Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Mechanism of mRNA deadenylation: evidence for a molecular interplay between translation termination factor eRF3 and mRNA deadenylases

Yuji Funakoshi,1,2,6 Yusuke Doi,1,6 Nao Hosoda,1,6 Naoyuki Uchida,3,6 Masanori Osawa,4 Ichio Shimada,4 Masafumi Tsujimoto,2 Tsutomu Suzuki,5 Toshiaki Katada,3 and Shin-ichi Hoshino1,7 1Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; 2Laboratory of Cellular Biochemistry, RIKEN, Wako, Saitama 351-0198, Japan; 3Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan; 4Department of Physical Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan; 5Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, Tokyo 113-0033, Japan

,In eukaryotes, shortening of the 3؅-poly(A) tail is the rate-limiting step in the degradation of most mRNAs and two major mRNA deadenylase complexes—Caf1–Ccr4 and Pan2–Pan3—play central roles in this process, referred to as deadenylation. However, the molecular mechanism triggering deadenylation remains elusive. Previously, we demonstrated that eukaryotic releasing factor eRF3 mediates deadenylation and decay of mRNA in a manner coupled to translation termination. Here, we report the mechanism of mRNA deadenylation. The eRF3-mediated deadenylation is catalyzed by both Caf1–Ccr4 and Pan2–Pan3. Interestingly, translation termination complexes eRF1–eRF3, Pan2–Pan3, and Caf1–Ccr4 competitively interact with polyadenylate-binding PABPC1. In each complex, eRF3, Pan3, and Tob, respectively, mediate PABPC1 binding, and a combination of a PAM2 motif and a PABC domain is commonly utilized for their contacts. A translation-dependent exchange of eRF1–eRF3 for the deadenylase occurs on PABPC1. Consequently, PABPC1 binding leads to the activation of Pan2–Pan3 and Caf1–Ccr4. From these results, we suggest a mechanism of mRNA deadenylation by Pan2–Pan3 and Caf1–Ccr4 in cooperation with eRF3 and PABPC1. [Keywords: Translation termination; deadenylation; eRF3; PABPC1] Supplemental material is available at http://www.genesdev.org. Received July 26, 2007; revised version accepted October 11, 2007.

Control of mRNA decay is a fundamentally important Two distinct enzyme complexes involved in the de- step in determining the amount of protein produced adenylation process have been identified in yeast from the mRNA by translation, and mRNA decay is in- (Daugeron et al. 2001; Tucker et al. 2001) and mammals timately linked to and regulated by translation. In eu- (Yamashita et al. 2005). One major cytoplasmic dead- karyotes, decay of most mRNAs is initiated by shorten- enylase complex is Ccr4–Caf1, which was originally im- ing of the poly(A)-tail at the 3Ј end (Shyu et al. 1991; plicated in the control of transcription (Denis and Chen Muhlrad and Parker 1992), which is followed by two gen- 2003). As suggested by the significant homology with eral pathways: the 5Ј-to-3Ј and 3Ј-to-5Ј exonucleolytic known 3Ј-to-5Ј exonucleases, both Ccr4 and Caf1 have degradation pathways. The poly(A) tail-shortening pro- catalytic activity of the deadenylase (Chen et al. 2002; cess, referred to as deadenylation, is the first and rate- Tucker et al. 2002; Thore et al. 2003; Viswanathan et al. limiting step, and is also the most efficient step in con- 2004; Bianchin et al. 2005). Another major deadenylase trolling mRNA decay (Decker and Parker 1993). complex, PAN, was identified in yeast (Sachs and Dear- dorff 1992) and in mammal (Uchida et al. 2004). PAN is a heterodimeric protein consisting of catalytic Pan2 and regulatory Pan3 subunits (Brown et al. 1996; Uchida et 6These authors contributed equally to this work. al. 2004). In contrast to Ccr4–Caf1, PAN is characterized 7Corresponding author. E-MAIL [email protected]; FAX 81-52-836-3427. by the requirement of PABPC1 for its deadenylating ac- Article is online at http://www.genesdev.org/cgi/doi/10.1101/gad.1597707. tivity: Pan2 is activated by binding to PABPC1 through

GENES & DEVELOPMENT 21:3135–3148 © 2007 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/07; www.genesdev.org 3135 Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Funakoshi et al. the interaction with Pan3 (Mangus et al. 2004; Uchida terplay between translation termination factor eRF3 and et al. 2004). mRNA deadenylases. We demonstrate that eRF3-medi- Since deadenylation is the rate-limiting step for ated deadenylation is catalyzed by both of the two major mRNA decay, the enzymatic activities of the two mRNA deadenylase complexes in yeast and humans. In- mRNA deadenylases constitute a major target for the terestingly, translation termination complex eRF1–eRF3 control of mRNA decay. Recent findings demonstrated and the two mRNA deadenylase complexes competi- that selective recruitment of deadenylating enzymes to tively bind to the PABC domain of PABPC1. The PAM2 the substrate mRNA is involved in the decay mecha- motifs found in both eRF3 and the two deadenylase com- nism for specific mRNAs (Semotok et al. 2005; Finoux plexes are responsible for their binding to PABPC1 and and Seraphin 2006; Goldstrohm et al. 2006). However, mRNA deadenylation. The termination complex and the the general mechanism triggering deadenylation of deadenylase complex are exchanged on PABPC1 in a mRNA has not been elucidated. Several lines of evidence translation-dependent manner. Recruitment of the two have revealed a direct link between translation termina- deadenylase complexes to PABPC1 leads to the activa- tion and mRNA decay. tion of both enzymes. These results provide a molecular In eukaryotes, translation termination is governed by basis for the mechanism of mRNA deadenylation. heterodimeric releasing factors eRF1 and eRF3 (Frolova et al. 1994; Stansfield et al. 1995; Zhouravleva et al. 1995). eRF1 directly recognizes all three stop codons in Results the ribosomal A site to release the completed polypep- The two major mRNA deadenylases, Pan2–Pan3 tide chain from the ribosome (Frolova et al. 1994), and and Caf1–Ccr4, are involved in the eRF3-mediated eRF3 stimulates the termination reaction in a GTP-de- deadenylation of mRNA in S. cerevisiae pendent manner (Zhouravleva et al. 1995). The eRF3 is conserved from yeast to mammals. In the yeast Using S. cerevisiae as a model organism, we showed pre- Saccharomyces cerevisiae, eRF3 is encoded by the essen- viously that eRF3 mediates deadenylation and decay of tial gene SUP35 (Stansfield et al. 1995), and the mam- normal mRNA in a manner coupled to translation ter- malian eRF3 gene GSPT was first identified by its ability mination (Hosoda et al. 2003). To identify the deadenyl- to complement the temperature-sensitive growth arrest ases involved in the eRF3-mediated deadenylation, we phenotype of a sup35/gst1-1 mutant S. cerevisiae that is first examined the possible involvement of the two defective in the G1-to-S-phase transition (Hoshino et al. major mRNA deadenylases, Pan2–Pan3 and Caf1–Ccr4, 1989). Later, two subtypes of GSPT , GSPT1 and by comparing phenotypes in strains pan2⌬ and ⌬, GSPT2, were identified (Hoshino et al. 1998). eRF3 con- and a strain in which the N-terminal domain of eRF3 sists of two domains: the unique N-terminal region (N (SUP35N) was overexpressed. In this study, we used the domain) and the C-terminal region (C domain), which strain overexpressing SUP35N, since this strain and the contains eEF1A-like GTP-binding motifs. The C domain eRF3 mutant strains sup35⌬N and sup35ts show the of eRF3 is sufficient to fulfill the functions of eRF3 in same phenotypes regarding mRNA deadenylation and termination (Zhouravleva et al. 1995). decay (Hosoda et al. 2003). We first analyzed the poly(A) In addition to translation termination, eRF3 functions tail length of total steady-state mRNA in vivo. After in translation termination-coupled events. We showed labeling with [32P]pCp, total RNA was digested with previously that eRF3 interacts with poly(A)-binding pro- RNaseA, and the remaining intact poly(A) tails were re- tein (PABPC1) through its N domain (Hoshino et al. solved on polyacrylamide gels (Fig. 1A,B). As reported 1999). The interaction is evolutionarily conserved from previously, the overexpression of SUP35N led to an in- yeast to mammals (Hoshino et al. 1999; Kozlov et al. crease in bulk mRNA poly(A) tail lengths (Hosoda et al. 2001; Cosson et al. 2002; Uchida et al. 2002; Hosoda et 2003). Similarly, a deletion in PAN2 also resulted in an al. 2003). In yeast, eRF3 regulates the initiation of nor- increase in the average poly(A) tail length. PhosphorImager mal mRNA decay at the poly(A) tail-shortening step analysis showed that the average length increased ∼20 through the interaction with PABPC1 in a manner nucleotides (nt) in both strains compared with the wild- coupled to translation termination (Hosoda et al. 2003). type strain. The changes in poly(A) tail length on mRNA Recently, Tpa1—which interacts with eRF1/eRF3 and in pan2⌬ overexpressing SUP35N were comparable with affects translation termination, deadenylation, and those found in either pan2⌬ or the SUP35N-overexpress- mRNA decay—was identified (Keeling et al. 2006). On ing strain. Thus, the deadenylation defect in the the other hand, in nonsense codon-containing mRNA, SUP35N-overexpressing strain was not exacerbated by translation termination is thought to occur via the ter- pan2⌬. These results suggest that Pan2 is involved at mination complex eRF1/eRF3 at the premature termina- least in part in eRF3-mediated deadenylation. tion codon. eRF1/eRF3 associates with Upf1/Upf2/Upf3 However, a previous study reported that pan2⌬ does to form a “surveillance complex” and triggers rapid not show any defect in the rate of mRNA deadenylation mRNA decay via the nonsense-mediated mRNA decay or decay (Tucker et al. 2001). Therefore, we next exam- (NMD) pathway (Wang et al. 2001; Kobayashi et al. 2004; ined the decay of an MFA2pG reporter transcript that Kashima et al. 2006). was under the control of a tetracycline-regulatable pro- In this study, we examined the mechanism of mRNA moter system. In this system, the reporter gene is spe- deadenylation and obtained evidence for a molecular in- cifically and rapidly suppressed in the presence of doxy-

3136 GENES & DEVELOPMENT Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Mechanism of mRNA deadenylation

Figure 1. Comparison of the length of poly(A) tails and decay rates in S. cerevisiae.(A) pKT10 or pKT10-SUP35N, which overexpresses the N domain of eRF3 (SUP35N) under the control of GAL1 UAS, was introduced into wild-type, pan2⌬, and ccr4⌬ strains. Total RNAs at the steady-state level from the strains were 3Ј-la- beled with [32P]pCp and digested with RNaseA. The remaining mRNA poly(A) tails were sepa- rated on a 12% polyacrylamide–7 M urea gel and visualized by autoradiography. (B) The resulting autoradiograph from A was analyzed by densito- metric tracing. (C) p9L-MFA2pG was introduced into the same strains as used in A. After transcrip- tional repression by adding doxycycline, the cells were collected at the indicated time points. (Left) Total RNAs were separated on a 1% formamide- agarose gel, and MFA2pG mRNA was detected by Northern blotting. (Right) The amount of MFA2pG mRNAs at each time point was mea-

sured. The half-lives (t1/2) of the mRNAs were derived from three independent experiments and are shown as the average and the standard error of the mean (SEM) in the left panel. (D) The dead- enylation rates of MFA2pG mRNA poly(A) tails in the strains shown in C were analyzed by a polyacrylamide Northern blot analysis. (Left) The RNA samples prepared in C were separated on a 4% polyacrylamide–8 M urea gel. The RNA samples from 0-min time points were treated with RNaseH and oligo(dT) to indicate the posi- tion of the deadenylated mRNA. (Right) The sizes of the longest poly(A) tails were measured.

⌬ cycline. The decay rate of the mRNA was measured after reduced in the strain ccr4 (t1/2 = 68 min and 0.35 nt per transcriptional shutoff by the addition of doxycycline. minute) (Tucker et al. 2001), which is similar to the phe- Consistent with previous reports, pan2⌬ did not show notype of the SUP35N-overexpressing strain. In addi- any apparent defect in the decay of the MFA2pG mRNA tion, SUP35N had no significant effect on the rates of ⌬ ⌬ (t1/2 = 15 min, cf. wild type and pan2 ) (Fig. 1C). In ad- both deadenylation and decay of the mRNA in the ccr4 dition, the poly(A) tail of MFA2pG examined using high- strain but led to an increase in the poly(A) tail length of resolution polyacrylamide Northern blot shortened at a either MFA2 mRNA or bulk mRNA in the ccr4⌬ strain rate of 2.0 nt per minute, which was comparable with at steady state (Fig. 1, cf. D and A). Thus, the phenotype the rate in wild-type cells (Fig. 1D). In sharp contrast, of the longer poly(A) tail of steady-state mRNA in the both the decay and the poly(A) tail-shortening rates of SUP35N-overexpressing strain is explained by the defect the MFA2pG mRNA were substantially reduced in the in Pan2, and the phenotype of the reduced rates of both strain expressing SUP35N (t1/2 = 46 min and 0.45 nt per deadenylation and decay is explained by the defect in minute), although the distribution of the poly(A) tail Ccr4. Taken together, these results indicate that two major length at time 0 was comparable with that of pan2⌬. mRNA deadenylases, Pan2–Pan3 and Caf1–Ccr4, are in- Furthermore, overexpression of SUP35N reduced the volved in the eRF3-mediated deadenylation of mRNA. rates of both the decay and the poly(A) tail shortening of the mRNA in the pan2⌬ strain. These results suggest The two major mRNA deadenylases, hPan2–hPan3 that additional deadenylases are involved in eRF3-medi- and hCaf1–hCcr4, are involved in the eRF3-mediated ated deadenylation and decay. In view of the defect in the deadenylation of mRNA in mammals deadenylation rates, Ccr4 is one such candidate. As re- ported previously, both the decay and the poly(A) tail Next, we examined whether hPan2–hPan3 and hCaf1– shortening of the MFA2pG mRNA were substantially hCcr4 are also involved in the eRF3-mediated mRNA

GENES & DEVELOPMENT 3137 Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Funakoshi et al. deadenylation in mammalian cells. We monitored deadenylation (Fig. 2B; Supplementary Fig. 1) and en- mRNA deadenylation and decay kinetics by using a tet- hanced the decay of the mRNA body in the second phase racycline regulatory transcriptional pulse-chase ap- (Fig. 2B, lane 7; Supplementary Fig. 1C). In sharp con- proach in HeLa cells. In this experiment, ␤-globin trast to this, a nuclease-deficient hCaf1 mutant in which mRNA was used as a reporter, since the transcript is well a catalytically essential aspartate residue was mutated characterized and therefore provides an ideal model sys- to alanine drastically reduced the rate of deadenylation tem to study mRNA stability. T-Rex-HeLa cells were and decay of mRNA body in the second phase and sta- transfected with the ␤-globin gene, transcription of the bilized the mRNA (Fig. 2B, lanes 13–15; Supplementary gene was driven by tetracycline for a short period of Fig. 1C). The expression of hPan2 or hCaf1 protein was time, and the metabolism of newly transcribed mRNAs confirmed by Western blot (Fig. 2D,E). These results con- was followed over time. As reported previously, ␤-globin firm the previous results showing that hPan2–hPan3 and mRNA showed slow and biphasic decay kinetics with a hCaf1–hCcr4 function in the first and second phases of half-life of ∼12 h (Fig. 2A, left panel). A slow and syn- mRNA deadenylation, respectively (Yamashita et al. chronous deadenylation occurred in the first phase (0–3 2005). h), which was followed by a fast and less-synchronous We then examined whether eRF3 mediates deadenyla- deadenylation with a concomitant decay of the mRNA tion of mRNA in HeLa cells. As shown in Figure 2C, body (second phase) (Supplementary Fig. 1C, left panel). ectopic expression of an N-terminal domain of eRF3 Overexpression of wild-type hPan2 increased the rate of (eRF3N) reduced the rate of deadenylation in both the deadenylation in the first phase (0–3 h) (Fig. 2A, lanes first and the second phases. The expression of the eRF3N 6,7), whereas a comparable amount of hPan2 mutant was confirmed by Western blot (Fig. 2F). Collectively, that had no deadenylase activity (Uchida et al. 2004) re- these results suggest that eRF3 mediates deadenylation duced the rate (Fig. 2A, lanes 11,12). On the other hand, through hPan2–hPan3 and hCaf1–hCcr4 in mammalian overexpression of wild-type hCaf1 increased the rate of cells.

Figure 2. Comparison of deadenylation rates in HeLa cells. (A) T-Rex-HeLa cells were transfected with pFlag-CMV5/TO-G1 reporter and either pFlag-CMV5 (lanes 1–5), pFlag-hPan2 (lanes 6–10), or pFlag-hPan2 D1085A (lanes 11–15). One day later, ␤-globin mRNA was induced by tetracycline for 2.5 h, and then cells were harvested at the specified time after ␤-globin mRNA transcription shutoff. (Left) ␤-Globin mRNA was detected by Northern blot analysis. (Lane 16)To determine the overall poly(A) tail length and the dead- enylation endpoint (arrow) of ␤-globin mRNA, ␤-glo- bin mRNAs at the steady-state level (24-h induction) are shown. (Right) The average length of poly(A) tails at each time point was measured. (B)AsinA, except that cells were transfected with pHA-CMV5 (lanes 1–5), pHA-hCaf1 (lanes 6–10), or pHA-hCaf1 D161A (lanes 11–15), and transcriptional pulse-chase analysis was performed. (C)AsinA, except that cells were transfected with pCMV-Myc (lanes 1–5) or pCMV- Myc-eRF3 N (lanes 6–10), and transcriptional pulse- chase analysis was performed. (D–F) Total cell lysates were analyzed by immunoblotting using anti-Flag (D, top), anti-HA (E, top), anti-Myc (F, top), or anti- GAPDH (D–F, bottom).

3138 GENES & DEVELOPMENT Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Mechanism of mRNA deadenylation hPan3 and eRF3 competitively bind to PABPC1 PABPC1 but not eRF3 (data not shown). These results suggest that both hPan3 and eRF3 bind with PABPC1, In mammals, we showed that eRF3 as well as hPan3 but each protein is part of a distinct complex, and eRF3 interacts with PABPC1 (Hoshino et al. 1999; Uchida et might compete with hPan3 for PABPC1 binding. To test al. 2002, 2004). The present results showing that eRF3- this idea, we examined whether hPan3 and eRF3 interact mediated deadenylation is catalyzed in part by hPan2– with the same region of PABPC1. Flag-PABPC1 or Flag- hPan3 (Fig. 2) led us to speculate that, in mammalian PABPC1⌬C lacking the eRF3-binding domain was coex- cells, eRF3 mediates mRNA deadenylation by hPan2– pressed with HA-hPan3, and an immunoprecipitation as- hPan3 through interaction with PABPC1. Therefore, we say using anti-Flag antibody was performed. As shown in investigated the interactions among eRF3, hPan2– Figure 3B, Flag-PABPC1, but not Flag-PABPC1⌬C was hPan3, and PABPC1. To this end, we first examined capable of coprecipitating with eRF3, which is consis- whether eRF3 and hPan3 could simultaneously interact tent with our previous study using a yeast two-hybrid with PABPC1. When an extract of COS-7 cells express- system (Hoshino et al. 1999). A similar result was also ing Flag-eRF3 and HA-hPan3 was immunoprecipitated obtained for the coprecipitation of HA-hPan3 (Fig. 3B, with anti-Flag antibody, the precipitated fraction con- top). Thus, both eRF3 and hPan3 require the C-terminal tained endogenous PABPC1 but not HA-hPan3 (Fig. 3A, domain of PABPC1 for their binding. lane 4). On the other hand, anti-Flag immunoprecipitate Next, we examined whether hPan3 actually competes from extracts of cells expressing Flag-hPan3 contained with eRF3 for PABPC1 binding. A complex consisting of

Figure 3. hPan3 and eRF3 competitively bind to PABC domain of PABPC1 via their PAM2 motifs. (A) COS-7 cells were transfected with pHA- hPan3 and either pFlag-CMV5 (lanes 1,3)or pFlag-eRF3 (lanes 2,4). The cell extracts were subjected to an immunoprecipitation assay (IP) using anti-Flag antibody. The immunoprecipi- tates (lanes 3,4) and the inputs (lanes 1,2, 20% of the amount immunoprecipitated) were analyzed by immunoblotting using the indicated antibod- ies. (B) COS-7 cells were transfected with pHA- hPan3 and either pFlag-CMV5 (lanes 1,4), pFlag- PABPC1 (lanes 2,5), or pFlag-PABPC1⌬C (lanes 3,6). The cell extracts were subjected to an im- munoprecipitation assay and immunoblot analy- ses as described in A.(C) COS-7 cells were trans- fected with pHA-hPan2 and either pFlag-CMV5 (lane 1) or pFlag-hPan3 (lanes 2–4), and the cell extracts were subjected to an immunoprecipita- tion assay using anti-Flag antibody. Coprecipi- tated were incubated with buffer alone

(lane 2), recombinant GST-eRF3 (58–204)-(His)6 (lane 3), or GST-eRF3 (80–204)-(His)6 (lane 4). The remaining proteins were separated by SDS- PAGE, and immunoblot analysis was carried out with the indicated antibodies. (D) COS-7 cells were transfected with pHA-hPan3 and either pFlag-CMV5 (lanes 1,6), pFlag-PABPC1 (lanes 2,7), pFlag-PABPC1 F567A (lanes 3,8), pFlag- PABPC1 K580A/L585A (lanes 4,9), or pFlag- PABPC1 F567A/K580A/L585A (lanes 5,10). The cell extracts were subjected to an immunopre- cipitation assay and immunoblot analysis as de- scribed in A.(Top) PABPC1 protein is schemati- cally represented. Gray bar indicates PABC do- main. The partial sequence of PABC is also shown. The amino acid residues that were mu- tated in the experiment are shaded. (E) COS-7 cells were transfected with either pHA-CMV5 (lanes 1,6), pHA-hPan3 (lanes 2,7), pHA-hPan3 Y57A (lanes 3,8), pHA-hPan3 F93A (lanes 4,9), or pHA-hPan3 Y57A/F93A (lanes 5,10). The cell extracts were subjected to an immunoprecipitation assay using anti-HA antibody. The immunoprecipitates (lanes 6–10) and the inputs (lanes 1–5, 10% of the amount immunoprecipitated) were analyzed by immu- noblotting using the indicated antibodies. (Top) hPan3 protein is schematically represented. Gray bars indicate putative PAM2 motifs. The sequences of the two putative PAM2 motifs are also shown. The amino acid residues that were mutated in the experiment are shaded.

GENES & DEVELOPMENT 3139 Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Funakoshi et al.

Flag-hPan3, HA-hPan2, and PABPC1 was prepared from of hPan2–hPan3 in vitro (Uchida et al. 2004). However, COS-7 cells expressing both Flag-hPan3 and HA-hPan2 the role of PABPC1 in the activity of the hCaf1–hCcr4 by means of immunoprecipitation with anti-Flag anti- complex is still controversial; in yeast, Pab1 is inhibitory body. Then the complex was incubated with recombi- to Ccr4 in vitro (Tucker et al. 2002), whereas a pab1⌬ nant eRF3 (58–204) (a fragment encompassing amino ac- strain severely blocks Ccr4-catalyzed deadenylation in ids 58–204) or eRF3 (80–204), which contained the vivo (Caponigro and Parker 1995). To address whether PABPC1-binding region or not, respectively (Uchida et PABPC1 is also involved in the activation of the hCaf1– al. 2002). As shown in Figure 3C, eRF3 (58–204) but not hCcr4 complex, we first examined the physical interac- eRF3 (80–204) effectively inhibited PABPC1 binding to tion between hCaf1 and PABPC1 by an immunoprecipi- Flag-hPan3, while the interaction between HA-hPan2 tation experiment. HeLa cells expressing Flag-PABPC1 and Flag-hPan3 was not affected. Thus, we conclude that and HA-Caf1 were immunoprecipitated using anti-HA hPan3 competes with eRF3 for PABPC1 binding. antibody. Flag-PABPC1 was capable of coprecipitating with HA-hCaf1 (Fig. 4A, lane 6). RNaseA treatment did not abolish the binding, which indicates that no RNA The PAM2 motif of hPan3 and PABC domain was required for the binding (Fig. 4A, lane 8). Moreover, of PABPC1 are responsible for the interaction the binding is indirect because a GST pull-down experi- between hPan3 and PABPC1 ment using purified recombinant PABPC1 and GST- hCaf1 showed no interaction (Fig. 4B, lane 1). We there- We reported previously that a 22-amino-acid region at fore searched for a factor that mediates hCaf1–PABPC1 the N terminus of eRF3 is critical for the binding to the binding. Previous findings independently demonstrated C-terminal domain of PABPC1 (PABC) (Uchida et al. that Tob binds to both PABPC1 (Okochi et al. 2005) and 2002). The PABC domain has been characterized as a hCaf1 (Ikematsu et al. 1999). This led us to speculate protein–protein interaction domain that recruits various that Tob mediates hCaf1–PABPC1 binding. As expected, translation or mRNA-processing factors possessing the purified recombinant PABPC1 bound to GST-hCaf1 in PABPC1-interacting motif (PAM2) to the mRNA poly(A) the presence of Tob (Fig. 4B). To further confirm that Tob tail (Kozlov et al. 2001). The PAM2 motif is also found in acts as a bridge between hCaf1 and PABPC1, we exam- the N-terminal 22-amino-acid region of eRF3 (Uchida et ined the structural requirements for the interaction. Ei- al. 2002). Thus, eRF3 binds to PABC through its PAM2 ther Flag-tagged Tob or its deletion mutants were ex- motif. hPan3 also binds to the C-terminal domain of pressed in COS-7 cells, and immunoprecipitation experi- PABPC1 competitively with eRF3. These results led us ments using anti-Flag antibody were performed. Flag- to speculate that hPan3 competes with eRF3 by binding Tob coprecipitated with both PABPC1 and HA-Caf1 (Fig. to the PABC domain of PABPC1. As expected, we iden- 4C, lane 6). On the other hand, truncation of the Tob C tified two putative PAM2 motifs at the N-terminal re- terminus (127 amino acids) led to a loss of interaction gion of hPan3 (Fig. 3E). We therefore tested the idea by with PABPC1 but not with HA-hCaf1. Conversely, N- site-directed mutagenesis of the PABC domain and im- terminal truncation of Tob (109 amino acids) led to a loss munoprecipitation with extracts from COS-7 cells ex- of interaction with HA-hCaf1 but not with PABPC1. pressing the mutants. As shown in Figure 3D, the bind- Taken together, these results indicate that PABPC1 and ing of hPan3 with PABPC1 was abolished by mutation of hCaf1 bind to the C- and N-terminal region of Tob, re- Phe567 to alanine (F567A) (lane 8), and also by a double spectively, to form a complex. (K580A/L585A) (lane 9) and a triple (F567A/K580A/ The above results demonstrate that a PAM2 motif of L585A) (lane 10) mutation in the PABC domain of hPan3 and the PABC domain of PABPC1 play essential PABPC1. The same results were obtained for eRF3 (Fig. roles for their binding. We therefore asked whether the 3D, top panel). These results indicate that hPan3 as well combination of PAM2 and PABC is also involved in the as eRF3 binds to PABC. Next, we examined whether interaction between Tob and PABPC1 first by searching hPan3 binds to PABC domain through its PAM2 motifs. for PAM2 motifs in the sequence of Tob. As expected, we As shown in Figure 3E, the binding of PABPC1 was not identified two putative PAM2 motifs in the C-terminal affected by mutation of Tyr57 to alanine (Y57A) (lane 8) region of Tob (Fig. 4D). Recently, PAM2 motifs in Tob2, in the first PAM2 motif of hPan3, but was almost com- a distinct subtype of Tob, have been reported (Lim et al. pletely abolished by F93A mutation in the second PAM2 2006). We therefore approached the above question by motif (lane 9) and also by their double mutation (Y57A/ site-directed mutagenesis and immunoprecipitation F93A) (lane 10). Thus, hPan3 binds to PABPC1 with the with extracts from COS-7 cells expressing the mutants. second PAM2 motif. From these results, we conclude As shown in Figure 4D, mutation of Phe139 to alanine that hPan3 and eRF3 competitively bind to the PABC (F139A) in the first PAM2 motif had no effect on the domain of PABPC1 through their PAM2 motifs. binding of PABPC1, whereas mutation of Phe274 to ala- nine (F274A) in the second PAM2 motif completely abol- ished the binding. These results indicate that PABPC1 Caf1–Ccr4 interacts with PABPC1 through Tob binds to the second PAM2 motif of Tob. Next we exam- by using PAM2–PABC contact ined whether Tob binds to the PABC domain of PABPC1. In a previous study, we demonstrated that PABPC1 is As shown in Figure 4E, the binding of Tob was com- required for the activation of the deadenylating activity pletely abolished by mutation of Phe567 to alanine

3140 GENES & DEVELOPMENT Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Mechanism of mRNA deadenylation

Figure 4. hCaf1 binds to PABC domain of PABPC1 via Tob with PAM2 motifs. (A) HeLa cells were transfected with pFlag-PABPC1 and ei- ther pHA-CMV5 (lanes 1,3,5,7) or pHA-hCaf1 (lanes 2,4,6,8). The cell extracts were subjected to an immunoprecipitation assay using anti-HA an- tibody in the presence or absence of RNaseA. The immunoprecipitates (lanes 5–8) and the in- puts (lanes 1–4, 10% of the amount immunopre- cipitated) were analyzed by immunoblotting us- ing the indicated antibodies. (B) Recombinant PABPC1 and GST-hCaf1 prepared from E. coli were subjected to a GST pull-down assay in the presence (lane 2) or absence (lane 1) of recombi- nant Tob (1–285). Immunoblot analyses were performed with the indicated antibodies. (C) COS-7 cells were transfected with pHA-hCaf1 and either pMEFlag (lanes 1,5), pMEFlag-Tob (lanes 2,6), pMEFlag-Tob (1–218) (lanes 3,7), or pMEFlag-Tob (110–345) (lanes 4,8). The cell ex- tracts were subjected to an immunoprecipitation assay using anti-Flag antibody. The immunopre- cipitates (lanes 5–8) and the inputs (lanes 1–4, 20% of the amount immunoprecipitated) were analyzed by immunoblotting using the indi- cated antibodies. (D) COS-7 cells were trans- fected with pHA-hCaf1 and either pMEFlag (lanes 1,6), pMEFlag-Tob (lanes 2,7), pMEFlag- Tob F139A (lanes 3,8), pMEFlag-Tob F274A (lanes 4,9), or pMEFlag-Tob F139A/F274A (lanes 5,10). The cell extracts were subjected to an immu- noprecipitation assay and immunoblot analyses as described in C.(Top) Tob protein is schematically represented. Gray bars indicate putative PAM2 motifs. The sequences of the two putative PAM2 motifs are also shown. The amino acid residues that were mutated in the experiment are shaded. (E) COS-7 cells were transfected with pMEMyc- Tob and either pFlag-PABPC1 (lanes 2,7), pFlag--PABPC1 F567A (lanes 3,8), pFlag-PABPC1 K580A/L585A (lanes 4,9), or pFlag-PABPC1 F567A/K580A/L585A (lane 5,10). (Lanes 1,6) As a control, COS-7 cells were transfected with pMEMyc and pFlag-PABPC1. The cell extracts were subjected to an immunoprecipitation assay using anti-Myc antibody. The immunoprecipitates (lanes 6–10) and the inputs (lanes 1–5, 5% of the amount immunoprecipitated) were analyzed by immunoblotting using the indicated antibodies.

(F567A) (lane 8), and also by a double (K580A/L585A) PABPC1 coprecipitated with Flag-Tob was inversely

(lane 9) and a triple (F567A/K580A/L585A) mutation correlated with the amount of eRF3-(His)6 expressed (lane 10) in PABPC1. Thus, we conclude that Tob binds in cells (Fig. 5B, top). These results suggest the possibil- to the PABC domain of PABPC1 through a PAM2 motif ity that hPan2–hPan3 and hCaf1–hCcr4 each com- and bridges between PABPC1 and Caf1 to form a com- petes with eRF3 for the binding with PABPC1 in living plex. cells. Since hPan2–hPan3 and hCaf1–hCcr4 are involved in eRF3-mediated mRNA deadenylation, which is coupled A translation-dependent exchange of eRF3 to translation termination, we expected that a change of for hPan2–hPan3 occurs on PABPC1 in living cells translation status might affect interactions of eRF3, Based on the structural evidence described above, we hPan3, and Tob with PABPC1. Therefore, we examined next examined the possibility that competition among the effect of a translation inhibitor, cycloheximide eRF3, hCaf1–hCcr4, and hPan2–hPan3 occurs on (CHX), on the interactions. Cells expressing Flag-eRF3 PABPC1 in living cells. First, Flag-hPan3 and GST-eRF3 were treated with CHX and subjected to immunoprecipi- (1–204) were expressed in COS-7 cells, and immuno- tation using anti-Flag antibody. Figure 5C shows that the precipitation experiments were performed with anti- amounts of Flag-eRF3 and eRF1 precipitated were almost Flag antibody. The amount of PABPC1 coprecipi- the same in the presence and absence of CHX, while tated with Flag-hPan3 was decreased by increasing the CHX treatment markedly increased the amount of co- amount of GST-eRF3 expressed in cells (Fig. 5A, top). precipitated PABPC1. In contrast, a comparative experi- Similar results were obtained with Tob; the amount of ment conducted using cells expressing Flag-hPan3 and

GENES & DEVELOPMENT 3141 Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Funakoshi et al.

Figure 5. Both hPan3 and Tob compete with eRF3 to form a complex with PABPC1 in a translation-depen- dent manner. (A) COS-7 cells were transfected with pFlag-hPan3 and increasing amounts of pEBG-eRF3 (1– 204) (0, 3, 6, and 12 µg in lane 2, 3, 4, and 5, respec- tively). The total amounts of plasmid introduced into the cells were maintained at the same level by cotrans- fecting the control vector. The cell extracts were sub- jected to an immunoprecipitation assay using anti-Flag antibody. (Top) Immunoblot analyses were performed with the indicated antibodies. (Bottom) The ratios of bound PABPC1 to precipitated Flag-hPan3 and the ex- pression level of GST-eRF3 (1–204) in the cell lysates were measured. Error bars indicate SEM of three inde- pendent experiments. (B, top) COS-7 cells were trans- fected with pMEFlag-Tob and increasing amounts of pcDNA3-eRF3-(His)6, and an immunoprecipitaion as- say was performed as described in A.(Bottom) The ra- tios of bound PABPC1 to precipitated Flag-Tob and the expression level of eRF3-(His)6 in the cell lysates were measured. Error bars indicate SEM of three independent experiments. (C) COS-7 cells were transfected with pFlag-CMV5 or pFlag-eRF3 and treated with or without cycloheximide (100 µg/mL) for 4 h. The cell extracts were subjected to immunoprecipitation assay using anti-Flag antibody. (Top) Immunoblot analyses were performed with the indicated antibodies. (Bottom) The ratios of bound PABPC1 to precipitated Flag-eRF3 were calculated. Error bars indicate SEM of at least four in- dependent experiments. (D) COS-7 cells were trans- fected with pHA-hPan2 and either pFlag-CMV5 or pFlag-hPan3, and CHX treatment and immunoprecipi- tation assay were performed as described in C.

HA-Pan2 showed that the CHX treatment remarkably extracts were subjected to immunoprecipitation with attenuated the interaction between PABPC1 and hPan3 anti-Flag IgG beads. To deplete the copurified PABPC1 without affecting that between hPan3 and hPan2 (Fig. from the hCaf1–Tob complex, the precipitated beads 5D). Thus, CHX treatment had opposite effects on eRF3– were washed with buffer containing 200 mM NaCl. This PABPC1 and hPan3–PABPC1 interactions. These results washing markedly attenuated the interaction between suggest that a translation-dependent exchange of eRF3 Tob and PABPC1 without affecting the interaction be- and hPan3 occurs on PABPC1. Tob was refractory to this tween hCaf1 and Tob (Fig. 6A). Thus, >85% of PABPC1 analysis, since Tob has a quick turnover through the could be removed from the immunopurified preparation ubiquitin–proteasome pathway and CHX treatment as compared with that prepared without the high-salt greatly affects the amount of Tob. wash. As shown in Figure 6B, Tob was copurified with hCaf1 as a complex (Tob–hCaf1 complex). For compari- son, we also prepared a nuclease-deficient hCaf1 mutant (D161A) as a complex with Tob (Tob–D161A complex). Deadenylating activities of both hCaf1–hCcr4 The RNase activities of the purified proteins were exam- and hPan2–hPan3 are stimulated by PABPC1 ined in the presence or absence of spermidine. Figure 6C We demonstrated previously that the binding of PABPC1 shows that the purified Tob–hCaf1 complex could de- to hPan2–hPan3 leads to the activation of the deadenyl- grade poly(A) RNA in the presence of spermidine (col- ase (Uchida et al. 2004). In order to assess the generality umn 5), whereas the Tob–D161A complex had no RNase of this observation for hCaf1–hCcr4, we tested the effect activity (column 6). As in the case of hPan2–hPan3, none of PABPC1 on the activity of hCaf1. We first prepared of the proteins exhibited any detectable RNase activity the hCaf1–Tob complex. Flag-tagged hCaf1 and HA- in the absence of spermidine (Fig. 6C, columns 1–3). Un- tagged Tob were coexpressed in COS-7 cells, and the cell der this assay condition, the effect of PABPC1 on RNase

3142 GENES & DEVELOPMENT Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Mechanism of mRNA deadenylation

Figure 6. Tob-mediated binding of PABPC1 leads to the activation of hCaf1. (A) COS-7 cells were transfected with pFlag-hCaf1 and pME-HA–Tob. The cell extracts were subjected to immunoprecipi- tation using anti-Flag IgG agarose. After washing with buffer containing the indicated concentrations of NaCl, proteins were eluted with the Flag peptide from the immunoprecipitated beads. The precipi- tated proteins were immunoblotted with the indi- cated antibodies. (B) COS-7 cells were transfected with pFlag-hCaf1 (lanes 2,3) or pFlag-hCaf1 D161A (lanes 4,5) and either pMEFlag (lanes 2,4) or pME- Flag-Tob (lanes 3,5). The cell extracts were subjected to immunoprecipitation as described in A. Buffer containing 200 mM NaCl was used for washing the beads. (C) The purified Flag-tagged hCaf1 and hCaf1 (D161A) (as shown in B), were incubated with inter- nally 32P-radiolabeled poly(A) RNA for 30 min at 30°C in the presence or absence of 1 mM spermi- dine, and 32P radioactivity released was measured. (D) The purified proteins (as shown in B) were incu- bated with the 32P-radiolabeled poly(A) RNA for 30 min at 30°C with or without 10 ng of GST-fused PABPC1 in the absence of spermidine. (E, left) COS-7 cells were transfected with pFlag-hCaf1 and either pMEHA–Tob (lane 1), pMEHA–Tob (F139A) (lane 2), or pMEHA–Tob (F139A/F274A) (lane 3). The cell extracts were subjected to immunoprecipi- tation as described in B.(Right) The purified pro- teins were incubated with the 32P-radiolabeled poly(A) RNA as described in D.

activity was assessed. As shown in Figure 6D, the recom- Significant roles of PABPC1-binding PAM2 motifs binant GST-fused PABPC1 prepared from Escherichia of Tob, hPan3, and eRF3 for mRNA deadenylation coli could markedly stimulate the RNase activity of the in cells Tob–hCaf1 complex (column 6). In sharp contrast, GST- PABPC1 could not stimulate the RNase activity of the The above results strongly suggest that PAM2 motifs of Tob–D161A complex (Fig. 6D, column 10) or hCaf1 Tob and hPan3 play essential roles in mRNA deadenyla- not complexed with Tob (Fig. 6D, column 4). These re- tion by recruiting hCaf1–hCcr4 and hPan2 to poly(A)- sults indicate that PABPC1 stimulates hCaf1 through its bound PABPC1. To test this possibility, we applied a association with Tob, consistent with the above finding dominant-negative approach. T-Rex-HeLa cells were that Tob mediates hCaf1 binding to PABPC1. To further transfected with a ␤-globin reporter plasmid, and the confirm the significance of Tob in the PABPC1-stimu- deadenylation and decay kinetics were examined by lated hCaf1 activity, we also prepared hCaf1 in a com- transcriptional pulse-chase as described in Figure 2. As plex with Tob whose PAM2 motifs were mutated. As shown in Figure 7A, ectopic expression of hPan3 with shown in Figure 6E, F139A mutation of the first PAM2 PAM2 mutations (Y57A/F93A) slowed the rate of dead- motif of Tob had no significant effect on the PABPC1- enylation in the first phase compared with the control stimulated RNase activity (column 4), whereas F139A/ case. The kinetics were similar to those seen with the F274A mutations, which abrogated the Tob–PABPC1 hPan2 D1083A mutant (Fig. 2A). In sharp contrast, binding, almost abolished the effect of PABPC1. Taken ectopic expression of Tob with PAM2 mutations together with previous findings on hPan2–hPan3, these (F139A/F247A) reduced the rate of deadenylation in the results suggest that recruitment of hPan2–hPan3 and second phase without affecting the rate in the first phase hCaf1–hCcr4 to the poly(A)-bound PABPC1 is the gen- (Fig. 7C), with kinetics resembling those seen with the eral mechanism leading to the activation of the mRNA hCaf1 D161A mutant (Fig. 2B). These results provide evi- deadenylases. dence that Tob actually functions with hCaf1–hCcr4 in

GENES & DEVELOPMENT 3143 Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Funakoshi et al.

Figure 7. hPan3, Tob, and eRF3 with mutations in the PABPC1-binding PAM2 motifs affect mRNA deadenylation in a dominant-negative fashion. (A) T-Rex-HeLa cells were transfected with pFlag-CMV5/TO-G1 reporter and either pHA-CMV5 (lanes 1–5) or pHA-hPan3 Y57A/ F93A (lanes 6–10). Transcriptional pulse-chase analysis was performed as described in Figure 2. (B) Total cell lysates were analyzed by immuno- blotting using anti-HA (top) and anti-GAPDH (bottom). (C)AsinA, except that the cells were transfected with pMEHA or pMEHA–Tob F139A/F274A, and transcriptional pulse-chase analysis was performed. (D)AsinB, total cell lysates were immunoblotted. (E)AsinA, except that the cells were transfected with pFlag-CMV5 or pFlag-eRF3 F66A/F75A, and transcriptional pulse-chase analysis was performed. (F)AsinB, except that anti-Flag antibody was used, and to- tal cell lysates were immunoblotted. the second phase of mRNA deadenylation, and further PABPC1 leads to the activation of the deadenylase activ- support the notion that PAM2 motifs play essential roles ity (Fig. 6; Uchida et al. 2004). Together, these new find- in triggering mRNA deadenylation. We also examined ings define PABPC1 as a regulatory platform for mRNA the effect of PAM2 mutations in eRF3 (F66A/F75A), deadenylation, and argue that the recruitment of dead- which abrogated the binding to PABPC1 (Supplementary enylating enzymes to PABPC1 is involved in the mecha- Fig. 3). Ectopic expression of the mutant slowed the rate nism of activation of mRNA deadenylation. The data of deadenylation mainly in the second phase (Fig. 7E). suggest that eRF3 mediates mRNA deadenylation by These results indicate that PAM2 motifs of Tob, hPan3, triggering the recruitment of the deadenylase complexes and eRF3—namely, PABPC1 binding—play key roles in to PABPC1; as translation proceeds, the termination eRF3-mediated mRNA deadenylation. complex is released from PABPC1 and in turn the dead- enylase complexes bind to PABPC1, which leads to the activation of the deadenylating enzymes. Discussion We demonstrated previously that eRF3 mediates dead- General mechanism of mRNA deadenylation enylation and decay of normal mRNA in a manner coupled to translation termination. In this study, we Several previous reports using specific mRNAs showed clarified the underlying mechanism of the eRF3-medi- that recruitment of deadenylating enzymes to the ated mRNA deadenylation. The main findings of this mRNA is involved in the mechanism of mRNA dead- study are (1) eRF3 mediates deadenylation through enylation. First, in Drosophila embryos, an RNA-bind- Pan2–Pan3 and Caf1–Ccr4 deadenylases, which are con- ing protein, Smaug, which physically interacts with the served from yeast to humans (Figs. 1, 2); (2) the termina- Caf1–Ccr4 complex, recruits the complex to Hsp83 tion complex eRF1–eRF3 and deadenylase complexes mRNA to trigger deadenylation and decay of the tran- hPan2–hPan3 and hCaf1–hCcr4 interact with PABPC1, script. The recruitment-induced enzyme activation was in all cases via PAM2–PABC contacts (Figs. 3, 4); (3) confirmed by tethering Smaug to reporter transcripts (Se- hPan2–hPan3 binds PABPC1 competitively with eRF3 motok et al. 2005). Second, a similar tethering strategy and in a manner dependent on translation (Fig. 5); and (4) was used to show that selective recruitment of Caf1– the binding of hPan2–hPan3 and hCaf1–hCcr4 with Ccr4 is sufficient to activate mRNA decay in yeast (Fi-

3144 GENES & DEVELOPMENT Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Mechanism of mRNA deadenylation noux and Seraphin 2006). Third, yeast Mpt5, a member ases for PABPC1 binding, dissociation of eRF3 from of the PUF family of RNA-binding proteins, also recruits PABPC1 is likely to be the key determinant in triggering the Caf1–Ccr4 complex to HO mRNA to trigger dead- deadenylation. As in the case for other G protein family enylation of the message (Goldstrohm et al. 2006). members, eRF3 is thought to function as a molecular Fourth, an RNA-binding protein, CUG-BP, specifically switch that changes binding partners, depending on binds to ARE-containing mRNAs to stimulate poly(A) whether it is in its GTP- or GDP-bound form. Since ri- shortening by recruiting another poly(A)-specific ribo- bosome and eRF1 stimulate GTP hydrolysis of eRF3 nuclease, PARN, to the mRNA (Moraes et al. 2006). For (Frolova et al. 1996), a conformational change of eRF3 the hPan2–hPan3 deadenylase complex, we demon- during or after translation termination might trigger dis- strated previously that hPan3 recruits the hPan2 cata- sociation of eRF3 from PABPC1. Consistent with this lytic subunit to poly(A)-bound PABPC1 to activate its idea, recent findings suggested that binding of PABPC1 poly(A) nuclease activity (Uchida et al. 2004), which is in and eRF1 to eRF3 might be mutually exclusive (Hau- accord with a previous report showing that Pan2–Pan3 is ryliuk et al. 2006). a Pab1-dependent poly(A) nuclease in yeast (Brown et al. Our present study has focused on the role of eRF3 in 1996). In this study, we further demonstrated that normal mRNA decay. However, eRF3 also functions in hCaf1–hCcr4 is also activated by its recruitment to NMD. NMD is a surveillance mechanism that degrades poly(A)-bound PABPC1. mRNA containing premature termination codons Our results indicate that anti-proliferative protein Tob (PTCs). Since PTCs are termination codons, recognition is a key component of hCaf1–hCcr4. We presented three of PTCs is thought to be mediated by translation termi- important observations showing physical and functional nation. Recent findings demonstrated that eRF1–eRF3 interaction between hCaf1–hCcr4 and PABPC1, which is binds the NMD factors SMG1–Upf1 to form a complex supported by structural evidence. First, hCaf1 indirectly termed SURF, and an association between SURF and the interacts with PABPC1 through Tob (Fig. 4A,B). Second, exon junction complex triggers phosphorylation of Upf1 the binding of PABPC1 leads to the stimulation of hCaf1 (Kashima et al. 2006). Although it is not known how deadenylase (Fig. 6). Third, the binding requires the Upf1 phosphorylation leads to rapid decay of PTC-con- PAM2 motif of Tob and the PABC domain of PABPC1 taining mRNA, eRF3 must play a pivotal role in trigger- (Fig. 4D,E). The PAM2–PABC contact is also utilized for ing NMD. the interaction between hPan3 and PABPC1 (Fig. 3D,E). We and others showed previously that a member of the Thus, recruitment of both of the two major mRNA dead- eRF3 family of G proteins, Ski7, interacts with the exo- enylase complexes to poly(A)-bound PABPC1 through some to function in 3Ј-to-5Ј decay of mRNA (van Hoof et adaptor proteins is an important determinant in the ac- al. 2000; Araki et al. 2001), and later, Ski7 was shown to tivation of the deadenylases. These results suggest the be involved in the decay of aberrant mRNAs lacking ter- biological significance of the mechanism of activation of mination codons, which is called “nonstop mRNA de- mRNA deadenylases by PABPC1 in the regulation of cay” (NSD) (Frischmeyer et al. 2002; van Hoof et al. mRNA deadenylation. One possible role of the adaptor 2002). In NSD, Ski7 is suggested to bind to a ribosome proteins (Tob and hPan3) is to recruit mRNA deadenyl- stalled at the 3Ј end of the mRNA and recruit the exo- ase specifically to the substrate mRNA poly(A) tails by some to the mRNA to trigger 3Ј-to-5Ј decay of the taking advantage of the poly(A) specificity of PABPC1. In mRNA. In addition, Hbs1, the closest relative of eRF3, the case of hPAN, the adaptor protein hPan3-mediated has similar roles in another type of aberrant mRNA. PABPC1 interaction actually increases its substrate mRNAs with ribosomes stalled before translation termi- specificity for poly(A) RNA (Uchida et al. 2004). The sec- nation are recognized and degraded by a pathway referred ond possible role is to reduce entropy in the reaction by to as “no-go decay” (NGD) (Doma and Parker 2006). In placing the deadenylase in close proximity to the sub- NGD, Hbs1 is suggested to recognize the stalled ribo- strate RNA. In the case of both deadenylases, >10-fold some to trigger endonucleolytic cleavage of the mRNA. stimulation of the enzyme activity was observed when Thus, eRF3 family members play key roles in triggering complexed with PABPC1 through the adapter. mRNA decay by functioning as a molecular switch con- necting translation to mRNA decay. Future challenges include clarifying the mechanism of The role of translation termination factor eRF3 sequential deadenylation by hPan2–hPan3 and hCaf1– in mRNA deadenylation hCcr4 during poly(A) tail shortening of mRNA. As re- Our previous and present results show that eRF3 medi- ported previously, our results also show that the poly(A) ates deadenylation and decay of normal mRNA in a man- tail is first shortened in a distributive manner by hPan2– ner coupled to translation termination and the eRF3-me- hPan3 to ∼100 nt (first phase, 0–3 h), and the remaining diated deadenylation is catalyzed by both hCaf1–hCcr4 poly(A) tail is then rapidly and processively degraded by and hPan2–hPan3. As translation proceeds, eRF3 is re- hCaf1–hCcr4 (Figs. 2, 7; Yamashita et al. 2005). It is not leased from PABPC1, and hPan2–hPan3 (and possibly clear, however, why eRF3 triggers activation of hPan2– hCaf1–hCcr4) associates with PABPC1, which leads to hPan3 in the first phase, but hCaf1–hCcr4 in the second the activation of the deadenylases. Thus, eRF3 triggers phase. Recent work by Keeling et al. (2006) may provide deadenylation by recruiting the deadenylases to poly(A)- a clue to this question. They found a novel factor (termed bound PABPC1. Since eRF3 competes with the deadenyl- Tpa1) that influences translation termination and

GENES & DEVELOPMENT 3145 Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Funakoshi et al. mRNA deadenylation in S. cerevisiae. Tpa1 interacts Cell culture and DNA transfection with both eRF1–eRF3 and Pab1, and is suggested to be COS-7, HEK-293T, and T-REx-HeLa cells (Invitrogen) were cul- involved in the termination-coupled mRNA deadenyla- tured in Dulbecco’s modified Eagle’s medium (Nissui) supple- tion (Keeling et al. 2006). Interestingly, Tpa1 serves as a mented with 10% fetal calf serum. Cells were transfected with positive regulator of Pan2–Pan3 and a negative regulator plasmid using Lipofectin or LipofectAMINE 2000 (Invitrogen). of Caf1–Ccr4. Thus, Tpa1, through the interaction with eRF3, might positively and negatively regulate the re- Immunoprecipitation cruitment of Pan2–Pan3 and Caf1–Ccr4 to Pab1, respec- The transfected cells were lysed with 10 µg/mL of boiled tively (Keeling et al. 2006). Further studies will be re- RNaseA in buffer A consisting of 20 mM Tris-HCl (pH 8.0), quired to test this possibility. 50 mM NaCl, 1% Nonidet P-40, 1 mM dithiothreitol (DTT), 2.5 mM EDTA-Na (pH 8.0), 100 µM PMSF, 2 µg/mL of aproti- nin, and 2 µg/mL of leupeptin. After centrifugation at 15,000g Note for 20 min, the lysate was incubated for1hat4°C with anti-Flag IgG agarose (Sigma) or Anti-HA Affinity Matrix (Roche Diag- During preparation of our manuscript, there appeared a nostics), and then the resin was washed with buffer A. As paper (Siddiqui et al. 2007) in which the authors report a needed, recombinant proteins were added and the resin was physicochemical analysis of PAM2–PABC contact be- further incubated for 60 min at 4°C. After washing with buffer tween hPan3 and PABPC1 by using NMR. They showed A, proteins retained on the resin were subjected to SDS-PAGE that the interaction is conserved from yeast to humans, and immunoblot analysis. The antibodies used for immunob- and is involved in the first trimming of mRNA poly(A) lotting were anti-Flag M2 (Sigma), anti-HA (3F10, Roche Diag- nostics), anti-Myc (9E10 [Sigma] or A-14 [Santa Cruz Biotech- tails in yeast. nology]), anti-GST (Santa Cruz Biotechnology), anti-His (MBL), anti-Tob (clone 4B1, Sigma), anti-PABP (kind gift from H. Imataka and N. Sonenberg), and anti-GSPT (Hoshino et al. Materials and methods 1998).

Plasmids RNA analysis To construct p9L-MFA2pG with the MFA2pG gene under the For the Tet-off transcriptional repression experiment, yeast control of the tetO promoter, the cassette harboring the tetO cells were grown in selective medium, which contained 2% promoter and CYC1 terminator was excised from pCM190 and galactose to produce the Sup35 N domain, for6hat30°C (yeast inserted into YEplac181 to give p9L. MFA2pG fragment was cells reached mid-log phase [OD600 = 0.5–0.6] in this condition). excised from pGAL-MFA2pG (Araki et al. 2001) and subcloned Cells were collected by centrifugation and resuspended in 10 into p9L. To construct pEBG-eRF3 (1–204) and pCMV-Myc- mL of selective media. After the cells were incubated for 10 min eRF3 N (1–204), the corresponding cDNA fragments of eRF3 at 30°C, doxycycline was added to a final concentration of 5 were inserted in pEBG and pCMV-Myc (Clontech), respectively. µg/mL to repress transcription. At the indicated times, cells In this study, we used the GSPT2 gene for the plasmids express- were harvested and immediately frozen in liquid nitrogen. For ing eRF3. To construct pFlag-PABP⌬C, a PCR fragment encod- preparation of total RNA in the steady state, yeast cells were ing the N-terminal 375 amino acids of PABPC1 was inserted in grown to mid-log phase (0.5–0.6) and collected by centrifuga- pFlag-CMV5 (Eastman Kodak Co.). To construct pMEFlag-Tob tion. RNA extraction from the cell pellet was performed as de- and pFlag-Caf1, the full-length Tob and hCAF1 cDNAs were scribed previously (Hosoda et al. 2003). To determine the amplified by RT–PCR using total RNA from HeLa cells and poly(A) tail length of MFA2pG mRNA, total RNA was separated inserted into pMEFlag (a Flag-tagged vector based on on a 4% polyacrylamide/8 M urea gel and electroblotted to Hy- pME18SFL3) and pFlag-CMV5, respectively. pMEHA-Tob and bond XL (Amersham Pharmacia). For the analysis of mRNA pHA-CAF1, which contain the HA tag instead of Flag, were also decay, total RNA was separated on a 1% agarose gel and blotted constructed. To generate pMEFlag-Tob (1–218) and pMEFlag- to Hybond XL as described previously. The reporters were de- Tob (110–345), the corresponding cDNA fragments of Tob were tected by Northern blotting using an end-labeled oligonucleo- amplified by PCR and inserted into pMEFlag. Point mutants in tide, oRP121 (5Ј-AATTCCCCCCCCCCCCCCCCCCA-3Ј), hCaf1, PABPC1, hPan3, Tob, and eRF3 were generated from which is complementary to the poly(G) sequence at the 3Ј-un- pHA-hCaf1, pFlag-PABPC1, pHA-hPan3, pMEFlag-Tob and translated region of the transcript, as a probe. The poly(A) tail pFlag-eRF3, respectively, using a QuickChange Site-Directed length analysis for total RNA was performed as described pre- Mutagenesis Kit and were confirmed by sequencing. pFlag- viously (Brown et al. 1996). CMV5/TO-G1 was generated from pFlag-CMV5 by inserting For the transcriptional pulse-chase analysis, T-Rex-HeLa cells the two Tet-operators and the ␤-globin gene downstream from were transfected with pFlag-CMV5/TO-G1. After 24 h, cells its CMV promoter. Construction of all other plasmids used in were treated with 10 ng/mL tetracycline for 2.5 h to induce this study has been described previously (Hosoda et al. 2003; transcription, and were harvested at the specified time after Uchida et al. 2004). transcription shutoff. Total RNA was isolated with TriPure Re- agents (Roche) and separated by 2.2% agarose gel electrophore- sis using a NorthernMax Kit (Ambion). ␤-Globin mRNA was Yeast strains and growth conditions detected by Northern blotting using [32P]-labeled ␤-globin ORF as a probe. The yeast strains used were derived from MBS (MATa ade2 his3 leu2 trp1 ura3 can1) and are listed in Supplementary Table 1. In vitro nuclease assay Strains were grown in standard media. Disruption of yeast genes was achieved using standard PCR-based methods and the dis- The production of internally 32P-radiolabeled poly(A) RNA was ruption was confirmed by PCR reactions. described previously (Uchida et al. 2004). In brief, oligo(A)23 was

3146 GENES & DEVELOPMENT Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Mechanism of mRNA deadenylation extended using [␣-32P]ATP and yeast poly(A) polymerase (USB Doma, M.K. and Parker, R. 2006. Endonucleolytic cleavage of Corp.). The labeled poly(A) RNA was purified by gel filtration eukaryotic mRNAs with stalls in translation elongation. Na- with S-400 HR MicroSpin columns (Amersham Bioscience) and ture 440: 561–564. ethanol precipitation. For the immunopurification of recombi- Finoux, A.L. and Seraphin, B. 2006. In vivo targeting of the yeast nant proteins, extracts prepared from COS-7 cells transfected Pop2 deadenylase subunit to reporter transcripts induces with pFlag-hCAF1 and with either pMEHA or pMEHA-Tob their rapid degradation and generates new decay intermedi- were immunoprecipitated with anti-Flag antibody in buffer A. ates. J. Biol. Chem. 281: 25940–25947. After washing three times with buffer A, the precipitated pro- Frischmeyer, P.A., van Hoof, A., O’Donnell, K., Guerrerio, A.L., teins were eluted with buffer B (10 mM HEPES-Na at pH 7.5, 2 Parker, R., and Dietz, H.C. 2002. An mRNA surveillance mM MgCl2, 1 mM DTT, 0.02% Nonidet P-40) containing 100 mechanism that eliminates transcripts lacking termination µg/mL Flag peptides (Sigma). The immunopurified proteins codons. Science 295: 2258–2261. were incubated for 30 min at 30°C in buffer B with 1 U/µL Frolova, L., Le Goff, X., Rasmussen, H.H., Cheperegin, S., RNasin (Promega) and 1 × 105 counts per minute (cpm) of la- Drugeon, G., Kress, M., Arman, I., Haenni, A.L., Celis, J.E., beled poly(A) RNA in the presence or absence of 1 mM spermi- Philippe, M., et al. 1994. A highly conserved eukaryotic pro- dine. If necessary, recombinant GST-fused PABPC1 was added tein family possessing properties of polypeptide chain re- to the reactions. The extent of nucleolysis was measured as lease factor. Nature 372: 701–703. follows: Aliquots were precipitated with 25% trichloroacetic Frolova, L., Le Goff, X., Zhouravleva, G., Davydova, E., Philippe, acid by centrifugation at 20,000g, and the radioactivity of the M., and Kisselev, L. 1996. Eukaryotic polypeptide chain re- soluble fraction was measured with a liquid scintillation lease factor eRF3 is an eRF1- and ribosome-dependent gua- counter. Purification of recombinant GST-PABPC1 was per- nosine triphosphatase. RNA 2: 334–341. formed as described previously (Uchida et al. 2004) Goldstrohm, A.C., Hook, B.A., Seay, D.J., and Wickens, M. 2006. PUF proteins bind Pop2p to regulate messenger RNAs. Nat. Struct. Mol. Biol. 13: 533–539. Acknowledgments Hauryliuk, V., Zavialov, A., Kisselev, L., and Ehrenberg, M. 2006. Class-1 release factor eRF1 promotes GTP binding by We thank Dr. Kazuyoshi Yonezawa and Dr. Tadashi Yamamoto class-2 release factor eRF3. Biochimie 88: 747–757. for plasmids. This work was supported in part by research grants Hoshino, S., Miyazawa, H., Enomoto, T., Hanaoka, F., Kikuchi, from the Japanese Ministry of Education, Culture, Sports, Sci- Y., Kikuchi, A., and Ui, M. 1989. A human homologue of the ence and Technology, and Grant-in-Aid for Research in Nagoya yeast GST1 gene codes for a GTP-binding protein and is City University. expressed in a proliferation-dependent manner in mamma- lian cells. EMBO J. 8: 3807–3814. Hoshino, S., Imai, M., Mizutani, M., Kikuchi, Y., Hanaoka, F., References Ui, M., and Katada, T. 1998. Molecular cloning of a novel member of the eukaryotic polypeptide chain-releasing fac- Araki, Y., Takahashi, S., Kobayashi, T., Kajiho, H., Hoshino, S., tors. Its identification as eRF3 interacting with eRF1. J. Biol. and Katada, T. 2001. Ski7p G protein interacts with the exo- Chem. 273: 22254–22259. some and the Ski complex for 3Ј-to-5Ј mRNA decay in yeast. Hoshino, S., Imai, M., Kobayashi, T., Uchida, N., and Katada, T. EMBO J. 20: 4684–4693. 1999. The eukaryotic polypeptide chain releasing factor Bianchin, C., Mauxion, F., Sentis, S., Seraphin, B., and Corbo, L. (eRF3/GSPT) carrying the translation termination signal to 2005. Conservation of the deadenylase activity of proteins of the 3Ј-Poly(A) tail of mRNA. Direct association of eRF3/ the Caf1 family in human. RNA 11: 487–494. GSPT with polyadenylate-binding protein. J. Biol. Chem. Brown, C.E., Tarun Jr., S.Z., Boeck, R., and Sachs, A.B. 1996. 274: 16677–16680. PAN3 encodes a subunit of the Pab1p-dependent poly(A) Hosoda, N., Kobayashi, T., Uchida, N., Funakoshi, Y., Kikuchi, nuclease in Saccharomyces cerevisiae. Mol. Cell. Biol. 16: Y., Hoshino, S., and Katada, T. 2003. Translation termina- 5744–5753. tion factor eRF3 mediates mRNA decay through the regula- Caponigro, G. and Parker, R. 1995. Multiple functions for the tion of deadenylation. J. Biol. Chem. 278: 38287–38291. poly(A)-binding protein in mRNA decapping and deadenyla- Ikematsu, N., Yoshida, Y., Kawamura-Tsuzuku, J., Ohsugi, M., tion in yeast. Genes & Dev. 9: 2421–2432. Onda, M., Hirai, M., Fujimoto, J., and Yamamoto, T. 1999. Chen, J., Chiang, Y.C., and Denis, C.L. 2002. CCR4, a 3Ј–5Ј Tob2, a novel anti-proliferative Tob/BTG1 family member, poly(A) RNA and ssDNA exonuclease, is the catalytic com- associates with a component of the CCR4 transcriptional ponent of the cytoplasmic deadenylase. EMBO J. 21: 1414– regulatory complex capable of binding cyclin-dependent ki- 1426. nases. Oncogene 18: 7432–7441. Cosson, B., Couturier, A., Chabelskaya, S., Kiktev, D., Inge- Kashima, I., Yamashita, A., Izumi, N., Kataoka, N., Morishita, Vechtomov, S., Philippe, M., and Zhouravleva, G. 2002. R., Hoshino, S., Ohno, M., Dreyfuss, G., and Ohno, S. 2006. Poly(A)-binding protein acts in translation termination via Binding of a novel SMG-1–Upf1–eRF1–eRF3 complex to the eukaryotic release factor 3 interaction and does not influ- exon junction complex triggers Upf1 phosphorylation and ence [PSI(+)] propagation. Mol. Cell. Biol. 22: 3301–3315. nonsense-mediated mRNA decay. Genes & Dev. 20: 355– Daugeron, M.C., Mauxion, F., and Seraphin, B. 2001. The yeast 367. POP2 gene encodes a nuclease involved in mRNA dead- Keeling, K.M., Salas-Marco, J., Osherovich, L.Z., and Bedwell, enylation. Nucleic Acids Res. 29: 2448–2455. D.M. 2006. Tpa1p is part of an mRNP complex that influ- Decker, C.J. and Parker, R. 1993. A turnover pathway for both ences translation termination, mRNA deadenylation, and stable and unstable mRNAs in yeast. Genes & Dev. 7: 1632– mRNA turnover in Saccharomyces cerevisiae. Mol. Cell. 1643. Biol. 26: 5237–5248. Denis, C.L. and Chen, J. 2003. The CCR4–NOT complex plays Kobayashi, T., Funakoshi, Y., Hoshino, S., and Katada, T. 2004. diverse roles in mRNA metabolism. Prog. Nucleic Acid Res. The GTP-binding release factor eRF3 as a key mediator cou- Mol. Biol. 73: 221–250. pling translation termination to mRNA decay. J. Biol. Chem.

GENES & DEVELOPMENT 3147 Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Funakoshi et al.

279: 45693–45700. van Hoof, A., Staples, R.R., Baker, R.E., and Parker, R. 2000. Kozlov, G., Trempe, J.F., Khaleghpour, K., Kahvejian, A., Ekiel, Function of the ski4p and Ski7p proteins in 3Ј-to-5Ј degra- I., and Gehring, K. 2001. Structure and function of the C- dation of mRNA. Mol. Cell. Biol. 20: 8230–8243. terminal PABC domain of human poly(A)-binding protein. van Hoof, A., Frischmeyer, P.A., Dietz, H.C., and Parker, R. Proc. Natl. Acad. Sci. 98: 4409–4413. 2002. Exosome-mediated recognition and degradation of Lim, N.S., Kozlov, G., Chang, T.C., Groover, O., Siddiqui, N., mRNAs lacking a termination codon. Science 295: 2262– Volpon, L., De Crescenzo, G., Shyu, A.B., and Gehring, K. 2264. 2006. Comparative peptide binding studies of the PABC do- Viswanathan, P., Ohn, T., Chiang, Y.C., Chen, J., and Denis, mains from the ubiquitin–protein isopeptide ligase HYD and C.L. 2004. Mouse CAF1 can function as a processive dead- poly(A)-binding protein. J. Biol. Chem. 281: 14376–14382. enylase/3Ј–5Ј-exonuclease in vitro but in yeast the deadenyl- Mangus, D.A., Evans, M.C., Agrin, N.S., Smith, M., Gongidi, P., ase function of CAF1 is not required for mRNA poly(A) re- and Jacobson, A. 2004. Positive and negative regulation of moval. J. Biol. Chem. 279: 23988–23995. poly(A) nuclease. Mol. Cell. Biol. 24: 5521–5533. Wang, W., Czaplinski, K., Rao, Y., and Peltz, S.W. 2001. The role Moraes, K.C., Wilusz, C.J., and Wilusz, J. 2006. CUG-BP binds of Upf proteins in modulating the translation read-through to RNA substrates and recruits PARN deadenylase. RNA 12: of nonsense-containing transcripts. EMBO J. 20: 880–890. 1084–1091. Yamashita, A., Chang, T.C., Yamashita, Y., Zhu, W., Zhong, Z., Muhlrad, D. and Parker, R. 1992. Mutations affecting stability Chen, C.Y., and Shyu, A.B. 2005. Concerted action of poly(A) and deadenylation of the yeast MFA2 transcript. Genes & nucleases and decapping enzyme in mammalian mRNA Dev. 6: 2100–2111. turnover. Nat. Struct. Mol. Biol. 12: 1054–1063. Okochi, K., Suzuki, T., Inoue, J., Matsuda, S., and Yamamoto, T. Zhouravleva, G., Frolova, L., Le Goff, X., Le Guellec, R., Inge- 2005. Interaction of anti-proliferative protein Tob with Vechtomov, S., Kisselev, L., and Philippe, M. 1995. Termi- poly(A)-binding protein and inducible poly(A)-binding pro- nation of translation in eukaryotes is governed by two inter- tein. Genes Cells 10: 151–163. acting polypeptide chain release factors, eRF1 and eRF3. Sachs, A.B. and Deardorff, J.A. 1992. Translation initiation re- EMBO J. 14: 4065–4072. quires the PAB-dependent poly(A) ribonuclease in yeast. Cell 70: 961–973. Semotok, J.L., Cooperstock, R.L., Pinder, B.D., Vari, H.K., Lip- shitz, H.D., and Smibert, C.A. 2005. Smaug recruits the CCR4/POP2/NOT deadenylase complex to trigger maternal transcript localization in the early Drosophila embryo. Curr. Biol. 15: 284–294. Siddiqui, N., Mangus, D.A., Chang, T.C., Palermino, J.M., Shyu, A.B., and Gehring, K. 2007. Poly(A) nuclease interacts with the C-terminal domain of polyadenylate-binding protein do- main from poly(A)-binding protein. J. Biol. Chem. 282: 25067–25075. Stansfield, I., Jones, K.M., Kushnirov, V.V., Dagkesamanskaya, A.R., Poznyakovski, A.I., Paushkin, S.V., Nierras, C.R., Cox, B.S., Ter-Avanesyan, M.D., and Tuite, M.F. 1995. The prod- ucts of the SUP45 and SUP35 genes interact to mediate translation termination in Saccharomyces cerevisiae. EMBO J. 14: 4365–4373. Shyu, A.B., Belasco, J.G., and Greenberg, M.E. 1991. Two dis- tinct destabilizing elements in the c-fos message trigger deadenylation as a first step in rapid mRNA decay. Genes & Dev. 5: 221–231. Thore, S., Mauxion, F., Seraphin, B., and Suck, D. 2003. X-ray structure and activity of the yeast Pop2 protein: A nuclease subunit of the mRNA deadenylase complex. EMBO Rep. 4: 1150–1155. Tucker, M., Valencia-Sanchez, M.A., Staples, R.R., Chen, J., De- nis, C.L., and Parker, R. 2001. The transcription factor asso- ciated Ccr4 and Caf1 proteins are components of the major cytoplasmic mRNA deadenylase in Saccharomyces cerevi- siae. Cell 104: 377–386. Tucker, M., Staples, R.R., Valencia-Sanchez, M.A., Muhlrad, D., and Parker, R. 2002. Ccr4p is the catalytic subunit of a Ccr4p/Pop2p/Notp mRNA deadenylase complex in Saccha- romyces cerevisiae. EMBO J. 21: 1427–1436. Uchida, N., Hoshino, S., Imataka, H., Sonenberg, N., and Katada, T. 2002. A novel role of the mammalian GSPT/eRF3 associating with poly(A)-binding protein in cap/poly(A)-de- pendent translation. J. Biol. Chem. 277: 50286–50292. Uchida, N., Hoshino, S., and Katada, T. 2004. Identification of a human cytoplasmic poly(A) nuclease complex stimulated by poly(A)-binding protein. J. Biol. Chem. 279: 1383–1391.

3148 GENES & DEVELOPMENT Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Mechanism of mRNA deadenylation: evidence for a molecular interplay between translation termination factor eRF3 and mRNA deadenylases

Yuji Funakoshi, Yusuke Doi, Nao Hosoda, et al.

Genes Dev. 2007, 21: Access the most recent version at doi:10.1101/gad.1597707

Supplemental http://genesdev.cshlp.org/content/suppl/2007/11/15/21.23.3135.DC1 Material

References This article cites 46 articles, 30 of which can be accessed free at: http://genesdev.cshlp.org/content/21/23/3135.full.html#ref-list-1

License

Email Alerting Receive free email alerts when new articles cite this article - sign up in the box at the top Service right corner of the article or click here.

Copyright © 2007, Cold Spring Harbor Laboratory Press