EIF2AK2 is a serine/threonine kinase activated by double-stranded RNA (dsRNA), playing pivotal roles in:
Antiviral Defense: Phosphorylates eIF2α to inhibit viral protein synthesis .
Tumor Regulation: Exhibits dual roles in cancer—suppressing metastasis in breast and lung cancers while promoting progression in hepatocellular carcinoma .
Immune Modulation: Activates NF-κB via IκB phosphorylation, enhancing interferon-β production .
The FITC conjugate enables precise subcellular localization:
Detects EIF2AK2 in HeLa cells with high signal-to-noise ratios .
Validated in pancreatic cancer studies, where elevated EIF2AK2 correlates with tumor progression and immune infiltration .
Pancreatic Cancer: Overexpression in tumor vs. normal tissues (validated via IHC and RNA sequencing) .
Immune Microenvironment: EIF2AK2 levels correlate with CD8+ T-cell infiltration, suggesting immune-modulatory roles .
Sensitivity: Detects endogenous EIF2AK2 at low concentrations (WB dilution up to 1:50,000) .
Versatility: Compatible with formaldehyde-fixed and frozen sections .
Specificity: No cross-reactivity reported with non-target kinases in validation assays .
EIF2AK2, also known as PKR (Protein Kinase R), is a eukaryotic translation initiation factor 2-alpha kinase that plays a crucial role in cellular stress responses. It functions as a serine/threonine protein kinase and is expressed in over 10 different cell types. EIF2AK2 is activated by various cellular stresses including viral infections, hypoxia, and nutritional shortages . The protein has a length of 551 amino acid residues and a molecular mass of 62.1 kDa in humans . Its significance in research stems from its dual role in both innate immune responses to viral infection and its complex involvement in cancer biology, making it a valuable target for immunological and oncological investigations .
EIF2AK2 Antibody, FITC conjugated is primarily designed for fluorescence-based applications due to the fluorescein isothiocyanate (FITC) conjugation. The most suitable applications include:
Immunofluorescence (IF) microscopy, particularly for fixed tissue sections (IHC-P) at recommended dilutions of 1:50-200
Flow cytometry (FACS) for analyzing EIF2AK2 expression in single-cell suspensions
Immunocytochemistry (ICC) for detecting the protein in cultured cells, as validated in HeLa cells
These applications allow researchers to visualize EIF2AK2 localization and expression patterns within cellular contexts without requiring secondary antibody incubation steps.
For rigorous experimental design when using EIF2AK2 Antibody, FITC conjugated, the following controls should be implemented:
Implementing these controls will significantly enhance data reliability and facilitate accurate interpretation of experimental results.
Optimizing sample preparation for detecting EIF2AK2 in pancreatic cancer tissues requires special consideration due to the unique characteristics of pancreatic tissue. Based on research findings:
Fixation Protocol: Use 10% neutral buffered formalin for 24-48 hours to preserve protein structure while maintaining tissue morphology. Extended fixation should be avoided as it may mask EIF2AK2 epitopes.
Antigen Retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0) for 20 minutes has shown optimal results for EIF2AK2 detection. This step is crucial as research has demonstrated significantly higher EIF2AK2 expression in pancreatic cancer tissues compared to adjacent normal tissues .
Section Thickness: 4-5 μm sections provide optimal resolution for distinguishing subcellular localization (nuclear vs. cytoplasmic), which is important since EIF2AK2 can be found in both compartments .
Blocking Protocol: Use 5% normal goat serum with 0.1% Triton X-100 for 1 hour to reduce non-specific binding while allowing antibody penetration.
Incubation Conditions: For FITC-conjugated antibodies, overnight incubation at 4°C in a humidified chamber protected from light yields the most consistent staining patterns.
When comparing staining intensity between cancerous and normal pancreatic tissue, researchers should implement quantitative image analysis to objectively measure fluorescence intensity differences that correlate with clinical outcomes .
Research has revealed contradictory roles for EIF2AK2 across different cancer types, functioning as both a tumor suppressor and an oncogene depending on the cellular context . To resolve these contradictions:
Multi-modal verification: Combine FITC-conjugated antibody immunofluorescence with orthogonal techniques such as RT-qPCR, western blotting, and RNAscope to verify expression levels through independent methodologies.
Cell type-specific analysis: Implement multiplexed immunofluorescence using the FITC-conjugated EIF2AK2 antibody alongside cell-type-specific markers to determine if expression patterns differ among various cellular components within heterogeneous tumor samples.
Functional validation: Combine expression data with functional assays that measure EIF2AK2 kinase activity rather than merely protein levels, as post-translational modifications may affect function without altering expression.
Contextual analysis: Evaluate EIF2AK2 expression in relation to:
Tumor microenvironment composition
Hypoxic conditions (measured by HIF-1α co-staining)
Inflammatory markers
Cellular stress indicators
Experimental models: When comparing results between studies, consider differences in:
Primary tumors versus established cell lines
Patient-derived xenografts versus genetically engineered mouse models
2D versus 3D culture systems
This integrated approach can help reconcile seemingly contradictory findings about EIF2AK2's role in different cancer types and provide insight into its context-dependent functions .
FITC is particularly susceptible to photobleaching, which can compromise data collection in extended imaging sessions. Researchers should implement the following strategies:
Antifade reagent optimization: Use antifade mounting media containing:
p-phenylenediamine at 1 mg/mL
ProLong Gold with specific pH adjustment (8.0-8.5)
Proprietary commercially available solutions optimized for FITC
Imaging parameters optimization:
Reduce exposure time and laser power to minimum effective levels
Utilize interval scanning rather than continuous illumination
Employ confocal aperture adjustment to minimize out-of-focus light exposure
Oxygen scavenging systems: Incorporate enzymatic oxygen scavenging components in live-cell imaging buffers:
Glucose oxidase (0.5 mg/mL) with catalase (40 μg/mL)
Glucose (10 mM) as substrate
Alternative workflows:
Capture regions of interest first at lower magnification
Reserve high-resolution imaging for pre-selected fields
Consider photoconversion to more stable fluorophores when compatible
Computational approaches:
Implement deconvolution algorithms
Apply photobleaching correction in post-processing
Utilize machine learning-based image restoration
These strategies significantly extend the viable imaging window for detecting EIF2AK2 in both fixed and live-cell applications while maintaining signal integrity.
Research data indicates complex relationships between EIF2AK2 expression and immune cell infiltration in pancreatic cancer:
Functional enrichment analysis of EIF2AK2-associated differentially expressed genes (DEGs) in pancreatic cancer reveals significant correlations with immune cell populations . The table below summarizes key findings:
| Immune Cell Type | Correlation with EIF2AK2 Expression | Functional Implication |
|---|---|---|
| CD8+ T cells | Positive correlation | Enhanced cytotoxic response |
| Natural Killer cells | Moderate positive correlation | Improved innate immune surveillance |
| M1 Macrophages | Strong positive correlation | Pro-inflammatory tumor microenvironment |
| Regulatory T cells | Variable correlation | Context-dependent immunosuppression |
| Neutrophils | Positive correlation | Potential pro-tumorigenic inflammation |
To investigate these correlations using FITC-conjugated EIF2AK2 antibodies, researchers should:
Perform multiplexed immunofluorescence with immune cell markers and EIF2AK2
Quantify spatial relationships between EIF2AK2-expressing tumor cells and immune infiltrates
Correlate findings with clinical outcomes and treatment responses
Consider three-dimensional analysis to assess the architectural relationship between EIF2AK2-positive cells and immune cell niches
These approaches can provide meaningful insights into how EIF2AK2 may modulate the pancreatic tumor immune microenvironment, potentially informing immunotherapeutic strategies .
Distinguishing between active and inactive EIF2AK2 is crucial for understanding its functional state in cancer research applications. The following methodological approaches can be utilized:
Phospho-specific antibody complementation:
Use phospho-specific antibodies (e.g., against Thr446) in multi-color immunofluorescence alongside the FITC-conjugated total EIF2AK2 antibody
Calculate activation ratio by dividing phospho-EIF2AK2 signal by total EIF2AK2 signal
Proximity ligation assay (PLA) adaptation:
Combine FITC-conjugated EIF2AK2 antibody with antibodies against known EIF2AK2 substrates like eIF2α
PLA signal indicates active kinase engaged with its substrate
Conformation-sensitive FRET reporters:
Design FRET-based reporters that change conformation upon EIF2AK2 activation
Co-localize FRET signal with FITC-conjugated EIF2AK2 antibody staining
Activity-based protein profiling:
Use biotinylated ATP mimetics that bind only to active kinase conformations
Detect co-localization with FITC-conjugated EIF2AK2 antibody
Downstream substrate phosphorylation:
Correlate EIF2AK2 expression with phosphorylation of known downstream targets (e.g., eIF2α)
Quantify correlation coefficients to infer activation state
These approaches allow researchers to move beyond simple detection of EIF2AK2 protein to gain functional insights into its activation status within the cellular context.
Non-specific staining can severely compromise data interpretation when using FITC-conjugated EIF2AK2 antibodies. The following systematic troubleshooting approaches address common issues:
Optimal antibody dilution determination:
Enhanced blocking protocols:
Extended blocking (2 hours) with combined blockers:
10% normal serum from the same species as secondary antibody
1% BSA
0.3% Triton X-100
0.05% Tween-20
Consider dual blocking with both normal serum and commercial blocking reagents
Autofluorescence management:
Pre-treatment with 0.1% Sudan Black B in 70% ethanol for 20 minutes
Sodium borohydride treatment (0.1% for 5 minutes) for formalin-fixed tissues
UV irradiation before antibody application
Spectral unmixing during image acquisition
Absorption controls optimization:
Pre-incubate antibody with recombinant EIF2AK2 protein at 5-10 μg/mL
Implement gradient absorption to determine specificity threshold
Sample preparation refinement:
Optimize fixative concentration and duration
Explore cryo-fixation alternatives when applicable
Adjust permeabilization protocols based on subcellular localization
Systematic implementation of these steps can significantly reduce background and improve specific detection of EIF2AK2 in research applications.
Accurate quantitative comparison of EIF2AK2 expression requires methodological standardization and appropriate controls. Here is a comprehensive approach:
Standardization of fluorescence measurements:
Use calibration beads with known fluorescence intensity before each imaging session
Include internal reference standards in each experiment
Maintain identical exposure settings across compared samples
Implement flat-field correction to account for illumination heterogeneity
Quantification methodology:
Mean fluorescence intensity (MFI) measurement in defined regions of interest
Integrated density calculation (area × mean intensity)
Thresholding-based binary quantification
Cell-by-cell analysis when possible
Normalization strategies:
Normalize to housekeeping protein expression
Use ratio to isotype control signal
Employ reference tissues with stable EIF2AK2 expression
Calculate fold-change relative to standardized control samples
Statistical analysis framework:
Determine appropriate sample sizes using power analysis
Apply appropriate statistical tests based on data distribution
Consider nested factors in experimental design
Address multiple testing correction when necessary
Methodological reporting standards:
Document all imaging parameters:
Exposure time
Gain settings
Laser power
Filter specifications
Objective specifications
Report software and algorithms used for quantification
Provide all normalization details
This comprehensive approach enables reliable quantitative comparisons of EIF2AK2 expression across different experimental conditions, ensuring reproducibility and validity of research findings.
Integrating FITC-conjugated EIF2AK2 antibodies with emerging single-cell technologies offers promising avenues for understanding tumor heterogeneity:
Single-cell sorting and analysis pipeline:
Live-cell FACS sorting based on EIF2AK2-FITC signal intensity
Downstream single-cell RNA sequencing of sorted populations
Correlation of protein expression with transcriptomic profiles
CITE-seq adaptation:
Convert FITC-conjugated antibodies to oligonucleotide-tagged formats
Perform simultaneous protein (including EIF2AK2) and RNA detection
Cluster cells based on multi-omic profiles
Spatial transcriptomics integration:
Combine immunofluorescence using FITC-conjugated EIF2AK2 antibodies with spatial transcriptomics platforms
Map EIF2AK2 protein expression to spatially resolved transcriptomes
Analyze spatial relationships between EIF2AK2-high cells and their microenvironment
Mass cytometry complementation:
Develop metal-tagged EIF2AK2 antibodies for CyTOF analysis
Create comprehensive immune-profiling panels including EIF2AK2
Identify rare subpopulations with unique EIF2AK2 expression patterns
Microfluidic-based approaches:
Capture single cells in droplets after EIF2AK2-FITC staining
Perform droplet-based functional assays
Correlate functional outcomes with expression levels
These integrated approaches can reveal how EIF2AK2 expression varies across tumor subpopulations and how this heterogeneity correlates with functional phenotypes and treatment responses.
Understanding EIF2AK2's distinct functions in different compartments of the tumor microenvironment requires sophisticated experimental designs:
Laser capture microdissection workflow:
Stain tissue sections with FITC-conjugated EIF2AK2 antibody
Identify and isolate tumor nests versus stromal regions
Perform downstream molecular analysis (RNA-seq, proteomics) on separated components
Compare EIF2AK2-associated pathways between compartments
Co-culture experimental systems:
Establish co-cultures of tumor cells with stromal components
Differentially label cell populations
Analyze EIF2AK2 expression and activation after various perturbations
Assess intercellular signaling effects on EIF2AK2 activity
Conditional knockout approaches in complex models:
Generate cell-type-specific EIF2AK2 knockout models
Compare phenotypes between epithelial-specific versus stromal-specific deletion
Use FITC-conjugated antibody to confirm knockout efficiency
Extracellular vesicle (EV) transfer studies:
Isolate EVs from EIF2AK2-high versus EIF2AK2-low cells
Track transfer of EVs between tumor and stromal components
Assess EIF2AK2-dependent content and functional consequences
Humanized mouse models:
Reconstitute immunodeficient mice with human immune components
Implant patient-derived tumor organoids
Use species-specific EIF2AK2 antibodies to distinguish host versus tumor expression
Correlate with treatment responses
These experimental designs can provide critical insights into the compartment-specific roles of EIF2AK2 in the complex tumor microenvironment, particularly relevant given its potential role as a diagnostic and prognostic biomarker in pancreatic cancer .