TFDP3 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Product dispatch occurs within 1-3 business days of order receipt. Delivery times may vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Synonyms
Cancer/testis antigen 30 antibody; CT30 antibody; DP4 antibody; E2F like protein antibody; E2F-like antibody; HCA661 antibody; Hepatocellular carcinoma-associated antigen 661 antibody; TFDP3 antibody; TFDP3_HUMAN antibody; Transcription factor Dp family member 3 antibody
Target Names
TFDP3
Uniprot No.

Target Background

Function
This antibody targets TFDP3, a competitive inhibitor of E2F-mediated transactivation, thereby impairing E2F-driven cell cycle progression from G1 to S phase.
Gene References Into Functions
  • A preclinical model of resistance to induction therapy was established to investigate the functional role of TFDP3 in chemoresistance within minimal residual disease (MRD) derived from Jurkat/E6-1 cells. PMID: 27902457
  • TFDP3 binds to E2F1 to form the E2F/TFDP3 complex. The subcellular localization of TFDP3 and E2F1, and their co-localization, vary across different cell cycle phases within the nucleus and cytoplasm, suggesting a role for the E2F/TFDP3 complex in cell cycle regulation. PMID: 28797103
  • Studies indicate TFDP3 expression in breast cancer, its classification as a cancer-testis antigen, and its function as an epithelial-mesenchymal transition regulator. PMID: 28114432
  • High TFDP3 expression was observed in various cancer tissues, including prostate cancer, often in coordination with E2F1 expression. PMID: 24406621
  • Research using cell lines, tumorigenicity studies, and primary hepatocellular carcinoma samples demonstrates a negative regulatory role of HIF-2α in tumors, mediated by the TFDP3/E2F1 pathway. PMID: 23212661
  • TFDP3 upregulates the autophagy gene LC3B and inhibits E2F1-induced apoptosis, potentially playing a significant role in prostate cancer. PMID: 22482402
  • DP-4 downregulates E2F-1 activity, contributing to a novel pRb-independent mechanism for regulating cell cycle progression. PMID: 20559320
  • TFDP3, a novel DP protein, inhibits E2F DNA binding and transactivation. PMID: 17062573
Database Links

HGNC: 24603

OMIM: 300772

KEGG: hsa:51270

STRING: 9606.ENSP00000385461

UniGene: Hs.142908

Protein Families
E2F/DP family
Subcellular Location
Nucleus. Cytoplasm. Note=Translocates to the nucleus on heterodimerization with E2F family members.
Tissue Specificity
Predominantly expressed in testis. Low level of expression in pancreas. Highly expressed in ovarian and colon cancer cell lines.

Q&A

What is TFDP3 and why is it significant in cancer research?

TFDP3, also known as HCA661 or CT30, is a cancer-testis antigen primarily expressed in normal testis and multiple cancer types. The TFDP3 gene (Gene ID: 51270) is located on chromosome X and shares high sequence homology with TFDP1 and TFDP2 . Unlike its family members, TFDP3 uniquely downregulates E2F-mediated transcriptional activation and can inhibit E2F1-mediated apoptosis . This protein is particularly significant in cancer research because it is involved in cell autophagy, epithelial-mesenchymal transition, and contributes to chemoresistance in various cancers, making it a potential target for cancer therapies .

What are the key differences between TFDP3 and other DP family members that researchers should consider when selecting antibodies?

TFDP3 differs significantly from TFDP1 and TFDP2 in both function and expression pattern. While TFDP1 and TFDP2 enhance DNA-binding activity of E2Fs, TFDP3 downregulates E2F-mediated transcriptional activation . This functional difference stems from four key amino acid substitutions in the DNA-binding domain and differences in the C-terminal region . Additionally, TFDP3 shows a restricted expression profile (primarily in testis and cancer cells), whereas TFDP1 is ubiquitously expressed in various tissues . When selecting antibodies, researchers should target epitopes unique to TFDP3 to avoid cross-reactivity with other DP family members, particularly focusing on regions containing these amino acid differences.

What are the recommended fixation and permeabilization methods when using TFDP3 antibodies for immunocytochemistry?

For optimal TFDP3 detection in immunocytochemistry, the following protocol is recommended based on research practices:

  • Fixation: 4% paraformaldehyde for 15-20 minutes at room temperature

  • Permeabilization: 0.1-0.5% Triton X-100 for 10 minutes

  • Blocking: 5% normal serum (matching secondary antibody host) with 1% BSA for 1 hour

This approach preserves TFDP3 epitopes while allowing antibody access to subcellular compartments. The choice of fixation method is particularly important as TFDP3 shows dynamic subcellular localization during different cell cycle phases, being predominantly nuclear during late mitosis and G1, and cytoplasmic during G2 phase . When analyzing TFDP3 co-localization with E2F1, these considerations become especially critical to accurately capture their interactions.

How can researchers effectively use TFDP3 antibodies to track subcellular localization changes throughout the cell cycle?

TFDP3 exhibits dynamic subcellular localization that changes throughout the cell cycle. Studies have shown that TFDP3 is primarily cytoplasmic when expressed without E2Fs, but translocates to the nucleus when co-expressed with E2F1-3 . Additionally, TFDP3 is expressed primarily in the nucleus at the end of mitosis, diffusively in G1 phase, cytoplasmic in G2 phase, and highly expressed during S phase .

To effectively track these changes:

  • Synchronize cells using thymidine double repression method to obtain populations at specific cell cycle phases

  • Perform immunofluorescence using anti-TFDP3 antibodies at each phase

  • Co-stain with cell cycle markers (e.g., cyclin proteins) and DNA stains

  • Use confocal microscopy for high-resolution imaging of subcellular compartments

For quantitative assessment, researchers should analyze at least 100 cells per condition and develop a scoring system for nuclear vs. cytoplasmic distribution patterns .

What controls should be included when validating TFDP3 antibody specificity for Western blot analysis?

When validating TFDP3 antibody specificity for Western blot analysis, researchers should include the following controls:

Control TypeDescriptionPurpose
Positive controlTestis tissue lysate or TFDP3-expressing cancer cell lines (e.g., HepG2)Confirms antibody can detect endogenous TFDP3
Negative controlNormal liver tissue lysate or other tissues with minimal TFDP3 expressionConfirms absence of non-specific binding
Knockdown/knockout controlLysates from cells with TFDP3 silenced via siRNAConfirms signal specificity
Overexpression controlLysates from cells transfected with TFDP3 expression vectorConfirms antibody detects overexpressed protein
Peptide competitionPre-incubation of antibody with immunizing peptideConfirms epitope specificity

Additionally, researchers should compare TFDP3 expression across synchronized cell populations, as TFDP3 shows dynamic expression levels throughout the cell cycle, with highest expression during S phase as determined by Western blot analysis of synchronized cells .

What are the methodological considerations for detecting TFDP3 in cancer tissue microarrays using immunohistochemistry?

When detecting TFDP3 in cancer tissue microarrays via immunohistochemistry, researchers should consider:

  • Antibody validation: First validate the TFDP3 antibody using positive controls (testis tissue) and negative controls (normal tissues with minimal expression) .

  • Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0) is recommended as TFDP3 may be masked by formalin fixation.

  • Detection system selection: Use a sensitive detection system (e.g., polymer-based) as TFDP3 expression can vary significantly between cancer types.

  • Scoring system implementation: Develop a standardized scoring system that accounts for:

    • Percentage of positive cells (0-100%)

    • Staining intensity (0: negative, 1+: weak, 2+: moderate, 3+: strong)

    • Subcellular localization (nuclear, cytoplasmic, or both)

  • Cancer type specificity: Consider that TFDP3 is significantly expressed in hepatocellular carcinoma, breast cancer, prostate cancer, and T-cell acute lymphoblastic leukemia, with expression patterns differing between cancer types .

  • EMT marker co-staining: For breast cancers, consider co-staining with EMT markers as TFDP3 preferentially expresses in mesenchymal rather than luminal types .

How can researchers design experiments to investigate the differential binding of TFDP3 to E2F family members using immunoprecipitation with TFDP3 antibodies?

To investigate differential binding of TFDP3 to E2F family members, researchers should design co-immunoprecipitation experiments with the following methodological considerations:

  • Cell model selection: Choose appropriate cell models expressing TFDP3 (e.g., HepG2, breast cancer cell lines) or transfect cells with TFDP3 expression vectors .

  • Cell synchronization: Synchronize cells at different cell cycle phases using thymidine double block or nocodazole treatment, as TFDP3-E2F interactions may vary throughout the cell cycle .

  • Immunoprecipitation protocol:

    • Use validated TFDP3 antibodies for immunoprecipitation

    • Include appropriate negative controls (isotype-matched IgG, TFDP3-knockout cells)

    • Optimize lysis conditions to preserve protein-protein interactions (use non-denaturing buffers)

    • Perform reverse IP with E2F antibodies to confirm interactions

  • Detection strategy:

    • Immunoblot for different E2F family members (E2F1-6)

    • Quantify relative binding affinities across E2F family members

    • Use truncated E2F constructs to map interaction domains

  • Functional validation: Confirm that observed interactions affect downstream E2F target genes using reporter assays or ChIP experiments.

Given that TFDP3 has been shown to interact with E2F1 and affects its nuclear translocation, researchers should pay particular attention to subcellular fractionation controls and nuclear/cytoplasmic distribution patterns of both proteins when analyzing results .

What approaches can be used to reconcile contradictory findings regarding TFDP3 subcellular localization in different studies?

The literature contains contradictory findings regarding TFDP3 subcellular localization, with some studies reporting cytoplasmic distribution while others report nuclear localization . To reconcile these contradictions, researchers should implement multiple complementary approaches:

  • Cell cycle-specific analysis: Synchronize cells at different cell cycle phases and analyze TFDP3 localization at each phase, as studies have shown TFDP3 exhibits dynamic subcellular localization throughout the cell cycle (nuclear at late mitosis, diffuse in G1, cytoplasmic in G2) .

  • Co-expression partners: Examine TFDP3 localization both with and without E2F co-expression, as TFDP3 translocates to the nucleus when co-expressed with E2F1-3 .

  • Multiple detection methods:

    • Immunofluorescence with different validated antibodies

    • Biochemical fractionation followed by Western blotting

    • Live-cell imaging with fluorescently tagged TFDP3

  • Cell type considerations: Compare localization across multiple cell types, as cell-specific factors may influence TFDP3 distribution.

  • Epitope accessibility analysis: Use antibodies targeting different TFDP3 epitopes to rule out epitope masking in certain cellular compartments.

  • Stimulus-dependent changes: Examine localization under various conditions (e.g., DNA damage, cell stress) as TFDP3 can be induced by DNA damage and may relocalize in response to cellular stressors .

By systematically addressing these variables, researchers can develop a more comprehensive model of TFDP3 localization dynamics that reconciles apparently contradictory findings.

How can researchers design ChIP-seq experiments using TFDP3 antibodies to identify differential genome-wide binding patterns of TFDP3/E2F complexes?

Designing effective ChIP-seq experiments for TFDP3 requires careful consideration of several methodological aspects:

  • Antibody validation for ChIP application:

    • Perform preliminary ChIP-PCR on known E2F target genes

    • Verify antibody specificity using TFDP3 knockdown controls

    • Test antibody under different crosslinking conditions

  • Experimental design considerations:

    • Include synchronized cell populations (G1, S, G2 phases)

    • Compare TFDP3 binding with and without DNA damage induction

    • Include parallel ChIP-seq for E2F family members to identify co-occupied sites

  • Control experiments:

    • Input DNA controls

    • IgG mock ChIP

    • TFDP3-depleted cells as negative control

  • Bioinformatic analysis pipeline:

    • Identify TFDP3-bound regions and compare with known E2F binding sites

    • Perform motif analysis to identify potential DNA binding motifs

    • Integrate with gene expression data to correlate binding with transcriptional effects

    • Compare TFDP3 binding profiles with TFDP1/TFDP2 patterns to identify unique targets

  • Validation of findings:

    • Confirm select binding sites via ChIP-qPCR

    • Perform reporter assays to validate functional effects on transcription

    • Use CRISPR-Cas9 to mutate binding sites and assess functional consequences

This approach would enable researchers to identify the genome-wide binding profile of TFDP3 and compare it with other DP family members, potentially uncovering mechanisms underlying TFDP3's unique role in downregulating E2F-mediated transcription .

What strategies can address false negative results when detecting low TFDP3 expression levels in early-stage cancers?

Detecting low TFDP3 expression in early-stage cancers can be challenging. To address false negative results, researchers should consider:

  • Signal amplification techniques:

    • Use tyramide signal amplification (TSA) for immunohistochemistry

    • Implement highly sensitive chemiluminescent substrates for Western blot

    • Consider RNAscope® for in situ mRNA detection with single-molecule sensitivity

  • Sample preparation optimization:

    • Optimize fixation times to prevent epitope masking

    • Test multiple antigen retrieval methods (heat vs. enzymatic)

    • Use fresh-frozen samples when possible to preserve antigenicity

  • Antibody selection and concentration:

    • Test multiple antibodies targeting different TFDP3 epitopes

    • Perform antibody titration to determine optimal concentration

    • Consider using antibody cocktails to improve detection

  • Pre-enrichment strategies:

    • Implement laser capture microdissection to isolate cancer cells

    • Use subcellular fractionation to concentrate samples based on known TFDP3 localization patterns

    • Consider immunoprecipitation followed by Western blot for enhanced sensitivity

  • Quantitative considerations:

    • Employ digital image analysis for precise quantification

    • Use highly sensitive qRT-PCR as a complementary approach

    • Consider droplet digital PCR for absolute quantification of low-abundance transcripts

Studies have shown that TFDP3 expression can be variable and may increase during cancer progression or following DNA damage , so temporal sampling may also be important for comprehensive detection.

How can researchers optimize co-immunoprecipitation protocols to preserve transient TFDP3-E2F interactions during different cell cycle phases?

To optimize co-immunoprecipitation protocols for capturing transient TFDP3-E2F interactions across different cell cycle phases:

  • Cell synchronization and crosslinking:

    • Implement precise synchronization methods to obtain populations at specific cell cycle phases

    • Use reversible protein crosslinkers (e.g., DSP) to stabilize transient interactions

    • Optimize crosslinking time to balance between interaction preservation and antibody epitope accessibility

  • Lysis buffer optimization:

    • Use mild, non-denaturing buffers to preserve protein-protein interactions

    • Include appropriate phosphatase inhibitors to maintain phosphorylation-dependent interactions

    • Test different detergent concentrations to optimize solubilization while preserving complexes

  • Antibody selection and binding conditions:

    • Choose antibodies targeting regions not involved in protein-protein interactions

    • Optimize antibody concentration and binding time/temperature

    • Consider using recombinant TFDP3 with affinity tags as an alternative approach

  • Washing stringency:

    • Determine optimal washing conditions that remove non-specific binding while preserving specific interactions

    • Consider using a gradient of salt concentrations in wash buffers

  • Controls and validation:

    • Include cycle-phase specific markers to confirm synchronization

    • Use TFDP3 knockdown cells as negative controls

    • Perform reverse IPs (using E2F antibodies) to confirm interactions

This approach is particularly important because TFDP3-E2F interactions show cell cycle-dependent patterns, with TFDP3 expression and localization varying significantly throughout the cell cycle .

What methodological approaches can distinguish between TFDP3's direct effects on E2F activity versus its indirect effects through autophagy induction?

Distinguishing between direct and indirect effects of TFDP3 requires systematic experimental design:

  • Temporal separation studies:

    • Perform time-course experiments after TFDP3 induction/depletion

    • Monitor both E2F activity and autophagy markers sequentially

    • Use mathematical modeling to determine which effects precede others

  • Genetic dissection approaches:

    • Generate TFDP3 mutants that selectively disrupt E2F binding but preserve other functions

    • Create domain-specific mutations that affect specific TFDP3 functions

    • Use these mutants to separate direct E2F effects from autophagy effects

  • Conditional manipulation systems:

    • Implement inducible TFDP3 expression systems

    • Use autophagy inhibitors (e.g., 3-methyladenine) and measure E2F activity

    • Conversely, use E2F activity modulators and measure autophagy

  • Mechanistic pathway analysis:

    • Investigate the p53 pathway as a potential bridge, as TFDP3 has been shown to promote autophagy by affecting p53 expression

    • Perform epistasis experiments with p53 manipulation to determine pathway order

    • Use ChIP-seq to identify direct TFDP3 binding sites on autophagy-related genes versus E2F targets

  • Single-cell analysis:

    • Implement single-cell analysis to identify heterogeneity in responses

    • Use dual reporters to simultaneously track E2F activity and autophagy

    • Correlate findings at single-cell level to determine relationship between processes

These approaches would help clarify the mechanism underlying the observation that TFDP3 inhibits E2F1-mediated apoptosis while promoting autophagy, which appears contradictory given TFDP3's general role as an E2F activity suppressor .

How might researchers develop TFDP3 antibody-based tools for monitoring treatment response in chemoresistant tumors?

Developing TFDP3 antibody-based tools for monitoring treatment response could leverage several key findings about TFDP3's role in chemoresistance:

  • Development of immunoassays for liquid biopsies:

    • Design sensitive ELISA or electrochemiluminescence immunoassays to detect circulating TFDP3

    • Correlate TFDP3 levels with treatment response in longitudinal patient samples

    • Develop multiplex assays that simultaneously measure TFDP3 and other chemoresistance markers

  • In vivo imaging applications:

    • Develop TFDP3 antibody-based imaging probes (e.g., antibody fragments labeled with imaging agents)

    • Employ these probes for non-invasive monitoring of TFDP3 expression in tumors during treatment

    • Correlate imaging signals with treatment response and resistance development

  • Tissue-based prognostic assessments:

    • Establish standardized immunohistochemistry protocols for TFDP3 detection in biopsies

    • Develop digital pathology algorithms to quantify TFDP3 expression patterns

    • Correlate expression with treatment outcomes in retrospective and prospective studies

  • Functional antibody applications:

    • Generate antibody drug conjugates targeting TFDP3-expressing cells

    • Evaluate their efficacy in eliminating chemoresistant tumor cells

    • Combine with conventional chemotherapy to prevent resistance development

These approaches leverage findings that TFDP3 confers chemoresistance in minimal residual disease within childhood T-cell acute lymphoblastic leukemia and can trigger autophagy during chemotherapy, potentially contributing to drug resistance by facilitating DNA repair .

What experimental design would best evaluate the potential of TFDP3 antibodies in immunotherapy approaches targeting cancer-testis antigens?

To evaluate TFDP3 antibodies for immunotherapy targeting this cancer-testis antigen:

  • Epitope identification and validation:

    • Identify HLA-restricted TFDP3 epitopes recognized by T cells

    • Validate these epitopes using synthetic peptides and T cell assays

    • Generate antibodies that specifically recognize these immunogenic epitopes

    • Previous research has identified two HLA-A*0201-restricted peptides, H110 and H246, as attractive candidates

  • In vitro assessment of antibody-dependent cellular cytotoxicity (ADCC):

    • Develop TFDP3-specific antibodies optimized for effector functions

    • Test ADCC using various immune effector cells against TFDP3-expressing cancer cell lines

    • Compare efficacy across different cancer types known to express TFDP3

  • Dendritic cell (DC) vaccination approaches:

    • Evaluate antibody-targeted delivery of TFDP3 to DCs

    • Compare with established recombinant adenovirus-expressing TFDP3 for DC transduction

    • Assess DC maturation and T cell activation capabilities

    • Building on findings that TFDP3-transduced DCs can activate T cells to target hepatoma

  • Bispecific antibody development:

    • Design bispecific antibodies linking TFDP3 recognition with T cell activation

    • Test efficacy in redirecting T cells against TFDP3-expressing tumor cells

    • Evaluate specificity using normal tissues with minimal TFDP3 expression

  • In vivo models:

    • Establish humanized mouse models with TFDP3-expressing tumors

    • Evaluate antibody-based therapies for efficacy and safety

    • Since mice lack a TFDP3 homolog, consider using rhesus or chimpanzee models that have TFDP3 homologs for safety assessment

This comprehensive approach builds on existing knowledge that TFDP3 is immunogenic and potentially useful for cancer immunotherapy .

How can researchers design experiments to study the impact of TFDP3 on immune cell function in the tumor microenvironment using antibody-based approaches?

To investigate TFDP3's impact on immune cells in the tumor microenvironment:

  • Multiplex immunohistochemistry/immunofluorescence:

    • Design panels combining TFDP3 antibodies with immune cell markers

    • Quantify spatial relationships between TFDP3-expressing tumor cells and immune infiltrates

    • Correlate patterns with clinical outcomes

  • Single-cell suspension analysis:

    • Develop protocols for simultaneous detection of TFDP3 and immune cell markers by flow cytometry

    • Isolate and characterize immune cells based on proximity to TFDP3+ tumor cells

    • Assess functional states of immune cells in relation to TFDP3 expression levels

  • Co-culture experimental systems:

    • Establish co-cultures of TFDP3-expressing tumor cells with various immune cells

    • Use TFDP3 antibodies to block or detect TFDP3 during interactions

    • Compare immune cell function when exposed to wild-type versus TFDP3-knockdown tumor cells

  • Secretome analysis:

    • Analyze how TFDP3 expression affects the tumor secretome

    • Investigate effects of these secreted factors on immune cell function

    • Use antibody-based cytokine arrays to profile differences

  • In vivo immune monitoring:

    • Generate TFDP3-reporter tumor models to track expression

    • Simultaneously monitor immune infiltration and activation

    • Employ antibody-based depletion of specific immune populations to determine contributions

This experimental approach would extend current knowledge about TFDP3's immunogenicity and potential as an immunotherapy target by characterizing its broader effects on anti-tumor immunity, an area that remains largely unexplored according to the current literature.

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.