The antibodies are validated for:
| Application | Dilution Range | Positive Controls |
|---|---|---|
| Western Blot | 1:5000–1:50,000 | HCT 116, HepG2, pig brain tissue |
| IF/ICC | 1:400–1:1600 | HepG2, PC-3 cells |
| ELISA | N/A | Human, mouse lysates |
Dilutions must be optimized for specific experimental systems .
Proteintech’s 68217-1-Ig antibody has been cited in 1 publication for WB studies .
Colon Cancer:
Therapeutic Potential:
GSPT1 forms a ternary complex with eRF1 and GTP to mediate termination at stop codons (UAA, UAG, UGA) .
Its degradation disrupts ribosomal recycling, impairing translation fidelity .
To ensure antibody specificity, employ a multi-validation approach:
Genetic validation: Perform GSPT1 knockdown or knockout experiments to confirm antibody specificity. This approach has been successfully demonstrated in studies with U87 glioblastoma cells where GSPT1-KO cells showed negative staining compared to wild-type cells .
Multiple detection methods: Cross-validate using different applications (WB, IF, IHC) to ensure consistent target detection. For instance, GSPT1 antibodies have been validated across multiple techniques showing consistent cytoplasmic localization patterns .
Molecular weight verification: GSPT1 should be detected at 80-85 kDa in Western blot applications, despite its calculated molecular weight of 4 kDa .
Enhanced validation techniques: Certain antibodies like HPA052488 have undergone enhanced validation including RNAi knockdown experiments to confirm specificity .
Recommended protocol table for multi-method validation:
| Validation Method | Experimental Approach | Expected Result |
|---|---|---|
| Western Blot | Lysates from control vs GSPT1-KO/KD cells | Single band at 80-85 kDa that disappears/diminishes in KO/KD samples |
| Immunofluorescence | Control vs GSPT1-KO/KD cells | Cytoplasmic staining that disappears in KO/KD samples |
| Peptide competition | Pre-incubation with immunogenic peptide | Signal elimination in all applications |
Selection between monoclonal and polyclonal GSPT1 antibodies should be based on experimental needs:
Monoclonal antibodies (e.g., CL488-68217, mouse IgG2a):
Offer higher batch-to-batch consistency for longitudinal studies
Provide exceptional specificity for a single epitope
Optimal for applications requiring high reproducibility such as clinical diagnostics or quantitative analyses
Polyclonal antibodies (e.g., 10763-1-AP, rabbit IgG):
Recognize multiple epitopes, beneficial for detecting proteins in different conformational states
Generally provide stronger signals in applications like IHC by binding multiple epitopes per target molecule
Better for detecting low-abundance proteins or partially denatured proteins
Validated across more published applications (16 publications for WB, 2 for IHC, 4 for IF)
The choice should be guided by specific experimental requirements:
For detecting GSPT1 in fixed tissues where epitope masking may occur, polyclonal antibodies might be preferable
For precise subcellular localization studies, monoclonal antibodies might offer cleaner backgrounds
When working with challenging samples like tumor tissues with variable protein expression, polyclonals may provide better detection sensitivity
GSPT1 epitope selection significantly impacts antibody performance across applications:
Epitope accessibility: Different regions of GSPT1 may be differentially accessible in various applications. For example, antibodies targeting amino acids 1-637 (ABIN1498536) provide broad epitope coverage suitable for multiple applications including WB, IHC, IF, and ICC .
Functional domains: GSPT1 contains distinct functional domains, including GTP-binding domains and regions that interact with eRF1. Antibodies targeting different regions may differentially detect:
Active vs. inactive conformations
Protein-protein interaction interfaces
Post-translationally modified forms
Cross-reactivity considerations: Some epitopes are more conserved across species. Antibodies targeting highly conserved regions show broader cross-reactivity across human, mouse, rat, and sometimes pig samples .
Application-specific performance:
For studying GSPT1's role in translation termination, antibodies targeting the C-terminal region may be more effective
For investigating cell cycle regulation functions, N-terminal targeted antibodies may be preferred
Research finding: Studies have shown that antibodies targeting different GSPT1 regions may yield different staining patterns in glioblastoma samples, with some patients showing strong cytoplasmic expression and others showing weak to moderate expression depending on the epitope targeted .
For optimal GSPT1 detection in cancer tissues, follow these evidence-based protocols:
Tissue Preparation and Antigen Retrieval:
Fix tissues in 10% neutral buffered formalin for 24-48 hours
Process and embed in paraffin following standard protocols
Section tissues at 4-5 μm thickness
Critical step: Perform heat-induced epitope retrieval using either:
Antibody Incubation and Detection:
Block endogenous peroxidase activity with 3% H₂O₂
Apply protein blocking solution for 20 minutes at room temperature
Incubate with primary GSPT1 antibody at validated dilutions:
Incubate 1 hour at room temperature or overnight at 4°C
Apply appropriate detection system (e.g., HRP-conjugated secondary antibody)
Develop with DAB and counterstain with hematoxylin
Validation Controls:
Include GSPT1-positive tissues (e.g., HepG2 cells, human liver cancer tissue)
Note: GSPT1 shows cytoplasmic localization in cancer cells but is typically absent in vascular tissue, providing an internal negative control
Research insight: GSPT1 expression varies among glioblastoma patients, with some showing strong expression and others showing weak to moderate expression. This heterogeneity should be considered when interpreting results .
Optimizing GSPT1 antibody protocols for flow cytometry requires attention to several critical factors:
Sample Preparation for Intracellular GSPT1 Detection:
Harvest cells (e.g., HeLa cells, which are validated for GSPT1 detection)
Fix cells with 4% paraformaldehyde for 15 minutes at room temperature
Critical step: Permeabilize with 0.1% Triton X-100 or commercial permeabilization buffer to access intracellular GSPT1
Block with 3-5% BSA or normal serum for 30 minutes
Antibody Titration and Staining:
Prepare serial dilutions of GSPT1 antibody centering around manufacturer's recommended concentration
For conjugated antibodies (e.g., CL488-68217): Use 0.40 μg per 10^6 cells in 100 μl as starting point
For unconjugated antibodies: Optimize primary antibody concentration followed by appropriate fluorochrome-conjugated secondary antibody
Incubate with antibody for 30-60 minutes at room temperature in the dark
Wash thoroughly to remove unbound antibody
Controls and Validation:
Include unstained, isotype, and single-color controls
Validate specificity with a GSPT1-knockdown sample
Use cells with known GSPT1 expression levels (e.g., HeLa cells) as positive controls
Gating Strategy:
Gate on viable single cells
Compare GSPT1 signal to isotype control to determine positive population
For cell cycle analysis applications, consider co-staining with DNA dye to correlate GSPT1 expression with cell cycle phases (relevant to GSPT1's role in G1 to S phase transition)
Effective immunofluorescence detection of GSPT1 requires optimized protocols for different cell types:
General Protocol:
Culture cells on coverslips or chamber slides
Fix with 4% paraformaldehyde for 15 minutes at room temperature
Permeabilize with 0.2% Triton X-100 for 10 minutes
Block with 5% normal serum in PBS for 1 hour
Incubate with primary GSPT1 antibody:
Incubate overnight at 4°C or 1-2 hours at room temperature
Wash 3× with PBS
For unconjugated antibodies, incubate with fluorophore-conjugated secondary antibody
Counterstain nuclei with DAPI
Mount with anti-fade mounting medium
Cell-Type Specific Considerations:
Research insight: In U87 glioblastoma cells, GSPT1 shows predominantly cytoplasmic localization. This pattern can be used to validate antibody specificity by comparing with GSPT1-knockout cells which should show negative staining .
GSPT1 antibodies serve as powerful tools for investigating its critical roles in cancer proliferation and apoptosis:
Cell Proliferation Studies:
Cell cycle analysis: Use GSPT1 antibodies in flow cytometry combined with DNA staining to correlate GSPT1 expression with cell cycle phases
Proliferation marker co-staining: Combine GSPT1 immunostaining with Ki67 to assess correlation with proliferative activity
Growth assays following GSPT1 modulation: Use GSPT1 antibodies to confirm knockdown/overexpression efficiency before assessing proliferation
Apoptosis Investigation:
Apoptosis marker co-detection: Use GSPT1 antibodies alongside apoptosis markers like cleaved PARP1 and cleaved caspase-3
In vivo apoptosis assessment: Use GSPT1 antibodies to confirm expression status in tumor models while measuring apoptotic markers
Drug sensitivity studies: Combine GSPT1 antibodies with apoptosis-inducing compounds
Methodological approach: For comprehensive analysis, employ GSPT1 antibodies in multiple techniques:
Western blot for quantitative expression analysis
Immunohistochemistry for spatial distribution in tumors
Immunofluorescence for subcellular localization studies
Recent research has identified GSPT1 as a promising therapeutic target in glioblastoma. The following methodologies utilize GSPT1 antibodies to investigate this potential:
1. Expression Profiling in Patient Samples:
Perform GSPT1 immunohistochemistry on glioblastoma tissue microarrays
Use validated antibody dilutions (1:50-1:500) with appropriate antigen retrieval
Quantify expression levels using digital pathology systems
Research finding: GSPT1 expression varies across glioblastoma patients, with expression confined to cytoplasm of tumor cells but absent in vascular tissue
2. Therapeutic Target Validation:
Generate GSPT1 knockout cells and confirm knockout efficiency using GSPT1 antibodies
Compare growth rates between wild-type, GSPT1-KO, and GSPT1-rescued cells
Assess survival in mouse models with transplanted cells of different GSPT1 status
Research data: Mice with transplanted GSPT1-KO U87 cells showed significantly longer survival compared to those with wild-type U87 cells
3. Drug Mechanism Studies:
Study GSPT1-targeting compounds like CC-885 (a cereblon modulator that degrades GSPT1)
Use GSPT1 antibodies to confirm protein degradation following drug treatment
Correlate GSPT1 levels with therapeutic response
Finding: CC-885 treatment significantly prolonged survival in mice with transplanted U87 glioblastoma tumors
4. Combination Therapy Assessment:
Investigate synergies between GSPT1-targeting approaches and standard therapeutics
Use GSPT1 antibodies to monitor expression during treatment
Research opportunity: Combining GSPT1-targeting with apoptosis-inducing agents may enhance therapeutic efficacy based on enhanced sensitivity to apoptosis in GSPT1-KO cells
The discrepancy between GSPT1 mRNA and protein expression levels in cancer tissues represents an important research area. GSPT1 antibodies enable detailed investigation of this phenomenon:
Methodological Approaches:
Dual RNA-protein analysis:
Single-cell analysis methods:
Combine RNA-FISH for GSPT1 transcript detection with immunofluorescence using GSPT1 antibodies
This allows simultaneous visualization of mRNA and protein in the same cells
Analyze correlation at single-cell level to identify regulatory patterns
Protein stability and turnover assessment:
Post-transcriptional regulation studies:
Investigate microRNA regulation of GSPT1
Use GSPT1 antibodies to measure protein levels while manipulating potential regulatory microRNAs
Research insights from clinical samples:
Analysis of 87 glioblastoma specimens showed variable GSPT1 protein expression despite mRNA level variations
Data from The Cancer Genome Atlas (TCGA) combined with protein analysis showed that GSPT1 protein levels, rather than mRNA levels, may be more relevant to biological function
This suggests post-transcriptional regulation plays a significant role in determining GSPT1 protein levels
Practical recommendation: When studying GSPT1 in cancer, researchers should employ both mRNA and protein detection methods, as protein levels may more accurately reflect functional relevance in disease processes.
Researchers working with GSPT1 antibodies may encounter several technical challenges that can be systematically addressed:
Problem: Variability in detecting the 80-85 kDa GSPT1 band
Solutions:
Optimize protein extraction: Use RIPA buffer with protease inhibitors to prevent degradation
Adjust loading amount: 20-50 μg total protein per lane is typically effective
Optimize antibody concentration: Begin with 1:2000-1:12000 dilution range for polyclonal antibodies
Extend transfer time for high molecular weight proteins
Use freshly prepared samples; avoid repeated freeze-thaw cycles
Problem: Non-specific staining reducing signal-to-noise ratio
Solutions:
Optimize blocking (5% normal serum from secondary antibody host species, 1 hour)
Titrate primary antibody concentration (starting with 1:50-1:500 for IHC)
Use antigen retrieval appropriate for target fixation (TE buffer pH 9.0 recommended)
Include proper negative controls (isotype control, GSPT1-knockout tissues)
Note: GSPT1 is absent in vascular tissue, providing an internal negative control
Problem: Different antibodies yield varying staining patterns
Solutions:
Verify epitope locations of different antibodies
Validate each antibody with GSPT1 knockdown/knockout samples
Use multiple antibodies targeting different epitopes for confirmation
Consider fixation effects on specific epitopes
Problem: Weak intracellular GSPT1 signal
Solutions:
Technical insight: The observed molecular weight of GSPT1 (80-85 kDa) differs significantly from the calculated weight (4 kDa) reported in some antibody datasheets, which represents a potential source of confusion. Always validate the correct band size against positive control cells with known GSPT1 expression .
Interpreting GSPT1 antibody staining patterns in heterogeneous tumor samples requires careful consideration of multiple factors:
Pattern Recognition and Quantification:
Subcellular localization: GSPT1 shows predominantly cytoplasmic localization in tumor cells. Nuclear staining may represent non-specific binding and should be validated
Staining intensity gradation:
Spatial heterogeneity assessment:
Evaluate multiple tumor regions (center vs. invasive margins)
Consider hot-spot analysis for areas of highest expression
Use digital pathology quantification when possible for objective measurement
Comparison with Internal Controls:
Vascular tissue: Acts as an internal negative control; GSPT1 is typically not expressed in vascular cells within tumor samples
Normal adjacent tissue: Compare tumor expression with surrounding non-malignant tissue
Infiltrating immune cells: May show different expression patterns than tumor cells
Multi-marker Analysis:
Co-staining approach: Combine GSPT1 with other markers
Cell type markers to confirm tumor cell expression
Proliferation markers (Ki67) to assess correlation with proliferative activity
Apoptosis markers (cleaved caspase-3) to evaluate relationship with cell death resistance
Correlation with Clinical Data:
Avoid overinterpretation: Research shows GSPT1 protein levels may not correlate with patient prognosis in glioblastoma
Consider technical variables:
Fixation artifacts can affect staining intensity
Batch effects between staining runs
Antibody lot-to-lot variations
Research insight: Studies have demonstrated that while GSPT1 is essential for glioblastoma growth, its expression levels alone may not predict aggressive behavior. Therefore, GSPT1 staining should be interpreted in the context of its functional role rather than simply as a prognostic biomarker .
Distinguishing specific from non-specific binding is critical for accurate GSPT1 antibody-based research, particularly in complex tissues:
Validation Controls for Specificity Confirmation:
Genetic validation controls:
Peptide competition assays:
Pre-incubate antibody with excess immunizing peptide
Specific staining should be abolished while non-specific binding remains
Most effective when the exact immunizing peptide sequence is known
Multiple antibody validation:
Technical Approaches to Minimize Non-specific Binding:
Optimized blocking protocols:
Use 5% serum from the same species as secondary antibody
Add 0.1-0.3% Triton X-100 to reduce hydrophobic interactions
Include 1% BSA to reduce non-specific protein interactions
Antibody titration:
Secondary antibody controls:
Include secondary-only control to identify non-specific secondary binding
Use isotype controls matched to primary antibody
Pattern Recognition for Specific vs. Non-specific Staining:
Subcellular localization assessment:
Specific GSPT1 staining is predominantly cytoplasmic in tumor cells
Non-specific staining often appears as:
Nuclear staining
Diffuse background staining
Edge artifacts
Tissue-specific patterns:
Research insight: When evaluating glioblastoma samples, researchers should expect GSPT1 to show cytoplasmic localization in tumor cells with variable intensity between patients. Complete absence of staining in vascular cells provides a reliable internal negative control for specificity assessment .
GSPT1 (eRF3a) plays critical roles in translation termination and mRNA degradation. Researchers can employ the following advanced approaches using GSPT1 antibodies to study these functions:
Translation Termination Studies:
Co-immunoprecipitation with translation factors:
Polysome profiling combined with GSPT1 detection:
Fractionate polysomes by sucrose gradient centrifugation
Analyze GSPT1 distribution across fractions using specific antibodies
Correlate with ribosome occupancy and translation efficiency
Proximity ligation assays:
Visualize GSPT1 interactions with translation machinery components
Use GSPT1 antibodies paired with antibodies against ribosomal proteins or other termination factors
Provides spatial information about interaction sites within cells
mRNA Degradation Pathway Investigation:
RNA immunoprecipitation (RIP):
Immunofluorescence co-localization with P-bodies:
Pulse-chase experiments:
Technical Recommendations:
Use fluorescently-conjugated GSPT1 antibodies (e.g., CoraLite® Plus 488) for live-cell imaging of translation dynamics
Consider the impact of fixation methods:
For capturing dynamic translation processes, mild fixation (2% PFA, 5 minutes)
For stable complexes, standard fixation (4% PFA, 15 minutes)
When studying translation complexes, use buffers that preserve protein-protein interactions:
Avoid harsh detergents during sample preparation
Include GTP in buffers to maintain functional interactions
GSPT1 is emerging as a promising therapeutic target, particularly in cancer research. The following approaches optimize GSPT1 antibody use in high-throughput drug screening:
Assay Development for High-Throughput Screening:
ELISA-based GSPT1 protein level quantification:
Develop sandwich ELISA using capture and detection GSPT1 antibodies
Optimize for 96/384-well format
Application: Screen compounds that induce GSPT1 degradation or modulate expression
High-content imaging assays:
AlphaLISA proximity assays:
Cellular Models for Screening:
Reporter cell lines:
Engineer cells with fluorescent/luminescent-tagged GSPT1
Validate with GSPT1 antibodies to ensure native behavior
Screen compounds for effects on GSPT1 stability
Cell line panel screening:
3D organoid models:
Validation and Secondary Screening:
Target engagement confirmation:
Cellular thermal shift assay (CETSA) with GSPT1 antibodies
Immunoprecipitation to verify compound-induced changes in GSPT1 interactions
Functional impact assessment:
Structure-activity relationship studies:
Use GSPT1 antibodies to quantify protein levels following treatment with compound derivatives
Correlate structural modifications with GSPT1 degradation efficiency
Technical recommendations: When screening cereblon modulators or other compounds targeting GSPT1 for degradation, ensure antibodies recognize epitopes that remain accessible during the degradation process to avoid false negatives.
Integrating GSPT1 antibody techniques with genomic and transcriptomic approaches enables comprehensive understanding of GSPT1's roles in cancer. The following methodologies facilitate this integration:
Multi-omics Integration Strategies:
Correlative analysis of protein and mRNA expression:
Quantify GSPT1 protein using validated antibodies (western blot, IHC)
Measure GSPT1 mRNA via RT-PCR or RNA-seq
Research finding: GSPT1 mRNA levels don't correlate with protein levels in glioblastoma, suggesting post-transcriptional regulation
Develop integrated models considering both expression modes
Chromatin immunoprecipitation sequencing (ChIP-seq) integration:
Identify transcription factors regulating GSPT1
Use GSPT1 antibodies to correlate protein levels with transcriptional regulation
Map regulatory networks controlling GSPT1 expression
CRISPR screens with antibody-based readouts:
Conduct genome-wide CRISPR screens affecting GSPT1 function
Use GSPT1 antibodies to quantify effects on protein level/localization
Identify synthetic lethal interactions with GSPT1 depletion
Translational Research Applications:
Patient-derived xenograft (PDX) models:
Tissue microarray analysis with multi-parameter readouts:
Single-cell multi-omics:
Combine single-cell RNA-seq with antibody-based protein detection
Resolve heterogeneity in GSPT1 expression at single-cell level
Identify cell populations with discordant RNA/protein expression
Methodological Framework for Integration:
Experimental design considerations:
Use identical or matched samples across platforms
Include calibration standards across batches
Design proper controls for each technology platform
Analytical approaches:
Employ multivariate statistical methods to correlate antibody-based protein data with genomic features
Use machine learning to identify patterns across data types
Pathway enrichment analysis incorporating both protein and genomic data
Validation strategy:
Verify key findings with orthogonal methods
Test predictions in independent cohorts
Functional validation of identified relationships
GSPT1 antibodies are finding expanding applications beyond traditional cancer research, opening new avenues for understanding diverse biological processes:
Neurodegenerative Disease Research:
Protein quality control mechanisms:
GSPT1's role in translation termination has implications for neurodegenerative diseases characterized by protein misfolding
GSPT1 antibodies enable investigation of translation fidelity in neuronal models
Research opportunity: Study GSPT1 expression in brain tissues using antibodies validated in mouse brain tissue
Stress granule dynamics:
Use GSPT1 antibodies to study its potential role in stress granule formation and dissolution
Implications for diseases like ALS where stress granule dysregulation occurs
Technical approach: Combine GSPT1 immunofluorescence with stress granule markers
Developmental Biology Applications:
Stem cell differentiation studies:
GSPT1's role in cell cycle regulation suggests importance in stem cell biology
Use antibodies to track GSPT1 expression changes during differentiation
Potential application: Investigate GSPT1's role in neural stem cell fate decisions
Embryonic development:
Emerging Therapeutic Areas:
Targeted protein degradation approaches:
Immunotherapeutic applications:
Explore GSPT1 as potential target for CAR-T or other immunotherapies
Use antibodies to characterize expression in immune-privileged tissues
Research question: Could GSPT1-targeted therapies synergize with immunotherapies?
New Technological Applications:
Single-molecule imaging:
Super-resolution microscopy with GSPT1 antibodies to visualize translation termination events
Enables study of spatial organization of translation termination complexes
In vivo imaging:
Development of GSPT1 antibody-based imaging agents for visualizing tumors
Research direction: Explore fluorescently-labeled antibody fragments for non-invasive imaging
Research insight: While GSPT1 has been primarily studied in cancer contexts, its fundamental roles in translation termination and cell cycle regulation suggest much broader biological significance. The availability of well-validated antibodies across multiple species facilitates these expanded research directions .
Recent methodological advances are significantly enhancing the specificity and sensitivity of GSPT1 antibody-based techniques:
Advanced Validation Technologies:
Enhanced genetic validation:
Multi-epitope targeting strategies:
Orthogonal validation methods:
Signal Amplification Innovations:
Tyramide signal amplification (TSA):
Enhances detection sensitivity for low-abundance GSPT1
Particularly valuable in tissues with weak expression
Compatible with multiplex immunofluorescence
Polymer-based detection systems:
Proximity ligation assays:
Dramatically improves signal-to-noise ratio
Enables detection of GSPT1 interactions with other proteins
Applications in studying translation termination complexes
Technical Optimization Strategies:
Automated antibody validation platforms:
High-throughput testing across multiple conditions
Standardized protocols reducing inter-laboratory variability
Systematic determination of optimal dilutions across applications
Specialized fixation protocols:
Digital pathology quantification:
Automated scoring of GSPT1 immunohistochemistry
Machine learning algorithms for pattern recognition
Reduces inter-observer variability in expression assessment