Skip to main content
Journal of Interferon & Cytokine Research logoLink to Journal of Interferon & Cytokine Research
. 2013 May;33(5):227–238. doi: 10.1089/jir.2012.0142

The Oncogene eIF4E: Using Biochemical Insights to Target Cancer

Martin Carroll 1,, Katherine LB Borden 2,
PMCID: PMC3653383  PMID: 23472659

Abstract

The eukaryotic translation initiation factor eIF4E is overexpressed in many human malignancies where it is typically a harbinger of poor prognosis. eIF4E is positioned as a nexus in post-transcriptional gene expression. To carry out these functions, eIF4E needs to bind the m7G cap moiety on mRNAs. It plays critical roles in mRNA translation, mRNA export, and most likely in mRNA stability as well. Through these activities, eIF4E coordinately modulates the expression of many transcripts involved in proliferation and survival. eIF4E function is controlled by interactions with protein cofactors in concert with many signaling pathways, including Ras, Mnk, Erk, MAPK, PI3K, mTOR, and Akt. This review describes the eIF4E activity and provides several examples of cellular control mechanisms. Further, we describe some therapeutic strategies in preclinical and clinical development.

Overview

As multiple lines of evidence build that cancer cells are much like normal cells, it becomes increasingly critical to identify molecular targets that may act as an Achilles' heel for malignancy. One such target is the eukaryotic translation initiation factor eIF4E. eIF4E is a potent oncogene elevated in up to 30% of human malignancies, including carcinomas of the breast, prostate, lung, head, and neck as well as in many leukemias and lymphomas (Crew and others 1996; Nathan and others 1997a; De Benedetti and Harris 1999; Crew and others 2000; Berkel and others 2001; Seki and others 2002; Lee and others 2005; Matthews-Greer and others 2005; Culjkovic and Borden 2009; Borden and Culjkovic-Kraljacic, 2010). Increased eIF4E expression is associated with poor prognosis in patients [reviewed in (Culjkovic and Borden 2009)]. Furthermore, its overexpression in cell culture and in animal models leads to oncogenic transformation and tumor formation, respectively (De Benedetti and Rhoads 1990; Lazaris-Karatzas and others 1990; Avdulov and others 2004). On the cellular level, eIF4E overexpression is characterized by increased proliferation and survival (De Benedetti and Rhoads 1990; Lazaris-Karatzas and others 1990; Berkel and others 2001; Li and others 2003; Polunovsky and others 1996; Tan and others 2000; Avdulov and others 2004). It is an important part of many growth factor and stress signaling pathways (Ras, Erk, Mnk, PI3K, Akt, and mTOR; Brunn and others 1996; Mendez and others 1996; Brunn and others 1997; Waskiewicz and others 1997; Gingras and others 1998). In this review, we will focus on biochemical activities of eIF4E and their dysregulation in cancer. We will also describe how this knowledge has been translated into the clinic and discuss ways to further utilize knowledge of this target in advancing cancer therapy.

Molecular Functions of eIF4E

The best-described function for eIF4E is as a translation initiation factor involved in directing ribosomes to capped mRNAs (Fig. 1). During mRNA translation, eIF4E, a 25-kDa protein forms the eIF4F complex with the cofactors eIF4G, eIF4A, and eIF4B to recruit these to the ribosome (Sonenberg and Gingras 1998). To carry out this function, eIF4E relies on its ability to bind the 5′ methyl-7-guanosine (m7G cap) found on transcripts (Filipowicz and others 1976). This promotes the more efficient translation of transcripts by loading more ribosomes per transcript (Sonenberg and Gingras 1998). Importantly, eIF4E differentially affects transcripts. For instance, overexpression of eIF4E does not lead to elevation of all proteins, as observed by 35S-Met incorporation (Kaufman and others 1993). In fact, there was no detectable change in 35S-Met incorporation between controls and eIF4E overexpressing cells highlighting the specificity (Kaufman and others 1993). Concomitantly, antisense-mediated knockdown of eIF4E does not lead to global decreases in protein levels (Graff and others 2007). Studies by the Sonenberg laboratory highlight this specificity. For example, eIF4E overexpression leads to increased translational efficiency of ornithine decarboxylase transcripts, but cyclin D1 and GAPDH transcripts are unaffected at the translation level (Rousseau and others 1996). Transcripts that are most sensitive to eIF4E at the translation level are typically characterized by highly structured untranslated region (5′UTRs) (Sonenberg and Gingras 1998). For example, deletion of these structured elements in Pim1 transcripts substantially reduces their requirement for stimulation by eIF4E (Hoover and others 1997). Studying model transcripts, the Pestova and Hellen laboratories have shown that as the complexity of the 5′UTR is reduced, there is a loss of sensitivity for eIF4E in their translation (Pestova and others 2001). Thus, the complex 5′UTR sensitizes transcripts to eIF4E regulation, while housekeeping transcripts with short unstructured UTRs are generally not affected. Additionally, eIF4E may play novel functions in cap-assisted internal translation of histones, which is somewhat akin to a mixture of cap dependent and internal ribosome entry site mechanisms (Martin and others 2011). Here eIF4E can contact the RNA independently of the cap structure, although the cap is still required for translation (Martin and others 2011). Additionally, a cap-dependent translation can occur via the cap-binding complex both in the pioneer round of translation (Maquat and others 2010) and during stress responses, for example, (Garre and others 2011). Importantly, eIF4E is also found as a component of P-bodies and stress granules implying that it also plays a role in mRNA sequestration and/or turnover (Ferraiuolo and others 2005; Parker and Sheth 2007). Overall, the cytoplasmic function of eIF4E can be summarized as an enhancer of translation for a select group of transcripts particularly during periods of cellular stress and proliferation.

FIG. 1.

FIG. 1.

Model of 2 known eIF4E functions: mRNA export and mRNA translation. See text for details.

Aside from the cytoplasm, eIF4E exists in nuclear bodies and throughout the nucleoplasm in most cell types examined (Lejbkowicz and others 1992; Iborra and others 2001; Topisirovic and others 2003b) (Fig. 1). In the nucleus, eIF4E acts to promote the export of a subset of growth promoting mRNAs, thereby increasing their cytoplasmic concentration (Rousseau and others 1996; Culjkovic and others 2005, 2006, 2007, 2008). Similar to the cytoplasm, eIF4E affects the export of only a specific subset of mRNAs encoding genes functioning in proliferation and survival. A ∼50 nucleotide structural element in the 3′UTR of transcripts, denoted an eIF4E sensitivity element (4E-SE), confers sensitivity to eIF4E at the export level (Culjkovic and others 2005, 2006, 2008). For instance, Lac-Z 4E-SE chimeras have their mRNA export promoted by eIF4E overexpression, whereas unmodified LacZ transcripts are unaffected. There may be other elements as well, but these have yet to be discovered. eIF4E-dependent mRNA export is independent of bulk mRNA export (Culjkovic and others 2005, 2006, 2008). Bulk mRNA export requires the association of export ribonucleoparticles (RNPs) with the TAP1 nuclear receptor, whereas eIF4E-dependent export uses the CRM1–Ran export pathway (Culjkovic and others 2006). In the nucleus, eIF4E associates with CRM1, a specific subset of transcripts and specific cofactors to facilitate export to the cytoplasm (Culjkovic and others 2006; Topisirovic and others 2009a). Interestingly, eIF4E overexpression alters the composition of the nuclear pore, which leads to increased mRNA export of sensitive transcripts (Culjkovic-Kraljacic and others 2012). In fact, the major constituent of the cytoplasmic fibrils of the nuclear pore RanBP2 is a suppressor of both the mRNA export and transformation activity of eIF4E (Culjkovic-Kraljacic and others 2012). This is consistent with spontaneous tumor formation observed in RanBP2 hypomorph mice (Chakraborty and others 2008). Here reduction in RanBP2 levels likely increases the efficiency of recycling export components back to the nucleus for subsequent rounds of export. Thus, eIF4E promotes gene expression of select groups of transcripts through both enhancement of nuclear export via the 4E-SE and through enhanced translation in the cytoplasm of transcripts with complex 5′UTR structures.

Combinatorial Changes in Gene Expression Underlie the Potent Biological Effects of eIF4E

The RNA regulon model postulates that groups of RNAs acting in the same biochemical pathways have their regulation coordinated post-transcriptionally to ensure that all factors necessary for a particular function are present at the correct time (Tenenbaum and others 2000; Keene and Tenenbaum 2002; Keene and Lager 2005; Keene 2007). For instance, eIF4E enhances nuclear export of transcripts involved in proliferation, including most of the cyclins because these contain 4E-SE elements. In the cytoplasm, many of the nuclear export targets are also translation targets of eIF4E because they also contain a highly complex 5′UTR (Culjkovic and others 2006). In this way, the production of these factors is coordinated enabling a robust biological effect. Thus, eIF4E via these 2 activities in mRNA processing can promote proliferation and survival (Culjkovic and others 2007) by ensuring adequate and balanced expression of critical regulators of these activities. Consistent with this idea, both the mRNA export and translation functions of eIF4E contribute to its effects in oncogenic transformation and apoptotic rescue in cell culture (De Benedetti and Rhoads 1990; Lazaris-Karatzas and others 1990; Polunovsky and others 1996; Tan and others 2000; Li and others 2003; Avdulov and others 2004; Culjkovic and others 2005, 2006, 2008).

Dysregulation of eIF4E in Cancer

eIF4E is highly elevated and dysregulated in many cancers, including those of the lung, bladder, colon, breast, prostate, cervix, ovary, thyroid, and squamous cell carcinoma of the head and neck and others (Crew and others 1996; Nathan and others 1997a; Nathan and others 1997b; De Benedetti and Harris 1999; Crew and others 2000; Berkel and others 2001; Li and others 2002; Seki and others 2002; Lee and others 2005; Matthews-Greer and others 2005; Culjkovic and Borden 2009; Pettersson and others 2011). Further, eIF4E is dysregulated in many hematological malignancies, including acute myeloid leukemia (AML), the blast crisis stage of chronic myeloid leukemia, and some lymphomas (Topisirovic and others 2003b; Assouline and others 2009; Culjkovic and Borden 2009). Importantly, in many cases, eIF4E is only dysregulated in a subset of each cancer type. For instance, eIF4E is upregulated in M4 and M5 AML subtypes, but not generally in M1, M2, or M3 subtypes (Topisirovic and others 2003b; Assouline and others 2009). eIF4E is elevated in only a subset of breast cancers, but in ∼98% of head and neck cancers [reviewed in (Culjkovic and Borden 2009; Pettersson and others 2011)]. Thus, an important question arises as to how eIF4E levels themselves are dysregulated and what is unique about this subset of tumors. Of note, it is unclear whether the eIF4E activity may be dysregulated in subsets of primary tumors where its expression is not elevated.

eIF4E transcript levels are known to be dysregulated by at least 3 mechanisms: gene amplification, transcriptional dysregulation, and mRNA stability [reviewed in (Culjkovic and Borden 2009)].These mechanisms are not mutually exclusive and can occur coincidently. Gene amplification has been observed in a subset of head and neck and breast cancers (Sorrells and others 1998; Sorrells and others 1999). The promoter of the eIF4E gene contains an E-box and is a direct transcriptional target of c-Myc (Schmidt 2004). Interestingly, c-Myc is an mRNA export and translation target of eIF4E suggesting a potential positive feedback mechanism (Culjkovic and others 2006, 2007). Transcriptional control goes beyond c-Myc as eIF4E is still produced in c-Myc null fibroblasts and its expression in these cells is still stimulated by addition of serum (Bush and others 1998). Other studies have implicated Sonic hedgehog signaling (Mainwaring and Kenney 2011) and p53 (Zhu and others 2005) in the control of EIF4E transcription, but these effects are likely mediated by the c-Myc interaction with the EIF4E promoter. The hnRNPK protein binds the EIF4E promoter and its regulation of eIF4E contributes to oncogenic transformation (Lynch and others 2005). Recent studies demonstrate that eIF4E is also a C/EBP target (Khanna-Gupta and others 2012). eIF4E is also an NFκB target where transduction of primary M4 and M5 AML cells with the IκB-SR results in relocalization of eIF4E as described below and also reduction in mRNA levels (Topisirovic and others 2003b). Finally, the stability of eIF4E mRNA is increased by the HuR and TIAR proteins (Mazan-Mamczarz and others 2006; Topisirovic and others 2009b). In primary M4 and M5 AML, increased HuR levels and activated NFκB coincident with the increased eIF4E expression are observed in these leukemias (Topisirovic and others 2009b). As with other observations of eIF4E, multiple mechanisms likely simultaneously alter transcript levels in human malignancies.

eIF4E-Cap Recognition

The defining biochemical feature of eIF4E is its specific association with the m7G cap on the 5′ end of mRNAs (Filipowicz and others 1976). eIF4E binds unmethylated cap with 100-fold less affinity than the m7G cap although this varies depending on the conditions used (Liu and others 2011). The m7G cap intercalates between 2 tryptophan residues of eIF4E (Marcotrigiano and others 1997, 1999). At pH 7.5, the m7G cap is a zwitterion and it is the positive charge component that is thought necessary for interaction with the negative pi electron clouds of the 2 tryptophans in the cap-binding site (Carberry and others 1990; Carberry and others 1991). However, crystal structures show that glycerol, a neutral molecule, binds eIF4E in the same pocket (Brown and others 2009) suggesting that other features can drive interactions within the cap-binding site. Interestingly, eIF4E also binds analogues with benzene or phenol substituted for the methyl group (Carberry and others 1990). The benzylate-7-G cap analogue binds eIF4E more tightly than m7G cap indicating that the cap-binding pocket is adjustable (Carberry and others 1990). Tri-methylated guanosine cap is found on many RNAs involved in splicing and also binds with about a 10-fold reduced affinity to human eIF4E relative to m7G cap. However, in nematodes, tri-methylated and m7G cap have similar affinities for eIF4E (Liu and others 2011). Despite the weaker affinity for eIF4E in humans, the tri-methylated cap still binds with a nearly 50-fold greater affinity than guanosine triphosphate (GTP). The crystal structures for eIF4E m7GTP or tri-methylated cap complexes are highly similar (Liu and others 2011). Thus, although m7G cap binding is the defining biochemical feature of eIF4E, it can bind other moieties adding to its list of known and possible functions.

Studies of other cap binding proteins can enhance our understanding of the critical structural elements of the eIF4E- m7G cap interaction. For example, although other m7G cap binding proteins, such as VP39 and CBP20, use the aromatic stacking model for cap recognition (Quiocho and others 2000), there are other cap binding proteins, which specifically bind the m7G cap that do not use this strategy. Reovirus polymerase lamba3 uses a pocket where the tryptophan is perpendicular rather than planar to the m7-guanosine moiety and also utilizes aliphatics for recognition to bind m7G cap (but not guanosine) (Tao and others 2002). Similarly, the decapping scavenger enzyme, DcpS, binds m7G cap and not guanosine through a half aromatic sandwich model whereby there is 1 tryptophan present, but for the other side, aliphatic residues are packed onto the ring (Gu and others 2004). A third member of the eIF4E family, eIF4E-3, has only 1 tryptophan with a cysteine in the other traditional aromatic position (Osborne and others, in press). Here the cap is recognized by the conserved tryptophan and sandwiched with backbone residues (Osborne and others, in press). Consistently, mutation of the conserved tryptophan 102 to leucine in eIF4E only modestly reduces m7G cap binding, while mutation to alanine completely abrogates these affects (Osborne and others, in press). Thus, principles for cap recognition are not restricted to the aromatic sandwich, and even in eIF4E, there is much to still learn about ligand recognition.

These findings have important implications for drug design and understanding the molecular basis of competitive inhibitors of cap binding, such as ribavirin. Ribavirin and its active metabolite ribavirin triphosphate (RTP) bind eIF4E with similar affinity to the m7G cap in a phosphate buffer pH 7.5 as measured by fluorescence quenching, cap chromatography, circular dichroism, mass spectrometry, and NMR (Kentsis and others 2004; Kentsis and others 2005; Borden and Culjkovic-Kraljacic 2010; Volpon and others, submitted). NMR studies with RTP indicate that the ligand binds in the same cap binding site as m7G cap, but its exact location in the pocket is likely slightly different (Volpon and others, submitted). Further, 3H ribavirin immunoprecipitated with endogenous eIF4E in live cells (Borden and Culjkovic-Kraljacic 2010). It may be feasible to generate more potent inhibitors of eIF4E binding to the m7G cap although whether such inhibitors would be safe for human therapy is unclear.

Direct Regulators of eIF4E

Regulation of eIF4E is extremely complex, as one would expect for a protein that modulates gene expression so potently. There are, to date, 3 classes of direct regulators of eIF4E described: those that contain an eIF4E consensus binding motif (YXXXXLΦ), those with zinc binding really interesting new gene (RING) domains, and a class of viral proteins that contain neither of these motifs (summarized in Table 1). All of these proteins bind the dorsal surface of eIF4E, which is about 35 Å away from the m7G cap-binding site. They mediate changes to activity by either blocking the ability of eIF4E to bind other factors, such as eIF4G, and/or mediate allosteric effects. These regulators can be present in the nucleus and/or cytoplasm suggesting that the biochemical basis for control is similar between compartments. The overall effect of these diverse regulators in specific tissues and organisms is incompletely understood; however, the plethora of regulators suggest that fine-tuning of the diverse activities of eIF4E is critical.

Table 1.

Classes of eIF4E Regulatorsa

Class I: Consensus YXXXXLΦ
eIF4G, BP1,b CUP,b Neurogidin, CYF1F1F, LRPPRC, GEMIN5, 4E-T, DDX3 Homeodomain subset: Bicoid, PRH/Hex, HoxA9, Emx-2, En2 (only a subset given, over 200 potential to date)
Class II: RING motif
PML, arenaviral Z, HHARI
Class III: Potyviral genome linked VPg- amphipathic helix
VPg from potyviruses
a

Other classes may also exist.

b

Additional binding sites act with the consensus site.

The first class of regulators is characterized by a consensus-binding motif first identified in eIF4G and eIF4E-BP1 (Mader and others 1995). The motif is YXXXXLΦ where X is any residue and Φ is any hydrophobic residue (Mader and others 1995). Structural studies indicate that the motif forms a helix and a reversed L-shape structure that interacts with the dorsal surface of eIF4E (Marcotrigiano and others 1999; Tomoo and others 2005). New structural studies demonstrate that BP1 and CUP, two proteins that use this motif, utilize the consensus sequence in concert with another part of the protein, in fact, having 2 distinct binding sites for eIF4E (Kinkelin and others 2012).

BP1 is the best-studied regulator of eIF4E. Its association with the dorsal surface competes for association with eIF4G, and thereby inhibits translation by blocking formation of the translation initiation complex (Haghighat and others 1995). Additionally, binding BP1 increases affinity of eIF4E for the cap through allosteric changes (Ptushkina and others 1998; Ptushkina and others 1999; Tomoo and others 2005). This is consistent with the model postulated by the McCarthy laboratory whereby BP1 inhibits protein translation by not only blocking binding of eIF4G, but also by sequestering the RNA bound to eIF4E from active translation complexes (Ptushkina and others 1998; Ptushkina and others 1999; Tomoo and others 2005). Phosphorylation of BP1, which can occur via multiple pathways (see Targeting eIF4E at multiple levels in the clinic), leads to unfolding of the recognition helix and thus impairs BP1 binding to eIF4E (Tait and others 2010). Of note, BP1 is found in both the nucleus and the cytoplasm indicating that it likely modulates mRNA export as well as translation (Rong and others 2008). BP1 overexpression suppresses the ability of eIF4E to transform cells. Highlighting the importance of other regulators of eIF4E, BP1 null mice do not have a substantial phenotype (Blackshear and others 1997) although there is some evidence of adipocyte tissue reduction (Tsukiyama-Kohara and others 2001). There are 3 homologues of BP1: BP1, BP2, and BP3, the latter of which has a very limited tissue distribution. The double knockout of BP1 and BP2 leads to diet-induced obesity and insulin resistance (Le Bacquer and others 2007). Interestingly, and highlighting the complex regulation of eIF4E, BP1 or BP1/2 null mice do not get cancers more readily than littermate controls (Tsukiyama-Kohara and others 2001; Le Bacquer and others 2007). Of note, BP1 is not conserved in Saccharomyces cervisiae, which use Caf20p and Eap1p to regulate translation. These proteins have no apparent sequence homology outside of the consensus binding motif, but they can still inhibit human eIF4E activity (Altmann and others 1989). Caf20p and Eap1p regulate the translation of specific transcripts suggesting that binding proteins can specifically regulate a subset of eIF4E-sensitive transcripts (Cridge and others 2010). These observations showcase the importance of other regulators to the control of eIF4E activity and specificity of these regulators.

In cancer, there are many surprising results with regard to BP1. BP1 levels are elevated in breast cancer where increased levels and phosphorylation are correlated with a more advanced stage of the disease (Armengol and others 2007). This correlation is actually stronger than the correlation with eIF4E levels themselves (Armengol and others 2007). Tissues from esophageal cancers have more eIF4E-BP1 complexes than adjacent normal tissue (Salehi and Mashayekhi 2006). Similar results are seen in primary M4 and M5 AML specimens that have highly elevated eIF4E (3–10-fold) (Assouline and others 2009) and similarly highly elevated BP1 and phospho-BP1 (Culjkovic-Kraljacic and Borden, unpublished). The basis for this increased expression of BP is not clear, but may reflect compensation within the tumor cells for increasing eIF4E levels. This and other observations make it challenging to come up with single variable models correlating eIF4E expression with tumor growth.

Other proteins that use the consensus motif to bind to and regulate eIF4E are described in a wide variety of functional classes of proteins that can both stimulate and repress eIF4E activity. For instance, about 200 homeodomain containing proteins include the consensus-binding site. Of these, HoxA9, Hex/PRH, Hox11, Bicoid, Emx-2, and Engrailed 2 are confirmed to directly bind eIF4E (Niessing and others 2002; Topisirovic and others 2003a; Nedelec and others 2004; Brunet and others 2005; Topisirovic and others 2005). HoxA9 is demonstrated to promote the mRNA export and translation activities of eIF4E (Topisirovic and others 2005). By contrast, Hex/PRH suppresses the nuclear function and does not modulate translation presumably due to the fact it is restricted to the nucleus (Topisirovic and others 2003a). Interestingly, in M4 and M5 AML, Hex/PRH is substantially downregulated and mislocalized suggesting that loss of this level of eIF4E regulation could contribute to leukemogenesis (Topisirovic and others 2003a). The RNA helicase DDX3 also uses a consensus-binding site to directly associate with eIF4E and modulates translation, and potentially functions in P-bodies and in mRNA export (Shih and others 2008; Topisirovic and others 2009a; Shih and others 2011). The mRNA export cofactor LRPPRC associates with eIF4E in the nucleus using the consensus site (Topisirovic and others 2009a). In fact, LRPPRC overexpression enhances eIF4E-dependent mRNA export and mutation of the consensus binding site abrogates this (Topisirovic and others 2009a). Other factors in this class include Gemin5, the translational repressor CYF1F1, Neurogidin, and many others (Culjkovic and Borden 2009; Rhoads 2009). Taken together, the consensus-binding motif is utilized in likely hundreds of regulators of eIF4E in tissue- and context-specific fashions.

A structurally distinct class of eIF4E regulators utilize their RING domains to interact with eIF4E and modulate its function (Kentsis and others 2001; Kentsis and others 2002a, 2002b; Volpon and others 2010; Culjkovic-Kraljacic and others 2012). To date, the promyelocytic leukemia protein (PML), and the arenaviral Z proteins from lymphocytic choriomeningitis virus and lassa fever virus have been shown to directly bind eIF4E via their RING domains, while human homologue of ariadne (HHARI) binds an eIF4E homologue 4EHP (Kentsis and others 2001; Kentsis and others 2002a; 2002b; Tan and others 2003; Volpon and others 2010). Biochemical and solution structure studies indicate that the RING domains of PML and Z directly bind the dorsal surface in a region that overlaps with, but is distinct from, the area bound by consensus site containing factors (Kentsis and others 2001; Kentsis and others 2002a, 2002b; Volpon and others 2010). The association of the RING with eIF4E leads to allosteric changes in the cap-binding site that correlated with up to a 100-fold reduction in cap binding. Cap binding is required for eIF4E-dependent mRNA export (Culjkovic and others 2005, 2006). This is consistent with previous observations that PML, which is almost entirely a nuclear protein suppresses the mRNA export functions of eIF4E (Cohen and others 2001). In fact, PML overexpression represses eIF4E-mediated oncogenic transformation through this interaction (Cohen and others 2001; Culjkovic and others 2006). Addition of purified PML RING or the viral Z protein in reticulocyte lysates leads to repression of translation, suggesting that PML and Z could impair translation when localized to the cytoplasm (Kentsis and others 2001). In the context of viral infection, Z binds other viral factors that likely impact on its eIF4E effects (Kentsis and others 2001). Importantly, PML, Z, and HHARI do not alter eIF4E/4EHP protein levels suggesting that these functions are independent of any functions they may play in ubiquitination or related pathways (Kentsis and others 2001; Kentsis and others 2002a, 2002b; Tan and others 2003).

The third class of known eIF4E binding proteins, which come from the plant potyviruses, comprise the potyviral genome-linked proteins VPg, which directly bind eIF4E and reduce its affinity for the m7G cap by about 15-fold (Michon and others 2006; Roudet-Tavert and others 2007; German-Retana and others 2008). VPg contains neither a consensus motif nor RING domain. Mapping studies suggest that VPg uses an amphipathic helix to associate with eIF4E and, importantly, does not bind to the same surface as the consensus binding proteins (Michon and others 2006; Roudet-Tavert and others 2007; German-Retana and others 2008). In support of this, VPg increases the affinity of eIF4E for eIF4G, a protein that does use the consensus-binding motif. Further structural studies will elucidate the molecular underpinnings of this interaction and will likely enable one to identify mammalian proteins that may use the same or similar strategies to associate with and regulate eIF4E.

Taken together, it is clear that the regulation of eIF4E is complicated with many context- and tissue-specific factors in play. Redundancy of regulation is the likely key for the normal control of both the nuclear and cytoplasmic functions of eIF4E. Further, in malignancies, it is likely that the overall level of eIF4E activity is critical and not simply the level of protein expression.

The Subcellular Localization of eIF4E

eIF4E is found throughout eukaryotic cells (Culjkovic and Borden 2009). It is found in the cytoplasm and can be distributed both diffusively through the cytosol or it can be found associated with P-bodies and stress granules under appropriate conditions (Ferraiuolo and others 2005; Parker and Sheth 2007). In the nucleus, eIF4E is found in both nuclear bodies and diffusely throughout the nucleoplasm, but is excluded from the nucleolus (Lejbkowicz and others 1992; Cohen and others 2001). A subset of nuclear bodies colocalize with those formed by PML. This latter localization is dependent on the RING domain of PML (Cohen and others 2001; Topisirovic and others 2003a). As noted above, PML is a potent repressor of eIF4E-dependent mRNA export and, subsequently, eIF4E-mediated oncogenic transformation (Cohen and others 2001). The relative cellular distribution of eIF4E is context and tissue-type dependent (Culjkovic and Borden 2009). Obviously, its subcellular distribution will have a profound impact on its function, but it is incompletely understood how its cellular localization is regulated.

Studies by the Borden laboratory have demonstrated one example of how eIF4E subcellular localization is dysregulated in malignancy. In primary leukemic blast cells isolated from patients with the M4 and M5 subtypes of AML, and in the M5 AML cell line THP-1, eIF4E is not only highly elevated, but also concentrated in the nucleus (Topisirovic and others 2003a, 2003b; Kraljacic and others 2011). By contrast, normal hematopoietic stem and progenitor CD34+ cells have discrete nuclear bodies and a cytoplasmic diffuse staining found in many other normal tissue types (Topisirovic and others 2003a; Topisirovic and others 2003b; Kraljacic and others 2011). Increased nuclear accumulation in these AML cells is associated with increased eIF4E-dependent mRNA export and, thus, this dysregulation contributes to oncogenesis (Topisirovic and others 2003b; Kraljacic and others 2011). In primary AML, the distribution of eIF4E is controlled, at least in part, by NFκB. Introduction of the NFκB repressor, IκB-SR, leads to a remarkable redistribution of eIF4E so that its localization is indistinguishable from normal CD34+ stem and progenitor cells, that is, there are discrete nuclear bodies with a diffuse cytoplasmic localization (Topisirovic and others 2003b). The effects of NFκB in this context are likely indirect. As described below, studies with ribavirin have shown activity in the therapy of AML patients with highly elevated nuclear eIF4E levels, and these molecular studies suggest that targeting mRNA export function can be clinically useful (Assouline and others 2009).

Basic laboratory studies have demonstrated that some direct regulators of eIF4E also modulate the subcellular distribution of eIF4E. The 4E-Transporter protein (4E-T) utilizes its consensus-binding site to bind eIF4E. 4E-T overexpression leads to relocalization of eIF4E to the cytoplasm where it is found in a subset of P-bodies (Ferraiuolo and others 2005; Cargnello and others 2012). Hex/PRH overexpression, again via its consensus binding site, causes relocalization of nuclear eIF4E to the cytoplasm (Topisirovic and others 2003a). This substantially impairs its mRNA export activity and its ability to transform cells (Topisirovic and others 2003a). LRPPRC utilizes its consensus-binding site to compete for PML RING binding to eIF4E in the nucleus (Topisirovic and others 2009a). Here LRPPRC overexpression leads to reduced PML-eIF4E association at nuclear bodies and more nucleoplasmic eIF4E. This correlates with increased mRNA export activity of eIF4E (Topisirovic and others 2009a). These studies demonstrate clearly that it is not simply nuclear versus cytoplasmic localization of eIF4E that is critical, but the exact distribution of the protein within the nucleus or cytoplasm that affects the overall function of eIF4E in modulating nuclear mRNA export, mRNA translation, and stress responses.

The molecular work above has led to the identification of inhibitors of eIF4E function. For example, treatment of cells with a cap analogue m7GpppG leads to disruption of eIF4E nuclear bodies (Dostie and others 2000; Cohen and others 2001). Similarly, and importantly, treatment with a competitive inhibitor of the m7G cap, ribavirin, redistributes nuclear eIF4E to the cytoplasm without altering other subnuclear organelles (Kentsis and others 2004; Assouline and others 2009). Ribavirin is a food and drug administration approved antiviral drug. The parent compound is actually a prodrug that is metabolized into RTP (Glue 1999). Leptomycin B, the CRM1 inhibitor, leads to a nuclear accumulation of eIF4E (Topisirovic and others 2009a). Thus, both ribavirin and leptomycin B treatment lead to reduced mRNA export by eIF4E; one by displacing eIF4E to the cytoplasm and the other by inhibiting its export function, which is CRM1 dependent (Kentsis and others 2004; Culjkovic and others 2006).

In summary, eIF4E localization within and between the nucleus and cytoplasm can be modulated by many factors. Ultimately, modulating eIF4E trafficking can have profound effects on its mRNA export, mRNA sequestration, and translation roles. Given that eIF4E only affects the expression of certain transcripts at one of these levels, for instance, vascular endothelial growth factor is a translation, but not an export target (Culjkovic and others 2006), this leads to differential effects on the proteome. Thus, modulators of the subcellular distribution of eIF4E differentially effect gene expression thereby modulating the biological effects of the cells tailoring responses to given stimuli or stresses.

Targeting eIF4E at multiple levels in the clinic

As described above, eIF4E can also be dysregulated at the functional level by post-translational modifications. Such modification can occur downstream of activation of a number of common signaling pathways, including RAS, mitogen-activated kinase (MAPK), phosphatidylinositol-3-kinase (PI3K), and Mnk kinases. A full description of the regulation of these pathways and their diverse effects is beyond the scope of this review, and we have selected a few critical points for discussion here. Conceptually, 3 steps regulate the eIF4E function—its phosphorylation, its binding to regulatory cofactors, and its binding to cap complexes. Under normal cellular conditions, eIF4F complex assembly is limited as eIF4E is sequestered from eIF4G by binding to BP1 and other regulators. Stimulation of the PI3K/AKT/mTOR pathway leads to hierarchical 4EBP phosphorylation, dislodging 4EBP from eIF4E and enabling assembly of the eIF4F complex. This process is regulated by the PI3K/mammalian target of the rapamycin (mTOR) pathway and other signaling networks. Many clinical studies have used mTOR inhibitors as cancer therapeutics and it is assumed, but not proven that they work in part through inhibition of mRNA translation. One difficulty in interpreting in vivo work with rapalogs is the difficulty of assessing 4EBP1 phosphorylation, eIF4E activity, or mRNA translation in primary tissue. Thus, phosphorylation of the highly abundant ribosomal S6 protein is the typical surrogate for mTORC1 inhibition (Perl and others 2012). Interesting work in AML is consistent, however, with the hypothesis that rapamycin is an incomplete inhibitor of cap-dependent translation suggesting that the inhibition of eIF4E activity through rapamycin-dependent inhibition of mTORC1 is likely less than complete (Tamburini and others 2009). This and other data suggest that combination of drugs that feed into mRNA translation regulation would have a strong rationale in the clinic. Consistent with this idea, recent work has shown that combinations of rapamycin with Mnk inhibition can augment cytotoxic effects in T cell lymphoma (Marzec and others 2011). New families of drugs that can inhibit both mTORC1 and mTORC2 are currently being tested with hopefully more potent effects.

eIF4E can also be dysregulated via its enhanced phosphorylation. eIF4E is phosphorylated at S209, although the necessity of this event for function of eIF4E is not entirely clear and may be different in normal and malignant tissues. Mnk kinases phosphorylate eIF4E when it is bound to eIF4G and perhaps other factors. The effect of phosphorylation on eIF4E cap binding affinity is controversial with some studies suggesting an increase in affinity, while others suggest reduced affinity (Scheper and Proud 2002). Mnk1 was identified by homology to other serine threonine kinases and quickly recognized as a target of both Erk and p38 MAP kinases. It was also demonstrated to phosphorylate eIF4E on S209 (Waskiewicz and others 1997). This phosphorylation event increases cap-dependent translation in some, but not all systems. As noted, in normal cells, S209 phosphorylation may not be critical as a Mnk1/Mnk2 deficient mouse strain has a normal phenotype despite the complete lack of detectable eIF4E S209 phosphorylation (Ueda and others 2004). On the other hand, mutation of this residue completely abrogates its ability to transform cell lines and lead to tumors in animal models (Topisirovic and others 2004; Wendel and others 2004). Here as elsewhere, the suggestion is that highly elevated eIF4E expression and function are associated with malignant transformation, but not normal mRNA translation/metabolism regulation. Such specificity again suggests that Mnk kinases may be appropriate targets for cancer therapy. A small molecular inhibitor (Tschopp and others 2000; Knauf and others 2001) was selected from a drug library and demonstrated to potently inhibit Mnk kinases, although nonspecifically. The compound, CGP57380, can inhibit transformation in cells overexpressing eIF4E (Topisirovic and others 2004), the growth of BCR/ABL-dependent cell lines (Zhang and others 2008), and survival of breast cancer cell lines (Wheater and others 2010). An orally available Mnk kinase inhibitor has recently been described. The compound inhibits the growth of melanoma and colon cancer xenografts without significant toxicity providing proof of principle that Mnk targeting is a promising avenue of investigation for cancer therapeutics (Konicek and others 2011). Interestingly, inhibition of Mnk kinase may be a double-edged sword. Elegant work by the Platanias laboratory has revealed that Mnk kinase plays a critical role in the growth inhibitory function of interferons (Joshi and others 2009; Altman and others 2010; Joshi and others 2011). Indeed these effects have been suggested to play a role in normal hematopoiesis (Joshi and others 2011).

Given these many associations with human malignancy, there has been substantial interest in targeting eIF4E both for therapeutic purposes and to increase understanding of its function. Studies dating back to 1991 by the De Benedetti and Zimmer laboratories demonstrated that antisense-mediated repression of eIF4E expression would slow cell growth in cancer cell lines (De Benedetti and others 1991; Rinker-Schaeffer and others 1993; Graff and others 1997). Graff and others (2007) took a substantial step forward in application of this approach with development of an eIF4E targeting antisense molecular containing a phosphorothioate backbone core (to promote RNase H–mediated degradation) flanked with five 2-methoxyethyl-modified (MOE-modified) bases to improve potency, nuclease resistance, and tissue half-life. Animal studies demonstrated that intravenous administration of this modified antisense oligonucleotide molecule decreased expression of eIF4E in multiple organs by nearly 80% and decreased growth of xeno-transplanted tumor cells without inducing toxicity (Graff and others 2007). This study demonstrated proof of principle that eIF4E targeting strategies would have a therapeutic index that would allow targeting to the molecule in cancer cells without the induction of toxicity (Graff and others 2007). This molecule (now designated LY2275796) has been taken forward into Phase I clinical trials and results were recently described (Hong and others 2011). In this trial, LY2275796 was given over an initial 3-day loading dose followed by weekly maintenance. The drug was well tolerated with only mild to modest toxicities observed. Importantly, the authors were able to obtain pretreatment and post-treatment biopsies and document slight decreases in eIF4E expression in most patients, but not nearly as extensive as observed in their animal models. No clinical responses were seen, perhaps due to inefficient knockdown of eIF4E or to synthetic lethal relationships with other pathways. There was no clear increase in efficacy at decreasing eIF4E between the top 2 doses and a dose of 1000 mg was chosen as both the maximum tolerated dose and the biologically effective dose. The authors conclude that administration of LY2275796 is feasible. However, the decrease in eIF4E expression in tumors was modest and it may be inadequate to provide therapeutic benefit by itself in humans. Combination studies of LY2275796 with chemotherapeutic agents are planned.

A more recent effort has involved characterization of a small molecular inhibitor, 4EGI-1 (Moerke and others 2007). Moerke and others initially identified this molecule and demonstrated that it could inhibit growth of cancer cell lines. Tamburini and others (2009) extended these results and demonstrated that 4EGI-1 inhibits the growth of primary AML cells. Of note, the authors primarily focused on the role of eIF4E in modulating mRNA translation and whether 4EGI-1 alters transcript export in AML cells is unknown. Relatively few other studies with this inhibitor have been published, although they are consistent with the data that 4EGI-1 can inhibit growth of cancer cells in myeloma, melanoma, and breast tumors (Pettersson and others 2011; Chen and others 2012; Descamps and others 2012). Subsequent work suggested that the molecule is not potent enough for in vivo work and may have other mechanisms of action, including effects on TRAIL (Blagden and Willis 2011).

Peptide inhibition using a novel tissue-specific targeting approach also suggests proof of principle that eIF4E is a potential target in ovarian cancer (Ko and others 2009). These studies demonstrate an elegant targeting mechanism by fusion of BP1 with a steroid agonist so that only ovarian cancer cells are targeted. These approaches, however, are not robust for clinical translation leading to a search for other inhibitors.

The Borden laboratory initially described the inhibitory effects of the antiviral, ribavirin, on eIF4E function (Kentsis and others 2004; Kentsis and others 2005; Assouline and others 2009). Direct binding studies indicate that ribavirin binds eIF4E in specific conditions, including at physiological pH and salt concentrations (Kentsis and others 2004; Kentsis and others 2005; Volpon and others, submitted). Studies have shown activity of ribavirin in inhibiting growth of breast cancer cell lines in an eIF4E-dependent manner (Pettersson and others 2011). Ribavirin is a nucleoside analog that also inhibits cellular inosine monophoshpate dehydrogenase (IMPDH) and leads to a decrease in cellular GTP levels (Hong and Cameron 2002). Thus, descriptions of its anticancer activity may reflect diverse mechanisms of action, a favorable characteristic of the drug. Importantly, a ribavirin analogue that is a potent IMPDH inhibitor, tiazofurin, has no effect on eIF4E activity in translation or mRNA export and is completely unable to repress eIF4E-mediated transformation (Tan and others 2008). Further, mycophenolic acid, another potent IMPDH inhibitor, does not modulate eIF4E activity (Culjkovic-Kraljacic and Borden, unpublished observation). Ribavirin is used for treatment of viral infections and its safety profile is well known. Investigators at McGill have led a multicenter trial studying ribavirin as a single agent for treatment of patients with AML of the M4/M5 subtype (Assouline and others 2009). Patients were refractory, relapsed, or unable to undergo chemotherapy. Among 11 evaluable patients, there were 1 complete remission and 2 partial remissions (overall 3/11 or 27% response rate in patients with high-risk disease). Importantly, ribavirin-induced relocalization of nuclear eIF4E to the cytoplasm and reduction of eIF4E levels were associated with a clinical response demonstrating a pharmacodynamics correlation with results described in vitro (Assouline and others 2009). Lack of response or relapse coincided with continued or renewed nuclear localization of eIF4E (Assouline and others 2009). Ribavirin similarly targeted the eIF4E activity in an ongoing combination study with low-dose ara-C, where prolonged remissions have been observed [ClinicalTrials.gov NCT01056523 & (Assouline and others 2011). Indeed, ribavirin combined well with standard chemotherapies in primary AML specimens ex vivo (Kraljacic and others 2011]. Development of an eIF4E inhibitor that can safely be used in cancer therapy is an important goal and this proof of principle experiment suggests that this widely used drug may be an important agent in cancer therapy although it is unlikely to be highly active as a single agent.

Conclusions

To summarize, multiple lines of evidence suggest that control of post-transcriptional mechanisms of gene expression is a critical node for regulation of cancer cell growth. This node is largely regulated through the diverse functions of eIF4E. Although many studies have focused on the role of eIF4E in regulation only of mRNA translation, its regulation of cell growth occurs at multiple levels, only some of which are elucidated. Regulation of eIF4E itself is complex and initial studies summarized above show that small molecule inhibitors of some of the functions of eIF4E may be promising cancer therapeutics. Although inhibitors of Mnk and mTOR kinases are also being used as approaches to inhibiting mRNA translation, these approaches will not inhibit all of the functions of eIF4E. Future studies focusing on drug combinations of ribavirin or other eIF4E inhibitors with signal transduction inhibitor studies should be of great interest.

Acknowledgments

MC is supported by the National Cancer Institute Grant R21CA153018-01 and RO1CA149566. KLBB is supported by grants from the NIH and LLS. KLBB holds a Canada Research Chair. The Institute for Research in Immunology and Cancer receives support from the CIHR and FRSQ.

Author Disclosure Statement

KLBB has no competing financial interests. MC has received research funding from Tetralogic Pharmaceuticals, Glaxo Smith Kline, and Sanofi Aventis Pharmaceuticals.

References

  1. Altman JK. Glaser H. Sassano A. Joshi S. Ueda T. Watanabe-Fukunaga R. Fukunaga R. Tallman MS. Platanias LC. Negative regulatory effects of Mnk kinases in the generation of chemotherapy-induced antileukemic responses. Mol Pharmacol. 2010;78(4):778–784. doi: 10.1124/mol.110.064642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Altmann M. Muller PP. Pelletier J. Sonenberg N. Trachsel H. A mammalian translation initiation factor can substitute for its yeast homologue in vivo. J Biol Chem. 1989;264(21):12145–12147. [PubMed] [Google Scholar]
  3. Armengol G. Rojo F. Castellvi J. Iglesias C. Cuatrecasas M. Pons B. Baselga J. Ramon y. Cajal S. 4E-binding protein 1: a key molecular “funnel factor” in human cancer with clinical implications. Cancer Res. 2007;67(16):7551–7555. doi: 10.1158/0008-5472.CAN-07-0881. [DOI] [PubMed] [Google Scholar]
  4. Assouline S. Culjkovic B. Cocolakis E. Rousseau C. Beslu N. Amri A. Caplan S. Leber B. Roy DC. Miller WH., Jr. Borden KL. Molecular targeting of the oncogene eIF4E in acute myeloid leukemia (AML): a proof-of-principle clinical trial with ribavirin. Blood. 2009;114(2):257–260. doi: 10.1182/blood-2009-02-205153. [DOI] [PubMed] [Google Scholar]
  5. Assouline S. Kraljacic-Culjkovic B. Cocolakis E. Amri A. Bergeron J. Jamal R. Miller WH., Jr. Borden KL. A Phase I Combination Study of Ribavirin and Low Dose Cytarabine Arabinoside (ara-C) in M4/M5 Acute Myeloid Leukemia (AML) and AML with High eIF4E. Blood (ASH Annual Meeting Abstracts) 2011;118:3606. [Google Scholar]
  6. Avdulov S. Li S. Michalek V. Burrichter D. Peterson M. Perlman DM. Manivel JC. Sonenberg N. Yee D. Bitterman PB. Polunovsky VA. Activation of translation complex eIF4F is essential for the genesis and maintenance of the malignant phenotype in human mammary epithelial cells. Cancer Cell. 2004;5(6):553–563. doi: 10.1016/j.ccr.2004.05.024. [DOI] [PubMed] [Google Scholar]
  7. Berkel HJ. Turbat-Herrera EA. Shi R. de Benedetti A. Expression of the translation initiation factor eIF4E in the polyp-cancer sequence in the colon. Cancer Epidemiol Biomarkers Prev. 2001;10(6):663–666. [PubMed] [Google Scholar]
  8. Blackshear PJ. Stumpo DJ. Carballo E. Lawrence JC., Jr Disruption of the gene encoding the mitogen-regulated translational modulator PHAS-I in mice. J Biol Chem. 1997;272(50):31510–31514. doi: 10.1074/jbc.272.50.31510. [DOI] [PubMed] [Google Scholar]
  9. Blagden SP. Willis AE. The biological and therapeutic relevance of mRNA translation in cancer. Nat Rev Clin Oncol. 2011;8(5):280–291. doi: 10.1038/nrclinonc.2011.16. [DOI] [PubMed] [Google Scholar]
  10. Borden KL. Culjkovic-Kraljacic B. Ribavirin as an anti-cancer therapy: acute myeloid leukemia and beyond? Leuk Lymphoma. 2010;51(10):1805–1815. doi: 10.3109/10428194.2010.496506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Brown CJ. Verma CS. Walkinshaw MD. Lane DP. Crystallization of eIF4E complexed with eIF4GI peptide and glycerol reveals distinct structural differences around the cap-binding site. Cell Cycle. 2009;8(12):1905–1911. doi: 10.4161/cc.8.12.8742. [DOI] [PubMed] [Google Scholar]
  12. Brunet I. Weinl C. Piper M. Trembleau A. Volovitch M. Harris W. Prochiantz A. Holt C. The transcription factor Engrailed-2 guides retinal axons. Nature. 2005;438(7064):94–98. doi: 10.1038/nature04110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Brunn GJ. Hudson CC. Sekulic A. Williams JM. Hosoi H. Houghton PJ. Lawrence JC., Jr. Abraham RT. Phosphorylation of the translational repressor PHAS-I by the mammalian target of rapamycin. Science. 1997;277(5322):99–101. doi: 10.1126/science.277.5322.99. [DOI] [PubMed] [Google Scholar]
  14. Brunn GJ. Williams J. Sabers C. Wiederrecht G. Lawrence JC., Jr. Abraham RT. Direct inhibition of the signaling functions of the mammalian target of rapamycin by the phosphoinositide 3-kinase inhibitors, wortmannin and LY294002. EMBO J. 1996;15(19):5256–5267. [PMC free article] [PubMed] [Google Scholar]
  15. Bush A. Mateyak M. Dugan K. Obaya A. Adachi S. Sedivy J. Cole M. c-myc null cells misregulate cad and gadd45 but not other proposed c-Myc targets. Genes Dev. 1998;12(24):3797–3802. doi: 10.1101/gad.12.24.3797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Carberry SE. Darzynkiewicz E. Goss DJ. A comparison of the binding of methylated cap analogues to wheat germ protein synthesis initiation factors 4F and (iso)4F. Biochemistry. 1991;30(6):1624–1627. doi: 10.1021/bi00220a026. [DOI] [PubMed] [Google Scholar]
  17. Carberry SE. Darzynkiewicz E. Stepinski J. Tahara SM. Rhoads RE. Goss DJ. A spectroscopic study of the binding of N-7-substituted cap analogues to human protein synthesis initiation factor 4E. Biochemistry. 1990;29(13):3337–3341. doi: 10.1021/bi00465a027. [DOI] [PubMed] [Google Scholar]
  18. Cargnello M. Tcherkezian J. Dorn JF. Huttlin EL. Maddox PS. Gygi SP. Roux PP. Phosphorylation of the eukaryotic translation initiation factor 4E-transporter (4E-T) by c-Jun N-terminal kinase promotes stress-dependent P-body assembly. Mol Cell Biol. 2012;32(22):4572–4584. doi: 10.1128/MCB.00544-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chakraborty P. Wang Y. Wei JH. van Deursen J. Yu H. Malureanu L. Dasso M. Forbes DJ. Levy DE. Seemann J. Fontoura BM. Nucleoporin levels regulate cell cycle progression and phase-specific gene expression. Dev Cell. 2008;15(5):657–667. doi: 10.1016/j.devcel.2008.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chen L. Aktas BH. Wang Y. He X. Sahoo R. Zhang N. Denoyelle S. Kabha E. Yang H. Freedman RY. Supko JG. Chorev M. Wagner G. Halperin JA. Tumor suppression by small molecule inhibitors of translation initiation. Oncotarget. 2012;3(8):869–881. doi: 10.18632/oncotarget.598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Cohen N. Sharma M. Kentsis A. Perez JM. Strudwick S. Borden KL. PML RING suppresses oncogenic transformation by reducing the affinity of eIF4E for mRNA. Embo J. 2001;20(16):4547–4559. doi: 10.1093/emboj/20.16.4547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Crew JP. Fuggle S. Bicknell R. Cranston DW. de Benedetti A. Harris AL. Eukaryotic initiation factor-4E in superficial and muscle invasive bladder cancer and its correlation with vascular endothelial growth factor expression and tumour progression. Br J Cancer. 2000;82(1):161–166. doi: 10.1054/bjoc.1999.0894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Crew JP. O'Brien TS. Harris AL. Bladder cancer angiogenesis, its role in recurrence, stage progression and as a therapeutic target. Cancer Metastasis Rev. 1996;15(2):221–230. doi: 10.1007/BF00437475. [DOI] [PubMed] [Google Scholar]
  24. Cridge AG. Castelli LM. Smirnova JB. Selley JN. Rowe W. Hubbard SJ. McCarthy JE. Ashe MP. Grant CM. Pavitt GD. Identifying eIF4E-binding protein translationally-controlled transcripts reveals links to mRNAs bound by specific PUF proteins. Nucleic Acids Res. 2010;38(22):8039–8050. doi: 10.1093/nar/gkq686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Culjkovic B. Borden KL. Understanding and targeting the eukaryotic translation initiation factor eIF4E in head and neck cancer. J Oncol. 2009;2009:981679. doi: 10.1155/2009/981679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Culjkovic B. Tan K. Orolicki S. Amri A. Meloche S. Borden KL. The eIF4E RNA regulon promotes the Akt signaling pathway. J Cell Biol. 2008;181(1):51–63. doi: 10.1083/jcb.200707018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Culjkovic B. Topisirovic I. Borden KL. Controlling gene expression through RNA regulons: the role of the eukaryotic translation initiation factor eIF4E. Cell Cycle. 2007;6(1):65–69. doi: 10.4161/cc.6.1.3688. [DOI] [PubMed] [Google Scholar]
  28. Culjkovic B. Topisirovic I. Skrabanek L. Ruiz-Gutierrez M. Borden KL. eIF4E promotes nuclear export of cyclin D1 mRNAs via an element in the 3′UTR. J Cell Biol. 2005;169(2):245–256. doi: 10.1083/jcb.200501019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Culjkovic B. Topisirovic I. Skrabanek L. Ruiz-Gutierrez M. Borden KL. eIF4E is a central node of an RNA regulon that governs cellular proliferation. J Cell Biol. 2006;175(3):415–426. doi: 10.1083/jcb.200607020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Culjkovic-Kraljacic B. Baguet A. Volpon L. Amri A. Borden KLB. The oncogene eIF4E reprograms the nuclear pore complext to promote mRNA export and oncogenic transformation. Cell Rep. 2012;2(2):207–215. doi: 10.1016/j.celrep.2012.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. De Benedetti A. Harris AL. eIF4E expression in tumors: its possible role in progression and malignacies. Int J Biochem Cell Biol. 1999;31:59–72. doi: 10.1016/s1357-2725(98)00132-0. [DOI] [PubMed] [Google Scholar]
  32. De Benedetti A. Joshi-Barve S. Rinker-Schaeffer C. Rhoads RE. Expression of antisense RNA against initiation factor eIF-4E mRNA in HeLa cells results in lengthened cell division times, diminished translation rates, and reduced levels of both eIF-4E and the p220 component of eIF-4F. Mol Cell Biol. 1991;11(11):5435–5445. doi: 10.1128/mcb.11.11.5435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. De Benedetti A. Rhoads RE. Overexpression of eukaryotic protein synthesis initiation factor 4E in HeLa cells results in aberrant growth and morphology. Proc Natl Acad Sci U S A. 1990;87(21):8212–8216. doi: 10.1073/pnas.87.21.8212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Descamps G. Gomez-Bougie P. Tamburini J. Green A. Bouscary D. Maiga S. Moreau P. Le Gouill S. Pellat-Deceunynck C. Amiot M. The cap-translation inhibitor 4EGI-1 induces apoptosis in multiple myeloma through Noxa induction. Br J Cancer. 2012;106(10):1660–1667. doi: 10.1038/bjc.2012.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Dostie J. Lejbkowicz F. Sonenberg N. Nuclear eukaryotic initiation factor 4E (eIF4E) colocalizes with splicing factors in speckles. J Cell Biol. 2000;148(2):239–247. doi: 10.1083/jcb.148.2.239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Ferraiuolo MA. Basak S. Dostie J. Murray EL. Schoenberg DR. Sonenberg N. A role for the eIF4E-binding protein 4E-T in P-body formation and mRNA decay. J Cell Biol. 2005;170(6):913–924. doi: 10.1083/jcb.200504039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Filipowicz W. Furuichi Y. Sierra JM. Muthukrishnan S. Shatkin AJ. Ochoa S. A protein binding the methylated 5′-terminal sequence, m7GpppN, of eukaryotic messenger RNA. Proc Natl Acad Sci U S A. 1976;73(5):1559–1563. doi: 10.1073/pnas.73.5.1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Garre E. Romero-Santacreu L. De Clercq N. Blasco-Angulo N. Sunnerhagen P. Alepuz P. Yeast mRNA cap-binding protein Cbc1/Sto1 is necessary for the rapid reprogramming of translation after hyperosmotic shock. Mol Biol Cell. 2011;23(1):137–150. doi: 10.1091/mbc.E11-05-0419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. German-Retana S. Walter J. Doublet B. Roudet-Tavert G. Nicaise V. Lecampion C. Houvenaghel MC. Robaglia C. Michon T. Le Gall O. Mutational analysis of plant cap-binding protein eIF4E reveals key amino acids involved in biochemical functions and potyvirus infection. J Virol. 2008;82(15):7601–7612. doi: 10.1128/JVI.00209-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Gingras AC. Kennedy SG. O'Leary MA. Sonenberg N. Hay N. 4E-BP1, a repressor of mRNA translation, is phosphorylated and inactivated by the Akt(PKB) signaling pathway. Genes Dev. 1998;12(4):502–513. doi: 10.1101/gad.12.4.502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Glue P. The clinical pharmacology of ribavirin. Semin Liver Dis. 1999;19(Suppl 1):17–24. [PubMed] [Google Scholar]
  42. Graff JR. De Benedetti A. Olson JW. Tamez P. Casero RA., Jr. Zimmer SG. Translation of ODC mRNA and polyamine transport are suppressed in ras-transformed CREF cells by depleting translation initiation factor 4E. Biochem Biophys Res Commun. 1997;240(1):15–20. doi: 10.1006/bbrc.1997.7592. [DOI] [PubMed] [Google Scholar]
  43. Graff JR. Konicek BW. Vincent TM. Lynch RL. Monteith D. Weir SN. Schwier P. Capen A. Goode RL. Dowless MS. Chen Y. Zhang H. Sissons S. Cox K. McNulty AM. Parsons SH. Wang T. Sams L. Geeganage S. Douglass LE. Neubauer BL. Dean NM. Blanchard K. Shou J. Stancato LF. Carter JH. Marcusson EG. Therapeutic suppression of translation initiation factor eIF4E expression reduces tumor growth without toxicity. J Clin Invest. 2007;117(9):2638–2648. doi: 10.1172/JCI32044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Gu M. Fabrega C. Liu SW. Liu H. Kiledjian M. Lima CD. Insights into the structure, mechanism, and regulation of scavenger mRNA decapping activity. Mol Cell. 2004;14(1):67–80. doi: 10.1016/s1097-2765(04)00180-7. [DOI] [PubMed] [Google Scholar]
  45. Haghighat A. Mader S. Pause A. Sonenberg N. Repression of cap-dependent translation by 4E-binding protein 1: competition with p220 for binding to eukaryotic initiation factor-4E. EMBO J. 1995;14(22):5701–5709. doi: 10.1002/j.1460-2075.1995.tb00257.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Hong DS. Kurzrock R. Oh Y. Wheler J. Naing A. Brail L. Callies S. Andre V. Kadam SK. Nasir A. Holzer TR. Meric-Bernstam F. Fishman M. Simon G. A phase 1 dose escalation, pharmacokinetic, and pharmacodynamic evaluation of eIF-4E antisense oligonucleotide LY2275796 in patients with advanced cancer. Clin Cancer Res. 2011;17(20):6582–6591. doi: 10.1158/1078-0432.CCR-11-0430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Hong Z. Cameron CE. Pleiotropic mechanisms of ribavirin antiviral activities. Prog Drug Res. 2002;59:41–69. doi: 10.1007/978-3-0348-8171-5_2. [DOI] [PubMed] [Google Scholar]
  48. Hoover DS. Wingett DG. Zhang J. Reeves R. Magnuson NS. Pim-1 protein expression is regulated by its 5′-untranslated region and translation initiation factor elF-4E. Cell Growth Differ. 1997;8(12):1371–1380. [PubMed] [Google Scholar]
  49. Iborra FJ. Jackson DA. Cook PR. Coupled transcription and translation within nuclei of mammalian cells. Science. 2001;293(5532):1139–1142. doi: 10.1126/science.1061216. [DOI] [PubMed] [Google Scholar]
  50. Joshi S. Kaur S. Redig AJ. Goldsborough K. David K. Ueda T. Watanabe-Fukunaga R. Baker DP. Fish EN. Fukunaga R. Platanias LC. Type I interferon (IFN)-dependent activation of Mnk1 and its role in the generation of growth inhibitory responses. Proc Natl Acad Sci U S A. 2009;106(29):12097–12102. doi: 10.1073/pnas.0900562106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Joshi S. Sharma B. Kaur S. Majchrzak B. Ueda T. Fukunaga R. Verma AK. Fish EN. Platanias LC. Essential role for Mnk kinases in type II interferon (IFNgamma) signaling and its suppressive effects on normal hematopoiesis. J Biol Chem. 2011;286(8):6017–6026. doi: 10.1074/jbc.M110.197921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Kaufman RJ. Murtha-Riel P. Pittman DD. Davies MV. Characterization of wild-type and Ser53 mutant eukaryotic initiation factor 4E overexpression in mammalian cells. J Biol Chem. 1993;268(16):11902–11909. [PubMed] [Google Scholar]
  53. Keene JD. RNA regulons: coordination of post-transcriptional events. Nat Rev Genet. 2007;8(7):533–543. doi: 10.1038/nrg2111. [DOI] [PubMed] [Google Scholar]
  54. Keene JD. Lager PJ. Post-transcriptional operons and regulons co-ordinating gene expression. Chromosome Res. 2005;13(3):327–337. doi: 10.1007/s10577-005-0848-1. [DOI] [PubMed] [Google Scholar]
  55. Keene JD. Tenenbaum SA. Eukaryotic mRNPs may represent posttranscriptional operons. Mol Cell. 2002;9(6):1161–1167. doi: 10.1016/s1097-2765(02)00559-2. [DOI] [PubMed] [Google Scholar]
  56. Kentsis A. Dwyer EC. Perez JM. Sharma M. Chen A. Pan ZQ. Borden KL. The RING domains of the promyelocytic leukemia protein PML and the arenaviral protein Z repress translation by directly inhibiting translation initiation factor eIF4E. J Mol Biol. 2001;312(4):609–623. doi: 10.1006/jmbi.2001.5003. [DOI] [PubMed] [Google Scholar]
  57. Kentsis A. Gordon RE. Borden KL. Control of biochemical reactions through supramolecular RING domain self-assembly. Proc Natl Acad Sci U S A. 2002a;99(24):15404–15409. doi: 10.1073/pnas.202608799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Kentsis A. Gordon RE. Borden KL. Self-assembly properties of a model RING domain. Proc Natl Acad Sci U S A. 2002b;15:15. doi: 10.1073/pnas.012317299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Kentsis A. Topisirovic I. Culjkovic B. Shao L. Borden KL. Ribavirin suppresses eIF4E-mediated oncogenic transformation by physical mimicry of the 7-methyl guanosine mRNA cap. Proc Natl Acad Sci U S A. 2004;101(52):18105–18110. doi: 10.1073/pnas.0406927102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kentsis A. Volpon L. Topisirovic I. Soll CE. Culjkovic B. Shao L. Borden KL. Further evidence that ribavirin interacts with eIF4E. RNA. 2005;11(12):1762–1766. doi: 10.1261/rna.2238705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Khanna-Gupta A. Abayasekara N. Levine M. Sun H. Virgilio M. Nia N. Halene S. Sportoletti P. Jeong JY. Pandolfi PP. Berliner N. Up-regulation of translation eukaryotic initiation factor 4E in nucleophosmin 1 haploinsufficient cells results in changes in CCAAT enhancer-binding protein alpha activity: implications in myelodysplastic syndrome and acute myeloid leukemia. J Biol Chem. 2012;287(39):32728–32737. doi: 10.1074/jbc.M112.373274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Kinkelin K. Veith K. Grunwald M. Bono F. Crystal structure of a minimal eIF4E-Cup complex reveals a general mechanism of eIF4E regulation in translational repression. RNA. 2012;18(9):1624–1634. doi: 10.1261/rna.033639.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Knauf U. Tschopp C. Gram H. Negative regulation of protein translation by mitogen-activated protein kinase-interacting kinases 1 and 2. Mol Cell Biol. 2001;21(16):5500–5511. doi: 10.1128/MCB.21.16.5500-5511.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Ko SY. Guo H. Barengo N. Naora H. Inhibition of ovarian cancer growth by a tumor-targeting peptide that binds eukaryotic translation initiation factor 4E. Clin Cancer Res. 2009;15(13):4336–4347. doi: 10.1158/1078-0432.CCR-08-2924. [DOI] [PubMed] [Google Scholar]
  65. Konicek BW. Stephens JR. McNulty AM. Robichaud N. Peery RB. Dumstorf CA. Dowless MS. Iversen PW. Parsons S. Ellis KE. McCann DJ. Pelletier J. Furic L. Yingling JM. Stancato LF. Sonenberg N. Graff JR. Therapeutic inhibition of MAP kinase interacting kinase blocks eukaryotic initiation factor 4E phosphorylation and suppresses outgrowth of experimental lung metastases. Cancer Res. 2011;71(5):1849–1857. doi: 10.1158/0008-5472.CAN-10-3298. [DOI] [PubMed] [Google Scholar]
  66. Kraljacic BC. Arguello M. Amri A. Cormack G. Borden K. Inhibition of eIF4E with ribavirin cooperates with common chemotherapies in primary acute myeloid leukemia specimens. Leukemia. 2011;25(7):1197–1200. doi: 10.1038/leu.2011.57. [DOI] [PubMed] [Google Scholar]
  67. Lazaris-Karatzas A. Montine KS. Sonenberg N. Malignant transformation by a eukaryotic initiation factor subunit that binds to mRNA 5′ cap. Nature. 1990;345(6275):544–547. doi: 10.1038/345544a0. [DOI] [PubMed] [Google Scholar]
  68. Le Bacquer O. Petroulakis E. Paglialunga S. Poulin F. Richard D. Cianflone K. Sonenberg N. Elevated sensitivity to diet-induced obesity and insulin resistance in mice lacking 4E-BP1 and 4E-BP2. J Clin Invest. 2007;117(2):387–396. doi: 10.1172/JCI29528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Lee JW. Choi JJ. Lee KM. Choi CH. Kim TJ. Lee JH. Kim BG. Ahn G. Song SY. Bae DS. eIF-4E expression is associated with histopathologic grades in cervical neoplasia. Hum Pathol. 2005;36(11):1197–1203. doi: 10.1016/j.humpath.2005.08.010. [DOI] [PubMed] [Google Scholar]
  70. Lejbkowicz F. Goyer C. Darveau A. Neron S. Lemieux R. Sonenberg N. A fraction of the mRNA 5′ cap-binding protein, eukaryotic initiation factor 4E, localizes to the nucleus. Proc Natl Acad Sci U S A. 1992;89(20):9612–9616. doi: 10.1073/pnas.89.20.9612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Li BD. Gruner JS. Abreo F. Johnson LW. Yu H. Nawas S. McDonald JC. DeBenedetti A. Prospective study of eukaryotic initiation factor 4E protein elevation and breast cancer outcome. Ann Surg. 2002;235(5):732–738. doi: 10.1097/00000658-200205000-00016. discussion 738–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Li S. Takasu T. Perlman DM. Peterson MS. Burrichter D. Avdulov S. Bitterman PB. Polunovsky VA. Translation factor eIF4E rescues cells from Myc-dependent apoptosis by inhibiting cytochrome c release. J Biol Chem. 2003;278(5):3015–3022. doi: 10.1074/jbc.M208821200. [DOI] [PubMed] [Google Scholar]
  73. Liu W. Jankowska-Anyszka M. Piecyk K. Dickson L. Wallace A. Niedzwiecka A. Stepinski J. Stolarski R. Darzynkiewicz E. Kieft J. Zhao R. Jones DN. Davis RE. Structural basis for nematode eIF4E binding an m(2,2,7)G-Cap and its implications for translation initiation. Nucleic Acids Res. 2011;39(20):8820–8832. doi: 10.1093/nar/gkr650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Lynch M. Chen L. Ravitz MJ. Mehtani S. Korenblat K. Pazin MJ. Schmidt EV. hnRNP K binds a core polypyrimidine element in the eukaryotic translation initiation factor 4E (eIF4E) promoter, and its regulation of eIF4E contributes to neoplastic transformation. Mol Cell Biol. 2005;25(15):6436–6453. doi: 10.1128/MCB.25.15.6436-6453.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Mader S. Lee H. Pause A. Sonenberg N. The translation initiation factor eIF-4E binds to a common motif shared by the translation factor eIF-4 gamma and the translational repressors 4E-binding proteins. Mol Cell Biol. 1995;15(9):4990–4997. doi: 10.1128/mcb.15.9.4990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Mainwaring LA. Kenney AM. Divergent functions for eIF4E and S6 kinase by sonic hedgehog mitogenic signaling in the developing cerebellum. Oncogene. 2011;30(15):1784–1797. doi: 10.1038/onc.2010.564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Maquat LE. Tarn WY. Isken O. The pioneer round of translation: features and functions. Cell. 2010;142(3):368–374. doi: 10.1016/j.cell.2010.07.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Marcotrigiano J. Gingras AC. Sonenberg N. Burley SK. Cocrystal structure of the messenger RNA 5′ cap-binding protein (eIF4E) bound to 7-methyl-GDP. Cell. 1997;89(6):951–961. doi: 10.1016/s0092-8674(00)80280-9. [DOI] [PubMed] [Google Scholar]
  79. Marcotrigiano J. Gingras AC. Sonenberg N. Burley SK. Cap-dependent translation initiation in eukaryotes is regulated by a molecular mimic of eIF4G. Mol Cell. 1999;3(6):707–716. doi: 10.1016/s1097-2765(01)80003-4. [DOI] [PubMed] [Google Scholar]
  80. Martin F. Barends S. Jaeger S. Schaeffer L. Prongidi-Fix L. Eriani G. Cap-assisted internal initiation of translation of histone H4. Mol Cell. 2011;41(2):197–209. doi: 10.1016/j.molcel.2010.12.019. [DOI] [PubMed] [Google Scholar]
  81. Marzec M. Liu X. Wysocka M. Rook AH. Odum N. Wasik MA. Simultaneous inhibition of mTOR-containing complex 1 (mTORC1) and MNK induces apoptosis of cutaneous T-cell lymphoma (CTCL) cells. PLoS One. 2011;6(9):e24849. doi: 10.1371/journal.pone.0024849. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  82. Matthews-Greer J. Caldito G. de Benedetti A. Herrera GA. Dominguez-Malagon H. Chanona-Vilchis J. Turbat-Herrera EA. eIF4E as a marker for cervical neoplasia. Appl Immunohistochem Mol Morphol. 2005;13(4):367–370. doi: 10.1097/01.pai.0000170625.98446.3e. [DOI] [PubMed] [Google Scholar]
  83. Mazan-Mamczarz K. Lal A. Martindale JL. Kawai T. Gorospe M. Translational repression by RNA-binding protein TIAR. Mol Cell Biol. 2006;26(7):2716–2727. doi: 10.1128/MCB.26.7.2716-2727.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Mendez R. Myers MG., Jr. White MF. Rhoads RE. Stimulation of protein synthesis, eukaryotic translation initiation factor 4E phosphorylation, and PHAS-I phosphorylation by insulin requires insulin receptor substrate 1 and phosphatidylinositol 3-kinase. Mol Cell Biol. 1996;16(6):2857–2864. doi: 10.1128/mcb.16.6.2857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Michon T. Estevez Y. Walter J. German-Retana S. Le Gall O. The potyviral virus genome-linked protein VPg forms a ternary complex with the eukaryotic initiation factors eIF4E and eIF4G and reduces eIF4E affinity for a mRNA cap analogue. FEBS J. 2006;273(6):1312–1322. doi: 10.1111/j.1742-4658.2006.05156.x. [DOI] [PubMed] [Google Scholar]
  86. Moerke NJ. Aktas H. Chen H. Cantel S. Reibarkh MY. Fahmy A. Gross JD. Degterev A. Yuan J. Chorev M. Halperin JA. Wagner G. Small-molecule inhibition of the interaction between the translation initiation factors eIF4E and eIF4G. Cell. 2007;128(2):257–267. doi: 10.1016/j.cell.2006.11.046. [DOI] [PubMed] [Google Scholar]
  87. Nathan CA. Carter P. Liu L. Li BD. Abreo F. Tudor A. Zimmer SG. De Benedetti A. Elevated expression of eIF4E and FGF-2 isoforms during vascularization of breast carcinomas. Oncogene. 1997a;15(9):1087–1094. doi: 10.1038/sj.onc.1201272. [DOI] [PubMed] [Google Scholar]
  88. Nathan CA. Liu L. Li BD. Abreo FW. Nandy I. De Benedetti A. Detection of the proto-oncogene eIF4E in surgical margins may predict recurrence in head and neck cancer. Oncogene. 1997b;15(5):579–584. doi: 10.1038/sj.onc.1201216. [DOI] [PubMed] [Google Scholar]
  89. Nedelec S. Foucher I. Brunet I. Bouillot C. Prochiantz A. Trembleau A. Emx2 homeodomain transcription factor interacts with eukaryotic translation initiation factor 4E (eIF4E) in the axons of olfactory sensory neurons. Proc Natl Acad Sci U S A. 2004;101(29):10815–10820. doi: 10.1073/pnas.0403824101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Niessing D. Blanke S. Jackle H. Bicoid associates with the 5′-cap-bound complex of caudal mRNA and represses translation. Genes Dev. 2002;16(19):2576–2582. doi: 10.1101/gad.240002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Osborne MJ. Volpon L. Kornblatt JA. Culjkovic-Kraljcic B. Baguet A. Borden KLB. eIF4E3 acts as a tumor suppressor by utilizing an atypical mode of methyl-7 guanosine cap recognition. Proc Natl Acad Sci. 2013 doi: 10.1073/pnas.1216862110. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Parker R. Sheth U. P bodies and the control of mRNA translation and degradation. Mol Cell. 2007;25(5):635–646. doi: 10.1016/j.molcel.2007.02.011. [DOI] [PubMed] [Google Scholar]
  93. Perl AE. Kasner MT. Shank D. Luger SM. Carroll M. Single-cell pharmacodynamic monitoring of S6 ribosomal protein phosphorylation in AML blasts during a clinical trial combining the mTOR inhibitor sirolimus and intensive chemotherapy. Clin Cancer Res. 2012;18(6):1716–1725. doi: 10.1158/1078-0432.CCR-11-2346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Pestova TV. Kolupaeva VG. Lomakin IB. Pilipenko EV. Shatsky IN. Agol VI. Hellen CU. Molecular mechanisms of translation initiation in eukaryotes. Proc Natl Acad Sci U S A. 2001;98(13):7029–7036. doi: 10.1073/pnas.111145798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Pettersson F. Yau C. Dobocan MC. Culjkovic-Kraljacic B. Retrouvay H. Puckett R. Flores LM. Krop IE. Rousseau C. Cocolakis E. Borden KL. Benz CC. Miller WH., Jr Ribavirin treatment effects on breast cancers overexpressing eIF4E, a biomarker with prognostic specificity for luminal B-type breast cancer. Clin Cancer Res. 2011;17(9):2874–2884. doi: 10.1158/1078-0432.CCR-10-2334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Polunovsky VA. Rosenwald IB. Tan AT. White J. Chiang L. Sonenberg N. Bitterman PB. Translational control of programmed cell death: eukaryotic translation initiation factor 4E blocks apoptosis in growth-factor-restricted fibroblasts with physiologically expressed or deregulated Myc. Mol Cell Biol. 1996;16(11):6573–6581. doi: 10.1128/mcb.16.11.6573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Ptushkina M. von der Haar T. Karim MM. Hughes JM. McCarthy JE. Repressor binding to a dorsal regulatory site traps human eIF4E in a high cap-affinity state. Embo J. 1999;18(14):4068–4075. doi: 10.1093/emboj/18.14.4068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Ptushkina M. von der Haar T. Vasilescu S. Frank R. Birkenhager R. McCarthy JE. Cooperative modulation by eIF4G of eIF4E-binding to the mRNA 5′ cap in yeast involves a site partially shared by p20. Embo J. 1998;17(16):4798–4808. doi: 10.1093/emboj/17.16.4798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Quiocho FA. Hu G. Gershon PD. Structural basis of mRNA cap recognition by proteins. Curr Opin Struct Biol. 2000;10(1):78–86. doi: 10.1016/s0959-440x(99)00053-6. [DOI] [PubMed] [Google Scholar]
  100. Rhoads RE. eIF4E: new family members, new binding partners, new roles. J Biol Chem. 2009;284(25):16711–16715. doi: 10.1074/jbc.R900002200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Rinker-Schaeffer CW. Graff JR. De Benedetti A. Zimmer SG. Rhoads RE. Decreasing the level of translation initiation factor 4E with antisense RNA causes reversal of ras-mediated transformation and tumorigenesis of cloned rat embryo fibroblasts. Int J Cancer. 1993;55(5):841–847. doi: 10.1002/ijc.2910550525. [DOI] [PubMed] [Google Scholar]
  102. Rong L. Livingstone M. Sukarieh R. Petroulakis E. Gingras AC. Crosby K. Smith B. Polakiewicz RD. Pelletier J. Ferraiuolo MA. Sonenberg N. Control of eIF4E cellular localization by eIF4E-binding proteins, 4E-BPs. RNA. 2008;14(7):1318–1327. doi: 10.1261/rna.950608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Roudet-Tavert G. Michon T. Walter J. Delaunay T. Redondo E. Le Gall O. Central domain of a potyvirus VPg is involved in the interaction with the host translation initiation factor eIF4E and the viral protein HcPro. J Gen Virol. 2007;88(Pt 3):1029–1033. doi: 10.1099/vir.0.82501-0. [DOI] [PubMed] [Google Scholar]
  104. Rousseau D. Kaspar R. Rosenwald I. Gehrke L. Sonenberg N. Translation initiation of ornithine decarboxylase and nucleocytoplasmic transport of cyclin D1 mRNA are increased in cells overexpressing eukaryotic initiation factor 4E. Proc Natl Acad Sci U S A. 1996;93(3):1065–1070. doi: 10.1073/pnas.93.3.1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Salehi Z. Mashayekhi F. Expression of the eukaryotic translation initiation factor 4E (eIF4E) and 4E-BP1 in esophageal cancer. Clin Biochem. 2006;39(4):404–409. doi: 10.1016/j.clinbiochem.2005.11.007. [DOI] [PubMed] [Google Scholar]
  106. Scheper GC. Proud CG. Does phosphorylation of the cap-binding protein eIF4E play a role in translation initiation? Eur J Biochem. 2002;269(22):5350–5359. doi: 10.1046/j.1432-1033.2002.03291.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Schmidt EV. The role of c-myc in regulation of translation initiation. Oncogene. 2004;23(18):3217–3221. doi: 10.1038/sj.onc.1207548. [DOI] [PubMed] [Google Scholar]
  108. Seki N. Takasu T. Mandai K. Nakata M. Saeki H. Heike Y. Takata I. Segawa Y. Hanafusa T. Eguchi K. Expression of eukaryotic initiation factor 4E in atypical adenomatous hyperplasia and adenocarcinoma of the human peripheral lung. Clin Cancer Res. 2002;8(10):3046–3053. [PubMed] [Google Scholar]
  109. Shih JW. Tsai TY. Chao CH. Wu Lee YH. Candidate tumor suppressor DDX3 RNA helicase specifically represses cap-dependent translation by acting as an eIF4E inhibitory protein. Oncogene. 2008;27(5):700–714. doi: 10.1038/sj.onc.1210687. [DOI] [PubMed] [Google Scholar]
  110. Shih JW. Wang WT. Tsai TY. Kuo CY. Li HK. Wu Lee YH. Critical roles of RNA helicase DDX3 and its interactions with eIF4E/PABP1 in stress granule assembly and stress response. Biochem J. 2011;441(1):119–129. doi: 10.1042/BJ20110739. [DOI] [PubMed] [Google Scholar]
  111. Sonenberg N. Gingras AC. The mRNA 5′ cap-binding protein eIF4E and control of cell growth. Curr Opin Cell Biol. 1998;10(2):268–275. doi: 10.1016/s0955-0674(98)80150-6. [DOI] [PubMed] [Google Scholar]
  112. Sorrells DL. Black DR. Meschonat C. Rhoads R. De Benedetti A. Gao M. Williams BJ. Li BD. Detection of eIF4E gene amplification in breast cancer by competitive PCR. Ann Surg Oncol. 1998;5(3):232–237. doi: 10.1007/BF02303778. [DOI] [PubMed] [Google Scholar]
  113. Sorrells DL. Ghali GE. Meschonat C. DeFatta RJ. Black D. Liu L. De Benedetti A. Nathan CO. Li BD. Competitive PCR to detect eIF4E gene amplification in head and neck cancer. Head Neck. 1999;21(1):60–65. doi: 10.1002/(sici)1097-0347(199901)21:1<60::aid-hed8>3.0.co;2-j. [DOI] [PubMed] [Google Scholar]
  114. Tait S. Dutta K. Cowburn D. Warwicker J. Doig AJ. McCarthy JE. Local control of a disorder-order transition in 4E-BP1 underpins regulation of translation via eIF4E. Proc Natl Acad Sci U S A. 2010;107(41):17627–17632. doi: 10.1073/pnas.1008242107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Tamburini J. Green AS. Bardet V. Chapuis N. Park S. Willems L. Uzunov M. Ifrah N. Dreyfus F. Lacombe C. Mayeux P. Bouscary D. Protein synthesis is resistant to rapamycin and constitutes a promising therapeutic target in acute myeloid leukemia. Blood. 2009;114(8):1618–1627. doi: 10.1182/blood-2008-10-184515. [DOI] [PubMed] [Google Scholar]
  116. Tan A. Bitterman P. Sonenberg N. Peterson M. Polunovsky V. Inhibition of Myc-dependent apoptosis by eukaryotic translation initiation factor 4E requires cyclin D1. Oncogene. 2000;19(11):1437–1447. doi: 10.1038/sj.onc.1203446. [DOI] [PubMed] [Google Scholar]
  117. Tan K. Culjkovic B. Amri A. Borden KL. Ribavirin targets eIF4E dependent Akt survival signaling. Biochem Biophys Res Commun. 2008;375(3):341–345. doi: 10.1016/j.bbrc.2008.07.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Tan NG. Ardley HC. Scott GB. Rose SA. Markham AF. Robinson PA. Human homologue of ariadne promotes the ubiquitylation of translation initiation factor 4E homologous protein, 4EHP. FEBS Lett. 2003;554(3):501–504. doi: 10.1016/s0014-5793(03)01235-3. [DOI] [PubMed] [Google Scholar]
  119. Tao Y. Farsetta DL. Nibert ML. Harrison SC. RNA synthesis in a cage—structural studies of reovirus polymerase lambda3. Cell. 2002;111(5):733–745. doi: 10.1016/s0092-8674(02)01110-8. [DOI] [PubMed] [Google Scholar]
  120. Tenenbaum SA. Carson CC. Lager PJ. Keene JD. Identifying mRNA subsets in messenger ribonucleoprotein complexes by using cDNA arrays. Proc Natl Acad Sci U S A. 2000;97(26):14085–14090. doi: 10.1073/pnas.97.26.14085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Tomoo K. Matsushita Y. Fujisaki H. Abiko F. Shen X. Taniguchi T. Miyagawa H. Kitamura K. Miura K. Ishida T. Structural basis for mRNA cap-binding regulation of eukaryotic initiation factor 4E by 4E-binding protein, studied by spectroscopic, X-ray crystal structural, and molecular dynamics simulation methods. Biochim Biophys Acta. 2005;1753(2):191–208. doi: 10.1016/j.bbapap.2005.07.023. [DOI] [PubMed] [Google Scholar]
  122. Topisirovic I. Culjkovic B. Cohen N. Perez JM. Skrabanek L. Borden KL. The proline-rich homeodomain protein, PRH, is a tissue-specific inhibitor of eIF4E-dependent cyclin D1 mRNA transport and growth. EMBO J. 2003a;22(3):689–703. doi: 10.1093/emboj/cdg069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Topisirovic I. Guzman ML. McConnell MJ. Licht JD. Culjkovic B. Neering SJ. Jordan CT. Borden KL. Aberrant eukaryotic translation initiation factor 4E-dependent mRNA transport impedes hematopoietic differentiation and contributes to leukemogenesis. Mol Cell Biol. 2003b;23(24):8992–9002. doi: 10.1128/MCB.23.24.8992-9002.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Topisirovic I. Kentsis A. Perez JM. Guzman ML. Jordan CT. Borden KL. Eukaryotic translation initiation factor 4E activity is modulated by HOXA9 at multiple levels. Mol Cell Biol. 2005;25(3):1100–1112. doi: 10.1128/MCB.25.3.1100-1112.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Topisirovic I. Ruiz-Gutierrez M. Borden KL. Phosphorylation of the eukaryotic translation initiation factor eIF4E contributes to its transformation and mRNA transport activities. Cancer Res. 2004;64(23):8639–8642. doi: 10.1158/0008-5472.CAN-04-2677. [DOI] [PubMed] [Google Scholar]
  126. Topisirovic I. Siddiqui N. Lapointe VL. Trost M. Thibault P. Bangeranye C. Pinol-Roma S. Borden KL. Molecular dissection of the eukaryotic initiation factor 4E (eIF4E) export-competent RNP. EMBO J. 2009a;28(8):1087–1098. doi: 10.1038/emboj.2009.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Topisirovic I. Siddiqui N. Orolicki S. Skrabanek LA. Tremblay M. Hoang T. Borden KL. Stability of eukaryotic translation initiation factor 4E mRNA is regulated by HuR, and this activity is dysregulated in cancer. Mol Cell Biol. 2009b;29(5):1152–1162. doi: 10.1128/MCB.01532-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Tschopp C. Knauf U. Brauchle M. Zurini M. Ramage P. Glueck D. New L. Han J. Gram H. Phosphorylation of eIF-4E on Ser 209 in response to mitogenic and inflammatory stimuli is faithfully detected by specific antibodies. Mol Cell Biol Res Commun. 2000;3(4):205–211. doi: 10.1006/mcbr.2000.0217. [DOI] [PubMed] [Google Scholar]
  129. Tsukiyama-Kohara K. Poulin F. Kohara M. DeMaria CT. Cheng A. Wu Z. Gingras AC. Katsume A. Elchebly M. Spiegelman BM. Harper ME. Tremblay ML. Sonenberg N. Adipose tissue reduction in mice lacking the translational inhibitor 4E- BP1. Nat Med. 2001;7(10):1128–1132. doi: 10.1038/nm1001-1128. [DOI] [PubMed] [Google Scholar]
  130. Ueda T. Watanabe-Fukunaga R. Fukuyama H. Nagata S. Fukunaga R. Mnk2 and Mnk1 are essential for constitutive and inducible phosphorylation of eukaryotic initiation factor 4E but not for cell growth or development. Mol Cell Biol. 2004;24(15):6539–6549. doi: 10.1128/MCB.24.15.6539-6549.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Volpon L. Osborne MJ. Capul AA. de la Torre JC. Borden KL. Structural characterization of the Z RING-eIF4E complex reveals a distinct mode of control for eIF4E. Proc Natl Acad Sci U S A. 2010;107(12):5441–5446. doi: 10.1073/pnas.0909877107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Waskiewicz AJ. Flynn A. Proud CG. Cooper JA. Mitogen-activated protein kinases activate the serine/threonine kinases Mnk1 and Mnk2. EMBO J. 1997;16(8):1909–1920. doi: 10.1093/emboj/16.8.1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Wendel HG. De Stanchina E. Fridman JS. Malina A. Ray S. Kogan S. Cordon-Cardo C. Pelletier J. Lowe SW. Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy. Nature. 2004;428(6980):332–337. doi: 10.1038/nature02369. [DOI] [PubMed] [Google Scholar]
  134. Wheater MJ. Johnson PW. Blaydes JP. The role of MNK proteins and eIF4E phosphorylation in breast cancer cell proliferation and survival. Cancer Biol Ther. 2010;10(7):728–735. doi: 10.4161/cbt.10.7.12965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Zhang M. Fu W. Prabhu S. Moore JC. Ko J. Kim JW. Druker BJ. Trapp V. Fruehauf J. Gram H. Fan HY. Ong ST. Inhibition of polysome assembly enhances imatinib activity against chronic myelogenous leukemia and overcomes imatinib resistance. Mol Cell Biol. 2008;28(20):6496–6509. doi: 10.1128/MCB.00477-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Zhu N. Gu L. Findley HW. Zhou M. Transcriptional repression of the eukaryotic initiation factor 4E gene by wild type p53. Biochem Biophys Res Commun. 2005;335(4):1272–1279. doi: 10.1016/j.bbrc.2005.08.026. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Interferon & Cytokine Research are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES