, and WP366 for EGF, WNT, insulin, MAPK, and TGFβ pathways, respectively). Understated-colored dots represent DTEGs. Highlighted-colored dots denote transcripts in specified pathways. Red and blue dots indicate genes whose TEs are significantly up-regulated and down-regulated, respectively (|log[2] FC| > 1, adjusted P < 0.05). n = 3. See full gene list, FC, and adjusted P value in data S5 to S9. (H) Phosphorylation status of key proteins in the signaling pathways across the timeline of EIF3D KD. To determine the translational changes due to EIF3D KD, we performed ribosome profiling (fig. S8, A to C). To consider translation in noncanonical manner, we identified 31,263 open reading frames (ORFs) undergoing translation in human primed iPSCs, classified as follows: 45% as annotated coding sequences (CDS), 42% as variant CDS, 2% as unidentified ORFs, and 8% as upstream ORFs (uORFs) ([113]Fig. 4D). EIF3D KD increased translation efficiency (TE) in 284 ORFs and decreased it in 1340 ORFs. The increased TE group featured a higher percentage of uORFs (35.07%), whereas the decreased TE group showed no significant preference in ORF classification. uORF is a cis-regulatory element that regulates TE of downstream ORF ([114]25). Therefore, we investigated the changes in TE of downstream ORFs of the up- and down-regulated uORFs and the nonsignificant uORFs, following EIF3D KD. The TE of ORFs located downstream of up-regulated uORFs showed minimal differences compared to that of ORFs located downstream of down-regulated uORFs (fig. S8D), indicating that EIF3D did not contribute to cis-regulatory changes between ORFs but rather influenced the TE of all ORFs across the transcripts. These results suggest that EIF3D is involved in the regulation of ribosome binding to uORFs in primed PSCs and also plays a role in the regulation of TE across the entire transcript, including uORFs and annotated ORFs such as CDS. We subsequently examined the transcript-wise translation status following EIF3D KD. Ribosome profiling revealed that EIF3D KD significantly altered the TE of 1321 genes (increased in 402; decreased in 919) ([115]Fig. 4E and data S3 and S4), which we term “differential translation efficiency genes” (DTEGs). Compared with a previous study that performed photoactivatable ribonucleoside-enhanced cross-linking and immunoprecipitation and identified JUN as a target of EIF3A, EIF3B, EIF3D, and EIF3G in 293T cells ([116]35), in this study, there are a small number of overlapped genes with the ribosome profiling data of EIF3D KD primed PSCs (fig. S8E). In addition, we confirmed that TE of JUN was not significantly changed in EIF3D KD primed PSCs (fig. S8F), suggesting a cellular context-dependent manner of translational regulation by EIF3D and supporting the importance of the cap-binding independent role of EIF3D in primed PSCs (Fig. S4). Pathway analysis indicated that down-regulated DTEGs (dDTEGs) were linked to several signaling pathways, including epidermal growth factor (EGF), WNT, insulin, mitogen-activated protein kinase (MAPK), and TGFβ, all of which are critical for the maintenance of primed pluripotency ([117]Fig. 4F) ([118]46). In addition, dDTEGs were also associated with several terms, including “pluripotency.” By contrast, up-regulated DTEGs (uDTEGs) did not show significantly enriched terms. Given the EIF3D KD phenotype, which includes the loss of primed pluripotency, it is plausible that these signaling pathways involving dDTEGs are implicated. On the basis of the pathway analysis results, we confirmed the TEs of EIF3D targets in each enriched pathway. The volcano plots revealed significant TE alterations in the transcripts of the EGF, WNT, insulin, MAPK, and TGFβ pathways ([119]Fig. 4G, fig. S8G, and data S5 to S9) ([120]47). Next, we examined the phosphorylation status of key proteins in these signaling pathways potentially regulated by EIF3D. EIF3D KD led to the hyperactivation of the MAPK, insulin (AKT, mTOR, and p70S6K), and WNT pathways while simultaneously suppressing the TGFβ pathway through SMAD2 ([121]Fig. 4H). These results confirm that EIF3D KD disrupts the balance of multiple signaling activities in primed PSCs through translational regulation. EIF3D inhibits p53 protein expression by modulating translation of p53 regulators Besides the dysregulation of multiple kinase pathways, there is a notable increase in p53 protein and subsequent activation of the p53 pathway due to EIF3D KD ([122]Fig. 1G and fig. S2G). However, ribosome profiling data revealed no significant change in p53 TE following EIF3D KD (likelihood ratio test, fold change = 1.01, adjusted P = 0.92). This led us to hypothesize about an indirect regulatory mechanism. To deepen our understanding, we performed a comparative analysis between DTEGs and a compilation of posttranscriptional regulators of p53 protein expression ([123]48). Setting a TE threshold of 1.5 fold change, EIF3D KD resulted in significant translation dysregulation of 207 of 818 genes, representing 25.3% of the list ([124]Fig. 5A and data S10). In addition, we found 54 genes under the condition of a TE threshold of twofold change between control and EIF3D KD ([125]Fig. 5A and data S10). The protein analysis confirmed the decreased expression of p53 regulator proteins in EIF3D KD primed PSCs ([126]Fig. 5B), suggesting that EIF3D indirectly affects p53 protein expression by regulating the translation of its regulators. Fig. 5. Indirect regulation of p53 protein through EIF3D-mediated translational regulation. [127]Fig. 5. [128]Open in a new tab (A) Venn diagrams display the overlap between DTEGs with significant TE (|log[2] FC| > 0.58) and p53 regulators. Genes with higher TE (|log[2] FC| > 1) are highlighted in red. See full gene list, FC, and adjusted P value in data S10. (B) Protein expression of p53 regulators translationally controlled by EIF3D. High TE (|log[2] FC| > 1) includes RBBP6, SSU72, and TCP1; moderate TE (|log[2] FC| > 0.58) includes KDM5C, WDR5, WDR82, and REST. (C) Luciferase reporter assays showing the effects of RBBP6 (P = 2.89 × 10^−9) and GAPDH (P = 0.85) 5′UTRs on translation in control and EIF3D KD primed PSCs (mean ± SD, n = 6). P values determined via unpaired t test. (D) Representative images of specified cells, 5 days post-KD induction. Scale bars, 100 μm. (E) Cell counts of the cells depicted in [129]Fig. 5D (mean ± SD, n = 6). (F) Relative gene expression in cells from [130]Fig. 5D. Values normalized to GAPDH and compared to control without Dox. n = 3. (G) Expression of specified proteins in cells from [131]Fig. 5D. For example, we identified RB binding protein 6 (RBBP6), a negative regulator of p53 protein stability in the dDTEG list (data S3) ([132]49). Luciferase reporter assays revealed that the 5′ untranslated region (5′UTR) of RBBP6 was responsible for EIF3D-mediated translational regulation in primed PSCs ([133]Fig. 5C). By contrast, the absence of EIF3D did not affect the translation of the GADPH 5′UTR-containing reporter ([134]Fig. 5C). These data together with the ribosome profiling results indicate that RBBP6 is a target of EIF3D in primed PSCs. As expected, RBBP6 KD mimicked EIF3D KD phenotypes, including cell growth defects with morphological changes, decreased expression of pluripotency markers, increased p53 protein, and elevated expression of CDKN1A/p21 and MDM2 ([135]Fig. 5, D to G). These findings indicate that EIF3D plays a role in modulating p53 protein expression by controlling the translation of its regulators. Artificial dysregulation of signaling balance recapitulates loss of pluripotency Last, we investigated whether deliberately disrupting the balance of pluripotency-associated signaling pathways and activating the p53 pathway could recapitulate the loss of primed pluripotency. We used chemical compounds and a growth factor to induce changes observed in primed EIF3D KD PSCs, including the activation of EGF/MAPK, mTOR, AKT, WNT, and p53, and the inhibition of TGFβ ([136]Fig. 6A). Treatment with five factors (5F consisted of EGF, AKT activator, mTOR activator, WNT activator, and TGFβ inhibitor) decreased cell number without obvious morphological changes compared to the dimethyl sulfoxide (DMSO)–treated control cells ([137]Fig. 6, B and C). Treatment solely with Nutlin-3a (N3a), a p53 activator, resulted in marked morphological changes with clear cell borders and irregular colony edges, yet it did not affect the expression of representative PSC markers ([138]Fig. 6, B and D). Notably, PSCs treated with N3a and EIF3D KD for 3 days exhibited clustering ([139]Fig. 6D). Furthermore, the conditions containing N3a (6F, N3a, N3a + 5F, and N3a + 6F) induced massive cell death, with a much greater reduction in cell numbers than 5F ([140]Fig. 6, B and C). The p53 overexpression and MDM2 KD also resulted in the death of undifferentiated primed PSCs, highlighting the critical role of p53 suppression in primed PSCs for survival and proliferation, rather than maintaining undifferentiation (fig. S9). Fig. 6. Disruption of primed pluripotency through manipulation of EIF3D’s target pathways. [141]Fig. 6. [142]Open in a new tab (A) Overview of chemical compound and growth factor treatments. Control cells were treated with DMSO. (B) Representative images of iPSCs treated with specified factors. Scale bars, 100 μm. (C) Cell counts on day 5 (mean ± SD, n = 9). *P < 0.0001 compared to the control; †P < 0.05 versus 5F determined by one-way ANOVA. (D) Sample clustering from (A), alongside control and sgEIF3D PSC core transcription factor (TF) KD iPSCs, as well as trilineage differentiated cells, highlighting selected PSC marker genes. n = 3. (E) Schematic representation of the model describing how EIF3D-mediated translational regulation balances homeostasis of critical signaling pathways in primed pluripotency. The dysregulation of multiple signaling pathways by the 5F and 6F treatment for 5 days substantially decreased the expression of pluripotency-associated genes, and they were clustered with PSCs after 5 days of EIF3D KD ([143]Fig. 6D). These results suggest that the disruption of multiple signaling pathways induced by EIF3D KD is responsible for the loss of primed pluripotency. Collectively, this study demonstrates that EIF3D plays a key role in safeguarding self-renewal and robust proliferation in primed pluripotency by coordinating essential signaling pathways and inhibiting the p53 pathway ([144]Fig. 6E). DISCUSSION This study demonstrates that EIF3D is critical for maintaining primed pluripotency through translational regulation that finely balances kinase signaling and suppresses the p53 pathway. The absence of EIF3D decreased global translation in primed PSCs by approximately 50%. However, previous research indicated that reduced global translation did not necessarily result in the loss of pluripotency, making it unlikely that the EIF3D KD phenotype is a consequence of stress due to a lowering translation ([145]24). On day 3 post-EIF3D KD, attenuated pluripotency, reduced translation, and changes in the activity of the p53 pathway were detected, whereas an increase in the phosphorylation of EIF2α, a sign of cellular stress, was not observed until day 5. The disparity in time indicates that the EIF3D KD phenotype in primed PSCs is not simply due to stress. Meanwhile, since translation factors such as EIF2B5, which are essential for self-renewal of primed PSCs, are targets of EIF3D, loss of EIF3D may affect the primed PSC identity by altering the translation machinery. Further research into comprehensive translational control mechanisms provides further insights. One of the remarkable phenotypes of EIF3D KD in primed PSCs shown in this study is the rapid loss of proliferation. We have also shown that the reduction in proliferative capacity caused by EIF3D KD also occurs in 293T/17 cells as non-PSCs. Previous studies have also shown that EIF3D promotes proliferation in some types of cancer cell lines ([146]50–[147]53). This is not unexpected given that EIF3D is expressed in many cell types and is responsible for regulating translation initiation. In this study, we did not analyze which signaling pathways are regulated by EIF3D KD in non-PSCs. Further analysis will provide more insight into the function of EIF3D on cellular proliferation. On the other hand, we have shown that the p53 pathway is activated by EIF3D KD in primed PSCs, leading to cell proliferation defects. Maintaining low p53 activity is essential in undifferentiated PSCs, although the exact regulatory mechanism remained unclear ([148]54). Our study sheds light on the indirect inhibition of p53 by EIF3D through targeted translation modulation of p53 regulators. Moreover, the marked increase in p53 protein in EIF3D KD, combined with the inverse correlation between increased p53 protein and decreased EIF3D expression during PSC differentiation, strongly suggests the pivotal role of EIF3D in suppressing p53. On the other hand, primed PSCs expressed a short isoform of p53 translated from an AUG start codon downstream of the canonical translation start site. Such a mechanism may also be involved in the tight regulation of p53 protein expression. The role of EIF3D, particularly its involvement in selective translation, has been recognized in the context of oncogenic and stress responses. On the other hand, we explicitly state that this study does not assert the importance of selective translation by EIF3D in primed PSCs. Recent studies have demonstrated the influence of EIF3D on the translational regulation of the MAPK pathway in commonly used cell lines, such as 293T and HeLa cells ([149]37, [150]55). These previous findings not only bolster our current results but also suggest a potential overlap of EIF3D targets in different cell types. The diverse abnormalities resulting from EIF3D depletion in different cell types may be due to the cellular context-dependent role of a particular pathway as a target of EIF3D. Together, the data from this and previous studies suggest that the regulation of the MAPK and p53 pathways, which are critical for proliferation in many cell types, is a universal rather than a selective role of EIF3D. The EIF3D target data in primed PSC provided in this work may serve as a resource for future research aimed at elucidating the selectivity of translational regulation by EIF3D. Recently, it has become possible to stop the proliferation of PSCs by inhibiting mTOR ([151]56, [152]57). This state, called diapause-like, slows proliferation while maintaining the proportion of cells in S phase. On the other hand, the decrease in proliferation caused by EIF3D KD in primed PSCs results in the loss of cells in S phase, which is different from the diapause-like state. In addition, while PSCs in the diapause-like state retained their differentiation potential, EIF3D depletion in primed PSCs resulted in failure to differentiate into the three germ layers. The differentiation failure of EIF3D KD PSCs may be due to proliferation arrest rather than its critical role in lineage commitment; thus, further studies are warranted to determine whether EIF3D directly contributes to differentiation potential. On the other hand, since EIF3D contributes to the regulation of protein expressions and their phosphorylation status in multiple pathways, including mTOR signaling, further research on the regulation of its activities (e.g., phosphatase) may reveal a comprehensive mechanism of proliferation control in PSC. Although EIF3D and EIF4G2 are known to interact ([153]26), the phenotype of EIF3D KD shown in this study differed from the previously reported phenotype of EIF4G2 KD in primed PSCs using the same experimental condition ([154]22). First, the time required for EIF3D KD to lose self-renewal was rapid (3 to 5 days), whereas that for EIF4G2 KD was relatively slow (6 to 12 days). Furthermore, while EIF4G2 was required for differentiation into NE but not EN and ME, EIF3D KD inhibited differentiation into all three germ layers. These results suggest that the simple interaction of EIF3D and EIF4G2 is not sufficient to maintain primed pluripotency. This study showed that both phosphorylation site mutants of EIF3D, which promote or inhibit Cap binding, can avoid the EIF3D KD phenotype in primed PSCs. Further research is warranted to clarify the detailed mechanism; this study provides the possible role of EIF3D in safeguarding pluripotency independent of Cap binding. Research on transcription factor networks and signaling pathways has advanced our understanding of pluripotency. This study highlights the importance of translational regulation as a link between these two aspects in pluripotency. Our CRISPRi screen indicated that, besides EIF3D, other translational regulators might play a role in maintaining primed pluripotency; deeper exploration into translational control will offer further insights into the fundamental nature of pluripotency. MATERIALS AND METHODS Experimental design To investigate the importance of translational regulation in primed PSCs, we performed functional analyses of EIF3D, which was identified through a functional screening process. We generated each of three independent lines of control and EIF3D KD iPSCs and used them for all comparative assays. In certain assays, the parental 1B4 iPSC line ([155]27) was also used. All experiments were performed with a minimum of two to three replicates with appropriate statistical analyses. Cell lines Human iPSC lines WTB6 and 1B4 (CRISPRi Gen 1 clone 4 derived from WTB6) were gifts from B. R. Conklin of Gladstone Institutes ([156]27, [157]58). HDFs derived from fetal (HDF1419) and adult (TIG-120) donors were acquired from Cell Applications Inc. and K. Kaji, respectively. The 293T/17 and HepG2 cells were purchased from American Type Culture Collection and Japanese Collection of Research Bioresources, respectively. Cell culture Human iPSC lines were maintained on tissue culture plates coated by iMatrix 511 silk (Matrixome) using StemFit AK02N media (Ajinomoto), as previously described ([158]59). For passaging, cells were washed once with Dulbecco’s phosphate-buffered saline (D-PBS; Nacalai Tesque) and incubated in TrypLE Express (Thermo Fisher Scientific) for 10 min at 37°C. Subsequently, cells were dissociated into single cells and washed in Dulbecco’s modified Eagle’s medium/Ham’s F-12 (DMEM/F-12; WAKO) containing 0.1% bovine serum albumin (BSA; WAKO). After cell counting and centrifugation, the cells were resuspended in StemFit AK02N media supplemented with iMatrix-511 silk (1.67 μg/ml) and 10 μM Y-27632 (Nacalai Tesque) ([159]60). G-banding tests conducted by Nihon Gene Laboratories confirmed that all PSC lines used in this study showed no apparent karyotypic abnormalities. HDFs, HepG2, and 293T/17 cells were maintained in DMEM (Nacalai Tesque) with 10% fetal bovine serum (Cosmo Bio). Routine testing confirmed the absence of mycoplasma infection. Transposon-mediated gene transfer We transfected 1 μg of a plasmid containing the inverted terminal repeats of either PiggyBac (PB) or Sleeping Beauty (SB) transposons, together with 0.5 μg of a plasmid encoding a hyperactive PB transposase (hyPBase) or SB transposase (SB100X), into 5 × 10^5 human PSCs. This was accomplished using the P3 Primary Cell 4D-Nucleofector X Kit S (Lonza) and Program CA-137 on the 4D Nucleofector device (Lonza). Two days posttransfection, the transfectants were selected with the appropriate drug until nontransfected cells were completely eradicated. Subsequently, single-cell–derived colonies that uniformly expressed the transduced fluorescent protein were isolated and expanded. CRISPR interference To generate inducible CRISPRi iPSC lines targeting a specific gene, we introduced a vector containing U6 promoter–driven sgRNA along with CAG promoter–driven fluorescence protein and a drug resistance marker into the 1B4 human iPSC line (passages 23 to 28) using the PB-mediated gene transfer method previously described ([160]27). Starting on day 2 posttransfection, drug selection was initiated and continued until nontransfected cells were eliminated. Subsequently, single-cell–derived colonies uniformly expressing the fluorescent protein were isolated and expanded. To induce KD, we administered Dox (1 μg/ml; WAKO) for the specified duration. KD clones within 20 passages postsubcloning were used for the study. The spacer sequences are listed in table S2. EN differentiation EN differentiation was conducted as previously described, with minor modifications ([161]61, [162]62). Primed PSCs were seeded at a density of 1 × 10^6 cells per well in iMatrix 511-coated six-well plates using StemFit AK02N media, supplemented with 10 μM Y-27632. The following day, cells were washed once with DMEM/F-12, and the media was replaced with differentiation media 1 (DM1) consisted of DMEM/F-12 (Thermo Fisher Scientific), 2% B27 supplement (Thermo Fisher Scientific), 1% MEM nonessential amino acids (NEAA; Thermo Fisher Scientific), and 0.1 mM 2-mercaptoethanol (2-ME; Thermo Fisher Scientific), supplemented with activin A (100 ng/ml; Nacalai Tesque), 3 μM CHIR99021 (Nacalai Tesque), basic fibroblast growth factor (bFGF; 20 ng/ml; Nacalai Tesque), and 50 nM PI-103 (Cayman Chemical). After 24 hours, the cells were washed with DMEM/F-12, and the medium was replaced with DM1 supplemented with activin A (100 ng/ml) and 250 nM LDN193189 (Stemgent). Two days later, following another wash with DMEM/F-12, the cells were cultured in DM1 with activin A (100 ng/ml) for an additional 48 hours. ME differentiation Directed differentiation to ME was carried out with minor modifications from previously described methods ([163]62, [164]63). A day before differentiation, primed PSCs were seeded at a density of 1 × 10^6 cells per well in iMatrix 511-coated six-well plates using StemFit AK02N media supplemented with 10 μM Y-27632. The following day, cells were washed once with DMEM/F-12 and then cultured in DM1 medium containing activin A (30 ng/ml), bone morphogenetic protein 4 (BMP4; 40 ng/ml; PeproTech), 6 μM CHIR99021, bFGF (20 ng/ml), and 100 nM PIK-90 (MedChemExpress) for 24 hours. Subsequently, after a wash with DMEM/F-12, the medium was replaced with DM1 supplemented with BMP4 (40 ng/ml), 1 μM A83-01, and 4 μM CHIR99021, and cells were maintained for an additional 48 hours. Then, the cells were washed once more with DMEM/F-12 and then cultured in DM1 medium supplemented with BMP4 (40 ng/ml) for another 48 hours. Ectoderm differentiation NE differentiation was conducted as previously described ([165]64, [166]65). A day before induction, primed PSCs were seeded at a density of 1 × 10^6 cells per well in iMatrix 511-coated six-well plates, using StemFit AK02N medium supplemented with 10 μM Y-27632. The following day, the cells were washed once with DMEM/F-12 and then cultured in Glasgow’s MEM (WAKO) containing 8% knockout serum replacement (KSR; Thermo Fisher Scientific), 1 mM sodium pyruvate (Sigma-Aldrich), 1% MEM NEAA, 0.1 mM 2-ME, 1 μM A83-01, and 250 nM LDN193189. This was maintained for 5 days, with daily media changes. Generation and maintenance of naïve PSCs Primed PSCs were converted to a naïve pluripotent state as previously described ([167]11). Before conversion, we maintained primed PSCs on γ-ray irradiated primary mouse embryonic fibroblasts (MEFs) in DFK20 media, composed of DMEM/F-12, 20% KSR, 1% NEAA, 0.1 mM 2-ME, and bFGF (4 ng/ml). For harvesting, cells were treated with CTK solution (ReproCELL) and dissociated into single cells. We then seeded 1.5 × 10^5 primed PSCs onto inactivated MEFs in a well of a six-well plate using DFK20 media supplemented with 10 μM Y-27632. The cells were incubated at 37°C in a hypoxic environment (5% O[2]). The following day, the media was replaced with NDiff227 (Takara) supplemented with 1 μM [168]PD325901 (Stemgent), leukemia inhibitory factor (LIF; 10 ng/ml; EMD Millipore), and 1 mM Valproic acid (WAKO). After 3 days, we switched the media to PXGL, consisting of NDiff227 supplemented with 1 μM [169]PD325901, 2 μM XAV939 (WAKO), 2 μM Gö6983 (Sigma-Aldrich), and LIF (10 ng/ml). Upon the emergence of round-shaped colonies, the cells were dissociated using a 1:1 mixture of TrypLE Express and 0.5 mM EDTA and then plated onto fresh inactivated MEF feeders in PXGL media containing 10 μM Y-27632. We replaced the media daily and passaged the cells every 3 to 5 days. The cells were used for assays after a minimum of 30 days postconversion. Differentiation of naïve PSCs to the primed state Before differentiating naïve PSCs into a primed state, we treated the cells, which were grown in PXGL media on iMatrix 511-coated plates, with Dox for 3 days. Subsequently, the media was replaced with StemFit AK02N, also supplemented with Dox, and the cells were incubated under normoxic conditions (20% O[2]). The media was changed daily. Treatment with chemical compounds and growth factors Before treatment, we seeded 1B4 iPSCs at a density of 2 × 10^5 cells per well in a six-well plate coated with iMatrix 511, using StemFit AK02N media supplemented with 10 μM Y-27632. On the following day, we replaced the media with StemFit AK02N containing various combinations of 10 μM SC79 (Sigma-Aldrich), 10 μM MHY1485 (MedChemExpress), 1 μM A83-01, 2 μM CHIR99021, EGF (50 ng/ml; WAKO), and 1 μM N3a (Sigma-Aldrich). As a control, cells were treated with DMSO at a 1/1000 volume ratio. The treatment was conducted for either 3 or 5 days, with daily media changes. RNA isolation and reverse transcription polymerase chain reaction Cells were washed once with D-PBS and lysed using QIAzol reagent (QIAGEN). Total RNA was extracted using the Direct-zol RNA Miniprep kit (Zymo Research), including on-column genomic DNA digestion as per the provided instructions. For reverse transcription, 1 μg of RNA was used, using the ReverTra Ace qPCR RT Master Mix (TOYOBO). qRT-PCR was conducted with gene-specific primers (refer to table S2) using either THUNDERBIRD Next SYBR qPCR Mix (TOYOBO) or TaqMan assays (Thermo Fisher Scientific) with TaqMan Universal Master Mix II, no UNG (Thermo Fisher Scientific) on a QuantoStudio 5 Real-Time PCR System (Applied Biosystems). Raw threshold cycle (Ct) values were normalized against ACTB or GAPDH expression using the delta-delta Ct method. Relative expression was then calculated as fold change relative to the control. Size-based protein analysis Cells were washed once with D-PBS and lysed using radioimmunoprecipitation assay (RIPA) buffer (Sigma-Aldrich) supplemented with a protease inhibitor cocktail (Sigma-Aldrich). The crude lysates were centrifuged at 15,300g for 15 min at 4°C, and the cleared supernatant was transferred to a new tube. The concentration of the cleared lysate was measured using a Pierce BCA Protein Assay Kit (Thermo Fisher Scientific) and an EnVision 2104 plate reader (PerkinElmer), following previously described methods. For quantitative and specific detection of target proteins, we used either a Wes or Jess automated capillary electrophoresis platform (ProteinSimple) with 12- to 230-kDa or 60- to 440-kDa Separation Modules (ProteinSimple). We loaded 2 μg of cell lysate per detection, along with the following antibodies (see also table S3): mouse monoclonal anti-OCT3/4 (1:500, Santa Cruz Biotechnology), goat polyclonal anti-SOX2 (1:40, R&D Systems), goat polyclonal anti-NANOG (1:40, R&D Systems), mouse monoclonal anti-p53 (DO-7) (1:200, Novus Biologicals), goat polyclonal anti-p53 (1:100, R&D Systems), rabbit monoclonal anti-p21 (1:50, Cell Signaling Technology), rabbit monoclonal anti-MDM2 (1:50, Cell Signaling Technology), rabbit polyclonal anti-EIF3D (1:250, Proteintech), rabbit polyclonal anti-eIF2α (1:50, Proteintech), rabbit polyclonal anti-phospho-eIF2α (Ser^51) (1:20, Cell Signaling Technology), rabbit polyclonal anti-SOX11 (1:500, Proteintech), rabbit polyclonal anti-KLF17 (1:200, Sigma-Aldrich), rabbit monoclonal anti-ERK1/2 (1:50, Cell Signaling Technology), rabbit monoclonal anti–phospho-ERK1/2 (Thr^202/Tyr^204) (1:50, Cell Signaling Technology), rabbit monoclonal anti-SMAD2 (1:50, Cell Signaling Technology), rabbit polyclonal anti–phospho-SMAD2 (Ser^245/250/255) (1:50, Cell Signaling Technology), rabbit monoclonal anti-mTOR (1:50, Cell Signaling Technology), rabbit monoclonal anti–phospho-mTOR (Ser2448) (1:50, Cell Signaling Technology), rabbit monoclonal anti-AKT (1:50, Cell Signaling Technology), rabbit monoclonal anti–phospho-AKT (Ser^473) (1:50, Cell Signaling Technology), rabbit polyclonal anti-p70S6K (1:50, Cell Signaling Technology), rabbit polyclonal anti–phospho-p70S6K (Thr^389) (1:50, Cell Signaling Technology), rabbit polyclonal anti–phospho-p70S6K (Thr^421/Ser424) (1:50, Cell Signaling Technology), rabbit monoclonal anti–β-catenin (1:50, Cell Signaling Technology), rabbit monoclonal anti–phospho-β-catenin (Ser^552) (1:50, Cell Signaling Technology), mouse monoclonal anti-puromycin (1:20, Developmental Studies Hybridoma Bank), rabbit polyclonal anti-RBBP6 antibody (1:50, Sigma-Aldrich), rabbit polyclonal anti-TCP1 (1:200, Proteintech), rabbit polyclonal anti-SSU72 (1:100, Proteintech), rabbit polyclonal anti-REST (1:100, Proteintech), rabbit polyclonal anti-KDM5C (1:250, Proteintech), rabbit polyclonal anti-WDR5 (1:100, Proteintech), rabbit polyclonal anti-WDR82 (1:50, Proteintech), rabbit monoclonal anti-VINCULIN (1:250, Cell Signaling Technology), and rabbit polyclonal anti–α tubulin (1:200, Proteintech). Data visualization and analysis were conducted using Compass for SW6.0 software (ProteinSimple). Immunocytochemistry The cells were washed once with D-PBS and fixed with 4% paraformaldehyde (Nacalai Tesque) for 15 min at room temperature. They were then blocked in D-PBS containing 1% BSA, 2% normal donkey serum (Sigma-Aldrich), and 0.2% Triton X-100 (Teknova) for 45 min at room temperature. Subsequently, the fixed cells were incubated overnight at 4°C with primary antibodies diluted in D-PBS containing 1% BSA. Following this, the cells were washed with D-PBS and incubated for 45 min at room temperature in 1% BSA containing fluorescence-conjugated secondary antibodies and Hoechst 33342 (1 μg/ml; Thermo Fisher Scientific). After a final wash in D-PBS, fluorescence was detected using a BZ-X810 imaging system (KEYENCE). Merged images were generated using a BZ-X Analyzer (KEYENCE). Nuclear size was quantified by analyzing Hoechst images with a Hybrid Cell Count Module (KEYENCE). The antibodies and their dilutions were as follows: mouse monoclonal anti-OCT3/4 (1:200), goat polyclonal anti-SOX2 (1:100), goat polyclonal anti-NANOG (1:100), goat polyclonal anti-p53 (1:200), rabbit monoclonal anti-p21 (1:400), goat polyclonal anti-SOX17 (1:100, R&D Systems), goat polyclonal anti-HAND1 (1:50, R&D Systems), rabbit polyclonal anti-PAX6 (1:100, BioLegend), Alexa 647 Plus anti-mouse immunoglobulin G (IgG; 1:500, Thermo Fisher Scientific), Alexa 647 Plus anti-rabbit IgG (1:500, Thermo Fisher Scientific), and Alexa 647 Plus anti-goat IgG (1:500, Thermo Fisher Scientific). Puromycin incorporation After washing the cells grown in three wells twice with prewarmed D-PBS, we added StemFit AK02N media containing cycloheximide (CHX; 100 μg/ml; Sigma-Aldrich) to one well and StemFit AK02N media alone to the other two wells. Following a 10-min incubation at 37°C, we added 1 μM puromycin (Thermo Fisher Scientific) to one well containing CHX-treated cells and to one of the two nontreated wells and then continued incubation for 30 min at 37°C. Postincubation, cells were washed with ice-cold D-PBS and lysed using RIPA buffer, supplemented with a protease inhibitor cocktail. Subsequently, the samples underwent size-based protein analysis as described previously. Cell cycle analysis As previously described ([170]66), we conducted cell cycle analysis using the Click-iT EdU Alexa Fluor 647 Flow Cytometry Assay Kit (Thermo Fisher Scientific). Cells seeded at a density of 5 × 10^5 cells per well in a six-well plate were cultured for 5 days in StemFit AK02N media supplemented with Dox. Subsequently, the cells were incubated in media containing 10 μM EdU for 135 min at 37°C. The cells were then harvested and washed with 1% BSA. Following centrifugation, the cells were resuspended in Click-iT fixative and incubated for 15 min at room temperature. After washing the fixed cells with 1% BSA, they were permeabilized with 1× Click-iT Perm/Wash reagent for 15 min at room temperature. For EdU detection, we added D-PBS containing Copper Protectant, Alexa Fluor 647 picolyl azide, and 1× Click-iT EdU buffer additive to the cell suspension. The samples were then washed with 1× Click-iT Perm/Wash reagent and stained with FxCycle Violet (1 μg/ml; Thermo Fisher Scientific) for 30 min at room temperature. We analyzed 1 × 10^4 cells using a FACS (fluorescence-activated cell sorting) Aria II (BD Biosciences) and BD FACSDiva software (BD Biosciences). EdU (detected with Alexa 647) and DNA (detected with FxCycle Violet) were analyzed using APC (650/660 nm) and Pacific Blue (405/455 nm) filters, respectively. Data analysis was conducted using FlowJo software (FlowJo LLC). Genome-wide CRISPRi screens Ten micrograms of the genome-wide CRISPRi library hCRISPRi-v2 (courtesy of J. Weissman; Addgene, #83969) which contains five sgRNA for each of 18,905 mRNAs along with 3.75 μg of psPAX2 (courtesy of D. Trono; Addgene, #12260) and 1.25 μg of pMD2.G (courtesy of D. Trono; Addgene, #12259) were transfected into 293T/17 cells (passage 27) using TransIT-Lenti Transfection Reagent (Mirus). Cells, plated at 5 million per 100-mm collagen I–coated dish, were transfected the day before. Two days posttransfection, the virus-containing supernatant was filtered through a 0.45-μm pore size polyvinylidene difluoride filter (Millipore), and lentiviral particles were concentrated using the Lenti-X Concentrator (Takara) and titrated using a Lenti-X qRT-PCR Titration Kit (Takara) as per the instructions. The lentivirus was then infected into 1B4 iPSCs (passage 23) at a multiplicity of infection of <0.4 (as determined by TagBFP fluorescence in the lentiviral vector) to achieve a coverage of >1000×. Three days postinfection, cells were selected with puromycin (1.5 μg/ml) until all noninfected cells perished ([171]28). Subsequently, the cells were plated at 10 million per 150-mm dish in StemFiT AK02N containing 10 μM Y-27632 and iMatrix-511 silk. The following day, the media was replaced with StemFiT AK02N supplemented with Dox. Cells were split every 2 to 3 days, maintaining a minimum of 100 million cells, corresponding to a 1000× coverage. Cells maintained without Dox (day 0) and those with Dox for 8 and 16 days were harvested, and SSEA-5 (+) cells were collected using an autoMACS Pro Separator (Miltenyi Biotec). We conducted the screening three times independently. Genomic DNA was purified from at least 100 million cells of each sample using NucleoSpin Blood XL (Takara) or QIAamp DNA Blood Midi Kit (QIAGEN). The purified DNA was digested overnight with SbfI-HF (New England Biolabs) and separated on a 0.8% Tris-acetate-EDTA (TAE) agarose gel. Postelectrophoresis, DNA fragments ranging from 350 to 700 bp were excised from the gel and purified using a QIAGEN gel extraction kit (QIAGEN). PCR and library preparation were conducted as previously described ([172]28). The libraries were sequenced using a NextSeq 500/550 High Output v2 Kit (Illumina) with custom primers, following the manufacturer’s protocol. Reads were aligned to the hCRISPRi-v2 sequences, counted, analyzed using MAGeCK (version 0.5.9.5), and then visualized using the MAGeCK flute (version 1.12.0) package in R (version 4.1.1) ([173]30, [174]31). GO analysis was conducted using clusterProfiler (version 4.2.2) ([175]67, [176]68). Polysome profiling The method used for polysome fractionation was based on a previously described method with minor modifications ([177]69). A single semiconfluent well of a 10-cm dish containing either control or sgEIF3D iPSCs was placed on a CoolBox XT Workstation (Biocision) to maintain a temperature of 4°C. This was followed by one gentle wash with 5 ml of ice-cold DPBS. The cells were then gently scraped and dissociated in 0.6 ml of ice-cold lysis buffer, consisting of 20 mM tris-HCl (pH 7.5), 150 mM NaCl, 5 mM MgCl[2] (Nacalai Tesque), 1 mM dithiothreitol (DTT; WAKO), a protease inhibitor cocktail, CHX (100 μg/ml), chloramphenicol (100 μg/ml), and 1% Triton X-100. The cell suspension was collected into a prechilled 1.5-ml DNA LoBind tube (Eppendorf). The lysate was incubated for 15 min on ice with TURBO deoxyribonuclease (DNase; 25 U/ml; Thermo Fisher Scientific) before centrifugation at 20,000g for 10 min at 4°C. The cleared supernatant was then transferred to a fresh 1.5-ml tube. The samples were rapidly frozen using liquid nitrogen and stored at −80°C. A continuous sucrose gradient ranging from 10 to 45% was prepared using 10 and 45% sucrose solutions (Sigma-Aldrich) in a 14 × 95 mm polyclear tube (Seton). The gradient was created in the presence of CHX (100 μg/ml) and 1 mM DTT in polysome buffer [25 mM tris-HCl, (pH 7.5), 150 mM NaCl, and 15 mM MgCl[2]] using the Biocomp Gradient Master program (Biocomp). Thawed cell lysates were measured for RNA concentration using the Qubit RNA BR Assay Kit (Thermo Fisher Scientific). A consistent volume of cell lysate containing 40 μg of RNA from each sample (300 μl) was layered onto the continuous sucrose gradient. The polysomes were separated by centrifugation in a Himac ultracentrifuge using a P40ST rotor (Himac) at 36,000 rpm for 2.5 hours at 4°C. The relative RNA abundance in ribosomal subunits, monosomes, and polysomes was detected using a 254-nm ultraviolet light with the Biocomp Piston Gradient Fractionator (Biocomp). Area under the curve was calculated using GraphPad Prism 8.0.2. KD via RNA interference To produce lentiviruses, pLKO.1-Blast (Addgene, #26655) encoding scramble shRNA (Addgene, #26701), EIF3D shRNA-1 or shRNA-2 (table S2) was cotransfected with psPAX2 and pMD2.G into 293T/17 cells as described above ([178]70). The collected viral supernatant was concentrated using a Lenti-X Concentrator and titrated using a Lenti-X qRT-PCR Titration Kit. These viral stocks were exposed to 293T/17 cells (passage 27), HDF1419 (passage 12), and HepG2 cells (passage 13) at 5000 viral particles per cell overnight at 37°C. Starting 2 days later, cells were selected with blasticidin S (10 μg/ml; Wako) until noninfected cells were completely dead. mRNA synthesis and luciferase assay For the luciferase reporter assay, a T7 promoter–driven NanoLuc luciferase (NLuc) was constructed. The 5′UTR sequences of RBBP6 (1061 bp from [179]NM_006910.5) and GAPDH (76 bp from [180]NM_002046.7) were inserted between the T7 promoter and NLuc using NEBuilder assembly cloning (New England Biolabs) according to the instructions. To prepare a control plasmid for normalization, we introduced a firefly luciferase (FLuc) gene into the linearized vector of the Takara IVTpro mRNA Synthesis System (Takara). Using these plasmids, mRNAs were synthesized according to the manufacturer’s protocol. We used CleanCap Reagent AG (Trilink BioTechnologies) as the cap analog and N1-methylpseudouridine 5′-triphosphate sodium salt (TCI Chemicals) instead of uridine triphosphate and performed quality control using a bioanalyzer (Agilent). After 2 days of Dox addition, the control and sgEIF3D iPSCs were replated at a density of 7.5 × 10^4 cells per well in iMatrix 511-coated 24-well plates with StemFit AK02N media, supplemented with 10 μM Y-27632 and Dox. The following day (day 3 post-Dox addition), we transfected the mRNA mix (125 ng each of NLuc reporter and FLuc control) into the cells using Lipofectamine RNAi Max reagent (Thermo Fisher Scientific). The next day, cells were lysed in 1× passive lysis buffer (Promega) and measured luciferase activities using a GloMax Discover Microplate Reader (Promega) and a Nano-Glo Dual-Luciferase Reporter Assay System (Promega). RNA sequencing Cells were lysed using QIAzol reagent, and total RNA was purified according to the protocol mentioned earlier. RNA quality was assessed using an Agilent RNA6000 Pico Kit on a Bioanalyzer 2100 (Agilent). The library preparation and subsequent analysis were carried out following methods outlined in previous studies ([181]71, [182]72). Briefly, 100 ng of DNase-treated total RNA was used for library preparation with the Illumina Stranded Total RNA Prep Ligation with Ribo-Zero Plus kit (Illumina). The libraries were evaluated using an Agilent High-Sensitivity DNA Kit (Agilent) and then sequenced using either a NextSeq 500/550 High Output v2 Kit (Illumina), NextSeq 1000/2000 P2 Reagents (100 cycles) v3 (Illumina), or HiSeq X (Illumina). The adapter sequence was trimmed using cutadapt-1.12 ([183]73). Reads mapping to ribosomal RNA (rRNA) were excluded using SAM tools (version 1.10) ([184]74) and Bowtie 2 (version 2.2.5) ([185]75). Reads were aligned to the hg38 human genome using STAR Aligner (version 2.7.10b) ([186]76). Quality checks were performed using RSeQC (version 4.0.0) ([187]77). Reads were counted with HTSeq (version 0.13.5) ([188]78) using the GENCODE annotation file (version 35) ([189]79). Counts were normalized using DESeq2 (version 1.34.0) in R (version 4.1) ([190]80). The DESeq2 package was also used to perform Wald tests. Principal components analysis and heatmaps were generated using prcomp and pheatmap, respectively. GO analysis was conducted and visualized using clusterProfiler ([191]67, [192]68). Hierarchical clustering was performed by using Euclidean distance and complete linkage method in R. Pearson’s correlation and P values of t test were calculated in R. Ribosome profiling Ribosome profiling was conducted as previously outlined ([193]81, [194]82). Cells were lysed in a buffer containing 20 mM tris-HCl (pH 7.5), 150 mM NaCl, 5 mM MgCl[2], 1 mM DTT, 1% Triton X-100, chloramphenicol (100 μg/ml), and CHX (100 μg/ml), followed by a 15-min DNase treatment on ice. RNA concentrations in the lysate were measured using the Qubit RNA BR assay kit (Thermo Fisher Scientific). We treated 10 μg of RNA with ribonuclease I (Epicentre) for 45 min at 25°C. The ribosome footprint RNA was then concentrated via ultracentrifugation using a sucrose cushion [20 mM tris-HCl (pH 7.5), 150 mM NaCl, 5 mM MgCl[2], 1 mM DTT, SUPERase-In (20 U/ml; Thermo Fisher Scientific), 1 M sucrose, chloramphenicol (100 μg/ml), and CHX (100 μg/ml)]. The resulting pellets were resuspended in pellet buffer [20 mM tris-HCl (pH 7.5), 300 mM NaCl, 5 mM MgCl[2], 1 mM DTT, 1% Triton X-100, and SUPERase-In (20 U/ml)] and purified using the Direct-zol RNA Microprep kit (Zymo Research). The RNA samples were separated by electrophoresis, and fragments ranging from 17 to 34 nt were excised and purified using Dr. GenTLE Precipitation Carrier (Takara). These purified ribosome footprint RNAs were ligated with linker oligonucleotides containing an inner index sequence and a unique molecular identifier (UMI), followed by rRNA depletion using riboPOOLs for Ribo-seq (siTOOLs Biotech). The residual RNAs were reverse-transcribed using ProtoScript II (New England Biolabs) and circularized with circLigase2 (Epicentre). The cDNA templates were amplified using Phusion polymerase (New England Biolabs) with index-sequenced primers. To calculate translational efficiency, corresponding RNA sequencing (RNA-seq) experiments were performed using RNA extracted from the lysis buffer. We used TRIZOL LS reagent (Thermo Fisher Scientific) and the Direct-zol RNA Microprep kit for RNA extraction. The RNA-seq libraries were prepared as instructed by manufacturer’s protocol, except using riboPOOLs for RNA-seq (siTOOLs Biotech) in the rRNA depletion step and xGen UDI-UMI Adapters (Integrated DNA Technologies) in the adapter ligation step. The cDNA libraries were sequenced following the RNA-seq protocol. The reads were demultiplexed using the inner index, and adapters were removed using fastp (version 0.22.0) and fastx-split ([195]81, [196]83). To filter out reads mapping to rRNA, Bowtie2 and SAMtools were used. The remaining reads were aligned to the human genome (hg38) using STAR (version 2.7.10b), and duplicates were removed on the basis of UMI using bam-suppress-duplicates. Quality control statistics were calculated using fp-framing. Read counting and normalization were performed using fp-count and DESeq2, respectively ([197]81). TE and fold change values were calculated using the average values of replicates as follows (i indicates a gene) [MATH: TEi=Ribosome profiling averaged normalized countiRNA sequencing averaged normalized counti :MATH] [MATH: Fold changei=TEi of sgEIF3DTEi of control :MATH] For identifying differentially translated transcripts, we used DESeq2, using a likelihood ratio test (full model: experiment + target + experiment:target; reduced model: experiment + target). In pathway enrichment analysis, Enrichr was used with the WikiPathways_2021_Human database. To analyze upstream, downstream, and newly identified ORFs, we first obtained a bed file using ORF-RATER with ribosome profiling data of the parental iPSC line WTB6, treated with CHX and harringtonine ([198]58, [199]84). Using this bed file, reads from control and sgEIF3D (Dox + 3 days) samples were counted by fp-count. We then conducted a statistical analysis to identify significantly different transcripts between these conditions, using DESeq2 with a Wald test. Statistics The quantitative measurement results are presented as individual data points, depicted by colored dots, with means indicated by bars. In some instances, bar graphs with individual data points and error bars representing SDs are used. Statistical analyses included unpaired two-tailed t tests to calculate P values, assessing differences between two groups. Furthermore, one-way analysis of variance (ANOVA) was used for multiple comparisons. These analyses were performed using GraphPad Prism version 8.0.2 (GraphPad) and Excel (Microsoft). Statistical significance was determined by P values or adjusted P values less than 0.05, denoted by asterisks in the figures. The specific values are detailed in the figure legends. Supplementary Material 20240409-1 [200]sciadv.adq5484.v1.pdf^ (9.8MB, pdf) Acknowledgments