EIF1B Human

Eukaryotic Translation Initiation Factor 1B Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Overview

Gene Characteristics

  • Location: Chromosome 3 (3p21.31) .

  • Aliases: GC20 .

  • Protein: Eukaryotic translation initiation factor 1B, a 113-amino acid protein .

Protein Structure

  • Domains: Contains a conserved SUI1 domain critical for ribosomal binding and translation initiation .

  • Functional Partners: Interacts with initiation factors (e.g., EIF1AX, EIF1AY) and ribosomal subunits (e.g., RPS3) .

FeatureDetails
Molecular Weight~12.5 kDa (predicted)
Isoforms1 primary transcript (ENSP00000232905)
ConservationOrthologs in vertebrates (e.g., zebrafish eif1b)

Functional Role in Translation Initiation

EIF1B contributes to the assembly of the 43S pre-initiation complex (PIC), a critical step in scanning mRNA for start codons . Key functions include:

  • Ribosome Binding: Stabilizes initiator tRNA binding to the 40S ribosomal subunit .

  • Context-Dependent Regulation: Modulates initiation site selection, particularly at non-AUG or suboptimal start codons .

  • Feedback Regulation: Autoregulates its own translation via a conserved upstream open reading frame (uORF) .

Mechanistic Insights

  • Overexpression reduces initiation at poor-context AUG codons by up to 25-fold, indicating sensitivity to cellular EIF1B levels .

  • Structural analogs in yeast (SUI1) and zebrafish highlight evolutionary conservation in translation fidelity .

Interaction Network

EIF1B forms a core interaction network with translation initiation factors and ribosomal proteins, as identified by STRING-DB :

Interacting ProteinFunctionInteraction Score
EIF1AXEnhances ribosome dissociation0.986
EIF3BRNA-binding component of eIF-3 complex0.963
EIF2S1Forms ternary complex with GTP and tRNA0.953
RPS340S ribosomal subunit component0.919

Regulatory Mechanisms

  • Autoregulation: EIF1B and its paralog EIF1 are regulated by negative feedback loops that adjust their synthesis in response to cellular abundance .

  • Post-Transcriptional Control: MicroRNAs (e.g., miR-30a-5p) target EIF1B, as predicted by TargetScan .

Clinical and Research Relevance

Cancer Associations

  • Expression in Tumors: Overexpressed in glioblastoma and colorectal cancer .

  • Functional Associations: Linked to 4,552 biological entities, including cell proliferation pathways and DNA repair mechanisms .

Disease Linkages

  • DNA Damage Response: Associates with RPS3, a ribosomal protein involved in DNA repair .

  • Pharmacogenomics: Identified as a dependency in specific cancer cell lines (e.g., Sanger Dependency Map) .

Expression Profiling

  • Tissue Specificity: Ubiquitous but enriched in tissues with high protein synthesis rates (e.g., liver, pancreas) .

  • Cancer vs. Normal: Differential expression patterns observed in TCGA datasets .

Cancer TypeExpression TrendClinical Correlation
GlioblastomaUpregulatedPoor prognosis
Colorectal CancerUpregulatedMetastasis-associated

Future Directions

  • Therapeutic Targeting: Explore EIF1B’s role in cancer-specific translation initiation.

  • Mechanistic Studies: Resolve structural interactions with the 43S PIC using cryo-EM.

Product Specs

Introduction
EIF1B plays a crucial role in the initiation of translation by facilitating the scanning process for the start codon. It is a key component of a complex that identifies the initiator codon during this process. Moreover, EIF1B regulates the activity of 43S, 48S, and 40S ribosomal subunits, which are essential for translation initiation. EIF1B enables the 43S ribosomal complexes to differentiate between correct and nearly correct initiation codons by recognizing the nucleotide sequence of these codons.
Description
Recombinant human EIF1B protein, expressed in E. coli, is fused with a 20 amino acid His tag at its N-terminus. This single, non-glycosylated polypeptide chain consists of 133 amino acids (residues 1-113) and has a molecular weight of 15 kDa. Purification of EIF1B is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The EIF1B solution is provided at a concentration of 1 mg/ml in a buffer containing 20mM Tris-HCl (pH 8.0), 10% glycerol, 2mM DTT, and 0.1M NaCl.
Stability
For short-term storage (2-4 weeks), the EIF1B vial can be kept at 4°C. For extended storage, freezing at -20°C is recommended. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of EIF1B is greater than 95.0%, as determined by SDS-PAGE analysis.
Synonyms
Eukaryotic translation initiation factor 1b, eIF1b, Protein translation factor SUI1 homolog GC20, EIF1B, GC20.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MSTIQNLQSF DPFADATKGD DLLPAGTEDY IHIRIQQRNG RKTLTTVQGI ADDYDKKKLV KAFKKKFACN GTVIEHPEYG EVIQLQGDQR KNICQFLLEV GIVKEEQLKV HGF.

Q&A

What is the function of EIF1B in human cells?

EIF1B (eukaryotic translation initiation factor 1B) is a protein that participates in the translation initiation process in human cells. It functions primarily by enabling RNA binding activity, ribosomal small subunit binding, and translation initiation factor activity . As part of the eukaryotic 43S preinitiation complex, EIF1B plays a crucial role in the accurate recognition of start codons during mRNA translation . Research indicates that EIF1B shares functional similarities with its paralog EIF1, which is known to be involved in the stringency of start codon selection .

To investigate EIF1B function experimentally, researchers typically employ knockdown or knockout approaches followed by analysis of global translation patterns using techniques such as ribosome profiling or proteomics. The protein contains a SUI1 domain that is essential for its function in translation initiation .

How does EIF1B differ from EIF1 in structure and function?

While EIF1B and EIF1 are paralogs with significant structural similarities, they exhibit distinct functional characteristics in translation regulation. Both contain the SUI1 domain critical for translation initiation , but research suggests they may have evolved different regulatory mechanisms and expression patterns.

Studies with knockout models demonstrate that while EIF1 knockdown leads to significant changes in translation patterns affecting 245 transcripts with upstream open reading frames (uORFs), EIF1B may have complementary or redundant functions . The differential effects of EIF1 versus EIF1B depletion on translation can be analyzed through:

ParameterEIF1 KnockdownEIF1B KnockoutMethodology
mRNA affected159 significant changesFewer changesmRNA-seq with Z-score analysis (p<0.01)
CDS translation125 significant changesDistinct patternRibosome profiling with LTM/CHX
uORF translation291 significant changesDifferent subsetPROTEOFORMER pipeline analysis
Protein expression238 regulated proteinsUnique signatureLC-MS/MS proteomics (ANOVA p<0.05)

These differences suggest that while the proteins share structural domains, they likely have context-specific roles in translation regulation.

What genomic and protein characteristics define human EIF1B?

Human EIF1B is characterized by specific genomic and protein features that influence its function:

  • Genomic location: Located on chromosome 24 according to ortholog mapping

  • Protein length: Approximately 113 amino acids in length

  • Domain structure: Contains the eukaryotic translation initiation factor SUI1 domain and SUI1 domain superfamily

  • Transcript variants: Multiple mRNA variants exist, including documented variants of 784 nt (eif1b-201) and 896 nt (zgc:56676-001)

The SUI1 domain is particularly significant as it mediates the protein's interaction with the ribosome and other translation initiation factors. This domain is highly conserved across species, underscoring its functional importance in the fundamental process of translation initiation across eukaryotes.

What are the most effective methods for studying EIF1B protein interactions?

For investigating EIF1B protein interactions, several complementary approaches yield comprehensive insights:

  • Immunoprecipitation coupled with mass spectrometry (IP-MS): This high-throughput approach is particularly effective for identifying novel interaction partners. When applying IP-HTMS to human proteins, researchers can express Flag-tagged EIF1B in human cells, followed by immunoprecipitation and LC-ESI-MS/MS analysis to identify co-precipitated proteins . This approach has successfully identified thousands of protein-protein interactions in human cells, with empirical filtering criteria and confidence scoring to minimize false positives .

  • Targeted co-immunoprecipitation: For validating specific suspected interactions, researchers can perform co-IP experiments using antibodies against endogenous proteins followed by Western blotting.

  • Proximity labeling methods: BioID or APEX2-based approaches can identify proteins in close proximity to EIF1B in living cells, providing spatial context to interactions.

  • Structural analysis: Techniques like cryo-EM can reveal the molecular basis of EIF1B interactions within the translation initiation complex.

When analyzing interaction data, application of confidence scoring systems is crucial for filtering out false positives. As demonstrated in genome-wide interaction studies, combining several experimental metrics with computational validation against known interactions enhances reliability .

How can researchers effectively perform knockdown or knockout studies of EIF1B?

To study the functional consequences of EIF1B depletion, researchers should implement a systematic experimental design:

  • siRNA-mediated knockdown: This approach typically achieves partial depletion (approximately 60% at transcript level and 50% at protein level) . Multiple siRNA sequences should be tested to identify the most effective while minimizing off-target effects.

  • CRISPR-Cas9 genome editing: For complete knockout studies, researchers can generate cell lines lacking functional EIF1B, similar to the EIF1BKO HAP1 cell lines referenced in literature .

  • Validation procedures:

    • qPCR to measure knockdown efficiency at the transcript level

    • Western blotting to confirm protein reduction

    • Rescue experiments with wild-type EIF1B to confirm specificity

  • Experimental controls:

    • Parallel EIF1 knockdown/knockout for comparative analysis

    • Non-targeting siRNA or guide RNA controls

    • Wild-type parental cell lines

For comprehensive analysis, researchers should examine multiple readouts including global translation patterns (polysome profiling), specific mRNA translation (ribosome profiling), and protein synthesis (pulse labeling). The effects should be assessed across multiple cell types to account for context-specific functions.

What approaches are most suitable for analyzing EIF1B's role in translation regulation?

To comprehensively analyze EIF1B's role in translation regulation, researchers should implement an integrated multi-omics strategy:

  • Ribosome profiling: This technique provides nucleotide-resolution information about ribosome positioning on mRNAs. Using translation inhibitors like lactimidomycin (LTM) and cycloheximide (CHX) enables discrimination between initiation and elongation events . The PROTEOFORMER pipeline can be applied to map ribosome-protected fragments to the genome and identify translation initiation sites .

  • mRNA sequencing: Parallel mRNA-seq allows researchers to distinguish translational from transcriptional effects .

  • Quantitative proteomics: Label-free or isotope-labeled mass spectrometry approaches can quantify protein-level changes resulting from EIF1B modulation .

  • Reporter assays: Constructs containing upstream open reading frames (uORFs) or suboptimal start codons can test how EIF1B affects start codon selection stringency.

  • Data integration: Combining these approaches allows categorization of affected genes into those regulated at transcriptional, translational, or post-translational levels. In studies of EIF1 knockdown, this integration revealed that 41% of changes in protein expression resulted from differential transcription, 28% from translation, and 18% from a combination of mechanisms .

This comprehensive approach provides a systems-level understanding of how EIF1B contributes to translation regulation and identifies specific mRNAs whose translation depends on proper EIF1B function.

How should researchers analyze multi-omics data to understand EIF1B function?

Multi-omics data analysis for EIF1B research requires sophisticated integration strategies:

  • Statistical framework: Implement Z-scoring strategies with adjustment for expression levels, as described in previous studies of translation initiation factors . A threshold P-value of 0.01 (corresponding to Z-score ≥ 2.58) is typically used to detect significant deviations in translational efficiency .

  • Comparative analysis workflow:

    • Map transcriptomic changes using mRNA-seq

    • Identify shifts in translation using ribosome profiling

    • Quantify protein-level alterations through proteomics

    • Apply ANOVA testing with appropriate thresholds (e.g., P < 0.05) for protein expression analysis

  • Classification of regulatory mechanisms: Systematically categorize affected genes based on the level of regulation:

    • Transcriptional (changes in mRNA levels)

    • Translational (changes in ribosome occupancy without mRNA changes)

    • Post-translational (changes in protein levels without corresponding changes in mRNA or translation)

  • Feature analysis: Examine sequence features of differentially translated mRNAs, particularly focusing on:

    • Start codon context (Kozak sequence strength)

    • Presence and characteristics of upstream open reading frames (uORFs)

    • 5' UTR structural elements

This systematic approach revealed that in EIF1 knockdown studies, 13% of proteins displayed significant changes in steady-state expression without notable changes in transcript or translation levels, suggesting effects on protein turnover or post-translational modifications .

What bioinformatics tools are most useful for analyzing EIF1B-related protein interaction networks?

For robust analysis of EIF1B-related protein interaction networks, researchers should employ several complementary bioinformatics approaches:

  • Network construction and visualization:

    • Cytoscape for network visualization and analysis

    • STRING database for interaction prediction and confidence scoring

    • IntAct and BioGRID for curated interaction data

  • Confidence assessment metrics:

    • Implement empirical filtering criteria based on peptide counts and reproducibility

    • Calculate interaction confidence scores combining experimental and identification metrics

    • Cross-validate using previously published human protein interactions

  • Functional enrichment analysis:

    • Gene Ontology (GO) enrichment for biological processes associated with interacting proteins

    • Pathway analysis using KEGG, Reactome, or other pathway databases

    • Protein domain enrichment to identify common structural features

  • Co-expression analysis:

    • Examine co-expression patterns of EIF1B and its interactors across tissues and conditions

    • Identify tightly co-expressed protein clusters that may represent functional complexes

When reporting interaction data, researchers should provide confidence metrics for each interaction rather than simply reporting binary interactions. This approach has been validated in large-scale human interactome studies, where filtering based on confidence scores significantly improved the biological relevance of identified interactions .

How can researchers resolve contradictory findings in EIF1B functional studies?

When facing contradictory findings in EIF1B research, a systematic troubleshooting approach is essential:

  • Methodological comparison:

    • Document differences in knockdown/knockout approaches (transient vs. stable, partial vs. complete)

    • Compare cell types used (different cell lines may have varying dependence on EIF1B)

    • Assess timing of analyses (acute vs. chronic depletion effects)

  • Compensatory mechanism assessment:

    • Measure expression changes in EIF1 and other translation factors that might compensate for EIF1B loss

    • Perform double knockdown/knockout experiments to identify redundant functions

    • Analyze isoform-specific effects if multiple EIF1B variants exist

  • Context-dependent function investigation:

    • Test EIF1B function under different stress conditions

    • Examine cell-cycle dependent effects

    • Assess nutrient availability influences

  • Technical validation:

    • Confirm antibody specificity through knockout controls

    • Validate key findings using orthogonal techniques

    • Reproduce critical experiments in independent laboratories

  • Systematic meta-analysis:

    • Pool data from multiple studies using standardized normalization

    • Weight findings based on methodological rigor and sample size

    • Identify consistent patterns despite apparent contradictions

This structured approach helps distinguish genuine biological complexity from technical artifacts, leading to a more nuanced understanding of EIF1B function across cellular contexts.

How does EIF1B contribute to translational control of upstream open reading frames (uORFs)?

The role of EIF1B in regulating translation through upstream open reading frames (uORFs) represents a frontier in translation control research:

EIF1B likely influences uORF translation by modulating start codon selection stringency. Studies of its paralog EIF1 have demonstrated that when EIF1 levels are reduced, the stringency of start codon selection diminishes, leading to increased translation of uORFs with suboptimal start codons . This regulatory mechanism affects hundreds of genes, with 313 uORFs showing altered translational efficiency upon EIF1 knockdown .

Investigating this aspect of EIF1B function requires:

  • Genome-wide uORF analysis:

    • Ribosome profiling with enhanced 5' UTR coverage

    • Comparative analysis of uORF vs. main ORF translation upon EIF1B modulation

    • Classification of affected uORFs based on start codon context and sequence features

  • Mechanistic studies:

    • In vitro reconstitution experiments to measure start codon selection fidelity

    • Single-molecule approaches to visualize scanning and initiation events

    • Structural studies of EIF1B interaction with the ribosome at near-cognate start codons

  • Functional consequences:

    • Analysis of how uORF de-repression affects main ORF translation

    • Identification of biological pathways particularly sensitive to EIF1B-mediated uORF regulation

    • Investigation of stress conditions that modulate this regulatory mechanism

This research direction has significant implications for understanding translational control in health and disease, as many stress-responsive and regulatory genes contain uORFs that control their expression.

What is the role of EIF1B in stress-responsive translation regulation?

EIF1B's potential role in stress-responsive translation represents a critical area for advanced investigation:

While direct evidence for EIF1B in stress response is limited, studies of EIF1 indicate that genes affected by eIF1 deprivation are implicated in energy production and sensing of metabolic stress . This suggests EIF1B may similarly participate in translational reprogramming during stress conditions.

A comprehensive research strategy should address:

  • Stress-specific expression patterns:

    • Quantify EIF1B expression changes across diverse stress conditions (oxidative, ER, nutritional, hypoxic)

    • Compare with changes in EIF1 and other initiation factors

    • Determine if EIF1B:EIF1 ratio shifts during specific stress responses

  • Integration with stress response pathways:

    • Investigate EIF1B interaction with eIF2α phosphorylation pathway components

    • Examine relationship with mTOR signaling under nutrient stress

    • Assess connection to integrated stress response (ISR) mediators

  • Selective translation regulation:

    • Identify mRNAs whose translation becomes particularly dependent on EIF1B during stress

    • Characterize sequence features that determine this dependency

    • Determine if EIF1B influences stress granule formation or composition

  • Temporal dynamics:

    • Map the time course of EIF1B involvement in acute versus chronic stress adaptation

    • Investigate whether EIF1B participates in translational memory of repeated stresses

This research direction has implications for understanding cellular adaptation mechanisms and may reveal EIF1B as a potential therapeutic target in diseases involving dysregulated stress responses.

How do post-translational modifications influence EIF1B function?

Post-translational modifications (PTMs) of EIF1B likely serve as key regulatory mechanisms that dynamically control its activity in response to cellular conditions:

While specific PTMs of EIF1B have not been extensively characterized in the provided search results, studies of translation initiation factors generally indicate that PTMs can dramatically alter their function, interactions, and localization. For EIF1B, potential regulatory modifications may include:

  • Phosphorylation dynamics:

    • Identification of specific residues subject to phosphorylation

    • Mapping of responsible kinases and phosphatases

    • Functional consequences for ribosome binding and start codon selection

  • Ubiquitination and stability regulation:

    • Characterization of ubiquitination sites and chain topologies

    • Effects on EIF1B turnover rates under different conditions

    • Potential non-degradative signaling roles of ubiquitination

  • Other potential modifications:

    • Methylation, acetylation, or SUMOylation events

    • Crosstalk between different modification types

    • Stress-induced modification patterns

  • Methodological approaches:

    • Mass spectrometry-based PTM mapping

    • Site-directed mutagenesis of modified residues

    • Expression of modification-mimicking variants

    • Temporal analysis of modification dynamics during cellular responses

Understanding these modifications would provide crucial insights into how cells fine-tune translation initiation in response to changing conditions and may reveal new regulatory mechanisms governing protein synthesis.

What emerging technologies will advance understanding of EIF1B function?

Several cutting-edge technologies are poised to transform our understanding of EIF1B biology:

  • Cryo-electron microscopy (Cryo-EM):

    • High-resolution structural analysis of EIF1B within the translation initiation complex

    • Visualization of conformational changes during start codon recognition

    • Comparison with EIF1-containing complexes to identify functional differences

  • Single-molecule techniques:

    • Real-time observation of EIF1B dynamics during scanning and start codon selection

    • FRET-based approaches to measure conformational changes

    • Optical tweezers to quantify binding forces and kinetics

  • Advanced proteomics:

    • Thermal proteome profiling to identify EIF1B targets and binding partners

    • Crosslinking mass spectrometry to map interaction interfaces

    • Targeted proteomics for absolute quantification of EIF1B:EIF1 ratios

  • Spatial transcriptomics and proteomics:

    • Localization of EIF1B-dependent translation events within cells

    • Tissue-specific functions in complex organisms

    • Subcellular compartment-specific roles

  • Systems biology approaches:

    • Machine learning models to predict EIF1B-dependent translation outcomes

    • Network analysis integrating multiple levels of regulation

    • Computational prediction of small molecule modulators of EIF1B function

These technologies, especially when combined in multi-modal approaches, promise to reveal new dimensions of EIF1B function beyond what conventional methods have uncovered.

How does evolutionary conservation inform EIF1B function across species?

Evolutionary analysis provides critical insights into EIF1B's core functions and species-specific adaptations:

The SUI1 domain found in EIF1B is highly conserved across eukaryotes, indicating fundamental importance in translation initiation . This conservation allows for comparative studies across model organisms, from yeast to mammals.

A comprehensive evolutionary analysis should examine:

  • Sequence conservation patterns:

    • Identification of invariant residues critical for core function

    • Variable regions that may confer species-specific regulation

    • Selective pressure analysis to detect positively selected sites

  • Paralog relationships:

    • Evolutionary history of EIF1 and EIF1B gene duplication events

    • Differential conservation of regulatory elements controlling expression

    • Species lacking EIF1B and potential compensatory mechanisms

  • Functional conservation testing:

    • Cross-species rescue experiments to test functional equivalence

    • Comparative biochemical studies of initiation factor properties

    • Heterologous expression to identify species-specific interactions

  • Co-evolution analysis:

    • Identification of co-evolving partners in the translation machinery

    • Correlation with translation initiation mechanism complexity

    • Relationship to genome complexity and regulatory sophistication

This evolutionary perspective not only informs fundamental understanding of EIF1B but also guides experimental design by highlighting the most functionally critical aspects of the protein.

What are the implications of EIF1B research for therapeutic development?

Translation initiation represents an emerging frontier for therapeutic intervention, with EIF1B research potentially contributing to several biomedical applications:

  • Cancer therapy opportunities:

    • Many cancers exhibit altered translation initiation patterns

    • EIF1B modulation might selectively affect translation of oncogenes with complex 5' UTRs

    • Combinatorial approaches targeting multiple initiation factors could increase specificity

  • Neurological disorder applications:

    • Protein synthesis dysregulation underlies several neurodegenerative conditions

    • EIF1B-dependent translation may affect specific neuron-enriched transcripts

    • Local translation regulation in neurons represents a potential intervention point

  • Viral infection countermeasures:

    • Many viruses manipulate host translation machinery

    • Understanding EIF1B's role in viral mRNA translation could reveal antiviral strategies

    • Small molecules targeting EIF1B-ribosome interaction might selectively inhibit viral protein synthesis

  • Development pathway:

    • Target validation through genetic and pharmacological approaches

    • High-throughput screening for EIF1B modulators

    • Structure-based drug design based on EIF1B binding pockets

    • Therapeutic window assessment comparing effects on normal versus disease-state cells

While direct EIF1B targeting remains speculative, the knowledge gained from basic research on this factor will contribute to the broader field of translation-targeted therapeutics, which has shown promising results in preclinical settings.

Product Science Overview

Introduction

Eukaryotic Translation Initiation Factor 1B (EIF1B) is a protein-coding gene that plays a crucial role in the initiation of translation in eukaryotic cells. This factor is essential for the proper assembly of the translation initiation complex, which is necessary for the accurate and efficient synthesis of proteins.

Gene and Protein Information

The EIF1B gene is located on chromosome 3 and encodes a protein that is involved in the binding of RNA and the initiation of translation . The protein is a part of the eukaryotic 43S preinitiation complex, which is responsible for the recognition of the start codon on mRNA and the assembly of the ribosome .

Function and Mechanism

EIF1B is predicted to be involved in the early stages of translation initiation. It enables RNA binding activity and is a crucial component of the translation initiation machinery . The protein interacts with other initiation factors to form a stable complex that facilitates the recruitment of the ribosome to the mRNA .

Diseases and Disorders

Mutations or dysregulation of the EIF1B gene have been associated with various diseases, including kidney fibrosarcoma and bladder lateral wall cancer . The gene’s involvement in these diseases highlights its importance in cellular processes and the potential consequences of its malfunction.

Research and Applications

Human recombinant EIF1B is used in various research applications to study the mechanisms of translation initiation and the role of initiation factors in protein synthesis. Recombinant proteins are produced using recombinant DNA technology, which involves inserting the gene encoding EIF1B into an expression vector and introducing it into a host cell to produce the protein.

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.