GTSE1 is a microtubule-associated protein that regulates cell cycle progression, particularly during G2 and S phases. It has emerged as a significant oncogenic factor across multiple cancer types. GTSE1 promotes malignant progression by:
Enhancing cell proliferation through modulation of cell cycle transition, particularly G1/S phase
Promoting cell migration and invasion via regulation of epithelial-mesenchymal transition (EMT)
Contributing to chemoresistance, particularly to cisplatin in clear cell renal cell carcinoma (ccRCC)
Correlating with immune cell infiltration in tumor microenvironments
Research significance: GTSE1 overexpression correlates with poor clinical outcomes in multiple cancers, making it a valuable prognostic biomarker and potential therapeutic target .
GTSE1 antibodies are versatile tools employed across multiple research applications:
Methodological approach: Selection of application should be guided by research question. For protein level quantification, WB is preferred; for spatial distribution in tissues, IHC/IF is optimal; for cell-by-cell analysis in heterogeneous populations, flow cytometry is recommended .
GTSE1 shows distinctive expression patterns that vary between normal and malignant tissues:
Normal tissues:
Low expression in G1 phase cells of non-transformed cell lines
Minimal detection during G1, with levels increasing during S phase and peaking in G2/M
Primary expression in proliferating tissues
Cancer tissues:
Significantly overexpressed in multiple cancer types, including:
Elevated expression across all cell cycle phases in transformed cells compared to normal cells
Particularly notable: GTSE1 is abundant in G1 phase of cancer cells, whereas it's nearly undetectable in G1 of normal cells
Methodological significance: When designing experiments, researchers should account for cell cycle phase-specific expression patterns and use appropriate synchronization techniques to accurately compare GTSE1 levels between normal and cancer samples .
Rigorous validation is crucial for reliable GTSE1 antibody-based experiments. Implement these methodological approaches:
Positive and negative controls:
Multiple antibody validation:
Specific validation techniques:
Cell cycle-specific validation:
Research applications demonstrate that proper validation can resolve discrepancies in reported molecular weights (66-77 kDa vs. 100 kDa) that may result from post-translational modifications or splice variants .
Successful GTSE1 detection requires optimization strategies tailored to sample type:
For Western blotting:
Protein extraction: RIPA lysis buffer containing 0.1M PMSF and 1% protease/phosphatase inhibitors
Sample preparation: Denature by boiling for 10min with 5x loading buffer
Membrane selection: 0.45μm PVDF membranes show superior protein retention
Primary antibody incubation: Overnight at 4°C after blocking with 5% nonfat milk
For IHC in tissue samples:
Antigen retrieval: TE buffer pH 9.0 or citrate buffer pH 6.0
Scoring system: Combine staining intensity (0-3+) with percentage of positive cells (0-4)
Final scoring calculation: Negative (0), weak (1-4), moderate (5-8), strong (9-12)
For cell line immunofluorescence:
Methodological insight: The expression pattern of GTSE1 is highly cell cycle-dependent, so researchers should consider cell synchronization or co-staining with cell cycle markers for accurate interpretation .
GTSE1 antibodies detect varying molecular weights (66-77 kDa, sometimes 100 kDa), which can create interpretation challenges . To resolve such discrepancies:
Understand biological factors affecting detection:
Technical approach to resolution:
Use phosphatase treatment on some samples to determine if phosphorylation causes the shift
Include positive control lysates from characterized cell lines (HeLa, MCF-7)
Use gradient gels for better separation of higher molecular weight proteins
Consider using knockout/knockdown controls to verify specificity
Analytical considerations:
Document exact experimental conditions when observing different molecular weights
Consider using mass spectrometry to identify the exact protein being detected
Cross-reference with transcriptomic data to identify potential splice variants
Research context: The electrophoretic mobility of GTSE1 increases during mitotic exit, suggesting dynamic post-translational modifications throughout the cell cycle .
GTSE1 antibodies enable mechanistic studies of cancer progression through multiple approaches:
EMT regulation investigation:
Cell cycle dynamics:
Migration and invasion studies:
Tumor microenvironment analysis:
Research data shows GTSE1 silencing significantly reduced colony formation (QGY-7703: 529.67 ± 59.53 vs. 262.67 ± 21.385, P = 0.002; SMMC-7721: 416.33 ± 21.962 vs. 139.00 ± 5.292, P = 0.001), while overexpression increased colony formation (QGY-7703: 102.33 ± 11.68 vs. 168.33 ± 9.29, P = 0.002) .
GTSE1 contributes to chemoresistance in multiple cancers, particularly cisplatin resistance in ccRCC . Recommended experimental approaches include:
Drug sensitivity assays:
Mechanistic pathway investigation:
Gene expression correlation analysis:
Analyze correlation between GTSE1 and DNA repair genes
Methodology: qRT-PCR validation of GTSE1-associated gene expression patterns
Clinical correlation approaches:
Research context: Functional enrichment analysis indicates GTSE1 and its co-expressed genes relate to cell cycle, DNA replication, and immunoreaction through multiple signaling pathways, including P53 and T-cell receptor signaling .
Recent research reveals significant correlations between GTSE1 expression and immune infiltration . Methodological approaches include:
Multiplex immunohistochemistry:
Flow cytometry-based approaches:
Combine GTSE1 staining with immune cell markers for single-cell analysis
Methodology: Dual or triple staining protocols to correlate GTSE1 with specific immune populations
Correlation analysis with immune checkpoint molecules:
Research data shows significant correlations between GTSE1 expression and:
B cells (r = 0.278, p = 1.9e−06)
CD8+ T cells (r = 0.165, p = 5.43e−04)
CD4+ T cells (r = 0.251, p = 4.89e−08)
Macrophages (r = 0.165, p = 4.44e−04)
Neutrophils (r = 0.285, p = 5.47e−10)
High GTSE1 expression correlates with increased infiltration of T cells (CD8+, follicular helper, Tregs), monocytes, macrophages (M0, M1, M2), resting dendritic cells, and neutrophils .
Robust controls are critical for reliable GTSE1 biomarker studies:
Tissue controls:
Cell line controls:
Technical controls:
Antibody validation: Omit primary antibody, use isotype control
Quantification controls: Include calibration standards for densitometry analysis
Reproducibility controls: Technical and biological replicates
Cell cycle-specific controls:
Methodological approach: When scoring GTSE1 expression in tissue samples, combine staining intensity (0-3+) with percentage of positive cells (0-4) for a comprehensive score .
When working with challenging samples, consider these optimization strategies:
Formalin-fixed, paraffin-embedded (FFPE) tissues:
Extended antigen retrieval: TE buffer pH 9.0 or citrate buffer pH 6.0
Signal amplification: Consider tyramide signal amplification for low abundance detection
Background reduction: Extended blocking (5% BSA or 10% normal serum)
Antibody concentration: Test higher concentrations (1:20-1:50) with extended incubation
Archived or degraded samples:
Cell lines with variable expression:
Multiplexed detection:
Sequential immunostaining with careful antibody stripping between rounds
Spectral unmixing for fluorescent detection of multiple markers
Consider using antibodies raised in different host species to avoid cross-reactivity
Research context: IHC scoring systems should account for both intensity and percentage of positive cells for comprehensive assessment of GTSE1 expression .
Accurate GTSE1 quantification requires careful technical considerations:
Standardized scoring methods for IHC:
Western blot quantification:
Transcriptional analysis integration:
Statistical analysis approaches:
Research application: In ccRCC tissue microarray analysis, GTSE1 IHC scoring successfully distinguished expression differences between tumor and normal tissues, correlating with clinical parameters and prognosis .
GTSE1 antibodies can facilitate several approaches to therapeutic development:
Target validation strategies:
Use antibodies to confirm GTSE1 overexpression in patient-derived samples
Correlate expression with treatment response in retrospective studies
Develop tissue microarray screening to identify patient populations most likely to benefit
Mechanism-based drug development:
Combination therapy approaches:
Biomarker development:
Standardize GTSE1 IHC protocols for patient stratification
Develop multiplexed assays combining GTSE1 with other prognostic markers
Create companion diagnostic approaches for future GTSE1-targeted therapies
Research significance: The association between GTSE1 and cisplatin resistance in ccRCC suggests potential as a predictive biomarker for treatment response and as a target to overcome resistance .
Developing GTSE1 as a clinical biomarker faces several technical and biological challenges:
Antibody standardization issues:
Different antibodies target different epitopes with varying specificity
Need for standardized protocols across laboratories
Validation across multiple sample types and preservation methods
Expression pattern complexity:
Cell cycle-dependent expression complicates interpretation
Heterogeneous expression within tumors requires careful sampling
Need to establish clinically relevant cutoff values for "high" vs. "low" expression
Technical standardization needs:
Automated vs. manual scoring systems
Digital pathology integration for quantitative assessment
Quality control measures for clinical laboratory implementation
Biological context considerations:
GTSE1's multiple biological functions (cell cycle, migration, EMT) require careful interpretation
Integration with other biomarkers for comprehensive assessment
Accounting for tumor microenvironment influences on expression
Research context: Studies have demonstrated GTSE1's prognostic value in multiple cancers, including ccRCC and HCC, but standardized clinical protocols need development .
Integrative approaches combining GTSE1 with other -omics data offer powerful insights:
Multi-omics integration strategies:
Correlate GTSE1 protein expression (antibody-based) with transcriptomic data
Integrate with genomic alterations affecting the GTSE1 locus
Combine with epigenomic profiles to understand regulatory mechanisms
Pathway analysis approaches:
Single-cell analysis techniques:
Combine GTSE1 antibody-based detection with single-cell RNA sequencing
Study heterogeneity of expression within tumors
Relate to cell states and differentiation hierarchies
Clinical data integration:
Correlate GTSE1 expression with treatment response data
Develop predictive models combining GTSE1 with other molecular and clinical factors
Network analysis to identify key interacting partners for combination targeting
Research application: GTSE1 functional enrichment analysis has already identified associations with cell cycle regulation, DNA replication, and immune response pathways, providing a foundation for integrated multi-omics approaches .