EIF4E Human

Eukaryotic Translation Initiation Factor 4E Human Recombinant
Shipped with Ice Packs
In Stock

Description

2.1. mRNA Translation Initiation

eIF4E recruits ribosomes to capped mRNAs by binding the m⁷G cap, facilitating the assembly of the eIF4F complex. This process is rate-limiting for translation and preferentially enhances "weak" mRNAs with structured 5' untranslated regions (UTRs), such as oncogenes . Key mechanisms include:

  • Helicase activation: eIF4E stimulates eIF4A helicase activity, enabling unwinding of mRNA secondary structures .

  • Dynamic regulation: eIF4E availability is controlled by 4E-BPs (eIF4E-binding proteins), which sequester eIF4E in non-phosphorylated states .

2.2. Nuclear Roles

Beyond cytoplasmic translation, eIF4E regulates nuclear RNA export and processing:

  • mRNA export: Binds the 4ESE element in 3' UTRs, facilitating CRM1/XPO1-dependent nuclear export .

  • RNA capping and splicing: Modulates m⁷G capping and alternative splicing of select transcripts .

Table 2: Functional Roles of eIF4E

FunctionMechanismImpact on Cellular ProcessesReference
Cap-dependent translationeIF4F complex assembly, ribosome recruitmentCell proliferation, survival
Helicase stimulationeIF4A activation via eIF4G autoinhibitionUnwinding of structured 5' UTRs
Nuclear RNA export4ESE element binding, CRM1/XPO1 interactionOncogene expression, viral response

3.1. Phosphorylation

Serine 209 phosphorylation (regulated by MAPK/ERK and PI3K/mTOR pathways) enhances cap-binding affinity and is linked to cell cycle progression (high in G₁/S phase, low in G₀/M phase) .

3.2. Interaction with 4E-BPs

4E-BPs (e.g., 4E-BP1, 4E-BP3) compete with eIF4G for eIF4E binding, suppressing cap-dependent translation. Phosphorylation of 4E-BPs (e.g., by mTORC1) releases eIF4E, promoting oncogenic mRNA translation .

Table 3: Regulatory Mechanisms of eIF4E

MechanismEffectorOutcomeReference
PhosphorylationSer209 modificationIncreased cap affinity, oncogenesis
4E-BP interactionCompetitive inhibitionTranslational suppression
eIF4G autoinhibitioneIF4E binding to eIF4GHelicase activation, mRNA unwinding

4.1. Cancer Biology

  • Overexpression: Elevated eIF4E levels (>2.5-fold) drive malignant transformation by upregulating oncoproteins (e.g., cyclin D1, c-Myc) .

  • Therapeutic targeting: Small-molecule inhibitors (e.g., 4EGI-1) disrupt eIF4E-eIF4G interactions, showing efficacy in leukemia and myeloma models .

4.2. Neurological Disorders

Dysregulated eIF4E activity is implicated in autism spectrum disorders and fragile X syndrome via excessive translation of synaptic proteins .

Table 4: Key Research Insights

Study FocusFindingsClinical RelevanceReference
eIF4E helicase roleeIF4E-eIF4G interaction enhances eIF4ATarget for structured mRNA cancers
Nuclear mRNA exportLRPPRC mediates 4ESE RNA exportAML progression marker
Inhibitor development4EGI-1 disrupts eIF4E/eIF4G bindingAnticancer therapy validation

Product Specs

Introduction
EIF4E, a member of the eukaryotic initiation factor 4 family, plays a crucial role in regulating the translation of maternal mRNAs during early embryonic development before zygotic transcription begins. This factor specifically recognizes and binds to the 7-methylguanosine (m7G) cap structure present at the 5' end of eukaryotic mRNAs. This interaction is essential for initiating translation as it facilitates the recruitment of ribosomes to the mRNA and aids in unwinding its secondary structures, allowing for efficient translation.
Description
Recombinant human EIF4E, expressed in E. coli, is a purified protein with a molecular weight of 27.2 kDa. It consists of a single polypeptide chain of 237 amino acids (residues 1-217), excluding any glycosylation modifications. A 20-amino acid Histidine tag is fused to the N-terminus to facilitate purification, which is achieved using proprietary chromatographic methods.
Physical Appearance
The product is a clear, colorless solution that has been sterilized through filtration.
Formulation
The EIF4E protein is supplied at a concentration of 0.5 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0) and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage to maintain protein stability. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the EIF4E protein is greater than 85%, as determined by SDS-PAGE analysis.
Synonyms
CBP, EIF4E1, EIF4EL1, EIF4F.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MATVEPETTP TPNPPTTEEE KTESNQEVAN PEHYIKHPLQ NRWALWFFKN DKSKTWQANL RLISKFDTVE
DFWALYNHIQ LSSNLMPGCD YSLFKDGIEP MWEDEKNKRG GRWLITLNKQ QRRSDLDRFW LETLLCLIGE SFDDYSDDVC GAVVNVRAKG
DKIAIWTTEC ENREAVTHIG RVYKERLGLP PKIVIGYQSH ADTATKSGST TKNRFVV.

Q&A

What is the molecular structure and function of human eIF4E?

Human eIF4E is the cap-binding component of the eukaryotic initiation factor 4F (eIF4F) complex that recognizes the 5' end of cellular mRNAs during translation initiation. The protein possesses a distinct architecture with dorsal and ventral surfaces that serve different functions . The ventral surface contains the cap-binding slot that interacts with the 7-methylguanosine cap structure on mRNAs, while the dorsal surface forms binding sites for protein partners including eIF4G and 4E-binding proteins (4E-BPs) . These interactions are critical for translation regulation, as the competition between eIF4G and 4E-BPs for the dorsal binding site determines whether eIF4E participates in active translation initiation complexes or remains sequestered in inactive complexes . Structurally, a conserved "allostery tract" of amino acids stretches from the dorsal binding site through the center of eIF4E to the cap-binding slot, enabling communication between these two functional regions .

How is human eIF4E activity regulated in normal cells?

In normal cells, eIF4E activity is tightly regulated through several mechanisms to ensure proper control of protein synthesis. The primary regulatory pathway involves 4E-binding proteins (4E-BPs), which function as competitive inhibitors of the eIF4E-eIF4G interaction . These 4E-BPs contain a conserved binding motif that mimics the eIF4E-binding region of eIF4G, allowing them to compete for the same dorsal binding site on eIF4E . The 4E-BPs' activity is controlled through phosphorylation events coordinated by the PI3K-AKT-mTOR signaling pathway . When mTOR complex 1 (mTORC1) is activated, it phosphorylates 4E-BPs, causing them to release eIF4E, which can then bind to eIF4G and participate in translation initiation . Different 4E-BPs (4E-BP1 and 4E-BP2) have distinct binding characteristics determined by three amino acid identities within their eIF4E-binding motifs, with 4E-BP2 showing approximately 3-fold higher affinity for eIF4E compared to 4E-BP1 (Kd values of 3×10^-9 and 10^-8, respectively) . These differences suggest that the 4E-BPs may be subject to distinct modulatory pathways during cellular processes such as myeloid cell differentiation .

Why is eIF4E considered a "rate-limiting factor" in translation initiation?

eIF4E is generally considered the rate-limiting factor in translation initiation due to its restricted availability relative to other translation factors and its critical role in recruiting mRNAs to ribosomes . This limitation makes eIF4E availability a key control point for regulating global and selective protein synthesis. Several factors contribute to this rate-limiting nature:

  • Competitive inhibition: The interaction between eIF4E and eIF4G (required for translation initiation) is regulated by competitive binding of 4E-BPs, which sequester eIF4E from active translation complexes .

  • Signal integration: eIF4E availability integrates signals from multiple cellular pathways, particularly the PI3K-AKT-mTOR axis, making it a convergence point for translational regulation in response to various stimuli .

  • Selective enhancement: Limited eIF4E particularly affects mRNAs with highly structured 5'-UTRs that depend heavily on the eIF4F complex for efficient translation, explaining why increased eIF4E levels selectively enhance translation of certain mRNAs involved in proliferation and cell survival .

  • Dual functionality: Beyond cap-binding, eIF4E stimulates eIF4A helicase activity, which is crucial for unwinding structured regions in mRNAs. This dual role makes eIF4E availability critical for both recruitment and restructuring of complex mRNAs .

The restriction of eIF4E availability thus serves as a checkpoint that ensures translation resources are appropriately allocated, particularly for complex mRNAs involved in growth and proliferation processes .

How does eIF4E stimulate eIF4A helicase activity independently of its cap-binding function?

eIF4E stimulates eIF4A helicase activity through a mechanism that is distinct from and independent of its cap-binding function . This unexpected second function was revealed through real-time fluorescence assays using highly purified initiation factors . The mechanism involves:

These findings provide a molecular explanation for how eIF4E abundance selectively enhances the translation of mRNAs with structured 5'-UTRs, as these mRNAs particularly benefit from increased helicase activity to unwind their complex structures prior to ribosome scanning .

What is the molecular basis for heterotropic cooperativity between eIF4E's cap-binding and protein-binding functions?

Heterotropic cooperativity between eIF4E's cap-binding and protein-binding functions represents a sophisticated regulatory mechanism that enhances translational control. The molecular basis for this cooperativity includes:

  • The allostery tract: A conserved tract of amino acids stretches from the dorsal binding site (where proteins bind) through the center of eIF4E to the ventral cap-binding slot . This tract contains fifteen amino acids that are fully conserved across 15 different eIF4E protein sequences, suggesting its fundamental importance in eIF4E function . This structural feature provides a physical connection that enables communication between the two binding sites.

  • Enhanced cap affinity: Binding of 4E-BPs to the dorsal site on eIF4E induces an allosteric shift that increases cap affinity, effectively trapping eIF4E in inactive complexes that still maintain high affinity for capped mRNA . This explains why 4E-BP-bound eIF4E can still bind to capped mRNAs but cannot participate in translation initiation.

  • Conformational changes: The binding of proteins to the dorsal surface of eIF4E likely induces conformational changes that are transmitted through the allostery tract to the cap-binding slot, altering its structure and affinity for the cap . This conformational coupling ensures that the cap-binding and protein-binding functions of eIF4E are coordinately regulated.

  • Evolutionary conservation: The mechanism of heterotropic cooperativity appears to be evolutionarily conserved, as it was initially described for yeast eIF4E with yeast eIF4G and later confirmed for human eIF4E with various protein ligands . This conservation suggests that the allosteric coupling between the two sites is a fundamental aspect of eIF4E function across species.

This cooperativity mechanism helps explain how binding events at one site on eIF4E can influence functions at the other site, providing a sophisticated means of regulating translation initiation through allosteric effects .

What are the distinct binding characteristics of 4E-BP1 and 4E-BP2, and how do they influence eIF4E function?

The 4E-binding proteins 4E-BP1 and 4E-BP2 display distinct binding characteristics when interacting with human eIF4E, despite sharing a conserved eIF4E-binding motif. These differences have important implications for eIF4E regulation:

Characteristic4E-BP14E-BP2Functional Significance
Binding affinity (Kd)10^-8 M3×10^-9 M4E-BP2 binds approximately 3-fold more tightly
Amino acid motif differencesContains MECContains LDRAffects binding specificity and strength
Interaction with eIF4E V69WeakerStrongerContributes to differential binding profiles
Charge characteristicsLess positively chargedMore positively chargedMay enhance 4E-BP2 interaction with acidic residues in eIF4E

The binding differences between 4E-BP1 and 4E-BP2 are determined primarily by three amino acids within an otherwise conserved 16-amino acid motif . The mutation of MEC in 4E-BP1 to LDR in 4E-BP2 adds an additional positive charge (R) that may contribute to the increased binding affinity of 4E-BP2 for eIF4E . Additionally, 4E-BP2, like eIF4G, has an L at the first position in this submotif, potentially explaining why both proteins have stronger interactions with V69 in the eIF4E dorsal binding site compared to 4E-BP1 .

These distinct binding characteristics suggest that 4E-BP1 and 4E-BP2 may have different functions in translational regulation, potentially responding to different cellular signaling pathways. This is supported by evidence that 4E-BP1 and 4E-BP2 activities may be subject to distinct modulatory pathways during human myeloid cell differentiation .

What are the most effective methods to measure eIF4E-eIF4A interactions and helicase activity?

Several sophisticated methods have been developed to measure eIF4E-eIF4A interactions and helicase activity in research settings. The most effective approaches include:

  • Real-time fluorescence unwinding assays: This methodology uses a modified fluorescence unwinding assay with an uncapped RNA duplex substrate to measure the kinetics of RNA strand separation in real-time . The assay design involves:

    • A fluorescently labeled RNA strand annealed to a complementary strand

    • Measurement of fluorescence increase as strands separate

    • The ability to test various combinations of initiation factors

    • Calculation of unwinding rate constants under different conditions

    This assay provided the critical evidence that eIF4E stimulates eIF4A helicase activity independently of cap binding .

  • Tethered-RNA translation assays: This approach bypasses the need for cap-binding by directly tethering the RNA to the translation machinery, allowing researchers to isolate and study specific functions of eIF4E beyond cap recognition . The method involves:

    • Creating fusion proteins with RNA-binding domains

    • Incorporating specific binding sites in reporter mRNAs

    • Measuring translation rates with and without cap analogs or eIF4E mutants

  • Fluorescence anisotropy binding assays: These assays measure the direct binding of various eIF4F components to RNA substrates by monitoring changes in the rotational diffusion of fluorescently labeled RNA upon protein binding . This allows determination of apparent equilibrium dissociation constants (Kd values) for different protein-RNA interactions.

  • Two-hybrid experiments: For measuring protein-protein interactions, modified two-hybrid systems have been effectively used to examine interactions between eIF4E and various binding partners . This approach involves:

    • Fusion of eIF4E to the Gal4-binding domain

    • Fusion of potential binding partners to the activation domain of Gal4

    • Introduction of mutations in specific regions of eIF4E to map interaction sites

    • Quantification of reporter gene expression to assess interaction strength

These methodologies collectively provide a comprehensive toolkit for investigating the complex interactions and activities within the eIF4F complex, enabling researchers to dissect the multiple functions of eIF4E in translation initiation .

How can researchers effectively study the relationship between eIF4E levels and cancer progression?

Researchers investigating the relationship between eIF4E levels and cancer progression can employ several effective methodological approaches:

  • Transgenic and xenograft mouse models:

    • Developing conditional eIF4E overexpression models in specific tissues

    • Using patient-derived xenografts with varying eIF4E expression levels

    • Employing CRISPR/Cas9 to create defined eIF4E mutations in cancer cell lines prior to implantation

    • Measuring tumor growth rates, invasion capacity, and metastatic potential in relation to eIF4E levels

  • Polysome profiling and ribosome footprinting:

    • Isolating mRNAs associated with multiple ribosomes (actively translated)

    • Comparing translational efficiency of different mRNAs in cells with normal versus elevated eIF4E

    • Identifying the "eIF4E-sensitive" mRNA signature in different cancer types

    • Correlating translation of specific mRNAs with structural characteristics of their 5'-UTRs

  • Structure-function analysis:

    • Creating eIF4E mutants that separately affect cap binding versus helicase stimulation

    • Testing these mutants in cellular transformation assays

    • Determining which function of eIF4E (cap binding or helicase stimulation) is more critical for oncogenic activity

    • Developing selective inhibitors based on structural insights

  • Clinical correlation studies:

    • Analyzing eIF4E expression levels in human tumor samples versus matched normal tissues

    • Correlating eIF4E levels with clinical outcomes (survival, treatment response)

    • Examining the phosphorylation status of 4E-BPs in relation to eIF4E activity

    • Developing tissue microarrays to efficiently screen large patient cohorts

  • Pharmacological inhibition experiments:

    • Testing compounds that block eIF4E-cap interaction

    • Evaluating agents that disrupt eIF4E-eIF4G binding

    • Assessing compounds that target the eIF4E-mediated stimulation of helicase activity

    • Measuring effects on cancer cell proliferation, survival, and response to standard therapies

These approaches provide complementary information about how eIF4E levels affect cancer progression through selective enhancement of the translation of proliferative and survival mRNAs with structured 5'-UTRs .

What techniques are most reliable for measuring the effect of eIF4E on structured 5'-UTR translation?

Measuring the effect of eIF4E on structured 5'-UTR translation requires specialized techniques that can accurately assess translational efficiency in relation to RNA structure. The most reliable approaches include:

  • Reporter construct systems:

    • Creation of reporter genes (luciferase, GFP) with various structured 5'-UTRs

    • Systematic variation of 5'-UTR structural complexity (stem-loops, G-quadruplexes)

    • Measurement of reporter expression under conditions of normal, elevated, or depleted eIF4E

    • Normalization to control reporters with unstructured 5'-UTRs to isolate eIF4E effects

  • In vitro reconstituted translation systems:

    • Assembly of purified translation components with defined eIF4E concentrations

    • Use of reporter mRNAs with characterized 5'-UTR structures

    • Real-time monitoring of translation initiation rates

    • Addition of cap analogs or eIF4E mutants to distinguish between cap-binding and helicase-stimulation functions

  • RNA structure probing with translation correlation:

    • SHAPE (Selective 2'-hydroxyl acylation analyzed by primer extension) or DMS (dimethyl sulfate) probing to map RNA structures

    • Correlating structural features with translational efficiency measurements

    • Performing structure probing in the presence and absence of eIF4F components

    • Identifying specific structural elements that confer eIF4E sensitivity

  • Ribosome profiling with structure analysis:

    • Deep sequencing of ribosome-protected mRNA fragments

    • Computational analysis of 5'-UTR structural complexity

    • Correlation of ribosome occupancy patterns with predicted structures

    • Comparison between conditions of normal and elevated eIF4E levels

  • Tethered function assays:

    • Tethering eIF4E directly to mRNAs via RNA-binding domains

    • Bypassing the cap-binding requirement

    • Testing the effect of tethered eIF4E on translation of structured versus unstructured reporters

    • Using eIF4E mutants that specifically affect helicase stimulation without altering tethering

These techniques allow researchers to quantitatively assess how eIF4E levels affect the translation of mRNAs with different 5'-UTR structures, providing insights into the mechanism by which elevated eIF4E selectively enhances the expression of proliferative and survival genes in cancer cells .

How are researchers exploring eIF4E as a therapeutic target in cancer?

Researchers are pursuing multiple strategies to target eIF4E in cancer therapy, based on its critical role in selectively enhancing the translation of proliferative and survival mRNAs:

  • Direct inhibition of eIF4E-cap interaction:

    • Development of cap analogs with improved stability and cellular uptake

    • Design of small molecules that compete with the cap structure for binding to eIF4E

    • Screening of natural product libraries for compounds that bind the cap-binding pocket

    • Structure-based design of inhibitors that lock eIF4E in an inactive conformation

  • Disruption of eIF4E-eIF4G interaction:

    • Peptide mimetics based on the conserved eIF4E-binding motif found in eIF4G and 4E-BPs

    • Small molecule inhibitors that target the protein-protein interaction surface

    • Stabilization of 4E-BP activity through inhibition of its phosphorylation

    • Development of stapled peptides with improved cellular penetration and stability

  • Targeting the helicase-stimulating function:

    • Design of molecules that specifically block eIF4E's ability to stimulate eIF4A

    • Compounds that prevent eIF4E from relieving the autoinhibition of eIF4G

    • Identification of the structural interfaces required for this function

    • Screening for inhibitors that specifically affect structured mRNA translation

  • Antisense oligonucleotides and RNAi approaches:

    • Second-generation antisense oligonucleotides targeting eIF4E mRNA

    • Nanoparticle delivery systems for improved targeting of cancer cells

    • Combination with conventional therapies to enhance treatment efficacy

    • Clinical trials evaluating safety and efficacy profiles

  • Targeting upstream signaling pathways:

    • mTOR inhibitors that prevent 4E-BP phosphorylation and maintain eIF4E sequestration

    • Dual PI3K/mTOR inhibitors that more effectively block the pathway

    • MNK inhibitors that prevent eIF4E phosphorylation

    • Combination strategies targeting multiple nodes in the signaling network

These approaches aim to exploit the dependency of cancer cells on elevated eIF4E activity, potentially providing therapeutic benefit with a favorable therapeutic window based on the differential requirement for eIF4E in normal versus cancer cells .

What are the emerging connections between eIF4E and RNA stress granules in cellular stress responses?

The relationship between eIF4E and RNA stress granules represents an emerging area of research with significant implications for understanding cellular stress responses and disease states:

  • Dynamic sequestration mechanisms:

    • During cellular stress, eIF4E can be sequestered in stress granules along with untranslated mRNAs

    • This sequestration involves interactions with 4E-BPs and other stress granule components

    • The process serves as a mechanism to rapidly inhibit cap-dependent translation

    • Upon stress resolution, eIF4E can be released to resume translation of specific mRNAs

  • Differential mRNA fate determination:

    • eIF4E may play a role in determining which mRNAs are targeted to stress granules versus processing bodies

    • Its interactions with different protein partners in these structures influence mRNA storage versus degradation

    • Cancer cells often show altered stress granule dynamics, potentially related to eIF4E dysregulation

    • This may contribute to selective translational advantages during stress conditions

  • Liquid-liquid phase separation (LLPS) regulation:

    • Recent research suggests eIF4E participates in the LLPS processes that drive stress granule formation

    • Its interactions with intrinsically disordered regions of binding partners contribute to condensate properties

    • Phosphorylation of eIF4E or its binding partners can modulate these phase separation behaviors

    • This provides another level of translational regulation during stress responses

  • Stress granule dysregulation in disease:

    • Altered eIF4E levels or activity may contribute to abnormal stress granule dynamics in cancer

    • This can confer resistance to stress-inducing therapies by modulating the stress response

    • Targeting eIF4E could potentially normalize stress granule function in disease states

    • Understanding these connections may reveal new therapeutic opportunities

This emerging research area connects eIF4E's known roles in translation initiation with its less understood functions in mRNP granule biology, potentially expanding our understanding of how translational control intersects with cellular stress responses .

How does post-translational modification of eIF4E impact its dual functionality?

Post-translational modifications of eIF4E serve as critical regulators of its activity, affecting both its cap-binding function and its ability to stimulate eIF4A helicase activity:

  • Phosphorylation at Ser209:

    • Catalyzed primarily by MAP kinase-interacting kinases (MNKs)

    • Occurs within a flexible C-terminal loop of eIF4E

    • Increases the affinity of eIF4E for the cap structure

    • May also influence protein-protein interactions with eIF4G and 4E-BPs

    • Often elevated in cancer cells, contributing to enhanced translation of specific mRNAs

    • Potentially modulates the helicase-stimulating function, though this needs further research

  • Ubiquitination:

    • Multiple lysine residues in eIF4E can be ubiquitinated

    • May regulate eIF4E stability and turnover

    • Could affect subcellular localization and incorporation into different complexes

    • Potentially creates binding sites for ubiquitin-recognition proteins that modulate eIF4E function

    • The impact on helicase stimulation remains largely unexplored

  • Sumoylation:

    • Modification with small ubiquitin-like modifier (SUMO) proteins

    • May alter eIF4E's interaction with protein partners

    • Could influence nuclear-cytoplasmic distribution of eIF4E

    • Potentially important for eIF4E's nuclear functions in mRNA export

    • May create competition with other modifications at the same residues

  • Acetylation:

    • Several lysine residues are potential targets for acetylation

    • May regulate protein-protein interactions

    • Could influence the conformational dynamics of eIF4E

    • Potentially creates a regulatory mechanism responsive to cellular metabolic state

    • The functional consequences for both cap binding and helicase stimulation require further study

These post-translational modifications create a complex regulatory network that fine-tunes eIF4E activity in response to various cellular signals . The interplay between these modifications likely coordinates eIF4E's dual functionality in different cellular contexts and may be altered in disease states, making them potential targets for therapeutic intervention.

What are the most significant knowledge gaps in our understanding of human eIF4E?

Despite substantial progress in understanding human eIF4E function, several significant knowledge gaps remain that present opportunities for future research:

  • Structural basis of allosteric communication: While we know that binding at the dorsal surface influences cap binding at the ventral surface, the exact structural mechanisms and conformational changes involved in this allosteric communication remain incompletely characterized . Detailed structural studies, particularly using techniques like cryo-EM and hydrogen-deuterium exchange mass spectrometry, could reveal the dynamic changes that occur during this process.

  • Differential regulation of 4E-BP isoforms: The distinct binding characteristics of 4E-BP1 and 4E-BP2 have been documented, but how these differences translate to physiological functions remains unclear . Investigation of tissue-specific expression patterns, unique regulatory pathways, and differential effects on specific mRNA subsets would provide greater insight into the specialized roles of these isoforms.

  • Mechanism of helicase stimulation: While we know that eIF4E stimulates eIF4A helicase activity by counteracting an autoinhibitory domain in eIF4G, the precise molecular interactions and conformational changes involved in this process remain to be fully elucidated . Structural studies of the complete eIF4F complex in different functional states would significantly advance our understanding.

  • mRNA specificity determinants: The structural features that make certain mRNAs particularly dependent on eIF4E levels are not fully characterized . Comprehensive analysis of 5'-UTR structures, combined with systematic measurement of translational efficiency under varying eIF4E conditions, would help identify the specific elements that confer eIF4E sensitivity.

  • Integration with other translation regulatory mechanisms: How eIF4E function coordinates with other translation regulatory mechanisms, such as uORF-mediated regulation, IRES-dependent initiation, and specialized ribosomes, represents an area requiring further investigation. This would provide a more complete understanding of the translational control network.

Addressing these knowledge gaps will require interdisciplinary approaches combining structural biology, biochemistry, molecular genetics, and systems biology to fully understand the multifaceted roles of eIF4E in normal physiology and disease.

How might artificial intelligence and computational approaches advance eIF4E research?

Artificial intelligence and computational approaches offer promising avenues to accelerate and deepen eIF4E research in several key areas:

  • Structure prediction and modeling:

    • AI-powered tools like AlphaFold can predict protein structures with unprecedented accuracy

    • Molecular dynamics simulations can model the dynamic interactions between eIF4E and its binding partners

    • These approaches could reveal how allosteric communication occurs between the dorsal and ventral surfaces of eIF4E

    • Virtual screening of potential inhibitors can accelerate drug discovery efforts targeting different eIF4E functions

  • RNA structure analysis and prediction:

    • Deep learning approaches can predict RNA secondary and tertiary structures

    • Computational methods can identify structural motifs in 5'-UTRs that confer eIF4E sensitivity

    • Simulation of RNA-protein interactions can model how eIF4F components interact with structured mRNAs

    • These insights could help explain the selectivity of eIF4E for certain mRNAs

  • Network analysis and systems biology:

    • AI can integrate multi-omics data to map the translational regulatory network

    • Machine learning algorithms can identify patterns in translational efficiency across the transcriptome

    • Network modeling can predict how perturbations in eIF4E levels propagate through cellular systems

    • These approaches could reveal unexpected connections between eIF4E and other cellular processes

  • Predictive biomarkers and personalized medicine:

    • AI analysis of cancer genomics and proteomics data can identify patients likely to benefit from eIF4E-targeted therapies

    • Machine learning algorithms can predict resistance mechanisms to eIF4E inhibitors

    • Computational approaches can design combination therapies based on molecular profiles

    • These tools could accelerate clinical applications of eIF4E research

  • Automated literature mining and hypothesis generation:

    • Natural language processing can extract information from the vast scientific literature

    • AI can identify knowledge gaps and suggest novel hypotheses

    • Automated analysis of experimental results can accelerate discovery

    • These approaches could overcome human cognitive limitations in integrating complex information

By leveraging these computational approaches, researchers can gain deeper insights into eIF4E function, accelerate drug discovery efforts, and develop more personalized treatment strategies for diseases involving dysregulated eIF4E activity .

What interdisciplinary approaches might drive breakthroughs in understanding eIF4E's role in disease?

Significant breakthroughs in understanding eIF4E's role in disease will likely come from interdisciplinary approaches that bridge traditional research boundaries:

  • Integration of structural biology with cellular systems:

    • Combining cryo-EM structures of eIF4F complexes with live-cell imaging

    • Correlating structural dynamics with functional outcomes in cellular contexts

    • Developing structure-based biosensors to monitor eIF4E activity in living cells

    • Using these tools to understand how structural perturbations affect disease processes

  • Translation-focused single-cell technologies:

    • Adapting single-cell RNA-seq to capture translational status (e.g., single-cell Ribo-seq)

    • Measuring eIF4E activity variations in heterogeneous tumor populations

    • Correlating translational profiles with cellular phenotypes and disease progression

    • Identifying cell-specific vulnerabilities for therapeutic targeting

  • Metabolomics and translational control:

    • Investigating how metabolic changes affect eIF4E activity through mTOR signaling

    • Exploring connections between nutrient sensing, eIF4E function, and disease states

    • Developing metabolic interventions that selectively target eIF4E-dependent translation

    • Creating integrated models of metabolism and translational control

  • Immunology and translational regulation:

    • Understanding how eIF4E affects the translation of cytokines and immune signaling molecules

    • Exploring the role of eIF4E in immune cell activation and differentiation

    • Investigating eIF4E as a target for modulating immune responses in cancer and autoimmunity

    • Developing immunotherapy approaches that consider translational control mechanisms

  • Clinical-basic science partnerships:

    • Creating biorepositories of patient samples with detailed translational profiling

    • Developing patient-derived models that preserve translational regulatory features

    • Conducting mechanistic studies in the context of clinical trials

    • Rapidly translating basic discoveries into clinical applications

Product Science Overview

Introduction

Eukaryotic Translation Initiation Factor 4E (eIF4E) is a crucial protein involved in the initiation of translation in eukaryotic cells. It is a part of the eIF4F complex, which plays a pivotal role in the recognition of the 5’ cap structure of messenger RNAs (mRNAs) and the recruitment of ribosomes to initiate protein synthesis . The human recombinant form of eIF4E is produced using recombinant DNA technology, allowing for its use in various research and therapeutic applications.

Structure and Function

eIF4E is a cap-binding protein that specifically recognizes and binds to the 7-methylguanosine (m7G) cap at the 5’ end of eukaryotic mRNAs . This binding is essential for the recruitment of the eIF4F complex, which includes eIF4A (a helicase) and eIF4G (a scaffolding protein) . The eIF4F complex facilitates the unwinding of secondary structures in the 5’ untranslated region (UTR) of the mRNA, allowing the ribosome to access the mRNA and initiate translation .

Biological Significance

eIF4E is a rate-limiting factor in cap-dependent translation, meaning its availability and activity directly influence the efficiency of protein synthesis . It is involved in various cellular processes, including cell growth, proliferation, and survival . Dysregulation of eIF4E has been implicated in several diseases, including cancer, where its overexpression is often associated with tumorigenesis and poor prognosis .

Recombinant Production

The human recombinant form of eIF4E is produced using recombinant DNA technology, typically in bacterial or yeast expression systems . This involves cloning the eIF4E gene into an expression vector, transforming the host cells, and inducing protein expression. The recombinant protein is then purified using affinity chromatography techniques, such as His-tag or GST-tag purification .

Applications

Recombinant eIF4E is widely used in research to study the mechanisms of translation initiation and its regulation . It is also used in drug discovery and development, particularly in the search for inhibitors that can modulate eIF4E activity for therapeutic purposes . Additionally, recombinant eIF4E can be used in structural biology studies to elucidate its interactions with other components of the translation machinery .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.