TCF3 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the shipping method and destination. Please consult your local distributors for specific delivery times.
Synonyms
AGM8 antibody; bHLHb21 antibody; Class B basic helix-loop-helix protein 21 antibody; E12 antibody; E2A antibody; E2A immunoglobulin enhancer binding factors E12/E47 antibody; E47 antibody; Helix loop helix protein HE47 antibody; Immunoglobulin enhancer-binding factor E12/E47 antibody; Immunoglobulin transcription factor 1 antibody; ITF1 antibody; Kappa-E2-binding factor antibody; MGC129647 antibody; MGC129648 antibody; Negative vitamin D response element binding protein antibody; NOL1-TCF3 fusion antibody; TCF-3 antibody; Tcf3 antibody; TFE2_HUMAN antibody; transcription factor 3 (E2A immunoglobulin enhancer binding factors E12/E47) antibody; Transcription factor 3 antibody; transcription factor 3 variant 3 antibody; Transcription factor E2-alpha antibody; Transcription factor ITF-1 antibody; VDIR antibody; VDR interacting repressor antibody; vitamin D receptor-interacting repressor antibody
Target Names
Uniprot No.

Target Background

Function
TCF3 is a transcriptional regulator that plays a critical role in the initiation of neuronal differentiation and mesenchymal to epithelial transition. TCF3 forms heterodimers with tissue-specific basic helix-loop-helix (bHLH) proteins, which are essential for determining tissue-specific cell fate during embryogenesis, such as muscle or early B-cell differentiation. In collaboration with TCF15, TCF3 is involved in the mesenchymal to epithelial transition. These dimers bind to DNA at E-box motifs: 5'-CANNTG-3'. TCF3 interacts with the kappa-E2 site in the kappa immunoglobulin gene enhancer, and it also binds to IEB1 and IEB2, which are short DNA sequences within the insulin gene transcription control region.
Gene References Into Functions
  1. hnRNP H/F are crucial for the maintenance and differentiation of embryonic stem cells. This is partly attributed to a shift in TCF3 alternative splicing, leading to repression of CDH1/E-cadherin. PMID: 30115631
  2. B-cell acute lymphoblastic leukemia patients with positive E2A-PBX1 fusion expression after transplant demonstrate a poor prognosis. PMID: 29705861
  3. Studies have shown that abnormal expression of endometrial E2A occurs in the mid-secretory endometrium of women with recurrent miscarriage. There is a positive correlation between E2A and FOXP3, and E2A and CTLA-4, suggesting a potential regulatory role of E2A in endometrial receptivity. PMID: 29270752
  4. TCF3 gene silencing inhibits esophageal cancer cell growth and proliferation, suppresses cell cycle progression, and promotes apoptosis. PMID: 28864779
  5. Research suggests that the upregulation of TCF3 is a significant prognostic factor for nasopharyngeal carcinoma. PMID: 28107608
  6. Findings indicate that TCF3 is clearly associated with the progression of cervical squamous cell carcinoma. This is the first report demonstrating that TCF3 can act as a tumor promoter in cervical cancer, potentially holding significance for the prognosis of CSCC. PMID: 28604457
  7. Evidence suggests that inactivation of TCF3 contributes to the oncogenic program of classical Hodgkin lymphoma. PMID: 27166193
  8. This study marks the first identification of protein partners for either E2A-PBX1 or HOXA9 oncoproteins through an unbiased biochemical approach. The discovery of translation initiation factors associated with HOXA9 might indicate a novel function for HOX proteins beyond their transcriptional activity. PMID: 28707666
  9. Poly (ADP-ribose) polymerase inhibitors selectively induce cytotoxicity in TCF3-HLF-positive leukemic cells. PMID: 27894958
  10. MiR-138 may serve as a tumor suppressor and a potential prognostic biomarker in cervical cancer. Its downstream target, TCF3, may also regulate cancer development in a reverse manner as miR-138. PMID: 28385388
  11. Elevated levels of TCF3 in gliomas promote glioma development through the Akt and Erk pathways. PMID: 27105323
  12. TWIST1-E12 protein heterodimers may constitute the primary active forms of TWIST1 concerning senescence inhibition throughout the course of breast tumorigenesis. PMID: 27237323
  13. This review focuses on the role of the E2A-PBX1 gene rearrangement in the prognosis of childhood acute lymphoblastic leukemia and its central nervous system relapse. PMID: 26509298
  14. E47 is a novel substrate of PAK5, and PAK5-mediated phosphorylation of E47 promotes epithelial-mesenchymal transition. High expression of phospho-E47 is associated with an aggressive phenotype of colon cancer and metastasis. PMID: 26212009
  15. Significant enrichment of the neuroactive ligand-receptor interaction pathway was observed in TCF3-PBX1, along with an enrichment of genes involved in immunity and infection pathways in the ETV6-RUNX1 subtype. PMID: 26237075
  16. Overexpression of E47 reprograms human pancreatic cancer cells to a quiescent acinar state with reduced tumorigenic potential. PMID: 25894862
  17. Drug response profiling of matched patient-derived xenografts revealed a distinct profile for TCF3-HLF ALL, characterized by resistance to conventional chemotherapeutics but sensitivity to glucocorticoids. PMID: 26214592
  18. When intensive chemotherapy was employed, TCF3-PBX1 was associated with a favorable outcome in childhood pre-B ALL. PMID: 25551271
  19. Inhibitor of differentiation 4 (ID4) acts as an inhibitor of ID-1, -2 and -3, promoting basic helix loop helix (bHLH) E47 DNA binding and transcriptional activity. PMID: 25778840
  20. Data indicate that microRNAs miR-155 and miR-16 downregulate activation-induced cytidine deaminase (AID) and transcription factor E47 in B Cells through binding of the 3'-untranslated regions. PMID: 26223652
  21. TCF3 is a novel susceptibility locus for Hodgkin lymphoma. PMID: 24920014
  22. Our findings suggest a collaborative action between autophagy and ubiquitination in the degradation of E2A/Pbx1, revealing a novel strategy for targeted preventive or treatment therapy in pediatric ALL. PMID: 25615280
  23. TCF3 rearrangement [t(1;19) (q23;p13)] was detected in 16 (20%) out of the 80 studied patients, and it was significantly associated with splenomegaly, lymphadenopathy, and CNS infiltration. PMID: 25116187
  24. Results demonstrate that the up-regulation of TCF3, primarily driven by promoter hypomethylation, is one of the molecular mechanisms involved in the development and progression of colorectal cancer. PMID: 25375219
  25. These results suggest that E47 exerts diverse effects in T-ALL, but functional deficiency of E47 is not a universal characteristic of Lmo2-induced T-ALL. PMID: 25499232
  26. Our study showed that TCF3-PBX1 positive pediatric BCP-ALL patients treated according to the JACLS ALL02 and CCLSG ALL2004 protocol exhibited favorable outcomes. PMID: 24578304
  27. Findings suggest that E2A suppresses CRC cell metastasis, at least partially if not entirely, by inhibiting YAP expression. PMID: 24369055
  28. E2A is an independent prognostic factor for colorectal cancer patients and targets miR-320a to regulate cell proliferation of colon cancer cells. PMID: 24454819
  29. Within the AETFC complex, AML1-ETO oligomerization is necessary for a specific interaction between the oligomerized NHR2 domain and a novel NHR2-binding (N2B) motif in E proteins, including HEB and E2A. Disruption of this interaction through point mutations abolishes AML1-ETO-induced hematopoietic stem/progenitor cell self-renewal and leukemogenesis. PMID: 23812588
  30. In t(8;21) leukemia cells, the two E proteins, HEB and E2A, function as components of the stable AML1-ETO-containing transcription factor complex (AETFC). The AETFC components collaboratively regulate gene expression and contribute to leukemogenesis. PMID: 23812588
  31. NF-kappaB potentiated the binding of E2A to an E-box motif located immediately downstream of the 2 closely-spaced transcription start sites for sustained 14-3-3gamma expression and CSR induction. PMID: 23851690
  32. Four patients with agammaglobulinemia exhibited the same de novo mutation in the broadly expressed transcription factor E47. The mutant protein (E555K) was stable in patient-derived EBV-transformed cell lines and cell lines transfected with expression vectors. PMID: 24216514
  33. E47 interacts with Id1 in E47 overexpressing MDCK cells that underwent a full epithelial-mesenchymal transition, as well as in mesenchymal breast carcinoma and melanoma cell lines. PMID: 23555842
  34. The E2A-PBX1 fusion gene caused by t(1;19)(q23;p13) may be a common genetic alteration in AIS and a survival determinant for female AIS patients in the early stage. PMID: 23688269
  35. High expression of TCF3 is associated with neuroblastoma progression. PMID: 23135478
  36. This research provides the first evidence that the portion of E2A proteins found within the oncogenic fusion protein E2A-PBX1, including the transcriptional activation domains, can enhance the HAT activity of CBP/p300. PMID: 22387215
  37. Our findings reveal that E2A proteins are acetylated at specific residues as a consequence of lysine acetyltransferases (CBP, p300, and PCAF) recruitment. PMID: 22207202
  38. The molecular mechanism for the effect of EB1089 on aromatase gene expression was identified as being mediated by the vitamin D receptor (VDR), vitamin D receptor interacting repressor (VDIR), and Williams syndrome transcription factor (WSTF). PMID: 23085504
  39. E2A-HLF sensitizes t(17;19)-positive acute lymphoblastic leukemia to graft-versus-leukemia effect by upregulating death receptors for TRAIL. PMID: 22743623
  40. The absence of T cell precursor potential, both in vivo and in vitro, is attributed to low Notch1 expression, which is secondary to the inhibition of E2A protein activity by members of the inhibitor of DNA binding (Id) protein family. PMID: 22972921
  41. Protein inhibitor of activated STAT, PIASy, regulates alpha-smooth muscle actin expression by interacting with E12 in mesangial cells. PMID: 22829926
  42. These results demonstrate for the first time that E2A can act as a tumor promoter, particularly in prostate cancer. PMID: 22564737
  43. Studies have shown that the entire ensemble of Id proteins has the ability to interact with E47, identify factors that associate with E47, and reveal a spectrum of phosphorylated residues in E47, including an AKT substrate site. PMID: 22354994
  44. The E2A, a key transcription factor associated with the B-cell activation profile, regulates apoptosis in CLL and may contribute to disease pathology. PMID: 21551245
  45. Altering the balance between Id3 and E47 is both necessary and sufficient to regulate the cell cycle in PDA cells. PMID: 21498546
  46. E2A exhibits a tumor suppressor function in human lymphoid cells. PMID: 21788410
  47. These results indicate that in cancer cells, E2A, FOXO1, and FOXP1 regulate RAG1 and RAG2 expression, which initiates Ig gene rearrangement in a manner similar to B lymphocytes. PMID: 21655267
  48. HEB and E2A bind the SCA motif at regions overlapping SMAD2/3 and FOXH1. PMID: 21828274

Show More

Hide All

Database Links

HGNC: 11633

OMIM: 147141

KEGG: hsa:6929

STRING: 9606.ENSP00000262965

UniGene: Hs.371282

Involvement In Disease
Agammaglobulinemia 8, autosomal dominant (AGM8)
Subcellular Location
Nucleus.

Q&A

What is TCF3 and why is it significant in research?

TCF3 (Transcription Factor 3), also known as E2A, is a critical transcription factor involved in early lymphocyte differentiation, particularly B cell development. It plays essential roles in neuronal differentiation and mesenchymal-to-epithelial transition . TCF3 is a member of the TCF/LEF family and functions as a component of the Wnt signaling pathway, serving as a dominant downstream effector in embryonic stem cells . The significance of TCF3 in research stems from its involvement in multiple biological processes, including:

  • Regulation of gene expression during developmental processes

  • Determination of tissue-specific cell fate during embryogenesis

  • B cell differentiation and immune system development

  • Transcriptional response to Wnt signaling

  • Role in certain cancer types, particularly colorectal cancer

What are the main isoforms of TCF3 and how do they differ functionally?

TCF3 exists in multiple isoforms, with E12 and E47 being the most well-characterized. These isoforms arise from alternative splicing of the TCF3 gene and have distinct functions:

  • E47 isoform: Facilitates ATOH7 binding to DNA at the consensus sequence 5'-CAGGTG-3', and positively regulates transcriptional activity .

  • Both isoforms bind to the kappa-E2 site in the kappa immunoglobulin gene enhancer and to IEB1 and IEB2 sequences in the insulin gene transcription control region .

The functional differences between these isoforms are important when selecting antibodies for specific research purposes, as some antibodies may target common regions while others may be isoform-specific.

What applications are TCF3 antibodies commonly used for in research?

TCF3 antibodies are utilized across multiple research applications, including:

ApplicationDescriptionCommon Uses
Western Blotting (WB)Detection of TCF3 protein in cell/tissue lysatesProtein expression analysis, verification of knockdown/overexpression
Immunoprecipitation (IP)Isolation of TCF3 protein complexesProtein-protein interaction studies, chromatin immunoprecipitation
Immunofluorescence (IF)Visualization of TCF3 localization in cellsSubcellular localization studies, co-localization with other proteins
Immunohistochemistry (IHC)Detection of TCF3 in tissue sectionsExpression patterns in normal vs. diseased tissues
ELISAQuantitative detection of TCF3Protein quantification in complex samples

Different antibodies may perform better in specific applications, so selection should be based on validated performance for the intended use .

How should I select the appropriate TCF3 antibody for my specific research question?

When selecting a TCF3 antibody, consider these critical factors:

  • Epitope specificity: Determine whether you need an antibody targeting the N-terminal or another region of TCF3 . This is particularly important if you want to distinguish between different isoforms or study specific domains.

  • Species reactivity: Confirm the antibody recognizes TCF3 in your experimental species. Some antibodies detect human, mouse, and rat TCF3, while others are species-specific .

  • Validated applications: Choose antibodies validated for your specific application (WB, IP, IF, IHC, etc.) through published literature or manufacturer validation data .

  • Clonality consideration:

    • Monoclonal antibodies (e.g., E-2 clone) offer high specificity and reproducibility

    • Polyclonal antibodies may provide better sensitivity through recognition of multiple epitopes

  • Conjugation requirements: Consider whether you need a non-conjugated antibody or specific conjugates (HRP, PE, FITC, Alexa Fluor®) for your detection system .

Reviewing published studies using TCF3 antibodies in similar experimental contexts can provide valuable guidance for antibody selection.

What are the critical controls needed for TCF3 antibody experiments?

Implementing proper controls is essential for generating reliable data with TCF3 antibodies:

Control TypePurposeImplementation
Positive ControlConfirms antibody functionalityUse tissues/cells known to express TCF3 (e.g., PANC-1 nuclear extract , lung, pancreas, or spleen tissue )
Negative ControlAssesses non-specific bindingUse tissues/cells with minimal TCF3 expression or TCF3 knockout samples
Isotype ControlEvaluates background from antibody classUse non-specific antibody of same isotype, species, and concentration
Blocking PeptideConfirms specificityPre-incubate antibody with immunizing peptide before application
siRNA/CRISPR KnockdownValidates signal specificityCompare signal between wildtype and TCF3-depleted samples

For TCF3 studies in immune contexts, comparing signals from normal and TCF3-deficient lymphocytes can serve as an excellent control system, as TCF3-deficient individuals show characteristic B cell developmental defects .

What are the optimal sample preparation methods for detecting TCF3 in different experimental systems?

Sample preparation varies by application and should be optimized for TCF3 detection:

For Western Blotting:

  • Use nuclear extraction protocols as TCF3 is predominantly nuclear

  • Include protease inhibitors to prevent degradation

  • Optimize lysis conditions (RIPA buffer for general use, gentler NP-40 buffer for complex studies)

  • For lymphocyte studies, consider subcellular fractionation to enrich nuclear fractions where TCF3 is primarily localized

For Immunoprecipitation:

  • Cross-linking may be beneficial for studying TCF3 interactions with DNA or RNA

  • For RNA immunoprecipitation (RIP) assays examining TCF3-RNA interactions (such as with ASBEL lncRNA), nuclear extraction followed by gentle lysis is recommended

For Immunofluorescence/Immunohistochemistry:

  • Optimize fixation method (4% paraformaldehyde for general use)

  • Consider antigen retrieval methods for formalin-fixed tissues

  • Test different permeabilization conditions for nuclear access

For Flow Cytometry (particularly in immunological studies):

  • Use gentle fixation to preserve epitopes

  • Ensure adequate permeabilization for nuclear TCF3 detection

  • Consider cell surface markers to identify specific lymphocyte populations when studying TCF3 in immune contexts

How can TCF3 antibodies be used to investigate TCF3 haploinsufficiency in immunodeficiency research?

TCF3 haploinsufficiency has been linked to immunodeficiency with incomplete clinical penetrance, making antibody-based approaches valuable for characterization:

  • Protein expression analysis: Western blotting with TCF3 antibodies can quantify reduced TCF3 protein levels in patient samples. This is critical for differentiating haploinsufficiency from dominant negative mutations, as the former shows approximately 50% reduction in wildtype protein expression .

  • Flow cytometry applications: TCF3 antibodies combined with B cell markers can identify characteristic B cell developmental abnormalities in TCF3 haploinsufficiency, such as:

    • Reduced total B cells

    • Decreased class-switched memory B cells

    • Reduced plasmablast populations

  • Protein-protein interaction studies: Immunoprecipitation using TCF3 antibodies can investigate how haploinsufficiency affects TCF3's interactions with other transcription factors and chromatin modifiers.

  • Functional analyses: TCF3 antibodies can be used in chromatin immunoprecipitation (ChIP) assays to examine how reduced TCF3 binding to target genes affects gene expression profiles in immune cells.

Research on TCF3 haploinsufficiency particularly benefits from combining antibody-based protein detection with functional immunological assays (e.g., plasmablast differentiation, immunoglobulin secretion) to correlate protein levels with phenotypic manifestations .

What methodological approaches can resolve conflicting TCF3 antibody results across different experimental systems?

When facing discrepancies in TCF3 antibody results, employ these methodological approaches:

  • Epitope mapping and antibody characterization:

    • Use multiple antibodies targeting different epitopes of TCF3 (N-terminal vs. other regions)

    • Determine if discrepancies relate to specific isoform detection (E12 vs. E47)

    • Consider epitope masking due to protein-protein interactions or post-translational modifications

  • Validation in genetic models:

    • Utilize TCF3 knockout or knockdown systems as negative controls

    • Compare results with the heterozygous Tcf3+/- mouse model to calibrate antibody sensitivity in detecting reduced expression

  • Context-dependent expression analysis:

    • Recognize that TCF3 expression varies during B cell development (high in bone marrow precursors, downregulated in splenic B cells)

    • Account for potential expression differences between human and mouse systems

  • Technical optimization:

    • Adjust antibody concentration and incubation conditions

    • Optimize sample preparation (nuclear extraction methods)

    • Consider non-denaturing conditions if conformational epitopes are involved

  • Cross-validation with non-antibody methods:

    • Correlate protein detection with mRNA expression (qRT-PCR)

    • Use tagged TCF3 constructs as positive controls

    • Apply mass spectrometry for unbiased protein identification

When studying TCF3 in immunological contexts, it's important to note that human and mouse systems show differences in TCF3 dependence, with murine models only partially recapitulating human TCF3 haploinsufficiency phenotypes .

How can ChIP-seq with TCF3 antibodies be optimized to study TCF3's transcriptional network?

Optimizing ChIP-seq with TCF3 antibodies requires careful consideration of several factors:

  • Antibody selection criteria:

    • Choose ChIP-validated antibodies with demonstrated specificity

    • Consider antibodies recognizing different TCF3 domains to capture various binding modes

    • Test multiple antibodies to identify those yielding highest signal-to-noise ratios

  • Chromatin preparation optimization:

    • Adjust crosslinking conditions (1% formaldehyde for 10-15 minutes is standard, but optimization may be necessary)

    • Optimize sonication parameters to generate 200-500bp fragments

    • Include appropriate controls (input chromatin, IgG control, TCF3-depleted cells)

  • Experimental design considerations:

    • For studying TCF3's role in Wnt signaling, consider parallel experiments with and without Wnt pathway activation

    • In B cell studies, analyze different developmental stages as TCF3 binding patterns change during development

    • For cancer studies, compare TCF3 binding patterns between normal and malignant cells

  • Data analysis approaches:

    • Integrate ChIP-seq with RNA-seq to correlate binding with expression changes

    • Perform motif analysis to identify canonical (5'-CANNTG-3') and non-canonical binding sites

    • Consider co-binding partners (for colorectal cancer, analyze ASBEL lncRNA-TCF3 complex binding at the ATF3 locus)

  • Validation strategies:

    • Confirm select binding sites with ChIP-qPCR

    • Perform functional validation using reporter assays or CRISPR-based approaches

    • For TCF3-regulated genes, verify protein level changes with Western blotting

TCF3 ChIP-seq is particularly informative when examining the impact of TCF3 gene dosage on transcriptional networks, as demonstrated in studies comparing wildtype, haploinsufficient, and dominant negative TCF3 mutations .

What are the common pitfalls in TCF3 Western blotting and how can they be addressed?

Western blotting for TCF3 can present several challenges:

ChallengePotential CauseSolution
Weak or absent signalInsufficient nuclear extractionUse specialized nuclear extraction buffers; verify extraction with other nuclear markers
Low TCF3 expressionIncrease sample loading; use more sensitive detection systems
Ineffective antibodyTry different antibodies (e.g., N-terminal specific)
Multiple unexpected bandsCross-reactivityIncrease blocking; optimize antibody dilution; try monoclonal antibodies
TCF3 isoforms/degradationCompare with literature for expected pattern; include protease inhibitors
High backgroundInsufficient blockingIncrease blocking time/concentration; use alternative blocking agents
Secondary antibody issuesTry different secondary antibody; optimize dilution
Size discrepancyPost-translational modificationsConsider phosphorylation or other modifications affecting migration
Isoform detectionCompare with expression pattern of specific isoforms (E12 vs. E47)

For lymphocyte studies, note that TCF3 expression varies during development - checking expression in specific B cell developmental stages can help in interpreting variable detection results .

How can I distinguish between genuine TCF3 signal and non-specific binding in immunohistochemistry?

To distinguish genuine TCF3 staining from non-specific binding:

  • Comprehensive controls:

    • Positive tissue controls: Use tissues with known TCF3 expression (pancreas, lung, spleen)

    • Negative controls: Omit primary antibody while maintaining all other steps

    • Absorption control: Pre-incubate antibody with immunizing peptide

    • Genetic controls: When possible, use tissue from TCF3-deficient models

  • Pattern recognition:

    • Authentic TCF3 staining should show predominantly nuclear localization

    • Staining pattern should correspond to expected cellular distribution (e.g., enriched in lymphoid tissues, particularly in B cell development zones)

    • Review literature for expected staining patterns in your tissue of interest

  • Technical optimization:

    • Titrate antibody to determine optimal concentration

    • Test multiple antigen retrieval methods

    • Compare multiple TCF3 antibodies targeting different epitopes

  • Advanced validation approaches:

    • Perform dual immunofluorescence with markers of expected TCF3-expressing cells

    • Correlate IHC results with Western blot or mRNA expression data

    • Consider RNAscope for parallel visualization of TCF3 mRNA and protein

When studying TCF3 in lymphoid tissues, correlate staining patterns with known B cell developmental markers to verify biological relevance of the observed signal .

How should I interpret discrepancies between TCF3 protein levels detected by antibodies and TCF3 mRNA expression?

Discrepancies between TCF3 protein and mRNA levels are not uncommon and can provide valuable biological insights:

  • Post-transcriptional regulation mechanisms:

    • Investigate microRNA-mediated regulation of TCF3 translation

    • Examine RNA-binding proteins that might affect TCF3 mRNA stability or translation

    • Consider the role of lncRNAs (such as ASBEL) that might interact with TCF3 at both protein and mRNA levels

  • Post-translational regulation:

    • Assess protein stability and turnover rates (pulse-chase experiments)

    • Investigate ubiquitination and proteasomal degradation pathways

    • Examine how Wnt signaling might affect TCF3 protein stability independent of transcription

  • Methodological considerations:

    • Verify antibody specificity using multiple approaches

    • Ensure appropriate subcellular fractionation (TCF3 is predominantly nuclear)

    • Consider temporal dynamics—protein levels may reflect earlier transcriptional events

  • Biological context interpretation:

    • In B cell development, TCF3 protein levels are tightly regulated despite relatively stable mRNA expression

    • In cancer contexts, post-transcriptional mechanisms may be altered, affecting the relationship between mRNA and protein levels

  • Integrated analysis approaches:

    • Combine proteomics and transcriptomics for comprehensive analysis

    • Use reporter systems to monitor translation efficiency

    • Apply ribosome profiling to assess translation rates

For TCF3 haploinsufficiency studies, note that gene-dosage effects may be more evident at the protein level than mRNA level, making antibody-based quantification particularly important .

How can TCF3 antibodies be utilized to study the ASBEL-TCF3 complex in colorectal cancer research?

The ASBEL-TCF3 complex represents an important target in colorectal cancer research, and antibody-based approaches offer valuable insights:

  • Co-immunoprecipitation strategies:

    • Use TCF3 antibodies to precipitate protein complexes, followed by RNA extraction and qRT-PCR for ASBEL detection

    • Perform RNA immunoprecipitation (RIP) assays with anti-TCF3 antibodies to verify direct interaction with ASBEL lncRNA

    • Combine with mass spectrometry to identify additional protein components of the complex

  • Chromatin studies:

    • Implement ChIP-seq with TCF3 antibodies to map genome-wide binding sites, focusing on the ATF3 locus and other potential targets

    • Use sequential ChIP (ChIP-reChIP) to identify regions co-bound by β-catenin and TCF3

    • Perform CHART (Capture Hybridization Analysis of RNA Targets) with ASBEL probes followed by TCF3 antibody detection

  • Functional analyses:

    • Combine TCF3 antibody-based detection with knockdown/overexpression of ASBEL to assess their interdependent functions

    • Use proximity ligation assays (PLA) to visualize and quantify ASBEL-TCF3 interactions in situ

    • Analyze TCF3 localization changes in response to ASBEL modulation

  • Therapeutic targeting assessment:

    • Monitor changes in the ASBEL-TCF3 complex formation using antibody-based assays after drug treatments

    • Assess effects of Wnt pathway modulators on TCF3 recruitment to target genes

    • Evaluate TCF3 post-translational modifications in response to targeted therapies

The study of ASBEL-TCF3 complex is particularly relevant for understanding the β-catenin–ASBEL–TCF3–ATF3 pathway, which may represent a promising target for colorectal cancer therapy .

What are the methodological considerations for studying TCF3 in the context of Wnt signaling pathways?

Investigating TCF3 in Wnt signaling contexts requires specialized methodological approaches:

  • Pathway activation controls:

    • Use validated Wnt pathway activators (Wnt3a, GSK3 inhibitors) and inhibitors

    • Include β-catenin stabilization verification (e.g., using phospho-specific antibodies)

    • Implement reporter assays (TOPFlash/FOPFlash) to monitor canonical Wnt activity

  • Protein interaction analyses:

    • Perform co-immunoprecipitation with TCF3 antibodies to assess β-catenin interaction under different signaling conditions

    • Use proximity ligation assays to visualize TCF3/β-catenin interactions in situ

    • Consider FRET/BRET approaches for real-time interaction monitoring

  • Chromatin dynamics assessment:

    • Conduct ChIP-seq before and after Wnt stimulation to track TCF3 redistribution

    • Analyze co-occupancy with other TCF/LEF family members

    • Implement ATAC-seq to examine chromatin accessibility changes at TCF3-bound regions

  • Functional readouts:

    • Monitor expression of established TCF3 target genes using qRT-PCR and Western blotting

    • Assess cellular phenotypes relevant to Wnt signaling (proliferation, differentiation)

    • In stem cell contexts, examine pluripotency marker expression and differentiation capacity

  • Model system considerations:

    • Choose appropriate cell models with intact or dysregulated Wnt signaling

    • For cancer studies, compare TCF3 function in APC-mutant versus APC-wildtype contexts

    • Consider the interplay between TCF3 and adenomatous polyposis coli (APC) tumor suppressor protein

Understanding that TCF3 can function as both a repressor and activator of transcription is critical when designing experiments and interpreting results in Wnt signaling contexts .

How can multiplexed antibody approaches advance our understanding of TCF3's role in developmental and disease processes?

Multiplexed antibody approaches offer powerful tools for elucidating TCF3's complex functions:

  • Multi-parameter flow cytometry:

    • Combine TCF3 antibodies with markers of B cell development stages to precisely map expression dynamics

    • Integrate with phospho-specific antibodies to correlate TCF3 expression with signaling pathway activation

    • Include lineage markers to identify specific cell populations affected by TCF3 mutations or dysregulation

  • Multiplex immunohistochemistry/immunofluorescence:

    • Perform sequential immunostaining or spectral unmixing to visualize TCF3 alongside multiple markers

    • Use tissue microarrays to analyze TCF3 expression across numerous samples simultaneously

    • Implement spatial transcriptomics to correlate protein localization with gene expression patterns

  • Mass cytometry (CyTOF) applications:

    • Develop metal-conjugated TCF3 antibodies for high-dimensional single-cell analysis

    • Profile dozens of proteins simultaneously to place TCF3 in broader signaling networks

    • Apply in developmental biology studies to track TCF3's role across differentiation trajectories

  • Proximity-based interactome mapping:

    • Use BioID or APEX2 fusions with TCF3 combined with antibody-based detection of biotinylated proteins

    • Implement protein-fragment complementation assays to screen for novel interaction partners

    • Apply cross-linking mass spectrometry to identify direct TCF3 protein complexes

  • Single-cell applications:

    • Combine index sorting with antibody staining to link TCF3 protein levels to transcriptomic profiles

    • Implement intracellular staining protocols compatible with single-cell RNA-seq

    • Use antibody-based cell sorting to isolate TCF3-expressing populations for further analysis

These multiplexed approaches are particularly valuable for understanding TCF3's diverse roles, from lymphocyte development to cancer progression and embryonic stem cell regulation .

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.