TERF2 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery times.
Synonyms
Telomeric DNA binding protein antibody; Telomeric DNA-binding protein antibody; Telomeric repeat binding factor 2 antibody; Telomeric repeat binding protein 2 antibody; Telomeric repeat-binding factor 2 antibody; TERF 2 antibody; Terf2 antibody; TERF2_HUMAN antibody; TRBF 2 antibody; TRBF2 antibody; TRF 2 antibody; TRF2 antibody; TTAGGG repeat binding factor 2 antibody; TTAGGG repeat-binding factor 2 antibody
Target Names
Uniprot No.

Target Background

Function
TERF2 (Telomeric Repeat Binding Factor 2) antibody binds to the telomeric double-stranded 5'-TTAGGG-3' repeat sequence. It plays a crucial role in telomere maintenance and preventing end-to-end fusion of chromosomes. Beyond its telomeric DNA-binding function, TERF2 is essential for recruiting various factors and enzymes involved in telomere protection, including the shelterin complex, TERF2IP/RAP1, and DCLRE1B/Apollo. TERF2 is a component of the shelterin complex (telosome), which regulates telomere length and safeguards chromosome ends. Shelterin binds to arrays of double-stranded 5'-TTAGGG-3' repeats added by telomerase, protecting chromosome ends from recognition as DNA damage. Without this protective mechanism, telomeres would be inappropriately processed by DNA repair pathways, compromising chromosomal stability. In collaboration with DCLRE1B/Apollo, TERF2 plays a key role in telomeric loop (T loop) formation by generating a 3' single-stranded overhang at the leading end of telomeres. T loops are thought to protect chromosome ends from degradation and repair. TERF2 is necessary for both recruiting DCLRE1B/Apollo to telomeres and activating its exonuclease activity. TERF2 preferentially binds to positively supercoiled DNA. Together with DCLRE1B/Apollo, it controls the amount of DNA topoisomerase (TOP1, TOP2A, and TOP2B) required for telomere replication during fork passage, preventing aberrant telomere topology. TERF2 recruits TERF2IP/RAP1 to telomeres, participating in the repression of homology-directed repair (HDR), which can affect telomere length. Overall, TERF2 plays a critical role in maintaining telomere integrity and protecting chromosomes from damage and degradation.
Gene References Into Functions
  1. ROC curve analysis identified TERF2 and RTA as promising markers in disease diagnosis. Univariate analysis of clinicopathological parameters and the genes TERF2 and RTA demonstrated significant association with disease progression and serve as reliable predictors of disease course, suggesting their potential utility in multiple myeloma (MM) prognosis. PMID: 29043869
  2. Both GEM and CAP induced telomere shortening and increased TRF2 expression, leading to improved ascites in nude mice and decreased in vitro clonogenic activity. TRF2 knockdown altered telomeres to a shortened but novel state, evading XPF-dependent telomere loss and conferring resistance to SKOV3.ip1 ascites cells against low-dose GEM and CAP. PMID: 28088627
  3. Research revealed that promoter binding by TRF2 mediates PCGF3 promoter activity, and both the N-terminal and C-terminal domains of TRF2 are crucial for this activity. PMID: 29589913
  4. The crystal structure of the N-terminal domain of TIN2 in complex with TIN2-binding motifs from TPP1 and TRF2 was determined, revealing how TIN2 interacts cooperatively with TPP1 and TRF2. PMID: 29160297
  5. Elevated levels of HULC and MALAT1 reduced the methylation of the TERC promoter, dependent on TRF2, increasing TERC expression and amplifying the interplay between TRET and TERC. PMID: 27782152
  6. RNA interference-mediated TERF2 knockdown had no effect on cell proliferation, migration, senescence, or apoptosis. However, it increased the expression of cytokines involved in inflammation and angiogenesis, except for vascular endothelial growth factor. PMID: 27329590
  7. Telomere stability is directly regulated by the major pro-oncogenic signaling pathway (RAS/RAF/MEK/ERK) via TRF2 phosphorylation. PMID: 27366950
  8. The B-domain enhances TRF2's interaction with DNA through enhanced long-range electrostatic interactions. PMID: 28981702
  9. Studies have identified TRF2 as a novel SIRT6 substrate, demonstrating that acetylation of TRF2 plays a crucial role in regulating TRF2 protein stability, providing a new avenue for modulating its expression during oncogenesis and damage response. PMID: 27923994
  10. Results suggest that dimerized TRF2 recruits the origin recognition complex and stimulates pre-replication complex formation at telomeres through the TRFH domain. PMID: 27836746
  11. The study investigated the disruption pattern of the 3D telomere-TRF2 interaction during the progression from mononuclear Hodgkin cells (H) to multinucleated Reed-Sternberg cells (RS). PMID: 28436953
  12. TRF2, a component of shelterin, binds to core histones to protect chromosome ends from inappropriate DNA damage response and loss of telomeric DNA. The N-terminal Gly/Arg-rich domain (GAR domain) of TRF2 directly interacts with the globular domain of core histones. PMID: 27514743
  13. TRF2 deficiency led to a 1.5-2 fold increase in the radiosensitivity of hMSC-telo1 cells due to telomere destabilization. PMID: 26598048
  14. DREEM imaging revealed that unlike chromatin with DNA wrapping around histones, large TRF2-DNA complexes (larger than TRF2 tetramers) compact DNA inside TRF2, with portions of folded DNA appearing at the edge of these complexes. PMID: 26856421
  15. Studies observed that Sp1 expression is downregulated in TRF2(DeltaBDeltaM)-induced senescence, mediated by ATM and p38 MAPK. PMID: 26906205
  16. Data indicate that isoform beta2 of heregulin (HRGbeta2) localizes at telomeres alongside the telomere-associated proteins TRF2 and RAP1. PMID: 26318724
  17. These findings suggest that TRF2 is a promising candidate for the attachment of telomeres to the nuclear envelope in somatic cells. PMID: 26572976
  18. Through a combination of biochemical, biophysical, and structural approaches, researchers unveiled a unique mode of assembly between RAP1 and TRF2. PMID: 26748096
  19. A dimer of the shelterin subunit TRF2 wraps approximately 90 bp of DNA through several lysine and arginine residues localized around its homodimerization domain. PMID: 26774283
  20. TRF2 overexpression results in replication stalling in duplex telomeric repeat tracts and the formation of telomeric ultrafine anaphase bridges, ultimately leading to stochastic loss of telomeric sequences. PMID: 26640040
  21. Anaphase bridge formation induced clustering of telomere repeats, telomere compaction, and concomitant depletion of the shelterin protein TRF2 (also known as TERF2). PMID: 25908860
  22. The study demonstrates that TRF2 interacts with and is sumoylated by PIAS1 and RNF4. RNF4 targets SUMO-conjugated TRF2 for ubiquitination, decreasing its stability, suggesting that TRF2 status provides a molecular switch regulating its levels at telomeres. PMID: 26450775
  23. TRF2 is a specific target of miR-23a, uncovering a previously unknown role for miR-23a in telomere regulation and cellular senescence. PMID: 25753893
  24. The variability of genes encoding TERF1 and TNKS2 is crucial for maintaining the integrity of the telomere structure and shows a significant association with longevity. PMID: 25631672
  25. TRF2 represses TERRA transcription through its homodimerization domain. PMID: 25359189
  26. TRF2 interacts with lamin A/C, impacting chromosome structure and cellular and organismal aging. PMID: 25399868
  27. Data suggest that TRF2 is a radioresistant protein in A549 and U2OS cells. PMID: 25609072
  28. The TRF2-mediated counterbalance between hREST4 and REST is essential for both the generation and maintenance of neural progenitor cells. PMID: 24740933
  29. TRF2 binds and transactivates the promoter of the angiogenic tyrosine kinase platelet-derived growth factor receptor beta (PDGFRbeta). PMID: 25437559
  30. Data show that full-length repressor activator protein 1 (Rap1) binds to full-length telomeric repeat binding factor 2 (TRF2) with high affinity and an equimolar ratio. PMID: 25675958
  31. TRF2 is involved in the recruitment of RTEL1 to facilitate t-loop disassembly at telomeres in S phase. PMID: 25620558
  32. These results suggest that altered nuclear staining of methylated TRF2 may reflect nuclear structural alterations associated with cellular senescence. PMID: 24721747
  33. KIP binding to TRF2 enhances its telomere-binding activity, suggesting that KIP acts as a positive regulator of TRF2 function. PMID: 25012820
  34. Findings lead researchers to propose a model in which telomeres are directly protected from c-NHEJ via TRF2 impeding Ku's ability to synapse telomere ends. PMID: 24095731
  35. Aging in hESC-derived CMs correlated with reduced membrane potential in mitochondria, accumulation of lipofuscin, a slower beating pattern, and downregulation of human telomerase RNA (hTR) and cell cycle regulating genes. PMID: 22843416
  36. The Wnt/beta-catenin signaling pathway maintains a level of TRF2 critical for telomere protection. PMID: 23429341
  37. High levels of TRF2 in tumor cells decrease their ability to recruit and activate natural killer cells, contributing to oncogenesis. PMID: 23792691
  38. TRF2 is recruited to sites of DNA damage and plays critical roles in the DNA damage response. PMID: 22841872
  39. Doxorubicin induces uPAR-mediated proteasomal activity and ubiquitination of TRF2 in VSMC from the umbilical artery. uPAR also controls nuclear accumulation and DNA binding of TRF2. PMID: 23172421
  40. Low TRF2 expression is associated with chronic lymphocytic leukemia. PMID: 22878603
  41. Results indicate that REST-dependent TRF2 expression renders cancer cells resistant to DNA damage during oxidative stress. PMID: 22821339
  42. As hTRF2 recruits hRap1 to telomeric sequences, hRap1 alters the affinity of hTRF2 and its binding preference on telomeric DNA. Moreover, the TRF2-Rap1 complex has a higher ability to remodel telomeric DNA. PMID: 23086976
  43. Gemcitabine causes telomere shortening by stabilizing TRF2, which is required for XPF-dependent telomere loss. Overexpression of TRF2 in the absence of gemcitabine also causes telomere shortening with simultaneous association of TRF2 with XPF/ERCC1. PMID: 22704123
  44. TRF2 stimulates the binding and enzymatic activities of the LP-BER proteins APE1, FEN1, and LigI, both individually and when they act together in reconstituted LP-BER using a telomeric substrate. PMID: 22336916
  45. The telomeric protein TRF2 binds Telomeric repeat-containing RNA (TERRA) via interactions requiring the formation of a G-quadruplex structure rather than the TERRA sequence itself. PMID: 22780456
  46. TRF2 negatively regulates the number of nucleosomes at human telomeres through a cell cycle-dependent mechanism that alters internucleosomal distance. PMID: 22536324
  47. Increased TRF2 immunostaining intensity in invasive carcinomas is associated with shorter telomeres, and shorter telomeres correlate with a higher TNM stage. PMID: 20625812
  48. Studies investigated the binding sites of telomeric proteins along human chromosomes; RAP1 and TRF2 were found on a small number of interstitial sites, including regions proximal to genes. PMID: 21423278
  49. Silencing TRF2 can inhibit proliferation of Hep-2 cells. PMID: 21395184

Show More

Hide All

Database Links

HGNC: 11729

OMIM: 602027

KEGG: hsa:7014

STRING: 9606.ENSP00000254942

UniGene: Hs.63335

Subcellular Location
Nucleus. Chromosome, telomere. Note=Colocalizes with telomeric DNA in interphase cells and is located at chromosome ends during metaphase.
Tissue Specificity
Ubiquitous. Highly expressed in spleen, thymus, prostate, uterus, testis, small intestine, colon and peripheral blood leukocytes.

Q&A

What is TERF2 and why is it significant in cellular biology research?

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

What applications can TERF2 antibodies be used for in laboratory research?

TERF2 antibodies have been validated for multiple experimental applications:

ApplicationTechnique DetailsCommon Dilutions
Western Blot (WB)Detection of TERF2 protein expression in cell/tissue lysates1:2000-1:50000
Immunohistochemistry (IHC)Visualization of TERF2 in FFPE tissues (human, mouse, rat)1:50-1:2000
Immunoprecipitation (IP)Isolation of TERF2 and interacting proteins0.5-4.0 μg for 1.0-3.0 mg total protein
Flow CytometryIntracellular detection of TERF2As recommended by manufacturer
Chromatin Immunoprecipitation (ChIP)Analysis of TERF2-DNA interactionsValidated in published studies
Immunofluorescence (IF)Subcellular localization of TERF2As recommended by manufacturer

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)

  • Autophagy regulation

What are the molecular characteristics and reactivity profiles of commonly available TERF2 antibodies?

Commercial TERF2 antibodies exhibit the following characteristics:

PropertyDetails
Host SpeciesTypically mouse monoclonal or rabbit polyclonal
Molecular WeightCalculated: 56 kDa; Observed: 60-69 kDa (variation due to post-translational modifications)
Immunogen RegionsVarious epitopes including AA 81-287, AA 78-238, AA 144-438, Full length
Species ReactivityPrimarily human, mouse, and rat; some also react with deer
Purification MethodCommonly antigen affinity or Protein A purification
Storage FormTypically liquid in PBS with sodium azide and glycerol (pH 7.3)

When selecting an antibody for specific applications, researchers should consider reactivity demonstrated in:

  • Cell lines: HeLa, MCF-7, NIH/3T3, Jurkat, K-562, HEK-293, Daudi, HSC-T6, 4T1

  • Tissues: Human gliomas, rat brain, mouse brain

How should TERF2 antibodies be stored and handled for optimal performance?

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:

    • Preferred method: TE buffer pH 9.0

    • Alternative method: Citrate buffer pH 6.0

Working Dilution Preparation:

  • Prepare fresh working dilutions on the day of the experiment

  • Discard any unused diluted antibody

What are the optimized protocols for TERF2 antibody applications in Western Blot and Immunohistochemistry?

Western Blot Protocol for TERF2 Detection:

  • Sample Preparation:

    • Lyse cells in appropriate buffer containing protease inhibitors

    • Validated cell lines: HeLa, MCF-7, NIH/3T3, Jurkat, K-562

  • Electrophoresis and Transfer:

    • Load 20-40 μg protein per lane

    • Expected molecular weight: 65-69 kDa (observed range)

  • Blocking and Antibody Incubation:

    • Block membrane in 5% non-fat milk or BSA for 1 hour at room temperature

    • Incubate with primary TERF2 antibody at recommended dilution (1:2000-1:12000)

    • Use secondary antibody appropriate for host species

  • Detection Considerations:

    • TERF2 may show multiple bands due to isoforms or processing (major bands at 65-69 kDa; minor bands at 32-35 kDa have been reported)

Immunohistochemistry Protocol for TERF2:

  • Tissue Preparation:

    • FFPE sections (4-5 μm thickness)

    • Validated tissues: human gliomas, rat brain, mouse brain

  • Antigen Retrieval (Critical Step):

    • Primary method: TE buffer pH 9.0

    • Alternative: Citrate buffer pH 6.0

    • Heat-induced retrieval (pressure cooker or microwave)

  • Staining Procedure:

    • Block endogenous peroxidase with peroxidase blocking solution

    • Block non-specific binding with protein blocking buffer (20 min)

    • Incubate with primary TERF2 antibody (1:50-1:500 dilution) for 1 hour at room temperature

    • Use appropriate detection system (e.g., Dako EnVision™ FLEX/HRP)

    • Develop with DAB and counterstain with Mayer's Hematoxylin

  • Analysis Guidelines:

    • Score staining by combining intensity and extent scores (0-12 scale)

    • Consider scores 0-4 as negative and 5-12 as positive

    • Have two independent pathologists assess the staining

How can researchers distinguish between different forms or post-translational modifications of TERF2?

Distinguishing between TERF2 variants requires specific methodological approaches:

  • Full-Length vs. Truncated Forms:

    • Use antibodies targeting different epitopes (N-terminal vs. C-terminal)

    • Compare with wild-type (pTERF2) and mutant (pTERF2ΔC) controls

    • Expected molecular weights:

      • Full-length TERF2: 65-69 kDa

      • Truncated forms: Various, including 32-35 kDa bands

  • 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:

    • HTH myb-type DNA-binding domain can be specifically targeted

    • For functional studies, compare wild-type TERF2 with domain-deletion mutants (e.g., TERF2ΔC)

  • Research Application Example:
    In autophagy studies, researchers distinguished TERF2's functional forms by:

    • Transfecting cells with wild-type (pTERF2) and mutant (pTERF2ΔC) forms

    • Using JetPEI reagent for 48h transfection

    • Employing pBabe-puro empty vector as control

What positive and negative controls should be included when working with TERF2 antibodies?

Positive Controls:

  • Cell Lines with Known TERF2 Expression:

    • Human: HeLa, MCF-7, Jurkat, K-562

    • Mouse: NIH/3T3, 4T1

    • Rat: HSC-T6

  • Tissue Sections:

    • Human gliomas

    • Rat brain tissue

    • Mouse brain tissue

  • Genetic Controls:

    • Overexpression systems: pBabe-puro-MYC-TERF2 transfected cells

    • For functional studies: Include both wild-type TERF2 and mutant TERF2ΔC

Negative Controls:

  • Antibody Controls:

    • Isotype-matched irrelevant antibody (same concentration)

    • Primary antibody omission

  • Expression Controls:

    • TERF2 knockdown/silenced cells:

      • siRNA approach: Use siTERF2 (Dharmacon, 5'-GCAGAAGUGGACUGUAGAAUU-3')

      • shRNA approach: shTERF2 stable cell lines

    • TERF2 knockout cells (if available)

  • 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

How do cancer-related mutations or expression changes impact TERF2 antibody detection?

TERF2 expression and mutations vary across cancer types, affecting antibody-based detection:

  • Altered Expression Patterns:

    • TERF2 is upregulated in multiple cancers including:

      • Cholangiocarcinoma (CHOL)

      • Diffuse large B-cell lymphoma (DLBC)

      • Pancreatic adenocarcinoma (PAAD)

      • Stomach adenocarcinoma (STAD)

      • Thymoma (THYM)

    • Expression correlates with tumor progression in specific cancers

  • 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:

    • Amplification and mutations in TERF2 particularly affect liver hepatocellular carcinoma (LIHC)

    • Mutations may alter antibody binding if they occur within the epitope region

    • Use multiple antibodies targeting different regions to verify results

  • Technical Considerations for Cancer Samples:

    • For IHC scoring in cancer tissues:

      • Use immunoreactive score (IRS) combining staining intensity and percentage of positive cells

      • Apply consistent criteria: negative (scores 0-4) and positive (scores 5-12)

    • For WB analysis:

      • Include stage-matched samples when comparing expression

      • Account for tumor heterogeneity through multiple sampling

How can TERF2 antibodies be optimally employed to study telomere maintenance and dysfunction?

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:

    • Lyse cells in non-denaturing buffer to preserve protein interactions

    • Immunoprecipitate with TERF2 antibody

    • Western blot for interacting partners (TRF1, POT1, TIN2, TPP1, RAP1)

    • Validate interactions with reverse IP

    • Recommended IP conditions: 0.5-4.0 μg antibody for 1.0-3.0 mg total protein

  • Immunofluorescence for Telomere Dysfunction-Induced Foci (TIF):

    • Co-stain for TERF2 and DNA damage markers (γ-H2AX, 53BP1)

    • Protocol example:

      • Fix cells in 4% formaldehyde (10 min, RT)

      • Permeabilize with 0.25% Triton X-100 (5 min, RT)

      • Incubate with rabbit anti-TERF2 and mouse anti-γ-H2AX antibodies

      • Use appropriate secondary antibodies (Alexa Fluor 488/555 conjugates)

      • Counterstain nuclei with DAPI

    • Quantify co-localization as indicator of telomere dysfunction

  • Telomere Dysfunction Analysis in Cancer Models:

    • Implement TERF2 knockdown approaches:

      • Transient: siTERF2 (100 nM, 24h with INTERFERin reagent)

      • Stable: shTERF2 cell lines or conditional knockout models

    • Assess telomere protection by measuring:

      • End-to-end chromosome fusions

      • Telomere length (Q-FISH, TRF analysis)

      • ATM pathway activation

What methodological considerations are critical when investigating TERF2's role in cancer?

TERF2's complex role in tumorigenesis requires sophisticated research approaches:

  • Expression Analysis Across Cancer Types:

    • Pan-cancer analysis reveals variable expression patterns:

      • Upregulated in: CHOL, DLBC, PAAD, STAD, THYM

      • Expression correlates with survival in specific cancers:

        • Poor OS in ACC, LUSC, and UVM

        • Better OS in CHOL and KIRC

    • Methodology:

      • Compare matched tumor/normal samples

      • Stratify by cancer stage and subtype

      • Correlate with clinical outcomes

  • TERF2 Knockdown/Overexpression in Cancer Models:

    • For knockdown studies:

      • siRNA approach: siTERF2 (Dharmacon, 5'-GCAGAAGUGGACUGUAGAAUU-3')

      • shRNA approach for stable knockdown

    • For overexpression studies:

      • Transfect with pBabe-puro-MYC-TERF2 (wild-type) or pBabe-puro-MYC-TERF2ΔC (mutant)

      • Assess proliferation, migration, invasion, and chemosensitivity

  • In Vivo Cancer Model Analysis:

    • Orthotopic injection model:

      • NSG mice injected with control (shSCR) and TERF2-silenced (shTERF2) cancer cells

      • Tumor volume monitoring

      • IHC analysis of extracted tumors with anti-TERF2 antibodies

    • Markers to assess in tumor samples:

      • Proliferation markers

      • DNA damage indicators (8-OHdG)

      • Autophagy markers (LC3B)

  • TERF2 and Tumor Immune Infiltration Analysis:

    • IHC multiplex staining for TERF2 and immune cell markers

    • Correlation analysis between TERF2 expression and:

      • Cancer-associated fibroblasts (positive correlation in BLCA, CESC, HNSC, PAAD, SKCM, STAD)

      • Lymphocyte infiltration (negative correlation in GBM, LGG, UCS)

    • Chemokine expression analysis:

      • TERF2 negatively correlates with CCL2, CCL4, CCL14, CCL15, CCL28, CXCL2, CXCL3, CXCL9, CXCL16

How can researchers effectively investigate TERF2's non-canonical functions using antibody-based approaches?

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:

      • Co-immunofluorescence using rabbit anti-TERF2 and mouse anti-HMGB1 antibodies

      • Secondary antibodies: Alexa Fluor 488 (anti-rabbit) and Alexa Fluor 555 (anti-mouse)

      • Nuclear counterstain with DAPI

  • TERF2's Role in Autophagy Assessment:

    • TERF2 silencing approach:

      • Transfect with siTERF2 (100 nM, 24h)

      • Autophagy stimulation: EBSS nutrient starvation

    • Autophagy markers analysis:

      • Transfect with autophagy reporters (EGFP-LC3B, mCherry-LC3B, mRFP-EGFP-LC3B)

      • Assess HMGB1 cytosolic translocation with:

        • EGFP-HMGB1 or EGFP-HMGB1 C106S constructs

        • Subcellular fractionation and western blotting

  • 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

What are the technical challenges and solutions in studying TERF2-protein interactions?

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:

    • Challenge: Identifying interaction domains requires domain-specific tools

    • Solution:

      • Use domain-specific antibodies where available

      • Express tagged domain constructs

      • Compare interaction profiles between wild-type and mutant TERF2:

        • Wild-type: pBabe-puro-MYC-TERF2

        • Mutant: pBabe-puro-MYC-TERF2ΔC

  • 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

How can researchers methodically investigate TERF2's role in regulating immune cell function and infiltration?

The emerging role of TERF2 in immune regulation requires specialized approaches:

  • Correlation Analysis of TERF2 and Immune Cell Markers:

    • Methodology:

      • Use TIMER, XCELL, MCPCOUNTER, and EPIC algorithms

      • Correlate TERF2 expression with infiltrating immune cell populations

      • Cancer types showing significant correlations:

        • Cancer-associated fibroblasts: BLCA, CESC, HNSC, PAAD, SKCM, STAD

        • Lymphocytes: GBM, LGG, UCS (negative correlation)

  • Multiparametric Analysis of Immune Markers:

    • Assess correlation between TERF2 and immune markers in tumor samples (Table from search result 5):

    Immune Cell TypeMarkerCorrelation with TERF2 in ESCA TumorCorrelation in Normal Tissue
    T cells (CD8+)CD8A, CD8BNo significant correlationPositive in normal tissue (GETX)
    T cells (general)CD2, CD3ENo significant correlationPositive in normal tissue
    Natural Killer cellsKIR2DL4, KIR3DL2No significant correlationPositive in normal tissue
    Th1 cellsT-betNo significant correlationPositive
    Th2 cellsSTAT5A, STAT6No significant correlationStrongly positive (p<0.01)
    Tfh cellsBCL6Positive (p<0.01)Strongly positive (p<0.01)
    Th17 cellsSTAT3Strongly positive (p<1e-6)Strongly positive (p<1e-4)
    T cell exhaustionPD-1, LAG3, TIM-3No significant correlationPositive for TIM-3 (p<0.05)
  • Chemokine Correlation Analysis:

    • Methodology:

      • Use TISIDB database

      • Analyze association between TERF2 and chemokine expression

      • Significant negative correlations with:

        • CCL2, CCL4, CCL14, CCL15, CCL28

        • CXCL2, CXCL3, CXCL9, CXCL16

  • 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

What are common issues in TERF2 antibody applications and their solutions?

IssuePossible CausesSolutions
No signal in Western BlotInsufficient protein expressionUse validated positive control cells (HeLa, MCF-7, K-562)
Ineffective protein extractionAdd DNase to release DNA-bound TERF2
Degraded proteinUse fresh samples with protease inhibitors
Multiple unexpected bandsProtein degradationAdd protease inhibitors, reduce sample processing time
Splice variantsCompare with literature reports of known variants
Non-specific bindingOptimize antibody dilution (1:2000-1:12000 for WB)
High background in IHCInsufficient blockingExtend blocking time, optimize blocker concentration
OverfixationOptimize fixation time, ensure proper antigen retrieval
Excessive antibodyFurther dilute antibody (test range 1:50-1:500)
Poor cellular localizationInadequate fixationOptimize fixation protocol (4% formaldehyde, 10 min, RT)
Inefficient permeabilizationAdjust Triton X-100 concentration (0.25%, 5 min, RT)
Epitope maskingTry different fixation methods or antibody clones

How can researchers address discrepancies in TERF2 detection between different experimental systems?

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:

    • When WB and IHC results differ:

      • Consider protein conformation differences (native vs. denatured)

      • Optimize antigen retrieval for IHC (TE buffer pH 9.0 or citrate buffer pH 6.0)

      • Validate with alternative methods (IF, flow cytometry)

  • 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

What emerging technologies are enhancing TERF2 detection and functional analysis?

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

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.