RNAP II CTD tyrosine 1 performs diverse functions in vertebrate cells

  1. Jing-Ping Hsin
  2. Wencheng Li
  3. Mainul Hoque
  4. Bin Tian
  5. James L Manley  Is a corresponding author
  1. Columbia University, United States
  2. Rutgers University New Jersey Medical School, United States

Abstract

The RNA polymerase II largest subunit (Rpb1) contains a unique C-terminal domain (CTD) that plays multiple roles during transcription. The CTD is composed of consensus Y1S2P3T4S5P6S7 repeats, in which Ser, Thr and Tyr residues can all be phosphorylated. Here we report analysis of CTD Tyr1 using genetically tractable chicken DT40 cells. Cells expressing an Rpb1 derivative with all Tyr residues mutated to Phe (Rpb1-Y1F) were inviable. Remarkably, Rpb1-Y1F was unstable, degraded to a CTD-less form; however stability, but not cell viability, was fully rescued by restoration of a single C-terminal Tyr (Rpb1-25F+Y). Cytoplasmic and nucleoplasmic Rpb1 was phosphorylated exclusively on Tyr1, and phosphorylation specifically of Tyr1 prevented CTD degradation by the proteasome in vitro. Tyr1 phosphorylation was also detected on chromatin-associated, hyperphosphorylated Rpb1, consistent with a role in transcription. Indeed, we detected accumulation of upstream antisense (ua) RNAs in Rpb1-25F+Y cells, indicating a role for Tyr1 in uaRNA expression.

https://doi.org/10.7554/eLife.02112.001

eLife digest

When a gene is expressed, the DNA is first transcribed to produce an intermediate molecule called a messenger RNA (mRNA), which is then translated to produce a protein. RNA Polymerase II is an enzyme that makes mRNA molecules in organisms as diverse as plants, animals and yeast.

RNA Polymerase II is a complex made of a number of proteins. The largest protein in this complex includes a ‘carboxy-terminal domain’ that has multiple repeats of seven amino acids one after the other. The first amino acid in each repeat, a tyrosine, is referred to as tyrosine-1. Adding various chemical tags to the amino acids in these repeats co-ordinates the steps involved in the transcription of genes. In yeast, for example, adding a phosphate groups to tyrosine-1 seems to help the polymerase to proceed to make long mRNA molecules. However, it is not known what these chemical tags do in humans or other animals.

Now Hsin et al. (and independently Descostes, Heidemann et al.) have shown that the same phosphate groups on tyrosine-1 perform functions in vertebrates (animals with backbones) that are different to those performed in yeast. These functions include protecting the carboxy-terminal domain from being broken down inside cells, and transcribing the DNA that is upstream of genes.

Hsin et al. replaced tyrosine-1 in RNA Polymerase II from chicken cells with a related amino acid that cannot have phosphate groups added to it. This mutant RNA Polymerase II was unstable and degraded by the molecular machinery in cells that breaks down damaged or unneeded proteins back into amino acids. Hsin et al. also compared the mRNA molecules that are made by the wild-type RNA Polymerase II with those produced by a related mutant. This comparison revealed an unexpected accumulation of RNA molecules that are transcribed in the opposite direction from mRNAs. These RNA molecules, known as ‘upstream antisense RNAs’, have been described only recently. And while the function of these RNAs remains mysterious, the results of Hsin et al. suggest that tyrosine-1 helps to ensure that these RNA molecules are rapidly broken down.

The results of Hsin et al. raise a number of important questions, and foremost among these questions is: how do these newly discovered properties of tyrosine-1 contribute to the control of gene expression in animals? Further work is needed to answer this question.

https://doi.org/10.7554/eLife.02112.002

Introduction

RNA polymerase II (RNAP II) is a multisubunit enzyme responsible in eukaryotes for transcription of all mRNAs and many non-coding RNAs. Rpb1, the largest subunit, contains a unique C-terminal domain (CTD), composed of up to 52 heptad repeats with the consensus sequence Tyr1-Ser2-Pro3-Thr4-Ser5-Pro6-Ser7 (YSPTSPS). The CTD performs multiple functions during transcription (Buratowski, 2009; Munoz et al., 2010; Egloff et al., 2012; Hsin and Manley, 2012; Heidemann et al., 2013), most of which are dependent on phosphorylation of specific CTD residues. For example, Ser5 phosphorylation (Ser5-P) promotes recruitment of capping enzyme (Fabrega et al., 2003) and Ser2-P can be important for 3′ mRNA processing (Kim et al., 2004). Ser7-P and Thr4-P also function in 3′ processing, of snRNAs (Egloff et al., 2007) and histone mRNAs (Hsin et al., 2011), respectively, with the later also functioning in transcription elongation (Hintermair et al., 2012). Tyr1 can be phosphorylated in mammals (by c-Abl; Baskaran et al., 1993) and yeast (Mayer et al., 2012), where it may prevent premature recruitment of termination factors. However, the function(s) of Tyr1 and Tyr1-P in metazoans are unknown.

Results and discussion

We previously utilized chicken DT40 cells to study properties of the Rpb1 CTD. We showed that an Rpb1 derivative containing a CTD with 26 YSPTSPS repeats (Rpb1-26r) plus the ten C-terminal non-consensus residues, important for stability (Chapman et al., 2005), confers cell viability, while a comparable derivative with all Thr4 residues changed to Val was inviable (Hsin et al., 2011). To investigate the functions of Tyr1, we constructed a plasmid encoding a Flag-tagged Rpb1 derivative, Rpb1-Y1F, identical to Rpb1-26r but with all Tyr1 residues replaced by Phe, and expressed this in Rpb1 conditional knock-out cells (DT40-Rpb1; Hsin et al., 2011). Tyr1 was suggested to be essential for viability in S. cerevisiae (West and Corden, 1995), but not in S. pombe (Schwer and Shuman, 2011). To determine whether Tyr1 is required for growth in vertebrate cells, DT40-Rpb1 cells were transfected with the Rpb1-Y1F vector, and tetracycline (tet) was added to turn off wild-type Rpb1 expression. Rpb1-Y1F was unable to complement Rpb1, whereas Rpb1-26r fully restored viability (Figure 1—figure supplement 1A).

We next established cell lines stably expressing Rpb1-Y1F to analyze how the Y1F mutation affects Rpb1 function. Cells expressing Rpb1-Y1F (Y1F) stopped growing around 24 hr in medium containing tet (Figure 1A). To examine whether the inviability of Y1F cells might result from different Rpb1 levels, we analyzed several independent Y1F cell lines by Western blot (WB) with anti-FLAG antibodies (Abs). Rpb1-Y1F levels were indeed significantly reduced compared to Rpb1-26r (Figure 1B). Importantly, accumulation of a lower molecular weight form (indicated by *) was observed in all Y1F cell lines. This corresponds to a derivative likely precisely lacking the CTD, as it migrated slightly more rapidly than an Rpb1 derivative containing six heptads (Figure 1B).

Figure 1 with 1 supplement see all
Growth properties of Rpb1 cell lines.

(A) Cells were cultured in medium containing 1 µg/ml tetracycline (tet). Control cells, 26r, were split on day 2. Average cell counts from two independent experiments were plotted. (B) Cells were treated with tet for 24 hr. Whole-cell lysates from 26r, Y1F cells, and cells (6r) expressing an Rpb1 with 6 YSPTSPS repeats were analyzed by western blotting. Flag-tagged Rpb1 proteins were detected using Flag antibody. Partially degraded CTD-less Rpb1 is indicated by an asterisk (*). Full-length and degraded Rpb1 isoforms were quantified using ImageJ, and % degradation is displayed. nd, Degraded Rpb1 not detected. (C) Cell lysates from four independent 25F+Y and four independent Y1F cell lines were analyzed as in (B). Asterisk indicates partially degraded Rpb1. The lower molecular weight species in the 26r sample is of unknown identity and was not observed reproducibly, but was included in the quantitation. (D) Growth curves of two independent 25F+Y cell lines and 26r cells were plotted as in (A).

https://doi.org/10.7554/eLife.02112.003

To begin to investigate the basis for Rpb1-Y1F instability, we determined how many Tyr1 residues were necessary to restore stability. We first analyzed an Rpb1-Y1F derivative (20F+6Y) in which the F residues in the C-terminal six heptads were reverted to Y, and found that this derivative was completely stable (Figure 1—figure supplement 1B), although cells expressing Rpb1-20F+6Y remained inviable (Figure 1—figure supplement 1A). Next, we analyzed an Rpb1-Y1F derivative in which only a single F, in the C terminal-most heptad, was changed back to Y (Rpb1-25F+Y). Strikingly, this single Tyr residue was sufficient to prevent Rpb1 degradation, as the truncated isoform, which we denote Rpb1-b, was absent, and Rpb1-25F+Y levels were comparable to Rpb1-26r in multiple 25F+Y cell lines (Figure 1C; quantitation of the amount of degraded Rpb1 observed in multiple experiments is shown in Figure 1—figure supplement 1C). However, despite the restoration of Rpb1 stability, 25F+Y cells remained inviable (Figure 1D).

We next set out to determine how Tyr1 residues stabilize Rpb1. A first question was whether Rpb1 is indeed Tyr1-phosphorylated in DT40 cells. To address this, we utilized an anti-phospho-Tyr1 Ab (Mayer et al., 2012) to examine Tyr1 phosphorylation (Tyr1-P) of Rpb1-25F+Y and Rpb1-26r by WB; both proteins were indeed Tyr1-phosphorylated (Figure 2A). We next investigated where in cells the Rpb1-b isoform accumulates. We analyzed cytoplasmic, nuclear and chromatin-bound fractions from 26r and Y1F cells by WB with an N-terminal Rpb1 Ab (N20). Rpb1-b (indicated by *) was detected in all three fractions from Y1F cells, but barely or not at all in the 26r fractions (Figure 2B). The relative (and absolute) Rpb1-b levels were lowest in the cytoplasm, while Rpb1-b was essentially the only form on Y1F chromatin. As anticipated, Rpb1-b was not detected in 25F+Y cell fractions (Figure 2—figure supplement 1A). We next determined whether Tyr1-P could also be detected on Rpb1 in all three fractions, in this case using extracts from wild-type DT40 (Figure 2C) and human HEK293 (Figure 2—figure supplement 1B) cells. Robust Tyr1-P was indeed detected in all three fractions in both cell types. Notably, in both cytoplasm and nucleoplasm, Tyr1-P was observed only on hypophosphorylated Rpb1 (the lower band), while it was found primarily on the hyperphosphorylated isoform on chromatin. This suggests both that CTD phosphorylation is limited to Tyr1 in the cytoplasm and nucleoplasm and that Tyr1-P is present on hyperphosphorylated RNAP II found on active genes. We also examined phosphorylation on Ser 2, 5 and 7 and Thr4 (Figure 2C, Figure 2—figure supplement 1B). All these modifications were nearly undetectable in cytoplasmic and nuclear fractions, present almost exclusively on chromatin-associated, hyperphosphorylated Rpb1. Together, our data show that Tyr1, and only Tyr1, is phosphorylated before RNAP II engages in transcription, and support the idea that Tyr1-P functions in stabilizing the CTD when RNAP II is not transcribing, and perhaps also plays a role during transcription. Consistent with this, Tyr1-P was detected on Rpb1 immunoprecipitated by Abs recognizing Ser5-P and Ser2-P (Figure 2—figure supplement 1C).

Figure 2 with 1 supplement see all
Rpb1 Tyr1 phosphorylation is found in all cell fractions.

(A) Flag-tagged Rpb1 proteins were immunoprecipitated from cells treated with tet for 24 hr, and analyzed using western blotting. Phosphorylation on Tyr1 (Tyr1-P) was detected by the 3D12 antibody. (B) 26r and Y1F cells were treated with tet for 24 hr, subcellular fractionation was performed, and cytoplasmic, nuclear, and chromatin fractions were analyzed by western blotting. U2AF65 (a nuclear protein), and chromatin bound histone H3 protein served as controls for subcellular fractionation. The asterisk (*) indicates partially degraded Rpb1. (C) Wild-type DT40 cells were subjected to subcellular fractionation. The localization of Rpb1 phosphorylated on Tyr1, Ser 2, 5 and 7 and Thr4 was determined using antibodies as described in ‘Materials and methods’.

https://doi.org/10.7554/eLife.02112.005

We next wished to determine how the CTD is degraded, and whether Tyr1-P indeed plays a role. Since one Tyr residue in the final heptad confers stability, it is unlikely that endoproteolytic cleavage occurred between the CTD and the Rpb1 body. One possibility is that the CTD is degraded by the proteasome, which has been shown to associate both with RNAP II and with active genes (Gillette et al., 2004). Additionally, certain naturally unstructured proteins can be degraded by the proteasome in a ubiquitin-independent manner (Sheaff et al., 2000; Tofaris et al., 2001). Since the CTD is considered a structure-less domain (Meinhart et al., 2005), we hypothesized that the CTD is a natural proteasome substrate, and that Tyr1-P prevents its proteasomal degradation. To test this directly, we performed in vitro proteasome assays using a GST-CTD substrate (containing full-length wt CTD) and purified 20S proteasomes, and detected products by WB. Using an anti-GST Ab, the amount of full-length GST-CTD was greatly diminished and a ladder-like pattern of low molecular weight bands was detected, indicating that the proteasome degraded the GST-CTD protein from the C-terminus (Figure 3A, lane 1 and 2). Consistent with this, the low molecular weight products were undetectable by WB using an anti-CTD Ab (8WG16; Figure 3B). Notably, the 20S proteasome used was in a latent status with a closed gate and minimal enzymatic activity (Groll et al., 2000; Forster and Hill, 2003). Thus, GST-CTD, like for example the unstructured protein p21 (Forster and Hill, 2003), was capable of activating the 20S proteasome. Low concentrations of SDS render the proteasome gate disordered, leading to proteasome activation (Groll et al., 2000; Forster and Hill, 2003). Indeed, addition of 0.01% SDS to reaction mixtures increased CTD degradation (Figure 3A, lane 3 and 4). In contrast, the proteasome inhibitor MG132 inhibited degradation (Figure 3A, lane 5 and 6).

Figure 3 with 1 supplement see all
Tyr1 phosphorylation functions in CTD stability.

(A) In vitro 20S proteasome assay. 200 nM purified GST-CTD was incubated with 5 nM bovine 20S proteasome, and the reaction was carried with or without 0.01% SDS. MG132 was used to inhibit the proteasome, and reaction with 2.5% ethanol (ETOH) served as control. Western blotting was performed using antibody against GST, and the CTD (B). Position of 25 and 75 kD molecular weight markers are indicated. (C) 40 nM GST-CTD phosphorylated by recombinant c-Abl or by purified CDK7 complex was incubated with 2 nM 20S proteasome with or without 0.01% SDS for 1 hr, and reactions were analyzed by western blotting. (D) Y1F cells grown in the absence of tet were treated with 50 nM epoxomicin or 5 µM MG132 for indicated time. DMSO treatment served as control. Cell lysates were analyzed by western blotting. Partially degraded Rpb1 is indicated.

https://doi.org/10.7554/eLife.02112.007

We next asked whether phosphorylation of GST-CTD affects its stability in the proteasome assay. For this, we first used a recombinant c-Abl derivative to phosphorylate GST-CTD. This resulted in conversion of a fraction of the GST-CTD to a low-mobility, Tyr1-P isoform, although the majority remained unphosphorylated (Figure 3C, lane 1, Figure 3—figure supplement 1A), consistent with the processive phosphorylation by c-Abl observed previously (Duyster et al., 1995). We then performed the proteasome assay described above using c-Abl-phosphorylated GST-CTD (Figure 3C). Strikingly, the Tyr1 hyperphosphorylated GST-CTD (top panel, upper band, and lower panel) was resistant to degradation (lane 2), while the remaining unphosphorylated GST-CTD (top panel, bottom band) was degraded. Addition of 0.01% SDS again promoted degradation of unphosphorylated GST-CTD, but the Tyr1-P isoform remained resistant (lane 3). Significantly, GST-CTD phosphorylated by the Ser5/Ser7 kinase CDK7, which converted essentially all of the substrate to the hyperphosphorylated form, was not protected from degradation (lanes 4–6), indicating a specific role of Tyr1-P in preventing proteasomal degradation.

We next investigated whether the proteasome functions in Rpb1-Y1F degradation in vivo. We added the proteasomal inhibitors epoxomicin and MG132 to Y1F cells, and measured intact Rpb1 Y1F levels by WB (Figure 3D). Both inhibitors led to approximately twofold elevated levels of Rpb1-Y1F (see Figure 3—figure supplemental 1B for quantification). Although considerable truncated Rpb1-Y1F remained, this likely reflects incomplete proteasomal inhibition and/or accumulation of the truncated form prior to addition of the inhibitors. In any event, our data provide evidence that Tyr1, and specifically Tyr1-P, prevents proteasomal degradation of the CTD in vitro and in vivo.

We next wished to determine the genome-wide impact of the 25F+Y mutation on transcript levels. Using 3′READS (Hoque et al., 2013), a deep sequencing method to quantitate poly(A)+ RNAs, we analyzed 25F+Y and 26r cells, as well as S2A, S5A and T4V cells (all of which, like 25F+Y, are inviable; Hsin et al., 2011; Hsin et al., 2014) for comparison. Cells were treated with tet for 24 hr, and a total of ∼5 million reads mapping to 3′ regions of genes were generated for each cell type (Figure 4—figure supplement 1). Reads were classified into sense RNAs and upstream antisense (ua) RNAs (Figure 4A). uaRNAs were defined as transcripts that did not overlap any known protein-coding genes and used a poly(A) site within 2 kb from the TSS (Figure 4—figure supplement 2). Unexpectedly, the number of genes with upregulated uaRNAs was significantly greater than the number of genes with downregulated uaRNAs, by ∼16-fold (p=10−21.5), in 25F+Y cells (Figure 4B). S2A and S5A cells showed similar trends but to much lesser extents, fourfold (p=10−6.7) and 5.6-fold (p=10−9.0), respectively, while T4V cells in fact showed a trend in the opposite direction (Figure 4B). Using RT-qPCR, we validated several of the uaRNAs (Figure 4C). uaRNAs associated with the ARGLU1, METTL14, SH3BP5 and WEE1 genes were upregulated about twofold in two independent 25F+Y cell lines, consistent with results from RNA-seq (Figure 4—figure supplement 3A). Levels of RPLP1- and CCNB2-associated uaRNAs were indistinguishable in 26r and 25F+Y cells by both methods.

Figure 4 with 6 supplements see all
Tyr1 functions in expression of upstream antisense transcripts.

(A) Schematic of sense RNA and upstream antisense (ua) RNAs analyzed. (B) Regulation of uaRNA expression. Cells treated with tet for 24 hr were processed for 3′READS RNA-seq analysis. The number of genes with upregulated (UP) and downregulated (DN) uaRNAs (compared to sense RNA expression) are shown. Their ratio (UP/DN) and p-value (Chi-squared test) are shown. (C) RT-qPCR was performed to measure uaRNA levels associated with select genes detected in the RNA-Seq analysis. Fold relative to 26r cells is plotted. N = 3. (D) Rpb1 levels on sense and antisense genes were determined by ChIP using primers as indicated. The number in parenthesis next to the examined gene indicates the distance between the amplicon and the TSS (minus sign denotes upstream). N = 3. Error bars display standard deviation.

https://doi.org/10.7554/eLife.02112.009

We next investigated the mechanism underling the enhanced accumulation of uaRNAs in 25F+Y cells. uaRNAs are low-abundant, usually rapidly degraded by the nuclear exosome (Preker et al., 2008; Seila et al., 2009; Wei et al., 2011; Ntini et al., 2013). However, protein levels of four exosome subunits were comparable in 26r and 25F+Y cells (Figure 4—figure supplement 4), suggesting that the increase in uaRNAs was unlikely due to decreased exosome levels. Also, poly(A) sites of uaRNAs were unchanged (Figure 4—figure supplement 3B), indicating that enhanced accumulation did not reflect altered poly(A) site utilization. Another possibility was that transcription of these transcripts was increased. However, ChIP assays indicated that Rpb1 levels were in fact reduced upstream of the ARGLU1, METTL14, SH3BP5 and WEE1 genes in 25F+Y cells (Figure 4D; see also Figure 4—figure supplement 5). Finally, ChIP analyses showed more Tyr1-P on these upstream genes than on the corresponding downstream sense genes (Figure 4—figure supplement 6). Our results point to a role for Tyr1-P in regulating accumulation of uaRNAs by contributing to their rapid turnover.

In this study, we described two important functions for Tyr1 residues: Protecting the CTD from proteolysis and ensuring turnover of uaRNAs. Both these functions are likely conserved throughout vertebrates, as Descostes et al. (Descostes et al., 2014) report remarkably similar results in human cells. While CTD stabilization requires only a single Tyr1 residue, and Tyr1 phosphorylation, more Tyr1 residues are required for other essential functions. As shown previously (Baskaran et al., 1993, 1999), c-Abl can phosphorylate the CTD in vitro, which stabilizes it from proteasomal degradation. But the role of c-Abl in Tyr1 phosphorylation in vivo is unclear, as c-Abl inhibitors had at most modest effects on Try1-P levels in cells (unpublished data). In any event, our results add additional and unexpected complexity to the multiple functions played by the CTD in controlling RNAP II activity.

Materials and methods

Cell culture and cloning

Request a detailed protocol

DT40 cells and HEK293 cells were cultured at 37°C with 5% CO2 in RPMI1640 medium containing 10% FBS and 1% chicken serum, and in DMEM medium containing 10% FBS, respectively.

Rpb1 CTD derivatives were cloned as previously described (Hsin et al., 2011). Briefly, a fragment of beta-actin promoter and FLAG tag was inserted into pBlueScript containing Neomycin resistance gene. The human Rpb1 body without the CTD was inserted immediately after the FLAG tag, and various CTD fragments were inserted directly 3′ to the Rpb1 body.

Complementation assay and construction of stable cell lines

Request a detailed protocol

Procedures for complementation assays and for constructing stable cell clines were followed as previously described (Hsin et al., 2011). Briefly, 107 cells were transfected with linearized DNA, and selected in the presence of appropriate antibiotics. Surviving cell clones were isolated, and the identity of these cells was further confirmed using western blotting.

Western blotting

Request a detailed protocol

Cells lysates, prepared by dissolving washed cells directly in SDS sample buffer, were resolved by SDS-PAGE with indicated percentage of acrylamide. Western blotting was performed using standard protocols. For quantification, western blots were analyzed by ImageJ. Antibodies used in this paper as follows: Flag tag (M2; Sigma, St. Louis, MO), actin (Sigma), phospho CTD Tyr1 (3D12; Active Motif, Carlsbad, CA), U2AF65 (Sigma), histone H3 protein (abcam, Cambridge, MA), phospho CTD Ser2 (3E10; Millipore, Billerica, MA), phospho CTD Ser5 (3E8; Millipore), phospho CTD Ser 7 (4E12, Millipore), Rpb1 CTD (8WG16; abcam), GST tag (Invitrogen, Carlsbad, CA), Rpb1 (N20; Santa Cruz, Santa Cruz, CA), Exosc10 (Rrp6) (Novus, Littleton, CO), Exosc9 (Rrp45) (Novus), Exosc3 (Rrp40) (Novus), and Dis3 (Novus).

Subcellular fractionation

Request a detailed protocol

Subcellular fractionation was performed using a modified protocol as described (Mapendano et al., 2010). Briefly, cells (1 ∼ 2 × 107) were harvested, washed in PBS, and resuspended in 0.5 ml of RSB100 (50 mM Tris–HCl PH 7.4, 100 mM NaCl) containing 40 µg/ml digitonin. Cell extracts were incubated on ice for 5 min. The cytoplasmic fraction was separated from nuclear fraction by centrifugation (2000×g, 5 min). The pellets were resuspended in 0.5 ml of RSB100 containing 0.5% Triton X-100, and the reactions were incubated on ice for 5 min. Separation of soluble nuclear proteins from insoluble chromatin bound proteins was carried by centrifugation (2000×g, 5 min). The pellets containing chromatin bound proteins were resuspended in 0.5 ml of RSB100 (0.5% Triton-X100), and sonicated briefly.

In vitro proteasome assay

Request a detailed protocol

In vitro proteasome assays were performed as described (Asher et al., 2005) with the following modifications. Briefly, GST-CTD or GST-CTD phosphorylated by abl tyrosine kinase was incubated with 2–10 nM bovine 20S proteasome (UBPBio, Aurora, CO) in a buffer (50 mM Tris–HCl PH 7.4, 100 mM NaCl, 0.5 mM EDTA) at 37°C for 1 hr. Reactions were stopped by adding equal volume of 2X SDS PAGE sampling buffer.

In vitro phosphorylation of GST-CTD

Request a detailed protocol

CDK7 complexes were expressed in insect cells, and purified using Ni-NTA agarose (QIAGEN, Valencia, CA) as described (Larochelle et al., 2006). GST-CTD was expressed in E. coli and purified using glutathione Sepharose 4B (GE Healthcare). Phosphorylation of GST-CTD by CDK7 complexes was carried out at 30°C for 1 hr in a kinase buffer (25 mM Hepes PH 7.5, 10 mM MgCl2, 150 mM NaCl, 1 mM ATP). GST-CTD phosphorylation by recombinant c-Abl kinase (NEB, Ipswich, MA) was performed as described in manual. Briefly, 500 nM GST-CTD was incubated with 25 U c-Abl at 30°C for 2 hr. Phosphorylated GST-CTD was purified using glutathione Sepharose 4B (GE Healthcare, Pittsburgh, PA).

RT-qPCR

Request a detailed protocol

RNA was extracted using Trizol (Invitrogen), and further treated with DNase I. Reverse transcription and qPCR analysis were performed as previously described (Hsin et al., 2011). Primer sequences are listed in Supplementary file 1.

Immunoprecipitation

Request a detailed protocol

About 2 × 107 cells were collected, and washed with PBS. Then, 1 ml cold RIPA (150 mM NaCl, 1 mM EDTA, 50 mM Tris–HCl pH 7.4, 0.5% NP-40, 0.25% sodium deoxycholate) buffer containing 1X PhosSTOP (Roche, Madison, WI), and 1X protease inhibitors (1.4 µg/ml Pepstatin A, 0.35 µg/ml Leupeptin, and 1.7 µg/ml Aprotinin). After brief sonication, debris was centrifuged at 12,000×g, 4°C, for 10 min, and the supernatant was removed to a new tube. 50 µl of the lysate were kept for input control, and the rest of the extract was incubated with 20 µl of pre-washed protein G Sepharose and 1–4 µg of antibody. Samples were rotated at 4°C for 1–2 hr, and beads were washed with cold RIPA buffer for 3 min three times, and then were resuspended in 100 ul of 1X SDS sample buffer for western blotting.

Chromatin immunoprecipitation (ChIP)

Request a detailed protocol

Cells were grown to 70% confluence (∼2 × 106/ml), cross-linked with 1% formaldehyde for 10 min, and processed for ChIP as previously described (Hsin et al., 2011). ChIP was performed using antibody against Flag tag (M2; Sigma), phospho CTD Tyr1 (3D12; Active Motif). Primers sequences are listed in Supplementary file 1.

Deep sequencing and data analysis

Request a detailed protocol

Total RNA was extracted from cells treated with tet for 24 hr. RNA was further processed, and analyzed by 3′READs, a deep sequencing method to analyze poly(A)+ RNAs using 3′ end regions, as described (Hoque et al., 2013) with some modifications for RNA fragmentation. Briefly, poly(A) RNA was selected using oligo d(T)25 magenetic beads (NEB), followed by fragmentation of RNA on-bead using RNaseIII (NEB). We generated two libraries for each cell type (biological replicates), and ∼4 million reads for each library. cDNA insert size range corresponded to RNA fragments of ∼100–200 nt. The reads were mapped to the chicken genome (version galGal4), and those with at least two non-genomic As at the 3′ end were considered as poly(A) site-supporting (PASS) reads. PASS reads were assigned to protein coding genes defined by Refseq. The 3′ end of each gene was extended by 4 kb if there was no gene on the same strand within this region. The PASS reads mapped to genic regions are called sense strand reads. Those mapped to the 2 kb upstream region of transcription start site (TSS) on the antisense strand were called upstream antisense (ua) RNA reads. We also required that uaRNA reads could not be assigned to any other annotated genes as sense strand reads. To examine expression change of uaRNAs vs sense RNAs, we grouped all uaRNA reads and compared them to all sense reads of a gene between two samples, for example, 25F+Y vs 26r. Genes with p-value <0.05 (Fisher′s exact test) were selected.

References

    1. Baskaran R
    2. Dahmus ME
    3. Wang JYJ
    (1993)
    Tyrosine phosphorylation of mammalian RNA polymerase II carboxyl-terminal domain
    Proceedings of the National Academy of Sciences of the United States of America 90:11167–11171.
    1. Baskaran R
    2. Escobar SR
    3. Wang JYJ
    (1999)
    Nuclear c-Abl is a COOH-terminal repeated domain (CTD)-tyrosine (CTD)-tyrosine kinase-specific for the mammalian RNA polymerase II: possible role in transcription elongation
    Cell Growth & Differentiation 10:387–396.
    1. Duyster J
    2. Baskaran R
    3. Wang JY
    (1995)
    Src homology 2 domain as a specificity determinant in the c-Abl-mediated tyrosine phosphorylation of the RNA polymerase II carboxyl-terminal repeated domain
    Proceedings of the National Academy of Sciences of the United States of America 92:1555–1559.
    1. Forster A
    2. Hill CP
    (2003)
    Proteasome degradation: enter the substrate
    Trends in Cell Biology 13:550–553.
    1. Seila AC
    2. Core LJ
    3. Lis JT
    4. Sharp PA
    (2009)
    Divergent transcription: a new feature of active promoters
    Cell Cycle 8:2557–2564.
    1. Sheaff RJ
    2. Singer JD
    3. Swanger J
    4. Smitherman M
    5. Roberts JM
    6. Clurman BE
    (2000)
    Proteasomal turnover of p21Cip1 does not require p21Cip1 ubiquitination
    Molecular Cell 5:403–410.
    1. Tofaris GK
    2. Layfield R
    3. Spillantini MG
    (2001)
    alpha-synuclein metabolism and aggregation is linked to ubiquitin-independent degradation by the proteasome
    FEBS Letters 509:22–26.
    1. West ML
    2. Corden JL
    (1995)
    Construction and analysis of yeast RNA polymerase II CTD deletion and substitution mutations
    Genetics 140:1223–1233.

Article and author information

Author details

  1. Jing-Ping Hsin

    Department of Biological Sciences, Columbia University, New York, United States
    Contribution
    J-PH, Conception and design, Acquisition of data, Analysis and interpretation of data, Drafting or revising the article, Contributed unpublished essential data or reagents
    Competing interests
    No competing interests declared.
  2. Wencheng Li

    Department of Biochemistry and Molecular Biology, Rutgers University New Jersey Medical School, Newark, United States
    Contribution
    WL, Acquisition of data, Analysis and interpretation of data, Contributed unpublished essential data or reagents
    Competing interests
    No competing interests declared.
  3. Mainul Hoque

    Department of Biochemistry and Molecular Biology, Rutgers University New Jersey Medical School, Newark, United States
    Contribution
    MH, Acquisition of data, Analysis and interpretation of data, Contributed unpublished essential data or reagents
    Competing interests
    No competing interests declared.
  4. Bin Tian

    Department of Biochemistry and Molecular Biology, Rutgers University New Jersey Medical School, Newark, United States
    Contribution
    BT, Acquisition of data, Analysis and interpretation of data, Drafting or revising the article, Contributed unpublished essential data or reagents
    Competing interests
    No competing interests declared.
  5. James L Manley

    Department of Biological Sciences, Columbia University, New York, United States
    Contribution
    JLM, Conception and design, Analysis and interpretation of data, Drafting or revising the article
    For correspondence
    jlm2@columbia.edu
    Competing interests
    JLM: Senior Editor, eLife.

Funding

National Institutes of Health (GM097174)

  • James L Manley

National Institutes of Health (GM084089)

  • Bin Tian

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Acknowledgements

We thank our lab members, especially Patricia Richard, for many insightful discussions, and Takashi Yamazaki for experimental support. We thank Drs Stephane Larochelle and Robert Fisher for CDK7 baculoviruses, and Drs Dirk Eick and Jean-Christophe Andrau for communicating results prior to publication. This work was funded by NIH grants GM 097174 to JLM and GM084089 to BT.

Reviewing Editor

  1. Danny Reinberg, Howard Hughes Medical Institute, New York University School of Medicine, United States

Version history

  1. Received: December 18, 2013
  2. Accepted: May 6, 2014
  3. Accepted Manuscript published: May 8, 2014 (version 1)
  4. Version of Record published: June 3, 2014 (version 2)

Copyright

© 2014, Hsin et al.

This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 2,771
    Page views
  • 279
    Downloads
  • 36
    Citations

Article citation count generated by polling the highest count across the following sources: Crossref, Scopus, PubMed Central.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Jing-Ping Hsin
  2. Wencheng Li
  3. Mainul Hoque
  4. Bin Tian
  5. James L Manley
(2014)
RNAP II CTD tyrosine 1 performs diverse functions in vertebrate cells
eLife 3:e02112.
https://doi.org/10.7554/eLife.02112

Share this article

https://doi.org/10.7554/eLife.02112

Further reading

    1. Biochemistry and Chemical Biology
    2. Structural Biology and Molecular Biophysics
    Karolina Honzejkova, Dalibor Kosek ... Tomas Obsil
    Research Article

    Apoptosis signal-regulating kinase 1 (ASK1) is a crucial stress sensor, directing cells toward apoptosis, differentiation, and senescence via the p38 and JNK signaling pathways. ASK1 dysregulation has been associated with cancer and inflammatory, cardiovascular, and neurodegenerative diseases, among others. However, our limited knowledge of the underlying structural mechanism of ASK1 regulation hampers our ability to target this member of the MAP3K protein family towards developing therapeutic interventions for these disorders. Nevertheless, as a multidomain Ser/Thr protein kinase, ASK1 is regulated by a complex mechanism involving dimerization and interactions with several other proteins, including thioredoxin 1 (TRX1). Thus, the present study aims at structurally characterizing ASK1 and its complex with TRX1 using several biophysical techniques. As shown by cryo-EM analysis, in a state close to its active form, ASK1 is a compact and asymmetric dimer, which enables extensive interdomain and interchain interactions. These interactions stabilize the active conformation of the ASK1 kinase domain. In turn, TRX1 functions as a negative allosteric effector of ASK1, modifying the structure of the TRX1-binding domain and changing its interaction with the tetratricopeptide repeats domain. Consequently, TRX1 reduces access to the activation segment of the kinase domain. Overall, our findings not only clarify the role of ASK1 dimerization and inter-domain contacts but also provide key mechanistic insights into its regulation, thereby highlighting the potential of ASK1 protein-protein interactions as targets for anti-inflammatory therapy.

    1. Biochemistry and Chemical Biology
    Jake W Anderson, David Vaisar ... Natalie G Ahn
    Research Article

    Activation of the extracellular signal-regulated kinase-2 (ERK2) by phosphorylation has been shown to involve changes in protein dynamics, as determined by hydrogen-deuterium exchange mass spectrometry (HDX-MS) and NMR relaxation dispersion measurements. These can be described by a global exchange between two conformational states of the active kinase, named ‘L’ and ‘R,’ where R is associated with a catalytically productive ATP-binding mode. An ATP-competitive ERK1/2 inhibitor, Vertex-11e, has properties of conformation selection for the R-state, revealing movements of the activation loop that are allosterically coupled to the kinase active site. However, the features of inhibitors important for R-state selection are unknown. Here, we survey a panel of ATP-competitive ERK inhibitors using HDX-MS and NMR and identify 14 new molecules with properties of R-state selection. They reveal effects propagated to distal regions in the P+1 and helix αF segments surrounding the activation loop, as well as helix αL16. Crystal structures of inhibitor complexes with ERK2 reveal systematic shifts in the Gly loop and helix αC, mediated by a Tyr-Tyr ring stacking interaction and the conserved Lys-Glu salt bridge. The findings suggest a model for the R-state involving small movements in the N-lobe that promote compactness within the kinase active site and alter mobility surrounding the activation loop. Such properties of conformation selection might be exploited to modulate the protein docking interface used by ERK substrates and effectors.