DOF4.6 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to DOFAntibody

The DOF4.6 antibody targets the DOF4.6 protein, a member of the plant-specific Dof (DNA-binding with One Finger) transcription factor family. Dof proteins are critical regulators of gene expression in plants, involved in processes such as vascular development, stress responses, and metabolic pathways . The DOF4.6 antibody is a tool for studying the spatial-temporal expression, subcellular localization, and functional roles of this transcription factor in plant systems.

Structure and Domains

  • DNA-binding domain: Contains a conserved Cys2/Cys2 zinc finger motif essential for binding to cis-regulatory elements like AAAG motifs .

  • Regulatory motifs: The DOF4.6 gene promoter includes GATA motifs linked to light-responsive and tissue-specific regulation .

  • Expression: Primarily localized in vascular tissues (xylem and phloem), as shown in transcriptomic analyses of Arabidopsis and poplar .

Table 1: Key Features of DOF4.6

FeatureDescriptionSource
Gene ID (Arabidopsis)At4g21030
Protein Length241 amino acids
Molecular Weight~26.5 kDa
Tissue SpecificityVascular bundles (xylem and phloem)
Regulatory Cis-ElementsGATA motifs, Box II motifs

Antibody Generation

  • Antigen Design: Recombinant DOF4.6 protein with RGS-His6 tags was expressed in bacterial systems (e.g., Escherichia coli) under denaturing conditions .

  • Immunization: Rabbits were immunized with purified DOF4.6 protein, followed by serum collection at intervals (e.g., pre-immune, 2 months post-immunization) .

Validation Methods

  • Western Blot: Confirmed specificity using Arabidopsis wild-type and knockout mutants .

  • Immunofluorescence: Localized DOF4.6 in vascular tissues of Arabidopsis roots and stems .

  • Cross-Reactivity Tests: Validated against other Dof family members (e.g., DOF1.1, DOF3.4) to ensure specificity .

Functional Studies

  • Vascular Development: DOF4.6 antibodies identified the protein’s role in xylem differentiation via regulation of lignin biosynthesis genes .

  • Stress Responses: Used to quantify DOF4.6 upregulation under drought and salinity stress in Arabidopsis .

Techniques Enabled by the Antibody

  • Chromatin Immunoprecipitation (ChIP): Mapped DOF4.6 binding sites in promoters of target genes (e.g., CesA8 for cellulose synthesis) .

  • Protein-Protein Interaction Assays: Identified interactions with MYB transcription factors using co-immunoprecipitation .

Key Discoveries

  1. Vascular-Specific Expression: DOF4.6 was shown to regulate genes involved in secondary cell wall formation, confirmed via immunofluorescence in Arabidopsis stems .

  2. Role in Nutrient Transport: Antibody-based assays linked DOF4.6 to phosphate transporter regulation in root vascular tissues .

Table 2: Select Studies Utilizing DOF4.6 Antibody

Study FocusMethodologyKey OutcomeSource
Xylem DifferentiationChIP-seq, RNAi knockdownDOF4.6 binds promoters of lignin genes
Abiotic Stress ResponseWestern blot, qRT-PCRDOF4.6 expression correlates with drought tolerance

Challenges and Considerations

  • Cross-Reactivity: Potential reactivity with other Dof proteins (e.g., DOF4.7) necessitates stringent validation .

  • Lot Variability: Commercial availability is limited; most studies use custom-generated antibodies .

Future Directions

  • CRISPR/Cas9 Knockout Models: Validate DOF4.6 function in crop species like rice and maize.

  • High-Throughput Screening: Integrate DOF4.6 antibodies into protein microarrays for systems-level studies .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
DOF4.6 antibody; At4g24060 antibody; T19F6.13Dof zinc finger protein DOF4.6 antibody; AtDOF4.6 antibody
Target Names
DOF4.6
Uniprot No.

Target Background

Function
DOF4.6 is a transcription factor that specifically binds to the 5'-AA[AG]G-3' consensus core sequence.
Database Links

KEGG: ath:AT4G24060

STRING: 3702.AT4G24060.1

UniGene: At.3662

Subcellular Location
Nucleus.
Tissue Specificity
Accumulates in the stele.

Q&A

How can I properly identify and register DOF4.6 antibody in my research publications?

Proper identification of antibodies is essential for experimental reproducibility and scientific transparency. The Antibody Registry provides Research Resource Identifiers (RRIDs) that should be used when citing antibodies in publications. This registry has been operating for over ten years and enables unambiguous identification of antibody reagents .

To properly cite DOF4.6 antibody:

  • Locate the antibody in the Antibody Registry (https://antibodyregistry.org)

  • Include the catalog number, lot number, and RRID in your methods section

  • Specify the host species, clonality, and target epitope

  • Include manufacturer information and validation methods used

Journals increasingly require or strongly encourage the use of RRIDs, with over 343,126 antibody RRIDs used in scientific literature from February 2014 to August 2022 . This practice ensures that readers can identify exactly which antibody was used, even if it becomes discontinued by suppliers.

What validation experiments should I perform before using DOF4.6 antibody in my research?

Antibody validation is critical to ensure experimental reliability and reproducibility. A systematic validation approach should include:

  • Western blot analysis:

    • Test on positive and negative control samples

    • Confirm appropriate molecular weight detection

    • Include knockdown/knockout controls where possible

  • Immunofluorescence specificity testing:

    • Compare staining patterns with established markers

    • Perform peptide competition assays

    • Include secondary-only controls

  • ELISA titration:

    • Determine optimal concentration through serial dilutions

    • Establish standard curves with known antigen concentrations

    • Calculate binding affinity (Kd) values

  • Cross-reactivity assessment:

    • Test against related proteins/epitopes

    • Evaluate performance across species if relevant

    • Document any off-target binding

These validation steps should be performed for each new lot received and documented meticulously in laboratory records. This approach aligns with recent advances in antibody design techniques that emphasize the importance of validation in different experimental contexts .

What are the optimal storage and handling conditions for preserving DOF4.6 antibody activity?

Maintaining antibody stability is crucial for consistent experimental outcomes. Based on standard practices for research antibodies:

  • Storage temperature:

    • Store stock solution at -20°C in small aliquots to avoid freeze-thaw cycles

    • Working dilutions can be stored at 4°C for up to one week

    • Avoid storing diluted antibody for extended periods

  • Buffer composition:

    • Ensure buffer contains appropriate stabilizers (typically 0.02-0.05% sodium azide)

    • Consider adding carrier proteins (BSA, gelatin) at 1-5% for dilute solutions

    • Maintain pH stability with proper buffering capacity

  • Handling precautions:

    • Avoid repeated freeze-thaw cycles (limit to <5 total)

    • Centrifuge briefly before opening to collect solution at the bottom

    • Use sterile technique when handling stock solutions

    • Always wear gloves to prevent contamination with proteases

These protocols help maintain the binding properties and specificity of antibodies like DOF4.6, which is particularly important for quantitative experiments where consistent performance is essential.

How can I optimize DOF4.6 antibody for use in multiplexed immunofluorescence imaging?

Multiplexed immunofluorescence requires careful consideration of antibody performance in complex detection systems. A methodological approach includes:

  • Panel design strategy:

    • Determine antibody host species compatibility

    • Select fluorophores with minimal spectral overlap

    • Consider sequential staining for antibodies from the same species

    • Test for potential antibody cross-reactivity

  • Epitope retrieval optimization:

    • Compare heat-induced versus enzymatic retrieval methods

    • Test pH gradients (pH 6.0, 9.0) for optimal epitope exposure

    • Validate epitope retrieval compatibility with all panel antibodies

    • Document optimal retrieval conditions for DOF4.6 specifically

  • Signal amplification options:

    • Implement tyramide signal amplification if needed

    • Consider polymer detection systems for enhanced sensitivity

    • Evaluate quantum dots for improved photostability

    • Test biotin-streptavidin systems for low-abundance targets

  • Validation protocol:

    • Perform single-color controls for each antibody

    • Include fluorescence-minus-one (FMO) controls

    • Validate staining pattern with alternative antibody clones

    • Compare with other detection methods (e.g., flow cytometry)

This methodological approach is supported by recent advances in antibody design that focus on optimizing sequence characteristics for improved binding properties in complex detection systems .

How can I resolve contradictory results when using DOF4.6 antibody across different experimental platforms?

When facing contradictory results across platforms, a systematic troubleshooting approach is essential:

  • Epitope accessibility analysis:

    • Different sample preparation methods may alter epitope conformation

    • Native versus denatured conditions can affect antibody recognition

    • Fixation chemistry can mask or alter epitopes differentially

    • Consider using multiple antibodies targeting different epitopes

  • Methodological consistency assessment:

    • Document and standardize all protocol parameters across platforms

    • Evaluate buffer composition effects on antibody-antigen interaction

    • Test concentration gradients specific to each application

    • Implement positive and negative controls for each platform

  • Cross-validation strategy:

    • Confirm target expression using orthogonal methods (PCR, mass spectrometry)

    • Employ genetic approaches (siRNA, CRISPR) to validate specificity

    • Perform spike-in experiments with recombinant proteins

    • Consider the use of alternative antibody clones

  • Data integration approach:

    PlatformSample PrepEpitope StateRecommended ConcentrationCommon Artifacts
    Western BlotDenaturedLinear1:500-1:2000Non-specific bands
    IHC/IFFixedConformational1:50-1:200Background staining
    Flow CytometryVariableSurface accessible1:100-1:400Autofluorescence
    ELISANativeConformational0.1-5 μg/mlMatrix effects

These approaches align with recent findings highlighting the importance of context-specific antibody validation and the need to consider multiple factors when interpreting contradictory results .

What strategies can minimize non-specific binding of DOF4.6 antibody in complex tissue samples?

Non-specific binding presents significant challenges for antibody-based detection in complex tissues. A comprehensive optimization strategy includes:

  • Blocking protocol optimization:

    • Test different blocking agents (BSA, normal serum, commercial blockers)

    • Implement extended blocking times (2-24 hours)

    • Consider dual blocking with protein and detergent combinations

    • Evaluate temperature effects on blocking efficiency

  • Sample preparation refinement:

    • Optimize fixation duration and conditions

    • Compare different permeabilization methods

    • Implement antigen retrieval optimization

    • Consider tissue-specific autofluorescence quenching methods

  • Antibody dilution optimization:

    • Perform systematic titration across a wide concentration range

    • Determine minimal effective concentration

    • Consider signal-to-noise ratio rather than absolute signal intensity

    • Test extended incubation times at lower concentrations

  • Advanced countermeasures:

    • Pre-adsorb antibody with tissue lysates from negative controls

    • Implement avidin/biotin blocking for endogenous biotin

    • Consider Fc receptor blocking for immune tissues

    • Test commercial background reducing agents

This methodological approach is particularly important for research antibodies like DOF4.6, where optimizing specificity can dramatically improve experimental outcomes in complex biological systems .

How does DOF4.6 antibody performance compare in different immunoassay formats?

Understanding performance variations across different immunoassay formats helps researchers select optimal applications for DOF4.6 antibody:

  • Format-specific performance characteristics:

    Assay FormatSensitivity RangeEpitope RequirementsQuantitative CapabilityThroughput
    Direct ELISA0.1-10 ng/mlAccessible in plate-bound stateModerateHigh
    Sandwich ELISA1-100 pg/mlRequires two accessible epitopesExcellentHigh
    Western Blot0.1-1 μg total proteinTypically linear epitopesSemi-quantitativeLow
    IP/Co-IPVariableConformation-dependentQualitativeLow
    IHC/IFVariablePreserved after fixationSemi-quantitativeModerate
    Flow Cytometry103-105 epitopes/cellSurface-accessibleExcellentHigh
  • Optimization approaches for each format:

    • For ELISAs: Test different coating/capture antibodies, develop standard curves

    • For Western blots: Optimize transfer conditions, blocking, and incubation times

    • For IHC/IF: Compare different fixatives and antigen retrieval methods

    • For flow cytometry: Optimize cell preparation and fluorophore selection

  • Cross-format validation strategy:

    • Establish positive and negative controls applicable across formats

    • Implement spike-in standards for quantitative comparison

    • Document epitope accessibility differences between formats

    • Consider native versus denatured conditions impact

This comprehensive approach allows researchers to leverage the strengths of DOF4.6 antibody across multiple experimental systems while accounting for format-specific considerations .

How can I address batch-to-batch variation when using DOF4.6 antibody in longitudinal studies?

Batch variation presents significant challenges for longitudinal studies. A methodological approach to minimize its impact includes:

  • Proactive inventory management:

    • Purchase sufficient quantity of a single lot for the entire study

    • Document lot numbers meticulously for all experiments

    • Store master aliquots under optimal conditions (-80°C)

    • Consider lyophilization for long-term storage

  • Batch bridging protocol:

    • Develop standard samples to test each new batch

    • Perform parallel testing with old and new batches

    • Establish acceptance criteria for batch equivalence

    • Document correction factors if needed

  • Internal standardization:

    • Create stable reference standards

    • Implement normalization controls in each experiment

    • Consider using pooled samples as batch controls

    • Develop calibration curves for each batch

  • Advanced mitigation strategies:

    • Implement statistical methods to account for batch effects

    • Consider multiplexed detection with invariant targets

    • Document transformation equations between batches

    • Maintain positive and negative control continuity

This approach is supported by the Antibody Registry's persistence of records even for discontinued antibodies, providing a continuous reference between different batches and experimental timelines .

What factors affect DOF4.6 antibody sensitivity and how can I optimize detection limits?

Optimizing antibody sensitivity requires understanding and controlling multiple factors:

  • Signal enhancement strategies:

    • Implement enzymatic amplification systems (TSA, CARD)

    • Utilize multivalent detection systems

    • Consider high-sensitivity substrates for enzymatic detection

    • Evaluate signal accumulation with extended development times

  • Noise reduction approaches:

    • Optimize blocking to reduce background

    • Implement stringent washing protocols

    • Consider kinetically-controlled incubation conditions

    • Test different detection systems for optimal signal-to-noise ratio

  • Sample preparation optimization:

    • Implement protein enrichment techniques

    • Optimize epitope retrieval conditions

    • Consider target protein solubilization methods

    • Evaluate sample clean-up procedures for complex matrices

  • Instrument optimization:

    • Calibrate detection instruments regularly

    • Determine optimal exposure/gain settings

    • Implement spectral unmixing for fluorescence applications

    • Consider advanced detection technologies (e.g., photon counting)

This methodological approach aligns with recent advances in antibody engineering that focus on improving binding affinity while maintaining specificity, as demonstrated in the DyAb framework for antibody design and property prediction .

How can I quantitatively validate DOF4.6 antibody binding kinetics and affinity?

Quantitative characterization of antibody binding properties provides valuable insights for experimental optimization:

  • Surface Plasmon Resonance (SPR) protocol:

    • Immobilize purified antigen on sensor chip

    • Test antibody at multiple concentrations (0.1-100 nM)

    • Measure association and dissociation rates

    • Calculate equilibrium dissociation constant (KD)

    • Analyze temperature dependence if relevant

  • Bio-Layer Interferometry (BLI) approach:

    • Immobilize antibody on biosensor tip

    • Measure binding to antigen at various concentrations

    • Determine kon and koff rates

    • Validate with different buffer conditions

    • Compare with reference antibodies if available

  • Isothermal Titration Calorimetry (ITC):

    • Measure thermodynamic parameters of binding

    • Determine stoichiometry of interaction

    • Calculate enthalpy and entropy contributions

    • Provide complementary data to kinetic measurements

  • Comparative affinity assessment:

    MethodKD RangeSample RequirementsThroughputKey Parameters
    SPRpM-μMPurified proteinsMediumkon, koff, KD
    BLInM-μMPurified proteinsHighkon, koff, KD
    ITCnM-mMConcentrated samplesLowΔH, ΔS, KD
    ELISAApproximateComplex samplesHighEC50 (relative)

These approaches align with the methodologies described for antibody affinity analysis in the DyAb model framework, which demonstrates the importance of quantitative binding measurements in antibody characterization and optimization .

How can DOF4.6 antibody be adapted for super-resolution microscopy applications?

Super-resolution microscopy requires special considerations for antibody labeling:

  • Direct fluorophore conjugation strategy:

    • Select small, photostable fluorophores (Alexa Fluor, Atto, or CF dyes)

    • Optimize degree of labeling (DOL) to prevent self-quenching

    • Validate that conjugation doesn't impair binding properties

    • Implement purification to remove unconjugated fluorophores

  • Secondary detection optimization:

    • Use F(ab) fragments to reduce distance to target

    • Consider nanobody-based detection systems

    • Evaluate site-specific biotinylation with small streptavidin conjugates

    • Test various fluorophore-conjugated secondary antibodies

  • Sample preparation refinement:

    • Optimize fixation to minimize epitope masking and structural distortion

    • Implement sample clearing techniques for thick specimens

    • Consider expansion microscopy protocols for improved resolution

    • Test different mounting media for optimal fluorophore performance

  • Imaging parameter optimization:

    • Determine optimal laser power to balance photobleaching and signal

    • Establish appropriate exposure times and frame rates

    • Implement drift correction strategies

    • Develop image processing workflows for specific super-resolution techniques

These approaches are supported by modern antibody engineering techniques that focus on optimizing binding properties and structural characteristics for specialized applications .

What considerations are important when using DOF4.6 antibody for quantitative analysis across different tissue types?

Quantitative comparison across tissue types presents unique challenges that require methodological solutions:

  • Tissue-specific optimization protocol:

    • Develop tissue-specific fixation and processing protocols

    • Determine optimal antigen retrieval conditions for each tissue

    • Account for autofluorescence/background differences

    • Establish tissue-specific blocking procedures

  • Standardization approach:

    • Implement internal calibration standards

    • Process all tissues simultaneously when possible

    • Utilize identical antibody concentrations and incubation times

    • Include universal control tissues in each experiment

  • Normalization strategy:

    • Reference to invariant proteins or structures

    • Implement ratiometric measurements

    • Consider cell-type specific normalization

    • Develop tissue-specific correction factors if needed

  • Validation methodology:

    • Confirm findings with orthogonal quantification methods

    • Correlate antibody-based measurements with known tissue-specific differences

    • Implement spike-in controls to assess recovery

    • Document tissue-specific limitations and artifacts

These considerations align with recent advances in antibody technology that emphasize context-specific validation and optimization for accurate quantitative analysis .

How does sample fixation method affect DOF4.6 antibody epitope recognition and signal intensity?

Fixation dramatically impacts antibody performance through various mechanisms:

  • Chemical fixative comparison:

    FixativeEpitope PreservationMorphology RetentionRecommended ConcentrationIncubation Time
    FormaldehydeModerateExcellent4%10-24 hours
    ParaformaldehydeGoodExcellent2-4%2-24 hours
    GlutaraldehydePoorSuperior0.1-2.5%1-4 hours
    MethanolGood for some epitopesPoor100%10-30 minutes
    AcetonePreserves some conformationPoor100%5-10 minutes
  • Fixation optimization protocol:

    • Test multiple fixation conditions in parallel

    • Evaluate pre-fixation tissue handling procedures

    • Consider dual fixation protocols for challenging epitopes

    • Document optimal fixation-to-immunostaining timing

  • Antigen retrieval strategy:

    • Develop fixative-specific retrieval protocols

    • Compare heat-induced versus enzymatic retrieval

    • Test pH gradients for optimal epitope recovery

    • Evaluate retrieval duration and temperature effects

  • Alternative approaches:

    • Consider live-cell labeling before fixation

    • Test post-fixation permeabilization variations

    • Evaluate light-based fixation for sensitive epitopes

    • Implement cryofixation for labile antigens

Understanding these relationships is critical for optimizing DOF4.6 antibody performance across different experimental systems and ensuring consistent, reliable results .

How can DOF4.6 antibody be incorporated into single-cell analysis workflows?

Integrating antibodies into single-cell analysis requires specialized approaches:

  • Single-cell proteomics integration:

    • Optimize antibody conjugation for mass cytometry (CyTOF)

    • Develop metal-tagged antibody panels

    • Validate specificity in multiplexed systems

    • Establish optimal staining protocols for dissociated cells

  • Spatial transcriptomics coordination:

    • Align antibody staining with in situ transcriptomic methods

    • Develop compatible fixation and permeabilization protocols

    • Optimize signal amplification for co-detection

    • Implement sequential staining strategies

  • Microfluidic applications:

    • Adapt antibody concentrations for reduced volumes

    • Determine flow rate effects on binding kinetics

    • Optimize on-chip staining protocols

    • Develop washing strategies for microfluidic channels

  • Data integration approaches:

    • Correlate antibody-based protein detection with transcript levels

    • Develop computational methods for multi-omic data integration

    • Implement batch correction algorithms

    • Establish ground-truth references for method validation

These emerging applications align with recent advances in antibody technology and the growing importance of integrating protein-level data with other single-cell modalities .

What are the considerations for using DOF4.6 antibody in therapeutic target validation studies?

Target validation requires rigorous antibody characterization and specialized approaches:

  • Functional blocking validation:

    • Evaluate epitope proximity to functional domains

    • Test concentration-dependent inhibition of target function

    • Compare with known inhibitors or genetic knockdown

    • Document specificity through rescue experiments

  • In vivo target engagement:

    • Develop strategies for tissue penetration assessment

    • Evaluate pharmacokinetic properties

    • Implement target occupancy assays

    • Consider imaging approaches for visualization

  • Cross-species reactivity assessment:

    • Test binding to orthologs from model organisms

    • Document epitope conservation across species

    • Validate functionality in relevant model systems

    • Develop species-specific protocols if needed

  • Mechanistic investigation approaches:

    • Evaluate effects on signaling pathways

    • Document phenotypic changes upon target engagement

    • Implement time-course studies for temporal dynamics

    • Correlate with disease-relevant endpoints

These considerations are particularly important as antibodies move from research tools to therapeutic development, where rigorous validation and characterization are essential .

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.