EIF2S2 Antibody

Shipped with Ice Packs
In Stock

Description

EIF2S2 Protein: Target Antigen Characteristics

EIF2S2 (Eukaryotic Translation Initiation Factor 2 Subunit Beta) functions as a critical component of the protein synthesis machinery in eukaryotic cells. Understanding the target antigen is essential for appreciating the functionality and specificity requirements of EIF2S2 antibodies.

Molecular and Functional Profile

EIF2S2 serves as the beta subunit of the eIF2 heterotrimer complex, which plays a central role in translational regulation. The protein facilitates the binding of initiator Met-tRNA to the 40S ribosomal subunit and participates in GTP/GDP exchange, recycling the eIF2 complex for subsequent rounds of translation initiation .

The following table summarizes key characteristics of the EIF2S2 protein:

PropertyDescription
Full NameEukaryotic Translation Initiation Factor 2 Subunit Beta
Alternative NamesEIF2S2, EIF2B, EIF-2-beta
Protein TypeTranslation initiation factor subunit
Molecular WeightApproximately 38 kDa (appears at ~50 kDa in experiments)
LocationCytoplasmic
Primary FunctionProtein synthesis initiation regulation
Role in TranslationFacilitates binding of initiator Met-tRNA to 40S ribosomal subunit
ComponentBeta subunit of eIF2 heterotrimer complex
Gene LocationHuman chromosome 20

Structure and Domains

The β-subunit contains multiple functional domains including an N-terminal domain with three lysine clusters important for interaction with eIF2B, a zinc finger motif crucial for ternary complex formation, and guanine nucleotide-binding sequences. The protein also comprises regions involved in both tRNA and mRNA interactions . These diverse structural features create multiple potential epitopes for antibody binding, influencing antibody design and production strategies.

EIF2S2 Antibody Types and Production Methods

Various types of EIF2S2 antibodies are commercially available, each produced through different methods that influence their specificity, sensitivity, and applications.

Antibody Categories

EIF2S2 antibodies are available in several formats, each with distinct characteristics and advantages:

Antibody TypeHost SpeciesProduction MethodImmunogenCommon Applications
Polyclonal EIF2S2 AntibodyPrimarily RabbitAntigen affinity purification from immunized animal seraSynthetic peptides or recombinant protein fragmentsWB, IHC, IF/ICC, IP, ELISA
Monoclonal EIF2S2 AntibodyMouseHybridoma technology with Protein G Magarose purificationRecombinant protein fragmentsWB, IHC, IF/ICC, ELISA
Recombinant EIF2S2 AntibodyVaries (E. coli/mammalian expression systems)Recombinant technology (phage display/antibody libraries)Recombinant protein/synthetic peptidesWB, IHC, IF/ICC, IP, ELISA
Conjugated EIF2S2 AntibodyVariesChemical conjugation of dyes/tags to purified antibodiesBase antibody before conjugationIF/ICC, Flow cytometry

Production Technologies

The production methods for EIF2S2 antibodies have evolved significantly, from traditional animal immunization approaches to advanced recombinant technologies:

Polyclonal antibodies are produced by immunizing animals (typically rabbits) with EIF2S2 synthetic peptides or recombinant protein fragments, followed by antibody purification from serum .

Monoclonal antibodies are generated using hybridoma technology, where antibody-producing B cells from immunized mice are fused with myeloma cells to create immortal antibody-producing cell lines .

Recombinant antibodies represent the newest generation, produced using molecular biology techniques such as phage display or antibody libraries. These methods offer several advantages, including: "increased sensitivity, confirmed specificity, high repeatability, excellent batch-to-batch consistency, sustainable supply, and animal-free production" .

Selection Criteria

When selecting an EIF2S2 antibody, researchers should consider:

  1. Target species and cross-reactivity requirements

  2. Intended applications (WB, IHC, IF, etc.)

  3. Epitope location (N-terminal, C-terminal, or specific domains)

  4. Format preference (polyclonal, monoclonal, or recombinant)

  5. Validation data availability

  6. Batch-to-batch consistency requirements

The immunogen information is particularly important for understanding antibody specificity. For example, the Proteintech antibody (10227-1-AP) uses "EIF2S2 fusion protein Ag0269" as immunogen , while antibodies-online's product (ABIN3030888) targets "an amino acid sequence from the C-terminus of human EIF2 beta (FQAVTGKRAQLRAKAN)" .

Research Applications of EIF2S2 Antibodies

EIF2S2 antibodies have been employed across multiple research techniques, contributing to both basic science understanding and disease investigations.

Technical Applications

The applications of EIF2S2 antibodies span various experimental techniques:

  1. Western Blotting (WB): Most commonly used application for detecting EIF2S2 protein expression levels. The protein typically appears at approximately 50 kDa despite its 38 kDa calculated molecular weight .

  2. Immunohistochemistry (IHC): Used to visualize EIF2S2 expression in tissue sections, particularly valuable in cancer research for examining expression patterns in tumor versus normal tissues .

  3. Immunofluorescence/Immunocytochemistry (IF/ICC): Enables subcellular localization studies of EIF2S2, confirming its predominantly cytoplasmic distribution .

  4. Immunoprecipitation (IP) and Co-IP: Employed to study protein interactions, as demonstrated in research confirming EIF2S2 interaction with SMAD4 in cervical cancer studies .

  5. ELISA: Used for quantitative measurement of EIF2S2 levels in various sample types .

Research Findings by Application

The following table summarizes key research findings utilizing EIF2S2 antibodies across different disease contexts:

Research AreaKey FindingsSignificanceAntibody Applications
Hepatocellular Carcinoma (HCC)EIF2S2 identified as prognostic biomarker associated with poor prognosis; correlation with immune cell infiltration and checkpointsPotential diagnostic and prognostic biomarker; ROC curve analysis confirms diagnostic valueIHC, Western blotting
Breast CancerEIF2S2 linked to poor prognosis; affects immune cell infiltrationMay serve as a prognostic indicator and therapeutic targetIHC, Western blotting
Cervical CancerStage-specific increase in EIF2S2 expression; confirmed interaction with SMAD4Potential role in cancer progression mechanismsWestern blotting, Co-IP
Translational RegulationEIF2S2 plays critical role in protein synthesis initiation and recycling eIF2 complexFundamental to cellular protein synthesis regulationWestern blotting, IF/ICC
Immune ResponseAssociation with CD8+ T cells, CD4+ memory T cells and immune checkpoints (PDCD1, TIGIT, CTLA4)Suggests involvement in tumor immune microenvironmentWestern blotting, ELISA

EIF2S2 Antibodies in Cancer Research

Recent studies have highlighted the potential of EIF2S2 as a biomarker in multiple cancer types, with antibodies serving as crucial tools in these investigations.

Hepatocellular Carcinoma Studies

EIF2S2 has emerged as a promising prognostic biomarker in hepatocellular carcinoma (HCC). Research utilizing EIF2S2 antibodies for immunohistochemistry and western blotting has demonstrated significantly higher expression levels in HCC tissues compared to normal liver tissue .

Analysis of The Cancer Genome Atlas (TCGA) database revealed that elevated EIF2S2 expression correlated with worse clinical outcomes in HCC patients. Multivariate COX regression analysis identified EIF2S2 as an independent risk factor for survival, and receiver operating characteristic (ROC) curve analysis confirmed its diagnostic value .

Particularly notable was the finding that EIF2S2 expression correlated with immune cell infiltration in the tumor microenvironment. Using the CIBERSORT-ABS algorithm, researchers demonstrated positive correlations between EIF2S2 expression and multiple immune cell populations, including memory B cells, plasma B cells, CD8+ T cells, CD4+ resting memory T cells, T follicular helper cells, regulatory T cells, M0 Macrophages, and M1 Macrophages .

Breast Cancer Investigations

Similar findings have been reported in breast cancer research, where EIF2S2 antibodies were employed to study expression patterns and clinical correlations. High expression levels of EIF2S2 were identified as a risk factor for poor prognosis in breast cancer patients .

The Gene Expression Profiling Interactive Analysis database revealed higher EIF2S2 expression in breast cancer compared to normal tissues, with expression levels correlating with both patient age and tumor stage. Moreover, EIF2S2 expression was associated with immune cell infiltration, including regulatory T cells, CD4+, CD8+, and natural killer cells .

Cervical Cancer Research

In cervical cancer studies, EIF2S2 antibodies have facilitated the discovery of a stage-specific increase in EIF2S2 expression. Immunoprecipitation and Bimolecular fluorescence complementation assays confirmed an interaction between EIF2S2 and SMAD4, with the N-terminus of EIF2S2 interacting with the MH-1 domain of SMAD4 .

Functional studies demonstrated that knockdown of EIF2S2 in human cervical cancer (SiHa) cells significantly reduced growth and migration properties, whereas overexpression enhanced these malignant characteristics .

Cross-Reactivity and Specificity Considerations

Understanding the cross-reactivity profile of EIF2S2 antibodies is essential for experimental design and interpretation.

Cross-Reactivity Fundamentals

Cross-reactivity occurs when an antibody raised against one specific antigen recognizes two antigens that have similar structural regions . For EIF2S2 antibodies, cross-reactivity can manifest in two primary ways:

  1. Species cross-reactivity: The ability to recognize the EIF2S2 protein across different species due to sequence conservation. This can be advantageous for comparative studies across model organisms.

  2. Protein cross-reactivity: The unintended binding to proteins other than EIF2S2 that share similar epitope structures, which can lead to false positive results.

Methods for Assessing Cross-Reactivity

Several approaches are used to evaluate and confirm the specificity of EIF2S2 antibodies:

  1. Sequence homology analysis: Pair-wise sequence alignment using NCBI-BLAST to check the percentage homology of the antibody immunogen to similar proteins or across species .

  2. Tissue cross-reactivity (TCR) screening: Testing antibody binding across approximately 38 different types of tissue sections to identify off-target and on-target binding sites .

  3. Knockout/knockdown validation: Using EIF2S2 knockout or knockdown samples as negative controls to confirm antibody specificity .

  4. Western blot analysis: Detecting a single band of the expected molecular weight (~50 kDa for EIF2S2) without significant additional bands .

  5. Comparison across antibodies: Using multiple antibodies targeting different epitopes of EIF2S2 to confirm consistent detection patterns .

Species Cross-Reactivity Profile

Most commercial EIF2S2 antibodies demonstrate cross-reactivity across human, mouse, and rat samples due to the high sequence conservation of this protein . The high degree of evolutionary conservation (pairwise amino acid identities ranging from 47 to 72% when comparing human and yeast proteins) contributes to this broad cross-reactivity .

Future Perspectives and Emerging Applications

The utility of EIF2S2 antibodies continues to expand, with several promising developments on the horizon.

Therapeutic and Diagnostic Potential

Recent research suggests EIF2S2 antibodies may have significant diagnostic applications. In hepatocellular carcinoma, ROC curve analysis has confirmed the diagnostic value of EIF2S2 as a biomarker . Similarly, in breast cancer, EIF2S2 expression has emerged as a potential prognostic indicator that could inform treatment decisions .

Additionally, the correlation between EIF2S2 expression and immune checkpoint molecules such as PDCD1, TIGIT, and CTLA4 suggests potential applications in immunotherapy response prediction . This relationship with the tumor immune microenvironment represents a particularly promising avenue for further investigation.

Drug Sensitivity Prediction

An intriguing finding from recent research is the relationship between EIF2S2 expression and drug sensitivity. Analysis using the Genomics of Drug Sensitivity in Cancer (GDSC) database revealed that high EIF2S2 expression correlated with increased sensitivity to several anticancer agents, including paclitaxel, sunitinib, S-Trityl-L-cysteine, VX-680, doxorubicin, cyclopamine, rapamycin, and gemcitabine .

This suggests that EIF2S2 antibodies could potentially serve as tools for predicting therapeutic response, although further validation is required before clinical implementation.

Technical Advancements

The development of recombinant EIF2S2 antibodies represents a significant technical advancement in the field. These antibodies offer several advantages over traditional polyclonal and monoclonal antibodies, including increased sensitivity, greater specificity, and improved batch-to-batch consistency .

Future developments may include the creation of more specialized EIF2S2 antibodies targeting specific phosphorylation sites or conformational epitopes, enabling more detailed studies of EIF2S2 regulation and function.

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method or location. For specific delivery times, please consult your local distributors.
Synonyms
DKFZp686L18198 antibody; eIF 2 beta antibody; eIF-2-beta antibody; EIF2 antibody; EIF2B antibody; EIF2beta antibody; EIF2S2 antibody; Eukaryotic initiation factor 2 beta antibody; eukaryotic initiation factor 2-beta antibody; Eukaryotic translation initiation factor 2 beta antibody; Eukaryotic translation initiation factor 2 subunit 2 antibody; Eukaryotic translation initiation factor 2 subunit 2 beta 38kDa antibody; Eukaryotic translation initiation factor 2 subunit 2 beta antibody; Eukaryotic translation initiation factor 2 subunit beta antibody; eukaryotic translation initiation factor 2; subunit 2 beta; 38kDa antibody; IF2B_HUMAN antibody; MGC8508 antibody; PPP1R67 antibody; protein phosphatase 1; regulatory subunit 67 antibody
Target Names
EIF2S2
Uniprot No.

Target Background

Function

eIF-2 plays a crucial role in the early stages of protein synthesis. It forms a ternary complex with GTP and initiator tRNA. This complex binds to a 40S ribosomal subunit, followed by mRNA binding to form a 43S preinitiation complex. The joining of the 60S ribosomal subunit to form the 80S initiation complex is preceded by the hydrolysis of GTP bound to eIF-2 and the release of an eIF-2-GDP binary complex. To enable eIF-2 to participate in another round of initiation, the GDP bound to eIF-2 must be replaced with GTP through a reaction catalyzed by eIF-2B.

Gene References Into Functions
  1. Research indicates that eukaryotic translation initiation factor 2 subunit beta (eIF2beta) is a potential therapeutic target for lung cancer. PMID: 29624814
  2. This study provides insights into the dynamic interaction between the eIF5-carboxyl terminal domain and eIF1 and eIF2beta. PMID: 22813744
  3. These findings suggest that the alpha and beta subunits of eIF2 interact, and the beta subunit plays a critical role in both the regulation and function of eIF2. PMID: 18639529
Database Links

HGNC: 3266

OMIM: 603908

KEGG: hsa:8894

STRING: 9606.ENSP00000364119

UniGene: Hs.429180

Protein Families
EIF-2-beta/eIF-5 family

Q&A

What is EIF2S2 and what is its biological significance?

EIF2S2 (Eukaryotic Translation Initiation Factor 2 Subunit Beta) functions as a critical subunit of the heterotrimeric G protein EIF2, which consists of α, β, and γ subunits. This protein plays an essential role in translation initiation by facilitating the binding of tRNA to ribosomes. Under various stress conditions, eukaryotic cells restrict protein synthesis through EIF2S2 inhibition . Recent research demonstrates that EIF2S2 is involved in cell proliferation and differentiation processes, with studies showing its deletion reduces the incidence of testicular germ cell tumors in mouse models .

How does EIF2S2 relate to cancer pathways and progression?

EIF2S2 participates in several cancer-related signaling pathways. Research has revealed that the PI3K/Akt/GSK-3β/ROS/EIF2S2 pathway regulates natural killer (NK) cell activity and tumor cell sensitivity to NK cells, directly influencing breast cancer growth and lung metastasis . In hepatocellular carcinoma (HCC), high EIF2S2 expression correlates with advanced pathological grade, higher clinical stage, and poorer prognosis . Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses demonstrate that EIF2S2 expression is closely associated with various immune pathways, indicating its important role in tumor microenvironment regulation .

What are the optimal experimental designs for studying EIF2S2 in hepatocellular carcinoma?

For comprehensive analysis of EIF2S2 in HCC, researchers should implement multi-omics approaches integrating:

  • Transcriptomic analysis: RNA-seq or microarray to quantify EIF2S2 mRNA expression levels

  • Proteomic validation: Western blot and immunohistochemistry with anti-EIF2S2 antibodies to confirm protein expression patterns

  • Clinical correlation studies: Analysis of EIF2S2 expression in relation to patient survival and clinicopathological characteristics

  • Functional assays: Knockdown or overexpression of EIF2S2 to assess effects on cell proliferation, migration, and invasion

  • Immune infiltration analysis: Flow cytometry or computational methods like CIBERSORT to correlate EIF2S2 expression with immune cell profiles

Research should include paired tumor and adjacent normal tissues, with clinical stage stratification for meaningful comparison. The study by Frontiers in Genetics demonstrated that EIF2S2 expression correlates with age, clinical stage, and pathological grade in HCC patients, providing a methodological framework for similar investigations .

How should researchers design EIF2S2 knockdown experiments to study its functional role?

When designing EIF2S2 knockdown experiments, researchers should:

  • Select appropriate vectors: Use siRNA, shRNA, or CRISPR-Cas9 systems targeting conserved regions of EIF2S2

  • Include multiple controls: Non-targeting control, mock transfection control, and wild-type control

  • Validate knockdown efficiency: Quantify EIF2S2 mRNA (RT-qPCR) and protein (Western blot) levels 48-72 hours post-transfection

  • Test multiple cell lines: Include both high and low EIF2S2-expressing HCC cell lines to observe differential effects

  • Design comprehensive functional assays: Analyze cell proliferation (CCK-8, EdU), migration (wound healing), invasion (transwell), apoptosis (Annexin V/PI), and cell cycle (flow cytometry)

  • Investigate downstream pathways: Examine effects on immune pathway components identified through GO and KEGG analyses

  • Perform rescue experiments: Re-express EIF2S2 in knockdown cells to confirm phenotype specificity

What are the optimal immunohistochemistry protocols for EIF2S2 antibody staining in HCC tissues?

For optimal immunohistochemical detection of EIF2S2 in HCC tissues:

  • Tissue preparation: Use formalin-fixed, paraffin-embedded (FFPE) tissues sectioned at 4-5 μm

  • Antigen retrieval: Perform heat-induced epitope retrieval using citrate buffer (pH 6.0) for 20 minutes

  • Blocking: Block endogenous peroxidase activity with 3% H₂O₂ for 10 minutes, followed by protein blocking with 5% normal goat serum

  • Primary antibody: Incubate with anti-EIF2S2 antibody (1:100-1:200 dilution) at 4°C overnight

  • Detection system: Apply HRP-conjugated secondary antibody and DAB substrate

  • Counterstaining: Use hematoxylin for nuclear visualization

  • Evaluation: Score staining intensity (0-3) and percentage of positive cells to generate H-score

  • Controls: Include positive controls (verified EIF2S2-expressing tissues), negative controls (antibody diluent only), and antibody validation controls

Based on research findings, EIF2S2 protein expression is significantly higher in HCC tissues compared to normal liver tissues, as confirmed by immunohistochemical analysis from the Human Protein Atlas database .

What techniques provide the most reliable quantification of EIF2S2 protein expression?

For reliable quantification of EIF2S2 protein expression, researchers should employ multiple complementary techniques:

TechniqueAdvantagesConsiderationsRecommended Protocol
Western blotQuantitative, size verificationRequires tissue lysisUse RIPA buffer with protease inhibitors; 1:1000 primary antibody dilution; quantify relative to loading controls (β-actin, GAPDH)
ImmunohistochemistrySpatial information, clinical correlationSemi-quantitativeUse validated scoring systems (H-score or Allred score); digital pathology for objective quantification
ELISAHigh sensitivity, high throughputNo spatial informationSandwich ELISA with capture and detection antibodies against different EIF2S2 epitopes
ImmunofluorescenceCo-localization studiesPhotobleaching concernsMulti-channel imaging with DAPI nuclear counterstain and specific EIF2S2 antibody (1:200)
Mass spectrometryAbsolute quantification, PTM detectionComplex sample preparationUse SILAC or TMT labeling for comparative proteomics

Research indicates that integrating multiple techniques provides the most comprehensive assessment of EIF2S2 expression patterns in tumor tissues .

How does EIF2S2 expression correlate with immune cell infiltration in HCC?

EIF2S2 expression demonstrates significant correlations with various immune cell populations in the tumor microenvironment:

  • Positive correlations: Multiple algorithms including CIBERSORT-ABS demonstrate that EIF2S2 expression positively correlates with:

    • Memory B cells

    • Plasma B cells

    • CD8+ T cells

    • CD4+ resting memory T cells

    • T follicular helper cells

    • Regulatory T cells

    • M0 Macrophages

    • M1 Macrophages

  • Negative correlations: EIF2S2 expression negatively correlates with:

    • Resting NK cells

    • Activated mast cells

These patterns suggest EIF2S2 may modulate the tumor immune microenvironment, potentially affecting immunotherapy responses. Researchers investigating tumor immune interactions should consider EIF2S2 expression as a potential factor influencing immune cell recruitment and function in the tumor microenvironment.

What is the relationship between EIF2S2 expression and immune checkpoint molecules?

Analysis reveals significant correlations between EIF2S2 expression and multiple immune checkpoint molecules:

  • Positive correlations: EIF2S2 expression positively correlates with:

    • PDCD1 (PD-1)

    • TIGIT

    • CTLA4

    • LAG-3

    • BTLA

This correlation pattern suggests that EIF2S2 may be involved in immune evasion mechanisms in HCC. The positive correlation with multiple immune checkpoints indicates that tumors with high EIF2S2 expression might create an immunosuppressive microenvironment. These findings have important implications for immunotherapy approaches, as patients with high EIF2S2 expression might benefit from immune checkpoint inhibitors targeting these pathways .

How should researchers integrate EIF2S2 expression analysis with chemosensitivity studies?

To effectively integrate EIF2S2 expression analysis with chemosensitivity studies, researchers should:

  • Stratify patients/samples: Divide samples into high and low EIF2S2 expression groups based on median expression values

  • Drug sensitivity testing: Using the GDSC database and "pRRophetic" R package approach, correlate EIF2S2 expression with drug sensitivity profiles

  • Focus on relevant drugs: Research has identified greater sensitivity to specific drugs in EIF2S2-high expression samples, including:

    • Paclitaxel

    • Sunitinib

    • S-Trityl-L-cysteine

    • VX-680

    • Doxorubicin

    • Cyclopamine

    • Rapamycin

    • Gemcitabine

  • Validation experiments: Conduct in vitro drug sensitivity assays using cell lines with manipulated EIF2S2 expression levels

  • Mechanistic investigation: Explore the molecular mechanisms underlying the differential drug responses, particularly focusing on pathways identified through co-expression analysis

This integrated approach can provide valuable insights for personalized treatment strategies based on EIF2S2 expression levels in HCC patients.

What are the common technical challenges when using EIF2S2 antibodies and how can they be addressed?

Researchers frequently encounter several technical challenges when working with EIF2S2 antibodies:

  • Cross-reactivity issues:

    • Challenge: EIF2S2 shares sequence homology with other EIF family members

    • Solution: Use monoclonal antibodies targeting unique epitopes; validate specificity using knockout/knockdown controls

  • Signal intensity variation:

    • Challenge: Variable immunostaining intensity across different tissue samples

    • Solution: Optimize antigen retrieval methods; titrate antibody concentrations; use automated staining platforms for consistency

  • Background staining:

    • Challenge: Non-specific binding causing high background

    • Solution: Increase blocking time (5% BSA or normal serum); optimize antibody dilution; include appropriate negative controls

  • Epitope masking:

    • Challenge: Fixation can mask EIF2S2 epitopes

    • Solution: Test multiple antigen retrieval methods (heat-induced vs. enzymatic); use fresh frozen samples when possible

  • Reproducibility concerns:

    • Challenge: Variation between experiments and laboratories

    • Solution: Standardize protocols; use automated imaging and quantification; implement positive and negative controls in each experiment

These technical considerations are especially important when comparing EIF2S2 expression across different HCC samples for prognostic or mechanistic studies .

How can researchers differentiate between specific and non-specific signals when using EIF2S2 antibodies?

To differentiate between specific and non-specific signals when using EIF2S2 antibodies:

  • Use multiple antibody validation approaches:

    • Genetic validation: Test antibody in EIF2S2 knockdown/knockout systems

    • Peptide competition: Pre-incubate antibody with immunizing peptide

    • Orthogonal validation: Compare results with alternative detection methods (e.g., mass spectrometry)

    • Independent antibody validation: Test multiple antibodies targeting different EIF2S2 epitopes

  • Implement comprehensive controls:

    • Positive controls: Tissues known to express EIF2S2 (e.g., HCC samples with confirmed high expression)

    • Negative controls: Primary antibody omission

    • Isotype controls: Matched isotype antibody at the same concentration

    • Absorption controls: Antibody pre-absorbed with recombinant EIF2S2

  • Analyze staining patterns:

    • Specific EIF2S2 staining should match expected subcellular localization

    • Non-specific staining often presents as diffuse background or unexpected localization

  • Quantitative assessment:

    • Compare signal-to-noise ratios across different antibody dilutions

    • Use digital imaging analysis to objectively quantify specific signals

Following these guidelines ensures reliable detection of EIF2S2 for accurate assessment of its expression in research and potential clinical applications .

How should researchers analyze EIF2S2 expression data to assess its prognostic value in HCC?

To properly assess the prognostic value of EIF2S2 in HCC, researchers should implement this methodological framework:

Research has demonstrated that high EIF2S2 expression correlates with shortened OS and PFS in HCC patients, and both univariate and multivariate analyses confirm EIF2S2 as an independent prognostic factor .

What methodologies should be used to investigate EIF2S2 as a potential therapeutic target in HCC?

To investigate EIF2S2 as a potential therapeutic target in HCC, researchers should employ these methodological approaches:

  • Target validation studies:

    • Gene silencing experiments (siRNA, shRNA, CRISPR-Cas9) to evaluate effects on cell proliferation, migration, invasion, and apoptosis

    • Overexpression studies to determine if increased EIF2S2 promotes oncogenic phenotypes

    • Patient-derived xenograft (PDX) models with EIF2S2 modulation to assess in vivo relevance

  • Drug discovery approaches:

    • Structure-based virtual screening to identify potential EIF2S2 inhibitors

    • High-throughput screening of compound libraries

    • Rational drug design based on EIF2S2 protein structure

  • Combinatorial therapy assessment:

    • Test combinations of EIF2S2 inhibition with:

      • Conventional chemotherapeutic agents (especially those showing sensitivity correlation with EIF2S2 expression, such as paclitaxel and sunitinib)

      • Immune checkpoint inhibitors (given the correlation between EIF2S2 and immune checkpoint molecules)

      • Molecularly targeted therapies

  • Biomarker development:

    • Develop companion diagnostic tools to identify patients likely to benefit from EIF2S2-targeted therapies

    • Establish standardized methods for EIF2S2 detection in clinical samples

  • Mechanism-based studies:

    • Investigate downstream signaling pathways affected by EIF2S2 inhibition

    • Explore the relationship between EIF2S2 and its co-expressed genes (TPD52L2, NOP56, CHMP4B, PDRG1, SNRPD1, RPN2)

This comprehensive approach will help determine whether EIF2S2 represents a viable therapeutic target for HCC treatment and identify the patient populations most likely to benefit.

How can researchers investigate the mechanism by which EIF2S2 influences immune cell populations in the tumor microenvironment?

To investigate EIF2S2's role in modulating the tumor immune microenvironment, researchers should implement:

  • Single-cell RNA sequencing: Analyze immune cell populations in EIF2S2-high versus EIF2S2-low tumors to characterize:

    • Immune cell composition differences

    • Activation states of various immune cell types

    • Cell-specific gene expression signatures

  • Spatial transcriptomics and multiplex immunofluorescence: Map the spatial relationship between EIF2S2-expressing tumor cells and infiltrating immune cells using:

    • GeoMx Digital Spatial Profiler

    • Multiplexed immunohistochemistry

    • In situ hybridization combined with immunostaining

  • Co-culture experiments: Design in vitro co-culture systems with:

    • EIF2S2-manipulated tumor cells (overexpression/knockdown)

    • Various immune cell populations (CD8+ T cells, memory B cells, NK cells)

    • Measurement of immune cell activation, proliferation, and function

  • Cytokine/chemokine profiling: Analyze secretome of EIF2S2-high versus EIF2S2-low tumor cells to identify:

    • Differentially secreted immune-modulatory factors

    • Chemokines affecting immune cell recruitment

    • Cytokines influencing immune cell function

  • Mechanistic studies: Investigate EIF2S2's role in:

    • Regulating translation of specific immune-modulatory factors

    • Stress response pathways affecting immune recognition

    • Post-translational modifications of immune signaling components

These approaches will help elucidate the mechanisms by which EIF2S2 influences the positive correlation with memory B cells, plasma B cells, CD8+ T cells, and CD4+ resting memory T cells observed in HCC .

What approaches should be used to resolve contradictory findings regarding EIF2S2 function across different cancer types?

When addressing contradictory findings about EIF2S2 function across cancer types, researchers should implement:

  • Systematic meta-analysis:

    • Compile all published data on EIF2S2 across cancer types

    • Apply standardized statistical methods to assess heterogeneity

    • Identify potential sources of variation (methodologies, patient populations, disease stages)

  • Cancer type-specific mechanistic studies:

    • Design parallel experiments across multiple cancer cell lines

    • Use identical methodologies to manipulate EIF2S2 expression

    • Compare downstream effects on signaling pathways

  • Context-dependent analysis:

    • Investigate tissue-specific interaction partners of EIF2S2

    • Analyze genetic background dependencies (mutation profiles)

    • Evaluate microenvironmental factors influencing EIF2S2 function

  • Integrated multi-omics approach:

    • Correlate EIF2S2 expression with:

      • Genomic alterations (mutations, CNVs)

      • Epigenetic modifications

      • Proteomic profiles

      • Metabolomic signatures

  • Pathway-focused analysis:

    • Compare the overlap between EIF2S2-associated pathways across cancer types

    • Identify common core functions versus cancer-specific roles

    • Analyze differences in stress response pathways

  • Isoform-specific investigation:

    • Determine if different EIF2S2 isoforms predominate in different cancer types

    • Assess isoform-specific functions and interactions

This systematic approach can help reconcile seemingly contradictory findings, such as why EIF2S2 shows prognostic significance in HCC but might have different implications in other cancer types.

How can researchers incorporate EIF2S2 expression data into multi-omics approaches for HCC characterization?

To effectively incorporate EIF2S2 expression data into multi-omics approaches for HCC characterization:

  • Integrated genomic analysis:

    • Correlate EIF2S2 expression with:

      • Mutation profiles (using whole-exome sequencing)

      • Copy number variations

      • Chromosomal instability metrics

    • Identify genetic alterations co-occurring with high EIF2S2 expression

  • Transcriptomic integration:

    • Perform weighted gene co-expression network analysis (WGCNA) to identify EIF2S2-associated gene modules

    • Construct EIF2S2-centered regulatory networks

    • Apply systems biology approaches to model EIF2S2's role in cellular pathways

  • Epigenomic correlation:

    • Analyze DNA methylation patterns of EIF2S2 promoter regions

    • Investigate histone modifications associated with EIF2S2 expression

    • Study chromatin accessibility at the EIF2S2 locus and related regulatory elements

  • Proteomic validation:

    • Perform reverse-phase protein arrays (RPPA) to validate EIF2S2 protein levels

    • Identify post-translational modifications of EIF2S2 in HCC

    • Study protein-protein interaction networks around EIF2S2

  • Metabolomic analysis:

    • Correlate EIF2S2 expression with metabolic signatures

    • Investigate alterations in protein synthesis-related metabolites

  • Integrative computational approaches:

    • Apply machine learning algorithms to integrate multi-omics data

    • Develop predictive models incorporating EIF2S2 expression

    • Use dimension reduction techniques to visualize integrated data

This multi-omics approach would extend beyond the current understanding of EIF2S2 in HCC, which has primarily focused on its correlation with clinical parameters and immune infiltration .

What advanced immunoprofiling methods should researchers use to further characterize the relationship between EIF2S2 and the immune microenvironment?

For advanced immunoprofiling to better characterize EIF2S2's relationship with the immune microenvironment:

  • Mass cytometry (CyTOF):

    • Analyze 40+ immune markers simultaneously in single cells

    • Compare immune cell phenotypes in EIF2S2-high versus EIF2S2-low tumor regions

    • Quantify rare immune cell subpopulations

  • Single-cell immune repertoire sequencing:

    • Characterize T-cell receptor (TCR) and B-cell receptor (BCR) diversity

    • Correlate clonal expansion patterns with EIF2S2 expression

    • Identify antigen specificity of infiltrating lymphocytes

  • Spatial proteomics:

    • Implement multiplexed ion beam imaging (MIBI) or Imaging Mass Cytometry

    • Map spatial relationships between EIF2S2-expressing cells and immune components

    • Quantify cell-cell interactions in the tumor microenvironment

  • Functional immunophenotyping:

    • Assess cytokine production profiles of tumor-infiltrating lymphocytes

    • Measure cytotoxic activity against EIF2S2-expressing tumor cells

    • Evaluate immune checkpoint receptor expression and functionality

  • In vivo immunocompetent models:

    • Develop syngeneic mouse models with modulated EIF2S2 expression

    • Track immune response dynamics using intravital microscopy

    • Test immunotherapeutic approaches in the context of EIF2S2 manipulation

  • Ex vivo tumor fragment platforms:

    • Culture tumor fragments with preserved immune microenvironment

    • Test effects of EIF2S2 inhibition on immune cell function

    • Evaluate combination approaches with immune checkpoint blockade

These advanced methodologies would build upon the established correlations between EIF2S2 expression and various immune cell populations (memory B cells, plasma B cells, CD8+ T cells) and immune checkpoints (PDCD1, TIGIT, CTLA4) , providing mechanistic insights that could inform immunotherapeutic strategies for HCC.

What are the key considerations for developing EIF2S2 as a clinical biomarker for HCC?

To develop EIF2S2 as a clinical biomarker for HCC, researchers must address these critical considerations:

  • Standardization of detection methods:

    • Establish standardized IHC protocols with validated antibodies

    • Develop quantitative PCR assays with appropriate reference genes

    • Create ELISA or other protein quantification methods for clinical samples

  • Determination of optimal cutoff values:

    • Conduct large-scale validation studies to determine clinically relevant expression thresholds

    • Use statistical methods like receiver operating characteristic (ROC) curve analysis to establish cutoff values with optimal sensitivity and specificity

  • Clinical validation studies:

    • Perform multicenter retrospective validation using tissue microarrays

    • Conduct prospective clinical trials to validate prognostic significance

    • Evaluate EIF2S2 in diverse patient populations and disease stages

  • Integration with existing biomarkers:

    • Compare performance against established HCC biomarkers (AFP, GPC3)

    • Develop multiparameter models incorporating EIF2S2 with other markers

    • Assess added prognostic or predictive value

  • Evaluation as a predictive biomarker:

    • Investigate correlation with response to specific therapies

    • Assess potential for predicting immunotherapy response given the correlation with immune checkpoints

    • Determine utility for predicting sensitivity to specific chemotherapeutics

  • Technical and practical considerations:

    • Develop less invasive detection methods (liquid biopsy approaches)

    • Assess EIF2S2 in circulating tumor cells or exosomes

    • Evaluate cost-effectiveness of implementation in clinical settings

With current evidence showing that EIF2S2 functions as an independent prognostic factor and correlates with clinicopathological features including pathological grade and clinical stage , it represents a promising biomarker candidate requiring rigorous validation.

What future research directions should be prioritized regarding EIF2S2 antibodies in cancer research?

Future research priorities for EIF2S2 antibodies in cancer research should include:

  • Development of therapeutic antibodies:

    • Engineer antibodies targeting extracellular domains or internalization pathways

    • Develop antibody-drug conjugates delivering cytotoxic payloads to EIF2S2-expressing cells

    • Investigate bispecific antibodies linking EIF2S2-expressing tumor cells to immune effectors

  • Improved diagnostic applications:

    • Create highly specific monoclonal antibodies for improved tissue diagnostics

    • Develop antibodies compatible with multiplexed imaging platforms

    • Engineer antibody fragments for enhanced tissue penetration

  • Mechanistic investigations:

    • Generate antibodies targeting specific EIF2S2 phosphorylation states

    • Develop antibodies distinguishing between EIF2S2 conformational states

    • Create antibodies recognizing EIF2S2 interaction interfaces

  • Translational medicine applications:

    • Establish companion diagnostic antibodies for potential EIF2S2-targeted therapies

    • Develop antibody-based imaging agents for non-invasive detection of EIF2S2-expressing tumors

    • Create standardized antibody-based assays for clinical implementation

  • Technological innovations:

    • Engineer nanobodies or alternative binding scaffolds with improved tissue penetration

    • Develop proximity-labeling antibodies to identify EIF2S2 interaction partners in situ

    • Create antibody-based sensors for real-time monitoring of EIF2S2 dynamics

  • Combinatorial therapeutic approaches:

    • Investigate antibodies targeting EIF2S2 in combination with immune checkpoint inhibitors

    • Explore synergistic effects with drugs showing sensitivity correlation (paclitaxel, sunitinib)

    • Develop rational combinations based on EIF2S2-associated pathways

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 2025 TheBiotek. All Rights Reserved.