Fact-checked by Grok 2 weeks ago

Eukaryotic initiation factor

Eukaryotic initiation factors (eIFs) are a diverse family of proteins that orchestrate the initiation phase of () translation in eukaryotic cells, enabling the precise assembly of the ribosomal initiation complex at the to begin protein synthesis. This highly regulated process involves at least 12 distinct eIFs working in concert to form the 43S preinitiation complex (PIC), recruit capped to the , scan the (UTR) for the AUG , and promote subunit joining, thereby controlling at the translational level. Dysregulation of eIFs is implicated in various diseases, including cancer, where they selectively enhance the translation of oncogenic . The core eIFs can be broadly classified into those involved in ternary complex formation and PIC assembly (eIF1, eIF1A, , eIF3, and eIF5), mRNA recruitment and scanning (eIF4A, eIF4B, , eIF4G, and eIF4H), and 60S subunit joining (eIF5B). For instance, eIF2 delivers the initiator methionyl-tRNA (Met-tRNAi) to the 40S ribosomal subunit as a GTP-bound ternary complex, while the eIF4F complex—comprising eIF4E (cap-binding protein), eIF4G (scaffold), and eIF4A (RNA helicase)—binds the mRNA 5' cap and unwinds secondary structures to facilitate ribosomal loading. eIF3, the largest complex, stabilizes the 43S PIC and promotes mRNA attachment, ensuring fidelity in start codon selection. These factors are targets of signaling pathways like mTOR and PI3K/AKT, allowing cells to adapt translation to stress, growth, or pathological conditions. Beyond their canonical roles, eIFs exhibit non-translational functions, such as eIF5A in elongation and polyproline motif synthesis, and eIF6 in ribosome biogenesis, highlighting their broader impact on cellular homeostasis. In cancer contexts, overexpressed eIFs like eIF4E drive tumorigenesis by prioritizing the translation of proliferation- and survival-related proteins, positioning them as attractive therapeutic targets for inhibitors that disrupt aberrant translation without excessive toxicity. Ongoing research continues to elucidate the structural dynamics and regulatory mechanisms of eIFs, underscoring their central role in eukaryotic gene expression.

Overview

Definition and Role in Translation

Eukaryotic initiation factors (eIFs) are a diverse group of more than 12 proteins or protein complexes that facilitate the initiation phase of protein synthesis in eukaryotic cells by promoting the assembly of the , (), and initiator () at the . These factors coordinate the ordered recruitment of the ribosomal subunit to the mRNA, scanning for the appropriate initiation codon, and joining of the 60S subunit to form the functional , thereby ensuring accurate and efficient onset. In total, the eIFs comprise approximately 25 distinct subunits, reflecting the complexity of machinery. The discovery of eIFs began in the 1970s through biochemical fractionation studies on rabbit reticulocyte lysates, which allowed the isolation and characterization of components from high salt-washed ribosomes essential for translation. A key milestone was the identification of in 1973 as the GTP-dependent factor responsible for binding and delivering the initiator methionyl-tRNA to the ribosomal subunit. Further progress in the 1980s included the purification and description of the eIF4F complex, a heterotrimeric assembly critical for cap-dependent mRNA recruitment. Beyond basic assembly, eIFs play a pivotal role in maintaining the fidelity of start codon selection, modulating the efficiency of , and integrating cellular signaling pathways to control both global protein synthesis rates and selective translation of specific mRNAs under varying conditions. This regulatory capacity allows eIFs to respond to stresses, nutrients, and growth signals, thereby linking to broader control. In contrast to prokaryotes, which rely on a simpler set of three initiation factors (IF1, IF2, IF3) for direct binding to the Shine-Dalgarno sequence on mRNAs, eukaryotes require a larger repertoire of eIFs due to unique features such as 5' mRNA capping, the need for 5'-3' scanning to locate the , and adaptation to nuclear mRNA processing and export. This increased complexity enables finer control but also introduces additional regulatory checkpoints absent in bacterial systems.

Classification of eIFs

Eukaryotic initiation factors (eIFs) are designated with the prefix "eIF" followed by a numeral that reflects the approximate chronological order of their discovery during early studies of translation initiation in eukaryotic systems. For instance, eIF1 was identified as a factor promoting methionyl-tRNAi binding, while was subsequently characterized for its role in GTP-dependent initiator tRNA delivery. This numbering system was standardized in the and to unify disparate naming conventions from rabbit lysate and systems, with subcomplexes like eIF4F denoted by combining individual factor identifiers (e.g., comprising , eIF4G, and eIF4A). Letters appended to the numerals indicate subunits, such as eIF2α, β, and γ. eIFs are broadly grouped by their primary functional roles in the sequential steps of translation initiation, providing a taxonomic framework that aligns with the pathway. The preinitiation group includes eIF1, eIF1A, , eIF3, and eIF5, which assemble with the ribosomal subunit and initiator tRNA to form the 43S preinitiation complex. The mRNA-related group encompasses eIF4A, eIF4B, , eIF4F, eIF4G, and eIF4H, which facilitate mRNA cap recognition, unwinding of secondary structures, and recruitment to the . The joining group consists of eIF5B and eIF6, promoting 60S subunit association to complete formation. Auxiliary factors, such as eIF5A, support these processes indirectly, often in specialized contexts like stress responses. Several eIFs exist as multisubunit complexes with defined architectures essential to their stability and activity. functions as a heterotrimeric complex composed of α (38 kDa), β (38 kDa), and γ (52 kDa) subunits, where the γ subunit harbors the activity critical for tRNA binding. is the largest, forming a ~800 kDa assembly of 13 non-identical subunits labeled a through m, with a core octameric subcomplex (a, b, c, e, f, h, i, k) that docks to the subunit solvent side. Similarly, operates as a heterotrimeric scaffold integrating (cap-binding), eIF4G (bridging), and (helicase) subunits to coordinate mRNA loading. In addition to eIFs, non-canonical variants like eIF2A and eIF2D mediate pathways, such as non-AUG selection or IRES-driven under stress, bypassing the eIF2-GTP-Met-tRNAi ternary complex.

The Eukaryotic Translation Initiation Pathway

Formation of the 43S Preinitiation Complex

The formation of the 43S preinitiation complex () is the foundational step in , assembling the ribosomal subunit with key initiation factors and the initiator methionyl-tRNA (Met-tRNAiMet) to create a scanning-competent entity. This ~2.5 MDa ribonucleoprotein complex integrates the subunit, eIF1, eIF1A, eIF3, eIF5, and the eIF2-GTP-Met-tRNAiMet ternary complex (), enabling subsequent mRNA recruitment without premature 60S subunit joining. Assembly commences with eIF1 and eIF1A binding to the 40S subunit, inducing an open conformation at the peptidyl (P) site to accommodate the initiator tRNA. eIF1 associates near the P site on the platform, while eIF1A binds adjacent to the aminoacyl (A) site, collectively promoting flexibility in the 40S head and body domains for tRNA entry and ensuring scanning fidelity by preventing stable accommodation of non-cognate tRNAs. The TC, formed by eIF2 bound to GTP and Met-tRNAiMet, then docks onto this eIF1/eIF1A-40S intermediate, with eIF2α contacting the 40S via ribosomal proteins uS15 and uS2 in a partial P/intramolecular (P/I) orientation. GTP binding to eIF2 is essential for TC stability, as the GDP-bound form exhibits markedly reduced affinity for Met-tRNAiMet, impairing complex formation. eIF3 subsequently integrates to stabilize the nascent multi-subunit structure, with its PCI/MPN core anchoring to the 40S solvent side via interactions with uS15, eL19, and uS2, while peripheral domains extend toward the intersubunit face to scaffold the TC and prevent 60S association. This large scaffold (~800 kDa) bridges eIF2 and the 40S, enhancing overall complex integrity. eIF5 then binds, functioning as a GTPase-activating protein to stimulate eIF2-mediated GTP hydrolysis, which induces a conformational shift, releases inorganic phosphate, and partially rearranges the PIC for downstream steps while eIF1 and eIF5 monitor tRNA positioning as a fidelity checkpoint. The open P-site conformation established early persists until mRNA engagement, ensuring accurate initiator tRNA placement. A cryo-EM structure of the human 43S PIC at ~3.3 resolution (PDB: 7A09) has illuminated the precise eIF arrangements, revealing eIF3's octameric core at the 40S back, eIF2- in an open POUT state, and eIF1/eIF1A maintaining latch openness for tRNA docking, providing a molecular blueprint for assembly dynamics. This visualization underscores eIF3's role in coordinating the and 40S, with ABCE1 occasionally present to potentially aid factor recruitment in native conditions.

mRNA Recruitment and 48S Formation

The recruitment of mRNA to the 43S preinitiation complex (PIC) begins with the activation of the mRNA's 5' cap structure, where eukaryotic initiation factor 4E (eIF4E) specifically binds the 7-methylguanosine cap (m⁷GpppN), forming the core of the eIF4F complex that anchors the mRNA to the translational machinery. This binding is essential for recruiting the 43S PIC, as eIF4E alone shows low affinity for the cap, but integration into eIF4F enhances specificity and stability. Scaffolding by eIF4G, a large multifunctional protein within eIF4F, bridges the cap-bound to other components, including poly(A)-binding protein (PABP) associated with the mRNA's 3' poly(A) tail, thereby promoting mRNA circularization. This closed-loop configuration facilitates efficient recycling and reinitiation, with disruption of the eIF4G-PABP interaction leading to approximately a 16-fold reduction in translation efficiency. Additionally, eIF4A, an ATP-dependent RNA helicase in eIF4F, unwinds secondary structures in the (UTR) to allow access for the 43S . The recruitment mechanism involves eIF4G directly interacting with eIF3 on the 43S PIC, tethering the activated mRNA-eIF4F complex to the small ribosomal subunit and enabling the initiator methionyl-tRNA (Met-tRNAi) in the ternary complex to enter the mRNA channel for scanning. eIF4B further stimulates this process by enhancing eIF4A's activity up to 4-fold and promoting 43S binding to the mRNA, thereby increasing the overall rate of attachment. Formation of the 48S initiation complex occurs as the mRNA is loaded onto the 43S PIC, positioning the ternary complex at the 5' end for downstream scanning of the UTR, which typically spans 100-200 in eukaryotic mRNAs before reaching the . This intermediate structure, stabilized by eIF1 and eIF1A, ensures accurate positioning and prepares the complex for subsequent steps in initiation.

Scanning and Start Codon Recognition

Following the attachment of the 48S preinitiation complex (PIC) to the 5' cap of the mRNA, the PIC undergoes a 5'-to-3' scanning process to locate the , during which the small ribosomal subunit moves along the (UTR) in an ATP-dependent manner. This scanning is facilitated by the activity of eIF4A, which unwinds secondary structures in the mRNA to allow progression, while eIF4G and eIF4B enhance this function by stimulating and stabilizing the unwound mRNA channel. The process maintains an open conformation of the mRNA entry channel on the subunit, primarily through the action of eIF1, which prevents premature base-pairing between the mRNA and the anticodon of initiator Met-tRNAi^Met^ until a suitable is encountered. Start codon recognition occurs when the triplet in the mRNA aligns with the anticodon of Met-tRNAi^Met^ in the of the subunit, forming stable codon-anticodon base pairs that trigger a conformational shift from the open scanning state to a closed, initiation-competent state. Optimal recognition is governed by the , typically GCCRCCAUGG (where R is a ), which surrounds the AUG and enhances initiation efficiency by promoting proper positioning and stability of the PIC at the start site; mutations in key positions, such as -3 or +4 relative to the AUG, can reduce fidelity and efficiency. This base-pairing is monitored closely, with near-perfect Watson-Crick pairing at the first two positions of the AUG being essential for commitment to translation initiation. Fidelity of start codon selection is ensured through dynamic interactions involving eIF1 and eIF1A, which stabilize the open conformation during scanning and destabilize non-optimal codon-anticodon matches, thereby rejecting non- codons or AUGs in suboptimal contexts. Upon encountering a valid , eIF1 dissociates from the , allowing eIF5-mediated on to lock the closed conformation and commit to initiation; this transition is reversible for mismatches, preventing erroneous starts. In vertebrates, these mechanisms achieve greater than 95% accuracy in selecting the correct , with mismatches at the leading to rejection rates exceeding 90% for non- initiators.

60S Subunit Joining and 80S Ribosome Assembly

Following start codon recognition by the 48S preinitiation complex, the GTPase-activating protein (GAP) activity of eIF5 stimulates hydrolysis of GTP bound to eIF2, which destabilizes eIF2-GDP interactions with the 40S subunit and promotes dissociation of eIF1, eIF2, eIF3, and eIF5 from the complex. This GTP hydrolysis event, triggered post-AUG alignment, converts the 48S complex into a state competent for large subunit association. Concurrently, eIF5B bound to GTP interacts with the 48S complex and the incoming 60S subunit, facilitating precise alignment of the intersubunit bridges and positioning the initiator Met-tRNAi in the P site of the 60S subunit. eIF5B functions as the eukaryotic ortholog of bacterial IF2, employing a similar architecture to catalyze GTP-dependent of the 60S subunit onto the 48S platform. Upon , eIF5B stabilizes the Met-tRNAi in a conformation suitable for center engagement, setting up the nascent 80S for the first formation during elongation. The cycle of eIF5B, which involves ribosomal subunit-stimulated , ensures irreversible commitment to subunit joining and subsequent factor release. During this joining step, eIF1A remains associated until after 60S docking, after which eIF5B promotes its dissociation to open the A site for . Additionally, eIF6, which binds the solvent-exposed interface of free 60S subunits to prevent premature association, is displaced to allow stable assembly. The resulting initiation complex is elongation-competent, with the initiator tRNA accommodated and most initiation factors recycled for subsequent rounds of translation. This subunit joining process occurs rapidly .

Individual eIFs and Their Functions

eIF1 and eIF1A

Eukaryotic initiation factor 1 (eIF1) is a small, universally conserved protein of approximately 12 kDa encoded by the SUI1 in , belonging to the SUI1 family found across eukaryotes, , and some . Its structure, determined by NMR for the ortholog, consists of a compact globular domain spanning residues 29–113 with two α-helices flanking a five-stranded mixed β-sheet that forms an antiparallel β-barrel, while the N-terminal 28 residues remain unstructured. A conserved basic patch on the first α-helix, comprising seven residues (K56–K58, K61, K64–K66), is positioned for potential electrostatic interactions with the 18S rRNA, contributing to its ribosomal binding. eIF1 binds to the interface side of the ribosomal subunit platform, near the , where it monitors the fidelity of selection during scanning. Eukaryotic initiation factor 1A (eIF1A), approximately 16 , serves as the functional eukaryotic ortholog of bacterial IF1 and features a core consisting of an oligonucleotide/oligosaccharide-binding (OB) fold domain and a helical , extended by flexible N- and C-terminal tails that are intrinsically disordered. The NMR structure of eIF1A reveals a large RNA-mimetic surface on the OB fold, facilitating interactions with single-stranded and promoting mRNA binding and scanning. eIF1A binds to the 40S subunit at a site analogous to the bacterial A-site, occupying a cleft between ribosomal proteins S12 and the 530 of 18S rRNA 44, thereby stabilizing the preinitiation complex. Together, eIF1 and eIF1A induce a scanning-competent open conformation of the subunit by stabilizing a ~3–5 lateral shift in the platform relative to the head domain, which widens the mRNA entry channel (between helices 18 and 34) and promotes an open geometry conducive to initiator tRNA movement during scanning. This conformation facilitates rapid association of the eIF2-GTP-Met-tRNAi ternary complex (with on-rates up to 3.8 × 10^7 M^{-1} s^{-1}) while preventing stable accommodation of non-cognate codons. Both factors enhance scanning fidelity, with eIF1 particularly critical for discriminating against non-AUG start sites; in sui1, such as D88Y or G112R, destabilize eIF1 binding and permit initiation at suboptimal non-AUG codons, as demonstrated in genetic screens. eIF1 and eIF1A interact dynamically with eIF5, a GTPase-activating protein for , to ensure during recognition. Specifically, eIF1 binds eIF5 via two distinct interfaces—one in its N-terminal tail and another on a /hydrophobic surface—allowing eIF5 to mimic eIF1's position on the upon eIF1 release, thereby stabilizing the closed conformation at authentic codons and promoting subunit joining. These interactions, mapped through NMR and mutagenesis, underscore their role in modulating conformational transitions for accurate initiation.69019-6/fulltext)

eIF2

eIF2 is a heterotrimeric GTPase composed of three subunits: the α subunit (eIF2α, approximately 36 kDa), which serves a regulatory role; the β subunit (eIF2β, approximately 38 kDa), which facilitates mRNA mimicry; and the γ subunit (eIF2γ, approximately 55 kDa), which harbors the GTPase activity essential for delivering the initiator methionyl-tRNA (Met-tRNAi) to the ribosome. The α subunit is particularly notable for its conserved serine residue at position 51 (S51), a key phosphorylation site that modulates eIF2 function under stress conditions, as highlighted in recent analyses of eIF2 dynamics. In its active form, eIF2 binds GTP and Met-tRNAi to form the ternary complex (TC: eIF2-GTP-Met-tRNAi), which exhibits high affinity with a dissociation constant (Kd) of approximately 1 nM, enabling efficient recruitment of the initiator tRNA to the 40S ribosomal subunit during formation of the 43S preinitiation complex. The β subunit plays a critical role in stabilizing this complex by mimicking mRNA anticodon-codon interactions, which helps position Met-tRNAi for accurate start codon recognition. This TC delivers Met-tRNAi to the P-site of the 40S subunit, setting the stage for mRNA scanning and AUG selection. The functional cycle of eIF2 involves GTP binding in the , followed by triggered upon codon recognition in the 48S preinitiation complex, where eIF5 acts as a GTPase-activating protein () to stimulate and release inorganic (Pi). This irreversible step commits the complex to initiation, leading to eIF2-GDP dissociation from the ; subsequent reactivation occurs via GDP release, catalyzed by eIF2B, a (GEF) composed of five subunits (α, β, γ, δ, ε). eIF2B facilitates the exchange of GDP for GTP, recycling eIF2 for the next round of initiation, with its decameric structure (two copies of each subunit) enhancing catalytic efficiency. Mutations at the eIF2α S51 phosphorylation site, such as substitution to alanine (S51A), abolish stress-induced regulation, underscoring its centrality in controlling translation under adverse conditions like nutrient deprivation or viral infection. Phosphorylation at this site briefly referenced here inhibits TC formation by sequestering eIF2B, a mechanism detailed further in discussions of stress responses. Recent structural and dynamic studies emphasize how S51 phosphorylation alters eIF2-eIF2B interactions, reducing GEF activity and global translation while selectively enhancing stress-response gene expression.

eIF3

The eukaryotic initiation factor 3 (eIF3) is a large, multi-subunit complex essential for translation in eukaryotes, comprising 13 subunits designated a through m and totaling approximately 800 kDa in mass. These subunits assemble into a modular scaffold that interacts extensively with the ribosomal subunit, facilitating the organization of other initiation factors and mRNA during the formation of the 43S preinitiation complex. The core of eIF3 consists of an octameric structure formed by subunits e, f, i, k, l, and m, along with a and c in mammals, which are highly conserved across species and stabilized by ( cop9-initiator) and MPN (MPN domain protease) domains. These domains enable the intricate assembly of the complex, with PCI domains forming a horseshoe-shaped scaffold and MPN domains (in subunits f and h) providing additional stability through metalloprotease-like folds, despite lacking catalytic activity in this context. eIF3 serves multiple critical functions initiation, primarily acting as a scaffold to stabilize the binding of the eIF2 ternary complex (TC) to the 40S subunit, thereby promoting the assembly of the 43S preinitiation complex. It also facilitates mRNA recruitment by binding directly to mRNA via subunits d and j, which position the mRNA near the ribosomal entry channel and enhance interactions with the 40S surface. Additionally, eIF3 prevents premature of the 60S ribosomal subunit with the 40S by occupying key interface sites, such as the mRNA decoding groove through eIF3j, ensuring fidelity in selection before subunit joining. This anti-association activity is particularly vital during scanning, where eIF3 maintains an open conformation of the 40S mRNA channel. The of eIF3 reveal significant conformational flexibility, allowing it to adapt during different stages of . Cryo-electron (cryo-EM) studies have elucidated these features, including a 2015 reconstruction of the 40S-eIF1-eIF1A-eIF3-eIF3j at approximately 6 Å resolution (PDB: 3JAP), which shows eIF3 encircling the 40S subunit from both solvent-exposed and interface sides. This structure highlights the modular arrangement, with the eIF3 "head" and "arms" projecting to contact multiple sites on the 40S, enabling coordinated recruitment of factors like eIF2-TC while accommodating mRNA threading. Such flexibility is evident in the pivoting of eIF3 domains, which shift between open and closed states to support scanning and codon recognition without dissociating from the . Dysregulation of eIF3, particularly overexpression of subunit b (eIF3b), has been implicated in various cancers, where it correlates with enhanced of oncogenic proteins, though detailed mechanisms are explored in clinical contexts. Overall, eIF3's role underscores its indispensability, as depletion of subunits disrupts multiple steps and impairs global protein synthesis.

eIF4 Group

The eIF4 group comprises several eukaryotic initiation factors essential for cap-dependent mRNA recruitment during translation initiation, primarily through the formation of the eIF4F complex. This complex facilitates the recognition of the 5' m7G cap structure on mRNAs and unwinds secondary structures in the (UTR) to enable ribosomal scanning. Key components include , a 24 kDa cap-binding protein that specifically recognizes the m7GpppN cap with a (Kd) of approximately 1 μM, thereby anchoring the mRNA to the initiation machinery. eIF4G serves as a large 160 kDa that bridges multiple interactions, binding to eIF4E via its N-terminal domain, to eIF4A through central domains, and to eIF3 and poly(A)-binding protein (PABP) via its C-terminal region, thus integrating mRNA ends and ribosomal components. Complementing these, eIF4A is a 46 kDa DEAD-box that hydrolyzes to unwind RNA duplexes, while eIF4B (approximately 50 kDa) and eIF4H (approximately 25 kDa) act as accessory stimulators, enhancing eIF4A's helicase efficiency by promoting RNA binding and activity without intrinsic helicase function themselves. The eIF4F complex assembles as a heterotrimeric core of , eIF4G, and eIF4A, where eIF4G's modular domains orchestrate the stable trimer formation critical for mRNA engagement. This assembly occurs post-cap recognition, with binding the cap first, followed by recruitment of eIF4G and eIF4A, enabling the complex to load onto the mRNA 5' end. The activity of eIF4A, stimulated within eIF4F, drives ATP-dependent unwinding of UTR secondary structures, countering the thermodynamic stability of RNA duplexes (typically ΔG ≈ -5 to -7 kcal/mol per ) to generate single-stranded templates for the 43S preinitiation complex. eIF4B and eIF4H further augment this process by binding and facilitating eIF4A's repetitive cycling on structured substrates, increasing unwinding rates by up to 10-fold . These functions collectively promote efficient mRNA recruitment to the , as briefly referenced in the broader pathway of 48S complex formation. Regulation of the eIF4 group occurs primarily through isoforms of eIF4E-binding proteins (4E-BPs), which competitively inhibit eIF4E-eIF4G interaction in their hypophosphorylated state, preventing eIF4F assembly and suppressing cap-dependent under nutrient-limiting conditions. Mammalian cells express three 4E-BP isoforms (4E-BP1, -2, and -3), with 4E-BP1 being the most abundant; by releases this inhibition, allowing eIF4F formation. eIF4G itself has two major isoforms (eIF4GI and eIF4GII) with overlapping but distinct binding affinities, while eIF4A exists as paralogs eIF4AI and eIF4AII, both capable of eIF4F integration but with subtle differences in efficiency. Recent advances as of have focused on targeting for therapeutic , particularly in dysregulated contexts, through fragment-based screening that identifies novel allosteric binding sites on to disrupt eIF4G association without affecting cap binding. These efforts include small-molecule inhibitors like second-generation cap analogues that selectively block activity in cancer cells, building on structural insights from cryo-EM of eIF4F-mRNA complexes. Such developments highlight eIF4E's , with preclinical studies demonstrating reduced tumor progression via eIF4F inhibition.

eIF5, eIF5A, and eIF5B

eIF5 is a monomeric protein of approximately 49 kDa that serves as the for during . Its N-terminal domain harbors the GAP activity, which stimulates on bound to the initiator tRNA in the 48S preinitiation complex, promoting phosphate (Pi) release upon recognition. This GTPase activation commits the to at the AUG codon. Mutations in eIF5, such as G31R in , can enhance its GAP function and reduce the fidelity of selection, leading to increased at non-AUG codons. eIF5A, a small protein of about 17 kDa, undergoes a unique called hypusination on a conserved residue, converting it to hypusine, which is essential for its function. Although primarily involved elongation and termination, eIF5A acts as an auxiliary factor by facilitating ribosomal progression through difficult sequences, such as polyproline stretches, thereby supporting overall translational efficiency. Hypusination enables eIF5A to bind the and alleviate stalling during formation , while also promoting peptidyl-tRNA at stop codons during termination. eIF5B is a large of approximately 140 kDa in humans, homologous to the bacterial IF2, and plays a central role in the final step of by catalyzing the docking of the 60S ribosomal subunit to the 48S complex. It binds GTP and stabilizes the interaction between the subunits in the intersubunit cleft of the emerging , with its G domain contacting the 60S subunit's GTPase-associated center. GTP hydrolysis by eIF5B, triggered upon subunit joining, drives a conformational change that releases eIF5B and associated factors like eIF1A, yielding an elongation-competent with Met-tRNA positioned in the . Cryo-EM structures reveal eIF5B's domain rearrangements during this process, including domain IV stabilizing the initiator tRNA. eIF5 interacts directly with eIF2 through its C-terminal domain binding to the β subunit of eIF2, forming a stable interface that positions the GAP domain near eIF2's γ subunit for efficient activation. This eIF5-eIF2 interaction also contributes to nucleotide dissociation inhibitor (GDI) activity, stabilizing the eIF2-GDP post-hydrolysis complex until recycling by eIF2B.

eIF6

Eukaryotic initiation factor 6 (eIF6), also known as p27BBP, is a conserved monomeric protein of approximately 25 kDa that plays a critical role in and translation initiation by acting as an anti-association factor for the 60S ribosomal subunit. Structurally, eIF6 adopts a compact fold resembling the SBDS domain of Shwachman-Bodian-Diamond syndrome protein, with a core composed of an N-terminal extension and a pentameric repeat that enables tight binding to the intersubunit face of the 60S subunit. This binding occurs primarily through interactions with ribosomal proteins uL14 and eL24, as well as proximity to the sarcin-ricin loop (helix 69) of the 28S rRNA, effectively occluding the interface to prevent premature association with the subunit. Following its function in the , free eIF6 undergoes nuclear export mediated by the CRM1 export receptor, facilitating its recycling for ongoing ribosomal maturation processes. The primary function of eIF6 is to maintain the integrity of the mature 60S subunit by sterically hindering its docking with the subunit prior to the completion of initiation, thereby preventing non-productive or aberrant ribosome assembly. This anti-association activity ensures that 60S subunits remain available only after proper 48S preinitiation complex formation and start codon recognition, with eIF6 displacement occurring during the subunit joining step to allow ribosome assembly. Cryo-EM structures from 2015 reveal how eIF6 occupies the region of the 60S subunit, blocking key intersubunit bridges (such as B2a and B3) and demonstrating its role in stabilizing the subunit against untimely joining. Regulation of eIF6 involves post-translational modifications that coordinate its release and shuttling. at serine residues S174 and S175, primarily by , promotes eIF6 dissociation from the 60S subunit and facilitates its nuclear re-import for reuse, although these sites may not be strictly essential in all contexts. Additionally, eIF6 is integral to 60S biogenesis, where it associates with pre-60S particles in the to support rRNA processing and subunit maturation before their nuclear export as part of the CRM1-dependent pathway. This dual localization—nucleolar for biogenesis and cytoplasmic for anti-association—highlights eIF6's essential role in linking production with translational fidelity.

Regulation of Translation Initiation

eIF2 Phosphorylation and Stress Responses

Phosphorylation of the α-subunit of eukaryotic initiation factor 2 (eIF2α) at serine 51 (Ser51) serves as a critical regulatory mechanism to attenuate global protein synthesis during cellular stress, thereby conserving resources and promoting adaptation. This modification is catalyzed by four specialized stress-activated kinases: protein kinase R-like endoplasmic reticulum kinase (PERK), general control nonderepressible 2 (GCN2), protein kinase R (PKR), and heme-regulated inhibitor (HRI). Each kinase responds to distinct environmental challenges, integrating signals through the integrated stress response (ISR) pathway. Upon activation, these kinases undergo autophosphorylation or transphosphorylation, enabling them to target the conserved Ser51 residue within the S1 domain of eIF2α. The stress-specific activation of these kinases links eIF2α phosphorylation to diverse physiological threats. PERK is primarily triggered by endoplasmic reticulum (ER) stress, where accumulation of unfolded proteins causes dissociation of the chaperone BiP/GRP78 from PERK's luminal domain, leading to its oligomerization and autophosphorylation at Thr980. GCN2 senses starvation through binding of uncharged tRNAs to its regulatory domain, inducing a conformational change that activates its kinase activity. PKR detects infections via double-stranded RNA binding to its dsRNA-binding motifs, resulting in dimerization and autophosphorylation at Thr446, while it can also respond to oxidative or ER stress through interaction with . HRI, in contrast, is activated by heme depletion in erythroid cells or , involving relief of heme-mediated autoinhibition and dimer formation. These pathways ensure that eIF2α phosphorylation is a versatile response to limitation, proteotoxic stress, invasion, and metabolic perturbations. Mechanistically, converts eIF2 from a to a potent competitive of its , eIF2B. In its unphosphorylated state, eIF2-GDP is recycled to eIF2-GTP by eIF2B, enabling formation of the (TC) with initiator methionyl-tRNA (Met-tRNAi) essential for translation initiation. However, phosphorylated eIF2α (eIF2α~P) binds eIF2B with higher affinity, sequestering it and drastically reducing TC availability. Since eIF2B is present in limiting amounts relative to eIF2 (approximately 10-fold lower), even modest levels of eIF2α phosphorylation (5-10%) can inhibit eIF2B activity by over 90%, corresponding to a 10-fold or greater reduction in formation and global translation rates. This attenuation suppresses cap-dependent mRNA translation, prioritizing during . Despite the broad suppression of translation, eIF2α phosphorylation paradoxically enables selective translation of specific mRNAs that harbor upstream open reading frames (uORFs) in their 5' untranslated regions. Under low TC conditions, ribosomes more efficiently bypass inhibitory uORFs, allowing reinitiation at the downstream main ORF. This mechanism preferentially translates transcription factors such as activating transcription factor 4 (ATF4), which upregulates genes involved in antioxidant defense, amino acid metabolism, and autophagy. Similarly, growth arrest and DNA damage-inducible 34 (GADD34) is selectively expressed, forming a feedback loop by recruiting protein phosphatase 1 (PP1) to dephosphorylate eIF2α. These adaptations enhance cellular resilience without fully halting protein synthesis. The reversal of eIF2α phosphorylation is mediated by the PP1-GADD34 phosphatase complex, which specifically targets phospho-Ser51 to restore eIF2B activity and resume as subsides. GADD34 acts as a regulatory subunit, PP1 to eIF2α via distinct motifs and enhancing its catalytic efficiency toward this . This prevents prolonged translational repression, allowing cells to recover. Studies as of 2024 have shown that in neuronal cells under integrated response, eIF2B forms distinct eIF2B bodies that modulate dynamics for long-term survival.

Other Regulatory Mechanisms Involving eIFs

The signaling pathway plays a central role in regulating initiation by controlling the activity of eukaryotic initiation factor 4E ()-binding proteins (4E-BPs), which sequester and prevent its interaction with eIF4G to form the eIF4F complex. Under nutrient-rich conditions, 4E-BPs at multiple sites, leading to their dissociation from and thereby promoting cap-dependent of mRNAs with structured 5' untranslated regions. Inhibition of by rapamycin disrupts this process, preventing 4E-BP and blocking eIF4F assembly, which selectively suppresses of proliferation-related transcripts. Viral infection provides another layer of regulation through proteolytic cleavage of eIF4G by viral proteases, which disrupts host cap-dependent while favoring viral protein synthesis. For instance, 2A protease directly cleaves eIF4G at a specific site between residues 681 and 682, separating the eIF4E-binding domain from the eIF3- and PABP-interacting regions, thereby inhibiting canonical initiation and redirecting to internal ribosome entry sites (IRES) on viral mRNA. This mechanism exemplifies how pathogens exploit eIFs to shut off cellular , with similar cleavage observed in other picornaviruses. Modulation of eIF3 activity further fine-tunes initiation through post-translational modifications, including ubiquitination of specific subunits that promotes complex disassembly and turnover. The eIF3f subunit, for example, undergoes ubiquitination and proteasomal mediated by the E3 ligase atrogin-1/MAFbx, reducing eIF3 stability and impairing 43S pre-initiation complex formation during or conditions. Additionally, microRNAs (miRNAs) indirectly regulate eIF3 function by targeting expression; miR-34c-3p binds the 3' untranslated region of eIF4E mRNA, suppressing its translation and thereby limiting eIF4F availability for eIF3-mediated mRNA recruitment. In hypoxic environments, such as those in solid tumors, hypoxia-inducible factor-1α (HIF-1α) transcriptionally upregulates expression via binding to hypoxia-responsive elements in its promoter, enhancing cap-dependent translation of pro-angiogenic mRNAs like (VEGF) to support tumor vascularization. As of 2025, clinical inhibitors of eIF4A, such as zotatifin, have shown promise in reducing tumor burden in models by repressing translation of oncogenic mRNAs.

Structural Insights

Cryo-EM and Crystal Structures of eIF Complexes

Advances in cryo-electron microscopy (cryo-EM) and since the have provided high-resolution insights into the architecture of eukaryotic initiation factor (eIF) complexes, revealing their interactions with the ribosomal subunit and mRNA during translation initiation. These structures have elucidated the modular of preinitiation complexes, such as the 43S and 48S, highlighting the scaffold roles of multi-subunit factors like eIF3 and the bridging functions of eIF4F. Seminal cryo-EM reconstructions have achieved resolutions below 4 Å, enabling atomic model building and visualization of dynamic interfaces. A landmark cryo-EM structure of the 43S preinitiation complex, resolved at approximately 3.5 Å, captured the subunit associated with eIF1, eIF1A, , eIF3, eIF5, and initiator tRNA, demonstrating how eIF3 encircles the solvent side of the to stabilize the assembly. This 2021 study on native ABCE1-bound 43S complexes from extended these findings, identifying multiple conformational states at resolutions of 3.2–4.0 Å that illustrate eIF positioning during early recruitment. In humans, a 2024 cryo-EM reconstruction of the 48S complex at 3.1 Å resolution revealed a critical bridge between eIF4F and the 43S via eIF3 subunits, positioning the mRNA cap proximal to the for scanning. This structure, published in , showed two distinct eIF4A helicases: one within eIF4F and a second recruited to the mRNA entry channel, facilitating unwinding. Crystal structures of eIF4F components have complemented these assemblies, with a 1.53 Å resolution X-ray structure of the human eIF4E-eIF4G complex (PDB: 5T46) illustrating the extended binding interface that enhances cap affinity and recruits the helicase eIF4A. Recent cryo-EM studies have further delineated eIF4A conformational cycles within scanning complexes, capturing ATP-dependent states that resolve mRNA secondary structures at the 40S mRNA channel. For multi-factor interactions, a 2007 cryo-EM reconstruction of the yeast 40S-eIF1-eIF1A complex at ~20 Å resolution first depicted an open 40S conformation primed for mRNA binding, later refined in higher-resolution models. A 2015 cryo-EM structure of the yeast 40S-eIF1-eIF1A-eIF3-eIF3j complex at 6 Å resolution positioned eIF3's PCI/MPN core along the 40S interface, underscoring its role in factor docking. Recent advances include 2024 cryo-EM structures of human scanning intermediates at 3.0–3.5 Å resolution, which reveal dynamic eIF2-eIF5 interfaces during codon recognition, with eIF5's GAP domain modulating GTP hydrolysis on eIF2 to commit the complex at the start codon. These maps, from reconstituted 48S assemblies, show transient contacts that prevent premature eIF2 release, providing a structural basis for fidelity in initiation. Such progress has transformed understanding of eIF complex modularity, with ongoing refinements expected to capture additional transient states.

Key Conformational Changes During Initiation

During the formation of the 43S preinitiation complex (), eIF1 and eIF1A induce an open conformation of the ribosomal subunit, characterized by a of the head domain by approximately 8–10° relative to the body, which widens the mRNA-binding channel and positions the initiator tRNA (Met-tRNAiMet) in a partially accommodated Pout state displaced about 10 Å from the . This openness facilitates the docking of the ternary complex (; eIF2-GTP-Met-tRNAiMet) and subsequent attachment of the -bound mRNA, closing the upon stable codon-anticodon pairing. During scanning of the (UTR), eIF4A, stimulated by eIF4G and eIF4B, acts as an RNA helicase to unwind secondary structures in a stepwise manner, advancing the by 1–3 per cycle while eIF3 pivots to guide mRNA entry into the decoding channel, ensuring processive movement toward the . Upon recognition of the AUG start codon, the undergoes a commitment step involving of eIF1, which shifts approximately 5 away from the to allow closure of the head domain and accommodation of Met-tRNAiMet into the Pin state. This conformational transition is triggered by eIF5-mediated on , stabilizing the codon-anticodon interaction and arresting scanning. Concurrently, eIF5B- binds to the , stabilizing key intersubunit bridges such as B2a and B3 to align the and incoming 60S subunits, promoting their without premature locking. The of eIF1 and partial reconfiguration of ensure fidelity in start-site selection by preventing non-AUG initiation. In the transition to the 80S ribosome, GTP hydrolysis by eIF5B drives a ~15° rotation of the 60S body relative to the 40S subunit, locking the initiation complex and facilitating the release of eIFs including eIF1A, eIF2-GDP, and eIF5 on a millisecond timescale (~100 ms for eIF1A ejection). This rapid kinetics ensures efficient handover of Met-tRNAiMet from eIF2 to the peptidyl transferase center, completing assembly of the elongation-competent 80S ribosome. Structures from recent studies illustrate these bridges in pre-80S states. Recent cryo-EM studies of human 48S complexes have revealed a two-step model for release: following GTP at the , -GDP initially remains dynamically bound, allowing eIF5B to engage and transfer Met-tRNAiMet before complete dissociation of and eIF5 in subsequent intermediates (48S-4 and 48S-5). This mechanism coordinates factor exchange, preventing premature 60S joining and enhancing translational accuracy.

Clinical and Therapeutic Relevance

eIFs in Cancer and Disease

Dysregulation of eukaryotic initiation factors (eIFs) plays a pivotal role in oncogenesis and various pathologies by altering translation of key regulatory proteins. Overexpression of eIF4E is frequently observed in multiple human cancers, including breast and prostate malignancies, where it enhances cap-dependent translation of oncogenic mRNAs such as cyclin D1 and those utilizing internal ribosome entry sites (IRES), thereby promoting cell proliferation and tumor progression. In prostate cancer, eIF4E levels are elevated in up to 78% of cases, correlating with advanced disease stages and resistance to therapy. Similarly, in breast cancer, eIF4E amplification and overexpression are linked to increased gene copy number and poorer patient outcomes, underscoring its oncogenic potential. Amplifications and overexpression of eIF3 subunits, particularly eIF3b and eIF3c, are implicated in development and progression. eIF3c on 8q, a region commonly altered in gastrointestinal tumors, leads to elevated eIF3c levels that enhance global translation and support malignant growth, with high expression serving as an independent prognostic marker for poor survival in patients. eIF3b overexpression similarly drives in colon cancer cells, and its downregulation inhibits tumor cell growth, highlighting its role in aggressive disease phenotypes. These alterations correlate with advanced tumor stages and reduced overall survival, emphasizing eIF3's contribution to colorectal oncogenesis. Chronic of eIF2α, often mediated by the GCN2, is associated with neurodegenerative disorders such as (ALS). In ALS models, persistent GCN2 activation leads to sustained eIF2α , which attenuates global protein synthesis while selectively translating stress-response factors, contributing to degeneration and disease progression. This dysregulation disrupts and exacerbates neuronal vulnerability, with GCN2 inhibition shown to reduce toxic and delay ALS onset in mutant SOD1 mouse models. Elevated eIF4A activity is prominent in -driven lymphomas, where upregulates eIF4A to boost of short, structured mRNAs essential for rapid and survival. Hypusination of eIF5A, a required for its function, is upregulated in fibrotic conditions, promoting collagen and deposition in cardiac and . Inhibiting eIF5A hypusination reduces activation and severity in preclinical models. Additionally, viruses such as (HCV) hijack machinery, with HCV core protein binding eIF4G to favor viral IRES-mediated over cap-dependent protein , facilitating persistent and associated liver .

Targeting eIFs for Therapeutic Intervention

Targeting eukaryotic initiation factors (eIFs) has emerged as a promising strategy for anticancer and antiviral therapies due to their central role in dysregulated protein synthesis in malignancies and . Inhibitors and modulators of specific eIFs have advanced through preclinical and clinical stages, focusing on disrupting cap-dependent to selectively impair tumor or viral growth while minimizing effects on normal cells. Tomivosertib, a selective inhibitor of interacting kinases (MNK1/2), blocks of at serine 209, thereby reducing the translation of oncogenic mRNAs such as those encoding and VEGF. This compound has demonstrated preclinical efficacy in inhibiting and tumor growth in models by decreasing phosphorylated levels. As of 2025, tomivosertib development has been terminated in frontline non-small cell lung cancer following Phase II failure, but it continues in Phase Ib evaluation for other solid tumors, including combinations with chemotherapies like in . For eIF4A, rocaglamides represent a class of natural product-derived inhibitors that bind to the helicase and stall its RNA unwinding activity, preferentially suppressing translation of structured mRNAs critical for cancer cell survival. In preclinical models of acute myeloid leukemia (AML), rocaglamides, including analogs like rocaglamide, have shown potent antiproliferative effects, particularly in cells with RUNX1 mutations or FLT3-ITD alterations, by inactivating heat shock proteins and disrupting bioenergetic homeostasis. Silvestrol, a related flavagline eIF4A inhibitor, has exhibited broad anticancer activity in preclinical studies across hematologic and solid tumors; efforts to optimize its pharmacokinetics have led to analogs entering early clinical development, with silvestrol itself explored in Phase I trials for refractory malignancies before progression to more stable derivatives like zotatifin, now in Phase I/II for advanced solid tumors. Modulators of eIF2 pathways offer therapeutic potential in neurodegenerative diseases linked to chronic integrated stress response (ISR) activation. ISRIB, a small-molecule activator of eIF2B, counteracts PERK-mediated eIF2α by stabilizing the eIF2B complex and restoring global protein synthesis without directly dephosphorylating eIF2α. In models, ISRIB has improved cognitive function and reduced neuronal toxicity by mitigating ISR-induced translational repression. As of 2025, ISRIB derivatives like DNL343 have been evaluated in Phase 2/3 clinical trials for , where it failed to meet the primary endpoint of functional decline, though it demonstrated penetration and ISR modulation in preclinical and early clinical data. Approaches targeting eIF3 and eIF6 subunits primarily involve (ASOs) for selective knockdown, exploiting their overexpression in cancers to induce and inhibit . For instance, ASOs against eIF3b have reduced tumor growth in , , and models by impairing and promoting , positioning eIF3 as a viable target for nucleic acid-based therapies. Similarly, knockdown of eIF3d via lentiviral delivery or ASOs has suppressed colon cancer cell in preclinical settings. Broad-spectrum antivirals targeting eIF4G, a key scaffolding factor in the eIF4F complex, disrupt host-virus interactions by blocking eIF4E-eIF4G binding or viral cleavage of eIF4G, showing efficacy against enteroviruses and other viruses in cell-based assays. Despite these advances, challenges in eIF-targeted therapies include achieving selectivity to avoid disrupting normal , managing off-target toxicity, and overcoming resistance mechanisms in heterogeneous tumors. A 2025 review highlights the potential of combining eIF inhibitors, such as tomivosertib, with inhibitors like to enhance antitumor immunity by modulating the , as evidenced by ongoing trials showing improved response rates in non-small cell and other solid tumors.

References

  1. [1]
    The Mechanism of Eukaryotic Translation Initiation: New Insights ...
    Translation initiation in eukaryotes is a highly regulated and complex stage of gene expression. It requires the action of at least 12 initiation factors.
  2. [2]
    Eukaryotic translation initiation factors as promising targets in cancer ...
    Nov 4, 2020 · eIFs are fundamental for the translation of mRNA and as such act as the primary targets of several signaling pathways to regulate gene expression.
  3. [3]
    Role of Eukaryotic Initiation Factors during Cellular Stress and ...
    Translation initiation is a highly regulated and rate-limiting step of protein synthesis that requires more than 12 eukaryotic initiation factors (eIFs).Missing: definition history
  4. [4]
    THE MECHANISM OF EUKARYOTIC TRANSLATION INITIATION ...
    It requires at least 9 eukaryotic initiation factors (eIFs; Table 1) and comprises two steps: first, formation of 48S initiation complexes with established ...
  5. [5]
    To initiate or not to initiate: A critical assessment of eIF2A, eIF2D ...
    Mar 3, 2024 · eIF2 is the canonical initiator tRNA (Met-tRNAiMet) carrier in eukaryotes. Other reported initiation factors, namely eIF2A, eIF2D, and the MCT-1 ...
  6. [6]
  7. [7]
    Structure of the mammalian ribosomal 43S preinitiation complex ...
    Eukaryotic translation initiation begins with assembly of a 43S preinitiation complex. First, Met-tRNAiMet, eukaryotic initiation factor 2 (eIF2) and GTP ...
  8. [8]
    Initiation Factor eIF2γ Promotes eIF2–GTP–Met-tRNAiMet Ternary ...
    To calculate dissociation constants for TC formation, 43S or 48S complex formation, or GDP binding, binding curves for Met-tRNAiMet and GDP were fit using ...
  9. [9]
    A multifactor complex of eukaryotic initiation factors, eIF1, eIF2, eIF3 ...
    Translation initiation factor 2 (eIF2) bound to GTP transfers the initiator methionyl tRNA to the 40S ribosomal subunit. The eIF5 stimulates GTP hydrolysis by ...
  10. [10]
    A structural inventory of native ribosomal ABCE1‐43S pre‐initiation ...
    In eukaryotes, the start of translation initiation requires the assembly of the 43S pre‐initiation complex (PIC). It consists of the 40S subunit, eIF3, eIF1, ...<|separator|>
  11. [11]
    Molecular mechanisms of translation initiation in eukaryotes - PNAS
    Jun 19, 2001 · Translation initiation is a complex process in which initiator tRNA, 40S, and 60S ribosomal subunits are assembled by eukaryotic initiation factors (eIFs) into ...
  12. [12]
  13. [13]
    Mammalian poly(A)-binding protein is a eukaryotic translation ... - NIH
    The eIF4G/PABP interaction, which brings about the circularization of the ... The ability of Krebs-2 extract to translate poly(A)+ mRNA decreased ∼16-fold as a ...
  14. [14]
    Untranslated regions of mRNAs | Genome Biology | Full Text
    Feb 28, 2002 · The average length of 5' UTRs is roughly constant over diverse taxonomic classes and ranges between 100 and 200 nucleotides, whereas the average ...
  15. [15]
    The Scanning Mechanism of Eukaryotic Translation Initiation
    Jun 2, 2014 · In eukaryotes, the scanning mechanism identifies the initiation codon by inspecting each triplet for complementarity to Met-tRNAi's anticodon.
  16. [16]
    Regulatory elements in eIF1A control the fidelity of start codon ...
    Regulatory elements in eIF1A control the fidelity of start codon selection by modulating tRNAiMet binding to the ribosome.
  17. [17]
    Eukaryotic translation initiation factor eIF5 promotes the accuracy of ...
    Eukaryotic translation initiation factor eIF5 promotes the accuracy of start codon recognition by regulating Pi release and conformational transitions of the ...
  18. [18]
    Full article: Why is start codon selection so precise in eukaryotes?
    The mechanistic basis for high accuracy in eukaryotic initiation is described based on recent findings concerning the role of the multifactor complex (MFC) in ...
  19. [19]
  20. [20]
    Initiation factor eIF5B catalyzes second GTP-dependent step ... - PNAS
    eIF5B, the eukaryotic ortholog of IF2, is a GTPase that promotes ribosomal subunit joining. Here we show that eIF5B GTPase activity is required for protein ...
  21. [21]
    Molecular cloning and functional expression of a human cDNA ...
    Eukaryotic translation initiation factor 6 (eIF6) binds to the 60S ribosomal subunit and prevents its association with the 40S ribosomal subunit.
  22. [22]
    Structure and interactions of the translation initiation factor eIF1
    eIF1 is a universally conserved translation factor that is necessary for scanning and involved in initiation site selection. We have determined the solution ...
  23. [23]
    The eIF1A Solution Structure Reveals a Large RNA-Binding Surface ...
    The activity of eIF1A in translation initiation is pleiotropic, stabilizing binding of Met–tRNA to 40S ribosomal subunits, promoting mRNA binding, and binding ...Article · Results · Eif1a Binds Single-Stranded...
  24. [24]
    The Eukaryotic Translation Initiation Factors eIF1 and eIF1A Induce ...
    A crystal structure of IF1 bound to the prokaryotic small (30S) ribosomal subunit showed that IF1 binds in the A site, blocking initiator tRNA from binding in ...
  25. [25]
  26. [26]
  27. [27]
    Exploring the interaction dynamics of eukaryotic translation initiation ...
    May 23, 2025 · The focus of this review is eukaryotic translation initiation factor 2 (eIF2), and due to space considerations, we will limit our discussion ...
  28. [28]
    Fail-safe control of translation initiation by dissociation of eIF2α ...
    Mar 18, 2017 · We show that eIF2B can compete with Met-tRNAi for eIF2-GTP and can destabilize TC. When TC is formed with unphosphorylated eIF2, eIF5 can out- ...
  29. [29]
    A multifactor complex of eukaryotic initiation factors, eIF1, eIF2, eIF3 ...
    Translation initiation factor 2 (eIF2) bound to GTP transfers the initiator methionyl tRNA to the 40S ribosomal subunit. The eIF5 stimulates GTP hydrolysis by ...Missing: composition | Show results with:composition
  30. [30]
    eIF2B is a decameric guanine nucleotide exchange factor with a γ 2 ...
    May 23, 2014 · eIF2B then acts as a guanine nucleotide exchange factor (GEF) catalyzing exchange ... GEF function is accomplished by the γ- and ε-subunits ( ...
  31. [31]
    Embraced by eIF3: structural and functional insights into the roles of ...
    eIF3 thus embraces the small ribosomal subunit from both sides to control most, if not all, initiation reactions. In detail, the head and left arm represented ...
  32. [32]
  33. [33]
    Cap‐Dependent Translation Initiation Factor eIF4E - PubMed Central
    eIF4E, a 25 kDa polypeptide, is an evolutionarily conserved protein.12 A growing body of experimental evidence has placed deregulated translation on the causal ...
  34. [34]
    Translation Initiation Factor eIF4G-1 Binds to eIF3 through the ... - NIH
    eIF4G forms specific complexes with eIF4E, eIF4A, and PABP, thereby linking the processes of cap recognition, poly(A) binding, and secondary structure melting.
  35. [35]
    The DEAD-box helicase eIF4A: Paradigm or the odd one out? - PMC
    eIF4A is the founding member of the DEAD-box protein family, and one of the few examples of DEAD-box proteins that consist of a helicase core only.
  36. [36]
    Eukaryotic initiation factor 4B is a multi-functional RNA binding ...
    Sep 3, 2024 · eIF4B stimulates the helicase activity of eIF4A, thereby promoting the unwinding of RNA structure within the 5′ untranslated regions of mRNAs.
  37. [37]
    The structure of a human translation initiation complex reveals two ...
    Jan 29, 2024 · Here we report the structure of a human 48S that shows that, in addition to the eIF4A that is part of eIF4F, there is a second eIF4A helicase bound at the mRNA ...Missing: 1980s | Show results with:1980s
  38. [38]
    Structural basis for the enhancement of eIF4A helicase activity by ...
    The eukaryotic translation initiation factors 4A (eIF4A) and 4G (eIF4G) are crucial for the assembly of the translationally active ribosome. Together with eIF4E ...
  39. [39]
    Synergistic activation of eIF4A by eIF4B and eIF4G - PMC - NIH
    Both mammalian and yeast eIF4G contain binding sites for the poly(A)-binding protein (PABP), eIF4A and eIF4E, while only mammalian eIF4G contains binding sites ...Missing: weight | Show results with:weight<|separator|>
  40. [40]
    Hierarchical phosphorylation of the translation inhibitor 4E-BP1
    The eIF4E-binding proteins (4E-BPs) interdict translation initiation by binding to the translation factor eIF4E, and preventing recruitment of the translation ...
  41. [41]
    Control of the eIF4E activity: structural insights and pharmacological ...
    A well-characterized group of proteins, named 4E-binding proteins (4E-BPs), inhibits the eIF4E–eIF4G interaction by competing for the same binding site on the ...
  42. [42]
    Advances in understanding and targeting eIF4E activity
    eIF4G acts as scaffold protein for recruitment of eIF4A RNA helicase which unwinds RNA secondary structure and promotes the binding of the 43 S pre-initiation ...
  43. [43]
    Integrating fragment-based screening with targeted protein ... - Nature
    Nov 20, 2024 · Here we screen a fragment library against eIF4E and identify a ligand-binding site with previously unknown function.
  44. [44]
    Second-Generation Cap Analogue Prodrugs for Targeting Aberrant ...
    Dec 12, 2024 · Advances in understanding and targeting eIF4E activity. Biochemical Society Transactions 2025, 53 (04) , 801-822. https://doi.org/10.1042 ...
  45. [45]
    Mutational Analysis of Mammalian Translation Initiation Factor 5 (eIF5)
    Eukaryotic initiation factor 5 (eIF5), a monomeric protein of 49 kDa in mammals (9, 10, 21) and 46 kDa in the yeast Saccharomyces cerevisiae (5, 6), plays an ...
  46. [46]
    Structure of the eukaryotic initiation factor (eIF) 5 reveals a ... - PubMed
    eIF5 plays multiple roles in translation initiation. Its N-terminal domain functions as a GTPase-activator protein (GAP) for GTP bound to eIF2.
  47. [47]
    Eukaryotic translation initiation factor 5 (eIF5) acts as a classical ...
    Moreover, these findings demonstrate that eIF5 acts as a GAP for the hydrolysis of GTP bound to eIF2 at the start codon of eukaryotic mRNAs. This is consistent ...
  48. [48]
    Hypusinated and unhypusinated isoforms of the translation factor ...
    Hypusination of eukaryotic translation initiation factor 5A (eIF5A) is essential for its role in translation elongation and termination.
  49. [49]
    eIF5A Functions Globally in Translation Elongation and Termination
    We conclude that eIF5A functions broadly in elongation and termination, rationalizing its high cellular abundance and essential nature.
  50. [50]
    eIF5A Functions Globally in Translation Elongation and Termination
    eIF5A functions globally in translation elongation and termination, alleviating stalling on many motifs, and stimulating peptidyl-tRNA hydrolysis.<|separator|>
  51. [51]
    EIF5B Gene - GeneCards | IF2P Protein | IF2P Antibody
    Protein attributes for EIF5B Gene. Size: 1220 amino acids; Molecular mass: 138827 Da ... kDa, though endogeneous hIF2 migrates anomalously on SDS/PAGE at 180 kDa.
  52. [52]
    Position of eukaryotic initiation factor eIF5B on the 80S ribosome ...
    eIF5B is located in the intersubunit cleft of the 80S ribosome: domain 1 is positioned near the GTPase activating center of the 60S subunit.
  53. [53]
    Structural basis for the transition from translation initiation to ... - Nature
    Oct 6, 2020 · All the mis-acylated tRNAs are active in 48S PIC and 80S formation (Fig. 4). However, the 60S subunit joining rate is altered in a way that ...
  54. [54]
    Specific interaction of eukaryotic translation initiation factor 5 (eIF5 ...
    Dec 12, 1997 · We observed that eIF5 specifically interacted with the beta subunit of initiation factor eIF2. No other initiation factors including the gamma subunit of eIF2 ...Missing: interface | Show results with:interface
  55. [55]
    Full article: eIF5 - Taylor & Francis Online
    The crystal structure of the carboxy-terminal domain of human translation initiation factor eIF5. J Mol Biol 2006; 360:457 - 465. (Open in a new window) ...<|control11|><|separator|>
  56. [56]
    Function and ribosomal localization of aIF6, a translational regulator ...
    Nov 26, 2008 · One of these is the small monomeric protein (about 25 kDa) called eIF6 in eukarya and aIF6 in archaea. In eukaryotes, eIF6 was identified ...Missing: CRM1 | Show results with:CRM1
  57. [57]
    Mechanism of eIF6-mediated Inhibition of Ribosomal Subunit Joining
    ... eIF6 prevents binding of the 40 S ribosomal subunit to the 60 S subunit. When the 40 S subunit in its 80 S-bound conformation is superimposed with the 60 S-eIF6 ...
  58. [58]
    Dynamic states of eIF6 and SDS variants modulate interactions with ...
    Structural studies show that eIF6 directly associates with uL14 (RPL23) and is proximal to the Sarcin-Ricin Loop (SRL) of the 28S rRNA and RPL24 (eL24) of 60S ( ...
  59. [59]
    Identification of CRM1-dependent Nuclear Export Cargos Using ...
    Here we present an approach for identifying targets of the CRM1-export pathway via quantitative mass spectrometry using stable isotope labeling with amino ...
  60. [60]
    Mechanism of eIF6 release from the nascent 60S ribosomal subunit
    Oct 19, 2015 · Upon release, eIF6 shuttles back to the nucleus, where it plays an essential part in the biogenesis and nuclear export of the 60S ribosomal ...Missing: monomer kDa CRM1
  61. [61]
    Uncoupling of GTP hydrolysis from eIF6 release on the ribosome ...
    Although we identified eIF6 phosphopeptides containing phosphates at residues S174, S175, T165, and S166 and unambiguously identified multiple tryptic ...
  62. [62]
    Eukaryotic initiation factor 6 mediates a continuum between 60S ...
    Apr 17, 2009 · In the nucleolus, eIF6 is a component of the pre-ribosomal particles and is required for the biogenesis of 60S subunits, whereas in the ...Missing: S174 S175
  63. [63]
    The eIF2α kinases: their structures and functions - PMC
    Phosphorylation of eIF2α on serine 51 results in a severe decline in de novo protein synthesis and is an important strategy in the cell's armory against ...The Eif2α Kinases · Eif2 And The Regulation Of... · Eif2α Regulation And Of The...
  64. [64]
    Integrated stress response of vertebrates is regulated by four eIF2α ...
    Sep 16, 2016 · PKR-like endoplasmic reticulum (ER) kinase (PERK/EIF2AK3) is activated during ER stress by the release of binding immunoglobulin protein from ...
  65. [65]
    REVIEW ARTICLE Heme-regulated eIF2α kinase in erythropoiesis ...
    Nov 14, 2019 · HRI is activated during heme deficiency to phosphorylate eIF2α (eIF2αP), which simultaneously inhibits the translation of globin messenger RNAs ...
  66. [66]
    The structural basis of translational control by eIF2 phosphorylation
    May 13, 2019 · As phosphorylated eIF2 has ten-fold higher affinity for eIF2B than unphosphorylated eIF2 (Kd, 3.5 vs. 32.2 nM), we first focused on this complex ...Missing: Kd | Show results with:Kd
  67. [67]
    Selective mRNA translation during eIF2 phosphorylation induces ...
    Mar 19, 2014 · ATF4 and ATF5 mRNAs, both of which are preferentially translated during ER stress, shifted 55 and 51% toward large polysomes, respectively ( ...
  68. [68]
    Structural and functional analysis of the GADD34:PP1 eIF2α ...
    As the stress declines, the GADD34 protein, by recruiting the ser/thr protein phosphatase 1 (PP1), facilitates the dephosphorylation of phospho-eIF2α, restoring ...
  69. [69]
    An eIF2α-binding motif in protein phosphatase 1 subunit GADD34 ...
    As indicated by its alternate name, GADD34 is a protein phosphatase 1 (PP1) targeting protein that directs PP1 to dephosphorylate eIF2α (7–11). Consistent with ...
  70. [70]
    Regulation of eIF-4E BP1 phosphorylation by mTOR - PubMed - NIH
    Oct 17, 1997 · We conclude that mTOR is an upstream regulator of eIF-4E BP1 as well as the p70 S6 kinase; moreover, these two mTOR targets are regulated in a parallel rather ...
  71. [71]
    The mTOR Pathway in the Control of Protein Synthesis | Physiology
    These proteins bind to the same region of eIF4E as eIF4G does, so binding of 4E-BPs to eIF4E prevents eIF4E from binding eIF4G and engaging in active ...
  72. [72]
    Poliovirus 2A proteinase cleaves directly the eIF-4G ... - PubMed - NIH
    Sep 11, 1998 · We report here that a recombinant form of poliovirus 2Apro fused to maltose binding protein (MBP) directly cleaves human eIF-4G from a highly purified eIF-4F ...
  73. [73]
    Cleavage of Eukaryotic Translation Initiation Factor 4G by ...
    Cleavage of initiation factor eIF4G by the poliovirus protease 2Apro has been proposed as the cause of this shutoff phenomenon (15). In fact, addition of ...
  74. [74]
    eIF3: a factor for human health and disease - PMC
    The eukaryotic initiation factor 3 (eIF3) is one of the most complex translation initiation factors in mammalian cells, consisting of several subunits (eIF3a to ...
  75. [75]
    miR‐34c‐3p functions as a tumour suppressor by inhibiting eIF4E ...
    In this study, we found that miR‐34c‐3p directly targeted and inhibited expression of eIF4E and played a significant role in regulating proliferation, migration ...
  76. [76]
    Hypoxia-inducible Factor-1α (HIF-1α) Promotes Cap-dependent ...
    Our study suggests that HIF-1α promotes cap-dependent translation of selective mRNAs through up-regulating eIF4E1, which contributes to tumorsphere growth of ...
  77. [77]
    Potential therapeutic targets of eukaryotic translation initiation ...
    Jul 5, 2025 · In this review, we summarize the roles of individual eIFs in cap-dependent translation and discuss their potential as therapeutic targets in cancer treatment.
  78. [78]
    5T46: Crystal structure of the human eIF4E-eIF4G complex
    Oct 26, 2016 · Here, we describe crystal structures of human and Drosophila melanogaster eIF4E-eIF4G complexes, which unexpectedly reveal that the eIF4G auxiliary sequences ...Missing: 2008 2W25
  79. [79]
    Structural basis for translational control by the human 48S initiation ...
    Sep 17, 2024 · This complex binds the 5′ end of the mRNA with the aid of eIF4F, forming the 48S initiation complex. The 48S complex then scans along the mRNA ...
  80. [80]
    Factor-dependent processivity in human eIF4A DEAD-box helicase
    Our findings support a memory-less stepwise mechanism for translation initiation and suggest that similar factor-dependent processivity may be shared by ...
  81. [81]
    Translational initiation factor eIF5 replaces eIF1 on the 40S ... - eLife
    Nov 26, 2018 · Recognition of an AUG start codon induces a major conformational change in the PIC to a scanning-arrested closed (PIN) complex made possible by ...
  82. [82]
    Translational initiation factor eIF5 replaces eIF1 on the 40S ...
    Thus, the structure of a eukaryotic translation initiation complex containing eIF5 would greatly aid our understanding of its multiple functions. The recent ...
  83. [83]
    Reconfiguration of yeast 40S ribosomal subunit domains by ... - PNAS
    The cryo-EM reconstruction of the 43S complex, at 20-Å resolution, indicates that major conformational changes occur in the 40S subunit on binding of the MFC ( ...<|separator|>
  84. [84]
    [PDF] eIF5B and eIF1A reorient initiator tRNA to allow ribosomal subunit ...
    Formation of the 80S ribosome triggers hydrolysis of GTP by eIF5B to accelerate ejection of. eIF1A by helix 69 of the 60S subunit (within ~100 ms), likely by ...
  85. [85]
    Targeting Eukaryotic Translation Initiation Factor 4E (eIF4E) in Cancer
    Clinical relevance of eIF4E in human cancers. eIF4E overexpression is common in multiple cancer types, including malignancies of the prostate, breast ...
  86. [86]
    eIF4E phosphorylation promotes tumorigenesis and is ... - PNAS
    We show that knock-in mice expressing a nonphosphorylatable form of eIF4E are resistant to tumorigenesis in a prostate cancer model.Results · Eif4e-Ki Mefs Are Resistant... · Eif4e-Ki Mice Are Resistant...
  87. [87]
    eIF4E Phosphorylation in Prostate Cancer - ScienceDirect.com
    It appears that eIF4E phosphorylation enhances the rate of translation of oncogene mRNAs to increase tumorigenicity.The Mtor Pathway · Increased Eif4e And Its... · Eif4e Targeted Therapies
  88. [88]
    Biological functions and research progress of eIF4E - Frontiers
    Overexpression of eIF4E in breast cancer samples was associated with increased gene copy number, which was not detected in benign breast tissue (67). EIF4E ...EIF4E and metabolic... · Main factors affecting eIF4E... · Targeted therapy of eIF4E
  89. [89]
    Silencing of translation initiation factor eIF3b promotes apoptosis in ...
    Several studies have shown that silencing of eIF3b inhibited the proliferation of different types of human cancers including bladder, prostate, and colon, as ...
  90. [90]
    Full article: eIF3: a factor for human health and disease
    Genome-wide analyses of colorectal cancers also identified amplification of the eIF3h gene. The oncogenic function of eIF3h was in fact confirmed ...Introduction · Eif3 Constitution · Cancer
  91. [91]
    GCN2 inhibition reduces mutant SOD1 clustering and toxicity ... - NIH
    Sep 20, 2024 · GCN2 inhibition reduces mutant SOD1 clustering and toxicity and delays disease progression in an amyotrophic lateral sclerosis mouse model.Missing: eIF2 | Show results with:eIF2
  92. [92]
    The Integrated Stress Response and Phosphorylated Eukaryotic ...
    This review examines evidence that dysregulated eIF2a phosphorylation acts as a driver of neurodegeneration.
  93. [93]
    Targeting Translation Initiation by Synthetic Rocaglates for Treating ...
    Jun 6, 2019 · In MYC-driven lymphomas, protein synthesis is enhanced to sustain rapid proliferation through MYC-mediated upregulation of eIF4E, eIF4A, and ...
  94. [94]
    Small-molecule modulators of B56-PP2A restore 4E-BP function to ...
    Jan 27, 2025 · 4E-BPs bind to eIF4E to block its interaction with eIF4G, thereby inhibiting eIF4F assembly. Three 4E-BP isoforms exist in mammalian cells, with ...
  95. [95]
    Ciclopirox Inhibition of eIF5A Hypusination Attenuates Fibroblast ...
    Jan 29, 2023 · In this study, we found that increased hypusinated eIF5A protein levels were associated with cardiac fibrosis and heart dysfunction in myocardial infarction ( ...
  96. [96]
    Ciclopirox Inhibition of eIF5A Hypusination Attenuates Fibroblast ...
    Jan 29, 2023 · In this study, we found that increased hypusinated eIF5A protein levels were associated with cardiac fibrosis and heart dysfunction in myocardial infarction ( ...
  97. [97]
    Regulation of host cell translation by viruses and effects on cell ...
    Infection of cells with picornaviruses causes dephosphorylation of 4E-BP resulting in the sequestration of eIF4E from eIF4G. Two hepatitis C viral proteins, ...
  98. [98]
    Protein translation: biological processes and therapeutic strategies ...
    Feb 23, 2024 · In particular, targeting eIFs has emerged as a promising strategy for therapeutic interventions against cancer, as supported by current studies.
  99. [99]
    Inhibition of eukaryotic initiation factor 4E by tomivosertib ... - PubMed
    Of note, tomivosertib inhibited glioblastoma angiogenesis and decreased p-eIF4E level in mice. We finally showed that p-eIF4E activation was a common molecular ...Missing: trials 2024
  100. [100]
    Targeting protein synthesis pathways in MYC-amplified ...
    Jan 8, 2025 · A recent study showed that decreased eIF4A1 levels suppress lymphomagenesis in murine MYC-driven lymphoma [58], suggesting that eIF4A1 is a ...Targeting Mtor Signaling... · 5.3 Targeting Ampk · 8 Future Perspective And...<|control11|><|separator|>
  101. [101]
    MNK, mTOR or eIF4E-selecting the best anti-tumor target for ...
    Aug 6, 2025 · 2024; Shuo et al. 2023) . Tomivosertib, BAY1143269, and ETC-206, have advanced to clinical trials for solid tumors and lymphomas. ...
  102. [102]
    A second-generation eIF4A RNA helicase inhibitor exploits ... - NIH
    Jan 17, 2024 · We report on MG-002, a rocaglate that shows superior properties relative to existing eIF4A inhibitors in pre-clinical models. Our study also ...Missing: preclinical | Show results with:preclinical
  103. [103]
    Effective therapy for AML with RUNX1 mutation by cotreatment with ...
    Feb 10, 2022 · ... inhibitors, including omacetaxine (HHT) and the eIF4A inhibitor rocaglamide. ... preclinical models of acute myeloid leukemia. Blood., 123 ...
  104. [104]
    EIF4A inhibition targets bioenergetic homeostasis in AML MOLM-14 ...
    Dec 9, 2022 · Our results reveal that eIF4A inhibition has a profound effect on the bioenergetic capacity of MOLM-14 AML cells and indicates that eIF4A ...
  105. [105]
    Investigational eIF2B activator DNL343 modulates the integrated ...
    Aug 18, 2025 · The original eIF2B activator, ISRIB, was shown to stabilize the eIF2B complex through a direct interaction and stimulate its GEF activity ...
  106. [106]
    Targeting the Unfolded Protein Response as a Disease-Modifying ...
    The small molecule ISRIB (integrated stress response inhibitor B) acts downstream of phosphorylated eIF2α, preventing its interaction with its target eIF2B, and ...
  107. [107]
    Small molecule ISRIB suppresses the integrated stress response ...
    Jan 23, 2019 · ISRIB enhances cognitive memory processes and has therapeutic effects in brain-injured mice without displaying overt side effects. While using ...Small Molecule Isrib... · Sign Up For Pnas Alerts · Results
  108. [108]
    Translation initiation factor eIF3b expression in human cancer and ...
    Amplification of EIF3S3 gene is associated with advanced stage in prostate cancer. ... (EIF3B) Gene Expression Inhibits Proliferation of Colon Cancer Cells. World ...
  109. [109]
    Silencing of translation initiation factor eIF3b promotes apoptosis in ...
    Mar 1, 2017 · Recent studies have shown that the expression of the eIF3b subunit is elevated in bladder and prostate cancer, and eIF3b silencing inhibited ...
  110. [110]
    Lentivirus-mediated knockdown of eukaryotic translation initiation ...
    eIF3D is essential for colon cancer cell growth, and knockdown of eIF3D resulted in a significant reduction in cell proliferation probably due to.
  111. [111]
    Discovery of RNA‐Targeting Small Molecules: Challenges and ...
    Aug 24, 2025 · Challenges remain in terms of compound selectivity, toxicity, and translating in vitro findings to clinical applications, but the integration ...
  112. [112]
    Emerging clinical and research approaches in targeted therapies for ...
    Challenges such as off-target effects, the need for more selective inhibitors, potential toxicity, and the complexity of targeting translation initiation in ...<|separator|>
  113. [113]
    A New Way to Unleash the Immune System against Cancer - NCI
    Feb 6, 2019 · One ongoing clinical trial is evaluating the combination of tomivosertib and immune checkpoint inhibitors in people with nonblood cancers. In ...<|separator|>