TCAF2 antibodies are polyclonal or monoclonal reagents designed for immunodetection methods. Key features include:
Biomatik CAC11544: Rabbit polyclonal antibody validated for ELISA, IHC, and IF. Targets residues 472–590 of human TCAF2 .
Sigma-Aldrich HPA038758: Rabbit antibody with IHC validation (1:1000–1:2500 dilution) and affinity-purified for specificity .
CUSABIO CSB-PA008015LA01HU: Biotin-conjugated antibody optimized for ELISA .
TCAF2 antibodies have been instrumental in elucidating the protein’s role in oncogenesis:
TCAF2 expression correlates with glioma aggressiveness and poor prognosis. Key findings include:
Mechanism: TCAF2 promotes epithelial-to-mesenchymal transition (EMT) via STAT3 activation, enhancing cell migration and invasion .
Clinical Relevance: Elevated TCAF2 in glioblastoma (GBM) tumor centers is linked to higher WHO grades and IDH mutation status .
In colorectal cancer (CRC), TCAF2 in tumor pericytes (TPCs) drives liver metastasis:
Mechanism: TCAF2 inhibits TRPM8 ion channel activity, upregulating Wnt5a secretion and activating STAT3 in tumor cells, which induces EMT .
Therapeutic Target: Menthol (a TRPM8 agonist) suppresses Wnt5a secretion, reducing metastasis in preclinical models .
TCAF2 antibodies enable precise quantification and localization in studies:
Glioma Samples: TCAF2 overexpression in GBM vs. low-grade glioma is confirmed via IHC, with tumor center dominance .
TPC Isolation: Microdissection combined with pericyte medium-based approaches isolates TPCs for TCAF2 detection in CRC .
Gain-/Loss-of-Function: TCAF2 overexpression in TPCs enhances CRC cell migration, while knockdown reduces metastasis in mice .
Protein Interactions: TCAF2 binds TRPM8, modulating its trafficking and ion channel activity .
Biomarker: High TCAF2+ TPC ratios (>30%) predict poor survival in CRC patients .
Therapeutic Strategies: Targeting TCAF2-TRPM8 axis or STAT3/Wnt5a pathways may inhibit metastasis .
For optimal TCAF2 detection in glioma tissue sections, paraformaldehyde fixation (4% PFA) for 24 hours followed by paraffin embedding provides consistent results. This approach preserves both tissue morphology and TCAF2 epitopes effectively. For frozen sections, brief fixation (10 minutes) with 4% PFA before antibody incubation optimizes signal-to-noise ratio. The choice between these methods should consider downstream applications and specific research questions. Notably, some epitopes may be sensitive to over-fixation, necessitating calibration with positive controls to establish ideal fixation duration for your specific TCAF2 antibody clone .
Comprehensive validation requires multiple complementary approaches. First, perform western blot analysis against recombinant TCAF2 protein alongside cell lysates from cells with known TCAF2 expression levels to confirm band specificity at the expected molecular weight (~100.9 kDa). Second, include positive and negative control tissues or cell lines with documented TCAF2 expression patterns. Third, conduct peptide competition assays by pre-incubating the antibody with purified TCAF2 antigen, which should eliminate specific staining. Finally, consider TCAF2 knockdown or knockout validation when possible, comparing antibody reactivity between wild-type and TCAF2-depleted samples .
Commercial TCAF2 antibodies differ primarily in their target epitopes, clonality, host species, and validated applications. Polyclonal antibodies typically recognize multiple TCAF2 epitopes, potentially offering higher sensitivity but variable batch-to-batch consistency. Monoclonal antibodies provide consistent reproducibility but might have more limited epitope recognition. When selecting an antibody, consider whether the target epitope falls within any of the three reported TCAF2 isoforms to ensure detection of your isoform of interest. Additionally, antibodies differ in their validated applications - some are optimized for Western blot analysis while others perform better in immunohistochemistry or immunofluorescence contexts .
For TCAF2 immunodetection, a blocking solution containing 5% normal serum (matched to the host species of the secondary antibody) with 0.3% Triton X-100 in PBS typically provides excellent background reduction. For tissues with high endogenous biotin, consider adding avidin/biotin blocking steps when using biotinylated detection systems. In cases of persistent background, incorporating 0.1% bovine serum albumin and 0.05% Tween-20 into the blocking solution can further minimize non-specific binding. The optimal blocking protocol should be empirically determined for each tissue type and fixation method, as TCAF2's membrane localization can sometimes require specialized blocking approaches .
To investigate TCAF2's role in immune cell infiltration within glioma, implement a multi-parameter approach combining immunohistochemistry, flow cytometry, and functional assays. Begin with multiplex immunofluorescence to simultaneously visualize TCAF2 expression alongside markers for tumor-associated macrophages (CD68, CD163), T-cells (CD3, CD8), and other immune components. Follow with flow cytometric analysis of dissociated tumor samples to quantify correlation between TCAF2 expression levels and immune cell populations. For mechanistic insights, conduct co-culture experiments with TCAF2-overexpressing or TCAF2-knockdown glioma cells alongside immune cells, measuring migration, activation, and cytokine production. This comprehensive approach will reveal whether TCAF2 directly influences immune cell recruitment or activation, providing deeper understanding of its immunomodulatory functions in the tumor microenvironment .
When evaluating potential cross-reactivity between TCAF2 antibodies and other TCAF family members (particularly TCAF1), implement a systematic control strategy. First, perform western blot analysis using recombinant proteins for all TCAF family members in parallel lanes. Second, conduct immunoprecipitation with the TCAF2 antibody followed by mass spectrometry to identify all captured proteins. Third, utilize cell lines with CRISPR-mediated knockout of individual TCAF family members as negative controls. Fourth, perform epitope mapping to identify the specific sequence recognized by your antibody and conduct in silico analysis to predict potential cross-reactivity with homologous regions in related proteins. Finally, include competitive blocking experiments with peptides derived from different TCAF family members to demonstrate specificity. This comprehensive approach ensures confident interpretation of results, particularly in tissues where multiple TCAF proteins are co-expressed .
Optimizing dual immunofluorescence for TCAF2 and TRPM8 requires careful consideration of antibody compatibility and signal separation. First, select primary antibodies raised in different host species (e.g., rabbit anti-TCAF2 and mouse anti-TRPM8) to enable species-specific secondary antibody detection. If only same-species primaries are available, consider sequential immunostaining with direct conjugated antibodies or implement tyramide signal amplification. Second, optimize fixation conditions that preserve both proteins' epitopes - typically, mild fixation (4% PFA for 10-15 minutes) works well for membrane proteins. Third, employ spectral unmixing during imaging to eliminate potential bleed-through between fluorophores. Finally, include appropriate controls: single-stained samples to establish signal specificity and samples treated with blocking peptides to confirm antibody specificity. This approach enables reliable co-localization analysis to investigate functional interactions between TCAF2 and TRPM8 channels .
Quantifying TCAF2 expression in patient-derived glioma samples requires robust standardization to account for sample heterogeneity. Implement a multi-modal approach beginning with IHC scoring systems (e.g., H-score or Allred) that account for both staining intensity and percentage of positive cells. Supplement this with digital image analysis using specialized software that can segment tissue compartments and provide objective intensity measurements. For protein quantification, consider targeted mass spectrometry with isotope-labeled TCAF2 peptide standards for absolute quantification. RNA-level quantification through qRT-PCR or RNA-seq should employ multiple reference genes validated for stability in glioma tissues. Critical factors influencing accurate quantification include: 1) consistent sample handling from collection through processing, 2) standardized staining protocols with batch controls, 3) inclusion of calibration standards, and 4) blinded analysis by multiple observers to minimize bias. This comprehensive approach enables reliable comparison of TCAF2 expression across patient cohorts for correlation with clinical parameters .
When encountering weak or inconsistent TCAF2 signal in Western blots, implement a systematic optimization strategy. First, evaluate protein extraction methods - TCAF2's membrane localization may require specialized extraction buffers containing ionic detergents (e.g., 1% SDS) or non-ionic detergents (e.g., 1% Triton X-100) supplemented with protease inhibitors. Second, optimize protein loading (50-100 μg total protein) and transfer conditions (consider extended transfer times of 2+ hours for this high molecular weight protein). Third, test different blocking solutions - 5% non-fat milk can mask some TCAF2 epitopes, so try 3-5% BSA alternatives. Fourth, increase primary antibody concentration or incubation time (overnight at 4°C often improves signal). Fifth, implement signal enhancement through high-sensitivity chemiluminescent substrates or consider HRP-polymer detection systems. If these approaches fail, test alternative TCAF2 antibodies targeting different epitopes, as protein modifications or isoform expression may affect epitope accessibility .
Non-specific binding in TCAF2 immunohistochemistry can be systematically addressed through multiple optimization strategies. First, implement more stringent blocking by increasing blocking solution concentration (5-10% normal serum) and extending blocking time (1-2 hours). Second, reduce primary antibody concentration and incorporate a 0.1-0.3% Triton X-100 wash step after primary antibody incubation to remove weakly bound antibodies. Third, add 0.1-0.5M NaCl to antibody diluent to increase stringency of binding. Fourth, pre-absorb the primary antibody with tissue powder from non-target tissues to remove antibodies with affinity for common epitopes. Fifth, if endogenous biotin causes background, implement avidin-biotin blocking or switch to polymer-based detection systems. For tissues with high endogenous peroxidase activity, extend hydrogen peroxide quenching (3% H₂O₂ for 15-30 minutes). Finally, include peptide competition controls to distinguish between specific and non-specific binding patterns. This systematic approach should significantly improve signal-to-noise ratio for TCAF2 detection .
Discrepancies between TCAF2 mRNA levels and protein detection occur frequently and require careful investigation. First, verify TCAF2 antibody specificity through knockout validation or peptide competition assays to confirm detection accuracy. Second, consider post-transcriptional regulation - TCAF2 may be subject to microRNA regulation or RNA binding protein control affecting translation efficiency. Third, evaluate protein stability and turnover using cycloheximide chase experiments to determine if rapid protein degradation explains low protein levels despite high mRNA expression. Fourth, examine potential alternative splicing producing isoforms not recognized by your antibody by using primers targeting different exons or antibodies targeting different epitopes. Fifth, consider technical factors like detection sensitivity limits - implement more sensitive detection methods such as proximity ligation assay or tyramide signal amplification. Finally, examine spatial-temporal dynamics, as mRNA and protein may peak at different timepoints or localize to different cellular compartments. This systematic troubleshooting approach helps reconcile seemingly contradictory TCAF2 expression data .
To comprehensively evaluate TCAF2's impact on glioma cell migration and invasion, implement a multi-assay approach. Begin with 2D wound healing assays comparing migration rates between TCAF2-overexpressing, wild-type, and TCAF2-knockdown glioma cells, quantifying wound closure kinetics over 24-48 hours. Follow with transwell migration and Matrigel invasion assays to assess directed cell movement and extracellular matrix penetration capabilities. For more physiologically relevant assessment, employ 3D spheroid invasion assays using fluorescently-labeled cells embedded in brain-mimetic matrices (hyaluronic acid/collagen composites), with time-lapse imaging to track invasion patterns and distances. Complement these with ex vivo brain slice models, where fluorescently-labeled glioma cells with varied TCAF2 expression are seeded onto mouse brain slices to observe invasion in neural tissue. Finally, validate findings using in vivo orthotopic xenograft models with inducible TCAF2 expression systems, followed by histological analysis of tumor margins and invasive fronts. This comprehensive approach provides robust evidence of TCAF2's specific contributions to glioma invasiveness .
To correlate TCAF2 expression with immune infiltrates in glioma tissue microarrays (TMAs), implement a multi-parameter digital pathology workflow. First, perform multiplex immunohistochemistry on sequential TMA sections using TCAF2 antibody alongside panels of immune markers (CD3, CD8, CD4, FOXP3, CD68, CD163) with spectral unmixing to distinguish overlapping signals. Second, employ whole-slide digital scanning with automated tissue recognition algorithms to identify tumor regions, necrotic areas, and perivascular zones. Third, implement machine learning-based cell classification to quantify both the density and spatial distribution of each immune cell population. Fourth, create spatial relationship maps analyzing distances between TCAF2-high tumor cells and specific immune populations. Fifth, perform correlation analyses between TCAF2 expression levels (using H-score or automated intensity quantification) and immune cell densities across different patient subgroups stratified by molecular subtypes, IDH status, and grade. This approach enables robust statistical correlation while preserving spatial context, providing insights into whether TCAF2 expression creates immunosuppressive or immunostimulatory microenvironments in glioma .
Investigating the relationship between TCAF2 expression and treatment response requires rigorous analytical approaches integrating multiple data types. First, establish standardized TCAF2 quantification protocols combining IHC scoring systems (H-score) with digital image analysis to minimize observer variability. Second, implement multivariate Cox regression models to assess TCAF2's predictive value while controlling for established prognostic factors (age, IDH status, MGMT methylation). Third, perform Kaplan-Meier survival analyses stratifying patients by TCAF2 expression levels and treatment modalities to identify potential interaction effects. Fourth, utilize longitudinal sampling (pre-treatment, mid-treatment, and recurrence) analyzed through linear mixed models to evaluate TCAF2 dynamics during treatment. Fifth, conduct in vitro drug sensitivity testing on patient-derived glioma cells with varied TCAF2 expression to establish direct causal relationships. Finally, develop machine learning algorithms integrating TCAF2 expression with other molecular markers to create predictive models for treatment response. This comprehensive analytical framework enables robust determination of TCAF2's value as a predictive biomarker for specific treatment modalities in glioma .
Designing effective CRISPR-Cas9 approaches for TCAF2 functional studies requires careful consideration of several factors. First, conduct thorough bioinformatic analysis to identify optimal gRNA target sites that minimize off-target effects while ensuring complete protein knockout - ideally targeting early exons (exons 2-4) that are present in all TCAF2 isoforms. Second, implement comprehensive validation strategies including Sanger sequencing of the targeted locus, western blot confirmation of protein depletion, and RNA-seq to verify absence of functional transcripts and identify potential compensatory mechanisms. Third, establish appropriate controls including non-targeting gRNAs and rescue experiments with CRISPR-resistant TCAF2 constructs to confirm phenotype specificity. Fourth, consider conditional knockout systems (e.g., doxycycline-inducible Cas9) to study temporal aspects of TCAF2 function. Fifth, design parallel approaches targeting individual TCAF2 domains to dissect their specific contributions to TCAF2 function. Particularly critical is considering TCAF2's interaction with TRPM8 channels - include experiments measuring TRPM8 activity to determine whether phenotypes result from direct TCAF2 functions or altered ion channel regulation .
Distinguishing between TCAF2 isoform functions requires a strategic approach combining molecular and functional analyses. First, design isoform-specific detection methods including qRT-PCR primers spanning unique exon junctions and antibodies targeting isoform-specific epitopes, validated using recombinant protein standards. Second, quantify relative isoform expression across normal tissues and glioma subtypes using targeted RNA-seq and isoform-specific western blotting to establish expression patterns. Third, create expression constructs for individual isoforms with consistent promoters and identical tags for comparable expression levels and detection sensitivity. Fourth, perform isoform-specific knockdown using siRNAs targeting unique regions complemented by rescue experiments with siRNA-resistant constructs. Fifth, conduct comparative functional assays (proliferation, migration, invasion, immunomodulation) with cells expressing single isoforms. Finally, implement proximity labeling techniques (BioID, APEX) with isoform-specific baits to identify differential protein interaction networks. This systematic approach enables attribution of specific cellular functions to individual TCAF2 isoforms, critical for understanding their potentially divergent roles in glioma biology .
To robustly demonstrate TCAF2-immune interactions in glioma, implement a multifaceted experimental design combining in vitro, ex vivo, and in vivo approaches. Begin with in vitro co-culture systems using TCAF2-modulated glioma cells (overexpression/knockdown) with various immune populations (macrophages, T-cells, microglia), assessing changes in immune cell phenotypes, cytokine production, and functional polarization. Next, employ organotypic brain slice cultures with fluorescently-labeled immune cells to observe migration and interaction behaviors in a more complex microenvironment. For in vivo studies, utilize orthotopic syngeneic mouse models with conditional TCAF2 expression systems, allowing temporal control of TCAF2 levels after tumor establishment. Analyze tumors using cytometry by time-of-flight (CyTOF) or single-cell RNA sequencing to comprehensively profile immune populations. Complement this with spatial transcriptomics or multiplex immunofluorescence to preserve information about cellular neighborhoods. Include functional studies with specific immune cell depletion (anti-CD8 antibodies, clodronate liposomes) to determine which immune populations mediate TCAF2's effects. This integrated approach comprehensively characterizes how TCAF2 modulates the immune landscape in glioma, potentially identifying targetable immune mechanisms .
| Application | Recommended Dilution Range | Sample Preparation | Incubation Parameters | Signal Detection Method | Common Troubleshooting |
|---|---|---|---|---|---|
| Western Blot | 1:500-1:2000 | 50-100μg total protein, RIPA or SDS buffer extraction | Primary: Overnight at 4°C; Secondary: 1h at RT | ECL or fluorescent detection | Insufficient transfer of high MW protein; Membrane proteins require complete denaturation |
| Immunohistochemistry (FFPE) | 1:50-1:200 | Heat-mediated antigen retrieval (citrate pH 6.0 or EDTA pH 9.0) | Primary: Overnight at 4°C; Secondary: 1h at RT | DAB or AEC chromogen | Background staining; Epitope masking; Fixation sensitivity |
| Immunofluorescence | 1:100-1:500 | 4% PFA fixation (10-15 min), 0.1% Triton X-100 permeabilization | Primary: Overnight at 4°C; Secondary: 2h at RT | Fluorophore-conjugated secondary antibodies | Autofluorescence; Photobleaching; Signal:noise issues |
| Flow Cytometry | 1:50-1:100 | Gentle fixation, membrane permeabilization for intracellular detection | 30-60 min at 4°C | Direct conjugates preferred | Poor cell permeabilization; Fixation-induced epitope alteration |
| Chromatin Immunoprecipitation | 1:50 | Crosslinked chromatin, sonication to 200-500bp fragments | Overnight at 4°C with rotation | qPCR or NGS of precipitated DNA | High background; Low enrichment; Antibody specificity |
| ELISA | 1:1000-1:5000 (capture); 1:5000-1:20000 (detection) | Recombinant protein standards, sample dilution series | Coating: Overnight at 4°C; Detection: 1-2h at RT | HRP-conjugated detection system | Hook effect at high concentrations; Matrix interference |