Distinct functional requirements of the SETDB1-binding region and the FNIII domain in ATF7IP-dependent retroelement silencing
We have previously established Atf7ip KO cells using mESCs infected with the murine stem cell virus (MSCV) carrying the GFP gene as a background (22) and observed that the Atf7ip KO ESCs showed increased expression of SETDB1-regulated ERVs and the MSCV-GFP reporter (16). For rescue experiments with ATF7IP, we used a piggyBac transposase-based vector for the expression of 3xFLAG-tagged mouse ATF7IP with either WT or each domain’s deletion mutants: dSETDB1 lacking residues 627-694, which is within the corresponding SETDB1 binding domain of mouse ATF7IP and covering two estimated a-helix regions, and dFNIII lacking residues 1190-1306 of the FNIII domain, which is highly conserved between human and mouse ATF7IP (Fig. 1A and Additional file 1: Fig. S1). These piggyBac transposase-based vectors can be integrated into the host genome and stably express exogenous FLAG-tagged ATF7IPs. By the transfection of each plasmid and continuous drug selection, we established Atf7ip KO mESCs expressing 3xFLAG-ATF7IP-WT, dSETDB1, or dFNIII mutants and performed a co-immunoprecipitation (co-IP) assay with these cell lines using a FLAG M2 affinity gel. As expected, the dSETDB1 mutant could not co-IP endogenous SETDB1, while the WT and the dFNIII mutant could (Fig. 1B). We then examined the RNA expression of exogenous MSCV-GFP and ERVs regulated by ATF7IP and SETDB1 under long-term culturing conditions (greater than two weeks). As shown in Fig. 1C, the ATF7IP WT and the dFNIII mutant re-silenced the de-repressed MSCV-GFP and ERV expression, while the dSETDB1 mutant did not, suggesting that its interaction with SETDB1 is essential for the role of ATF7IP in transcriptional silencing.
We next examined the silencing kinetics of these rescued cells by taking advantage of the MSCV-GFP reporter (Fig. 1D). We transfected 3xFLAG-ATF7IP with either WT, dSETDB1, or dFNIII expression vectors into Atf7ip KO mESCs, and the transfected cells were selected by continuous puromycin treatment. At day 5 after transfection, GFP expression levels were analyzed by flow cytometry. As expected, the WT-rescued cells showed low GFP expression, similar to that of the parental Atf7ip WT cells, and the dSETDB1-rescued cells showed higher GFP expression, as seen in the Atf7ip KO cells. Unexpectedly, the expression of the dFNIII mutant could not re-silence MSCV-GFP expression on day 5. However, the higher GFP expression in the dFNIII-expressing cells was repressed to the WT level after culturing them for an additional seven days (at day 12). Consistent with this observation, the dFNIII-rescued cells at day 5 showed higher expression of GFP mRNA compared to that of the WT-rescued cells (Fig. 1E). We then confirmed the expression of exogenous ATF7IP protein between the WT- and dFNIII-rescued cells at day 5 by western blot analysis (Fig. 1F). Interestingly, ERVs were re-silenced by the expression of the dFNIII mutant, similar to WT expression, even at day 5 (Fig. 1E). These results suggest that the FNIII domain plays a role in ATF7IP-mediated transcriptional silencing under certain condition.
Interaction with SETDB1, but not the FNIII domain of ATF7IP, is required for ATF7IP-dependent SETDB1 nuclear localization
We have recently reported that ATF7IP regulates SETDB1’s nuclear localization by antagonizing and enhancing its nuclear export and import, respectively (16). We therefore determined whether the expression of the dSETDB1 or FNIII mutant can rescue the cytoplasmic accumulation phenotype of SETDB1 in Atf7ip KO mESCs. We examined SETDB1 localization in the long-cultured 3xFLAG-tagged ATF7IP WT-, dSETDB1-, and dFNIII-rescued cells by immunofluorescence (IF) analysis. The WT and the two mutants of 3xFLAG-tagged ATF7IP were all localized in the nucleus (Fig. 2A; quantification in Fig. 2B, right). Expression of the ATF7IP WT and the dFNIII mutant in Atf7ip KO cells restored the SETDB1’s nuclear localization (Fig. 2A; quantification in Fig. 2B, left) without significant changes in nuclear foci numbers (Fig. 2C and D, left). Both the ATF7IP WT and the dFNIII mutant also co-localized with SETDB1 in the nucleus and in the nuclear foci (Fig. 2A). In contrast, the dSETDB1 mutant could not rescue the cytoplasmic localization of SETDB1 (Fig. 2A; quantification in Fig. 2B, left), and the number of dSETDB1 nuclear foci was reduced (Fig. 2A; quantification in Fig. 2C and D, right). The dFNIII-rescued cells also showed that SETDB1 and exogenous ATF7IP were localized in the nucleus as efficiently as those in the ATF7IP WT-rescued Atf7ip KO cells five days after transfection (Additional file 2: Fig. S2A-C). These results suggest that the regulation of SETDB1 nuclear localization by ATF7IP requires their interaction and that a delayed silencing of MSCV-GFP in dFNIII mutant-rescued cells does not seem to be caused by SETDB1’s delayed nuclear localization or mislocalization.
Identification of ATF7IP FNIII domain-binding proteins
We further sought to reveal the underlying mechanism for the inefficient re-silencing of the MSCV-GFP reporter transgene by the dFNIII mutant (Fig.1D). Since the FNIII domain functions as a binding domain for MBD1 in human cell lines (9,18), we searched for binding proteins for the FNIII domain of ATF7IP in mESCs. For this, we performed a proteomic analysis with the recombinant FNIII domain of mouse ATF7IP and nuclear lysates of mESCs (Fig. 3A). The nuclear fractions from Atf7ip KO mESCs were incubated with a GST-tagged FNIII domain, produced in, and purified from E. coli. After purification with glutathione beads, the bound proteins with the FNIII domain were identified by liquid chromatography followed by tandem mass spectrometry (LC-MS/MS) analysis (Fig. 3B, the full list is in Additional file 3: Table S1). We identified over 20 proteins enriched in the FNIII domain-pulled-down sample, with a high coverage, and found some known protein networks, including a ZMYM2 (also known as ZNF198)-LSD1 (also known as KDM1A)-HDAC complex (23,24), by STRING analysis (Additional file 4: Fig. S3). Unexpectedly, we could not recover MBD1 in our proteomic analysis, suggesting that MBD1 may not exist in Atf7ip KO mESCs or that their interaction may not occur significantly in mESCs. Using a co-IP experiment with transient ectopic expression in HEK293T cells, we validated the interaction of full-length ATF7IP with several high-ranked candidates, including ZMYM2, MGA (residues 2362-3003), ZFP518A, and KIAA1551, and a known interactor MBD1 (Fig. 3C). As expected, the dFNIII mutant could not bind to these proteins, but it could bind to SETDB1 (Fig. 3D), supporting the hypothesis that these newly identified interactors, as well as MBD1, can bind to the FNIII domain of ATF7IP.
To examine which region of ZMYM2, which is a top-ranked protein in our proteomic analysis, is essential for its interaction with ATF7IP, we performed co-IP experiments with a series of truncated mutants of ZMYM2 using HEK293T cells (Additional file 5: Fig. S4A-D). These results suggest that residues 181-350 within ZMYM2 seem to be important for its interaction with ATF7IP. Visual inspection of this region revealed that it contains two sequences (referred to as FNIII domain of ATF7IP-interacting motif 1 (FAM1) and FAM2, explained later) similar to an “ITEFSL” sequence within the TRD of MBD1, which was shown to be essential for its binding to the FNIII domain (18). As substitutions of isoleucine (I) and leucine (L) to arginine (R) within this sequence perturbed the interaction between MBD1 and the FNIII domain (9,18), we wondered whether similar mutations at the corresponding “V” and “L” residues of FAM1 and/or FAM2 within ZMYM2 can affect the interaction between ZMYM2 and ATF7IP (see Additional file 6: Fig. S5A). We transfected 3xFLAG-ATF7IP with either the control empty vector, V5-ZMYM2-WT, FAM1 mutant (V187R/L190R), FAM2 mutant (V217R/L220R), FAM1 and FAM2 double mutant, or a d39aa mutant that lacks residues 182-220 into HEK293T cells and performed co-IP experiments with an anti-FLAG M2 affinity gel (Fig. 3E). The results showed that the mutations at either FAM1 or FAM2 impaired the interaction, and that the FAM1 and 2 mutant or the d39aa mutant completely failed to co-IP ATF7IP, suggesting that both FAM1 and FAM2 contribute to the binding with ATF7IP. We further examined the interaction of ZMYM2 with ATF7IP using a GST pull-down assay (Fig. 3F). The HEK293Tcell lysates transfected with V5-ZMYM2 WT or FAM1&2 Mut were pulled-down with a recombinant GST or a recombinant GST-FNIII domain. Western blot analysis showed that the GST-FNIII domain, but not GST alone, bound to ZMYM2 WT, and that the binding of the FAM1 and 2 mutant to ATF7IP was severely impaired compared to that of the ZMYM2 WT. These data suggest that ZMYM2 binds to the FNIII domain of ATF7IP via its FAM1 and FAM2 motifs. We then examined primary sequences of the identified FNIII-binding proteins and found that MGA, ZMYM4, and ZFP518A possess an “ITEFSL”-like sequence (Fig. 3G). We showed that mutations on the motif abolished the interaction of those proteins with ATF7IP (Additional file 6: Fig. S5A-C). We therefore proposed that the “ITEFSL”-like sequences are a consensus binding motif for the FNIII domain of ATF7IP, and referred to as FAM.
ZMYM2 is involved in the efficient silencing of exogenous provirus reporter by ATF7IP
Among the identified binding proteins for the FNIII domain of ATF7IP, we focused our attention on the top-ranked protein ZMYM2, which has two FAMs, and may function with the LSD-HDAC1 repressor as a complex (23,24). We established Zmym2 KO mESCs using CRISPR/Cas9 technology. mESCs harboring the hCas9 and MSCV-GFP reporter were transfected with an expression vector for gRNA targeting the mouse Zmym2 gene. We observed a slight increase in the MSCV-GFP reporter in the Zmym2-gRNA-transfected cells by flow cytometry analysis (Additional file 7: Fig. S6A) and sorted the cell populations with high GFP intensity. The sorted cells were cloned, and the ZMYM2 expression in the cloned cell lines were subsequently analyzed. We finally isolated two independent clones of Zmym2 KO mESCs, as evidenced by western blot using an anti-ZMYM2 antibody (Fig. 4A). We found that both established Zmym2 KO cell lines showed GFP expression equivalent to the parental WT cells (Additional file 7: Fig. S6B), resembling the case of the ATF7IP FNIII-rescued Atf7ip KO mESCs after long-term culture (Fig.1D). We then transfected Zmym2 KO cells with a 3xFLAG-tagged ZMYM2 WT expression vector and confirmed their expression (Fig. 4A). By using the 3xFLAG-tagged ZMYM2-rescued cells, we observed the co-localization of 3xFLAG-ZMYM2 with endogenous SETDB1 at the foci in the nucleus by IF analysis (Fig. 4B), suggesting a potential function of ZMYM2 in interacting with SETDB1 and ATF7IP. Furthermore, we found an enrichment of the FLAG-tagged ZMYM2 at the SETDB1/ATF7IP-target genomic regions, including the LTR of MSCV-GFP (Fig. 4C).
To examine the potential involvement of ZMYM2 in the re-silencing of the MSCV-GFP reporter by ATF7IP, we further inactivated Zmym2 in Atf7ip KO mESCs. We confirmed the depletion of ZMYM2 protein in the two Zmym2/Atf7ip DKO cell lines by western blot analysis (Fig. 4D). Furthermore, we found an upregulation of ZMYM2 protein in Atf7ip KO mESCs compared to the parental WT cells (Fig. 4D), suggesting the existence of a potential negative feedback mechanism. We then transfected either Atf7ip KO cells or Zmym2/Atf7ip DKO cell lines with 3xFLAG-ATF7IP WT and analyzed them by flow cytometric analysis five days after transfection. We found that the re-expression of ATF7IP WT in the Zmym2/Atf7ip DKO cell lines incompletely silenced the expression of MSCV-GFP reporter (Fig. 4E). Consistent with this, we observed, by RT-qPCR analysis that although the expression levels of 3xFLAG-ATF7IP WT mRNA were similar, the 3xFLAG-ATF7IP WT-rescued Atf7ip/Zmym2 DKO cells showed ~3-fold increase in the expression of GFP mRNA compared to the 3xFLAG-ATF7IP WT-rescued Atf7ip KO cells at day 5 after transfection (Fig. 4F). Taken together, these results suggest that ZMYM2 partly mediates the efficient re-silencing of the MSCV-GFP reporter by ATF7IP in mESCs.
The FNIII domain of ATF7IP contributes to the efficient silencing of SETDB1 target ERVs and some MGA/MAX-targeted germ cell-related genes
To further elucidate the role of the FNIII domain in ATF7IP-mediated transcriptional regulation, we performed RNA-seq analysis of WT, Atf7ip KO, and Atf7ip KO stably rescued with WT or FNIII domain mutant of ATF7IP and Zmym2 KO mESCs (Additional file 9: Fig. S7). In comparison with parental WT ESCs, 87 and 69 genes were commonly up- and downregulated, respectively, (FDR < 0.05, FC ≥ 2) in two independent Atf7ip KO mESC clones, TT#2-5 and TT2-12 (16) (Fig. 5A and Additional file 8: Table. S2). A majority of the upregulated genes (76/87) in Atf7ip KO cells were repressed by exogenous ATF7IP WT expression (Fig. 5B). Complementation with ATF7IP WT induced a greater number of up- or downregulated genes compared to the Atf7IP KO (Fig. 5A). When the FNIII domain mutant was introduced into Atf7ip KO ESCs, a majority of the upregulated genes (70/87) in Atf7ip KO mESCs were reversed to WT levels (Fig. 5B). Interestingly, Gene Ontology (GO) term enrichment analysis using DAVID 6.7 (25) showed that GO terms related to the meiotic cell cycle or spermatogenesis were enriched in the 12 genes that were re-silenced in the WT rescued cells, but not in the dFNIII mutant-rescued cells (Fig. 5C). These include Rec114, Tex11, Tex15, Fkbp6, Sycp1, Stra8, and Mael (Additional file 8: Table. S2). Such germ cell-related genes were also de-repressed in Setdb1 KO mESCs (3). Furthermore, it has been reported that these genes were also induced in the Max knockdown (KD) mESCs, and that some of them were de-repressed in Atf7ip KD mESCs (26). RT-qPCR analysis confirmed that the de-repressed germ cell-related genes were not repressed by the FNIII domain mutant in Atf7ip KO ESCs at day 5 after transfection or were only partially silenced over longer culture conditions, whereas these genes were efficiently silenced by ATF7IP WT even at day 5 post-transfection (Fig. 5D). Thus, the FNIII domain has an indispensable role in the ATF7IP-mediated silencing of some MAX-regulated germ cell-related genes.
In the case of the retroelements, 10 different classes of repeats were upregulated in Atf7ip KO mESCs, which was consistent with previous findings (15), and all of them were repressed by exogenous ATF7IP WT expression (Fig. 5A and E). The introduction of the FNIII domain mutant also repressed most of the de-repressed repeats (9/10) (Fig. 5A and E). Interestingly, the other 10 different classes of retroelements were further downregulated in Atf7ip KO mESCs rescued with ATF7IP WT (Fig. 5A and F). The additionally downregulated retroelements were also SETDB1-targeted and de-repressed in Setdb1 KO mESCs (3,15). Since most of the additionally downregulated repeats by exogenous ATF7IP WT expression were indeed weakly (< 2-fold) de-repressed in Atf7ip KO ESCs (Fig. 5F), SETDB1-mediated retroelement silencing might be enhanced by the overproduction of ATF7IP WT in mESCs. However, the majority of additionally downregulated repeats by exogenous ATF7IP WT expression were more mildly repressed by the FNIII domain mutant in comparison with ATF7IP WT (Fig. 5F), even though the two molecules were similarly expressed (Fig. 1B), supporting the notion that the FNIII domain of ATF7IP contributes to efficient transcriptional silencing mediated by the SETDB1 complex.
In Zmym2 KO mESCs, multiple genes were also upregulated and downregulated (Fig. 5A). More than half of the upregulated genes (52/87) in Atf7ip KO mESCs were also upregulated in Zmym2 KO mESCs, whereas a smaller portion of upregulated genes (52/178) in Zmym2 KO mESCs were also upregulated in Atf7ip KO mESCs (Additional file 10: Fig. S8A, B, and Additional file 8: Table. S2), suggesting that the regulation of the majority of the upregulated genes in Zmym2 KO mESCs is independent of FNIII domain interaction. More importantly, however, among the 12 upregulated genes in Atf7ip KO mESCs that were not silenced by the FNIII domain mutant, 11 genes were also upregulated by ZMYM2 depletion (Additional file 8: Table. S2). We confirmed that the genes commonly upregulated in Atf7ip KO mESCs expressing the dFNIII mutant and the Zmym2 KO mESCs are repressed in the Zmym2 KO mESC complemented with 3xFLAG-ZMYM2 (Additional file 7: Fig. S6C). RNA-seq analysis showed that six different classes of repeats, which were mainly L1 elements, were upregulated in Zmym2 KO mESCs (Fig. 5A and Additional File 10: Fig. S8C). Furthermore, the IAPEy-int retroelement, which was upregulated in Atf7ip KO mESCs and was not silenced by the FNIII domain mutant, was also de-repressed in Zmym2 KO mESCs (Additional file 10: Fig. S8C and D). Thus, ZMYM2 contributes to ATF7IP FNIII domain-dependent transcriptional silencing, including MAX-targeted germ cell-related gene regulation.
Finally, we performed ChIP-seq analysis of Atf7ip KO mESCs rescued with 3xFLAG-tagged WT or the FNIII domain mutant of ATF7IP with an anti-FLAG antibody. We analyzed two samples for each cell type, and commonly detected peaks between two samples were defined as stringent peaks and utilized for subsequent informatics analysis. As shown in Fig. 5G, more than 90% of 3xFLAG-ATF7IP WT stringent peaks (1831/2015) overlapped with 3xFLAG-dFNIII mutant peaks. Because the number of 3xFLAG-ATF7IP WT peaks for one ChIP-seq sample was about 1/3 that of the other sample (2229 vs. 6331), the stringent peaks of 3xFLAG-ATF7IP WT might be underrepresented. When the total peaks of 3xFLAG-ATF7IP WT from two ChIP-seq samples were used for the same comparison analysis, 94.1% of the 3xFLAG-dFNIII mutant stringent peaks (3177/3376) overlapped with the 3xFLAG-WT peaks (Additional file 11: Fig. S9). 3xFLAG-ATF7IP WP was enriched on the transcription start site of some of the upregulated germ cell-related genes in Atf7ip KO mESCs, but the enrichment of 3xFLAG-dFNIII mutant was lost or diminished on them (Fig. S9B). We further examined 3xFLAG-ATF7IP accumulation on the retroelements that were major targets of the SETDB1/ATF7IP complex and de-repressed in Atf7ip or Setdb1 KO mESCs. As shown in Fig. 5H, 3xFLAG-ATF7IP WT was enriched in the retroelements and the binding profiles of the 3xFLAG-dFNIII mutant were mostly maintained. These data indicate that the deletion of the FNIII domain does not have a strong impact on ATF7IP targeting and accumulation, especially on SETDB1 target retroelements.