EIF5A antibodies are specialized tools designed to detect and study the eukaryotic translation initiation factor 5A (eIF5A), a critical protein involved in mRNA translation elongation and termination. These antibodies are engineered to bind specifically to eIF5A, enabling its detection in various experimental contexts. Key characteristics include:
EIF5A antibodies are pivotal in studying eIF5A’s roles in cellular processes and diseases. Below are key applications supported by experimental data:
Translation Regulation: Required for elongation of polyproline motifs and termination of specific mRNAs, including those encoding IFNγ and cytotoxic T cell proteins .
Viral Replication: Essential for replication of RNA viruses (e.g., influenza, Ebola) by facilitating viral mRNA translation .
Autophagy: Mediates ATG3 translation, critical for autophagosome formation .
Immune Response: Regulates T cell survival and cytokine production (e.g., IFNγ) .
Detection Sensitivity: Monoclonal antibodies (e.g., AB01/2G8) detect ~17 kDa bands in Western blotting, while polyclonal antibodies (e.g., AF7558) show broader cross-reactivity .
Optimal Dilution:
T Cell Activation: Hypusinated eIF5A is dynamically regulated in naïve CD8+ T cells, enabling IFNγ production and cytotoxicity .
Senescence Surveillance: Regulates transcription factor EB (TFEB) synthesis in B cells and autophagy in aged T cells .
EIF5A (eukaryotic translation initiation factor 5A) is an mRNA-binding protein primarily involved in translation elongation rather than initiation, despite its name. Its significance stems from being the only known protein containing hypusine, a unique amino acid formed through post-translational modification of a specific lysine residue. This hypusination is essential for cellular proliferation and responses to extracellular stressors . In vertebrates, there are two isoforms: eIF5A1 (constitutively expressed in all tissues) and eIF5A2 (primarily expressed in gonads) . The protein plays crucial roles in various cellular processes and has been proposed as a potential target for pharmacologic therapy in conditions like infections, cancer, and obesity .
Several types of EIF5A antibodies are available for research, including:
Mouse monoclonal antibodies:
Rabbit antibodies:
Specialized antibodies:
These antibodies vary in their specificity, reactivity with different species, and optimal applications, allowing researchers to select the most appropriate tool for their specific experimental needs.
The key difference lies in epitope specificity and the biological information they provide:
Hypusine-specific antibodies (such as IU-88) selectively recognize only the hypusinated or deoxyhypusinated forms of eIF5A. These specialized antibodies bind specifically to the modified lysine residue (hypusine) or its immediate surrounding region. They provide crucial information about the functional state of eIF5A, as only hypusinated eIF5A is biologically active .
In experimental contexts, total eIF5A antibodies help assess expression levels, while hypusine-specific antibodies reveal the proportion of eIF5A that is post-translationally modified and functionally active. The IU-88 antibody, for example, has been characterized to specifically recognize either deoxyhypusine or hypusine forms of eIF5A in vitro and the hypusinated form in cellular extracts .
Different eIF5A antibodies demonstrate optimal performance in specific applications based on their characteristics:
Antibody Type | Optimal Applications | Recommended Dilutions | Species Reactivity |
---|---|---|---|
Mouse monoclonal [4E10F6] (ab204939) | Western Blot, IHC-P, Flow Cytometry | WB: 1/500-1/2000 | Human |
Mouse monoclonal (67214-1-Ig) | Western Blot, IF/ICC, ELISA | WB: 1:5000-1:50000, IF/ICC: 1:400-1:1600 | Human, Mouse, Rat |
Rabbit polyclonal (11309-1-AP) | Western Blot, IHC, IF/ICC, ELISA | WB: 1:5000-1:50000, IHC: 1:50-1:500, IF/ICC: 1:50-1:500 | Human, Mouse, Rat |
Hypusine-specific (IU-88) | Western Blot, Immunocytochemistry | Experiment-dependent | Human, potentially others |
For optimal results, consider these application-specific recommendations:
Western Blot: Mouse monoclonal antibodies typically provide cleaner bands with less background. The expected molecular weight for eIF5A is approximately 18 kDa .
Immunohistochemistry: Rabbit polyclonal antibodies often provide stronger signals in fixed tissues. For 11309-1-AP, antigen retrieval with TE buffer pH 9.0 is recommended, though citrate buffer pH 6.0 may also work .
Immunofluorescence: Both monoclonal and polyclonal antibodies work well; selection should be based on species compatibility with other antibodies in multi-labeling experiments.
Functional studies: For assessing the active form of eIF5A, hypusine-specific antibodies like IU-88 are essential .
Validating antibody specificity is crucial for reliable results. For eIF5A antibodies, implement these validation approaches:
Positive controls: Use cell lines known to express eIF5A, such as HeLa, HEK-293T, NIH/3T3, or PC-3 cells .
Molecular weight verification: Confirm detection at the expected molecular weight (18 kDa for endogenous eIF5A) .
Genetic validation:
siRNA/shRNA knockdown: Compare antibody signal between control and eIF5A-depleted samples
Overexpression: Transfect cells with eIF5A expression vectors and verify increased signal intensity
Mutant controls: Use eIF5A(K50A) mutant (which cannot be hypusinated) as a negative control for hypusine-specific antibodies
Hypusination-specific validation (for hypusine-specific antibodies):
Cross-reactivity assessment: Test the antibody against samples from multiple species if working with non-human models.
Multiple detection methods: Confirm findings using different techniques (e.g., WB, IF, IHC) to ensure consistent detection.
For hypusine-specific antibodies like IU-88, additional validation can include co-transfection with DHS (deoxyhypusine synthase) to enhance hypusination in cell types where DHS may be limiting .
Optimal protocols for eIF5A detection vary by sample type and application:
Sample preparation:
Cell lysis: Use RIPA buffer with protease inhibitors
Protein quantification: Bradford or BCA assay
Loading: 20-30 μg total protein per lane
Electrophoresis and transfer:
12-15% SDS-PAGE (to resolve the 18 kDa eIF5A protein)
Transfer to PVDF or nitrocellulose membrane (0.2 μm pore size recommended)
Immunoblotting:
Fixation: 10% neutral buffered formalin
Antigen retrieval: TE buffer pH 9.0 (recommended for 11309-1-AP) or citrate buffer pH 6.0
Primary antibody: Dilute 1:50-1:500, incubate overnight at 4°C
Detection: Standard polymer detection system with DAB or other chromogen
Fixation: 4% paraformaldehyde (10 minutes)
Permeabilization: 0.1-0.5% Triton X-100 in PBS (5-10 minutes)
Blocking: 1-5% BSA in PBS (1 hour)
Primary antibody: Dilute 1:50-1:500 for polyclonal or 1:400-1:1600 for monoclonal antibodies
Secondary antibody: Fluorophore-conjugated, species-appropriate
Counterstain: DAPI for nuclei
Mounting: Anti-fade medium
For hypusinated eIF5A detection:
When using hypusine-specific antibodies like IU-88, include appropriate controls (GC7-treated samples) and consider cell type-specific factors (e.g., DHS availability may be limiting in some cell types like 293T cells but not in INS-1 β cells) .
Studying hypusination dynamics requires sophisticated approaches combining hypusine-specific antibodies with other methods:
Time-course analysis after inhibition/stimulation:
Treat cells with GC7 (DHS inhibitor) to block hypusination, then remove the inhibitor and monitor the restoration of hypusinated eIF5A using IU-88 or other hypusine-specific antibodies
Time-resolved Western blotting with parallel detection of total eIF5A and hypusinated eIF5A
Quantify the hypusine/total eIF5A ratio to assess modification efficiency
Cell-type comparative analysis:
Different cell types show varying hypusination capacity. For example, research shows that DHS is limiting in 293T cells but not in INS-1 β cells
Design experiments that compare hypusination levels across tissue/cell types using Western blot or immunofluorescence with hypusine-specific antibodies
Include co-transfection with DHS to assess whether DHS availability limits hypusination in specific cell types
Stress response studies:
Expose cells to various stressors (oxidative stress, nutrient deprivation, etc.)
Monitor changes in hypusination status and correlate with cellular responses
Compare stress effects across normal and diseased cell models
Combined immunoprecipitation approaches:
Perform sequential immunoprecipitation with hypusine-specific and total eIF5A antibodies
Analyze eIF5A-interacting proteins under different conditions to identify hypusination-dependent interactions
Use proximity ligation assays to visualize interactions of hypusinated eIF5A with partner proteins in situ
Pulse-chase analysis:
Label newly synthesized proteins with metabolic labels
Track the kinetics of eIF5A hypusination using timecourse immunoprecipitation with hypusine-specific antibodies
These approaches can reveal how hypusination is regulated in response to developmental cues, stress conditions, or in disease states where translation regulation is altered.
Studying eIF5A isoforms presents several challenges and requires careful experimental design:
Antibody specificity challenges:
eIF5A1 and eIF5A2 share approximately 84% amino acid sequence identity
Most commercially available antibodies may cross-react with both isoforms
Verify isoform specificity through:
Expression pattern considerations:
eIF5A1 is constitutively expressed in all tissues, making it easier to detect
eIF5A2 has restricted expression (mainly gonads) and may require more sensitive detection methods
In cancer tissues, eIF5A2 is often overexpressed and can be detected more readily
Experimental design strategies:
Use RT-qPCR to confirm transcript levels of each isoform before protein analysis
Consider isoform-specific siRNA knockdown to validate antibody specificity
For tissues with low eIF5A2 expression, consider enrichment strategies (e.g., immunoprecipitation before Western blot)
Functional analysis considerations:
Design experiments that can distinguish functional differences between isoforms
Consider the different roles of each isoform in normal physiology versus disease states
When studying hypusination, remember both isoforms can be hypusinated
Subcellular localization studies:
Use high-resolution imaging with validated isoform-specific antibodies
Consider co-staining with organelle markers to detect differential localization
A methodologically sound approach would involve using multiple antibodies targeting different epitopes, combined with genetic manipulation of each isoform to establish specificity and reliability of the detection system.
eIF5A antibodies provide powerful tools for investigating disease associations through several methodological approaches:
Cancer research applications:
Compare hypusination levels between normal and tumor tissues using hypusine-specific antibodies like IU-88
Correlate eIF5A and/or hypusinated eIF5A levels with cancer stage, prognosis, or treatment response
Assess eIF5A2 overexpression in cancers using isoform-specific antibodies, as eIF5A2 has been implicated as an oncogene
Methodology: Tissue microarray immunohistochemistry with quantitative image analysis
Viral infection studies:
Monitor changes in eIF5A hypusination during viral infections, as eIF5A serves as a cellular cofactor for HTLV-1 Rex and HIV-1 Rev proteins
Track the interaction between viral proteins and eIF5A using co-immunoprecipitation with eIF5A antibodies
Methodology: Time-course immunoblotting with both total and hypusine-specific antibodies in infected versus uninfected cells
Metabolic disorders research:
Assess changes in hypusination in obesity and diabetes models, where translation regulation may be altered
Examine the relationship between polyamine metabolism (required for hypusination) and disease progression
Methodology: Western blot analysis of tissue samples with parallel detection of hypusination enzymes (DHS, DOHH)
Pharmacological intervention studies:
Use hypusine-specific antibodies to monitor the efficacy of DHS inhibitors (like GC7) or other drugs targeting the hypusination pathway
Assess downstream effects of hypusination inhibition on disease-relevant cellular processes
Methodology: Dose-response and time-course studies combining Western blot, IF, and functional assays
Multi-parametric analysis in patient samples:
Combine eIF5A/hypusinated eIF5A detection with markers of cellular stress, inflammation, or disease progression
Develop prognostic/diagnostic panels that include eIF5A status
Methodology: Multiplex immunofluorescence or sequential immunohistochemistry
For robust disease-related studies, researchers should consider longitudinal sampling when possible and include proper controls (both disease and treatment controls). Statistical analysis should account for patient heterogeneity and potential confounding factors in the interpretation of eIF5A-related findings.
Researchers frequently encounter several technical challenges when working with eIF5A antibodies:
High background in Western blots:
Problem: Non-specific binding creating diffuse signal
Solutions:
Increase blocking time/concentration (try 5% BSA instead of milk)
Dilute primary antibody further (especially for high-sensitivity antibodies)
Include 0.1% Tween-20 in antibody dilution buffer
Try shorter incubation times at room temperature instead of overnight at 4°C
Multiple bands in Western blot:
Problem: Detection of post-translational modifications or degradation products
Solutions:
Use freshly prepared lysates with complete protease inhibitors
For hypusine-specific antibodies, verify with GC7-treated controls to identify which band represents hypusinated eIF5A
Run longer gels (15-20%) for better resolution of the small (18 kDa) eIF5A protein
Consider phosphatase treatment if phosphorylation may contribute to band shifts
Weak signal from hypusine-specific antibodies:
Problem: Limited hypusination or technical issues
Solutions:
Inconsistent immunofluorescence results:
Problem: Variable signal intensity or localization patterns
Solutions:
Standardize fixation time precisely (overfixation can mask epitopes)
Test different permeabilization reagents and conditions
For hypusine-specific antibodies, include proper controls (e.g., GC7-treated cells)
Use confocal microscopy with Z-stacking to assess true signal distribution
Poor reproducibility between experiments:
Problem: Variable results across repeated experiments
Solutions:
Standardize lysate preparation methods rigorously
Prepare larger antibody aliquots to reduce freeze-thaw cycles
Include appropriate loading controls and normalize signal across blots
Consider internal standards (e.g., recombinant eIF5A) for quantitative comparisons
For hypusine-specific antibodies like IU-88, remember that cell type-specific factors (such as DHS levels) can significantly impact results, requiring customized optimization for each experimental system .
When facing contradictory results between different detection methods using eIF5A antibodies, follow this systematic approach to interpretation:
Understand epitope-specific differences:
Application-specific considerations:
Western blot: Denaturating conditions expose all epitopes but may destroy conformation-dependent epitopes
IHC/IF: Fixation and antigen retrieval can differentially affect epitope accessibility
Flow cytometry: Measures intact cells, potentially limiting access to intracellular epitopes
Methodological approach to resolving contradictions:
Systematic validation: Test multiple antibodies targeting different epitopes
Genetic controls: Use siRNA knockdown or overexpression systems
Orthogonal techniques: Confirm findings using non-antibody methods (e.g., mass spectrometry)
Control for hypusination status: Compare GC7-treated (hypusination inhibited) versus untreated samples
Experimental design to resolve conflicts:
Sequential application: Apply multiple detection methods to the same sample
Parallel controls: Include positive and negative controls in each method
Titration experiments: Test multiple antibody concentrations to rule out saturation effects
Cross-validation: Have different laboratory members repeat critical experiments
Biological interpretation framework:
When reporting contradictory results, transparently describe all methods, antibodies used, and potential limitations. This approach not only enhances scientific rigor but may uncover previously unknown aspects of eIF5A biology.
Quantitative analysis of eIF5A and its hypusinated form requires rigorous methodology to ensure accuracy and reproducibility:
Western blot quantification best practices:
Sample preparation standardization:
Consistent lysis buffer composition and protein extraction protocol
Precise protein quantification (duplicate measurements)
Equal loading verified by total protein staining (REVERT, Ponceau S)
Technical considerations:
Use mid-range exposures avoiding saturation
Include calibration curves with recombinant protein when possible
Apply appropriate normalization (total protein preferred over single housekeeping proteins)
Analysis methodology:
Calculate hypusine/total eIF5A ratio to assess modification efficiency
Use triplicate biological samples minimum
Apply appropriate statistical tests for comparisons
Immunofluorescence quantification approaches:
Image acquisition standards:
Fixed exposure settings across all comparable samples
Z-stack acquisition for accurate signal integration
Multichannel imaging with appropriate controls for bleed-through
Analysis methods:
Single-cell analysis rather than field averages when possible
Automated segmentation of subcellular compartments
Colocalization analysis for functional studies
Quantitative parameters:
Mean fluorescence intensity
Nuclear/cytoplasmic signal ratio
Correlation with functional markers
Flow cytometry analysis:
Standardization:
Use calibration beads for consistent instrument settings
Include fluorescence-minus-one (FMO) controls
Gating strategy:
Exclude cell doublets and debris
Subpopulation analysis when relevant
Parameters to report:
Median fluorescence intensity
Percent positive cells
Ratio of hypusinated/total eIF5A
Normalization strategies:
For Western blot:
Total protein normalization (preferred)
Multiple housekeeping proteins if total protein staining is unavailable
For cellular imaging:
Cell area or volume normalization
Nuclear counterstain as reference
For multiple experiments:
Include common reference sample across experiments
Report relative rather than absolute values when combining experiments
Reporting standards:
Clearly state quantification method, software used, and statistical approach
Include representative images with scale bars
Present both raw data and normalized results when possible
Report both technical and biological replicate numbers
When specifically quantifying hypusinated eIF5A, always include appropriate controls (GC7-treated samples, K50A mutants) to validate the specificity of the signal being quantified .
Cutting-edge research is employing eIF5A antibodies to uncover its selective functions in mRNA translation through several sophisticated approaches:
Translational Complex Immunoprecipitation (IP) Methods:
Polysome IP: Using eIF5A antibodies to pull down polysome-associated complexes followed by RNA-seq to identify mRNAs whose translation depends on eIF5A
Cross-linking and Immunoprecipitation (CLIP): UV cross-linking RNA-protein complexes before IP with eIF5A antibodies to identify directly bound mRNAs
Proximity-dependent biotinylation: Expressing BioID-eIF5A fusion proteins to identify proteins in close proximity to eIF5A during translation
Differential Analysis Approaches:
Comparing translation efficiency of mRNAs (by ribosome profiling) in the presence and absence of hypusinated eIF5A (using GC7 treatment)
Using antibodies against both total and hypusinated eIF5A to fractionate cellular lysates and analyze associated mRNAs
Correlating changes in protein synthesis (by pulse labeling) with hypusination levels detected by antibodies like IU-88
Structural and Functional Studies:
Using eIF5A antibodies in structural studies (e.g., cryo-EM) to visualize eIF5A's position in translation complexes
Combining antibody-based detection with site-specific labeling techniques to track eIF5A movement during translation elongation
Employing hypusine-specific antibodies to assess how post-translational modifications affect eIF5A's interactions with the ribosome and other factors
Disease-relevant Translation Regulation:
Investigating how viral infections impact eIF5A's role in translation using both total and hypusine-specific antibodies
Examining altered translation of specific mRNAs in cancer models where eIF5A or eIF5A2 is overexpressed
Correlating stress-induced changes in hypusination with preferential translation of stress-response mRNAs
These approaches are revealing that eIF5A appears to preferentially facilitate the translation of mRNAs containing consecutive proline codons or other challenging sequence motifs, highlighting its specialized role in translation elongation rather than initiation.
Researchers are implementing innovative techniques to study the full spectrum of eIF5A post-translational modifications:
Mass Spectrometry-based Approaches:
Top-down proteomics: Analyzing intact eIF5A protein to maintain all modification relationships
Middle-down proteomics: Limited proteolysis followed by MS analysis to better characterize hypusination in context with other modifications
Parallel reaction monitoring (PRM): Targeted MS approach to quantify specific modified forms of eIF5A
Antibody-enrichment MS: Using eIF5A antibodies to enrich the protein before MS analysis for increased sensitivity
Antibody-based Multi-modification Detection:
Sequential immunoprecipitation: Using hypusine-specific antibodies for first IP, followed by enrichment with antibodies against other modifications
Multiplex immunofluorescence: Simultaneous detection of hypusinated eIF5A with other PTM-specific antibodies
Proximity ligation assays: Detecting co-occurrence of hypusination with other modifications on the same eIF5A molecule
Genetic and Chemical Biology Approaches:
Site-specific incorporation of PTM mimetics: Using amber suppression technology to introduce PTM-mimicking amino acids
Development of antibodies against multiple eIF5A modifications: Beyond hypusine, antibodies targeting phosphorylation, acetylation, or ubiquitination sites
Chemical probes: Small molecules that selectively bind modified forms of eIF5A for visualization or pull-down
Functional Correlation Methods:
FRET/BRET biosensors: Engineered eIF5A constructs that report on conformational changes associated with specific modifications
Real-time tracking: Following PTM dynamics using antibodies conjugated to quantum dots or other stable fluorophores
Modification-specific interactome analysis: Using antibodies to isolate differently modified eIF5A pools and identify differential protein interactions
These emerging techniques are revealing that eIF5A undergoes several modifications beyond hypusination, including phosphorylation, acetylation, and potentially ubiquitination, which may fine-tune its function in different cellular contexts and create a complex "PTM code" that regulates its activity.
Integration of eIF5A antibody-based detection with multi-omics approaches enables comprehensive systems-level understanding:
Integrative Multi-omics Frameworks:
Antibody-based proteomics + Transcriptomics: Correlate eIF5A protein levels/modifications with mRNA expression profiles
Translatomics integration: Combine ribosome profiling data with eIF5A and hypusinated eIF5A immunoprecipitation to identify translationally regulated targets
Antibody-based spatial proteomics + Metabolomics: Map the subcellular distribution of eIF5A in relation to metabolic activity using multiplexed imaging and metabolite profiling
Methodological Integration Strategies:
Sequential analysis pipeline:
Antibody-based FACS sorting of cells based on eIF5A modification status
Subsequent multi-omics analysis of sorted populations (transcriptomics, proteomics, metabolomics)
Parallel multi-modal data collection:
Simultaneous quantification of eIF5A modifications and global protein synthesis rates
Correlation with transcriptome and translatome data from the same samples
Computational Analysis Approaches:
Network analysis: Placing eIF5A within protein-protein interaction networks using antibody-based interactome data
Integrative visualization tools: Developing dashboards that display eIF5A modification status alongside transcriptomic and proteomic changes
Machine learning models: Training algorithms to predict cellular outcomes based on eIF5A modification patterns detected by specific antibodies
Temporal and Perturbation-based Integration:
Time-course studies: Following eIF5A modifications using antibodies while simultaneously tracking transcriptome and proteome changes
Perturbation analysis: Systematic inhibition of hypusination (using GC7) or eIF5A depletion combined with multi-omics profiling
Drug response profiling: Using hypusine-specific antibodies to monitor eIF5A modification alongside transcriptional and translational responses to therapeutic agents
Single-cell Multi-modal Analysis:
Single-cell antibody-based detection: Using hypusine-specific antibodies in single-cell proteomics workflows
Multi-modal single-cell analysis: Combining antibody-based detection of eIF5A with single-cell transcriptomics or metabolomics
Spatial transcriptomics integration: Correlating spatial distribution of modified eIF5A with spatially resolved transcriptomes
These integrative approaches provide a holistic view of how eIF5A and its post-translational modifications function within the broader cellular context, revealing its role in coordinating translational responses to various stimuli and how its dysregulation contributes to disease states.
Eukaryotic Translation Initiation Factor 5A (eIF5A) is a highly conserved protein found in eukaryotic cells and archaea. It plays a crucial role in the initiation of translation, a fundamental process in protein synthesis. eIF5A is unique due to its post-translational modification, known as hypusination, which is essential for its activity .
eIF5A is a small, acidic protein with a molecular weight of approximately 17 kDa and consists of 157 amino acids. It is highly conserved across species, including plants, indicating its vital physiological role. This conservation suggests that eIF5A has been maintained throughout evolution due to its essential functions .
eIF5A is involved in various cellular processes, including:
eIF5A has been studied extensively for its role in various physiological processes and diseases:
Mouse anti-human eIF5A antibodies are used in research to study the protein’s function and its role in various diseases. These antibodies are crucial for detecting and quantifying eIF5A in different experimental settings, aiding in the understanding of its physiological and pathological roles.