key: cord-0779879-ef6n59rb authors: Hong, Hea Jin; Guevara, Matthew G.; Lin, Eric; O’Leary, Seán E. title: Single-Molecule Dynamics of SARS-CoV-2 5’ Cap Recognition by Human eIF4F date: 2021-05-27 journal: bioRxiv DOI: 10.1101/2021.05.26.445185 sha: 93d2f65b185219488cb1643144db6830dbbd4873 doc_id: 779879 cord_uid: ef6n59rb Coronaviruses initiate translation through recognition of the viral RNA 5’ m7GpppAm cap by translation factor eIF4F. eIF4F is a heterotrimeric protein complex with cap-binding, RNA-binding, and RNA helicase activities. Modulating eIF4F function through cellular regulation or small-molecule inhibition impacts coronavirus replication, including for SARS-CoV-2. Translation initiation involves highly coordinated dynamics of translation factors with messenger or viral RNA. However, how the eIF4F subunits coordinate on the initiation timescale to define cap-binding efficiency remains incompletely understood. Here we report that translation supported by the SARS-CoV-2 5’-UTR is highly sensitive to eIF4A inhibition by rocaglamide. Through a single-molecule fluorescence approach that reports on eIF4E–cap interaction, we dissect how eIF4F subunits contribute to cap-recognition efficiency on the SARS-CoV-2 5’ UTR. We find that free eIF4A enhances cap accessibility for eIF4E binding, but eIF4G alone does not change the kinetics of eIF4E–RNA interaction. Conversely, formation of the full eIF4F complex significantly alters eIF4E–cap interaction, suggesting that coordinated eIF4E and eIF4A activities establish the net eIF4F–cap recognition efficiency. Moreover, the eIF4F complex formed with phosphomimetic eIF4E(S209D) binds the viral UTR more efficiently than with wild-type eIF4E. These results highlight a dynamic interplay of eIF4F subunits and mRNA that determines cap-recognition efficiency. A virus of unknown origin was first reported on December 31, 2019 in the city of Wuhan, China. Ensuing worldwide reports of the same virus alarmed the international community due to their rapid spread and associated sudden increase of severe pneumonia cases. 1 The virus was confirmed to belong to the Coronaviridae family, and was initially named 2019-nCoV; it was later renamed as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the associated disease was named COVID-19 by WHO. 1, 2 More than a year after the outbreak was declared a pandemic, more than 162 million confirmed cases have been reported, including more than 3.3 million deaths globally, in addition to significant impacts on the global economy. 2, 3 As obligate intracellular parasites, coronaviruses rely entirely on the host protein synthesis machinery for translation of viral proteins. 4 The SARS-CoV-2 genome is a large, single-stranded positive-sense RNA -at 30 kb, one of the largest known viral RNA genomes. 5 The positive-sense genomic RNA encodes, among others, 16 non-structural viral proteins. 5 The viral RNA must compete with host mRNAs for cellular ribosomes in order to produce these proteins at the correct abundance and stoichiometry to ensure successful viral replication. 6 Host-cell translation is also central to a major breakthrough in combating the SARS-CoV-2/COVID-19 pandemic: the rapid development of efficacious vaccines. 7 Several vaccines with emergency approval for widespread use are mRNA-based. 8, 9 mRNA vaccines rely on host cell ribosomes and the associated translation apparatus to synthesize an antigenic viral protein fragment encoded by the vaccine mRNA in sufficient amounts to allow immune-system activation. 8 Since in eukaryotes mRNA selection for translation is heavily regulated at its initiation step to control the efficiency of protein synthesis, 10 understanding the molecular mechanisms of mRNA interaction with the human translation initiation machinery is important in understanding fundamental aspects of both the viral life cycle and vaccine efficacy. Initiation on most cellular mRNAs involves recognition of an mRNA 5' m 7 G(5')ppp(5')N cap structure 11, 12 where N is the +1 nucleotide of the transcript. The cap is recognized by the heterotrimeric eukaryotic translation initiation factor 4F (eIF4F). 13 eIF4F binds the cap and recruits the small ribosomal subunit to mRNA, forming a 48S pre-initiation complex which is thought to scan toward and locate the start codon for ribosomal subunit joining and translation elongation. [14] [15] [16] eIF4F is composed of a cap-binding subunit, eIF4E, which interacts with the terminal N 7 -methylguanosine base and the triphosphate bridge. [17] [18] [19] [20] eIF4E also binds the large, multidomain eIF4F "scaffold" subunit, eIF4G, which also possesses domains that bind eIF4A, eIF3, the poly(A)-binding protein, and RNA; [21] [22] [23] [24] in humans the eIF4G interaction with 40S subunit-bound eIF3 leads to physical attachment of the pre-initiation complex to the mRNA. 14, 25, 26 The third eIF4F subunit, eIF4A, is a DEAD-box RNA helicase. 13, 27, 28 mRNAs encoding different proteins vary in their dependence on eIF4F for translation efficiency -in particular, greater extent of structure in the 5'-untranslated region is associated with a greater relative eIF4F requirement for efficient translation. 13, 29 Physical accessibility of the 5' cap due to varying structural propensity has been proposed to enhance the recruitment efficiency of translation factors to the mRNA, and thus of translation initiation. 13, 14, 30 mRNA sequence elements have also been proposed to modulate cap-recognition efficiency. [31] [32] [33] However, the molecular logic relating specific mRNA features to cap-recognition efficiency remains incompletely understood. Owing to their centrality in the initiation mechanism, eIF4F subunits are important components of translational control mechanisms. Both cell-signaling pathways and small-molecule inhibitors modulate eIF4E activity to rapidly alter cellular translation patterns. eIF4E is phosphorylated at Ser 209 , in an eIF4G-dependent manner, by MNK (MAP kinase-interacting) kinases MNK1 and MNK2, and downstream of the p38 MAP kinase and/or ERK cell signaling pathway. 34 Phospho-eIF4E has been found associated with polysomes and has been implicated in enhancing translation, 35 though the specific role of eIF4E phosphorylation in promoting translation has been questioned. 36 Nevertheless, small-molecule eIF4F inhibitors have been of significant interest, particularly as anticancer agents. 37 Coronaviruses, including SARS-CoV-1, initiate translation through a cap-and eIF4F-dependent mechanism. 38 Accordingly, disruption of eIF4E-eIF4G interaction by the small-molecule inhibitor 4E2RCat abolished HCoV-229E replication in a cell-based assay, while host protein synthesis was inhibited only by 40%. 39 Likewise, the eIF4A inhibitor silvestrol inhibited expression of MERS-CoV and HCoV-229E nonstructural proteins and formation of viral transcription complexes. 40 eIF4F inhibition was also identified in a comprehensive survey of potential small-molecule therapeutic strategies for SARS-CoV-2. 6 However, the dynamics of eIF4F interaction with a capped coronavirus 5'-untranslated region have not been elucidated. Coronavirus infection activates the p38 pathway MAP kinase, leading to eIF4E phosphorylation at residue Ser 209 . 38 However, while eIF4E phosphorylation is frequently thought to promote translation, 41 global cellular protein synthesis is attenuated following coronavirus infection. 38 Furthermore, a MAP kinase inhibitor failed to reduce viral translation in SARS-CoV-1-infected cells even though it inhibited MAP kinase-dependent eIF4E phosphorylation. 42 Thus, the role of eIF4E phosphorylation specifically in coronavirus translation remains unclear. In this study we developed a single-molecule fluorescence assay for human eIF4E interaction with capped mRNA, to gain insights into how the eIF4F subunits coordinate in cap recognition. We then applied this assay to characterize eIF4F interaction with the SARS-CoV-2 5ʹ untranslated region. DNA templates for in vitro RNA transcription of SARS-CoV-2 RNAs were obtained commercially (Genewiz, Inc.). For preparation of the luciferase fusion construct, DNA fragments containing the class II T7 promoter F2.5, followed by the SARS-CoV-2 5' untranslated region sequence, the coding sequence for firefly luciferase, and the SARS-CoV-2 5' untranslated region sequence were assembled in a pUC57 vector. A HindIII restriction site was placed at the 3' end of the insert, to allow template linearization prior to transcription. The construct for transcription of the isolated 5' UTR was prepared by the same strategy, by inserting class II T7 promoter F2.5 at the 5' end and an EcoRI restriction site at the 3' end to linearize the DNA template. The full-length human GAPDH transcript sequence, obtained from UCSC Genome Browser, was purchased as a gBlock from IDT. The sequence was inserted into pUC119 via SalI and EcoRI restriction sites incorportated during gBlock synthesis. A T7 promoter sequence was also included at the 5ʹ end, along with an extra 5' dG to facilitate efficient in vitro transcription. The EcoRI site placed at the 3' end allowed linearization of the template for the subsequent transcription reaction. Plasmids were isolated at preparative scale from E. coli DH5a transformants selected with ampicillin (100 µg/mL), using a maxi-prep kit (Macherey Nagel). The insert DNA sequences for each construct are given in Supplementary Information. The sequences of all plasmid inserts used in this study were confirmed by Sanger sequencing (Genewiz, Inc.). Purified plasmids were digested at the 3' end of the insert sequence with EcoRI-HF (NEB) to linearize the template with a 5' overhang for in vitro transcription. For 60 µL transcription reactions, about 11 µg of linearized DNA template was incubated for 4 h at 37 °C with 12.5 mM of each NTP, 3% (v/v) DMSO, 25 mM MgCl2, 17 mM DTT, and T7 RNA polymerase (12,500 units/mL) in transcription buffer composed of 0.4 M Tris-HCl (pH 8.1), 10 mM spermidine, and 0.01% (v/v) Triton X-100. The transcription product was extracted using acidic phenol chloroform (pH 4.5) and precipitated overnight in ethanol at -20 °C. RNA was redissolved in water. RNAs were capped using the Vaccinia capping system (NEB) and poly(A)-tailed at their 3' ends with poly(A) polymerase (NEB), following the manufacturer's protocol. RNA concentration was quantified by UV absorbance spectrophotometry using a NanoDrop instrument. 100 nM CoV firefly luciferase fusion RNA was added to 12 µL of HeLa lysate reaction mix from an in vitro protein expression kit (Thermo Scientific, 88882), along with 0.5 µL of rocaglamide/RocA solution stock solutions were prepared in 10% (v/v) and 100% DMSO, respectively; the final DMSO concentration in all reactions was 0.4% and 4 % respectively (v/v). The reaction was incubated at 30 °C for 5 hours and 2.5 µL of the reaction mix was added to 50 µL of luciferase assay reagent (Promega) pre-equilibrated in room temperature. The luminescence was then measured using a Luminometer Turner (Turner Biosystems). Luciferase assays were performed in triplicate and the luminescence of each replicate was determined as the average of three separate measurements. Luminescence was normalized to a DMSO-only control reaction, by dividing the luminescence measured from the reactions containing the inhibitors with the luminescence measured from the sample only containing the DMSO at the appropriate concentration for each inhibitor. The pET-28a(+) vectors containing the sequences of eIF4E or eIF4E(S209D), each fused with an Nterminal Protein G tag, were designed similarly to Feokistova et. al., (2013). 28 The constructs contained a sequence encoding the peptide MA(pAzF) between the Protein G tag and the N-terminus of eIF4E, where pAzF is p-azidophenylalanine (pAzF). pAzF was encoded by the amber stop codon (TAG), for decoding by pAzF-tRNA CUA . The insert sequences are given in the Supplementary Information. The pULTRA expression vector, containing inserts encoding tRNA CUA and pAzF-tRNA CUA -synthetase -a generous gift from Abhishek Chatterjee, Boston College -was co-transformed into an E. coli BL95∆A∆fabR strain with the pET28a(+)-eIF4E plasmid, and transformants were selected on LB agar containing Kanamycin (50 µg/mL) and Spectinomycin (100 µg/mL). A single colony from this selection was used to inoculate a 10 mL 2×YT broth starter culture containing both antibiotics, which was grown overnight at 37 ˚C. The following day, the starter culture was used to inoculate 1 L of 2xYT broth containing the antibiotics. The culture was grown at 37 ˚C to OD600 ~0.6, and then 1 mM pAzF (BAChem, for further purification. Fractions were assessed for purity by SDS-PAGE, and for labelling efficiency by measuring the ratio of 640 nm to 280 nm absorbance. The 1,2,3-triazole formed in the azide-alkyne cycloaddition reaction that conjugates the fluorophore to the protein is expected to absorb at 280 nm. Considering the contribution of this additional absorbance, the typical labelling efficiency was ~50%; this assessment was supported by SDS-PAGE analysis of the labelled protein, which contained two bands, only one of which was conjugated to Cy5 (Fig. S1A) . Purified Cy5-eIF4E and Cy5-eIF4E(S209D) were stored at 4 ˚C in darkness and used for a maximum of one week after each purification. Preparation of human eIF4A. pHis APEX2-eIF4A1 was a gift from Nicholas Ingolia (Addgene plasmid #12964; Ingolia et. al., 2019). 43 The plasmid was transformed into BL21(DE3) CodonPlus cells and transformants were selected on LB-agar plates containing ampicillin (100 µg/mL) and chloramphenicol (25 µg/mL). A single colony from this selection was used to grow a 10 mL overnight culture at 37 ˚C. This starter culture was then used to inoculate 1 L LB containing the selective antibiotics. The cells were grown to OD600 of ~0.6, then protein overexpression was induced with 1 mM IPTG for 3 h at 37 °C. After induction, the cells were harvested by centrifugation at 4,000 rpm for 15 min and were frozen and stored at -80 °C until purification. The frozen cell pellet was resuspended in lysis buffer (20 mM HEPES-KOH, pH 7.5, 500 mM NaCl, 10 mM imidazole, 10 mM β-mercaptoethanol, 0.5% (v/v) NP-40). The cells were lysed by sonication with a microtip attachment (80 % amplitude, setting 3, for 45 seconds at 5 min intervals) and the lysate was immediately clarified by centrifugation (22,000 rpm for 30 min). During centrifugation, a gravity-flow column containing 1 mL Ni-NTA agarose (Thermo Scientific) was washed and preequilibrated with the lysis buffer. The cell-free supernatant was diluted twofold with this lysis buffer, and filtered through a 0.22 µm syringe filter (Corning). The filtrate was then applied to the equilibrated Ni-NTA column, which was washed with a further ~20 mL lysis buffer. The column was further washed with 40 mL high-salt buffer (20 mM HEPES-KOH, pH 7.5, 1 M NaCl, 20 mM imidazole, 10 mM βmercaptoethanol), followed by low-salt buffer (20 mM HEPES-KOH, pH 7.5, 500 mM NaCl, 20 mM imidazole, 10 mM β-mercaptoethanol). The protein was then eluted with elution buffer (50 mM Naphosphate buffer, pH 7.5, 500 mM NaCl, 100 mM Na2SO4, 250 mM imidazole, 2 mM DTT). After elution, the eluate was buffer-exchanged into low-salt buffer (20 mM Tris HCl pH 7.5, 100 mM KCl, 5% glycerol, 2 mM DTT, 0.1 mM EDTA) using a 10 DG column (Bio-Rad), and was TEV-protease (50 units; NEB) cleaved overnight at 4°C to remove the APEX tag. The TEV-protease cleaved sample was loaded onto a 5 mL Q-Sepharose HP column (GE Healthcare Life Sciences), equilibrated in low-salt buffer and maintained at ~4 ˚C. The column was washed with 20 column volumes of low-salt buffer, then eluted with a linear gradient from 0% to 100% high-salt buffer (20 mM Tris HCl pH 7.5, 500 mM KCl, 5% glycerol, 2 mM DTT, 0.1 mM EDTA) over 50 column volumes. eIF4A typically eluted at ~265 mM KCl. Eluate fractions were analyzed by SDS-PAGE, and fractions containing pure eIF4A were then buffer exchanged into a storage buffer (20 mM Tris HCl, pH 7.5, 2 mM DTT, 0.1 mM EDTA, 10% glycerol, 100 mM KCl) using Superdex-75, flash frozen under liquid nitrogen and was stored at -80 °C. A pET-28a(+) vector was constructed with an insert containing a sequence encoding a hexahistidine tag, followed by the sequence encoding residues 557 to 1137 of human eIF4G1, codon-optimized for E. coli.. The plasmid was transformed into BL21 (DE3) CodonPlus cells. Transformants were selected on LB-agar plates containing kanamycin (50 µg/mL) and chloramphenicol (25 µg/mL). A single transformant colony was used to inoculate a 10 mL LB starter culture, which was grown overnight at 37 ˚C. The next day the starter culture was used to inoculate six 1 L LB cultures containing the selective antibiotics. Cells were grown at 37°C to OD600 of ~0.6, then protein overexpression was induced by addition of 1 mM IPTG, and allowed to proceed overnight at 16 °C. The cells were pelleted by centrifugation at 4,000 rpm for 15 minutes and stored at -80°C until purification. Frozen cell pellets were resuspended in lysis buffer (20 mM sodium phosphate, pH 7.5, 300 mM NaCl, 10% (v/v) glycerol, 10mM β-mercaptoethanol, 1× EDTA-free protease inhibitor (Roche), 1× PMSF, 10mM imidazole) and lysed using the microtip sonicator described above (80 % amplitude, setting 3, for 30 seconds in 2 minute intervals). Approximately 3 mL Ni-NTA agarose (Thermo Scientific) was washed and pre-equilibrated with lysis buffer while the lysate was clarified by centrifugation (22,000 rpm for 30 min). The clarified lysate was filtered first through 0.80 µm syringe filter (Corning) then by a 0.22 µm filter. The filtered lysate was added to the equilibrated column and washed with ~15 mL lysis buffer. At room temperature, the column was pre-treated with 25 mL micrococcal nuclease buffer (20 mM sodium phosphate, pH 7.5, 100 mM NaCl, 2 mM CaCl2) prior to treating with 1.5 mL micrococcal nuclease buffer containing 3 U/µL micrococcal nuclease (pre-warmed to 37°C) followed by immediate incubation at 37°C for 30 mins. After this incubation, at room temperature, the column was washed with lysis buffer supplemented with 5 mM EGTA to stop the reaction. The remaining washes (15 mL each) were carried out at 4 °C: wash buffer 1 (20 mM sodium phosphate, pH 7.5, 800 mM NaCl, 10% (v/v) glycerol, 10 mM βmercaptoethanol), wash buffer 2 (20 mM sodium phosphate, pH 7.5, 100 mM NaCl, 10 % Glycerol, βmercaptoethanol), wash buffer 3 (20 mM sodium phosphate, pH 7.5, 100 mM NaCl, 10% (v/v) glycerol, 20 mM imidazole, 10 mM β-mercaptoethanol). Ni-NTA-bound proteins were eluted with 0.75 mL aliquots of elution buffer (20 mM sodium phosphate, pH 7.5, 100 mM NaCl, 10% (v/v) glycerol, 500 mM Imidazole) and DTT was added to each elution fraction to a final concentration of 2 mM. Protein fractions containing the eIF4G fragment, as assessed by SDS-PAGE, were pooled and buffer exchanged to remove excess imidazole (20mM sodium phosphate, pH 7.5, 100mM NaCl, 10% glycerol) using a 10-DG column (Bio-Rad; pre-equilibrated in elution buffer without imidazole and DTT). The protein eluted from the 10-DG column (3.5 mL) was divided into four samples, which were each applied to ~400 µL of magnetic Ni-NTA beads (NEB, product number S1423S), equilibrated at 4 ˚C in binding buffer (50 mM sodium phosphate, 300 mL NaCl, 10 mM imidazole, pH 8.0). The mixture was rocked on ice for one hour, then the supernatant was removed. The beads were washed with three 500 µL aliquots of wash buffer (50 mM sodium phosphate, 300 mM NaCl, 20 mM imidazole, pH 8.0), then bound proteins were eluted by rocking the beads on ice for 10 minutes after addition of 100 µL of elution buffer (50 mM sodium phosphate, 300 mM NaCl, 500 mM imidazole, pH 8.0). This magnetic-bead step allowed the protein to be concentrated under mild conditions; repeated attempts at concentration by centrifugal ultrafiltration resulted in very low levels of protein recovery. The resulting protein was then dialyzed (Thermo Scientific; Product No. 69552) into storage buffer (20 mM sodium phosphate pH 7.5, 100 mM NaCl, 10 % glycerol, 2 mM DTT), flash-frozen in liquid nitrogen, and stored at -80 ˚C until use. 25 nM SARS-CoV-2 5' UTR was added to varying concentration of eIF4G(557-1137) (0 to 300 nM) in The solutions were mixed in a 100 µL quartz cuvette with 1 cm pathlength and immediately monitored using a UV spectrophotometer (Shimadzu) at 340 nm for 15 min, at 0.7-s time intervals. The time course of absorbance change was then fit by linear regression to obtain the slope. This and the background NADH conversion (i.e., in the absence of proteins) were used to compute the observed ATPase rates with the following formulae: is the pathlength, is the background corrected slope, B