Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Mar 13.
Published in final edited form as: J Mol Biol. 2022 Mar 28;434(10):167564. doi: 10.1016/j.jmb.2022.167564

Eukaryotic initiation factor 5A2 regulates expression of antiviral genes

Dorian Farache 1,2, Luochen Liu 1,2, Amy SY Lee 1,2,*
PMCID: PMC11906106  NIHMSID: NIHMS2056840  PMID: 35358571

Summary

Translation factors are essential for regulation of protein synthesis. The eukaryotic translation initiation factor 5A (eIF5A) family is made up of two paralogues – eIF5A1 and eIF5A2 – which display high sequence homology but distinct tissue tropism. While eIF5A1 directly binds to the ribosome and regulates translation initiation, elongation, and termination, the molecular function of eIF5A2 remains poorly understood. Here, we engineer an eIF5A2 knockout allele in the SW480 colon cancer cell line. Using ribosome profiling and RNA-Sequencing, we reveal that eIF5A2 is functionally distinct from eIF5A1 and does not regulate transcript-specific or global protein synthesis. Instead, eIF5A2 knockout leads to decreased intrinsic antiviral gene expression, including members of the IFITM and APOBEC3 family. Furthermore, cells lacking eIF5A2 display increased permissiveness to virus infection. Our results uncover eIF5A2 as a factor involved regulating the antiviral transcriptome, and reveal an example of how gene duplications of translation factors can result in proteins with distinct functions.

Introduction

mRNA translation is regulated through the efforts of over a dozen translation factors [1],[2], which coordinate to regulate the loading, elongation, and termination of the ribosome on transcripts. In agreement to their importance, dysregulation of many translation factors is linked to development disorders and carcinogenesis [3]. Intriguingly, several translation factors and ribosomal proteins exist as gene duplications [4],[5]. However, it remains poorly understood if these duplications allow for gain of additional functions, or instead the paralogues exist as functionally redundant factors.

The eukaryotic translation initiation factor 5A (eIF5A) family contains two paralogue genes, eIF5A1 and eIF5A2, which share 84 % identity at the amino acid level [6],[7] and are found duplicated throughout Eukarya (Fig. 1A) [6]. The eIF5A proteins are notably the only proteins to be post-translationally modified with hypusine [6]. eIF5A1 has multiple functions in translation elongation and termination [8]. eIF5A1 binds directly to the ribosome and increases peptidyl transferase activity, along with stimulating the release of newly synthesized peptides during termination [8]. eIF5A1 is also essential for relieving ribosome pausing at motifs such as poly-proline stretches [8]. In agreement with these functions in translation regulation, eIF5A1 depletion leads to alterations to critical cellular pathways, including autophagy, macrophage activation, neurodevelopment [9],[10],[11]. Additionally, eIF5AL1, an eIF5A1 isoform with 98% sequence conservation, is linked to carcinogenesis [12].

Figure 1. Knockout of eIF5A2 from SW480 cells reduces cellular migration.

Figure 1.

(A) Sequence alignment of eIF5A1 and eIF5A2. The SH3-fold domain is highlighted in blue, and the OB-fold domain is highlighted in purple. (B) Crystal structure of human eIF5A1 (PDB 3CPF), with amino acid divergence between eIF5A1 and eIF5A2 colored in red [13]. (C) Representative western blot of wild type (WT) and eIF5A2KO SW480 cells, showing loss of eIF5A2 expression but no change to eIF5A1 levels. (D) Representative image of transwell cell migration by WT and eIF5A2KO SW480, using 30 % FBS as chemoattractant. (E) Quantification of cell migration. The results are given as the mean ± standard deviation (s.d.) of at least three independent experiments. (F) MTT assay measuring cell proliferation in WT and eIF5A2KO SW480 cells. The P value is calculated by unpaired t-test. ***P<0.001, **P<0.005.

In contrast to eIF5A1, the molecular function of eIF5A2 remains poorly characterized. Structurally, eIF5A1 is composed of two domains, an N-terminal SRC Homology 3 (SH3)-fold domain, and a C-terminal domain composed of an oligonucleotide-Binding (OB) fold [13]. The majority of the residues that are divergent between eIF5A1 and eIF5A2 are clustered within the C-terminal domain, suggesting these mutations could alter nucleic-acid binding specificity (Fig. 1B). Notably, unlike eIF5A1, which is ubiquitously expressed, eIF5A2 expression is limited to the brain and testes. However, the genomic locus of eIF5A2, chromosome 3q26, is commonly amplified in many cancers and found upregulated in many cancers, including colorectal, bladder, gastric, hepatocellular, pancreatic and lung cancers [14]. In addition, eIF5A1 and eIF5A2 also have differing functions in supporting cancer growth. For example, injection of nude mice with p53−/− hepatocytes overexpressing eIF5A2 leads to increased tumor volume, when compared with cells overexpressing eIF5A1 [15]. Furthermore, overexpression of eIF5A2, but not eIF5A1, modulates the epithelial mesenchymal transition (EMT) and increases expression levels of EMT-related proteins such as the metastasis-associated protein 1 (MTA1) [16].

Despite these genetic studies highlighting a distinct role of eIF5A2 during cancer development and cell fate decisions, it remains unclear if eIF5A1 and eIF5A2 differ in molecular function. Here, we generated eIF5A2 knockout cell lines and use ribosome profiling to demonstrate that, unlike eIF5A1, eIF5A2 does not regulate mRNA translation. Using RNA-Sequencing, we reveal that knockout of eIF5A2 induces widespread changes to the transcriptome, including reducing levels of factors involved in cellular migration, development, and the antiviral response. We further demonstrate that eIF5A2 knockout leads to increased cellular permissiveness to infection by RNA viruses.

Results

eIF5A2 is not required for global protein synthesis

To identify the gene program controlled by eIF5A2, we used CRISPR-Cas9 gene editing to construct a knockout allele in the colorectal cancer SW480 cell line, which overexpress eIF5A2 (Fig. 1C). In agreement with the function of eIF5A2 in the epithelial to mesenchymal transition [16],[17], eIF5A2 knockout (SW480 eIF5A2KO) cells expressed lower levels of the mesenchymal marker protein vimentin, and exhibited a 48-fold decrease in cellular migration in a transwell assay, using FBS as the chemoattractant (Fig. 1D, E). Furthermore, knockout of eIF5A2 significantly decreased cellular proliferation (Fig. 1F). Importantly, the expression of eIF5A1 is unaltered in eIF5A2 knockout cells, indicating that the two paralogs are not coregulated in SW480 cells (Fig. 1C).

eIF5A1 was previously shown to be involved in numerous steps of translation, including elongation and termination [8]. Given the high sequence conservation between the two proteins (Fig. 1A), we first examined if eIF5A2 knockout altered global protein synthesis. By [35S]-methionine/cysteine metabolic labeling, we did not observe a significant change in protein synthesis between SW480 and eIF5A2 knockout cell lines (Fig. 2A, B). Furthermore, polysome profile analysis showed similar monosome to polysome ratios between wild type and eIF5A2 knockout cell lines, indicating general translation is unaffected by loss of eIF5A2 (Fig. 2C,D). Altogether, our results reveal that, unlike eIF5A1, eIF5A2 does not regulate general protein synthesis.

Figure 2. eIF5A2 is not required for global translation.

Figure 2.

(A) [35S]-Met/Cys labeling of global protein synthesis in WT and eIF5A2KO SW480 cells. Coomassie staining is showed as a loading control. (B) Quantification of [35S]-Met/Cys metabolic labeling. The intensity of [35S] labeling is normalized to Coomassie staining, and expressed as fold change to WT SW480 cells. (C) Polysome profile of WT and eIF5A2KO SW480 cells. The results in (A), (C) are representative of three independent experiments. (D) Quantification of the monosome to polysome ratio, as calculated by area under the curve. The results in (B), (D) are given as the mean ± s.d. of three independent experiments. The P value is calculated by unpaired t-test. ns, non-significant.

Knockout of eIF5A2 leads to widespread changes to the transcriptome

Given that eIF5A2 is not essential for cell viability, one possibility is that eIF5A2 regulates transcript-specific translation rather than protein synthesis on a global level. To obtain high resolution information of translational changes that occur upon knockout of eIF5A2, we performed ribosome profiling, an approach that uses deep-sequencing of ribosome-protected footprints to identify changes in the association of ribosomes on mRNAs on a genome-wide level [18]. By mapping ribosome-protected footprints to the transcriptome and normalizing these numbers to total mRNA level, as determined by RNA-Sequencing, we obtained translation efficiency measurements for each mRNA. Less than thirty mRNAs were translationally downregulated upon eIF5A2 knockout, and manual inspection of these genes revealed that the footprints were mapped to intronic regions and thus likely attributed to mapping artefacts (Fig. 3A).

Figure 3. Knockout of eIF5A2 alters expression of genes involved in migration, development, and anti-viral response.

Figure 3.

(A) Volcano plot comparing log-fold change in translation efficiency with adjusted P values for mRNAs in WT versus eIF5A2KO SW480 cells.

(B) Volcano plot comparing log-fold change in transcript expression levels with adjusted P values for genes in WT versus eIF5A2KO SW480 cells. (C) Gene ontology analysis of transcripts downregulated in eIF5A2KO SW480 cells. Functional categories below P value = 0.025 are shown. (D) Read mapping to transcripts downregulated in eIF5A2KO SW480. GAPDH is a negative control mRNA that is unaffected by eIF5A2 knockout.

Given that we do not observe functions for eIF5A2 in mRNA translation, we next asked if eIF5A2 regulates the transcriptome. Analysis of the RNA-Sequencing data from wild type and eIF5A2KO SW480 cells revealed widespread transcriptional changes, with 377 genes exhibiting upregulated expression and 477 genes being downregulated more than 2-fold and with a P value cutoff of 0.05 (Fig. 3B). We performed gene ontology analysis of the downregulated genes, and found three major functional categories regulated by eIF5A2 (Fig. 3C). In agreement to the decreased cellular migration (Fig. 1D, E), transcript levels of mRNAs that encode proteins involved in migration and adhesion was blocked in eIF5A2 KO cells, including the intermediate filament protein vimentin. Furthermore, eIF5A2 knockout decreases expression of genes involved in development, such as homeobox family proteins [19],[20]. Finally, eIF5A2 regulates genes involved in defense against viruses, a function that has not previously been reported for eIF5A2. Intriguingly, eIF5A2 appears to co-regulate multiple proteins within a gene family. For example, eIF5A2 regulates the level of the interferon-induced transmembrane (IFITM)-Family transcripts (IFITM1, IFITIM2, IFITM3) (Fig. 3D), which are restriction factors that block the cellular entry of various viruses including influenza A virus [21], flaviviruses [21], rhabdoviruses [22], and filoviruses [23]. In addition, eIF5A2 regulates expression of five genes in the Apolipoprotein B Editing Complex (APOBEC3) family, which are cytidine deaminases that restrict retrovirus replication by leading to genomic instability [24].

eIF5A2 regulates basal expression of antiviral genes

We next used quantitative RT-PCR and western blot to validate candidate transcripts identified by RNA-Sequencing as being dependent on eIF5A2 for efficient expression (Fig. 4A, B, S1A). We validated that vimentin was downregulated by 42-fold, and that the migration-related gene Transforming Growth Factor Beta Induced (TGFBi) was downregulated by 11-fold. Notably, the TGF-β pathway is a key regulator of EMT and migration, and modulation of eIF5A2 levels by knockdown [17],[25] or knockout [26] inhibits both cellular processes (Fig. 1C-E). For the development cluster, we validated that homeobox B6 (HoxB6), homeobox B9 (HoxB9), Wnt family member 6 (Wnt6), and nemo like kinase (NLK) are all downregulated by 2.3 to 5 fold upon eIF5A2 knockout (Fig. 4A, B). Intriguingly, the HoxB family members are important for proper skeletal development, and overexpression of eIF5A2 in transgenic mice impairs ossification during development leading to premature skeletal degeneration [27]. In addition, Wnt6 is a critical regulator of organ development and embryogenesis [28] and NLK is a kinase that is involved in diverse developmental cellular processes [29]. We also confirmed that the IFITM and APOBEC3 family proteins, along with interferon alpha inducible protein 27 (IFI27), are all substantially downregulated upon knockout of eIF5A2 (Fig. 4A, B). Finally, as a control for potential off-target effects of gene-editing, we also examined two additional eIF5A2 knockout clonal cell lines, and found that all three lines exhibit the same downregulation of eIF5A2 target genes (Fig. S1B, C). As eIF5A1 and eIF5A2 are divergent in their role on general protein synthesis, we next asked if they exhibit conserved roles in regulating the transcriptome. Depletion of eIF5A1 in SW480 cells does not alter the levels of the eIF5A2 targets HoxB9 or APOBEC3G (Fig. S2). Altogether, our results indicate that eIF5A2 is functionally distinct from eIF5A1 through its regulation of the transcriptome.

Figure 4. SW480 cells display increased permissivity to viral infection upon knockout of eIF5A2.

Figure 4.

(A) Fold change in transcript expression levels in eIF5A2KO versus WT SW480 cells as measured by quantitative RT-PCR. (B) Representative western blot of WT and eIF5A2KO SW480 cells, showing decreased expression of target genes identified by RNA-Sequencing. Hsp90 is a loading control. (C) Fold change in IFN-β-mediated induction of transcripts in eIF5A2KO and WT SW480 cells, as measured by quantitative RT-PCR. Fold change induction is compared to corresponding vehicle-treated cells. (D) Representative images of eIF5A2KO and WT SW480 cells infected with rVSV-EGFP. (E) Fold change in viral titer output from WT and eIF5A2KO SW480 cells infected with rVSV-EGFP. PFU, Particle forming units. Cells in (D) and (E) were infected at MOI 3 and images or cell supernatant were taken at 4.5 h post infection. (F) Fold change in luciferase activity in WT and eIF5A2KO SW480 cells infected with murine leukemia virus pseudotyped with the Marburg virus glycoprotein and expressing Firefly luciferase as a reporter for infection. The results in (A), (C), (E–G) are given as the mean ± s.d. of three independent experiments. The P value is calculated by unpaired t-test. **P<0.005, *P<0.05.

Antiviral interferon-stimulated genes (ISG) can be transcriptionally controlled in response to cytokine stimulation or at a constitutive basal level. As ISG levels are already altered in unstimulated wild type versus eIF5A2KO SW480 cells, we next asked if eIF5A2 is additionally important for transcription upregulation upon treatment with interferon beta (IFN-β). As expected, stimulation with IFN-β led to massive upregulation of the IFITM and IFI27 transcripts [30],[31]. This upregulation is observed regardless of the expression of eIF5A2, and there are not statistical differences in the magnitude of transcriptional activation (Fig. 4C). Thus, these results indicate that eIF5A2 is important for the basal expression of antiviral genes but not for transcriptional upregulation upon IFN-β stimulation.

Loss of eIF5A2 makes cells more permissive for viral infection

Given our findings that eIF5A2KO SW480 cells exhibit downregulated expression of antiviral genes, we next tested if eIF5A2 controls cellular susceptibility to viral infection and levels of viral replication. Vesicular stomatitis virus (VSV) is a rhabdovirus which has previously been shown to be exquisitely sensitive to interferon [32]. We infected wild type and eIF5A2KO SW480 with a recombinant VSV strain that is engineered to express eGFP as a reporter for viral replication [33],[34]. eIF5A2KO SW480 cells display increased numbers of infected cells, as assessed by eGFP expression, when compared to wild type SW480 cells (Fig. 4D). Furthermore. In addition, infected eIF5A2KO SW480 cells produce ~4-fold higher virus titers compared to wild type cells (Fig. 4E). Thus, knockout of eIF5A2 causes cells to become more permissive for VSV replication and viral output.

As the IFITM family is known to restrict viral entry mediated by filovirus glycoproteins, we next tested if eIF5A2KO SW480 cells are more susceptible to infection mediated by the Marburg virus (MARV) glycoprotein [23]. We rescued recombinant murine leukemia virus which express Firefly luciferase as a reporter for infection, and were pseudotyped with the MARV glycoprotein. Knockout of eIF5A2 lead to a 3.5-fold increase luciferase activity when compared to WT cells, indicating there was an increase in MARV glycoprotein-mediated entry, in agreement with a decrease in IFITM levels (Fig. 4F). Altogether, our results reveal that eIF5A2 expression regulates expression of an antiviral gene signature important for restriction of viral infection.

Discussion

Here, we reveal that eIF5A2 has a primary function in regulation of the transcriptome but not protein synthesis. In agreement with the genetic link between eIF5A2 upregulation and cancer, we find that eIF5A2 knockout modulates the expression of genes important for cellular migration and differentiation (Fig. 3C, 4A, S1B, C). Unexpectedly, we also discover that eIF5A2 expression is inhibitory towards viral replication, through promoting expression of antiviral genes like the IFITM and APOBEC3 family member. Notably, eIF5A2 knockdown leads to loss of constitutive ISG expression while still permitting IFN-induced signaling, and thus eIF5A2 has a molecular role in intrinsic, but not inducible, antiviral gene regulation. Given that eIF5A2 expression in non-cancer cells is limited to the testes and brain, it is of interest to examine if eIF5A2 is important for the antiviral response in these tissues. Indeed, certain viruses infect testes, including Zika Virus and mumps virus [35],[36]; or are neurotropic, including poliovirus, and rhabdoviruses closely related to VSV such as rabies [37].

Our results show that eIF5A2 functionally differs from eIF5A1 in human cells on a molecular and cellular level (Fig. 2, S2B). Intriguingly, eIF5A1 and eIF5A2 are particularly divergent in their cellular function during viral replication, with eIF5A1 supporting replication of viruses that can be inhibited by eIF5A2 expression. For example, while eIF5A2 promotes the expression of APOBEC3 proteins that block HIV-1 replication (Fig. 4A), eIF5A1 binds to the HIV-1 Rev protein and enhances retroviral genome nuclear export and replication [38],[39]. Furthermore, while eIF5A2 expression is required for basal transcription of IFITM proteins that restrict filovirus entry (Fig. 4C, 4F), eIF5A1 is necessary for the synthesis of the ebolavirus VP30 polymerase component protein and subsequently acts in a pro-viral replication capacity [39]. Finally, modulation of eIF5A1 activity through treatment of cells with chemical inhibitors reduces replication of diverse RNA viruses, including rhabdoviruses, flaviviruses, picornaviruses, and bunyaviruses [41].

How can the two paralogs have different roles despite high sequence conservation? The clustering of divergent residues is in the C-terminal OB-fold, suggesting this is the major domain involved in dictating the unique functions of the eIF5A family proteins. Given that OB-fold domains can be involved in direct nucleic acid binding, this domain could explain why eIF5A2 knockout leads to specific changes in the transcriptome. Additionally, eIF5A2 could have differential functions from eIF5A1 due to cellular localization. Indeed, both eIF5A1 and eIF5A2 show dynamic cellular localization that is regulated through post-translational modifications. Hypusination of the eIF5A proteins at position K50 leads to cytoplasmic localization, while acetylation at position K47 promotes nuclear localization [42]. These post-translational modifications are not equally distributed for both family members, as eIF5A1 has a ~6-fold higher affinity for deoxyhypusine synthase and thus is a better substrate for hypusination than eIF5A2 [6]. Finally, knockdown of the nuclear export protein Xpo4 lead to nuclear accumulation of eIF5A2, and subsequently promotion of tumor growth, suggesting eIF5A2 cellular localization is also important for its role in carcinogenesis [15]. It will be of interest to examine the biochemical mechanisms that allow eIF5A2 to function differentially from eIF5A1 in gene expression.

Indeed, the role of eIF5A2 in controlling the transcriptome could be through direct or indirect regulation of transcription, mRNA stability, or mRNA localization. Notably, prior work showed that eIF5A2 can bind directly to the HIF-1ɑ promoter upon hypoxic stress in esophageal squamous cell carcinomas [43]. Furthermore, eIF5A2 shuttles to the nucleus upon cellular perturbations such as hypoxia, and eIF5A2 overexpression leads to increased nuclear localization of the transcription factor STAT3[17],[43]. Finally, single amino acid mutations in the C-terminus of the yeast eIF5A1 homolog block mRNA decay and lead to accumulation of uncapped mRNAs, suggesting the non-conserved residues in the C-terminal domain of eIF5A2 could also be involved in regulating mRNA stability [44],[45]. Overall, our work reveals that eIF5A2 is molecularly distinct from eIF5A1, and demonstrates how two translation factor paralogs can mediate divergent cellular functions.

Methods

Reagent/Resource Source Identifier
Antibodies
Rabbit anti eIF5A Proteintech 17069-1-AP
Mouse anti GAPDH Proteintech 60004-1-Ig
Rabbit anti Vimentin Proteintech 10366-1-AP
Mouse anti E-Cadherin Proteintech 60335-1-1g
Mouse anti IFITM1 Proteintech 60074-1-Ig
Rabbit anti IFITM3 Proteintech 11714-1-AP
Mouse anti Hsp90 BD bioscience BD 610418
Rabbit anti APOBEC3G Cell signaling 43584S
Mouse anti HoxB9 Santa Cruz Biotechnology sc-398500
Goat anti Mouse IgG HRP Fisher G-21040
Goat anti Rabbit HRP Fisher G21234
 
Experimental models organism/strains
SW480 ATCC CCL-228
Hek293T ATCC CRL-11268
BHK-21 ATCC CCL-10
Vero ATCC CCL-81
 
rVSV-EGFP Kind gift from Whelan lab
 
Plasmids
pLBH269 SpCas9 N-Term 5A2 sgRNA Plasmid derivative of pX330 N/A
pBA392 shRNA 5A1
pBA392 shRNA non targeting
psPAX2
pMD2.G
Chemicals, Peptides and recombinant proteins
DMEM Invitrogen 11995073
OptiMEM Fisher 31985070
FBS Biowest S1620
Trypsin Genesee 25-510
100X Penicillin-Streptomycin Genesee 25-512
Polybrene Sigma H9268-5G
Puromycin Dihydrochloride Fisher AAJ61278MC
Polyethylenimine, Linear, MW 25,000 Polysciences, Inc 23966-1
Lipofectamine 2000 Fisher LMRNA001
Emetine Dihydrochloride Hydrate Med Chem Express HY-B1479B
Cycloheximide VWR 97064-724
EXPRE35S35S Protein Labeling Mix Perkin Elmer NEG072002MC
Crystal Violet Fisher AC405830250
Turbo Dnase Fisher AM2238
RNase I Lucigen N6901K
SUPERase-In Rnase Inhibitor Fisher AM2696
TRIzol Reagent Fisher 15596026
Glycogen VWR 97063-256
Mth RNA Ligase NEB M2611A
T4 PNK NEB M0201L
T4 RNA Ligase 2, truncated K227Q NEB M0351L
Yeast 5´ Deadenylase NEB M0331S
Rec J Exonuclease Lucigen RJ411250
Murine RNase inhibitor NEB M0314L
CircLigaseTM ssDNA Ligase Lucigen CL4111K
Dynabeads Biotin Binder Life Technologies 11047
Q5 Polymerase NEB M0491L
Firefly luciferase kit Genecopoeia LF012 LF012
DMSO VWR 97061-250
Thiazolyl blue tetrazolium bromide, 98% Fisher AC158990010
qPCR primers Sequence Origin
eIF5A2 Forward TTTGCCAGCTGAAAGTTCCCA This paper
eIF5A2 Reverse TGCATGGTCGTCCTTTGAGC This paper
IFITM1 Forward CCCCAAAGCCAGAAGATGCACAAGGAG This paper
IFITM1 Reverse CGTCGCCAACCATCTTCCTGTCCCTAG This paper
IFITM2 Forward AGAAAACGGAACTACTGGGGAA This paper
IFITM2 Reverse GAGCATCTCGTAGTTGGGAGG This paper
IFITM3 Forward GTCCACCGTGATCCACATCC This paper
IFITM3 Reverse CATAGGCCTGGAAGATCAGCAC This paper
APOBEC3B Forward AGCGCTTCAGAAAAGAGTGG This paper
APOBEC3B Reverse AAGTTTCGTTCCGATCGTTG This paper
APOBEC3F Forward TCCGTGGAGATCATGGGCTACAAA This paper
APOBEC3F Reverse TGCAGCTTGCTGTCCAGGAATAGA This paper
APOBEC3G Forward GCTGTGCCCAGGAAATGGCTAAAT This paper
APOBEC3G Reverse ACAAAGGTGTCCCAGCAGTGCTTA This paper
IFI27 Forward TCTCCTTCTTTGGGTCTGGC This paper
IFI27 Reverse TGGCCACAACTCCTCCAATC This paper
Vimentin Forward TGTCCAAATCGATGTGGATGTTTC This paper
Vimentin Reverse TTGTACCATTCTTCTGCCTCCTG This paper
HOXB6 Forward GGAGCACTGTCGTCCTTCAG This paper
HOXB6 Reverse ATTGTCCCAACGTGAGAGCC This paper
HOXB9 Forward TACCTCACCAGGGACCGTAG This paper
HOXB9 Reverse GGGAGGACTGGGGGTAATCT This paper
WNT-6 Forward GGTTATGGACCCTACCAGCA This paper
WNT-6 Reverse AACTGGAACTGGCACTCTCG This paper
NLK Forward TTGGCCAGAGTGGAAGAATTAG This paper
NLK Reverse CCCACAGACCAGATGTCAATAG This paper
TGFBi Forward GTCCTGACTGATGAGCTGAAA This paper
TGFBi Reverse TGTTGGTGATGGTGGAGATG This paper
GAPDH Forward GAAGGTGAAGGTCGGAGTCAAC This paper
GAPDH Reverse CAGAGTTAAAAGCAGCCCTGGT This paper
PGK1 Forward TTGACCGAATCACCGACCTC This paper
PGK1 Reverse CATAACGACCCGCTTCCCTT This paper
ROBO1 Forward CCAGAGAGAGCTGGGAAATG This paper
ROBO1 Reverse CCAGTCTGATTCTCCGTGGT This paper
c-myc Forward TGAGGAGACACCGCCCAC This paper
c-myc Reverse CAACATCGATTTCTTCCTCATCTTC This paper
GPX2 Forward GACTTCACCCAGCTCAACGA This paper
GPX2 Reverse ATGCTCGTTCTGCCCATTCA This paper
rRNA Depletion oligos Sequence Origin
LSU_rRNA_1 ggatctttcccgccccccgttcctcccgacccctccacccgccctcccttcccc This paper
LSU_rRNA_2 aaggtggctcggggggccccgtccgtccgtccgtccgtcctcctcctcccccgtctccgc This paper
LSU_rRNA_3 ccgcgaggggggtctcccccgcgggggcgcgccggcgtctcctcgtgggg This paper
LSU_rRNA_4 ggcgacggggggggtgccgcgcgcgggtcggggggcggggcggactgtccccagtgcgcc This paper
LSU_rRNA_5 actctggtggaggtccgtagcggtcctgacgtgcaaatcggtcgtccgacctgggtatag This paper
LSU_rRNA_6 ccgacgcacccccgccacgcagttttatccggtaaagcgaatgattagag This paper
LSU_rRNA_7 cgcgaagcggggctgggcgcgcgccgcggctggacgaggcgccgccgccccccccacgcc This paper
LSU_rRNA_8 cggcgggggcggcgcgcgcgcgcgcgcgtgtggtgtgcgtcggagggcggcggcggcggc This paper
LSU_rRNA_9 ccgcgaggggggcccgggcacccggggggccggcggcggcggcgactctg This paper
LSU_rRNA_10 ccgggcccttcccgtggatcgccccagctgcggcgggcgtcgcggccg This paper
LSU_rRNA_11 cgcgtgccccgccgcgcgccgggaccggggtccggtgcggagtgcccttcgtcctgggaa This paper
LSU_rRNA_12 gagaggcggccgccccctcgcccgtcacgcaccgcacgttcgtggggaacctggcgctaa This paper
rRNA_depl_1 GGGGGGATGCGTGCATTTATCAGATCA [49]
rRNA_depl_2 TTGGTGACTCTAGATAACCTCGGGCCGATCGCACG [49]
rRNA_depl_3 GAGCCGCCTGGATACCGCAGCTAGGAATAATGGAAT [49]
rRNA_depl_4 TCGTGGGGGGCCCAAGTCCTTCTGATCGAGGCCC [49]
rRNA_depl_5 GCACTCGCCGAATCCCGGGGCCGAGGGAGCGA [49]
rRNA_depl_6 GGGGCCGGGCCGCCCCTCCCACGGCGCG [49]
rRNA_depl_7 GGGGCCGGGCCACCCCTCCCACGGCGCG [49]
rRNA_depl_8 CCCAGTGCGCCCCGGGCGTCGTCGCGCCGTCGGGTCCCGGG [49]
rRNA_depl_9 TCCGCCGAGGGCGCACCACCGGCCCGTCTCGCC [49]
rRNA_depl_10 AGGGGCTCTCGCTTCTGGCGCCAAGCGT [49]
rRNA_depl_11 GAGCCTCGGTTGGCCCCGGATAGCCGGGTCCCCGT [49]
rRNA_depl_12 GAGCCTCGGTTGGCCTCGGATAGCCGGTCCCCCGC [49]
rRNA_depl_13 TCGCTGCGATCTATTGAAAGTCAGCCCTCGACACA [49]
rRNA_depl_14 TCCTCCCGGGGCTACGCCTGTCTGAGCGTCGCT [49]
Softwares and informatic resources
Rstudio 1.4.1717
CFX Maestro Biorad
ImageQuant TL GE Healthcare lifescience
IGV Robinson, 2011
Windaq DataQ intruments
DAVID Bioinformatics Resources 6.8 Huang, 20091; Huang, 20092

Contact for Reagent and Resource Sharing

Further information and requests for reagents may be directed to and will be fulfilled by the Lead Contact, Amy S.Y. Lee (amysy_lee@dfci.harvard.edu)

Cell culture.

All cells were cultured in complete media (DMEM, high glucose and L-glutamine (Gibco) with 10 % (v/v) FBS (Biowest) and 500 U mL−1 penicillin/streptomycin). For generation of the eIF5A2KO cell line, SW480 were transfected with pLBH269 SpCas9 N-Term 5A2 sgRNA plasmid, which expresses Cas9 and an eIF5A2-targeting sgRNA, using Lipofectamine 2000 (Thermo Fisher Scientific) following manufacturer’s protocol. After 1 day post transfection, media was changed to complete media supplemented with 1μg mL−1 puromycin to select for transfected cells. After 48 h of selection, cells were trypsinized and seeded in 96-well plates for single cell cloning. Single colony clones were harvested and genotyped by Sanger sequencing to identify eIF5A2 knockout. pLBH269 was a kind gift from L. Harrington and J. Doudna (UC Berkeley). For interferon-β treatment, cells were seeded the day before to be 70 % confluent on the day of treatment. Media was changed to fresh complete media supplemented with 500 U mL−1 interferon-β, and cells were harvested after 24 h of incubation for isolation of total RNA. shRNA-expressing lentiviruses were produced as previously described [46] using lentivirus transfer plasmids encoding the shRNA sequence (non-targeting: 5′-TACAACAGCCACAACGTCTAT-3′, eIF5A1 targeting: 5′-GCTGGACTCCTCCTACACA-3′). To make stable knockdown SW480 cell lines, lentivirus infections were performed in the presence of 10 μg ml−1 hexadimethrine bromide (Sigma), and at 24 h post infection, cells selected with puromycin (Fisher) at 1 μg ml−1 for one week before subsequent experiments.

Quantitative RT-PCR.

Total RNA was isolated by phenol-chloroform extraction and ethanol precipitation. cDNA was reverse transcribed using random hexamers and Moloney Murine Leukemia virus M5 reverse transcriptase (MMLV M5 RT) [47] using standard conditions [46]. Quantitative PCR was performed with Luna Universal qPCR Master Mix (NEB) according to manufacturer’s protocol using a CFX Connect Real-Time PCR Detection System (Biorad).

Western blots.

Cell extracts were lysed in RIPA buffer (50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 % (v/v) NP-40, 0.5 % (v/v) sodium deoxycholate, 0.1 % (w/v) SDS) for 5 minutes on ice. Western blot analysis was performed using the following antibodies: anti-eIF5A1/eIF5A2 (Abclonal A4864), anti-Hsp90 (BD bioscience BD 610418), anti-GAPDH (Proteintech 60004-1-Ig), anti-Vimentin (Proteintech 10366-1-AP), anti-E-cadherin (Proteintech 60335-1-1g), anti-APOBEC3G (cell signaling 43584S), anti-IFITM1 (Proteintech 60074-1-Ig), anti-IFITM3 (Proteintech 11714-1-AP), anti-HoxB9 (Santa Cruz Biotechnology sc-398500).

Migration assay.

SW480 cells were trypsinized and resuspended to seed 5 × 105 cells per well in 500 μL of DMEM supplemented with 1 % (v/v) FBS in the upper chamber of 8 μm pore size Corning Transwell (Fisher) inserts. The inserts were placed in a Corning Companion plate (VWR) and 600 μL of DMEM supplemented with 30 % (v/v) FBS was added to the lower chamber as the chemoattractant factor. After 48 h at 37 °C with 5 % CO2, the cells on the upper surface were removed with a cotton swab, and migrated cells on the lower surface of the insert were fixed with 4 % (v/v) paraformaldehyde in PBS and stained with 0.1 % (w/v) crystal violet. Images of the migrated cells were taken with an EVOS imaging system (Thermo Fisher) and the cells were counted four randomly selected fields.

MTT assay.

Cells were seeded at a density of 10,000 cells per well in a 96-well plate in 100 μL of complete DMEM media and incubated for the indicated timepoints. At each timepoint, media was replaced with 30 μL of complete DMEM supplemented with methyl thiazolyl tetrazolium (MTT) at a concentration of 0.5 mg per well. After 1 h of incubation, 150 μL of DMSO was added to each well and cells were incubated for 10 min with gentle shaking protected from light. The plate was read at a wavelength of 570 nm using a microplate reader (Biotek Synergy H1).

Virus Production and Infection.

Pseudotyped murine leukemia virus (MLV) was rescued as previously described [23]. Plasmids for virus production were a kind gift from M. Farzan (Scripps Research). Briefly, HEK293T cells in a 10-cm plate were transfected with 12 μg of pCMV plasmid DNA encoding MLV gag and pol, 12 μg of pQC plasmid DNA encoding firefly luciferase, and 2.4 μg of plasmid DNA encoding Marburg virus glycoprotein lacking the mucin domain using PEI (1 mg ml−1 linear polyethylenimine (Polysciences)) [47]. Cells were incubated at 37 °C for 8 h and existing media was replaced with fresh complete media. Cells were incubated 32 °C for 72 h, supernatant was harvested, and debris was removed by centrifugation at 3000 × g for 15 min at 4 °C in an Eppendorf 5810R centrifuge. Virus was pelleted by centrifugation in a JA-17 rotor at 28,000 × g for 160 min at 4 °C, and virus was resuspended in NTE buffer (100 mM NaCl, 1 mM EDTA-NaOH pH 8, 10 mM Tris-HCl pH 7.4). For infection of SW480 cells with pseudotyped MLV, cells were seeded the day before in a 96-well plate, and subsequently infected with 1 μL of concentrated virus in 30 μL of complete media supplemented with 10 μg mL−1 polybrene. After 1 h of virus absorption at 37 °C, virus was removed and 100 μL complete media was added. Cells were harvested at 24 h post infection (hpi) and Firefly luciferase activity was measured following manufacturer’s protocol (Genecopoeia) using a Glomax Multi+ plate reader (Promega).

Recombinant VSV virus expressing GFP (rVSV-EGFP) was a kind gift from S. Whelan (Washington University) [33]. Briefly, for propagation of rVSV-EGFP, BHK-21 cells were infected at MOI of 0.01, and supernatant was harvested 16-20 h post infection upon visualization of cytopathic effect. Cellular debris was clarified and virus was pelleted as described above. For infection of SW480 cells with rVSV-EGFP, cells were infected at multiplicity of infection of 3. After 1 h of virus absorption at 37 °C in DMEM, virus was removed and complete media was added. At 4.5 hpi, images were taken on an EVOS imaging system (Thermo Fisher) and supernatant was harvested. Viral titers were determined by plaque assay on Vero cells as previously described [34].

Polysome profiling.

Polysome profiling was performed as previously described with the following alterations [48]. Cells were harvested on ice by scraping into ice cold PBS with 5 μM emetine, and pelleted by centrifugation for 3 min at 300 × g at 4 °C. Cell pellets were lysed in 180 μl ice cold lysis buffer (20 mM HEPES-KOH pH 7.4, 150 mM KOAc, 5 mM MgOAc2, 0.2 mM spermidine, 1 % (v/v) Triton-X 100, 1 mM DTT, 5 μM emetine) for 10 min on ice, passed five times through a 21-g needle, and nuclei were pelleted by spinning at 15,000 × g for 5 min at 4 °C. Lysates were loaded onto a 10-50 % (w/v) sucrose gradient made in lysis buffer lacking detergent or emetine, and centrifuged for 2 h at 4 °C at 38,000 rpm in a SW41 Ti rotor. The gradient was fractioned using a Brandel gradient fractionator and A254 profiles were acquired using a Dataq data acquisition system. Ribosomes were pelleted from fraction by centrifugation for 2 h at 4 °C at 100,000 rpm in a TLA110 rotor. Quantification of the monosome to polysome ratio was performed through analysis of individual peaks using the trapezoidal rule.

Metabolic labeling.

Cells were seeded the day before to be 80 % confluent the day of the experiment. Media was changed to DMEM without methionine or cysteine (Invitrogen) supplemented with 10 μg mL−1 actinomycin D. After 30 min, 10 μL of EXPRE35S35S Protein Labeling Mix (Perkin Elmer) was added to each plate to start the metabolic labeling. After 2 h, cells were harvested by scraping into ice cold PBS and pelleted by centrifugation for 3 min at 300 × g at 4 °C. Cells were lysed in 3× the pellet volume of NP40-NaCl lysis buffer (50 mM HEPES-KOH pH 7.5, 150 mM NaCl, 2 mM EDTA-NaOH pH 8, 0.5 % (v/v) NP40, 0.5 mM DTT) for 5 min on ice. Nuclei were pelleted by centrifugation for 5 min at 15,000 × g at 4 °C, and equal amounts of clarified lysate were separated by 12 % SDS-PAGE. The gel was Coomassie blue stained prior to imaging using a phosphorimager, and lane intensity was quantified using ImageQuant TL software.

Ribosome profiling and RNA-Sequencing.

Ribosome profiling libraries were constructed as previously described [49]. Briefly, cells were harvested in lysis buffer (20 mM Tris-HCl pH 7.4, 150 mM NaCl, 5 mM MgCl2, 1 mM DTT, 100 ug mL−1 cycloheximide, 1 % (v/v) Triton X-100, 25 U mL−1 Turbo DNase1) and ribosome footprints were isolated by RNase 1 digestion and pelleting by centrifugation through a 1 M sucrose cushion at 100,000 rpm for 1 h at 4 °C in TLA-110. 17-34 nucleotide long footprints were size-selected using a 15 % polyacrylamide TBE–urea gel and footprints were dephosphorylated and ligated to a barcoded adenylated RNA linker. Samples were pooled, and cDNA was made by reverse transcription. cDNA products were circularized using CircLigase ssDNA Ligase (Lucigen), contaminating rRNA sequences were removed using biotinylated rRNA depletion oligos [50], and sequencing libraries were constructed by PCR using primers to add Illumina adapters. For RNA-Sequencing libraries, total RNA was isolated using phenol-chloroform extraction and ethanol precipitation. Libraries were prepared and sequenced by Novogene using standard polyA-RNA Illumina sequencing protocol. Experiments were performed in duplicate.

Bioinformatics analysis.

For RNA-Sequencing pre-processing, adapters were trimmed using Cutadapt [51], and contaminating rRNA reads were removed using bowtie2 [52],[53]. For ribosome profiling pre-processing, adapters were trimmed and reads were demultiplexed using Cutadapt. Reads were collapsed with FASTX-Toolkit [54] and UMI were removed with Cutadapt. Contaminating rRNA reads were removed using bowtie2. Reads were mapped to the human transcriptome (GRCh38) using STAR [55] and differential gene analysis was performed using the DESeq2 package [56] and Babel package [57] in R Studio. Gene ontology analysis of RNA-Sequencing data was performed using DAVID [58],[59]. Mapped reads were sorted and indexed using samtools [60], bigwig files were created using DeepTools [61], and reads were visualized using the Integrated Genomics Viewer (Broad Institute) [62].

Supplementary Material

SI Table 1

Table 1. Table of changes in translation efficiency upon knockout of eIF5A2 in SW480 cells. The baseMean indicates the average of the normalized count values, dividing by size factors, taken over all samples. The log2FoldChange is the Fold change between the comparison and control group, reported on a logarithmic base 2 scale. The lfcSE is the standard error estimate for the fold change estimate. The change type column indicates if both the ribosome footprint and RNA-sequencing levels are changed (“both”), or only the ribosome footprint levels are altered (“translational_only”).

SI Table 2

Table 2. Table of changes in transcript levels upon knockout of eIF5A2 in SW480 cells.

3

Figure S1. Analysis of gene expression changes in eIF5A2 knockout clonal cell lines. (A) Fold change in transcript expression levels of eIF5A2KO versus WT SW480 cells as measured by quantitative RT-PCR of upregulated genes (GPX2 and ROBO1) or non-target genes (PGK1 and c-Myc) as identified by RNA-Sequencing. (B) Representative western blot of WT and two different clonal eIF5A2KO SW480 cell lines, showing decreased expression of target genes identified by RNA-Sequencing. Hsp90 is a loading control. (C) Fold change in transcript expression levels in two different clonal eIF5A2KO cell lines versus WT SW480 cells as measured by quantitative RT-PCR.

Figure S2. Analysis of cell proliferation and gene expression changes in SW480 cells upon eIF5A1 knockdown. (A) MTT assay measuring cell proliferation in SW480 cells transduced with a non-targeting or eIF5A1-targeting shRNA. The results are given as the mean ± standard deviation (s.d.) of at least three independent experiments. The P value is calculated by unpaired t-test. **P<0.005. (B) Representative western blot of SW480 cells transduced with a non-targeting or eIF5A1-targeting shRNA, showing that decreased expression of eIF5A1 does not affect eIF5A2 target genes. Hsp90 is a loading control.

Acknowledgments:

The authors thank K. Chat, J. Chen, P.J. Kranzusch, J.K. Nuñez, and members of the Lee lab for discussions.

Funding:

This work was funded by the Charles H. Hood Foundation, the Searle Scholars Program, the Pew Biomedical Scholars Program, and a Sloan Research Fellowship (A.S.Y.L).

Footnotes

Competing interests: The authors declare no competing interests.

Data and materials availability: All sequencing data have been deposited in the NCBI Gene Expression Omnibus under GEO Series accession number GSE196982.

References

  • [1].Merrick WC, & Pavitt GD Protein Synthesis Initiation in Eukaryotic Cells. Cold Spring Harbor perspectives in biology, 10 (12) (2018), Article a033092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Dever TE, Dinman JD, & Green R Translation Elongation and Recoding in Eukaryotes. Cold Spring Harbor perspectives in biology, 10 (8) (2018), Article a032649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Chu J, Cargnello M, Topisirovic I, & Pelletier J Translation Initiation Factors: Reprogramming Protein Synthesis in Cancer. Trends in cell biology, 26 (12) (2016), pp. 918–933. [DOI] [PubMed] [Google Scholar]
  • [4].Ho J, & Lee S A Cap for Every Occasion: Alternative eIF4F Complexes. Trends in biochemical sciences, 41 (10) (2016), pp. 821–823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Mills A, & Gago F On the Need to Tell Apart Fraternal Twins eEF1A1 and eEF1A2, and Their Respective Outfits. International journal of molecular sciences, 22 (13) (2021), p. 6973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Clement PM, Henderson CA, Jenkins ZA, Smit-McBride Z, Wolff EC, Hershey JW, Park MH, & Johansson H E Identification and characterization of eukaryotic initiation factor 5A-2. European journal of biochemistry, 270 (21) (2003), pp. 4254–4263. [DOI] [PubMed] [Google Scholar]
  • [7].Jenkins ZA, Hååg PG, & Johansson HE Human eIF5A2 on chromosome 3q25-q27 is a phylogenetically conserved vertebrate variant of eukaryotic translation initiation factor 5A with tissue-specific expression. Genomics, 71 (1) (2001), pp. 101–109. [DOI] [PubMed] [Google Scholar]
  • [8].Schuller AP, Wu CC, Dever TE, Buskirk AR, & Green R eIF5A Functions Globally in Translation Elongation and Termination. Molecular cell, 66 (2) (2017), pp. 194–205 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Lubas M, Harder LM, Kumsta C, Tiessen I, Hansen M, Andersen JS, Lund AH, & Frankel LB eIF5A is required for autophagy by mediating ATG3 translation. EMBO reports, 19 (6) (2018), Article e46072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Puleston DJ, Buck MD, Klein Geltink RI, Kyle RL, Caputa G, O’Sullivan D, Cameron AM, Castoldi A, Musa Y, Kabat, et al. Polyamines and eIF5A Hypusination Modulate Mitochondrial Respiration and Macrophage Activation. Cell metabolism, 30 (2) (2019), pp. 352–363 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Kar RK, Hanner AS, Starost MF, Springer D, Mastracci TL, Mirmira RG, & Park MH Neuron-specific ablation of eIF5A or deoxyhypusine synthase leads to impairments in growth, viability, neurodevelopment and cognitive functions in mice. The Journal of biological chemistry, 297 (5) (2021), Article 101333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Zhou B, & Guo R (2018). Integrative analysis of significant RNA-binding proteins in colorectal cancer metastasis. Journal of cellular biochemistry, 119 (12) (2018), pp9730–9741 [DOI] [PubMed] [Google Scholar]
  • [13].Tong Y, Park I, Hong BS, Nedyalkova L, Tempel W, & Park HW Crystal structure of human eIF5A1: insight into functional similarity of human eIF5A1 and eIF5A2. Proteins, 75 (4) (2009), pp. 1040–1045 [DOI] [PubMed] [Google Scholar]
  • [14].Wu GQ, Xu YM, & Lau A Recent insights into eukaryotic translation initiation factors 5A1 and 5A2 and their roles in human health and disease. Cancer cell international, 20 (2020), p. 142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Zender L, Xue W, Zuber J, Semighini CP, Krasnitz A, Ma B, Zender P, Kubicka S, Luk JM, Schirmacher P, et al. An oncogenomics-based in vivo RNAi screen identifies tumor suppressors in liver cancer. Cell, 135 (5) (2008), pp. 852–864 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Zhu W, Cai MY, Tong ZT, Dong SS, Mai SJ, Liao YJ, Bian XW, Lin MC, Kung HF, Zeng YX, et al. Overexpression of EIF5A2 promotes colorectal carcinoma cell aggressiveness by upregulating MTA1 through C-myc to induce epithelial-mesenchymaltransition. Gut, 61 (4) (2012), pp. 562–575 [DOI] [PubMed] [Google Scholar]
  • [17].Wei JH, Cao JZ, Zhang D, Liao B, Zhong WM, Lu J, Zhao HW, Zhang JX, Tong ZT, Fan S, et al. EIF5A2 predicts outcome in localised invasive bladder cancer and promotes bladder cancer cell aggressiveness in vitro and in vivo. British journal of cancer, 110 (7) (2014), pp. 1767–1777 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Ingolia NT, Ghaemmaghami S, Newman JR, & Weissman JS Genome-wide analysis in vivo of translation with nucleotide resolution using ribosome profiling. Science (New York, N.Y.), 324 (5924) (2009), pp. 218–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Jurberg AD, Aires R, Varela-Lasheras I, Nóvoa A, & Mallo M Switching axial progenitors from producing trunk to tail tissues in vertebrate embryos. Developmental cell, 25 (5) (2013), pp. 451–462 [DOI] [PubMed] [Google Scholar]
  • [20].Casaca A, Nóvoa A, & Mallo M Hoxb6 can interfere with somitogenesis in the posterior embryo through a mechanism independent of its rib-promoting activity. Development (Cambridge, England), 143 (3) (2016), pp. 437–448. [DOI] [PubMed] [Google Scholar]
  • [21].Brass AL, Huang IC, Benita Y, John SP, Krishnan MN, Feeley EM, Ryan BJ, Weyer JL, van der Weyden L, Fikrig E, Adams DJ, Xavier RJ, Farzan M, & Elledge SJ The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus. Cell, 139 (7) (2009), pp. 1243–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Weidner JM, Jiang D, Pan XB, Chang J, Block TM, & Guo JT Interferon-induced cell membrane proteins, IFITM3 and tetherin, inhibit vesicular stomatitis virus infection via distinct mechanisms. Journal of virology, 84 (24) (2010), pp. 12646–12657 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Huang IC, Bailey CC, Weyer JL, Radoshitzky SR, Becker MM, Chiang JJ, Brass AL, Ahmed AA, Chi X, Dong L, et al. Distinct patterns of IFITM-mediated restriction of filoviruses, SARS coronavirus, and influenza A virus. PLoS pathogens, 7 (1) (2011), Article e1001258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Chiu YL, & Greene WC The APOBEC3 cytidine deaminases: an innate defensive network opposing exogenous retroviruses and endogenous retroelements. Annual review of immunology, 26 (2008), pp. 317–353. [DOI] [PubMed] [Google Scholar]
  • [25].Bao Y, Lu Y, Wang X, Feng W, Sun X, Guo H, Tang C, Zhang X, Shi Q, & Yu H Eukaryotic translation initiation factor 5A2 (eIF5A2) regulates chemoresistance in colorectal cancer through epithelial mesenchymal transition. Cancer cell international, 15 (2015), p. 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Zhao G, Zhang W, Dong P, Watari H, Guo Y, Pfeffer LM, Tigyi G, & Yue J EIF5A2 controls ovarian tumor growth and metastasis by promoting epithelial to mesenchymal transition via the TGFβ pathway. Cell & bioscience, 11 (1) (2021), p. 70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Chen M, Huang JD, Deng HK, Dong S, Deng W, Tsang SL, Huen MS, Chen L, Zan T, Zhu GX, et al. Overexpression of eIF-5A2 in mice causes accelerated organismal aging by increasing chromosome instability. BMC cancer, 11(2011), p. 199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Wei M, Zhang C, Tian Y, Du X, Wang Q, & Zhao H Expression and Function of WNT6: From Development to Disease. Frontiers in cell and developmental biology, 8 (2020), Article 558155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Ishitani T, & Ishitani S Nemo-like kinase, a multifaceted cell signaling regulator. Cellular signalling, 25 (1) (2013), pp. 190–197 [DOI] [PubMed] [Google Scholar]
  • [30].Lewin AR, Reid LE, McMahon M, Stark GR, & Kerr IM Molecular analysis of a human interferon-inducible gene family. European journal of biochemistry, 199 (2) (1991), pp. 417–423 [DOI] [PubMed] [Google Scholar]
  • [31].Sellebjerg F, Krakauer M, Hesse D, Ryder LP, Alsing I, Jensen PE, Koch-Henriksen N, Svejgaard A, & Soelberg Sørensen P Identification of new sensitive biomarkers for the in vivo response to interferon-beta treatment in multiple sclerosis using DNA-array evaluation. European journal of neurology, 16 (12) (2009), pp. 1291–1298 [DOI] [PubMed] [Google Scholar]
  • [32].Faul EJ, Lyles DS, & Schnell MJ Interferon response and viral evasion by members of the family rhabdoviridae. Viruses, 1 (3) (2009), pp. 832–851 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Whelan SP, Ball LA, Barr JN, & Wertz GT Efficient recovery of infectious vesicular stomatitis virus entirely from cDNA clones. Proceedings of the National Academy of Sciences of the United States of America, 92 (18) (1995), pp. 8388–8392 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Lee AS, Burdeinick-Kerr R, & Whelan SP A genome-wide small interfering RNA screen identifies host factors required for vesicular stomatitis virus infection. Journal of virology, 88 (15) (2014), pp. 8355–8360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Matusali G, Houzet L, Satie AP, Mahé D, Aubry F, Couderc T, Frouard J, Bourgeau S, Bensalah K, Lavoué S, et al. Zika virus infects human testicular tissue and germ cells. The Journal of clinical investigation, 128 (10) (2018), pp. 4697–4710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Le Tortorec A, Denis H, Satie AP, Patard JJ, Ruffault A, Jégou B, & Dejucq-Rainsford N Antiviral responses of human Leydig cells to mumps virus infection or poly I:C stimulation. Human reproduction (Oxford, England), 23 (9) (2008), pp. 2095–2103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Milora KA, & Rall GF Interferon Control of Neurotropic Viral Infections. Trends in immunology, 40 (9) (2019), pp. 842–856 [DOI] [PubMed] [Google Scholar]
  • [38].Malim MH, Tiley LS, McCarn DF, Rusche JR, Hauber J, & Cullen BR HIV-1 structural gene expression requires binding of the Rev trans-activator to its RNA target sequence. Cell, 60 (4) (1990), pp. 675–683 [DOI] [PubMed] [Google Scholar]
  • [39].Bevec D, Jaksche H, Oft M, Wöhl T, Himmelspach M, Pacher A, Schebesta M, Koettnitz K, Dobrovnik M, Csonga R, et al. Inhibition of HIV-1 replication in lymphocytes by mutants of the Rev cofactor eIF-5A. Science (New York, N.Y.), 271 (5257) (1996), pp. 1858–1860. [DOI] [PubMed] [Google Scholar]
  • [40].Olsen ME, Filone CM, Rozelle D, Mire CE, Agans KN, Hensley L, & Connor JH Polyamines and Hypusination Are Required for Ebolavirus Gene Expression and Replication. mBio, 7 (4) (2016), Article e00882–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Mounce BC, Cesaro T, Moratorio G, Hooikaas PJ, Yakovleva A, Werneke SW, Smith EC, Poirier EZ, Simon-Loriere E, Prot M, et al. Inhibition of Polyamine Biosynthesis Is a Broad-Spectrum Strategy against RNA Viruses. Journal of virology, 90 (21) (2016), pp. 9683–9692 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Ishfaq M, Maeta K, Maeda S, Natsume T, Ito A, & Yoshida M Acetylation regulates subcellular localization of eukaryotic translation initiation factor 5A (eIF5A). FEBS letters, 586 (19) (2012), pp. 3236–3241 [DOI] [PubMed] [Google Scholar]
  • [43].Li Y, Fu L, Li JB, Qin Y, Zeng TT, Zhou J, Zeng ZL, Chen J, Cao TT, Ban X, et al. Increased expression of EIF5A2, via hypoxia or gene amplification, contributes to metastasis and angiogenesis of esophageal squamous cell carcinoma. Gastroenterology, 146 (7) (2014), pp. 1701–13 [DOI] [PubMed] [Google Scholar]
  • [44].Zuk D, & Jacobson A A single amino acid substitution in yeast eIF-5A results in mRNA stabilization. The EMBO journal, 17 (10) (1998), pp 2914–2925 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Hoque M, Park JY, Chang YJ, Luchessi AD, Cambiaghi TD, Shamanna R, Hanauske-Abel HM, Holland B, Pe’ery T, Tian B, & Mathews MB Regulation of gene expression by translation factor eIF5A: Hypusine-modified eIF5A enhances nonsense-mediated mRNA decay in human cells. Translation (Austin, Tex.), 5(2) (2017), Article e1366294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Lamper AM, Fleming RH, Ladd KM, & Lee A A phosphorylation-regulated eIF3d translation switch mediates cellular adaptation to metabolic stress. Science (New York, N.Y.), 370 (6518) (2020), pp. 853–856. [DOI] [PubMed] [Google Scholar]
  • [47].Arezi B, & Hogrefe H Novel mutations in Moloney Murine Leukemia Virus reverse transcriptase increase thermostability through tighter binding to template-primer. Nucleic acids research, 37 (2) (2009), pp. 473–481 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Lee AS, Kranzusch PJ, Doudna JA, & Cate JH eIF3d is an mRNA cap-binding protein that is required for specialized translation initiation. Nature, 536 (7614) (2016), pp. 96–99 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].McGlincy NJ, & Ingolia NT Transcriptome-wide measurement of translation by ribosome profiling. Methods (San Diego, Calif.), 126 (2017), pp. 112–129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Ingolia NT, Brar GA, Rouskin S, McGeachy AM, & Weissman JS The ribosome profiling strategy for monitoring translation in vivo by deep sequencing of ribosome-protected mRNA fragments. Nature protocols, 7 (8) (2012), pp. 1534–1550 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Kechin A, Boyarskikh U, Kel A, & Filipenko M cutPrimers: A New Tool for Accurate Cutting of Primers from Reads of Targeted Next Generation Sequencing. Journal of computational biology : a journal of computational molecular cell biology, 24 (11) (2017), pp. 1138–1143 [DOI] [PubMed] [Google Scholar]
  • [52].Langmead B, Wilks C, Antonescu V, & Charles R Scaling read aligners to hundreds of threads on general-purpose processors. Bioinformatics (Oxford, England), 35 (3) (2019), pp. 421–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Langmead B, & Salzberg SL Fast gapped-read alignment with Bowtie 2. Nature methods, 9 (4) (2012), pp. 357–359 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Hannon GJ (2010) FASTX-Toolkit. http://hannonlab.cshl.edu/fastx_toolkit. [Google Scholar]
  • [55].Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, & Gingeras TR STAR: ultrafast universal RNA-seq aligner. Bioinformatics (Oxford, England), 29 (1) (2013), pp. 15–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Love MI, Huber W, & Anders S Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome biology, 15 (12) (2014), p. 550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Olshen AB, Hsieh AC, Stumpf CR, Olshen RA, Ruggero D, & Taylor BS Assessing gene-level translational control from ribosome profiling. Bioinformatics (Oxford, England), 29(23) (2013), pp 2995–3002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Huang D, Sherman BT, & Lempicki RA Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature protocols, 4 (1) (2009), pp. 44–57 [DOI] [PubMed] [Google Scholar]
  • [59].Huang D, Sherman BT, & Lempicki RA Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic acids research, 37 (1) (2009), pp. 1–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R, & 1000 Genome Project Data Processing Subgroup The Sequence Alignment/Map format and SAMtools. Bioinformatics (Oxford, England), 25 (16) (2009), pp. 2078–2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Ramírez F, Ryan DP, Grüning B, Bhardwaj V, Kilpert F, Richter AS, Heyne S, Dündar F, & Manke T deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic acids research, 44(W1) (2016), W160–W165 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, & Mesirov JP Integrative genomics viewer. Nature biotechnology, 29 (1) (2011), pp. 24–26 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

SI Table 1

Table 1. Table of changes in translation efficiency upon knockout of eIF5A2 in SW480 cells. The baseMean indicates the average of the normalized count values, dividing by size factors, taken over all samples. The log2FoldChange is the Fold change between the comparison and control group, reported on a logarithmic base 2 scale. The lfcSE is the standard error estimate for the fold change estimate. The change type column indicates if both the ribosome footprint and RNA-sequencing levels are changed (“both”), or only the ribosome footprint levels are altered (“translational_only”).

SI Table 2

Table 2. Table of changes in transcript levels upon knockout of eIF5A2 in SW480 cells.

3

Figure S1. Analysis of gene expression changes in eIF5A2 knockout clonal cell lines. (A) Fold change in transcript expression levels of eIF5A2KO versus WT SW480 cells as measured by quantitative RT-PCR of upregulated genes (GPX2 and ROBO1) or non-target genes (PGK1 and c-Myc) as identified by RNA-Sequencing. (B) Representative western blot of WT and two different clonal eIF5A2KO SW480 cell lines, showing decreased expression of target genes identified by RNA-Sequencing. Hsp90 is a loading control. (C) Fold change in transcript expression levels in two different clonal eIF5A2KO cell lines versus WT SW480 cells as measured by quantitative RT-PCR.

Figure S2. Analysis of cell proliferation and gene expression changes in SW480 cells upon eIF5A1 knockdown. (A) MTT assay measuring cell proliferation in SW480 cells transduced with a non-targeting or eIF5A1-targeting shRNA. The results are given as the mean ± standard deviation (s.d.) of at least three independent experiments. The P value is calculated by unpaired t-test. **P<0.005. (B) Representative western blot of SW480 cells transduced with a non-targeting or eIF5A1-targeting shRNA, showing that decreased expression of eIF5A1 does not affect eIF5A2 target genes. Hsp90 is a loading control.

RESOURCES