TERF2 (also known as TRF2) is a 542 amino acid protein containing an HTH myb-type DNA-binding domain that localizes primarily in the nucleus. It serves as a critical component of the shelterin complex (telosome) that binds to telomeric double-stranded 5'-TTAGGG-3' repeats .
TERF2's biological significance stems from its central role in:
Telomere maintenance and protection against chromosome end-to-end fusion
Recruiting factors required for telomere protection, including TERF2IP/RAP1 and DCLRE1B/Apollo
Telomeric loop (T-loop) formation by generating 3' single-stranded overhangs at leading end telomeres
Regulating telomere topology during replication by controlling DNA topoisomerase activity (TOP1, TOP2A, TOP2B)
Beyond telomeric functions, recent research has revealed TERF2's involvement in:
Regulating gene expression through binding to interstitial telomeric sequences
Modulating immune responses and angiogenesis
Controlling microRNA expression
Regulating macroautophagy/autophagy through interaction with HMGB1
TERF2 antibodies have been validated for multiple experimental applications:
Research applications include studying:
Telomere maintenance mechanisms
Cancer progression (TERF2 is overexpressed in multiple cancer types)
Immune cell infiltration in tumors
Protein-protein interactions (particularly with telomeric proteins)
Commercial TERF2 antibodies exhibit the following characteristics:
When selecting an antibody for specific applications, researchers should consider reactivity demonstrated in:
For maximum stability and efficacy:
Storage Conditions:
Store at -20°C (most commercial antibodies remain stable for one year after shipment)
Aliquoting is generally unnecessary for -20°C storage
Some preparations (typically smaller 20μl sizes) contain 0.1% BSA as a stabilizer
Storage Buffer Composition:
Typically PBS with 0.02% sodium azide and 50% glycerol, pH 7.3
Avoid repeated freeze-thaw cycles
Handling Guidelines:
Allow antibody to equilibrate to room temperature before opening
For dilution, use appropriate buffer recommended for each application
For IHC applications with TERF2 antibodies, antigen retrieval is crucial:
Working Dilution Preparation:
Prepare fresh working dilutions on the day of the experiment
Discard any unused diluted antibody
Western Blot Protocol for TERF2 Detection:
Sample Preparation:
Electrophoresis and Transfer:
Blocking and Antibody Incubation:
Detection Considerations:
Immunohistochemistry Protocol for TERF2:
Tissue Preparation:
Antigen Retrieval (Critical Step):
Staining Procedure:
Analysis Guidelines:
Distinguishing between TERF2 variants requires specific methodological approaches:
Full-Length vs. Truncated Forms:
Post-Translational Modifications:
Phosphorylation: Use phospho-specific antibodies or treat samples with phosphatase
Ubiquitination: Use immunoprecipitation followed by ubiquitin-specific antibodies
Acetylation: Employ acetylation-specific antibodies
Domain-Specific Analysis:
Research Application Example:
In autophagy studies, researchers distinguished TERF2's functional forms by:
Positive Controls:
Cell Lines with Known TERF2 Expression:
Tissue Sections:
Genetic Controls:
Negative Controls:
Antibody Controls:
Isotype-matched irrelevant antibody (same concentration)
Primary antibody omission
Expression Controls:
Application-Specific Controls:
For IP experiments: Use non-immune IgG from same species
For ChIP: Include IgG control and primers for non-bound regions
For dual labeling: Single antibody controls
Technical Validation:
Cross-reactivity assessment: Test on samples known to lack TERF2
Peptide competition assay: Pre-incubate antibody with immunizing peptide
Antibody titration: Perform dilution series to establish optimal concentration
TERF2 expression and mutations vary across cancer types, affecting antibody-based detection:
Altered Expression Patterns:
Detection Challenges in Cancer Tissues:
Heterogeneous expression requires careful interpretation
Higher antibody concentrations may be needed for low-expression regions
Background staining must be distinguished from specific signal
Mutation Impact on Epitope Recognition:
Technical Considerations for Cancer Samples:
For IHC scoring in cancer tissues:
For WB analysis:
Include stage-matched samples when comparing expression
Account for tumor heterogeneity through multiple sampling
TERF2 antibodies are valuable tools for investigating telomere biology through advanced methodological approaches:
Chromatin Immunoprecipitation (ChIP) for Telomere Analysis:
Crosslink proteins to DNA using formaldehyde (1%, 10 min)
Sonicate chromatin to ~500 bp fragments
Immunoprecipitate with TERF2 antibody
Analyze by qPCR using telomere-specific primers or dot blot with telomeric probe
Include input and IgG controls
Co-Immunoprecipitation for Shelterin Complex Analysis:
Immunofluorescence for Telomere Dysfunction-Induced Foci (TIF):
Co-stain for TERF2 and DNA damage markers (γ-H2AX, 53BP1)
Protocol example:
Quantify co-localization as indicator of telomere dysfunction
Telomere Dysfunction Analysis in Cancer Models:
TERF2's complex role in tumorigenesis requires sophisticated research approaches:
Expression Analysis Across Cancer Types:
Pan-cancer analysis reveals variable expression patterns:
Methodology:
Compare matched tumor/normal samples
Stratify by cancer stage and subtype
Correlate with clinical outcomes
TERF2 Knockdown/Overexpression in Cancer Models:
In Vivo Cancer Model Analysis:
Orthotopic injection model:
Markers to assess in tumor samples:
TERF2 and Tumor Immune Infiltration Analysis:
IHC multiplex staining for TERF2 and immune cell markers
Correlation analysis between TERF2 expression and:
Chemokine expression analysis:
Recent research has revealed TERF2's involvement in non-telomeric functions, requiring specialized methodological approaches:
TERF2-HMGB1 Interaction in Autophagy Regulation:
Co-immunoprecipitation protocol:
Lyse cells in non-denaturing buffer
Immunoprecipitate with anti-TERF2 or anti-HMGB1 antibodies
Western blot to detect interacting partner
Co-localization analysis:
TERF2's Role in Autophagy Assessment:
TERF2 silencing approach:
Transfect with siTERF2 (100 nM, 24h)
Autophagy stimulation: EBSS nutrient starvation
Autophagy markers analysis:
Interstitial Telomeric Sequence Binding:
ChIP-seq methodology:
Immunoprecipitate with TERF2 antibody
Sequence and analyze for non-telomeric binding sites
Validate with ChIP-qPCR for selected targets
Gene expression analysis:
Compare expression profiles in TERF2-depleted vs. control cells
Correlate with TERF2 binding at specific loci
TERF2 and microRNA Regulation:
RNA immunoprecipitation (RIP):
Crosslink RNA-protein complexes
Immunoprecipitate with TERF2 antibody
Extract and analyze associated RNAs
miRNA expression analysis:
Compare miRNA profiles in TERF2-manipulated cells
Validate key targets with qRT-PCR
TERF2's interactions with multiple proteins present specific technical challenges:
Preservation of Native Interactions:
Challenge: TERF2 interactions may be disrupted during extraction
Solution:
Use gentle lysis buffers (low detergent concentration)
Employ in situ proximity ligation assay (PLA) for direct visualization
Consider crosslinking approaches for transient interactions
Nuclear Protein Extraction Efficiency:
Challenge: TERF2 is predominantly nuclear, requiring efficient nuclear extraction
Solution:
Optimize nuclear extraction protocol
Verify extraction efficiency with nuclear markers
Include DNase treatment to release DNA-bound proteins
Distinguishing Direct vs. Indirect Interactions:
Challenge: Co-IP may detect both direct and indirect interactions
Solution:
Perform reciprocal IPs
Use domain-specific mutants (e.g., TERF2ΔC)
Employ in vitro binding assays with purified proteins
Domain-Specific Interaction Mapping:
Validation of Novel Interactions:
Challenge: Confirming biological relevance of detected interactions
Solution:
Functional validation through knockdown/overexpression
Subcellular co-localization studies
Mutational analysis of interaction interfaces
The emerging role of TERF2 in immune regulation requires specialized approaches:
Correlation Analysis of TERF2 and Immune Cell Markers:
Multiparametric Analysis of Immune Markers:
Assess correlation between TERF2 and immune markers in tumor samples (Table from search result 5):
| Immune Cell Type | Marker | Correlation with TERF2 in ESCA Tumor | Correlation in Normal Tissue |
|---|---|---|---|
| T cells (CD8+) | CD8A, CD8B | No significant correlation | Positive in normal tissue (GETX) |
| T cells (general) | CD2, CD3E | No significant correlation | Positive in normal tissue |
| Natural Killer cells | KIR2DL4, KIR3DL2 | No significant correlation | Positive in normal tissue |
| Th1 cells | T-bet | No significant correlation | Positive |
| Th2 cells | STAT5A, STAT6 | No significant correlation | Strongly positive (p<0.01) |
| Tfh cells | BCL6 | Positive (p<0.01) | Strongly positive (p<0.01) |
| Th17 cells | STAT3 | Strongly positive (p<1e-6) | Strongly positive (p<1e-4) |
| T cell exhaustion | PD-1, LAG3, TIM-3 | No significant correlation | Positive for TIM-3 (p<0.05) |
Chemokine Correlation Analysis:
Experimental Validation in Immune Contexts:
In vitro approaches:
Co-culture TERF2-manipulated cancer cells with immune cells
Measure immune activation markers and effector functions
Assess cytokine/chemokine production
In vivo approaches:
Immunocompetent mouse models with TERF2-manipulated tumors
IHC analysis of tumor microenvironment
Flow cytometry of tumor-infiltrating immune cells
Methodological approaches to resolve inconsistencies:
Between antibody clones:
Test multiple antibodies targeting different epitopes
Compare monoclonal (for specificity) and polyclonal (for sensitivity) options
Verify with genetic validation (siRNA knockdown)
Between detection methods:
Between cell/tissue types:
Adjust protocols for specific sample types:
Cell lines: Optimize lysis conditions
FFPE tissues: Extend antigen retrieval time
Fresh tissues: Adjust fixation parameters
Consider tissue-specific expression levels and adjust antibody concentration
Between in vitro and in vivo results:
Account for microenvironment influences
Compare primary cells with established cell lines
Validate findings across multiple experimental models
Advanced technological approaches:
Single-cell protein analysis:
Mass cytometry (CyTOF) for multi-parameter analysis
Imaging mass cytometry for spatial protein detection
Single-cell Western blotting for heterogeneity assessment
Super-resolution microscopy:
Visualize TERF2 localization at telomeres with nanometer precision
Techniques include STORM, PALM, and STED microscopy
Enables co-localization studies with other telomeric proteins
Proximity-based protein interaction detection:
BioID or APEX2 proximity labeling
FRET/BRET for real-time interaction monitoring
In situ proximity ligation assay for visualizing interactions
CRISPR-based approaches:
Endogenous tagging of TERF2 for live-cell imaging
CUT&RUN or CUT&Tag for improved chromatin binding profile
CRISPR interference/activation for functional studies
Combinatorial approaches for cancer studies:
Multiplex IHC for simultaneous detection of TERF2 and immune markers
Spatial transcriptomics combined with protein detection
Patient-derived organoids for personalized functional studies