EIF4G2 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Applications

The EIF4G2 Antibody, FITC conjugated is validated for research applications requiring precise detection of eIF4G2 in human samples. Key uses include:

  • Immunofluorescence (IF): Co-localization studies to map eIF4G2 within cellular compartments (e.g., cytosol or translation initiation complexes) .

  • Flow Cytometry (FCM): Quantitative analysis of eIF4G2 expression levels in cell populations .

  • Cell Biology Studies: Investigating eIF4G2’s role in translation regulation, stress responses, or disease pathways .

While the FITC-conjugated variant is not explicitly cited in published studies, other eIF4G2 antibodies (e.g., unconjugated forms) have been used in:

  • Cancer Research: Detecting low eIF4G2 expression in endometrial cancer (EC) tissues, correlating with poor prognosis .

  • Neuropathic Pain Models: Linking eIF4G2 to downregulation of ion channels (e.g., Kv1.2, MOR) in dorsal root ganglion neurons .

Role in Disease Pathology

  • Endometrial Cancer: Low eIF4G2 expression correlates with reduced survival in Grade 2 EC patients, mediated by therapy resistance and stem cell enrichment .

  • Neuropathic Pain: eIF4G2 modulates ion channel expression, contributing to hypersensitivity in chronic pain models .

Mechanistic Insights

  • Translation Regulation: eIF4G2 represses cap-dependent translation while promoting IRES-mediated initiation, critical during stress or mitosis .

  • Interaction Networks: Binds eIF4A and eIF3 to form translationally inactive complexes, opposing the canonical eIF4G1 pathway .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
DAP-5 antibody; DAP5 antibody; Death associated protein 5 antibody; Death-associated protein 5 antibody; eIF-4-gamma 2 antibody; eIF-4G 2 antibody; eIF4G 2 antibody; EIF4G2 antibody; Eukaryotic translation initiation factor 4 gamma 2 antibody; IF4G2_HUMAN antibody; Nat1 antibody; Novel APOBEC-1 target 1 antibody; p97 antibody; Translation repressor NAT1 antibody; Translation repressor NAT1 antibody
Target Names
EIF4G2
Uniprot No.

Target Background

Function
EIF4G2, also known as DAP5, plays a significant role in the transition from cap-dependent to IRES-mediated translation during cellular processes such as mitosis, apoptosis, and viral infection. This protein can be cleaved by certain caspases and viral proteases.
Gene References Into Functions
  1. Studies have shown that miR-379 mimics and EIF4G2 shRNA groups lead to a decrease in the percentage of cells in the S-phase of the cell cycle. PMID: 28381518
  2. Knockdown of DAP5 in human embryonic stem cells (hESCs) resulted in persistent expression of pluripotent genes, delayed induction of differentiation-associated genes in various cell lineages, and impaired embryoid body formation. PMID: 27664238
  3. Research indicates that microRNA miR-379 enhances lung cancer (LCa) chemosensitivity by modulating cisplatin (CDDP)-induced apoptosis through direct targeting of the 3' UTR of eukaryotic translation initiation factor 4 gamma 2 (EIF4G2). PMID: 28117895
  4. These findings suggest that miR-139 inhibits chondrocyte proliferation and migration, potentially serving as a therapeutic target for osteoarthritis (OA). The mechanism of miR-139 in chondrocytes may involve its regulation of EIF4G2 and IGF1R. PMID: 27105918
  5. The Coxsackievirus B3 protease 2A-mediated cleavage of DAP5 leads to the production of two truncated proteins that differentially affect the protein translation of IRES-containing genes, resulting in enhanced host cell death. PMID: 26586572
  6. Knockdown of EIF4G2 replicated the effects of mir-139, while restoring EIF4G2 expression rescued the mir-139 phenotype. Elevated miR-139-5p expression is associated with a favorable outcome in acute myeloid leukemia. PMID: 26165837
  7. These findings provide the first mechanistic insights into DAP5's function as a selective regulator of cap-independent translation. PMID: 25779044
  8. Research demonstrates that the tumor suppressor effect of miR-520c-3p is mediated by repressing translation while inducing senescence, with eIF4GII serving as a key effector of this anti-tumor activity. PMID: 24497838
  9. DAP5, a translation initiation factor known to positively regulate the translation of various internal ribosome entry sites (IRES)-containing mRNAs, promotes IRES-driven translation of p53 mRNA. PMID: 23318444
  10. DAP5/p97 and DAP5/p86 enhance the translation of the anti-apoptotic protein Bcl-2 and inhibit cisplatin-induced apoptosis. PMID: 22555068
  11. Multiple isoforms of eIF4GII arise from alternative promoters and splicing events. A non-canonical CUG initiation codon extends the eIF4GII N-terminus, which plays an alternative role in initiation factor assembly. PMID: 22909319
  12. The crystal structure of the C-terminal region (residues 540-897) of p97/EIF4G2 (at 2.0 Angstrom resolution) shows sequence/structure homology with EIF4G1. PMID: 20544972
  13. Crystallization and preliminary X-ray diffraction analysis of the MIF4G domain of DAP5 have been reported. PMID: 20057060
  14. This includes the functional analysis of a similar protein in yeast. PMID: 11331597
  15. The caspase-cleaved DAP5 protein supports IRES-mediated translation of death proteins. PMID: 11943866
  16. Endogenous eIF4GII is phosphorylated by Ca(2+)/calmodulin-dependent protein kinase I. PMID: 14507913
  17. Cleavage of eIF4GII is induced in cells and cell extracts by the FMDV leader protease (L(pro)) alone. PMID: 15016848
  18. EIF4G2 transcripts exhibit three distinct expression patterns, with downregulation observed in approximately 50% of the cases analyzed. EIF4G2 gene transcription was associated with invasive tumors. PMID: 16410684
  19. p97 differs functionally from the closely related C-terminal two-thirds of eIF4GI and can positively promote protein synthesis and cell proliferation. PMID: 16932749
  20. These findings indicate that full-length p97 is a translational activator with essential roles in unstressed cells, suggesting a reassessment of current models of p97 function. PMID: 17237356
  21. Research demonstrates that DAP5 promotes cap-independent translation of Bcl-2 and CDK1 to facilitate cell survival during mitosis. PMID: 18450493
  22. This research investigates the role and regulation of death-associated protein-5 (DAP5/p97/NAT1), a novel inhibitor of translational initiation, in acute promyelocytic leukemia cell differentiation and apoptosis. PMID: 18491231
  23. Results report the crystal structure of the C-terminal region of DAP5/p97 extending between amino acids 730 and 897. PMID: 18722383
  24. The exclusive use of a GUG translation initiation codon is conserved across human, chicken, Xenopus, and zebrafish. PMID: 15718103
  25. Translation initiation occurs exclusively at a non-AUG translation initiation codon (GUG). PMID: 9049310

Show More

Hide All

Database Links

HGNC: 3297

OMIM: 602325

KEGG: hsa:1982

STRING: 9606.ENSP00000340281

UniGene: Hs.183684

Protein Families
Eukaryotic initiation factor 4G family
Tissue Specificity
Ubiquitously expressed in all adult tissues examined, with high levels in skeletal muscle and heart. Also expressed in fetal brain, lung, liver and kidney.

Q&A

What is EIF4G2 and why is it important in research?

EIF4G2 (eukaryotic translation initiation factor 4 gamma 2) is a crucial protein involved in cap-independent translation initiation and represents a significant area of interest in molecular biology and cellular research. This 102.4 kilodalton protein, also known as DAP5, NAT1, P97, and DAP-5, functions as a scaffolding protein that facilitates the assembly of translation initiation complexes in the absence of the canonical cap-binding eIF4E protein . The significance of EIF4G2 in research stems from its critical role in regulating protein synthesis under various cellular stress conditions when cap-dependent translation is compromised. Researchers investigating cellular stress responses, apoptosis, and cancer biology frequently examine EIF4G2 as it maintains protein synthesis of specific mRNAs containing internal ribosome entry sites (IRES) during cellular stress. The study of EIF4G2 has revealed its involvement in translational control of genes associated with cell survival, proliferation, and programmed cell death, making it a valuable target for understanding disease mechanisms and potential therapeutic interventions .

What experimental applications are suitable for FITC-conjugated EIF4G2 antibodies?

FITC-conjugated EIF4G2 antibodies enable direct visualization of EIF4G2 protein in various experimental applications without requiring secondary antibody detection steps. Immunofluorescence microscopy represents one of the primary applications, allowing researchers to directly visualize the subcellular localization of EIF4G2 within fixed or live cells, providing insights into its distribution patterns under different experimental conditions or cellular states . Flow cytometry applications benefit from FITC-conjugated EIF4G2 antibodies by enabling quantitative analysis of EIF4G2 expression levels across cell populations, particularly useful for examining expression differences between normal and diseased states or following experimental manipulations. Immunocytochemistry (ICC) with these antibodies allows detailed examination of EIF4G2 distribution within cellular compartments, especially valuable for co-localization studies with other proteins involved in translation initiation complexes . Additionally, high-content screening approaches can leverage the direct fluorescence of FITC-conjugated EIF4G2 antibodies for large-scale analysis of EIF4G2 expression or localization changes in response to drug treatments, genetic modifications, or other experimental variables.

How should I validate the specificity of my FITC-conjugated EIF4G2 antibody?

Validating the specificity of FITC-conjugated EIF4G2 antibodies requires a comprehensive approach combining multiple complementary techniques to ensure reliable experimental results. Western blot analysis using the unconjugated version of the same antibody clone should be performed first to confirm that the antibody recognizes a single band of approximately 102.4 kDa, corresponding to the expected molecular weight of EIF4G2 . Peptide competition assays provide another critical validation method, where pre-incubation of the antibody with the immunizing peptide should significantly reduce or eliminate the fluorescent signal, confirming that the observed staining is specific to the EIF4G2 epitope. Knockout or knockdown validation, considered the gold standard for antibody specificity, involves comparing staining patterns between wild-type samples and those where EIF4G2 expression has been eliminated through CRISPR-Cas9 knockout or significantly reduced using siRNA/shRNA . Cross-reactivity testing with related proteins, particularly other eIF4G family members, should be conducted to ensure the antibody does not recognize structurally similar proteins, which can be accomplished by overexpression systems or testing in samples with known expression profiles. Finally, orthogonal detection methods using antibodies targeting different epitopes of EIF4G2 should produce similar staining patterns, further confirming specificity.

What controls should I include when using FITC-conjugated EIF4G2 antibodies?

Implementing appropriate controls is essential for generating reliable and interpretable data when using FITC-conjugated EIF4G2 antibodies in research applications. An isotype control using a FITC-conjugated antibody of the same isotype but with irrelevant specificity allows researchers to assess the level of non-specific binding due to the antibody class rather than actual target recognition . Unstained controls (cells without any antibody treatment) are necessary to establish the baseline autofluorescence of the sample, particularly important when working with tissues or cells with naturally high autofluorescence in the FITC emission spectrum. Secondary antibody-only controls are not needed with direct FITC conjugates, representing an advantage over unconjugated primary antibodies that require labeled secondary detection systems . Positive control samples known to express EIF4G2 at detectable levels (such as HeLa cells) should be included to verify that the staining protocol is functioning correctly and to establish the expected staining pattern. Negative control samples where EIF4G2 expression has been knocked down or is naturally absent provide critical validation that the signal observed depends on the presence of the target protein. Additionally, researchers should include a blocking peptide control where the antibody is pre-incubated with the immunizing peptide to demonstrate that the observed signal is specifically due to recognition of the EIF4G2 epitope.

How should I optimize fixation and permeabilization for FITC-conjugated EIF4G2 antibody staining?

Optimizing fixation and permeabilization conditions is crucial for preserving both antigenicity and structural integrity when using FITC-conjugated EIF4G2 antibodies. Paraformaldehyde fixation (4%) for 15-20 minutes at room temperature generally preserves EIF4G2 epitopes while maintaining cellular architecture, making it the recommended starting point for most applications involving this antibody . Methanol fixation (-20°C for 10 minutes) offers an alternative that may enhance detection of certain EIF4G2 epitopes, particularly those that might be masked in the protein's tertiary structure, though researchers should be aware that this approach can disrupt membrane structures and potentially alter the localization pattern of membrane-associated proteins. For permeabilization, Triton X-100 (0.1-0.5% for 5-10 minutes) effectively facilitates antibody access to intracellular EIF4G2 without excessive protein extraction, while saponin (0.1-0.2%) provides a gentler alternative that may better preserve fine subcellular structures and membrane associations relevant to EIF4G2 function . The optimal fixation-permeabilization protocol should be determined empirically for each cell type or tissue, as variations in membrane composition and protein expression levels can significantly impact accessibility. Importantly, researchers should verify that their chosen protocol does not introduce artifacts by comparing results across multiple fixation methods, particularly when examining EIF4G2 subcellular localization or interactions with translation machinery components.

How can I use FITC-conjugated EIF4G2 antibodies to investigate stress granule formation?

FITC-conjugated EIF4G2 antibodies provide a powerful tool for investigating the dynamics of stress granule formation and EIF4G2's role in stress response pathways. Researchers can implement live-cell imaging protocols using these antibodies with membrane-permeabilizing agents to track the real-time recruitment of EIF4G2 to stress granules following various cellular stressors such as arsenite treatment, heat shock, or oxidative stress . Dual-labeling approaches combining FITC-conjugated EIF4G2 antibodies with far-red labeled stress granule markers (like G3BP1 or TIA-1) allow for precise colocalization analysis without spectral overlap, enabling quantification of the temporal relationship between EIF4G2 recruitment and stress granule assembly. Super-resolution microscopy techniques such as STORM or STED using FITC-conjugated EIF4G2 antibodies can resolve the nanoscale organization of EIF4G2 within stress granules, potentially revealing functional domains or protein interaction networks not visible with conventional microscopy . Fluorescence recovery after photobleaching (FRAP) experiments with these antibodies can measure the mobility and exchange rates of EIF4G2 within stress granules, providing insights into whether EIF4G2 serves as a stable structural component or exhibits dynamic association. Researchers should implement quantitative analysis protocols that measure parameters such as stress granule size, number, EIF4G2 content, and colocalization coefficients with other stress granule components across different time points and stress conditions to fully characterize the relationship between EIF4G2 and stress granule biology.

What are the optimal conditions for detecting low-abundance EIF4G2 using FITC-conjugated antibodies?

Detecting low-abundance EIF4G2 requires specialized protocols that maximize signal-to-noise ratios when using FITC-conjugated antibodies. Signal amplification through tyramide signal amplification (TSA) can be particularly effective, as this approach can increase detection sensitivity by 10-100 fold by depositing multiple fluorophores at the antibody binding site, though researchers must carefully titrate reagents to avoid excessive background . Extended antibody incubation periods (overnight at 4°C rather than standard 1-2 hour protocols) significantly improve detection of low-abundance targets by allowing more complete antibody penetration and epitope binding, particularly important when examining EIF4G2 expression in tissues or cells where it is minimally expressed. Background reduction strategies, including extended blocking steps (2-3 hours) with specialized blocking reagents containing both proteins and non-ionic detergents, effectively minimize non-specific binding of the FITC-conjugated antibody to cellular components . Advanced microscopy approaches such as spectral unmixing can separate true FITC signal from autofluorescence, particularly valuable in tissues with high natural fluorescence. Researchers working with tissues should consider implementing antigen retrieval methods specifically optimized for EIF4G2, such as citrate buffer (pH 6.0) or Tris-EDTA buffer (pH 9.0) heat-induced epitope retrieval, which can significantly improve antibody access to EIF4G2 epitopes that may be masked during fixation. Additionally, researchers should consider using photostable FITC variants or anti-fade mounting media containing radical scavengers to minimize photobleaching during extended imaging sessions necessary for detecting weak signals.

What are the considerations for multiplexing FITC-conjugated EIF4G2 antibodies with other fluorescent probes?

Successful multiplexing of FITC-conjugated EIF4G2 antibodies with other fluorescent probes requires careful consideration of spectral properties and staining protocols to generate interpretable multi-parameter data. Spectral compatibility represents the primary consideration, where FITC (excitation ~495nm, emission ~520nm) should be paired with fluorophores having minimal spectral overlap, such as far-red dyes (Cy5, Alexa 647) or blue fluorophores (DAPI, Hoechst), rather than yellow-orange emitters (PE, TRITC) that could complicate signal separation . Sequential staining protocols often produce cleaner results than simultaneous incubation with multiple antibodies, particularly when combining antibodies from the same host species or when working with samples having high background autofluorescence. Antibody panel validation should be performed by comparing staining patterns of individual antibodies alone versus in combination to ensure that multiplexing does not alter the expected staining pattern for any component, which can occur due to steric hindrance or unexpected cross-reactivity . Signal balancing represents another critical consideration, where researchers should match the intensities of different fluorophores by adjusting antibody concentrations, as vastly different signal intensities can complicate both visual interpretation and computational analysis of colocalization. Advanced spectral unmixing algorithms can be employed when using fluorophores with partial spectral overlap, allowing computational separation of overlapping signals post-acquisition based on the known spectral signatures of each fluorophore.

How can I troubleshoot weak or non-specific staining with FITC-conjugated EIF4G2 antibodies?

Troubleshooting weak or non-specific staining with FITC-conjugated EIF4G2 antibodies requires a systematic approach to identify and address potential technical issues affecting antibody performance. Antibody concentration optimization should be the first step, typically performing a titration series (ranging from 1:50 to 1:1000 dilutions) to identify the optimal concentration that maximizes specific signal while minimizing background, as both insufficient and excessive antibody concentrations can compromise staining quality . Epitope masking during fixation represents a common cause of weak staining, particularly with aldehyde-based fixatives that can form protein cross-links, which can be addressed by implementing epitope retrieval techniques such as heat-induced epitope retrieval with citrate buffer (pH 6.0) or enzymatic retrieval using proteinase K or trypsin at carefully controlled concentrations and incubation times . Storage and handling factors significantly impact antibody performance, as FITC is susceptible to photobleaching and quenching; therefore, researchers should store antibodies protected from light at appropriate temperatures (typically 4°C short-term, -20°C long-term) and minimize freeze-thaw cycles. Non-specific binding can be reduced through implementation of more effective blocking protocols, including extended blocking times (1-2 hours) with species-appropriate normal sera supplemented with BSA, non-fat milk, or commercial blocking reagents, followed by thorough washing steps. When persistent background remains problematic, researchers should consider using competitive binding controls (using the immunizing peptide) to differentiate between specific and non-specific signals, or potentially switching to alternative detection methods such as tyramide signal amplification or quantum dot-based detection systems that may provide improved signal-to-noise characteristics.

How can FITC-conjugated EIF4G2 antibodies be utilized in studying translation regulation during cellular stress?

FITC-conjugated EIF4G2 antibodies offer valuable tools for investigating the complex dynamics of translation regulation during cellular stress responses. Stress-induced relocalization studies can track EIF4G2 movement between different subcellular compartments following exposure to stressors (oxidative agents, ER stress inducers, nutrient deprivation), revealing how its spatial distribution correlates with changes in translation activity as measured by complementary techniques such as polysome profiling or ribosome footprinting . Co-immunoprecipitation followed by fluorescence microscopy allows visualization of stress-dependent changes in EIF4G2's interaction partners, which can be quantified through intensity correlation analysis between FITC-EIF4G2 signals and other labeled components of the translation machinery. Stress-recovery dynamics can be examined through time-lapse imaging with FITC-conjugated EIF4G2 antibodies in permeabilized cells, documenting the kinetics of EIF4G2 recruitment to stress granules during stress exposure and its subsequent release during recovery phases, providing insights into the temporal regulation of translation during stress adaptation . Puromycin incorporation assays performed in conjunction with EIF4G2 immunofluorescence enable direct correlation between EIF4G2 localization patterns and local translation activity at the single-cell level, revealing functional consequences of EIF4G2 redistribution. Researchers should consider implementing automated image analysis pipelines that quantify multiple parameters (EIF4G2 intensity, granule formation, colocalization with translation factors) across hundreds of cells to achieve statistically robust characterization of stress-induced translation regulation patterns that might be missed in small-scale qualitative analyses.

What are the considerations for using FITC-conjugated EIF4G2 antibodies in flow cytometry?

Using FITC-conjugated EIF4G2 antibodies in flow cytometry requires specific technical considerations to obtain meaningful quantitative data about EIF4G2 expression across cell populations. Fixation and permeabilization protocols need careful optimization for flow cytometry applications, with paraformaldehyde (2-4%) followed by methanol or commercial permeabilization buffers containing saponin typically providing good access to intracellular EIF4G2 while preserving cell morphology necessary for proper light scatter properties . Titration of antibody concentration is particularly critical in flow cytometry to achieve optimal separation between positive and negative populations while minimizing background, typically requiring testing of multiple dilutions to identify the concentration providing the highest signal-to-noise ratio as measured by stain index calculations. Compensation controls are essential when multiplexing with other fluorophores to correct for spectral overlap, requiring single-color controls for each fluorophore in the panel prepared under identical conditions as the experimental samples . Gating strategies should incorporate viability dyes to exclude dead cells, which often exhibit increased autofluorescence and non-specific antibody binding that can confound EIF4G2 signal interpretation. Reference controls including fluorescence-minus-one (FMO) preparations help establish proper gate boundaries by accounting for spillover contributions from other fluorophores, while biological controls comparing samples with known differential EIF4G2 expression (such as stressed versus unstressed cells) confirm the assay's ability to detect biologically relevant expression changes. Researchers should consider potential cell cycle dependencies of EIF4G2 expression, which may necessitate additional staining for DNA content to correlate EIF4G2 levels with specific cell cycle phases when studying processes where both parameters could be relevant.

How can I apply FITC-conjugated EIF4G2 antibodies in tissue microarray (TMA) analysis?

Implementing FITC-conjugated EIF4G2 antibodies in tissue microarray analysis requires specialized protocols to achieve consistent staining across multiple tissue cores while enabling quantitative expression profiling. Antigen retrieval optimization is particularly critical for TMA applications, as tissues from different sources may exhibit varying fixation histories; therefore, systematic comparison of different retrieval methods (heat-induced epitope retrieval with citrate buffer pH 6.0 versus Tris-EDTA pH 9.0, or enzymatic retrieval) should be performed on representative tissue sections before proceeding with full TMA analysis . Automated staining platforms offer significant advantages for TMA applications by ensuring consistent antibody concentration, incubation times, and washing steps across all tissue cores, minimizing technical variability that could confound biological interpretation. Multispectral imaging systems capable of spectral unmixing provide superior results when working with tissue samples by effectively separating FITC signal from tissue autofluorescence, which can be particularly problematic in tissues rich in collagen, elastin, or lipofuscin . Digital image analysis workflows specifically designed for TMAs should be implemented, incorporating tissue segmentation algorithms that identify regions of interest (tumor versus stroma, specific cell types) followed by quantification of EIF4G2 staining intensity, percentage of positive cells, and subcellular localization patterns within each defined compartment. Reference controls including both positive and negative tissue cores should be incorporated into each TMA to validate staining performance and provide internal standards for normalization, enabling reliable comparison of EIF4G2 expression across different tissue types or disease states represented in the array.

What advanced microscopy techniques enhance the utility of FITC-conjugated EIF4G2 antibodies?

Advanced microscopy techniques significantly extend the research applications of FITC-conjugated EIF4G2 antibodies beyond conventional imaging approaches. Super-resolution microscopy methods including Structured Illumination Microscopy (SIM), Stimulated Emission Depletion (STED), and Single Molecule Localization Microscopy (SMLM) overcome the diffraction limit of conventional microscopy, revealing nanoscale organization of EIF4G2 within translation initiation complexes that would remain obscured with standard approaches, though researchers must consider that FITC is not optimal for all super-resolution methods and may need to be replaced with more photostable alternatives for techniques requiring extensive illumination . Fluorescence lifetime imaging microscopy (FLIM) provides additional information beyond simple intensity measurements by capturing the excited-state decay kinetics of FITC, which can change when EIF4G2 interacts with binding partners, offering a quantitative approach to study protein-protein interactions in situ. Fluorescence correlation spectroscopy (FCS) permits analysis of EIF4G2 mobility and concentration in living cells by measuring fluorescence fluctuations as molecules diffuse through a small observation volume, providing insights into its diffusion characteristics under different cellular conditions . Förster resonance energy transfer (FRET) approaches combining FITC-conjugated EIF4G2 antibodies with acceptor fluorophore-labeled antibodies against putative interaction partners can directly demonstrate proximity (<10nm) between proteins, providing strong evidence for functional interactions within the cellular context. Light-sheet fluorescence microscopy enables rapid three-dimensional imaging of EIF4G2 distribution with minimal phototoxicity, particularly valuable for capturing dynamic processes in thick samples such as spheroids or small model organisms expressing human EIF4G2.

How can EIF4G2 phosphorylation states be analyzed using modified immunofluorescence approaches?

Analyzing EIF4G2 phosphorylation states requires specialized immunofluorescence approaches that can distinguish between different post-translational modifications of this translation factor. Phospho-specific antibody panels represent the primary approach, where researchers would use a combination of FITC-conjugated general EIF4G2 antibodies alongside antibodies specifically recognizing phosphorylated forms at key regulatory sites (particularly at Ser540, Thr508, and Ser730), allowing direct visualization of the relationship between total EIF4G2 and its phosphorylated subpopulations . Lambda phosphatase treatment controls provide critical validation for phospho-specific staining, where parallel samples treated with this enzyme to remove phosphate groups should show significantly reduced or absent signal with phospho-specific antibodies while maintaining reactivity with general EIF4G2 antibodies. Proximity ligation assays (PLA) offer a powerful alternative approach where antibodies against EIF4G2 and phosphorylated epitopes are used in combination with oligonucleotide-conjugated secondary antibodies, generating fluorescent signals only when the two target proteins are within 40nm proximity, thereby providing exquisite specificity for detecting phosphorylated EIF4G2 species . Phos-tag™ supplement protocols, where EIF4G2 immunofluorescence is performed on cells previously treated with Phos-tag™ reagents that specifically bind phosphorylated proteins, can enhance detection sensitivity for phosphorylated EIF4G2 forms by effectively concentrating them or reducing their mobility. Researchers should implement quantitative image analysis approaches measuring the ratio of phosphorylated to total EIF4G2 signal across different subcellular compartments, providing insights into the spatial regulation of EIF4G2 phosphorylation and its correlation with functional outcomes such as translation initiation complex formation or stress granule recruitment.

How should I interpret heterogeneous EIF4G2 staining patterns within a cell population?

Heterogeneous EIF4G2 staining patterns within cell populations require nuanced analytical approaches to extract meaningful biological insights from apparent variability. Cell cycle correlation analysis represents an essential first step, as EIF4G2 expression and localization can vary substantially across different cell cycle phases; researchers should implement dual staining with cell cycle markers (such as PCNA for S-phase or phospho-histone H3 for mitosis) to determine whether observed heterogeneity correlates with cell cycle progression . Single-cell quantification approaches provide more robust characterization than visual assessment alone, where automated image analysis measuring parameters such as integrated intensity, nuclear/cytoplasmic ratio, and granule formation for hundreds of individual cells can reveal distinct subpopulations with characteristic EIF4G2 expression patterns. Microenvironment correlation should be considered, particularly in tissue samples or three-dimensional culture systems, where spatial analysis can determine whether EIF4G2 heterogeneity corresponds to local environmental factors such as hypoxia gradients, nutrient availability, or cell-cell contact patterns . Time-lapse analysis of living cells using membrane-permeable FITC-conjugated antibodies can distinguish between stable subpopulations versus temporal fluctuations in EIF4G2 patterns, providing insights into the dynamic versus static nature of the observed heterogeneity. Researchers should consider implementing computational clustering approaches such as t-SNE or UMAP to identify natural groupings of cells based on multiple EIF4G2 parameters (intensity, localization, granularity), followed by correlation of these clusters with biological variables to determine the functional significance of the observed heterogeneity patterns.

What are the best practices for quantitatively comparing EIF4G2 expression between experimental conditions?

Quantitative comparison of EIF4G2 expression between experimental conditions requires rigorous methodological approaches to ensure measurements reflect true biological differences rather than technical artifacts. Normalization strategies represent the cornerstone of reliable comparison, with several options available: global intensity normalization using total cellular protein content (measured through parallel protein staining), internal reference normalization using housekeeping proteins unaffected by the experimental conditions, or structural reference normalization based on stable cellular features like nuclear area . Batch effect correction is essential when analyzing samples processed on different days or by different operators, typically implemented through inclusion of identical reference samples in each batch or through computational approaches such as ComBat or linear mixed models that mathematically adjust for systematic batch-related variations. Statistical power considerations should guide experimental design, with power calculations based on preliminary data determining the minimum number of cells/fields required to detect expected effect sizes with appropriate statistical confidence, generally requiring hundreds to thousands of cells for detecting subtle expression changes . Advanced statistical approaches addressing non-normal distributions frequently encountered in immunofluorescence data should be implemented, including non-parametric tests (Mann-Whitney U, Kruskal-Wallis) or data transformation strategies when appropriate. Researchers should implement blinded analysis protocols where the analyst quantifying EIF4G2 staining is unaware of sample identity until after measurements are complete, minimizing the risk of unconscious bias influencing the results. Multi-parameter context analysis provides more meaningful interpretation by simultaneously measuring EIF4G2 alongside markers of cellular processes (proliferation, stress, differentiation) to determine how expression changes relate to functional cellular states across experimental conditions.

How can I distinguish between specific EIF4G2 stress granules and non-specific aggregates?

Distinguishing genuine EIF4G2-containing stress granules from non-specific protein aggregates requires systematic analysis incorporating multiple validation criteria. Co-localization with established stress granule markers provides the primary validation approach, as genuine stress granules should show significant overlap with canonical components such as G3BP1, TIA-1, or PABP, quantified through colocalization coefficients (Manders' overlap or Pearson's correlation) with values typically exceeding 0.6 for authentic association . Stress-dependent formation dynamics represent another critical criterion, as legitimate stress granules should appear rapidly following exposure to known stress granule-inducing conditions (arsenite, heat shock, thapsigargin) and dissolve following stress removal or treatment with translation inhibitors like cycloheximide, unlike non-specific aggregates that typically form more slowly and persist despite stress removal. Size and morphology parameters can provide additional discrimination, as stress granules typically exhibit characteristic dimensions (0.5-2μm) and irregular but roughly spherical morphology, distinguishable from smaller protein aggregates or larger non-specific precipitates through quantitative image analysis . RNA-dependency tests offer definitive validation, as treatment with RNase should disrupt genuine stress granules containing RNA while leaving protein-only aggregates intact. Researchers should implement a comprehensive analytical framework incorporating all these criteria, ideally using automated image analysis platforms that can process large numbers of cells to generate statistically robust discrimination between true stress granules and non-specific structures that might appear similar through casual visual inspection.

What analytical approaches can reveal EIF4G2's role in cap-independent translation?

Sophisticated analytical approaches combining FITC-conjugated EIF4G2 antibody staining with functional translation assays can elucidate EIF4G2's role in cap-independent translation mechanisms. Bicistronic reporter systems represent a powerful approach where cells are transfected with constructs containing two reporter genes separated by an IRES element, with the first reporter dependent on cap-dependent translation and the second on IRES-mediated translation; correlation of EIF4G2 staining intensity with the ratio of second-to-first reporter expression across individual cells can reveal the quantitative relationship between EIF4G2 levels and IRES activity . Polysome fractionation followed by immunofluorescence analysis of the different fractions can determine EIF4G2's association with actively translating ribosomes under conditions where cap-dependent translation is inhibited, such as during viral infection or cellular stress, providing direct evidence for its role in maintaining translation of specific mRNAs. Puromycylation proximity assays combining FITC-EIF4G2 antibodies with antibodies recognizing puromycin-labeled nascent peptides can reveal spatial correlation between EIF4G2 localization and active translation sites at subcellular resolution, particularly valuable for identifying cap-independent translation hot spots during stress conditions . Ribosome profiling correlation, where EIF4G2 immunoprecipitation is performed followed by sequencing of associated mRNAs and comparison with ribosome-protected fragments, can identify specific transcripts dependent on EIF4G2 for their translation under different cellular conditions. Researchers should implement comprehensive data integration frameworks that synthesize evidence from multiple analytical approaches to develop mechanistic models of how EIF4G2 contributes to the complex landscape of translation regulation, particularly focusing on how its activity changes during transitions between normal and stressed cellular states.

How can FITC-conjugated EIF4G2 antibodies be used in studying viral hijacking of the translation machinery?

FITC-conjugated EIF4G2 antibodies provide valuable tools for investigating the intricate mechanisms through which viruses manipulate the host translation apparatus. Temporal colocalization studies tracking the association between EIF4G2 and viral components (proteins or RNA) at different stages of infection can reveal how viruses recruit or modify this translation factor to support their replication cycle, with quantitative image analysis measuring the progressive redistribution of EIF4G2 from host structures to viral replication compartments . Integration with functional translation assays where cells are pulse-labeled with puromycin or methionine analogs can correlate EIF4G2 redistribution with shifts from host to viral protein synthesis, providing mechanistic insights into how EIF4G2 manipulation contributes to the virus-induced shutoff of host translation. Viral mutant comparison studies, where cells are infected with wild-type versus mutant viruses lacking specific host-interaction domains, can identify viral determinants responsible for EIF4G2 recruitment or modification, particularly valuable for understanding the molecular basis of viral translation strategies . Pharmacological intervention studies combining EIF4G2 immunofluorescence with compounds targeting specific steps in translation initiation can identify potential vulnerabilities in viral takeover mechanisms that might be exploitable for antiviral development. Researchers should consider implementing high-resolution time-lapse imaging approaches capturing the earliest stages of viral interaction with the translation machinery, as these initial events often establish critical virus-host interactions that determine the subsequent course of infection and represent potential targets for therapeutic intervention.

What approaches can reveal EIF4G2's role in cancer-specific translation programs?

Investigating EIF4G2's involvement in cancer-specific translation programs requires multifaceted approaches that connect its expression patterns with functional outcomes in malignant cells. Cancer tissue microarray analysis using FITC-conjugated EIF4G2 antibodies across diverse cancer types and matched normal tissues can identify patterns of overexpression or altered subcellular localization, with quantitative image analysis measuring parameters such as expression level, nuclear/cytoplasmic ratio, and association with ribosomal markers across hundreds of patient samples to identify cancer-specific alterations . Correlation with patient outcome data linking EIF4G2 expression patterns with clinical parameters such as survival, metastasis, or treatment response can identify prognostic signatures and potential therapeutic opportunities, particularly valuable when combined with molecular subtyping to identify cancer subsets where EIF4G2-dependent translation may play a crucial role. Therapeutic response visualization, where cancer cells are treated with targeted therapies (mTOR inhibitors, eIF4A inhibitors) followed by analysis of EIF4G2 redistribution and associated changes in translation activity, can reveal how cancer cells adapt their translation apparatus to overcome treatment effects . Stress adaptation studies comparing how normal versus cancer cells redistribute EIF4G2 in response to microenvironmental stresses (hypoxia, nutrient deprivation) can identify cancer-specific stress response mechanisms that might represent vulnerabilities for therapeutic targeting. Researchers should implement integrated analytical frameworks combining EIF4G2 immunofluorescence with complementary approaches such as polysome profiling, ribosome footprinting, and proteomic analysis to develop comprehensive models of how altered EIF4G2 function contributes to the dysregulated translation programs supporting cancer cell survival, proliferation, and therapeutic resistance.

How can FITC-conjugated EIF4G2 antibodies contribute to studying translational regulation in neurodegenerative diseases?

FITC-conjugated EIF4G2 antibodies provide valuable tools for investigating the complex relationship between disrupted translation regulation and neurodegenerative pathology. Pathological co-localization analysis examining the spatial relationship between EIF4G2 and disease-specific protein aggregates (tau in Alzheimer's, α-synuclein in Parkinson's, TDP-43 in ALS) can reveal whether translational machinery components become sequestered or abnormally associated with pathological structures, with quantitative co-localization metrics determining the strength and specificity of these associations across different disease stages . Stress response profiling in disease models, comparing how neurons with disease-associated mutations or protein aggregates redistribute EIF4G2 under stress conditions compared to healthy neurons, can identify compromised translational stress responses that might contribute to neuronal vulnerability. Regional vulnerability mapping, where brain sections are analyzed for EIF4G2 distribution patterns across differentially affected brain regions, can determine whether translation dysregulation correlates with the selective vulnerability characteristic of many neurodegenerative diseases . Single-cell translation activity correlation, where EIF4G2 immunofluorescence is combined with markers of active translation (puromycin incorporation) and cellular stress (phospho-eIF2α), can reveal how individual neurons maintain or fail to maintain protein synthesis under disease conditions, potentially explaining the progressive nature of neurodegeneration. Researchers should implement longitudinal analysis approaches in progressive disease models to track how EIF4G2 distribution and function change over the disease course, potentially identifying early alterations that precede overt pathology and might represent opportunities for preventive intervention before irreversible neuronal damage occurs.

What cellular processes can be studied using EIF4G2 antibodies in stem cell differentiation research?

FITC-conjugated EIF4G2 antibodies enable investigation of translation regulation dynamics during stem cell differentiation, revealing how protein synthesis mechanisms adapt during cell fate transitions. Differentiation time-course analysis tracking EIF4G2 expression levels and subcellular distribution as stem cells progress through defined differentiation stages can reveal how translation factor dynamics correlate with changing protein synthesis requirements, with quantitative image analysis measuring parameters such as total EIF4G2 content, nuclear/cytoplasmic ratio, and association with actively translating polysomes across the differentiation trajectory . Single-cell heterogeneity characterization measuring EIF4G2 patterns across individual cells within differentiating populations can identify whether translation factor expression anticipates or follows lineage commitment, potentially revealing how translation regulation contributes to differentiation decisions at branch points where cells choose between alternative fates. Integration with transcription factor staining, combining EIF4G2 immunofluorescence with antibodies against lineage-specifying transcription factors, can determine whether cells establishing specific translation factor configurations preferentially activate certain transcriptional programs, providing insights into the coordination between transcriptional and translational control during differentiation . Stress resilience profiling, examining how differentiating cells at various stages respond to stressors that challenge translation systems, can reveal how differentiation state influences cellular capacity to maintain protein synthesis under adverse conditions. Researchers should implement computational integration approaches synthesizing EIF4G2 staining data with transcriptomic and proteomic profiles across differentiation to develop comprehensive models of how coordinated regulation at multiple levels guides cells through complex differentiation processes.

How can FITC-conjugated EIF4G2 antibodies be utilized in high-throughput drug screening applications?

FITC-conjugated EIF4G2 antibodies enable development of high-throughput screening platforms for compounds affecting translation regulation, with applications in multiple disease contexts. Automated screening workflow implementation represents the foundation of this approach, where cells grown in multi-well formats are treated with compound libraries, fixed, stained with FITC-EIF4G2 antibodies alongside markers for cellular compartments and translation activity, then imaged using automated microscopy systems capable of capturing thousands of cells per well across hundreds of wells . Multi-parameter phenotypic profiling measuring numerous features of EIF4G2 staining (intensity, localization, granularity) alongside cellular parameters (viability, proliferation, morphology) can identify compounds inducing specific signatures that correlate with desired biological effects, such as selective inhibition of stress-induced translation pathways without disrupting basal translation. Target validation studies combining EIF4G2 immunofluorescence with genetic approaches (siRNA, CRISPR) targeting specific translation pathway components can confirm the mechanisms through which hit compounds exert their effects, essential for distinguishing on-target from off-target activities . Disease-relevant cell model screening, where patient-derived cells or genetically engineered disease models are used as screening platforms, can identify compounds with selective effects in disease contexts, potentially revealing therapeutic windows for translation-targeting therapies. Researchers should implement machine learning approaches for image analysis and hit identification, as these computational methods can identify subtle but meaningful changes in complex staining patterns that might be missed by conventional threshold-based analysis, potentially revealing novel compound classes with unique mechanisms of action affecting EIF4G2 function or localization.

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.