The PTTG1 antibody was first characterized in 2001 using a polyclonal antiserum (SK601) generated against a His-tagged PTTG1 protein expressed in E. coli . Key features include:
Specificity: Detects recombinant PTTG1 in ELISA (titer 1:100,000) and Western blot (52 kDa band for GST-PTTG1 fusion protein).
Epitope Targeting: Recognizes both phosphorylated and unphosphorylated forms of PTTG1.
Validation: Confirmed via immunoprecipitation and immunohistochemistry in COS-7 cells transfected with PTTG1-GFP chimeric constructs .
A second antibody developed in 2011 targeted the C-terminal fragment (residues 108–202) of PTTG1, enabling subcellular localization studies. This antibody demonstrated colocalization with cis-Golgi markers (GM130) and centrosomal proteins (γ-tubulin) .
The SK601 antibody revealed intense PTTG1 staining in tumor tissues (breast, ovarian, testicular) but minimal staining in normal tissues except testis . This suggests utility as a diagnostic marker for malignancies.
In prostate cancer models, PTTG1 overexpression correlated with higher Gleason scores and tumor aggressiveness. The antibody confirmed PTTG1’s role in promoting cell cycle progression (G1 arrest reversal) and tumor formation in nude mice .
Using the C-terminal antibody, PTTG1 was localized to the cis-Golgi and centrosome, interacting with microtubule nucleation complexes (e.g., γ-tubulin, AKAP450) . This highlights its role in cell migration and cytoskeletal dynamics.
| Antibody | Epitope | Production Method | Applications |
|---|---|---|---|
| SK601 | Full-length PTTG1 | His-tagged protein in E. coli | ELISA, Western blot, IHC |
| C-terminal | Residues 108–202 | GST-fused fragment in E. coli | Immunofluorescence, subcellular localization |
The PTTG1 antibody has enabled critical insights into oncogenic mechanisms:
PTTG1 (also known as securin) functions as a key regulator of sister chromatid separation during cell division. It plays critical roles in normal physiological conditions, particularly in regulating human embryonic stem cells and maintaining the stem cell compartment . As a multifunctional protein, PTTG1 contains 202 amino acids with no significant similarity to other known proteins . Its expression is highly restricted in normal tissues, being predominantly expressed in testis with minimal expression in normal ovary and breast tissues .
The molecular weight profile of PTTG1 varies depending on its form:
These differences are important when validating antibody specificity in Western blotting applications, as the detection of bands at incorrect molecular weights could indicate non-specific binding or protein degradation.
PTTG1 is referenced under several alternative names in research papers and databases:
Researchers should be aware of these alternative designations when conducting literature searches to ensure comprehensive coverage of relevant studies.
PTTG1 shows a consistent pattern of upregulation during tumor progression. Studies examining normal ovary (NO) samples compared to benign tumor (BN), borderline tumor (BL), and high-grade tumors (HG) have documented a graded increase in PTTG1 expression . While there is considerable inter-patient variation, this pattern of progressive upregulation makes PTTG1 a potential biomarker for tumor progression. The expression increase is often concurrent with upregulation of cancer stem cell markers like CD133 and CD24 .
PTTG1 has been demonstrated to co-localize with multiple stem cell/cancer stem cell markers including:
This co-localization occurs in both normal ovarian samples and ovarian tumors at various stages. Notably, PTTG1 is highly expressed in ALDH1+ cancer stem cells compared to ALDH1- cells, suggesting it may function as a previously overlooked marker for stem cells and cancer stem cells . Research indicates PTTG1 may directly regulate self-renewal mechanisms and epithelial-mesenchymal transition (EMT) pathways in cancer stem cells.
Both in vitro and in vivo models have proven valuable for PTTG1 research:
When selecting a PTTG1 antibody, researchers should consider:
Epitope specificity: Some antibodies target the full-length protein (AA 1-202), while others target specific epitopes
Validated applications: Confirm the antibody has been validated for your specific application (WB, IHC, IP, etc.)
Host species: Consider compatibility with other antibodies for dual-labeling experiments
Reactivity: Ensure the antibody recognizes PTTG1 from your species of interest (human, mouse, etc.)
Published validation: Review literature using the antibody to assess reliability and reproducibility
Clonality: Monoclonal antibodies offer consistency, while polyclonal antibodies may provide increased sensitivity
Rigorous validation controls include:
Positive tissue controls: Testis tissue (known to express PTTG1)
Negative tissue controls: Normal breast or ovary tissue (minimal PTTG1 expression)
Recombinant protein: Purified PTTG1 protein as a positive control for Western blotting
Genetically modified cells: PTTG1 knockout/knockdown cells as negative controls
Overexpression systems: Cells transfected with PTTG1 expression constructs as positive controls
Peptide competition: Pre-incubation of antibody with immunizing peptide should abolish specific signal
| Application | Recommended Dilution Range | Optimization Strategy | Considerations |
|---|---|---|---|
| Western Blot | 1:1,000 - 1:5,000 | Titration series | Lower for weak expression; higher for abundant expression |
| IHC/ICC | 1:100 - 1:500 | Dilution series on positive control tissue | Optimize antigen retrieval method concurrently |
| ELISA | 1:10,000 - 1:100,000 | Checkerboard titration | Determine optimal coating concentration and antibody dilution |
| Flow Cytometry | 1:50 - 1:200 | Titration with positive control cells | Consider fixation and permeabilization effects |
| Immunoprecipitation | 1:50 - 1:200 | Antibody:lysate ratio optimization | Ensure antibody excess for complete target capture |
For optimal dual immunofluorescence co-localization:
Fixation method: Use 4% paraformaldehyde for 15-20 minutes to preserve epitope accessibility
Permeabilization: 0.1-0.3% Triton X-100 for intracellular antigens
Blocking: Use 5-10% serum from secondary antibody host species for 1 hour
Primary antibodies: Apply PTTG1 antibody alongside stem cell marker antibodies (ALDH1, CD44, etc.)
Secondary antibodies: Use species-specific secondaries with distinct fluorophores
Nuclear counterstain: DAPI for nuclear localization context
Controls: Single-antibody controls to verify absence of cross-reactivity
Imaging: Confocal microscopy for accurate co-localization analysis
Research has successfully demonstrated co-localization of PTTG1 with multiple stem cell markers in both ovarian surface epithelium and cortex regions in normal ovary and ovarian tumors .
To investigate PTTG1's regulatory functions in cancer stem cells:
Gene expression manipulation:
Functional assays:
Sphere formation assays to assess self-renewal capacity
Colony formation assays for clonogenicity
Invasion/migration assays to evaluate EMT properties
In vivo tumor initiation studies with limiting dilution analysis
Molecular analysis:
qRT-PCR for stem cell markers and self-renewal pathway genes
Western blotting for protein expression changes
ChIP-seq to identify PTTG1 binding sites on chromatin
RNA-seq for global transcriptional changes
Studies have shown that PTTG1 manipulation affects the expression of self-renewal mechanisms and EMT-related genes in cancer stem cells, suggesting its critical role in maintaining stemness properties .
| Approach | Methodology | Application | Considerations |
|---|---|---|---|
| Co-immunoprecipitation | Pull-down with PTTG1 antibody followed by Western blot | Protein-protein interactions | Use appropriate lysis buffers to preserve interactions |
| Mass spectrometry | IP-MS or TAP-MS | Interactome analysis and PTM identification | Requires high antibody specificity |
| Proximity ligation assay | In situ detection of protein-protein interactions | Visualization of interactions in tissue context | Needs highly specific antibodies for both proteins |
| Phospho-specific antibodies | Western blot or IHC with phospho-PTTG1 antibodies | Detection of phosphorylation status | Validate phospho-specificity extensively |
| FRET/BRET | Fluorescence/bioluminescence resonance energy transfer | Real-time interaction dynamics | Requires protein tagging which may affect function |
| ChIP-seq | Chromatin immunoprecipitation with PTTG1 antibody | DNA binding and transcriptional regulation | Optimize crosslinking and sonication conditions |
To minimize non-specific bands in Western blots:
Optimize blocking conditions: Try different blocking agents (5% milk, 5% BSA, or commercial blockers)
Increase washing stringency: More frequent washes with higher TBST concentration (0.1-0.3% Tween-20)
Titrate antibody concentration: Determine minimum effective concentration through serial dilutions
Use PVDF membranes: May provide better signal-to-noise ratio than nitrocellulose for some antibodies
Include protein extraction controls: Use protease inhibitors to prevent degradation products
Try alternative antibodies: Test antibodies targeting different epitopes
Use gradient gels: Better separation of proteins with similar molecular weights
Optimize transfer conditions: Adjust time and voltage for complete transfer of protein
| Antigen Retrieval Method | Protocol | Tissue Types | Considerations |
|---|---|---|---|
| Heat-induced (HIER) - Citrate | 10mM Citrate buffer (pH 6.0), 95-100°C for 20 minutes | FFPE ovarian tissue | Optimal for preserving morphology |
| Heat-induced (HIER) - EDTA | 1mM EDTA buffer (pH 8.0-9.0), 95-100°C for 20 minutes | FFPE testicular tissue | May give stronger signal for some epitopes |
| Enzymatic retrieval | Proteinase K (10-20 μg/ml) for 10-15 minutes at 37°C | Frozen sections | Use when heat-induced methods fail |
| Combined approach | Mild enzymatic treatment followed by HIER | Highly fixed tissues | For difficult tissues with extensive crosslinking |
Research has shown that proper antigen retrieval is critical for detecting PTTG1 in various tumor tissues, with heat-induced methods generally providing superior results .
When facing discrepancies between PTTG1 protein and mRNA levels:
Consider post-transcriptional regulation:
microRNA-mediated suppression of translation
RNA-binding protein effects on mRNA stability
Alternative splicing generating different isoforms
Evaluate post-translational regulation:
Protein stability differences (proteasomal degradation)
Subcellular localization affecting antibody accessibility
Post-translational modifications altering epitope recognition
Technical considerations:
Antibody specificity for particular isoforms
Sample preparation differences between protein and RNA extraction
Tissue heterogeneity and sampling differences
Research has noted high variation in PTTG1 expression levels between patients, suggesting complex regulatory mechanisms beyond transcriptional control .
PTTG1 shows significant potential as a therapeutic target based on several lines of evidence:
Knockdown effects: Various methods of PTTG1 downregulation have been shown to inhibit ovarian cell proliferation and suppress tumor growth in nude mice
Stem cell targeting: PTTG1's expression in cancer stem cells suggests targeting it could eliminate the tumor-initiating population
Signaling pathway modulation: PTTG1 mediates AKT activation, which is implicated in stemness and EMT properties of cancer cells
Differential expression: The minimal expression in most normal tissues compared to high expression in tumors provides a therapeutic window
Future therapeutic approaches may include siRNA-based therapies, small molecule inhibitors of PTTG1 or its interaction partners, and antibody-drug conjugates targeting PTTG1-expressing cells.
Emerging technologies with potential for PTTG1 research include:
Single-cell proteomics: Measuring PTTG1 protein levels in individual cells to assess heterogeneity
Spatial transcriptomics: Mapping PTTG1 mRNA expression with spatial resolution in tissue sections
Imaging mass cytometry: Multiplexed protein detection including PTTG1 with spatial information
CODEX multiplexed imaging: Simultaneous visualization of PTTG1 with dozens of other markers
Live-cell imaging with fluorescent PTTG1 reporters: Monitoring dynamic changes in expression
Organoid models: 3D culture systems to study PTTG1 in more physiologically relevant contexts
These approaches could reveal new insights into PTTG1's spatial distribution within the tumor microenvironment and its relationship to specific cellular niches.
Integrative multi-omics strategies for PTTG1 research:
Correlate PTTG1 protein expression with:
Genomic alterations (mutations, CNVs)
DNA methylation patterns at the PTTG1 promoter
Chromatin accessibility at PTTG1 regulatory regions
Global transcriptomic profiles
Metabolomic signatures
Data integration methods:
Network analysis to identify PTTG1-centered regulatory networks
Machine learning approaches to predict PTTG1-high tumor phenotypes
Systems biology modeling of PTTG1 pathway interactions
Patient stratification based on PTTG1 expression and multi-omics profiles
Clinical correlations:
Integrate PTTG1 expression with treatment response data
Develop predictive biomarker signatures incorporating PTTG1
Correlate with immune infiltration patterns and immunotherapy response
These integrative approaches could provide comprehensive understanding of PTTG1's role in tumor biology and identify novel therapeutic strategies targeting PTTG1-dependent pathways.