DOF4.2 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of Antibodies Targeting Transcription Factors

Antibodies targeting transcription factors like DOF (DNA-binding One Zinc Finger) proteins are critical tools for studying gene regulation. While no direct reference to "DOF4.2 Antibody" exists, antibodies against DOF homologs or analogous transcription factors are characterized by their specificity for conserved DNA-binding domains. For example:

  • PCRP-E2F4-2H10 (DSHB Hybridoma Product) targets human transcription factor E2F4, a protein with roles in cell cycle regulation .

  • Anti-CD132 monoclonal antibody 2D4 blocks IL-21/IL-15 signaling by targeting a shared cytokine receptor subunit .

These examples highlight the importance of epitope specificity and functional validation in antibody development.

DOF Transcription Factor Family Context

DOF proteins are plant-specific transcription factors characterized by a single zinc finger domain. Key structural features include:

  • A conserved Cys2/Cys2 zinc finger domain recognizing AAAG DNA motifs .

  • Regulatory roles in plant development, stress responses, and metabolic pathways .

While DOF4.2 is not explicitly mentioned, studies on DOF homologs (e.g., AtDOF4 in Arabidopsis) suggest potential functional parallels.

Antibody Characterization Data

The following table summarizes antibody properties from analogous studies:

AntibodyTargetHost SpeciesApplicationsBinding AffinitySource
PCRP-E2F4-2H10 E2F4 (Human)MouseWB, IP, ELISAHigh (0.2–5 µg/mL)DSHB
2D4 CD132 (Human)HumanizedImmunotherapyIC₅₀: ≤10 nMNature
Anti-PF4 rAbs PF4 (Human)ChimericPlatelet activationKD: 1–10 nMBlood Journal

Research Findings on Antibody Function

  • Neutralization Mechanisms: Antibodies like 2D4 block cytokine signaling by sterically hindering receptor binding .

  • Epitope Specificity: Anti-PF4 antibodies from VITT patients target arginine-22 (R22) on PF4, enabling heparin-independent platelet activation .

  • Cross-Reactivity: Anti-DOF antibodies in plants show high specificity for conserved zinc finger domains, but cross-reactivity risks exist due to motif conservation .

Technical Considerations for Antibody Validation

  • Titration: Optimal concentrations vary by application (e.g., 0.2–0.5 µg/mL for WB) .

  • Binding Kinetics: Advanced methods like biolayer interferometry (BLI) quantify affinity (e.g., KD values) .

  • Neutralization Assays: Functional validation (e.g., platelet activation tests) confirms biological impact .

Gaps in Current Knowledge

No peer-reviewed studies explicitly describe "DOF4.2 Antibody." Potential explanations include:

  • Typographical errors (e.g., "E2F4.2" or "DOF4" miswritten as "DOF4.2").

  • Reference to an unpublished or proprietary antibody.

  • Species-specific nomenclature (e.g., DOF4.2 in non-model plants).

Recommendations for Further Research

  • Screen databases like the Developmental Studies Hybridoma Bank (DSHB) for DOF-related antibodies .

  • Validate cross-reactivity of existing anti-DOF antibodies (e.g., anti-AtDOF4) using immunoblotting .

  • Characterize hypothetical "DOF4.2 Antibody" via epitope mapping and functional assays .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
DOF4.2 antibody; At4g21030 antibody; T13K14.190Dof zinc finger protein DOF4.2 antibody; AtDOF4.2 antibody
Target Names
DOF4.2
Uniprot No.

Target Background

Function
DOF4.2 is a transcription factor that exhibits specific binding to the 5'-AA[AG]G-3' consensus core sequence.
Gene References Into Functions
  1. Research has indicated that overexpression of AtDOF4.2 promotes increased branching through the upregulation of three branching-related genes: AtSTM (SHOOT MERISTEMLESS), AtTFL1 (TERMINAL FLOWER1), and AtCYP83B1 (CYTOCHROME P450 83B1). PMID: 23095045
Database Links

KEGG: ath:AT4G21030

STRING: 3702.AT4G21030.1

UniGene: At.54449

Subcellular Location
Nucleus.

Q&A

What are monoclonal antibodies and how are they produced?

Monoclonal antibodies are laboratory-produced molecules engineered to serve as substitute antibodies that can restore, enhance, or mimic the immune system's attack on specific targets. They are typically produced through a process involving:

  • Immunization of an animal (often mice) with the target antigen

  • Collection of B cells that produce antibodies against the target

  • Fusion of these B cells with myeloma cells to create hybridomas

  • Selection and cultivation of hybridoma clones that produce the desired antibody

  • Humanization of the antibody (for therapeutic applications) to reduce immunogenicity

Most therapeutic monoclonal antibodies are developed on IgG1 platforms due to improved solubility, greater complement-fixation, low nonspecific immunity, and better immune effector cell receptor (FcγR)-binding efficiencies, which can play a crucial role in their therapeutic activity . Some antibodies use IgG4 platforms despite potential reduced efficacy due to Fab-arm exchange, as seen with Gemtuzumab ozogamicin and Inotuzumab ozogamicin .

How is antibody specificity determined and optimized?

Antibody specificity refers to the ability of an antibody to bind to its intended target antigen while avoiding cross-reactivity with other antigens. Specificity is determined through:

  • Direct binding assays (ELISA, BLI, SPR) against the target and potential cross-reactive antigens

  • Cell-based assays to confirm target engagement in a biological context

  • Tissue cross-reactivity studies to evaluate potential off-target binding

Optimization of antibody specificity can be achieved through:

  • Phage display selection against multiple ligands to identify binding modes associated with particular targets

  • Computational modeling to predict and design specific binding profiles

  • Directed evolution approaches with high-throughput screening

Recent advances combine biophysics-informed modeling with extensive selection experiments to design antibodies with desired physical properties and binding profiles . This approach allows researchers to generate antibodies with either specific high affinity for a particular target ligand or cross-specificity for multiple target ligands .

What validation methods should be used to confirm antibody functionality?

A comprehensive validation approach should include:

  • Biochemical validation:

    • ELISA to confirm binding to the target antigen

    • Western blotting to verify specific recognition of the target protein

    • Immunoprecipitation to demonstrate ability to pull down the target from complex mixtures

  • Cellular validation:

    • Flow cytometry to confirm binding to cell-surface targets

    • Immunocytochemistry to assess cellular localization

    • Functional assays to demonstrate biological activity (e.g., receptor blocking)

  • In vivo validation:

    • Target engagement studies in relevant animal models

    • Pharmacokinetic/pharmacodynamic (PK/PD) studies

    • Efficacy in disease models

For example, in a study of anti-CD132 monoclonal antibody 2D4, researchers validated its functionality by demonstrating its ability to block IL-21 and IL-15 with limited effectiveness against IL-2, showing suppression of T and B cells without disrupting immune tolerance . They further validated it in mouse immunization models and lupus murine models, confirming its ability to mitigate inflammation by suppressing pro-inflammatory cytokines and anti-dsDNA antibody titers .

How can computational approaches enhance antibody design and specificity?

Modern antibody engineering increasingly relies on computational methods to optimize binding specificity and physicochemical properties:

  • Machine learning for binding prediction:

    • Utilization of high-throughput sequencing data to identify binding patterns

    • Development of models that predict binding affinity based on sequence features

  • Structural modeling for rational design:

    • Homology modeling and molecular dynamics simulations to predict antibody-antigen interactions

    • In silico mutagenesis to identify key binding residues

  • Integrated experimental-computational approaches:

    • Combining phage display experiments with computational analysis

    • Iterative optimization based on experimental feedback

Recent research demonstrates the successful use of computational approaches to design antibodies with customized specificity profiles. This involves identifying different binding modes associated with particular ligands against which the antibodies are either selected or not . Using data from phage display experiments, researchers have shown that computational models can successfully disentangle these modes, even when they are associated with chemically very similar ligands .

These approaches allow for:

  • Design of antibodies with specific high affinity for particular target ligands

  • Creation of antibodies with cross-specificity for multiple target ligands

  • Mitigation of experimental artifacts and biases in selection experiments

What are the considerations for developing antibody-drug conjugates (ADCs) for therapeutic applications?

Antibody-drug conjugates (ADCs) combine the selectivity of monoclonal antibodies with the potency of cytotoxic drugs, requiring careful consideration of multiple factors:

  • Antibody selection:

    • Target antigen expression profile (tumor vs. normal tissue)

    • Internalization rate and efficiency

    • Binding affinity optimization

  • Linker chemistry:

    • Stability in circulation

    • Cleavable vs. non-cleavable linkers

    • Site-specific conjugation technologies

  • Payload selection:

    • Mechanism of action

    • Potency requirements

    • Bystander effect considerations

  • Formulation and manufacturing:

    • Drug-to-antibody ratio (DAR) optimization and control

    • Stability during storage and administration

    • Scalable manufacturing processes

ADCs are currently among the most promising drug classes in oncology, with ongoing efforts to expand their application to non-oncological indications and in combination therapies . Beyond cancer treatment, ADCs have shown potential as:

  • Antibody-antibiotic conjugates (AACs) for targeting intracellular bacteria like Staphylococcus aureus

  • Immunomodulatory agents delivering glucocorticoids to specific immune cells, as demonstrated with anti-TNF-α based ADCs for autoimmune diseases

These applications demonstrate the versatility of the ADC platform beyond traditional cancer therapies.

How should researchers address ARIA risks when developing anti-amyloid monoclonal antibodies?

Amyloid-related imaging abnormalities (ARIA) represent a significant safety concern with anti-amyloid monoclonal antibodies for Alzheimer's disease. Researchers should consider:

  • Patient stratification based on genetic risk factors:

    • APOE genotyping to identify high-risk patients, particularly those with two copies of the APOE-4 allele

    • Exclusion of highest-risk populations from treatment

  • Monitoring protocols:

    • Baseline and periodic MRI assessments

    • Standardized reporting and grading of ARIA findings

    • Clear management algorithms for ARIA detection

  • Dose optimization strategies:

    • Titration protocols to minimize rapid amyloid clearance

    • Evaluation of dosing frequency to balance efficacy and safety

The APOE-4 allele, especially when two copies are present, is associated with a form of cerebral amyloid angiopathy in which amyloid protein is present in the smooth muscle of small blood vessels, making them susceptible to leak or rupture when amyloid is removed by treatment . Clinical experience with anti-amyloid antibodies like aducanumab, lecanemab, and donanemab has shown that while most ARIA cases are asymptomatic or mild, approximately 1.5% of cases are severe, sometimes resulting in hospitalization or death . Seven cases of death associated with severe ARIA have been reported, all in patients with two copies of the APOE-4 allele .

This data suggests that the risk-benefit ratio may not support the use of anti-amyloid antibodies in patients with two copies of the APOE-4 allele .

What analytical methods are most effective for characterizing antibody heterogeneity?

Comprehensive characterization of antibody heterogeneity requires multiple complementary analytical approaches:

  • Primary structure analysis:

    • Peptide mapping with LC-MS/MS

    • Intact mass analysis

    • N- and C-terminal sequencing

  • Post-translational modification analysis:

    • Glycan profiling (HILIC, MS)

    • Charge variant analysis (cIEF, CEX)

    • Oxidation and deamidation assessment

  • Higher-order structure analysis:

    • Circular dichroism (CD)

    • Fourier-transform infrared spectroscopy (FTIR)

    • Differential scanning calorimetry (DSC)

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS)

  • Aggregation and particle analysis:

    • Size-exclusion chromatography (SEC)

    • Analytical ultracentrifugation (AUC)

    • Dynamic light scattering (DLS)

    • Flow imaging microscopy for subvisible particles

Each method provides unique insights into specific aspects of antibody heterogeneity, and a combination of techniques is typically required for comprehensive characterization.

How can researchers optimize antibody phage display experiments?

Phage display is a powerful technique for identifying novel antibodies with desired properties. To optimize these experiments:

  • Library design considerations:

    • Diversity (theoretical vs. actual)

    • Scaffold selection (scFv, Fab, nanobody)

    • Randomization strategy (CDR-focused vs. whole variable domain)

  • Selection strategy optimization:

    • Antigen presentation (purified vs. cell-surface)

    • Washing stringency

    • Elution conditions

    • Number of selection rounds

  • Counter-selection strategies:

    • Depletion against related antigens to improve specificity

    • Alternating selection between related antigens for cross-reactive antibodies

    • Differential selection conditions for customized binding profiles

  • Output analysis:

    • Next-generation sequencing of libraries before and after selection

    • Computational analysis to identify enriched sequences and binding motifs

Recent research demonstrates that combining phage display experiments with computational analysis can enable the identification of different binding modes associated with particular ligands . This approach allows researchers to disentangle binding modes even when they are associated with chemically very similar ligands, facilitating the design of antibodies with customized specificity profiles .

What experimental controls are essential when conducting antibody-based research?

Rigorous experimental controls are critical for ensuring the validity and reproducibility of antibody-based research:

  • Antibody validation controls:

    • Isotype controls to assess non-specific binding

    • Knockout/knockdown systems to verify specificity

    • Peptide blocking experiments to confirm epitope specificity

    • Secondary antibody-only controls to assess background

  • Assay-specific controls:

    • For ELISA: blank wells, non-specific binding controls, standard curves

    • For Western blotting: molecular weight markers, positive/negative controls

    • For immunohistochemistry: tissue sections known to express/not express the target

  • Experimental design controls:

    • Technical replicates to assess assay variability

    • Biological replicates to account for biological variation

    • Positive and negative controls for each experimental condition

  • Data analysis controls:

    • Blinding during image analysis or scoring

    • Appropriate statistical tests with multiple comparison corrections

    • Randomization of samples to minimize batch effects

The importance of proper controls is exemplified in the study of antibody responses to SARS-CoV-2, where researchers used flow cytometry to isolate B lymphocytes with receptors that bound to RBD from convalescent individuals . They validated their findings by comparing to pre-COVID-19 controls, where RBD-binding B cells were undetectable, confirming the specificity of their isolation approach .

How should researchers analyze antibody binding kinetics data?

Accurate analysis of antibody binding kinetics requires:

When interpreting binding data, researchers should be aware that extremely high affinity (sub-picomolar KD) may indicate problems with the experimental setup or analysis, and validation with orthogonal methods is recommended.

What approaches can resolve discrepancies between different antibody characterization methods?

When faced with discrepancies between different characterization methods:

  • Systematic troubleshooting:

    • Evaluate reagent quality and experimental conditions

    • Assess method-specific limitations and biases

    • Consider epitope accessibility differences between methods

  • Orthogonal method validation:

    • Implement additional complementary techniques

    • Compare results across multiple experimental platforms

    • Validate findings with functional assays

  • Method-specific considerations:

    • For binding assays: antigen presentation format (solid-phase vs. solution)

    • For cell-based assays: expression level, conformational state

    • For in vivo studies: species differences, biodistribution, clearance

  • Integrated data analysis approaches:

    • Weight evidence based on method reliability and relevance

    • Develop a consensus model that accounts for all observations

    • Identify experimental conditions that reconcile discrepancies

Understanding the limitations of each method is crucial. For example, ELISA assays may not accurately reflect binding to cell-surface antigens due to differences in protein conformation or accessibility. In a study examining antibody responses to SARS-CoV-2, researchers used both ELISA assays and flow cytometry to characterize antibody binding, finding that 95% of antibodies bound to SARS-CoV-2 RBD with an average EC50 of 6.9 ng/mL .

How can researchers predict and mitigate immunogenicity risks with therapeutic antibodies?

Immunogenicity remains a significant challenge for therapeutic antibodies. Researchers can address this through:

  • In silico prediction methods:

    • T-cell epitope analysis

    • Sequence-based aggregation prediction

    • Structural stability assessment

  • Deimmunization strategies:

    • Humanization of non-human antibodies

    • Removal of T-cell epitopes

    • Framework region optimization

    • Identification and elimination of post-translational modification sites

  • Experimental risk assessment:

    • HLA binding assays

    • T-cell proliferation assays

    • Dendritic cell activation assays

    • Transgenic animal models

  • Clinical mitigation strategies:

    • Concomitant immunosuppression

    • Optimization of dosing regimen

    • Patient-specific risk assessment based on HLA type

The importance of immunogenicity management is illustrated in the development of therapeutic antibodies like the humanized anti-CD132 monoclonal antibody 2D4, which was specifically humanized to reduce immunogenicity while maintaining its ability to block IL-21 and IL-15 signaling .

What factors determine optimal antibody dosing in different therapeutic contexts?

Determining optimal antibody dosing requires consideration of multiple factors:

  • Pharmacokinetic parameters:

    • Half-life and clearance mechanisms

    • Volume of distribution

    • Target-mediated drug disposition (TMDD)

  • Pharmacodynamic considerations:

    • Minimum effective concentration

    • Receptor occupancy requirements

    • On-target vs. off-target effects

    • Duration of response

  • Disease-specific factors:

    • Target expression level and turnover

    • Tissue penetration requirements

    • Immune system status

  • Patient factors:

    • Genetic polymorphisms affecting clearance

    • Body weight and composition

    • Co-morbidities and co-medications

    • Age and organ function

For example, with anti-amyloid monoclonal antibodies for Alzheimer's disease, dosing considerations must balance efficacy in amyloid plaque removal against the risk of ARIA, particularly in patients with genetic risk factors like the APOE-4 allele . Some patients, particularly those with two copies of the APOE-4 allele, may have an unfavorable risk-benefit ratio that precludes treatment altogether .

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.