GSPT1 Antibody

Shipped with Ice Packs
In Stock

Description

Applications and Validation

The antibodies are validated for:

ApplicationDilution RangePositive Controls
Western Blot1:5000–1:50,000HCT 116, HepG2, pig brain tissue
IF/ICC1:400–1:1600HepG2, PC-3 cells
ELISAN/AHuman, mouse lysates

Notes:

  • Dilutions must be optimized for specific experimental systems .

  • Proteintech’s 68217-1-Ig antibody has been cited in 1 publication for WB studies .

Cancer Biology

  • Colon Cancer:

    • Overexpression of GSPT1 promotes G1 to S phase transition by regulating CyclinD1, CDK4/6, and p21/p27 pathways .

    • Depletion reduces tumor cell proliferation, migration, and invasion (HCT116/SW480 models) .

  • Therapeutic Potential:

    • GSPT1 degraders induce TP53-independent cell death in acute myeloid leukemia (AML), leveraging translation initiation dependencies .

Translation Termination

  • 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 .

Neurological Roles

  • A novel Rac1-GSPT1 signaling pathway regulates astrogliosis (CNS injury response) .

Disease Associations

  • Duane-Radial Ray Syndrome: GSPT1 mutations linked to congenital malformations .

  • Myelodysplastic Syndrome: Implicated in hematopoietic stem cell dysfunction .

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 time may vary depending on the purchasing method and location. Please contact your local distributor for specific delivery information.
Synonyms
GSPT1 antibody; ERF3AEukaryotic peptide chain release factor GTP-binding subunit ERF3A antibody; Eukaryotic peptide chain release factor subunit 3a antibody; eRF3a antibody; G1 to S phase transition protein 1 homolog antibody
Target Names
GSPT1
Uniprot No.

Target Background

Function
GSPT1 (eRF3) plays a crucial role in translation termination, specifically in response to the stop codons UAA, UAG, and UGA. It enhances the activity of ETF1 and is involved in regulating mammalian cell growth. GSPT1 is also a component of the transient SURF complex, which facilitates the recruitment of UPF1 to stalled ribosomes during nonsense-mediated decay (NMD) of mRNAs containing premature stop codons. Additionally, GSPT1 is essential for SHFL-mediated translation termination, which inhibits programmed ribosomal frameshifting (-1PRF) of mRNA from viruses and cellular genes.
Gene References Into Functions
  1. PABP promotes the efficient binding of the eRF1-eRF3 complex to the ribosome through interactions with the N-terminal domain of eRF3a, which itself actively participates in translation termination. PMID: 27418677
  2. Research indicates that the N-terminal glycine repeat of eRF3a influences eRF3a-PABP interaction. The eRF3a 12-GGC allele exhibits a reduced binding affinity for PABP. PMID: 26818177
  3. GAB2, GSPT1, TFDP2, and ZFPM1 have been identified as four novel susceptibility loci for testicular germ cell tumor. PMID: 26503584
  4. The survivin and eRF3 complex may play a role in spindle formation, chromosome segregation, and cytokinesis. PMID: 23377885
  5. Overexpression of ID1 in two different cell lines led to increased transcriptional levels of STMN3 and GSPT1, whereas depletion of ID1 resulted in reduced expression of these genes. PMID: 25028095
  6. Proteolytic cleavage of eRF3a and eRF3b into p-eRF3 releases an amino-terminal fragment containing a nuclear export signal, enabling the relocalization of eRF3 into the nucleus to interact with p14ARF. PMID: 24569073
  7. ERF3 is targeted for caspase-mediated proteolytic cleavage and degradation during apoptosis induced by DNA damage. PMID: 23054082
  8. Studies have shown that the presence of the longer allele of eRF3a is associated with a threefold increased risk of developing breast cancer. PMID: 22101789
  9. The overlapping ERF3 PAM2 motifs have a biological role in regulating the accessibility of deadenylase to PABPC1 at the 3' end of poly(A). PMID: 23019593
  10. Crystal structures have been determined for the MLLE domain from PABPC1 in complex with the two PAM2 regions of eRF3. PMID: 20418951
  11. GSPT1/eRF3 undergoes proteolytic processing to generate an IAP-binding protein. PMID: 12865429
  12. Human translation termination factor eRF3/GSPT1 is an interacting partner of RNase L. PMID: 15908960
  13. Polyglycine expansions in eukaryotic translation release factor 3 have been linked to an increased susceptibility to gastric cancer. PMID: 15987717
  14. eRF3a regulates the formation of the termination complex by modulating eRF1 protein stability. PMID: 15987998
  15. Further research has identified a novel complex containing the NMD factors SMG-1 and Upf1, as well as the translation termination release factors eRF1 and eRF3 (SURF). An association between SURF and the EJC is required for SMG-1-mediated Upf1 phosphorylation and NMD. PMID: 16452507
  16. GSTP1 inhibits the binding of MAP kinase kinase kinase 5 (MAP3K5) to the 14-3-3 protein, a MAP3K5 inhibitor, while GSPT1 plays a role in regulating apoptosis signal-regulating kinase 1 (ASK1)-mediated apoptosis. PMID: 17700517
  17. eRF3a is degraded by the proteasome when not associated with eRF1. Proteasomal degradation of eRF3a regulates translation termination complex formation by adjusting the eRF3a level to that of eRF1. PMID: 18083835
  18. The eRF3a/GSPT1 12-GGC allele increases the susceptibility for breast cancer development. PMID: 19424636
  19. The GSPT1 GGC(12) allele was found in 2.2% of colorectal cancer patients but was absent in Crohn disease patients and the control group. PMID: 19963113

Show More

Hide All

Database Links

HGNC: 4621

OMIM: 139259

KEGG: hsa:2935

STRING: 9606.ENSP00000398131

UniGene: Hs.528780

Protein Families
TRAFAC class translation factor GTPase superfamily, Classic translation factor GTPase family, ERF3 subfamily

Q&A

What are the most reliable approaches for validating GSPT1 antibody specificity?

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 MethodExperimental ApproachExpected Result
Western BlotLysates from control vs GSPT1-KO/KD cellsSingle band at 80-85 kDa that disappears/diminishes in KO/KD samples
ImmunofluorescenceControl vs GSPT1-KO/KD cellsCytoplasmic staining that disappears in KO/KD samples
Peptide competitionPre-incubation with immunogenic peptideSignal elimination in all applications

How should researchers select between monoclonal and polyclonal GSPT1 antibodies for different experimental 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

  • May be less affected by background in specific tissues

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

How do epitope selection and target regions affect GSPT1 antibody performance in experimental applications?

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 .

What are the optimal protocols for using GSPT1 antibodies in cancer tissue immunohistochemistry?

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:

    • TE buffer at pH 9.0 (recommended as primary option)

    • Citrate buffer at pH 6.0 (alternative option)

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:

    • For polyclonal antibodies (e.g., 10763-1-AP): 1:50-1:500 dilution

    • For monoclonal antibodies: Follow manufacturer's recommended dilution

  • 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 .

How can researchers effectively optimize GSPT1 antibody protocols for flow cytometry applications?

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)

What are the most effective protocols for immunofluorescence detection of GSPT1 in different cell types?

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:

    • For polyclonal antibodies: 1:200-1:800 dilution

    • For monoclonal conjugated antibodies: 1:50-1:500 dilution

  • 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:

Cell TypeValidated AntibodiesSpecial Considerations
HeLa cellsCL488-68217, 10763-1-APStrong cytoplasmic expression, good positive control
HepG2 cellsCL488-68217Effective for hepatocellular studies
MCF-7 cells10763-1-APValidated for breast cancer studies
Glioblastoma cellsMultiple antibodiesExpression varies between cell lines (U87 > LN229)

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 .

How can GSPT1 antibodies be used to investigate its role in cancer cell proliferation and apoptosis?

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

    • Research finding: GSPT1 overexpression decreases G1 phase cells and increases S and G2/M phase cells, confirming its role in G1 to S transition

  • Proliferation marker co-staining: Combine GSPT1 immunostaining with Ki67 to assess correlation with proliferative activity

    • Research evidence: GSPT1 knockdown tumors show fewer Ki67-positive cells compared to control tumors

  • Growth assays following GSPT1 modulation: Use GSPT1 antibodies to confirm knockdown/overexpression efficiency before assessing proliferation

    • Data: GSPT1 knockdown decreased cell viability by over 50% in colorectal cancer cells

Apoptosis Investigation:

  • Apoptosis marker co-detection: Use GSPT1 antibodies alongside apoptosis markers like cleaved PARP1 and cleaved caspase-3

    • Research finding: GSPT1-KO U87 cells show enhanced cleaved PARP1 expression compared to wild-type cells

  • In vivo apoptosis assessment: Use GSPT1 antibodies to confirm expression status in tumor models while measuring apoptotic markers

    • Evidence: Brain tumors from GSPT1-KO U87 cells showed significantly higher cleaved caspase-3 positive areas compared to wild-type or GSPT1-rescued tumors

  • Drug sensitivity studies: Combine GSPT1 antibodies with apoptosis-inducing compounds

    • Research data: GSPT1-KO U87 cells showed enhanced sensitivity to staurosporine-induced apoptosis

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

What methodologies effectively utilize GSPT1 antibodies to study its potential as a therapeutic target in glioblastoma?

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

How can researchers use GSPT1 antibodies to investigate the discrepancy between mRNA and protein expression in cancer tissues?

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:

    • Perform immunohistochemistry with GSPT1 antibodies on serial tissue sections

    • Extract RNA from adjacent sections for RT-PCR analysis

    • Research finding: Studies in glioblastoma patients revealed no association between GSPT1 immunostaining levels and relative mRNA expression values

  • 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:

    • Use GSPT1 antibodies to track protein levels following transcription or translation inhibition

    • Determine protein half-life through cycloheximide chase experiments

    • Western blot analysis with GSPT1 antibodies (recommended dilution 1:2000-1:12000) provides quantitative measurement

  • 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.

What are the most common technical challenges when using GSPT1 antibodies, and how can they be resolved?

Researchers working with GSPT1 antibodies may encounter several technical challenges that can be systematically addressed:

Challenge 1: Inconsistent Western Blot Detection

  • 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

Challenge 2: High Background in Immunohistochemistry/Immunofluorescence

  • 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

Challenge 3: Inconsistent Results Between Different GSPT1 Antibodies

  • 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

Challenge 4: Flow Cytometry Signal Optimization

  • Problem: Weak intracellular GSPT1 signal

  • Solutions:

    • Ensure thorough permeabilization (0.1% Triton X-100 for 15 minutes)

    • Optimize antibody concentration (0.40 μg per 10^6 cells for conjugated antibodies)

    • Extend incubation time (45-60 minutes at room temperature)

    • Use signal amplification systems for unconjugated primary antibodies

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 .

How should researchers interpret GSPT1 antibody staining patterns in heterogeneous tumor samples?

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:

    • Use a standardized scoring system (0=negative, 1=weak, 2=moderate, 3=strong)

    • Research finding: Glioblastoma patients show variable GSPT1 expression from strong to moderate/weak cytoplasmic staining

  • 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 .

How can researchers distinguish between specific and non-specific binding when using GSPT1 antibodies in complex tissue samples?

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:

    • GSPT1 knockout/knockdown tissues or cells as negative controls

    • Research application: GSPT1-KO U87 cells showed negative staining compared to wild-type cells, providing definitive validation

    • GSPT1-rescued cells (GSPT1-KO cells with reintroduced GSPT1) restore staining, confirming specificity

  • 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:

    • Use antibodies targeting different GSPT1 epitopes

    • Consistent staining patterns across antibodies suggest specificity

    • Example: Compare monoclonal antibody CL488-68217 results with polyclonal antibody 10763-1-AP

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:

    • Test serial dilutions to identify optimal concentration

    • Follow validated dilution ranges (e.g., 1:50-1:500 for IHC)

    • Use the lowest concentration giving specific signal

  • 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:

    • Internal negative controls: GSPT1 is absent in vascular tissue within samples

    • Expected positive regions: Strong expression in epithelial tumor cells

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 .

How can researchers effectively use GSPT1 antibodies to study its role in translation termination and mRNA degradation?

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:

    • Use GSPT1 antibodies to pull down protein complexes

    • Identify interaction partners like eRF1

    • Western blot analysis: Use 1:2000-1:12000 dilution of anti-GSPT1 antibodies

    • Research insight: GSPT1 functions as a GTP-dependent stimulator of eRF1 in translation termination

  • 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):

    • Use GSPT1 antibodies to isolate ribonucleoprotein complexes

    • Identify associated mRNAs through sequencing or qPCR

    • Research basis: GSPT1 plays a key role in initiating mRNA degradation machinery during ribosome recycling

  • Immunofluorescence co-localization with P-bodies:

    • Use GSPT1 antibodies (1:200-1:800 dilution) for immunofluorescence

    • Co-stain with P-body markers

    • Analyze co-localization during normal conditions and stress responses

  • Pulse-chase experiments:

    • Label newly synthesized proteins

    • Use GSPT1 antibodies to track translation termination efficiency

    • Compare wild-type cells with GSPT1-depleted cells

    • Research application: GSPT1-KO cells show disrupted translation termination patterns

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

What are the recommended approaches for using GSPT1 antibodies in high-throughput screening of potential therapeutic compounds?

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:

    • Use fluorescently labeled GSPT1 antibodies (e.g., CoraLite® Plus 488)

    • Automated image acquisition and analysis

    • Measure changes in:

      • GSPT1 protein levels

      • Subcellular localization

      • Co-localization with interaction partners

  • AlphaLISA proximity assays:

    • Pair GSPT1 antibodies with antibodies against potential degradation machinery

    • Screen for compounds that promote GSPT1-ubiquitin ligase interaction

    • Research basis: Cereblon modulators like CC-885 induce GSPT1 degradation by bridging it to CRL4 E3 ubiquitin ligase

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:

    • Select diverse cancer cell lines with variable GSPT1 expression

    • Research finding: U87 glioblastoma cells show higher GSPT1 expression than LN229 cells

    • Correlate compound sensitivity with baseline GSPT1 levels detected by antibodies

  • 3D organoid models:

    • More physiologically relevant than 2D culture

    • Use GSPT1 antibodies for immunofluorescence (1:200-1:800 dilution) to monitor effects in 3D context

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:

    • Measure effects on translation termination efficiency

    • Assess impact on cell cycle progression (relevant to GSPT1's role in G1-S transition)

    • Monitor apoptosis markers like cleaved PARP1 (elevated in GSPT1-depleted cells)

  • 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.

How can researchers integrate GSPT1 antibody-based techniques with genomic and transcriptomic approaches for comprehensive cancer studies?

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:

    • Characterize GSPT1 protein expression using antibodies

    • Correlate with genomic alterations and response to therapy

    • Research basis: GSPT1 expression varies across glioblastoma patients and may affect therapeutic response

  • Tissue microarray analysis with multi-parameter readouts:

    • GSPT1 immunohistochemistry (1:50-1:500 dilution)

    • Genomic analysis of same samples

    • Digitally quantify GSPT1 expression and correlate with genomic features

    • Statistical modeling of relationships between GSPT1 protein levels and genomic/clinical data

  • 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

What emerging applications for GSPT1 antibodies are being developed beyond traditional cancer research?

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:

    • Monitor GSPT1 expression patterns across developmental stages

    • Cross-reactivity of antibodies with mouse, rat, and zebrafish samples enables comparative studies

Emerging Therapeutic Areas:

  • Targeted protein degradation approaches:

    • GSPT1 degraders represent a new therapeutic modality

    • Antibodies provide critical tools for measuring degradation efficiency

    • Research basis: Cereblon modulators like CC-885 induce GSPT1 degradation

  • 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 .

What methodological advances are improving the specificity and sensitivity of GSPT1 antibody-based techniques?

Recent methodological advances are significantly enhancing the specificity and sensitivity of GSPT1 antibody-based techniques:

Advanced Validation Technologies:

  • Enhanced genetic validation:

    • CRISPR/Cas9-mediated GSPT1 knockout cells provide definitive negative controls

    • Research application: GSPT1-KO U87 cells with rescued GSPT1 expression offer paired controls

    • Commercial antibodies increasingly validated using genetic approaches (e.g., RNAi knockdown)

  • Multi-epitope targeting strategies:

    • Use of antibody panels targeting different GSPT1 epitopes

    • Confirmation of findings with antibodies recognizing distinct regions:

      • N-terminal regions (AA 1-637)

      • Internal regions (AA 138-420, 390-499)

      • C-terminal domains

  • Orthogonal validation methods:

    • Combining antibody-based detection with mass spectrometry

    • Validation across multiple applications (WB, IHC, IF)

    • Research approach: 16 publications validating polyclonal antibody 10763-1-AP in Western blot applications

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:

    • Enhanced sensitivity for immunohistochemistry applications

    • Reduces background compared to traditional avidin-biotin methods

    • Optimizes detection of variable GSPT1 expression in heterogeneous samples

  • 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:

    • Epitope-preserving fixation methods

    • Dual fixation approaches (combining crosslinking and precipitating fixatives)

    • Application-specific optimization (e.g., TE buffer pH 9.0 for IHC)

  • Digital pathology quantification:

    • Automated scoring of GSPT1 immunohistochemistry

    • Machine learning algorithms for pattern recognition

    • Reduces inter-observer variability in expression assessment

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.