EIF2AK2 plays a critical role in:
Antiviral defense: Detects dsRNA during viral replication, triggering translation shutdown and apoptosis in infected cells .
Cellular homeostasis: Modulates mRNA translation under stress conditions, such as nutrient deprivation or oxidative damage .
Cancer biology: Elevated expression correlates with immune cell infiltration and prognosis in pancreatic cancer, suggesting its potential as a diagnostic marker .
A 2023 study using unconjugated EIF2AK2 antibodies identified:
Prognostic significance: High EIF2AK2 expression in tumor tissues associated with improved survival in pancreatic ductal adenocarcinoma (PDAC) patients (p < 0.05) .
Functional validation: Knockdown of EIF2AK2 in PANC-1 cells reduced cell proliferation and migration (CCK-8 assay, p < 0.01) .
Cross-reactivity: Antibodies from Proteintech (e.g., 18244-1-AP) show reactivity across human, mouse, and rat samples, with no cross-reactivity reported for other kinases .
Optimization: Dilution ranges must be titrated for each experimental system, as recommended in manufacturer protocols .
EIF2AK2, also known as PKR (Protein Kinase R), is a serine/threonine protein kinase activated by autophosphorylation after binding to double-stranded RNA. This 62 kDa protein (calculated MW) typically appears as 65-74 kDa bands in Western blots due to post-translational modifications .
EIF2AK2 functions by phosphorylating translation initiation factor EIF2S1, inhibiting protein synthesis in response to various cellular stresses. Its biological significance spans multiple areas:
For research applications, HRP-conjugated EIF2AK2 antibodies offer direct enzymatic detection without secondary antibodies, enabling more consistent results when analyzing expression patterns across different experimental conditions.
Selection should be methodologically driven by your experimental goals:
Methodological approach:
Examine validation data (specificity verified with knockout cells is ideal)
Review published applications in similar experimental systems
Test multiple antibodies targeting different epitopes when possible
For phosphorylation studies, select antibodies specifically validated for distinguishing activation states
Sample preparation significantly impacts antibody performance. The following methodological approaches optimize detection:
For Western Blot samples:
Cell lysis buffer selection:
RIPA buffer with phosphatase inhibitors preserves phosphorylation status
Add protease inhibitors to prevent degradation (particularly important for EIF2AK2)
Include 1-5 mM DTT as EIF2AK2 contains multiple cysteine residues
Protein quantification and loading:
SDS-PAGE conditions:
8-10% gels provide optimal resolution around 62-74 kDa
Complete transfer to membrane at 30V overnight (or 100V for 1 hour) for proteins >60 kDa
For Immunohistochemistry:
Tissue preparation:
Staining controls:
EIF2AK2 frequently shows discrepancies between calculated (62 kDa) and observed (65-74 kDa) molecular weights. Understanding these variations is critical for accurate interpretation:
Molecular weight interpretation guide:
Methodologically, verify these interpretations by:
Treating lysates with lambda phosphatase to eliminate phosphorylation-dependent bands
Stimulating cells with IFN-α or poly(I:C) to enrich activated forms
Running gradient gels for better resolution of closely migrating forms
EIF2AK2 exhibits complex, context-dependent roles in cancer. Recent research shows it may function as either tumor suppressor or oncogene depending on cancer type . For pancreatic cancer specifically, EIF2AK2 expression is significantly higher in tumor tissues than adjacent normal tissue and correlates with immune infiltration .
Methodological approach for cancer studies:
Expression analysis across cancer types:
Correlation with clinical outcomes:
Immune infiltration analysis:
Investigate correlations with immune cell populations using ssGSEA
Analyze relationship with immune checkpoints:
| Immune Checkpoint | Correlation with EIF2AK2 | Statistical Significance |
|---|---|---|
| CD274 (PD-L1) | Strong positive (rs=0.601) | P<0.001 |
| PDCD1LG2 | Strong positive (rs=0.501) | P<0.001 |
| HAVCR2 | Moderate positive (rs=0.423) | P<0.001 |
| TIGIT | Moderate positive (rs=0.341) | P<0.001 |
| CTLA4 | Moderate positive (rs=0.311) | P<0.001 |
| PDCD1 | Weak positive (rs=0.221) | P=0.003 |
| LAG3 | Weak positive (rs=0.207) | P=0.006 |
Functional validation:
Knockdown/knockout EIF2AK2 in cancer cell lines
Evaluate effects on proliferation, migration, and response to therapy
Analyze changes in immune checkpoint expression after EIF2AK2 modulation
EIF2AK2 activation involves a complex series of phosphorylation events. Recent research has identified novel regulatory phosphorylation sites that significantly impact activity .
Key phosphorylation sites and their functions:
Primary activation sites:
Thr446 and Thr451: Critical for kinase activation following dimerization
Phospho-specific antibodies targeting these sites indicate active EIF2AK2
Newly discovered regulatory sites:
Ser6: Located 3 amino acids upstream of DRBM1
Ser97: Located in same position relative to DRBM2
Regulatory mechanisms:
| Phosphorylation Site | Effect When Phosphorylated | Effect When Mutated to Alanine |
|---|---|---|
| Ser6/Ser97 | Maintains inactive conformation | Spontaneous activation |
| Thr446/Thr451 | Required for kinase activity | Prevents activation |
Methodological approach with phospho-specific antibodies:
Activation state assessment:
Use phospho-Thr446/Thr451 antibodies to quantify active EIF2AK2
Normalize to total EIF2AK2 to calculate activation percentage
Regulatory phosphorylation analysis:
Develop antibodies targeting newly identified Ser6/Ser97 sites
Monitor changes in regulatory phosphorylation under different stimuli
Mutational analysis validation:
Generate phosphomimetic (S→D) and phospho-dead (S→A) mutants
Compare antibody reactivity with wild-type and mutant proteins
Correlate phosphorylation patterns with functional outcomes
When interpreting results, remember that negative charges at positions 6 and 97 appear to tighten interactions between RNA-binding motifs and kinase domain, maintaining EIF2AK2 in an inactive conformation even in the presence of dsRNA .
EIF2AK2 plays a critical role in antiviral immunity, making it an important target for infectious disease research. The protein is activated by viral dsRNA and inhibits viral protein synthesis through eIF2α phosphorylation.
Methodological approach for viral infection studies:
Expression and activation monitoring:
Track total EIF2AK2 levels during infection time course
Monitor phosphorylation status using phospho-specific antibodies
Correlate with viral replication markers
Intervention studies:
Compare wild-type vs. EIF2AK2-deficient cells
Assess viral replication efficiency
Measure interferon response pathways
Interferon stimulated gene (ISG) analysis:
Recent findings show EIF2AK2 regulates ISG expression through mRNA splicing :
| ISG | Regulation by EIF2AK2 | Verification Method |
|---|---|---|
| Mx1 | Positive regulation through splicing | qRT-PCR of specific variants |
| OAS1 | Positive regulation through splicing | Western blot protein detection |
| PKR (auto-regulation) | Self-regulation through splicing | Expression rescue experiments |
Mechanistic investigations:
When designing experiments, consider that IFN-α treatment increases EIF2AK2 expression, making it a useful positive control for antibody validation .
Recent discoveries have linked EIF2AK2 missense variants with early onset generalized dystonia (DYT33) , opening new research directions in neurological disorders.
Key research strategies:
Genetic variant analysis:
Screen for EIF2AK2 variants in dystonia patients
Focus on variants identified in previous studies:
c.388G>A (p.Gly130Arg): Found in multiple families, including a de novo case
c.413G>C (p.Gly138Ala): Different heterozygous variant
c.95A>C (p.Asn32Thr): Homozygous variant in consanguineous family
Functional characterization:
Express variant proteins in cellular models
Compare phosphorylation patterns with wild-type EIF2AK2
Assess kinase activity toward eIF2α
Splicing analysis:
Examine potential splicing effects of variants near exon-intron boundaries
Perform cDNA studies to detect aberrant splicing products
Use minigene assays to confirm variant effects on splicing
Neuronal model systems:
Develop neuronal models expressing EIF2AK2 variants
Assess impact on protein synthesis regulation
Investigate effects on neuronal morphology and function
When using HRP-conjugated antibodies in these studies, ensure they can detect the variant proteins equally well as wild-type, possibly by targeting conserved epitopes away from the mutation sites.
When facing detection challenges with EIF2AK2 antibodies, a systematic troubleshooting approach is essential:
Signal enhancement methods:
Sample preparation optimization:
Increase protein loading (up to 25-30 μg per lane)
Add phosphatase inhibitors to preserve phosphorylated forms
Use fresh lysates to minimize degradation
Antibody conditions adjustment:
Reduce antibody dilution (start with 1:500 for weak signals)
Extend incubation time (overnight at 4°C instead of 1-2 hours at RT)
Switch to more sensitive detection substrates (enhanced ECL)
Signal amplification techniques:
For HRP-conjugated antibodies: Use high-sensitivity substrates with longer exposure times
Consider tyramide signal amplification for IHC/IF applications
Try reduced-size format blots to concentrate protein
Expression enhancement:
Decision matrix for troubleshooting weak signals:
| Observation | Likely Cause | Solution |
|---|---|---|
| Signal in positive control only | Low expression in sample | Use more sensitive detection or increase protein amount |
| No signal in any sample | Antibody or detection issue | Test antibody with verified positive control |
| Signal at wrong molecular weight | Non-specific binding | Increase stringency of washes; use different antibody |
| Weak signal with high background | Suboptimal blocking | Increase blocking time/concentration; try different blocking agent |
HRP-conjugated antibodies require specific optimization strategies for maximum sensitivity and specificity:
Optimized Western blot protocol:
Membrane preparation:
Antibody incubation:
Dilute HRP-conjugated EIF2AK2 antibody in fresh blocking buffer
Start with manufacturer's recommended dilution (typically 1:1000-1:2000)
Incubate 1 hour at room temperature or overnight at 4°C with gentle agitation
Avoid sodium azide in antibody dilution buffer as it inhibits HRP
Washing optimization:
Perform 5-6 washes with TBST, 5 minutes each
Increase wash volume to at least 4 mL per cm² of membrane
Use fresh wash buffer for each wash
Detection conditions:
Validation controls:
Include wild-type and EIF2AK2 knockout cell lysates (e.g., A549 or HeLa KO lines)
Use GAPDH (36 kDa) as loading control to verify equal protein loading
Thorough validation is essential before using any new EIF2AK2 antibody for research applications. The following methodological approach ensures reliable results:
Comprehensive validation strategy:
Specificity verification:
Functional validation:
Application-specific testing:
HRP conjugation-specific tests:
Verify HRP activity with direct substrate test
Compare signal-to-noise ratio with unconjugated primary + HRP-secondary approach
Assess stability over time with repeated testing of the same antibody lot
Literature cross-validation:
EIF2AK2 undergoes multiple post-translational modifications (PTMs) that significantly impact its detection:
Major PTMs affecting detection:
Phosphorylation:
Other potential modifications:
Ubiquitination: May create higher MW bands
SUMOylation: Can alter protein migration
Acetylation: May affect antibody binding to specific epitopes
Experimental strategies to address PTM-related issues:
PTM-specific analysis:
Use phospho-specific antibodies to detect activation status
Employ phosphatase treatment to confirm phosphorylation-dependent bands
Run samples under conditions that preserve or remove specific modifications
Protocol adjustments:
| PTM Consideration | Methodological Adjustment | Rationale |
|---|---|---|
| Phosphorylation | Add phosphatase inhibitors to lysis buffer | Preserves phosphorylation status |
| Include phosphorylated controls | Positive control for activation | |
| Degradation | Add protease inhibitors | Prevents protein degradation |
| Multiple forms | Use gradient gels (4-15%) | Better separation of different forms |
| Activation analysis | Compare resting vs. stimulated cells | Demonstrates dynamic changes |
Visualization strategies:
For total EIF2AK2: Use antibodies targeting conserved regions away from PTM sites
For activation studies: Compare phospho-specific to total EIF2AK2 signal
For PTM mapping: Consider 2D gel electrophoresis followed by Western blot
Stimulation protocols for validation:
Recent research has revealed EIF2AK2's involvement in regulating gene expression through mRNA splicing, particularly for interferon-stimulated genes (ISGs) . This opens new research directions requiring specialized experimental approaches:
Comprehensive experimental design:
Gene expression analysis:
Splicing event characterization:
Perform RT-PCR with primers spanning potential splice junctions
Use exon-specific primers to quantify individual splicing events
Design primers for specific transcript variants identified in RNA-seq
Functional validation:
Interaction studies with splicing machinery:
When using HRP-conjugated EIF2AK2 antibodies in these studies, ensure specificity by including appropriate knockout controls and comparing results with unconjugated antibodies to rule out any HRP-related artifacts.
Multiplexed detection allows simultaneous analysis of EIF2AK2 and related proteins, providing comprehensive pathway insights:
Multiplexed analysis strategies:
Multi-color Western blotting:
Combine HRP-conjugated EIF2AK2 antibody with differently labeled antibodies
Detect total and phospho-EIF2AK2 simultaneously
Include downstream markers (p-eIF2α, ATF4, CHOP)
Pathway activation profiling:
| Target | Significance | Expected Pattern in Activation |
|---|---|---|
| EIF2AK2 (total) | Expression level | Increased after IFN treatment |
| p-EIF2AK2 (T446/T451) | Activation marker | Rapidly increased after dsRNA exposure |
| p-eIF2α (S51) | Downstream effect | Follows p-EIF2AK2 with slight delay |
| ATF4 | Integrated stress response | Increased translation despite global inhibition |
| CHOP | Terminal stress response | Increased in prolonged activation |
Multi-parameter flow cytometry:
Permeabilize cells to detect intracellular EIF2AK2
Combine with surface markers to identify responsive cell populations
Include viability markers to distinguish stress responses
Tissue microarray analysis:
When designing multiplexed assays with HRP-conjugated antibodies, careful planning is needed to avoid signal overlap. Consider sequential detection protocols or use HRP inactivation between detection steps.
Studying EIF2AK2 activation dynamics requires specialized techniques to capture real-time changes:
Live cell imaging methodologies:
Fluorescent reporter systems:
Generate EIF2AK2-fluorescent protein fusion constructs
Create phospho-specific biosensors based on FRET technology
Use destabilized fluorescent proteins under control of ATF4 regulatory elements
Activation dynamics monitoring:
| Phenomenon | Measurement Approach | Expected Timeline |
|---|---|---|
| Dimerization | FRET or BiFC between tagged EIF2AK2 molecules | Minutes after dsRNA exposure |
| Phosphorylation | Phospho-specific biosensors | 15-30 minutes after activation |
| Translational inhibition | Fluorescent translation reporters | 30-60 minutes after activation |
| Stress granule formation | Co-localization with G3BP1 markers | 1-2 hours after sustained activation |
Advanced microscopy techniques:
Use confocal microscopy to track subcellular localization changes
Employ FRAP (Fluorescence Recovery After Photobleaching) to measure mobility changes upon activation
Implement light-sheet microscopy for 3D visualization of activation patterns
Quantitative analysis methods:
Track intensity changes in specific cellular compartments
Measure nuclear-cytoplasmic ratios over time
Quantify stress granule formation and composition
While HRP-conjugated antibodies aren't directly applicable to live cell imaging, fixed-cell validation experiments using these antibodies can confirm the specificity of fluorescent tags and biosensors.
The search results reveal apparently contradictory roles for EIF2AK2 across different diseases and cellular contexts . Resolving these contradictions requires careful methodological consideration:
Framework for reconciling contradictory findings:
Context-dependent function analysis:
Tissue-specific effects:
Pathway interaction considerations:
Experimental approach harmonization:
Use consistent cell models across studies
Apply multiple methodologies to verify findings
Consider kinetics and dose-response relationships
Distinguish between correlation and causation through mechanistic studies
When conducting your own research on EIF2AK2, clearly define the cellular context, document all experimental conditions, and employ multiple complementary methods to build confidence in your findings.
Robust EIF2AK2 research requires carefully selected controls and reference materials:
Essential reference materials:
Cell line controls:
Activation controls:
Technical controls:
Reference datasets:
Different tissue types require specific optimization strategies for reliable EIF2AK2 detection:
Tissue-specific optimization guidelines:
Formalin-fixed paraffin-embedded (FFPE) tissues:
Frozen tissue sections:
Fixation: 4% paraformaldehyde for 10 minutes
Permeabilization: 0.1-0.5% Triton X-100 for intracellular access
Blocking: 1-2 hours with 5-10% normal serum
Higher antibody dilutions (1:200-1:500) may be effective
Tissue-specific considerations:
| Tissue Type | Special Considerations | Protocol Adjustments |
|---|---|---|
| Pancreatic tissue | High endogenous peroxidase | Extended peroxidase blocking (15 min) |
| High background with milk | Use BSA or commercial blockers | |
| Brain tissue | Lipid-rich environment | Add 0.1% Tween-20 to antibody diluent |
| Autofluorescence | Treat with Sudan Black B before antibody | |
| Tumor tissues | Variable expression | Include internal controls within section |
| Heterogeneous expression | Analyze multiple fields (≥5) |
Tissue microarrays (TMAs):
Single standardized protocol applies to all samples
Include known positive and negative controls in each TMA
Score intensity using standardized scales (0-3+)
Consider digital image analysis for quantification
For all tissue preparations, validate staining patterns with multiple antibodies and correlate with mRNA expression data when available.
Accurate quantification is essential for meaningful EIF2AK2 research. Multiple approaches offer complementary advantages:
Comprehensive quantification methods:
Western blot quantification:
Densitometric analysis guidelines:
| Measurement | Calculation Method | Application |
|---|---|---|
| Total EIF2AK2 | EIF2AK2 band intensity / GAPDH intensity | Expression level changes |
| Activation ratio | Phospho-EIF2AK2 / Total EIF2AK2 | Activation state analysis |
| Relative expression | Sample intensity / Reference sample intensity | Cross-comparison between conditions |
Flow cytometry quantification:
Use geometric mean fluorescence intensity (gMFI)
Include isotype controls to set negative gates
Calculate stain index: (Sample MFI - Background MFI) / 2× SD of background
Compare with standard curves for absolute quantification
Real-time PCR correlation:
Primer design for specific EIF2AK2 transcript variants
Normalize to validated reference genes
Calculate relative expression using 2^(-ΔΔCt) method
Correlate mRNA with protein levels to assess post-transcriptional regulation
Digital pathology approaches:
Whole slide scanning of immunohistochemistry
Automated detection of positive cells
Intensity scoring (0, 1+, 2+, 3+)
Both percentage of positive cells and intensity should be reported
When quantifying HRP-conjugated antibody signals, ensure substrate isn't depleted (non-linear range) by testing multiple exposure times or dilution series.