Phospho-E2F1 (T433) Antibody

Shipped with Ice Packs
In Stock

Description

E2F1 Protein Overview

E2F1 is a transcription factor regulating genes involved in cell cycle progression (e.g., CCNE1, CDC6) and apoptosis (e.g., p73, Apaf1) . Its activity is modulated by phosphorylation at multiple sites, including T433, which may influence interactions with regulatory partners like TOPBP1 or chromatin remodeling factors .

Phosphorylation at T433

  • Role in DNA Damage Response: While phosphorylation at Ser31/Ser364 by ATM/ATR stabilizes E2F1 during DNA damage , T433 phosphorylation’s functional significance remains less characterized.

  • Detection Utility: The antibody specifically recognizes T433-phosphorylated E2F1, enabling studies on stress-induced signaling pathways .

Western Blot (WB)

  • Protocol: Used at 1:500 dilution in HeLa cell extracts treated with DNA-damaging agents like etoposide .

  • Key Observation: A 47 kDa band corresponding to phosphorylated E2F1 is detectable in damage-induced samples (Fig. 1A) .

Immunohistochemistry (IHC-P)

  • Localization: Nuclear staining in human pancreas tissue sections, consistent with E2F1’s role in transcriptional regulation .

  • Controls: Specificity confirmed using isotype-matched antibodies and peptide competition .

DNA Damage-Induced Phosphorylation

  • Etoposide Treatment: T433 phosphorylation increases in HeLa cells exposed to etoposide, correlating with E2F1’s activation in genotoxic stress .

  • Kinase Associations: While ATM/ATR phosphorylate E2F1 at Ser31 , the kinase responsible for T433 modification remains unidentified.

Functional Implications

  • Transcriptional Regulation: Phosphorylated E2F1 may alter DNA-binding affinity or cofactor recruitment, modulating target gene expression (e.g., RRP1B) .

  • Nontranscriptional Roles: E2F1 localizes to DNA damage foci independent of its DNA-binding domain, suggesting phosphorylation-dependent recruitment to repair sites .

Comparative Data

StudyPhosphorylation SiteFunctional OutcomeMethod Used
Blattner et al. (1999)Ser31Stabilization via ATM/ATR; apoptosis inductionRadioactive labeling
Abcam (ab55325)T433Detection in DNA damage modelsWB, IHC-P
Stevens & La Thangue (2003)Ser364CHK2-mediated stabilizationKinase assays

Limitations and Future Directions

  • Uncharacterized Kinase: The upstream kinase targeting T433 requires identification.

  • Pathological Relevance: Whether T433 phosphorylation influences cancer progression or therapy resistance remains unexplored.

Product Specs

Buffer
Liquid in PBS containing 50% glycerol, 0.5% BSA and 0.02% sodium azide.
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method or location. For specific delivery times, please consult your local distributor.
Synonyms
Dmel\CG6376 antibody; Dmel_CG6376 antibody; drosE2F1 antibody; E(Sev-CycE)3A antibody; E(var)3-93E antibody; E2-promoter binding facto antibody; E2F 1 antibody; E2F transcription factor 1 antibody; E2F-1 antibody; E2f-PA antibody; E2f-PB antibody; E2f-PC antibody; E2F1 antibody; E2f1 E2F transcription factor 1 antibody; E2F1_HUMAN antibody; Evar(3)164 antibody; KIAA4009 antibody; l(3)07172 antibody; l(3)j3B1 antibody; l(3)j3C2 antibody; l(3)rM729 antibody; mKIAA4009 antibody; OTTHUMP00000030661 antibody; PBR3 antibody; PRB binding protein E2F 1 antibody; PRB-binding protein E2F-1 antibody; RBAP 1 antibody; RBAP-1 antibody; RBAP1 antibody; RBBP-3 antibody; RBBP3 antibody; RBP 3 antibody; RBP3 antibody; Retinoblastoma-associated protein 1 antibody; Retinoblastoma-binding protein 3 antibody; Transcription factor E2F1 antibody
Target Names
Uniprot No.

Target Background

Function
E2F1, a transcription activator, cooperatively binds to DNA with DP proteins through the E2 recognition site (5'-TTTC[CG]CGC-3'). This site is found in the promoter regions of various genes involved in cell cycle regulation and DNA replication. The DRTF1/E2F complex plays a crucial role in controlling the transition from the G1 to S phase of the cell cycle. E2F1 exhibits a cell cycle-dependent preference for binding to RB1. It can mediate both cell proliferation and TP53/p53-dependent apoptosis. E2F1 inhibits adipocyte differentiation by binding to specific promoters, thereby repressing the binding of CEBPA to its target gene promoters. Furthermore, E2F1 positively regulates the transcription of RRP1B.
Gene References Into Functions
  1. XPC, an RNA polymerase II cofactor, recruits the ATAC coactivator complex to promoters by interacting with E2F1. PMID: 29973595
  2. Research indicates that the rs3213173 (C/T) and rs3213176 (G/A) polymorphisms in the E2F1 gene are genetic risk factors for susceptibility to lung cancer and Head and Neck cancer in the North Indian Population. PMID: 30036075
  3. High E2F1 expression is associated with the progression of hepatocellular carcinoma. PMID: 30106440
  4. The expression of miR175p has been shown to inhibit high glucose-induced endothelial cell injury by targeting E2F1. PMID: 29786752
  5. The nuclear transcription factor Y subunit beta (NFYB)-E2F transcription factor 1 (E2F1) pathway plays a crucial role in the chemoresistance of oxaliplatin-resistant colorectal cancer (OR-CRC) by inducing the expression and activation of checkpoint kinase 1 (CHK1), suggesting a potential therapeutic target for oxaliplatin resistance in CRC. PMID: 29203250
  6. If, as expected, the consequences of deregulation of the CDKN1C-E2F1-TP53 axis were the same as those experimentally demonstrated in mouse models, disrupting this axis might be useful for predicting tumor aggressiveness and providing the basis for developing potential therapeutic strategies in human Precursor T-cell lymphoblastic lymphomas. PMID: 29661169
  7. Lapatinib and cytotoxic agents have an antitumor effect by suppressing E2F1 in HER2-positive breast cancer. PMID: 29845287
  8. Data indicate that mRNA translation stress induces E2F transcription factor 1 (E2F1) via PI3-kinase p110 subunit delta (PI3Kdelta). PMID: 29235459
  9. PPM1B plays a negative role in the activation of the p38-RB1-E2F1 pathway, and targeting PPM1B could be beneficial in certain types of cancer by stimulating chemotherapy-induced cell death. PMID: 29654756
  10. Studies have examined the impact of BET proteins and E2F transcription factor 1 (E2F1) on neoplastic genetic transcription in glioblastoma. PMID: 29764999
  11. E2F1 knockdown decreased the expression of discoidin domain receptor 1 (DDR1), which plays a crucial role in various fundamental processes, including cell differentiation, adhesion, migration, and invasion. PMID: 29039472
  12. Findings suggest that downregulation of E2F1 may be a key factor in the celastrol-mediated inhibitory effects in HepG2 cells, and celastrol could serve as a lead compound for developing compounds designed to inactivate E2F1 for hepatocellular carcinoma therapy. PMID: 29048668
  13. SNHG16 promotes glioma tumorigenesis by sponging miR-20a-5p, leading to the enhancement of its endogenous targets, including E2F1. PMID: 29685003
  14. Results demonstrate that gambogic acid sensitizes pancreatic cancer cells to gemcitabine in vitro and in vivo by inhibiting the activation of the ERK/E2F1/RRM2 signaling pathway. PMID: 28797284
  15. Reports have shown that high expression of the transcription factor E2F1 is involved in the invasion and metastasis of small cell lung cancer (SCLC). Further results provide evidence that E2F1 promotes EMT by regulating ZEB2 gene expression in SCLC. PMID: 29115924
  16. In HeLa cells, Dox induced apoptosis through upregulation of endogenous E2F1 involving post-transcriptional mechanisms, while E2F6 was downregulated with induction of the Checkpoint kinase-1 and proteasome degradation. These data imply that E2F6 serves to modulate E2F activity and protect cells, including cardiomyocytes, from apoptosis and improve survival. PMID: 28964969
  17. This study reveals a molecular pathway involving lncRNA GAS5/E2F1/P27(Kip1) that regulates cell proliferation and could be a potential therapeutic target in prostate cancer. PMID: 28396462
  18. E2F1 induces TINCR transcriptional activity and accelerates gastric cancer progression via activation of the TINCR/STAU1/CDKN2B signaling axis. PMID: 28569791
  19. Data show that lncRNA-HIT acts as an oncogene through association with E2F transcription factor 1 protein (E2F1). PMID: 28429752
  20. Cell proliferation and apoptosis were almost completely abolished in the PAa cells cotreated with TRIM28 siRNA and etoposide following knockdown of E2F1. The results of this study demonstrated that the combination of TRIM28 siRNA and etoposide may be effective against non-small cell lung cancer (NSCLC) and has the potential to be a new therapeutic tool for future treatment. PMID: 28498400
  21. These results reveal an additional level of regulation of the stability and activity of E2F1 by a non-degradative K63-poly-ubiquitination and uncover a novel function for the E3-ubiquitin ligase cIAP1. PMID: 28542143
  22. Higher levels of miR-135a in gastric cancer (GC) are associated with shorter survival times and reduced times to disease recurrence. The mechanism whereby miR-135a promotes GC pathogenesis appears to be the suppression of E2F1 expression. PMID: 27683111
  23. Results provide mechanistic insight into a series of complex, differentiation-specific molecular mechanisms that regulate E2F1 during keratinocyte maturation via multiple events. These events include nucleocytoplasmic transport and changes in ubiquitinylation patterns specifically orchestrated through S403 and T433, and which differ from other mechanisms that regulate E2F1 turnover in undifferentiated cells. PMID: 27903963
  24. These results suggest that the E2F1/miR19a/PPARalpha feedback loop is critical for glioma progression. PMID: 27835866
  25. This study suggests for the first time an involvement of E2F1 copy number variations in testicular germ cell tumor susceptibility and supports previous preliminary data on the importance of AKT/mTOR signaling pathway in this cancer. PMID: 28104681
  26. High E2F1 expression is associated with gastric cancer. PMID: 27036039
  27. Data suggest that the protein arginine methyltransferase 5 (PRMT5)-E2F1 transcription factor (E2F-1) pathway may act as a common target for exogenous lectins, including Anguilla japonica lectin 1 (AJL1), and the cellular response to exogenous AJL1 may suggest a novel agent for cancer gene therapy. PMID: 26990556
  28. Low levels of E2F1 are sufficient to induce numerous cell cycle-promoting genes, intermediate levels induce growth arrest genes (e.g., p18, p19, and p27), whereas higher levels are necessary to induce key apoptotic E2F1 targets, including APAF1, PUMA, HRK, and BIM. PMID: 28211871
  29. This review focuses on the relationship between E2F1, growth factors, and cytokines. PMID: 26947516
  30. Research indicates that E2F1 mRNA stability and E2F1 protein levels are reduced in cells lacking RALY expression. PMID: 28972179
  31. Results indicate that E2F1 is an important downstream gene of ISX in hepatoma progression. PMID: 27175585
  32. Studies have found that human E2F1 competes with YAP for TEAD1 binding, affecting YAP activity, indicating that this mode of cross-regulation is conserved. PMID: 29207260
  33. E2F1-mediated hPOMC transcription is a potential target for suppressing ACTH production in ectopic Cushing's syndrome. PMID: 27935805
  34. Results demonstrate that E7 recruited CUL2, driven by the CUL2/E2F1/miR-424 regulatory loop, is overexpressed and accelerates HPV16-induced cervical carcinogenesis. PMID: 27153550
  35. Results demonstrated that E2F-1 mediated PEG10 overexpression promotes pancreatic cancer cell proliferation via accelerating cell cycle progression and increases migration and invasion through the ERK/MMP7 pathway. PMID: 28193232
  36. Researchers conclude that high expression of S18-2 and free E2F...1 might be a good set of prognostic markers for endometrial cancer. PMID: 26959119
  37. COMMD9 participates in TFDP1/E2F1 activation and plays a critical role in non-small cell lung cancer. PMID: 27871936
  38. Using iterative experimental and computational analyses, researchers have shown physical and functional interactions between NF-kappaB and the E2 Factor 1 (E2F-1) and E2 Factor 4 (E2F-4) cell cycle regulators. PMID: 27185527
  39. Heavy ion irradiation could induce p53(-/-) hepatoma cells to undergo apoptosis via the E2F1/Bax/Casp3 signaling pathway. PMID: 28500630
  40. Researchers propose that E2F1 interacts with the BRCA1 indirect pathway to induce two different small molecule metabolic pathways or cell cycle regulation pathways in hepatocellular carcinoma. PMID: 28474358
  41. The pyruvate dehydrogenase kinases (PDKs) PDK1 and PDK3 are direct targets of KDM4A and E2F1 and modulate the switch between glycolytic metabolism and mitochondrial oxidation. PMID: 27626669
  42. Taken together, this study reveals evidence demonstrating a mechanism by which the LPR6/ GSK3beta/E2F1 axis-upregulated LSH promoted gliomas. PMID: 28042322
  43. High E2F1 expression is associated with melanoma. PMID: 28068326
  44. p63alpha protein up-regulates heat shock protein 70 expression via E2F1 transcription factor 1, promoting Wasf3/Wave3/MMP9 signaling and bladder cancer invasion. PMID: 28794159
  45. E2F1 couples immune cell development to the immune response. E2f1 plays a role in inflammation-associated cancers [review]. PMID: 26881929
  46. These data suggest a model in which cells experiencing oncogene-induced replication stress through deregulation of E2F-dependent transcription. PMID: 27160911
  47. Researchers describe a regulatory loop miR-218-CDK6/CyclinD1-E2F1 whose disruption may contribute to cell cycle progression in gastric cancer. PMID: 28634044
  48. Results demonstrated that somatic mutation within the E2F1:MIR136-5p target site impairs miRNA-mediated regulation and leads to increased gene activity. PMID: 28704519
  49. Specific E2Fs also have prognostic value in breast cancer, independent of clinical parameters. This section discusses recent advances in understanding the RB-E2F pathway in breast cancer. It also discusses the application of genome-wide genetic screening efforts to gain insight into synthetic lethal interactions of CDK4/6 inhibitors in breast cancer for the development of more effective combination therapies. PMID: 26923330
  50. Results provide evidence that the E2F1rs3213180 polymorphism may influence susceptibility to HPV-associated oral squamous cell carcinoma, particularly oropharynx tumors. PMID: 27677255

Show More

Hide All

Database Links

HGNC: 3113

OMIM: 189971

KEGG: hsa:1869

STRING: 9606.ENSP00000345571

UniGene: Hs.654393

Protein Families
E2F/DP family
Subcellular Location
Nucleus.

Q&A

What is the biological significance of E2F1 phosphorylation at Threonine 433?

E2F1 phosphorylation at Threonine 433 plays a critical role in regulating this transcription factor's stability and activity. E2F1 functions as a transcription activator that binds DNA cooperatively with DP proteins through the E2 recognition site (5'-TTTC[CG]CGC-3') found in promoter regions of genes involved in cell cycle regulation and DNA replication . The phosphorylation of T433 specifically contributes to:

  • Regulation of E2F1 protein turnover, particularly during cellular differentiation processes

  • Modulation of E2F1's subcellular localization

  • Alteration of E2F1's interaction with ubiquitination machinery

Research has shown that T433 phosphorylation, often in conjunction with S403 phosphorylation, is involved in differentiation-specific degradation pathways that differ from those activated during DNA damage responses . In keratinocytes, for example, phosphorylation at both S403 and T433 appears necessary for proper E2F1 turnover during differentiation, but not in undifferentiated cells .

What detection methods are most suitable for analyzing Phospho-E2F1 (T433) in experimental systems?

Multiple detection methods have been validated for Phospho-E2F1 (T433) analysis, with varying applications across experimental systems:

MethodApplicationsAdvantagesConsiderations
Western BlottingProtein expression level detectionQuantifiable, size verificationRequires careful optimization of lysis conditions to preserve phosphorylation
Immunohistochemistry (IHC)Tissue localizationSpatial context within intact tissueTypically requires 1:50-1:200 dilution of antibody
Immunofluorescence (IF/ICC)Subcellular localizationHigh-resolution imagingFixation method critical for phospho-epitope preservation
Cell-Based ELISAQuantitative high-throughput screeningScalable, conserves reagentsRequires careful normalization to cell number

For optimal results when detecting phosphorylated E2F1, researchers should consider supplementing antibody-based detection with phosphatase treatment controls to confirm specificity of phosphorylation-dependent signals .

How can researchers validate the specificity of Phospho-E2F1 (T433) antibodies in their experimental system?

Validating antibody specificity for phosphorylation-specific epitopes requires a multi-faceted approach:

  • Phosphatase treatment control: Treat one sample with lambda phosphatase before immunoblotting to confirm the phospho-specific nature of the signal. Research has shown that treatment with λ phosphatase causes E2F1 to collapse into a single band on denaturing polyacrylamide gels, confirming that the mobility shift is due to phosphorylation .

  • Phospho-null mutant comparison: Compare detection between wild-type E2F1 and a T433A mutant (where threonine is replaced with non-phosphorylatable alanine). The phospho-specific antibody should not detect the T433A mutant .

  • Phospho-mimetic comparison: A T433D mutant (where threonine is replaced with aspartic acid to mimic phosphorylation) can provide additional validation of phosphorylation-dependent effects .

  • Stimulation conditions: Apply conditions known to induce or reduce T433 phosphorylation, such as differentiation cues in keratinocytes, and confirm expected changes in signal intensity .

  • Molecular weight verification: Confirm that the detected band appears at approximately 47 kDa, which is the expected molecular weight of E2F1 .

What is the difference between phospho-specific and total E2F1 antibodies in research applications?

Understanding the distinction between phospho-specific and total E2F1 antibodies is crucial for experimental design and data interpretation:

For comprehensive analysis of E2F1 regulation, researchers should consider using both phospho-specific and total E2F1 antibodies in parallel. This approach enables calculation of the proportion of E2F1 that is phosphorylated at T433 relative to the total E2F1 pool, providing insight into the activation state of relevant signaling pathways .

How does T433 phosphorylation regulate E2F1 protein stability and degradation during cellular differentiation?

T433 phosphorylation plays a critical role in regulating E2F1 stability specifically during differentiation processes. Research on keratinocytes has provided detailed insights into this mechanism:

This phosphorylation-dependent regulation represents a specialized mechanism for controlling E2F1 levels during cellular differentiation processes, which is separate from other regulatory pathways affecting this protein.

What techniques are most effective for analyzing dynamic changes in E2F1 T433 phosphorylation in response to cellular stimuli?

Analyzing dynamic phosphorylation changes requires specialized techniques that capture temporal aspects of modification:

  • Time-course immunoblotting with quantification:

    • Collect samples at multiple time points after stimulus application

    • Use phospho-T433-specific antibody alongside total E2F1 antibody

    • Employ digital imaging and quantification software to calculate phospho/total ratios

    • Consider normalization to housekeeping proteins like GAPDH or β-actin

  • Cell-Based Colorimetric ELISA:

    • Enables high-throughput analysis of phosphorylation changes

    • Can be performed in 96-well microplates for multiple timepoints/conditions

    • Uses target-specific primary antibody and HRP-conjugated secondary antibody

    • Crystal violet staining serves as normalization for cell number

  • Phospho-proteomic mass spectrometry:

    • Provides comprehensive, unbiased assessment of phosphorylation

    • Can detect multiple phosphorylation sites simultaneously

    • Requires specialized equipment and expertise

    • Consider enrichment strategies for phosphopeptides

  • Pulse-chase analysis for phosphorylation turnover:

    • Particularly useful for determining phosphorylation half-life

    • Can be combined with phosphatase inhibitor treatment to assess phosphorylation persistence

    • Enables calculation of apparent half-life (t½) of phosphorylated species

  • Microscopy-based approaches:

    • Immunofluorescence with phospho-specific antibodies

    • Live-cell imaging with phospho-sensors (if available)

    • Provides spatial information about phosphorylation events

    • Can reveal subcellular relocalization following phosphorylation

For optimal results, researchers should consider employing multiple complementary techniques to validate findings across different methodological approaches.

How do T433 and S403 phosphorylation sites on E2F1 cooperate in regulating protein function during differentiation?

The cooperative action of T433 and S403 phosphorylation represents a sophisticated regulatory mechanism for E2F1 function during differentiation:

  • Cooperative effects on protein stability:

    • Individual phosphorylation at either site is insufficient to trigger complete degradation

    • Double phosphorylation at both S403 and T433 appears necessary for full degradation during differentiation

    • The S403A/T433A double mutant shows dramatically enhanced stability (t½>240 min) compared to wild-type or single-site mutants in differentiated cells

  • Differential impacts on ubiquitination patterns:

    • Both sites together influence K11 and K48 ubiquitin linkage formation

    • The absence of phosphorylation at both sites (S403A/T433A) significantly reduces K11 and K48 linkages while preserving K63 linkages

    • The pseudophosphorylated mutant (S403D/T433D) maintains ubiquitylation patterns similar to wild-type E2F1

  • Protein conformation and complex formation:

    • Phosphorylation at both sites likely induces conformational changes

    • These changes may affect E2F1's interaction with the APC/C^Cdh1 complex

    • Proper positioning within ubiquitin ligase complexes appears dependent on phosphorylation status

  • Subcellular localization effects:

    • Research in keratinocytes shows that pseudophosphorylation at S403, T433, or both produces proteins that localize primarily to the nucleus in undifferentiated cells

    • Upon differentiation, these proteins redistribute to the cytoplasm similarly to wild-type E2F1

This coordinated phosphorylation system illustrates the complexity of post-translational modifications in fine-tuning transcription factor activity during cellular differentiation processes.

What are the methodological considerations for distinguishing between phosphorylation-dependent and phosphorylation-independent functions of E2F1?

Distinguishing between these functional modes requires carefully designed experimental approaches:

  • Site-directed mutagenesis systems:

    • Generate phospho-null mutants (T433A) to abolish phosphorylation

    • Create phospho-mimetic mutants (T433D) to simulate constitutive phosphorylation

    • Employ site-specific mutations at individual and combined sites to parse their contributions

    • Use retroviral transduction systems in E2F1-null backgrounds for clean functional analysis

  • Domain-specific functional assays:

    • DNA binding assays to assess impact on transcriptional activity

    • Co-immunoprecipitation to evaluate protein-protein interactions

    • Chromatin immunoprecipitation (ChIP) to measure promoter occupancy

    • Cell cycle analysis to determine effects on proliferation control

  • Comparative phosphorylation analysis:

    • Implement parallel assays with wild-type and mutant proteins

    • Use λ phosphatase treatment to remove all phosphorylations

    • Employ mobility shift analysis on denaturing gels to assess phosphorylation status

    • Consider 2D gel electrophoresis to separate different phospho-forms

  • Temporal separation of functions:

    • Design experiments that exploit different temporal windows of E2F1 activity

    • Compare early (often phospho-independent) versus late (potentially phospho-dependent) functions

    • Use synchronized cell populations to align cell cycle stages

  • Stimulus-specific phosphorylation triggers:

    • Compare DNA damage-induced versus differentiation-induced phosphorylation

    • Utilize specific kinase activators or inhibitors to manipulate phosphorylation

    • Research shows that etoposide treatment increases abundance of wild-type and S403A/T433A E2F1, indicating that these sites are not required for DNA damage-induced stabilization

These methodologies allow researchers to dissect the complex relationship between phosphorylation status and functional outcomes for E2F1 in different cellular contexts.

How does the phosphorylation of E2F1 at T433 influence its interactions with cell cycle regulatory proteins?

E2F1 phosphorylation at T433 modulates its interactions with key cell cycle regulatory proteins through several mechanisms:

  • Interaction with pocket proteins:

    • E2F1 normally binds preferentially to retinoblastoma protein (RB1) in a cell-cycle dependent manner

    • T433 phosphorylation may alter the strength or dynamics of this interaction

    • The DRTF1/E2F complex functions in controlling cell-cycle progression from G1 to S phase

  • Association with ubiquitination machinery:

    • Phosphorylation at T433 affects E2F1's interaction with the APC/C^Cdh1 ubiquitin ligase complex

    • Both wild-type and S403A/T433A E2F1 bind to Cdh1, but their ubiquitination patterns differ significantly

    • This suggests that phosphorylation alters how E2F1 is positioned within the complex rather than preventing binding entirely

  • Impact on dimer formation:

    • E2F1 functions as a heterodimer with DP proteins

    • Research indicates that physiological E2F is a heterodimer composed of an E2F subunit together with a DP subunit

    • T433 phosphorylation may influence the stability or transcriptional activity of these heterodimers

  • Regulation by kinase signaling pathways:

    • E2F1 is regulated by PI3-kinase-like kinases such as ATM/ATR

    • These stress-responsive pathways may trigger T433 phosphorylation under specific conditions

    • Understanding the kinases responsible for T433 phosphorylation provides insight into upstream regulatory pathways

The dynamic phosphorylation of T433 creates a molecular switch that helps determine whether E2F1 promotes cell proliferation or apoptosis, depending on cellular context and the presence of additional regulatory signals.

What are common technical challenges when working with Phospho-E2F1 (T433) antibodies and how can they be addressed?

Researchers frequently encounter several challenges when working with phospho-specific antibodies:

ChallengeCauseSolution
Weak or absent signalPhosphorylation loss during sample preparationInclude phosphatase inhibitors in all buffers; keep samples cold; use fresh reagents
High backgroundNon-specific bindingOptimize blocking conditions; increase antibody dilution (1:500-1:2000 for WB ); use alternative blocking agents
Multiple bandsCross-reactivity or degradationVerify with phospho-null mutant controls; use freshly prepared samples; add protease inhibitors
Inconsistent resultsVariable phosphorylation levelsStandardize cell culture conditions; synchronize cells if possible; validate stimulation protocols
Poor reproducibilityTechnical variabilityStandardize lysate preparation; use consistent antibody lots; implement quantitative controls

Additional optimization strategies:

  • Sample preparation optimization:

    • For Western blotting, 1 mg/ml in Phosphate buffered saline (PBS) with 0.05% sodium azide (approx. pH 7.2) is recommended

    • Consider using specialized phospho-protein extraction buffers

    • Maintain cold chain throughout sample processing

  • Antibody validation:

    • Verify specificity with phospho-null mutants (T433A)

    • Conduct phosphatase treatment controls

    • Confirm expected molecular weight (~47 kDa)

  • Application-specific considerations:

    • For IHC applications, dilutions of 1:50-1:200 are typically recommended

    • For Western blotting, dilutions of 1:500-1:2000 are often optimal

    • For ELISA applications, higher dilutions (1:5000) may be appropriate

How can researchers optimize detection of low-abundance phosphorylated E2F1 in complex biological samples?

Detecting low-abundance phosphorylated proteins requires specialized approaches:

  • Enrichment strategies:

    • Immunoprecipitation with total E2F1 antibody before probing with phospho-specific antibody

    • Phospho-protein enrichment columns prior to standard detection methods

    • Subcellular fractionation to concentrate nuclear proteins where E2F1 is predominantly located

  • Signal amplification techniques:

    • Enhanced chemiluminescence (ECL) systems with extended exposure times

    • Tyramide signal amplification for immunohistochemistry

    • Biotin-streptavidin amplification systems

    • Consider highly sensitive detection reagents for low-abundance targets

  • Sample optimization:

    • Increase starting material quantity

    • Reduce sample dilution during processing

    • Use phosphatase inhibitor cocktails at maximum recommended concentrations

    • Process samples rapidly at cold temperatures to preserve phosphorylation

  • Technical considerations:

    • Utilize high-sensitivity imaging systems with extended dynamic range

    • Consider alternate membrane types (PVDF vs. nitrocellulose) for Western blotting

    • Optimize transfer conditions to ensure complete protein transfer

    • Use freshly prepared antibody dilutions for each experiment

  • Alternative detection methods:

    • Consider Cell-Based Colorimetric ELISA which can be more sensitive than Western blotting

    • Explore immunofluorescence with tyramide signal amplification

    • Investigate proximity ligation assay (PLA) for detecting protein-protein interactions involving phosphorylated E2F1

These approaches can significantly improve detection of low-abundance phosphorylated E2F1 in complex biological samples.

How does E2F1 T433 phosphorylation contribute to cancer development and progression?

E2F1 phosphorylation at T433 may play complex roles in cancer biology, with implications for both tumor suppression and oncogenesis:

  • Dysregulated phosphorylation in cancer cells:

    • Altered phosphorylation patterns may contribute to E2F1's dual role in both promoting cell proliferation and inducing apoptosis

    • Cancer cells might exploit phosphorylation-dependent regulation to favor proliferative over apoptotic functions

  • Cell cycle regulatory impact:

    • E2F1 is essential for regulation of the cell cycle, particularly the G1 to S phase transition

    • Altered T433 phosphorylation could disrupt normal cell cycle control, contributing to unrestrained proliferation

    • The E2F family plays a crucial role in the control of cell cycle and action of tumor suppressor proteins

  • Interaction with tumor suppressor pathways:

    • E2F1 can induce both p53-dependent and p53-independent apoptosis

    • T433 phosphorylation may influence whether E2F1 activates pro-apoptotic target genes like TA-p73 and ARF

    • Phosphorylation could affect E2F1's interaction with retinoblastoma protein (RB1), a critical tumor suppressor

  • Colon cancer implications:

    • Research suggests specific roles for E2F1 in colon cancer development

    • Pharmacological molecules that influence E2F1 expression may have therapeutic potential in colon cancer

    • Understanding phosphorylation-specific functions could lead to more targeted therapeutic approaches

  • Cancer-specific phosphorylation patterns:

    • Different cancer types may exhibit distinct patterns of E2F1 phosphorylation

    • These patterns could serve as potential biomarkers or therapeutic targets

    • Phospho-specific antibodies may help characterize these patterns in patient samples

Further research into the cancer-specific roles of T433 phosphorylation could yield new diagnostic and therapeutic approaches based on this post-translational modification.

What novel methodologies are being developed to study the dynamics of E2F1 phosphorylation in live cells?

Several cutting-edge approaches are advancing our ability to study phosphorylation dynamics in real-time:

  • Phospho-specific fluorescent biosensors:

    • Genetically encoded FRET-based sensors designed to detect specific phosphorylation events

    • These could allow real-time visualization of E2F1 phosphorylation in living cells

    • Enable correlation of phosphorylation events with cellular processes like cell cycle progression

  • Optogenetic control of kinase activity:

    • Light-controlled activation of kinases that target E2F1

    • Permits precise temporal control over phosphorylation events

    • Allows determination of immediate versus delayed effects of phosphorylation

  • High-content imaging platforms:

    • Automated microscopy systems for tracking phosphorylation in living cells

    • Machine learning algorithms to identify subtle changes in protein localization or interactions

    • Integration with other cellular markers to contextualize phosphorylation events

  • CRISPR-based endogenous tagging:

    • Knock-in of fluorescent tags at the endogenous E2F1 locus

    • Combined with phospho-specific antibodies for immunofluorescence

    • Provides physiologically relevant expression levels for more accurate assessment

  • Single-cell phosphoproteomic approaches:

    • Mass cytometry (CyTOF) with phospho-specific antibodies

    • Single-cell Western blotting techniques

    • Spatial proteomics approaches to map phosphorylation events within cellular compartments

These emerging technologies promise to provide unprecedented insights into the spatial and temporal dynamics of E2F1 phosphorylation in cellular contexts.

Which experimental controls are essential when using Phospho-E2F1 (T433) antibodies in various applications?

Control TypeDescriptionApplicationRationale
Phosphatase treatmentTreat sample with λ phosphataseWB, IHC, IFConfirms signal is phosphorylation-dependent
Phospho-null mutantUse E2F1 T433A expression constructWB, IP, IFValidates antibody specificity for phosphorylated form
Phospho-mimeticUse E2F1 T433D expression constructFunctional studiesMimics constitutive phosphorylation for comparison
Blocking peptidePre-incubate antibody with immunizing phosphopeptideWB, IHC, IFDemonstrates epitope-specific binding
Stimulation controlsTreat cells with conditions known to modulate phosphorylationAll applicationsConfirms biological responsiveness
Total E2F1 detectionParallel detection with antibody to total E2F1WB, IHC, IFDistinguishes between changes in phosphorylation vs. total protein
Loading/transfection controlsHousekeeping proteins or co-transfection markersWB, functional assaysEnsures equal loading or transfection efficiency
Non-specific IgGSame species IgG at equivalent concentrationIHC, IF, IPControls for non-specific binding

Implementation of these controls ensures reliable and interpretable results when working with phospho-specific antibodies in research applications.

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.