E2F6 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Stored at -20°C, Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receiving them. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
Synonyms
E2F 6 antibody; E2F binding site modulating activity protein antibody; E2F transcription factor 6 antibody; E2F transcription factor 6 isoform 1 antibody; E2F-6 antibody; E2F6 antibody; E2F6_HUMAN antibody; EMA antibody; MGC111545 antibody; Transcription factor E2F6 antibody
Target Names
E2F6
Uniprot No.

Target Background

Function
E2F6 Antibody is an inhibitor of E2F-dependent transcription. It binds to DNA cooperatively with DP proteins through the E2 recognition site, 5'-TTTC[CG]CGC-3'. It exhibits a preference for the 5'-TTTCCCGC-3' E2F recognition site. E2F6 lacks the transcriptional activation and pocket protein binding domains. It appears to regulate a subset of E2F-dependent genes that are essential for cell cycle entry but not for normal cell cycle progression. E2F6 represses the expression of some meiosis-specific genes, including SLC25A31/ANT4. It may silence gene expression by recruiting a chromatin remodeling complex containing histone H3-K9 methyltransferase activity. Overexpression of E2F6 delays cell exit from the S-phase.
Gene References Into Functions
  1. This analysis revealed a relative increase in the expression of E2F6 in gastric adenocarcinoma without lymph node metastasis (chi (2), P = 0.04 and OR, P = 0.08). Conversely, overexpression of RhoA and SMUG1 was more prevalent in the diffuse subtype of gastric adenocarcinoma compared to the intestinal subtype. PMID: 27909884
  2. Following replication stress, the checkpoint kinase Chk1 phosphorylates E2F6, causing its dissociation from promoters. This facilitates E2F-dependent transcription, promoting cell survival by preventing DNA damage and cell death. PMID: 23954429
  3. Findings suggest an inhibitory role of E2F6 in the regulation of IL-13 and allergy. PMID: 22981205
  4. E2F6 may recruit BRG1 in the transcriptional regulation of genes crucial for the G1/S phase transition of the cell cycle. PMID: 23082233
  5. Data suggests that E2F- and Myc-responsive genes are coregulated by the E2F6 complex in quiescent cells. PMID: 12004135
  6. Results indicate that E2F6 represses transcription of the brca1, ctip, art27, hp1alpha, and the rbap48 genes. Depletion of E2F6 led to the recruitment of E2F1 to the target promoters. PMID: 12909625
  7. E2F6 comprises nine exons distributed along 20.4kbp of genomic DNA on chromosome 2, resulting in the transcription of six alternatively spliced E2F6 mRNAs that encode four different E2F6 proteins. PMID: 15081404
  8. E2F6 lacks the domains required for the modulation of squamous differentiation. PMID: 15474455
  9. Studies revealed that E2F6, DP1, EPC1, EZH2, and Sin3B co-elute, indicating the presence of a novel E2F6 complex in vivo in both normal and transformed human cell lines. PMID: 15536069
  10. E2F6 functions as a repressor of E2F-dependent transcription during the S phase. PMID: 15574595
  11. PHC3 and E2F6 exhibited nuclear colocalization in a punctate pattern, which was not observed in proliferating cells. This suggests that PHC3 may be part of an E2F6-polycomb complex that silences target promoters in G(0). PMID: 17001316
  12. E2F-6 inhibits Apaf-1 upregulation by competing with E2F-1 for promoter binding. It enhances the clonogenic growth of granulocyte, erythroid, macrophage, & megakaryocyte cells. It may prevent hematopoietic progenitor cell loss during proliferation. PMID: 17600109
  13. E2F6 binding sites are located within 2 kb of a transcription start site, both in normal and tumor cells. PMID: 17908821

Show More

Hide All

Database Links

HGNC: 3120

OMIM: 602944

KEGG: hsa:1876

STRING: 9606.ENSP00000370936

UniGene: Hs.603093

Protein Families
E2F/DP family
Subcellular Location
Nucleus.
Tissue Specificity
Expressed in all tissues examined. Highest levels in placenta, skeletal muscle, heart, ovary, kidney, small intestine and spleen.

Q&A

What distinguishes E2F6 from other E2F family members, and how does this affect antibody selection?

E2F6 is structurally distinct from other E2F family members as it lacks the C-terminal sequences responsible for transcriptional activation and binding to retinoblastoma protein (pRb) family members. This unique structure means E2F6 primarily functions as a transcriptional repressor through association with polycomb group proteins . When selecting an antibody, researchers should consider epitopes within E2F6's distinct domains:

  • Repression domain (interacts with RYBP)

  • DNA binding domain (recognizes 5'-TTTC[CG]CGC-3' sequences)

  • Marked box domain (critical for silencing germline genes)

For optimal specificity, select antibodies raised against regions that have minimal homology with other E2F family members to avoid cross-reactivity . Validation should include western blot analysis comparing E2F6 detection against other E2F proteins (E2F1-E2F5) to confirm specificity.

How should researchers validate E2F6 antibodies before experimental use?

Thorough validation is essential for reliable E2F6 antibody performance. A comprehensive validation approach includes:

  • Western blot analysis: Test the antibody against recombinant E2F6 and cell lysates known to express E2F6 (e.g., Jurkat, K-562, ML-1 cells). Observe for a distinct band at approximately 32 kDa .

  • Specificity testing: Ensure the antibody recognizes E2F6 but not other E2F family members (E2F1-E2F5) in western blots and immunoprecipitation assays .

  • Peptide competition assay: Pre-incubate the antibody with the immunizing peptide to confirm signal elimination in western blots or immunohistochemistry .

  • Sibling antibody comparison: When possible, validate results with multiple antibody clones (e.g., LLF6-1 and LLF6-2 as used in published research) .

  • Positive control tissues/cells: Use cells known to express high levels of E2F6, such as proliferating cells at G1/S transition or breast cancer cell lines that overexpress E2F6 .

TechniqueKey Validation PointsExpected Results
Western BlotBand specificity31-32 kDa band
ImmunoprecipitationPull-down specificityRecovers only E2F6
ELISASensitivity testingDetects recombinant E2F6
ChIPTarget enrichmentEnrichment at E2F target genes

How effective are E2F6 antibodies in chromatin immunoprecipitation (ChIP) studies, and what experimental considerations should be addressed?

E2F6 antibodies have proven effective in ChIP assays to identify E2F6 binding sites across the genome. Research shows E2F6 predominantly binds to core promoter regions, particularly within 2 kb of transcription start sites . When designing ChIP experiments:

  • Antibody amount optimization: Titration experiments (1-5 μg per ChIP) should be conducted to determine optimal antibody concentration. Research indicates 5 μg per ChIP often yields optimal results for polyclonal E2F6 antibodies .

  • Appropriate controls: Include IgG controls and positive control targets such as known E2F6-regulated genes. E2F6 binds to a subset of E2F target genes that are activated at G1/S during S phase of the cell cycle .

  • Cell cycle synchronization: For accurate mapping of E2F6 binding dynamics, synchronize cells at different cell cycle phases, as E2F6 shows cell cycle-dependent binding patterns with peak association during S phase .

  • Crosslinking conditions: Standard 1% formaldehyde for 10 minutes is typically sufficient, but optimization may be required for specific experimental questions.

  • Target verification: Confirm enrichment at known E2F6 target genes using qPCR before proceeding to genome-wide analyses .

ChIP-seq studies have shown that E2F6 preferentially binds to CpG islands in embryonic cells, particularly at germline genes, information that should guide experimental design when studying E2F6's epigenetic roles .

What are the optimal conditions for using E2F6 antibodies in co-immunoprecipitation experiments to study protein-protein interactions?

E2F6 forms complexes with multiple proteins, particularly members of the polycomb repressive complex. For optimal co-immunoprecipitation results:

  • Lysis buffer selection: Use E1A lysis buffer or similar non-denaturing buffers that preserve protein-protein interactions while effectively solubilizing nuclear proteins .

  • Antibody selection considerations:

    • Some E2F6 antibodies (like LLF6-1) may destabilize certain E2F6 protein complexes

    • Anti-RYBP antibodies have shown better recovery of E2F6-RYBP complexes than anti-E2F6 antibodies

  • Pre-clearing protocol: Pre-clear lysates with protein A-Sepharose beads at 4°C for 30 minutes to reduce non-specific binding .

  • Complex stabilization: Consider crosslinking approaches for transient interactions.

  • Interaction verification: Confirm interactions by reciprocal immunoprecipitation (e.g., both anti-E2F6 and anti-partner protein antibodies) .

Published research has identified several E2F6 interaction partners that could be verified through co-IP experiments:

  • RYBP (Ring 1 and YY1 binding protein)

  • Bmi1

  • Ring1

  • MEL-18

  • mph1

  • DP proteins (heterodimeric partners)

How can E2F6 antibodies be used to investigate cell cycle-dependent E2F6 functions?

E2F6 exhibits cell cycle-dependent activity, particularly during the S phase where it represses G1/S-activated genes. When designing experiments to study this dynamic regulation:

  • Cell synchronization protocols:

    • Double thymidine block for G1/S boundary synchronization

    • Nocodazole treatment for G2/M arrest

    • Serum starvation for G0/G1 synchronization

  • Time-course sampling: Collect samples at multiple time points after synchronization release (typically 0, 3, 6, 9, 12, 15, 18, 21, 24 hours) to track E2F6 binding dynamics throughout the cell cycle .

  • Dual analysis approach:

    • ChIP with E2F6 antibodies to identify temporal binding patterns

    • Western blot analysis to monitor E2F6 protein levels throughout the cell cycle

    • qRT-PCR of E2F target genes to correlate binding with transcriptional outcomes

  • Target gene selection: Focus on known E2F6 targets that are activated at G1/S but not those activated at G2/M, as research shows E2F6 specifically associates with G1/S-activated E2F target genes during S phase .

Research demonstrates that E2F6 accumulates during G1, reaching peak levels at the G1/S transition, and then interacts with E2F target genes during S phase to restrict their expression and promote cell cycle progression .

What methodological approaches can resolve contradictory findings regarding E2F6 expression in cancer tissues?

Contradictory findings exist regarding E2F6 expression in cancer. For example, some studies report elevated E2F6 expression in breast tumors , while others found no significant difference between tumor and normal tissues . To address such contradictions:

  • Multiple detection methods: Employ independent techniques:

    • qRT-PCR for transcript level quantification

    • Western blot for protein level assessment

    • Immunohistochemistry for tissue distribution patterns

    • ChIP-seq for genome-wide binding patterns

  • Transcript variant analysis: Specifically design primers to distinguish E2F6 variants, as research has shown differential expression of E2F6 variants in breast cancer (variant a overexpressed, variant b underexpressed) .

  • Cancer subtype stratification: Analyze E2F6 expression across different cancer subtypes (e.g., ER-positive vs. ER-negative breast cancers) since significant differences in expression have been observed between subtypes .

  • Statistical approach: Use appropriate statistical tests (e.g., Mann-Whitney U test) and sufficient sample sizes to detect significant differences.

  • Controls and normalization: Include multiple reference genes for qPCR normalization and appropriate positive controls (e.g., Jurkat cells known to overexpress E2F6) .

A comprehensive approach using these methods can help resolve seemingly contradictory findings by accounting for cancer heterogeneity, transcript variants, and cell-type specific E2F6 functions.

How can E2F6 antibodies be leveraged to study epigenetic silencing mechanisms in early embryonic development?

E2F6 plays a critical role in targeting and initiating epigenetic silencing of germline genes during early embryonic development. To investigate these mechanisms:

  • ChIP-seq experimental design: Use E2F6 antibodies in ChIP-seq experiments on embryonic cells to identify genome-wide binding patterns, focusing on CpG islands where E2F6 preferentially binds .

  • Sequential ChIP (ChIP-reChIP): Perform sequential ChIP with E2F6 antibodies followed by antibodies against polycomb complex proteins (Bmi1, Ring1, MEL-18) or histone modifications (H3K27me3, H3K9me3) to identify co-occupied regulatory regions .

  • Developmental time-course: Analyze E2F6 binding and associated epigenetic modifications across different embryonic stages to track the establishment of epigenetic silencing.

  • DNA methylation correlation: Combine E2F6 ChIP-seq with bisulfite sequencing to correlate E2F6 binding with DNA methylation patterns at CpG islands during implantation .

  • Functional studies: Use E2F6 knockout/knockdown models to assess the consequences of E2F6 loss on epigenetic silencing patterns of germline genes.

Research has demonstrated that E2F6 cooperates with MGA to silence germline genes in mouse embryonic stem cells and embryos, with this function critically depending on the E2F6 marked box domain. E2F6 inactivation leads to failure in depositing CpG island DNA methylation during implantation .

What approaches can distinguish between direct and indirect effects when using E2F6 antibodies in chromatin studies?

Distinguishing direct from indirect E2F6 effects requires sophisticated experimental designs:

  • Motif analysis of binding sites: Identify canonical E2F binding motifs (5'-TTTC[CG]CGC-3') within ChIP-seq peaks to identify direct binding events, as E2F6 shows preference for the 5'-TTTCCCGC-3' recognition site .

  • DNA binding domain mutants: Compare binding patterns of wild-type E2F6 with DNA binding domain mutants to identify sites depending on direct DNA interaction.

  • Rapid depletion systems: Use degron-tagged E2F6 for rapid protein depletion and analyze immediate versus delayed changes in chromatin and transcriptional states.

  • Protein-protein interaction disruption: Target specific interaction domains (e.g., the region interacting with RYBP) to disrupt specific E2F6 functions while preserving others .

  • Integration of multiple datasets:

    • E2F6 ChIP-seq

    • RNA-seq after E2F6 depletion

    • ATAC-seq or DNase-seq for chromatin accessibility

    • Histone modification ChIP-seq (e.g., H3K27me3, H3K9me3)

These integrated approaches can determine which genomic sites are directly bound by E2F6 versus those affected through secondary mechanisms or downstream effects of E2F6-mediated repression.

What are the most common technical challenges when using E2F6 antibodies, and how can researchers overcome them?

Several technical challenges may arise when working with E2F6 antibodies:

  • Complex destabilization during immunoprecipitation:

    • Problem: Some E2F6 antibodies (e.g., LLF6-1) may destabilize certain E2F6 protein complexes

    • Solution: Use antibodies directed against E2F6 partner proteins (e.g., anti-RYBP) for more stable complex recovery

  • Nuclear protein extraction efficiency:

    • Problem: Incomplete extraction of nuclear E2F6

    • Solution: Use specialized nuclear extraction buffers with appropriate salt concentration (typically 300-420 mM NaCl) and add nuclease treatment

  • Cross-reactivity with other E2F family members:

    • Problem: Antibody recognizes multiple E2F proteins

    • Solution: Validate antibody specificity with western blots against recombinant E2F1-E2F6 proteins; use knockout/knockdown controls

  • Fixation sensitivity in ChIP experiments:

    • Problem: Over-fixation can mask epitopes

    • Solution: Optimize formaldehyde concentration (0.5-1%) and fixation time (8-12 minutes) for nuclear transcription factors

  • Cell-type specific differences in epitope accessibility:

    • Problem: Variable antibody performance across cell types

    • Solution: Validate antibody in each new cell type before proceeding with full experiments

How should researchers interpret E2F6 antibody results in the context of cellular heterogeneity and cell cycle dynamics?

Interpreting E2F6 antibody results requires careful consideration of cellular context:

  • Single-cell resolution techniques:

    • Consider coupling immunofluorescence staining with cell cycle markers (PCNA, EdU incorporation)

    • Use flow cytometry with E2F6 antibodies and DNA content staining to correlate E2F6 levels with cell cycle phases

  • Cell synchronization considerations:

    • Account for synchronization method artifacts

    • Include asynchronous cell populations as controls

    • Perform time-course experiments after synchronization release

  • Integration with cell cycle markers:

    • Correlate E2F6 binding with markers of specific cell cycle phases

    • Analyze co-binding of E2F6 with other cell cycle regulators

  • Data normalization approaches:

    • For ChIP-seq, normalize to input and account for cell cycle differences in chromatin accessibility

    • For expression analyses, consider cell cycle-dependent reference genes

  • Biological versus technical variation:

    • Use biological replicates to distinguish natural heterogeneity from technical artifacts

    • Apply appropriate statistical methods for heterogeneous cell populations

Research has demonstrated that E2F6 accumulates during G1, peaks at G1/S transition, and then associates with specific target genes during S phase . This dynamic regulation must be considered when interpreting E2F6 antibody results from heterogeneous or asynchronous cell populations.

How can novel antibody-based approaches advance our understanding of E2F6's role in the initiation versus maintenance of epigenetic silencing?

Innovative antibody applications can address this fundamental question:

  • Temporal knockout systems paired with ChIP-seq:

    • Use inducible E2F6 knockout/knockdown systems at different developmental timepoints

    • Perform ChIP-seq for associated chromatin modifications before and after E2F6 depletion

    • Track persistence of epigenetic marks following E2F6 removal

  • Proximity-based labeling with E2F6 antibodies:

    • Develop BioID or APEX2 fusion proteins with E2F6

    • Identify phase-specific protein interactions during initiation versus maintenance phases

    • Correlate interaction partners with silencing functionality

  • Cell type-specific ChIP approaches:

    • Use genetic tools for cell type-specific E2F6 tagging in vivo

    • Perform ChIP-seq in developmentally distinct cellular populations

    • Compare binding patterns across developmental stages

Research has demonstrated that E2F6 is required to initiate epigenetic silencing in early embryonic cells but becomes dispensable for maintenance in differentiated cells . These approaches would further elucidate the molecular mechanisms behind this transition.

What methodological considerations are important when studying E2F6's dual roles in cell cycle regulation and apoptosis using antibody-based techniques?

E2F6 exhibits context-dependent functions in both cell cycle regulation and apoptosis resistance. To effectively study these dual roles:

  • Stress condition optimization:

    • Standardize UV exposure protocols (wavelength, dose, recovery time) when studying E2F6's role in UV-induced apoptosis

    • Compare multiple stress inducers (DNA damage, replication stress, metabolic stress) to identify condition-specific E2F6 functions

  • Target protein selection:

    • Monitor both cell cycle targets (CDC6, RR1, p68) and apoptosis-related targets (BRCA1)

    • Develop multiplex detection systems for simultaneous monitoring of multiple targets

  • Post-translational modification analysis:

    • Use phospho-specific antibodies to track E2F6 modifications under different cellular conditions

    • Combine with mass spectrometry to identify stress-induced modifications

  • Temporal resolution approaches:

    • Implement time-resolved ChIP following stress induction

    • Track real-time protein interactions using proximity ligation assays with E2F6 antibodies

Research has shown that E2F6 can inhibit UV-induced apoptosis by preventing BRCA1 expression and cleavage while also playing critical roles in cell cycle regulation . These methodological considerations will help dissect the molecular mechanisms underlying these seemingly distinct functions.

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.