(C) PLOS One This story was originally published by PLOS One and is unaltered. . . . . . . . . . . The RNA helicase DDX6 controls early mouse embryogenesis by repressing aberrant inhibition of BMP signaling through miRNA-mediated gene silencing [1] ['Jessica Kim', 'Department Of Biological Sciences', 'Graduate School Of Science', 'The University Of Tokyo', 'Tokyo', 'Masafumi Muraoka', 'Mammalian Development Laboratory', 'Department Of Gene Function', 'Phenomics', 'National Institute Of Genetics'] Date: 2022-12 The evolutionarily conserved RNA helicase DDX6 is a central player in post-transcriptional regulation, but its role during embryogenesis remains elusive. We here show that DDX6 enables proper cell lineage specification from pluripotent cells by analyzing Ddx6 knockout (KO) mouse embryos and employing an in vitro epiblast-like cell (EpiLC) induction system. Our study unveils that DDX6 is an important BMP signaling regulator. Deletion of Ddx6 causes the aberrant upregulation of the negative regulators of BMP signaling, which is accompanied by enhanced expression of Nodal and related genes. Ddx6 KO pluripotent cells acquire higher pluripotency with a strong inclination toward neural lineage commitment. During gastrulation, abnormally expanded Nodal and Eomes expression in the primitive streak likely promotes endoderm cell fate specification while inhibiting mesoderm differentiation. We also genetically dissected major DDX6 pathways by generating Dgcr8, Dcp2, and Eif4enif1 KO models in addition to Ddx6 KO. We found that the miRNA pathway mutant Dgcr8 KO phenocopies Ddx6 KO, indicating that DDX6 mostly works along with the miRNA pathway during early development, whereas its P-body-related functions are dispensable. Therefore, we conclude that DDX6 prevents aberrant upregulation of BMP signaling inhibitors by participating in miRNA-mediated gene silencing processes. Overall, this study delineates how DDX6 affects the development of the three primary germ layers during early mouse embryogenesis and the underlying mechanism of DDX6 function. Gene expression occurs through two steps: transcription (DNA to RNA) and translation (RNA to protein). Cells have highly sophisticated regulatory processes working on various levels for accurate gene expression. Post-transcriptional regulation, which includes all RNA-related controls, is crucial because it enables fine-tuning and rapid alteration of gene expression. RNA-binding proteins and non-coding RNAs are the two main players in post-transcriptional regulation. DDX6, the subject of our study, is an RNA-binding protein, more specifically an RNA helicase, which can unwind or rearrange RNA secondary structures. Its diverse molecular and cellular functions have been reported, but its embryogenic role is unknown. Here, we describe DDX6 function during early mouse embryogenesis and the underlying mechanism using genetic methodology. DDX6 enables proper cell lineage specification of pluripotent stem cells by mainly regulating BMP signaling through miRNA-mediated gene silencing. As DDX6-mediated RNA regulation affects signaling pathways, the loss of DDX6 has a wide impact on developmental processes from pluripotency to embryo patterning. In addition to showing the developmental role of DDX6, we succeeded in the modular segregation of its various RNA-regulatory pathways. Considering the presence of DDX6 in diverse contexts, such as cancer, virus infection, and stem cells, this new knowledge forms a foundation for DDX6 being a good therapeutic target. However, these examinations were conducted on ESCs or very limited cell types. To deepen our understanding of the role of DDX6 as a key post-transcriptional regulator during early mouse embryogenesis, we examined Ddx6 KO embryos. Gastrulation stages were investigated in embryos and the earlier time points were assessed using an ESC-to-EpiLC induction model. This study revealed that DDX6 exerts potent effects on the development of the three primary germ layers by preventing aberrant inhibition of the BMP signaling pathway. Furthermore, through genetic dissection of the DDX6 pathways, we found that DDX6 works through the miRNA pathway, but P-bodies are dispensable during early development. There are a few studies that examined the developmental roles of DDX6 in vivo. DDX6 function is crucial for maintaining adult spermatogonial stem cells [ 45 ] and for helping NANOS2 to induce male-type germ cell differentiation in embryonic testes [ 46 ]. During female germ cell development, deletion of Ddx6 causes defects in primordial follicle formation [ 47 ]. Two previous studies investigated the role of DDX6 in mouse and human pluripotent cells. In mESCs, DDX6 is necessary for translational repression of miRNA targets and Ddx6 knockout (KO) cells exhibited similar phenotypes to Dgcr8 KO ESCs, which lack all miRNAs [ 48 ]. Another study elucidated the relationship between stem cell potency and P-body-mediated translational regulation. As an essential factor of P-body formation, once DDX6 was depleted, P-bodies were disassembled and translationally suppressed target mRNAs, including many transcription factors and chromatin regulators, re-entered the translation pool. The resulting increased expression of target genes altered chromatin organization and made both human and mouse primed ESCs become more naively pluripotent and resistant to differentiation [ 20 ]. From the 8-cell stage, transcription factors and signaling pathways play a major role in cell fate determination [ 32 ]. The transforming growth factor-β (TGF-β) superfamily is one of the major signaling pathways involved in early mammalian development. Bone morphogenetic protein (BMP) and Nodal belong to different subgroups, and they utilize distinctive serine/threonine kinase receptors for signal transduction. Activated receptors phosphorylate downstream intracellular mediators, receptor SMADs 1, 5, and 8 for BMP signaling, and SMADs 2 and 3 for Nodal signaling [ 33 , 34 ]. BMP and Nodal signaling are known to mutually antagonize each other, and this antagonism is generally thought to occur through competition for the common signal mediator SMAD4 [ 35 – 38 ]. BMP signaling has multiple roles during early post-implantation development. It is required for extra-embryonic mesoderm formation [ 39 ], primordial germ cell (PGC) induction [ 40 ], mesoderm development, and patterning [ 41 ], and inhibiting premature neural differentiation [ 42 ]. BMP signaling is dispensable for ESC self-renewal but is required for proper differentiation [ 43 , 44 ]. The Nodal pathway exerts its influence when ESCs transition from naive pluripotency. Nodal is important for securing primed pluripotency with the capacity to differentiate into multiple lineages [ 30 ]. Gastrulation is a milestone in embryogenesis because the primary germ layers that give rise to all cell types develop during this developmental event. The three germ layers of the embryo, the ectoderm, mesoderm, and endoderm, basically originate from the inner cell mass (ICM) of the blastocyst. Pluripotent embryonic stem cells (ESCs) can be derived from the ICM of E3.5 early blastocysts or the epiblast of E4.5 late blastocysts [ 24 – 27 ]. Cells at these stages are in the naive (ground) pluripotent state. Soon after implantation, naive epiblasts differentiate into the primed pluripotent state in which cells become capable of committing to a certain lineage during gastrulation [ 28 – 30 ]. The epiblast-like cells (EpiLCs) permit more precise staging of mouse pluripotent cells. Transcriptomic analysis demonstrated that EpiLCs, induced from ESCs, have similar properties to the epiblast of post-implanted, pre-gastrulating (E5.5~6.0) embryos [ 31 ]. These in vitro systems enable detailed examination of pre- and early post-implantation embryos, which are normally difficult to investigate in vivo due to their small size. DDX6 participates in many aspects of RNA metabolism: processing body (P-body) formation [ 7 , 8 ], stress granule assembly, mRNA storage, mRNA decay [ 9 ], translational repression [ 10 , 11 ], microRNA (miRNA) pathway [ 12 , 13 ], and translational promotion [ 14 , 15 ]. P-bodies are membrane-less cytoplasmic mRNP granules thought to be the place of the storage or decay of translationally inactive mRNAs [ 13 , 16 ]. Many translational repression and decapping-related proteins, miRISC components, and translationally repressed mRNAs are accumulated in P-bodies. DDX6 is necessary for the formation and maintenance of P-bodies [ 17 ]. The helicase core of DDX6 and all other DEAD-box RNA helicases is composed of two RecA-like globular domains and they share conserved motifs [ 6 ]. The characteristic DEAD (Asp-Glu-Ala-Asp) sequence, which has helicase activity in vitro [ 18 ], is in Motif II. Motif VI is known to be associated with conformational changes coupled with ATP hydrolysis and unwinding activity [ 19 ]. DEAD helicase activity is necessary for P-body assembly in human iPSCs [ 20 ]. The tethering assay using only the RecA2 domain of Xenopus homologue p54 showed that this domain alone can repress the translation of target mRNAs without defects in P-body localization, but it was unable to form de novo P-bodies [ 8 ]. Another important finding was the functional defects of different helicase mutants were dependent on the changes in the interaction with other gene-repressing proteins. This is consistent with the notion that DDX6 acts as a partner protein of numerous RNA-regulatory proteins and a remodeler of mRNP complexes. As RNA helicases mostly interact with the sugar-phosphate backbone of RNAs, the target specificity comes from cofactors [ 5 , 21 , 18 ]. Regarding its mRNA decay functions, DDX6 predominantly interacts with 5’-to-3’ decay proteins [ 17 ]. In this direction of mRNA decay, decapping of the m7G cap structure is the rate-limiting step. Decapped mRNAs are finally degraded by the 5’ monophosphate-dependent 5’-to-3’ exoribonuclease, Xrn1 [ 22 , 23 ]. Although many molecular and cellular studies on DDX6 have been reported, investigation of its functions in embryonic development is limited. Post-transcriptional regulation, located in the middle layer of gene expression, is a critical controlling point where many mRNA regulatory processes occur. RNA-binding proteins (RBPs) and non-coding RNAs are the two main players [ 1 , 2 ]. Among diverse RBPs, RNA helicases are characterized by their wide range of involvement in RNA metabolism by binding RNA or remodeling ribonucleoprotein complexes (RNPs) [ 3 , 4 ]. DEAD box proteins compose the largest RNA helicase family sharing the Asp-Glu-Ala-Asp (DEAD) motif and having ATP-dependent RNA unwinding activity [ 5 ]. Mice have 43 DEAD box RNA helicases. Among them, we focused on DDX6 (Rck/p54 in human, Xp54 in Xenopus, Me31B in Drosophila, Cgh-1 in C. elegans, and Dhh1 in S. cerevisiae), an evolutionarily conserved RNA helicase throughout eukaryotes [ 6 ]. As Ddx6 KO and Dgcr8 KO had similar transcriptomic changes in both ESC and EpiLC stages, we further examined their similarity in embryo samples. We generated Dgcr8 KO embryos by injecting Dgcr8 targeting sgRNAs into eggs via electroporation. Dgcr8 KO embryos were smaller and more malformed than Ddx6 KO embryos ( Fig 6C ), but we were able to find common defects with Ddx6 KO: ectopically expanded and increased Nodal and Eomes expression in E7.5 embryos ( Fig 6D ). Unlike Ddx6 KO, which exhibited a delay in primitive streak induction, Dgcr8 KO embryos had a relatively normal and strong Brachyury induction at E7.5 despite their small body size. We also conducted detailed analyses during ESC-to-EpiLC differentiation through qRT-PCR. Dgcr8 KO cells exhibited similar characteristics to Ddx6 KO cells: enhanced pluripotency and stronger expression of the neural lineage-inducing factors with a decreased differentiation capacity to the mesendoderm lineage ( Fig 6E ). As we identified that repression of BMP signaling is the primary change in cellular condition of Ddx6 KO, we examined Dgcr8 KO for a similar change. Dgcr8 KO cells also had upregulated expression of the negative regulators of BMP signaling, and pSMAD1/5 target genes were de-repressed. Moreover, GSEA of ESC RNA-seq data revealed that ‘the negative regulation of BMP signaling’ gene set (Fbn1, Grem2, Grem1, Wnt5a, Htra1, Sorl1, Fstl3, Bmper, Nbl1, Htra3, Crim1, Rbpms2, Spart) was highly upregulated only in Ddx6 KO and Dgcr8 KO ESCs, but not in Eif4enif1 KO and Dcp2 KO ( Fig 6F ). As such, DDX6-mediated RNA regulation and miRNA-mediated gene silencing share a common role, especially in preventing aberrant upregulation of the negative regulators of BMP signaling. (A) PCA plot of the ESC and EpiLC Day2 samples of each genotype group. (B) Summary of the GSEA results of ESC RNA-seq data. The gene sets that were changed in Ddx6 KO, Dgcr8 KO, Eif4enif1 KO, and Dcp2 KO were compared. (C) Dgcr8 KO embryos exhibited similar morphological defects to Ddx6 KO embryos at E7.5, but they became more malformed at E8.5 (Scale: 100 μm for E7.5; 200 μm for E8.5). (D) Whole-mount ISH of E7.5 Dgcr8 KO embryos with Nodal, Eomes, and Brachyury probes (Scale: 100 μm, n = 3 for Nodal, n = 4 for Eomes, n = 5 for Brachyury). (E) Comparison of gene expression between Ddx6 KO and Dgcr8 KO. qRT-PCR analysis of several key genes during the EpiLC induction period. Each bar represents the relative expression of KO cells to WT cells at the indicated time point. Mean ± SEM. Student’s t-test (n ≥ 3) (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001). (F) GSEA enrichment plot of the “negative regulation of BMP signaling pathway” gene set in four mutant ESCs. Black bars represent the position of the genes that belong to this gene set (n = 45) in the whole ranked gene list. The green line shows the overall distribution of this gene set (whether over-represented at the top (left) or bottom (right) of the ranked list of genes). To assess the effects on the transcriptomic landscape over the time course of development, we generated cDNA libraries of ESC and EpiLC Day2 stage from all mutant groups and WT. Principle component analysis (PCA) demonstrated that Ddx6 KO (Ddx6 △/△ ) is greatly different from WT, Eif4enif1 KO, and Dcp2 KO, but highly similar to Dgcr8 KO ( Fig 6A ). Eif4enif1 KO and Dcp2 KO, whose protein functions directly affect P-body functions, resulted in similar transcriptomes. Eif4enif1 KO, in which translational repression on transcripts is disrupted, resulted in the disassembly of P-bodies, whereas Dcp2 KO caused the enlargement of P-bodies due to the blockage of mRNA degradation ( S3D Fig ). Even though their transcriptomes were different from WT, gastrulation occurred normally in KO embryos ( S5A Fig ). Based on the above, we concluded that P-bodies are dispensable until the peri-gastrulation stage. Disruption of P-body functions altered the transcriptome of cells, but the ultimate differentiation capacity of pluripotent stem cells was not affected. We examined differentially expressed genes in detail through gene set enrichment analysis (GSEA). A greater number of gene sets was affected in Ddx6 KO and Dgcr8 KO than in Eif4enif1 KO and Dcp2 KO ( Fig 6B ). The high portion of the differentially expressed gene sets of Ddx6 KO ESCs was shared with Dgcr8 KO ESCs (87% of upregulated, 84% of downregulated), indicating that DDX6 function is largely inclusive to the miRNA pathway. To identify the most affected gene sets, we filtered with adjusted p-value 0.05 and then aligned in order of normalized enrichment score (NES). The list of Top10 upregulated and downregulated gene sets in three different groups is shown in Table 2 : Ddx6 KO and Dgcr8 KO only (regulated by miRNAs/ P-body-independent), all four KO groups (regulated by miRNAs/P-body-dependent), Eif4enif1 KO and Dcp2 KO only (regulated by P-body functions/miRNA-independent). We cannot go over these lists in detail, but they provide useful information for the functions of each pathway (Tables 2 and S1 – S5 ). Next, we aimed to identify which DDX6 pathway is most important during early development. DDX6 functions as a hub of post-transcriptional regulation. Due to its wide range of involvement, it is difficult to pinpoint which pathway is responsible when DDX6 is depleted. Therefore, we individually disrupted three main DDX6-associated pathways by knocking out a key gene of each pathway ( S3C Fig ). Translational repression along with P-body formation was impaired in Eif4enif1 KO, 5’-to-3’ mRNA degradation was impaired in Dcp2 KO, and miRNA-mediated gene silencing was disrupted in Dgcr8 KO [ 64 – 67 , 17 ]. (A) Tamoxifen was injected at E6.5. Whole-mount DDX6 immunostaining confirmed that the complete depletion of DDX6 takes approximately 1 day (Scale: 30 μm, n = 3). DDX6 in green, DAPI in blue. (B) E8.5 cKO embryos exhibited similar phenotypes to conventional KO embryos (Scale: 500 μm for group, 100 μm for KO17, 200 μm for KO12) (n = 6). (C) qRT-PCR analysis of several key genes in Ddx6 cKO E8.5 embryos. Embryos exhibiting similar morphology to the KO12 were used for analysis. Mean ± SEM. Significance was calculated by the Wilcoxon rank-sum test (n = 9~10) (* α = 0.05 significance level, ** α = 0.01). To further ask whether the aberrant activation of BMP signaling inhibition is a primary property of Ddx6 △/△ cells, we conditionally deleted Ddx6 using the Rosa-CreER T2 ; Ddx6 flox/flox mouse line. As we expected the loss of DDX6 to cause mesoderm formation defects, we removed DDX6 during gastrulation. We first examined the time required for the complete depletion of DDX6. When tamoxifen was administered to the pregnant female via oral gavage at E6.5, Ddx6 deletion and depletion of existing DDX6 proteins were completed by E7.5 ( Fig 5A ). We then injected tamoxifen at E6.5 and collected embryos at E8.5 to examine their phenotypes. Conditional knockout (cKO) embryos of the same litter exhibited variable phenotypes like conventional KO embryos ( Fig 5B ). A few had marked posterior truncation (KO2, KO17). The others developed the mid-to-posterior body part, but it was shorter and smaller, and the head and heart were abnormal (KO7, KO12). Although the phenotypes were milder than those of conventional KO embryos, conditional KO embryos also demonstrated characteristic gene expression of Ddx6 mutants. The expression of the negative regulators of BMP (Chrd, Noggin, Lgals9, Xdh, Pai), Nodal, and Eomes increased after the depletion of DDX6 ( Fig 5C ). Therefore, along with the features of Ddx6 △/△ ESCs, this conditional KO experiment revealed that these characteristic gene expression changes are closely related to the absence of DDX6 rather than the cumulative result of indirect effects. It also reconfirmed that DDX6 is essential during gastrulation and that the loss of DDX6 makes cells activate inhibitory regulation of BMP signaling. BMP signaling is important to prevent the differentiation of ESCs to the neuronal lineage [ 62 ]. Thus, the strong inclination of Ddx6 △/△ ESCs toward the neuronal cell fate suggests that this brake is nonfunctional. We investigated whether the SMAD-dependent BMP signaling pathway was repressed in Ddx6 △/△ cells. In ESCs and the initial differentiation stage, BMP signaling has a transcriptional repressive role for these SMAD1/5 target genes [ 43 ]. We examined the expression pattern of known SMAD1/5 target genes in ESCs, and found that the expression of Accn4, Alx3, Dpysl2, and Kdm6b was higher in Ddx6 △/△ cells ( Fig 4H ). This suggested that target genes were transcriptionally de-repressed due to reduced BMP signaling. In particular, this characteristic higher expression was prominent on EpiLC Day1, which corresponds to the earliest differentiation state. DPYSL2 and the H3K27 demethylase KDM6B are early neural differentiation regulators [ 63 , 43 ]; therefore, their high expression is consistent with the phenotype of Ddx6 △/△ ESCs preferring neural lineage commitment. We next investigated whether aberrant upregulation of a set of BMP signaling inhibitors also occurred in Ddx6 △/△ pluripotent cells. Like Ddx6 △/△ embryos, Ddx6 △/△ ESCs exhibited a significant increase in the expression of negative regulators of the BMP pathway during EpiLC induction ( Fig 4I ). Lastly, we confirmed that this increased expression of BMP inhibitors indeed reduced BMP signaling via Western analysis ( Fig 4J ). The p-SMAD1/5 protein amount was significantly reduced in Ddx6 △/△ ESCs. There was no difference in SMAD5 protein amount, and transcript levels of the BMP signaling components, such as ligands, receptors, and Smad mediators, were similar ( S2J Fig ). Along with suppressed BMP signaling, Ddx6 △/△ ESCs displayed high expression levels of Nodal and Nanog, analogous to Ddx6 △/△ embryos ( Fig 4D and 4E ). We hypothesized that Nodal signaling was promoted in Ddx6 △/△ ESCs, but we did not observe a significant difference in the amount of p-SMAD2 ( S4A Fig ). In conclusion, the common features of Ddx6 △/△ ESCs and embryos suggest that suppression of BMP signaling with an increased Nodal expression occurs when DDX6 is absent. As Ddx6 △/△ blastocysts and ESCs did not exhibit notable abnormalities, we examined their next developmental capacity. We conducted ESC-to-EpiLC induction to mimic natural in vivo development. The transition of the naive ground state ESCs to the primed pluripotent state EpiLCs takes two days and EpiLC Day1 is regarded as a transition state that exhibits a distinctive open chromatin landscape and transcriptome [ 60 ]. During EpiLC induction, the difference in cell number between WT and Ddx6 △/△ cells markedly increased ( Fig 4C ). We then investigated the expression pattern of several key genes during EpiLC induction by qRT-PCR. First, we checked well-known epiblast markers Fgf5 and Nodal, and an important regulator of the transition state, Zic3, which exhibits peak expression on EpiLC Day1 [ 60 ]. Ddx6 △/△ cells had significantly higher expression of Nodal and Zic3, but there was no difference in Fgf5 expression ( Fig 4D ). After confirming induction, we examined the expression profile of pluripotency and early differentiation-related genes. As shown in Fig 4B , Ddx6 △/△ ESCs exhibited slightly higher expression of pluripotency genes and this difference increased during EpiLC induction ( Fig 4E ). They had much higher expression of naive pluripotency markers (Klf4, Rex1), suggesting that even though an overall transition was made to the EpiLC state, cells failed to completely exit from the ground state. We also noted a change in the differentiation capacity of Ddx6 △/△ cells. Neural lineage-inducing genes, such as Sox1, Sox2, and Pax6, were highly upregulated in Ddx6 △/△ cells, whereas the mesendoderm lineage inducer T was significantly downregulated ( Fig 4F ). We also conducted a monolayer differentiation experiment. ESCs favor neuronal differentiation in low-density, serum-free, and feeder-free culture conditions [ 61 ]. Compared with WT ESCs, Ddx6 △/△ ESCs differentiated and developed into neurons quickly. Ddx6 △/△ cells exhibited stronger expression of TuJ1 with the morphology of well-developed dendrites and axons on differentiation Day1 ( Fig 4G ). This was similar to the premature neural differentiation observed in E8.5 Ddx6 △/△ embryos. Taken together, Ddx6 △/△ embryos developed normally until the blastocyst stage and ESCs had no defects in self-renewal. However, the differentiation capacity of Ddx6 △/△ pluripotent cells was strongly skewed to neuronal lineage commitment. (A) Cell counting of ESCs over a three-day culture period. Mean ± SEM. Significance was calculated by the Student’s t-test (n = 5). (B) qRT-PCR examining the relative expression of pluripotency markers in Ddx6 △/△ ESCs to WT ESCs. Mean ± SEM. Student’s t-test (n = 7~9). (C) Cell counting during the ESC-to-EpiLC induction period. Mean ± SEM. Student’s t-test (n = 13 for WT, n = 7 for Ddx6 △/△ ). (D) qRT-qPCR examining the expression pattern of Nodal, Fgf5, and Zic3. Mean ± SEM. Student’s t-test (n = 9 for Nodal & Fgf5, n = 7 for Zic3) (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001). (E-F, I) qRT-PCR analysis of the expression trend of several key genes during the EpiLC induction period. Each bar represents the relative expression of Ddx6 △/△ cells to WT cells at the indicated time point. Mean ± SEM. Student’s t-test. (E) Major pluripotency genes (n = 7~9). (F) Early neuroectoderm and mesendoderm lineage markers (n = 7~9). (I) The negative regulators of BMP signaling (n = 6~9). (G) TuJ1 ICC (immunocytochemistry) on Day1 of monolayer differentiation (Scale: 50 μm) (n = 3). (H) qRT-PCR analysis of the expression of the known BMP-SMAD1/5 target genes in ESCs. Each value is the relative expression to WT ESCs (n = 5). (J) Western blot for endogenous SMAD5 and phosphorylated SMAD1/5 in mESCs. Four conditions: LDN (LDN193189: BMP Type I receptor ALK2/3 inhibitor)/—(non-treated)/ BMP4 1/10 (1/10 dilution of rBMP4 treated sample)/ BMP4 (rBMP4 treated). The quantified signal intensity of the band is displayed on the right-side graph (n = 6). We examined E8.5 Ddx6 △/△ embryos and considered repressed BMP signaling as a major cause of their developmental defects. We then looked for the earliest time point of inhibited BMP signaling in Ddx6 △/△ . There were no morphological abnormalities until E3.5 blastocysts and ESCs were successfully established. DDX6 was highly expressed in ESCs and EpiLCs in P-bodies ( S3A Fig ), which were disassembled in Ddx6 △/△ cells ( S3B Fig ). The proliferation rate of Ddx6 △/△ ESCs was lower ( Fig 4A ), but they had no defects in maintaining pluripotency over many passages. However, like E8.5 Ddx6 △/△ embryos, they expressed higher levels of pluripotency genes, such as Oct4, Nanog, and Sox2, than WT ESCs ( Fig 4B ). Of note, the expression pattern of TuJ1 (TUBB3) was abnormal, being another indication of the failure of Ddx6 △/△ embryos exiting from the pluripotent state ( Fig 3D ). In E6.5 WT embryos, low but positive TuJ1 expression was detected in the epiblast and its expression increased by E7.5. In E7.5 embryos, T-positive mesendoderm progenitors retained TuJ1 expression, but TuJ1 expression was eventually turned off with the progression of differentiation and only remained in the neuroepithelium. This suggests that a well-known marker of neuroectoderm and neurons, TuJ1, is also expressed in the primed pluripotent epiblast. In Ddx6 mutants, the expression level of TuJ1 was much higher than in WT at all time points. The stronger expression in Ddx6 KOs may have resulted from the promoted pluripotency and premature neuroectoderm differentiation. Enhanced Nodal expression also affected pluripotency. Nodal signaling is important for regulating primed pluripotency. Activin/Nodal signaling is necessary to induce Nanog transcription in mEpiSCs [ 58 ]. NODAL is also required to activate and maintain Nanog expression in the proximal epiblast of peri-gastrula mouse embryos [ 59 ]. Therefore, we examined whether the expression of this other NODAL target gene also increased in Ddx6 △/△ embryos. The core pluripotency factors Nanog and Pou5f1 (Oct4) and naive pluripotency marker Klf4 were upregulated in Ddx6 △/△ ( S2I Fig ). The retained expression of the naive pluripotency-specific genes in E8.5 embryos suggested that exit from the ground pluripotent state did not occur properly in Ddx6 mutants. We examined NANOG expression by section IHC ( Fig 3C ). In WT embryos, NANOG expression was strongest in the primitive streak at E6.5, but at E7.5, its expression moved anteriorly, and the posterior part was negative. Contrarily, E7.5 Ddx6 △/△ embryos exhibited strong NANOG expression in the posterior epiblast and this expression was maintained at E8.5. (A-B) Whole-mount ISH of E7.5 & E8.5 embryos with (A) a Nodal probe (Scale: 100 μm, n = 10 for E7.5; 200 μm, n = 10 for E8.5) and (B) an Eomes probe (Scale: 100 μm, n = 11 for E7.5; 200 μm, n = 10 for E8.5). Expressions of Nodal and Eomes were maintained in E8.5 Ddx6 KO embryos, which were younger than the head-fold stage (left), but down-regulated as in WT at the head-fold stage (right). (C) E6.5~E8.5 embryo frozen section IHC for NANOG (Scale: 50 μm for E6.5, n = 2; 100 μm for E7.5, n = 3; 100 μm for lower magnification of E8.5, 50 μm for higher magnification, n = 3). Square parts are enlarged in the right panels. (D) TuJ1 expression in the epiblast. E6.5~E8.5 embryo frozen section IHC for TuJ1 & T (BRACHYURY). The signal intensity is comparable only between the same embryonic day samples. (Scale: 50 μm for E6.5, n = 2; 100 μm for lower magnification, 50 μm for higher magnification, n = 3 for E7.5 & E8.5). Square parts are enlarged in the right panels. Another indication of suppressed BMP signaling in Ddx6 △/△ embryos was increased Nodal expression ( S2F Fig ). BMP and Nodal signaling are often in a competitive relationship and can suppress each other. During gastrulation, a gradient of NODAL activity patterns the primitive streak and allocates mesendoderm progenitors. NODAL and its downstream target EOMES together define the anterior primitive streak (APS), from which cardiac mesoderm and definitive endoderm progenitors are specified [ 53 – 56 ]. WISH demonstrated that Nodal expression was confined to the node in E7.5 WT embryos, but its expression was highly spread over the proximal-posterior region in Ddx6 △/△ embryos ( Fig 3A ). The expression was eventually restricted to the node at E8.5 in mutants in the head fold stage, but one mutant without a head fold retained high Nodal expression. Eomes is highly expressed in the extraembryonic ectoderm and the posterior part of the epiblast at E6.5. Its expression moves distally to the primitive streak at E7.5 [ 57 ], and is reduced by E8.5 in the WT embryos. However, some E8.5 Ddx6 △/△ embryos retained high-level expression in the primitive streak ( Fig 3B ). The expression pattern in E7.5 KO embryos resembled that of earlier stage (~E6.5) control embryos. Some E8.5 Ddx6 △/△ embryos were morphologically similar to the control E7.5 embryos, but expression levels of Nodal and Eomes were much stronger, suggesting that these genes are highly upregulated regardless of certain embryonic stages. RNA-seq analyses showed that the KO2 sample had downregulated expression of differentiated mesoderm genes, whereas the expression of endoderm lineage genes was upregulated ( S2G Fig ). KO2 exhibited higher expression of mesendoderm progenitor markers (Mixl1, and Gsc), endoderm progenitor marker (Lhx1), and definitive endoderm markers (Sox17, and Foxa2). The ‘Endoderm cell fate specification’ category was also enriched in GO term analysis of most upregulated genes in Ddx6 △/△ ( S2A Fig ). Therefore, posteriorly expanded high expression of the APS marker Nodal and Eomes disturbed the patterning of the primitive streak, which likely directed mesendoderm progenitors toward the endodermal lineage. Transcript levels of some key genes were further assessed by qRT-PCR using separately prepared Ddx6 △/△ embryos and the results were consistent with the RNA-seq data ( S2H Fig ). BMP signaling also prevents premature neural induction [ 42 ]. We examined whether this function was also impaired in Ddx6 △/△ embryos. SOX1 is the earliest neuroectoderm marker and is normally not detected until E7.5 in WT embryos [ 51 , 42 ]. However, E6.5 Ddx6 △/△ embryos exhibited clear SOX1 expression in all epiblast cells ( Fig 2D ). Additionally, premature neuronal differentiation was detected in Ddx6 △/△ based on RNA-seq. The markers of neural stem cells (NSCs) and neural progenitor cells (NPCs), such as Sox1 and Pax6, were downregulated ( S2D Fig ), but genes of neuron-restricted progenitors and differentiated post-mitotic neuronal cells were upregulated ( S2E Fig ). Section IHC confirmed that protein expression resembled transcript levels ( Fig 2E ). The earliest neuroectoderm marker, SOX1, and a persistent marker of NSC and NPC, SOX2 [ 52 ], were weakly expressed in E8.5 Ddx6 △/△ embryos. In summary, in Ddx6 △/△ embryos, the neural lineage was precociously induced as in BMP receptor mutant embryos [ 42 ]. Moreover, Ddx6-deficient NSCs showed defective self-renewal maintenance and prematurely differentiated. BMP signaling is required for mesoderm formation and posterior body development [ 41 , 49 ]. Whole-mount in situ hybridization (WISH) with a Brachyury (T) probe showed that the primitive streak existed in E8.5 Ddx6 △/△ but it did not extend anteriorly (Figs 2A and S1F ). The somites were barely developed. We also examined the induction timepoint of Brachyury. Gastrulation begins around E6.5 as the primitive streak forms and is preceded by Brachyury expression [ 50 ]. Unlike WT, Ddx6 △/△ embryos started expressing BRACHYURY from E7.5 ( Fig 2B ). E7.5 WISH also revealed a small amount of Brachyury in the very proximal posterior region of the embryo ( Fig 2A ). Visualization of Otx2 expression, which marks the head region, indicated the lack of posterior body in mutants, which have not formed head fold yet ( Fig 2C ). In summary, there was a delay of primitive streak formation in Ddx6 △/△ embryos, and their shortened and widened primitive streak ( S1F Fig ) suggested that the nascent mesoderm population has defects in differentiation and subsequent ingression. Ddx6 △/△ embryos had defects in posterior body development and mesoderm differentiation similar to BMP signaling mutant embryos. To characterize Ddx6 mutant embryos at the transcript-level and further find the possible causes of defects, we performed RNA sequencing (RNA-seq). Two E8.5 Ddx6 KO cDNA libraries (KO1 and KO2) were generated. KO1 comprised one Panel 1 in Fig 1 -like embryo and one Panel 2 in Fig 1 -like embryo, whereas KO2 was composed of three Panel 1 in Fig 1 -like embryos. As there was variability between the two samples, we concluded them to be insufficient to provide statistical significance and decided to utilize RNA-seq data just to provide a rough trend and guide subsequent experiments. Gene ontology (GO) term enrichment analysis indicated that genes of major developmental processes, especially cell fate commitment and the formation of mesoderm derivatives, such as muscle tissue development, muscle organ morphogenesis, cardiac myofibril assembly, skeletal muscle organ development, and connective tissue development, were downregulated in Ddx6 KO libraries ( S2A Fig ). In contrast, the terms associated with cell death, immune response, cell metabolism, and negative regulation of BMP signaling pathway-related genes were highly upregulated. Negative regulation of the BMP signaling pathway was notable because BMP signaling has multiple important roles during early embryogenesis. Several BMP negative regulators were upregulated in Ddx6 △/△ embryos ( S2B Fig ). Based on the reported functions, genes that are listed as the negative regulators of BMP signaling were classified into five clusters: receptor-related (Inhbb, and Tmprss6), secreted BMP antagonists (Cer1, Chrd, Chrdl2, Noggin, Grem2, and Htra1), TGF-β signaling-related (Lgals9, Xdh, Pai1, Hpgd, and miR-382), FGF signaling-related (Fgf5), and intracellular inhibitor (Nanog) ( S2C Fig ). As genes related to the inhibition of BMP signaling were highly upregulated, we examined whether BMP signaling is repressed in Ddx6 △/△ embryos. We reasoned that if BMP signal transduction is indeed dysfunctional, then Ddx6 △/△ embryos should exhibit the representative phenotypes of BMP signaling mutant embryos. Developmental defects were observed from E6.5 ( Fig 1 ). At E7.5, mutants were smaller, but an extraembryonic body part developed and embryos formed a cylindrical shape. E8.5 Ddx6 KO embryos exhibited developmental delay and posterior defects with some variability ( S1D Fig ). They were broadly divided into two embryonic stages: before (Panel 1 in Fig 1 and Panels 1–3 in S1D Fig ) and after (Panel 2 in Fig 1 and Panels 4–6 in S1D Fig ) head-fold formation. The variance in posterior defects was also observed in E9.5 mutants. Some developed a mid-posterior body ( Fig 1 and Panel 1 in S1E Fig ), whereas others exhibited marked posterior truncation (Panels 2–3 in S1E Fig ). In conclusion, DDX6 is necessary for mouse embryonic development. To clarify the role of DDX6 in embryonic development, we generated a Ddx6 △/+ mouse line and crossed heterozygous mice to get Ddx6 △/△ (KO) embryos. The genotyping results of the collected embryos are displayed in Table 1 . No Ddx6 △/△ embryo was found from E11.5. The proportion of homozygous KO embryos at E7.5~E8.5 (12.8% and 15.4%) was already lower than the expected Mendelian ratio, which signifies that some mutant embryos died even earlier than E7.5. Before investigating the functions of DDX6 during embryogenesis, we examined its expression pattern through DDX6 immunohistochemistry (IHC). In embryonic day (E) 6.5 embryos, DDX6 was highly and ubiquitously expressed, localizing in cytoplasmic foci ( S1A Fig ). All DDX6 foci co-localized with DCP1A foci, a P-body-specific marker, indicating that DDX6 is expressed in P-bodies. At E7.5, DDX6 expression was strongest in the epiblast and there were many distinct P-body foci ( S1B Fig ). BRACHYURY-positive emerging and migrating mesoderm cells had relatively weaker DDX6 expression with a fewer number of P-bodies. In E8.5 embryos, DDX6 expression was observed in all areas, including the neuroepithelium, tailbud, and somites ( S1C Fig ). DDX6 was ubiquitously expressed in early embryos in forming P-bodies. Discussion This study delineated the essential role of DDX6 in proper cell lineage specification and differentiation during early mouse embryogenesis (Fig 7A and 7B). We propose that DDX6 prevents cells from activating negative regulation of BMP signaling through miRNA-mediated gene silencing. The genes that are related to ‘negative regulation of BMP signaling’ were upregulated in both Ddx6 KO E8.5 embryos and ESCs. Their phenotypes, including posterior body and mesoderm developmental defects, premature neural induction, and de-repression of BMP-SMAD1/5 target genes, indicate that the BMP pathway is dysfunctional in Ddx6 KO. Another characteristic of Ddx6 KO was the increased expression of Nodal and its downstream targets. Promoted pluripotency, such as increased Nanog expression, and disruption of the primitive streak patterning by overexpanded Eomes expression can be attributable to high Nodal. However, there was no increase in the level of SMAD2 phosphorylation in Ddx6 KO ESCs (S4A Fig). We now hypothesize that the increased expression of Nodal and its known downstream targets Nanog and Eomes in Ddx6 KO is due to an altered SMAD3 activity level. Previously, Liu et al. reported a marked difference between SMAD2 and SMAD3. They showed that the main mediator of TGF-β signaling is SMAD2, whereas SMAD3 has a TGF-β signaling- and SMAD4-independent working mechanism [68]. Thus, we briefly analyzed the public ChIP-seq data for SMAD2 and SMAD3 in embryoid bodies, which are similar to early-stage embryos. When we assayed potential target genes using ‘ChIP-Atlas,’ Eomes, whose expression was significantly increased in Ddx6 KO embryos, was only bound by SMAD3 (S4B and S4C Fig). Nanog was more preferentially bound by SMAD3, whereas Nodal was equally regulated by SMAD3 and SMAD2. Based on these results, Ddx6 KO phenotypes may arise from the increased SMAD3-induced transcriptional activation rather than Nodal signaling itself. Further detailed analyses are required to clarify this possibility. PPT PowerPoint slide PNG larger image TIFF original image Download: Fig 7. Schemes showing developmental defects caused by loss of DDX6-mediated RNA regulation. (A) Development of the three primary embryonic germ layers is largely affected by DDX6. Neuroectoderm is specified earlier than WT, whereas formation of the primitive streak is delayed (The smaller size of the Ddx6 mutant is not reflected in the images). (B) Changes in cell-lineage specification from pluripotent stem cells caused by Ddx6 loss are depicted on a horizontal diagram. Uncommitted Ddx6△/△ pluripotent cells possess promoted pluripotency and strongly favor commitment to the neuronal lineage. In WT embryos, the mesendoderm lineage arises at ~E6.5 as the primitive streak is formed, and three germ layers simultaneously develop at ~E7.5. In Ddx6 KO embryos, premature neural induction occurs with a one-day delay in primitive streak formation. During mesendoderm segregation, definitive endoderm specification is increased, whereas mesoderm specification is greatly reduced due to the patterning defect of the primitive streak. Posterior epiblast cells cannot exit pluripotency on time, impeding the differentiation processes. (C) The interaction of DDX6 with other gene silencing effector protein complexes. https://doi.org/10.1371/journal.pgen.1009967.g007 Since DDX6 functions in multiple pathways, it is difficult to find the most crucial one for a certain cellular process. To overcome this problem, we examined three gene-regulating pathways, in which DDX6 is known to have a role: translational repression, mRNA degradation, and miRNA-mediated gene silencing by knocking out the key gene. EIF4ENIF1 (4E-T, eIF4E-Transporter) is a kind of eIF4E-binding proteins (4E-BPs), which inhibit the initiation step of translation by binding to eIF4E, a component of the translational initiation factor eIF4F complex [69–72]. When eIF4E is captured by 4E-T, an mRNA whose cap is bound by eIF4E is also brought into P-bodies. Importantly, eIF4E-4E-T-bound transcripts are protected from decapping even after deadenylation. Hence it is a distinct mechanism that specifies mRNAs to be stored in P-bodies instead of being degraded [73]. 4E-T binds to DDX6 via Cup homology domain (CHD) and is necessary for the assembly of P-bodies [17]. Thus, we wanted to examine the degree of involvement of DDX6 in translational repression process and the dependency of DDX6 functions on P-body formation via Eif4enif1 KO. DCP2 (NUDT20) is a major mRNA decapping enzyme in mammalian cells along with another Nudix hydrolase protein, NUDT16 [74–76]. Knockdown of DCP2 in human U2OS cells caused the enlargement of P-bodies with the accumulation of the MS2-tagged β-actin transcripts, leading to the conclusion that a constant mRNA decay occurs inside the P-bodies, and when DCP2 is depleted, this decay process is inhibited [67]. DCP2 is connected to DDX6 through EDC3 [77]. As Dcp2 KO also changes the morphology of P-bodies, we were able to study the effects of the disruption of mRNA decay pathway as well as functional defect of P-bodies through Dcp2 KO. Even though this genetic approach cannot directly explore molecular functions of P-bodies or target transcripts, we tried to indirectly examine their roles in cellular processes by conducting transcriptomic analyses. The RNA-seq analyses identified the similarity between Ddx6 KO and Dgcr8 KO. Only Dgcr8 KO, but neither Eif4enif1 KO nor Dcp2 KO, closely phenocopied Ddx6 KO, indicating that DDX6 mainly works through the miRNA pathway among the various RNA regulatory means during early embryogenesis. The double-stranded RNA-binding protein DGCR8 forms the microprocessor complex with the RNase III enzyme Drosha to process long primary miRNAs (pri-miRNAs) into precursor miRNAs (pre-miRNAs) [78,79]. In Dgcr8 KO, miRNA-mediated gene silencing becomes nonfunctional because of the failure of miRNA generation [66]. miRNAs repress target gene expression through either translational repression or mRNA degradation [80–83]. In mESCs, the loss of DDX6 impaired only miRNA-induced translational repression [48]. Thus, the greater number of upregulated gene sets in Dgcr8 KO ESC than in Ddx6 KO (1211 vs 828 gene sets) may have resulted from the disruption of miRNA-mediated mRNA destabilization. DDX6 participates in the effector step of miRNA-mediated gene silencing by binding to CNOT1, the scaffold subunit of the CCR4-NOT complex. The CCR4-NOT complex, a predominant generic deadenylase [84], cooperates with miRISC by binding to GW182/TNRC6 protein [80], and then recruits DDX6. The binding of CNOT1 and DDX6 activates the ATPase activity of DDX6, and this change is significant for target gene repression [85]. Our study is connected to previous studies which demonstrated the important role of miRNAs during early embryogenesis. miR-302 and miR-290 are highly expressed in pluripotent cells and are required for neurulation. Loss of miR-302 causes precocious neuronal differentiation [86]. Moreover, miR-302 stimulates BMP signaling by repressing BMP inhibitors TOB2, DAZAP2, and SLAIN1 in hESCs [87]. Although these genes were not upregulated in Ddx6 KO or Dgcr8 KO mESCs, probably due to the species difference, it is a good example that miRNAs can indirectly affect BMP signaling by targeting BMP signaling inhibitors. Embryos had abundant P-bodies from blastocyst to peri-gastrula embryos, but disruption of P-body function had marginal effects on early embryogenesis. Based on our results, we refute Di Stefano et al. [20]’s statement that disruption of P-bodies is the cause of Ddx6-deficient PSCs being “hyper-pluripotent.” Ayache et al. [17] investigated multiple conditions for P-body assembly in HEK293 cells and found that DDX6, 4E-T (EIF4ENIF1), and LSM14A (RAP55) are the three key factors of P-body formation. Eif4enif1 KO mESCs from our study and LSM14A knockdown hESCs from Di Stefano et al. completed the puzzle of the relationship between DDX6 and the P-body. The characteristics of Ddx6 KO mESCs and DDX6 KD hESCs were similar. As described earlier, our mESCs also exhibited hyper-pluripotent properties by having high expression of Nanog and failing to exit the ground state properly. However, the two P-body mutants exhibited different phenotypes. Knockdown of LSM14A made hESCs hyper-pluripotent with the significantly higher expression of NANOG like DDX6-depleted hESCs. In our case, deletion of Eif4enif1 led to the disassembly of P-bodies in mESCs (S3D Fig), but Eif4enif1 KO pluripotent cells did not display hyper-pluripotent properties (S5B Fig). Furthermore, Eif4enif1 KO embryos lacked the developmental defects of Ddx6 mutants (S5A Fig). As such, P-bodies are not essential for development at least until the gastrulation stage. This result is consistent with a previous study demonstrating that P-body formation is not the cause but a consequence of RNA-mediated gene silencing activities [13]. However, we do not totally agree that P-bodies are merely a resultant complex. Our RNA-seq analyses showed that the gene expression profiles of Eif4enif1 KO and Dcp2 KO moved into a similar direction and clustered together despite the apparent functional differences. We speculated that this change may have come from the disruption of overall P-body function. From the GSEA, there were a few gene sets that are commonly changed only in Eif4enif1 KO and Dcp2 KO. As these gene sets were not affected in Ddx6 KO or Dgcr8 KO, we considered them as genes that are associated with P-bodies but not related to miRNA-mediated gene silencing (Tables 2 and S1). miRNA-mediated gene silencing activity was functional when P-bodies were disrupted, suggesting miRNA function is P-body-independent [13]. However, the presence of distinctive gene sets that are altered by both miRNA pathways (Ddx6 KO & Dgcr8 KO) and P-body-related functions (Eif4enif1 KO & Dcp2 KO) may suggest that GW182 and AGO2, the essential components of the miRISC, locate to P-bodies for certain functional purposes rather than just being stored there. Unlike in plants, direct cleavage by AGO2 endonuclease of miRISC is rare in animal cells [88,89]. Animal miRISCs instead recruit other effector proteins to facilitate either translational repression or mRNA decay [90]. We conjectured that DDX6, which interacts with various mRNA repressing proteins and is incorporated into the miRISC, would work as a bridge coupling miRNA target with the RNA repression effector. We initially expected that depletion of 4E-T (disruption of translational repression pathway) or DCP2 (disruption of mRNA decay pathway) would show the similar phenotypes to DDX6 loss. Different from our expectation, these downstream effector pathways did not phenocopy Ddx6 KO and Dgcr8 KO. On the other hand, the loss of LSM14A resulted in a similar phenotype to Ddx6-depleted cells [20]. These results led to the following hypotheses: 1) According to GSEA, distinct gene sets were commonly affected in Eif4enif1 and/or Dcp2 KO (Tables 2 and S1). These gene sets may not be important for early developmental processes. 2) There are diverse translational repressors in the cytoplasm. The 4E-T-associated mechanism may not be the major method of translational repression in pluripotent cells. DDX6 uses the same surface (RecA2) for binding to different RNA regulatory proteins in a mutually exclusive manner (Fig 7C) [91–97,8,77,21,11]. Among various binding partners, 4E-T and LSM14A are the two translation repression proteins whose interaction with DDX6 is well documented [21,11,97]. Taken together, we surmise that the DDX6-LSM14A complex is necessary for facilitating translation repression targeted by miRNAs, but P-body assembly/maintenance is not required for this process. Although DDX6, miRNA-mediated gene silencing (DGCR8), 4E-T, DCP2 are highly intercorrelated, they also have other molecular functions independent of P-bodies or proteins that are analyzed here. To have a clue about each protein’s unique role, we searched genes that were only affected in each KO ESCs (Fig 6B and S2–S5 Tables). For instance, in Ddx6 KO ESCs (S2 Table), some of upregulated gene sets were related to the immune response (MYD88-independent toll like receptor signaling pathway, positive regulation of small GTPase-mediated signal transduction, myeloid dendritic cell activation, positive regulation of leukocyte apoptotic process), which is consistent with the known DDX6 role of preventing aberrant activation of interferon-stimulated genes [98]. Many downregulated gene sets were related to nucleoside and nucleotide biosynthesis and metabolism, which implies that DDX6 is deeply involved in nucleoside/nucleotide processing. Lastly, we discuss the significance of DDX6 studies. DDX6 is ubiquitously expressed with well conserved functional capabilities, but its function often becomes specific in a certain cell type depending on interacting protein complexes. For example, DDX6 is necessary for parental RNA degradation in fibroblasts for iPSC reprogramming in concert with the RO60/RNY1 complex [99]. It also acts as a promoter of translation in epidermal progenitor cells along with the YBX1/EIF4E complex [15]. DDX6 has another role in packaging the retroviral RNA genome by interacting with the viral RNA-Gag ribonucleoprotein complex [100]. One of the key findings of our study is that DDX6 molecular functional pathways can be relatively clearly segregated. We need to exploit this potent endogenous gene expression regulator. DDX6 participates in various RNA metabolisms and has a wide expression pattern (e.g., in pluripotent cells, adult tissue stem/progenitor cells, cancer, and RNA-virus infected cells). Deciphering DDX6-mediated regulatory pathways and modular manipulation of a specific pathway may make DDX6 a useful genetic therapeutic target. [END] --- [1] Url: https://journals.plos.org/plosgenetics/article?id=10.1371/journal.pgen.1009967 Published and (C) by PLOS One Content appears here under this condition or license: Creative Commons - Attribution BY 4.0. via Magical.Fish Gopher News Feeds: gopher://magical.fish/1/feeds/news/plosone/