DOF5.6 Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Development and Validation

DOF5.6 antibodies are typically raised against synthetic peptides or recombinant protein fragments. For example:

Table 1: Example Antigen Design for DOF Antibodies

Target RegionHost SpeciesClonalityApplicationCross-ReactivitySource
C-terminal peptide (AA 1324–1352)RabbitPolyclonalWB, IHCHuman, Arabidopsis
Recombinant protein (AA 1–376)MousePolyclonalWBHuman
  • Validation: Antibodies are tested via Western blot (WB), immunoprecipitation (IP), and immunolocalization in mutant backgrounds to confirm specificity .

Role in Vascular Development

  • DOF5.6/HCA2 interacts with TMO5/LHW transcription factors to regulate periclinal and radial cell divisions in the procambium .

  • Mutant analysis: Triple mutants (dof2.1 dof6 tmo6) show disrupted vascular patterning, highlighting functional redundancy among DOF proteins .

Cytokinin Signaling

  • DOF5.6 expression is induced by cytokinin, linking it to hormone-mediated vascular development .

  • Mechanism: Acts downstream of TMO5/LHW to activate cell proliferation genes (e.g., CYCD3) .

Applications in Plant Research

  • Western blotting: Detects ~32–35 kDa DOF5.6 protein in Arabidopsis root extracts .

  • Immunolocalization: Localizes DOF5.6 to procambium and xylem precursor cells .

  • Functional studies: Used to characterize loss-of-function mutants and overexpression lines .

Limitations and Challenges

  • Redundancy: Functional overlap with homologs (e.g., DOF2.1, DOF6) complicates phenotype analysis .

  • Antibody specificity: Cross-reactivity with other DOF family members requires rigorous validation .

Future Directions

  • Structural studies: Resolving DOF5.6-DNA complexes to elucidate binding specificity.

  • Agricultural applications: Engineering DOF5.6 expression to modulate vascular development in crops.

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Components: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (Made-to-order)
Synonyms
DOF5.6 antibody; HCA2 antibody; At5g62940 antibody; MQB2.26Dof zinc finger protein DOF5.6 antibody; AtDOF5.6 antibody; Protein HIGH CAMBIAL ACTIVITY 2 antibody; Protein PHLOEM EARLY DOF HCA2 antibody
Target Names
DOF5.6
Uniprot No.

Target Background

Function
DOF5.6 is a transcription factor that specifically binds to the 5'-AA[AG]G-3' consensus core DNA sequence, thereby promoting gene expression. PEAR proteins, including DOF2.4, DOF5.1, DOF3.2, DOF1.1, DOF5.6, and DOF5.3, activate gene expression crucial for the radial growth of protophloem sieve elements. DOF5.6 plays a significant role in regulating interfascicular cambium formation and vascular tissue development, particularly during the early stages of inflorescence stem development. Its function promotes both cambium activity and phloem differentiation while inhibiting xylem differentiation.
Gene References Into Functions
  1. The transcription factor gene, HCA2, is implicated in the regulation of interfascicular cambium formation and vascular tissue development in Arabidopsis. [HCA2] PMID: 19915089
Database Links

KEGG: ath:AT5G62940

STRING: 3702.AT5G62940.1

UniGene: At.29026

Subcellular Location
Nucleus.
Tissue Specificity
The PEAR proteins (e.g. DOF2.4, DOF5.1, DOF3.2, DOF1.1, DOF5.6 and DOF5.3) form a short-range concentration gradient that peaks at protophloem sieve elements (PSE). Preferentially expressed in the vasculature of all organs, including seedlings, roots, ste

Q&A

How do I select the appropriate antibody clone for my specific research application?

Antibody selection should be based on several critical factors including target specificity, application compatibility, and experimental conditions. For instance, when studying protein-protein interactions or specific signaling pathways, consider whether the epitope recognized by the antibody might be masked in your experimental system. The choice between monoclonal and polyclonal antibodies depends on your specific research needs - monoclonal antibodies offer high specificity for a single epitope, while polyclonal antibodies recognize multiple epitopes and may provide stronger signals.

When selecting clones, review validation data specific to your application. For example, the D5/16B4 clone for Cytokeratin 5 & 6 antibodies has been validated for both paraffin-embedded and frozen tissue sections, making it versatile for different sample preparations . Additionally, consider the antibody's isotype (e.g., IgG1) and reactivity conditions, as these parameters influence experimental design and compatibility with secondary detection systems .

What validation steps should I perform to ensure antibody specificity before proceeding with experiments?

Rigorous validation is essential to prevent experimental artifacts and ensure reproducible results. Implement these critical validation procedures:

  • Positive and negative controls: Use tissues or cell lines with known expression patterns. For Cytokeratin 5 & 6 antibodies, squamous cell carcinoma (SCC) or basal cell carcinoma (BCC) samples serve as effective positive controls .

  • Secondary antibody-only controls: Essential to detect non-specific binding.

  • Concentration gradients: Test various antibody dilutions to determine optimal signal-to-noise ratios.

  • Cross-reactivity testing: Particularly important when studying closely related proteins.

  • Method compatibility: Validate antibodies specifically for your experimental conditions. For example, antibodies validated for ChIP-Seq may not work effectively in native conditions used in CUT&Tag assays .

  • Epitope verification: Confirm that sample preparation does not alter the target epitope. For instance, certain fixation methods may mask epitopes or change their availability .

This systematic approach ensures reliable experimental outcomes and prevents resource waste on non-specific antibodies.

How should sample preparation be modified when using antibodies for different experimental techniques?

Sample preparation must be tailored to both the antibody properties and the experimental technique:

For immunohistochemistry using frozen sections (as with TintoFast Cytokeratin 5 & 6 antibody D5/16B4):

  • Embed specimen in OCT inside a cryostat

  • Cut sections at 4-5 μm thickness and mount on positively charged slides

  • Air dry at room temperature for 2 minutes followed by incubation at 60°C for 3 minutes

  • Fix using either 100% acetone or 10% neutral buffered formalin (NBF) for 2 minutes at room temperature, with NBF providing better morphology

For CUT&Tag assays:

  • Permeabilize native/unfixed cells (unlike traditional ChIP-Seq which uses fixed cells)

  • Immobilize cells on concanavalin A-coated magnetic beads

  • Maintain cells on ice after permeabilization to preserve nuclear integrity

  • Optimize digitonin concentration to 0.01% during later steps to prevent clump formation

For traditional ChIP assays versus newer techniques:

  • ChIP typically requires crosslinking with formaldehyde and sonication

  • Native approaches like CUT&Tag omit fixation, which may affect epitope availability

  • The native conditions of CUT&Tag may not be suitable for all targets, particularly transiently binding transcription factors or those with weak DNA interactions

These protocol adjustments can significantly impact experimental outcomes and should be carefully optimized for each target protein and antibody combination.

What are the key differences between using antibodies in ChIP-Seq versus CUT&Tag assays?

These two methods represent different approaches to studying protein-DNA interactions, with important methodological distinctions:

ParameterChIP-SeqCUT&Tag
Sample preparationFormaldehyde fixation to crosslink protein-DNA interactionsUses native/unfixed cells
Chromatin processingSonication or enzymatic digestion to fragment chromatinNo sonication; uses antibody-directed Tn5 transposase for tagmentation
Material requirementsGenerally requires high cell numbers (10⁵-10⁷)Works with low cell numbers (as few as 500 cells)
Antibody conditionsMost antibodies validated for crosslinked conditionsRequires antibodies that work in native conditions
Target suitabilityWorks for most proteins including transient DNA bindersBest for abundant proteins with stable DNA binding
Workflow durationTypically 2-3 daysCan be completed in 1 day
Bias considerationsLess intrinsic bias toward open chromatinTn5 transposase has affinity for open chromatin regions
Library preparationSeparate step after immunoprecipitationIntegrated with the protein targeting step via Tn5 tagmentation

CUT&Tag might preferentially detect factors associated with active genomic regions due to the Tn5 transposase's affinity for open chromatin, potentially introducing bias when studying heterochromatic regions or silenced genes . This limitation should be considered when selecting between methods, especially for targets associated with repressed chromatin states.

How can I diagnose and resolve high background issues when using antibodies in immunoassays?

High background represents a common challenge that can obscure meaningful signals. Systematic troubleshooting includes:

  • Antibody concentration optimization: Excessive antibody concentration often leads to high background. Perform titration experiments to identify the minimum concentration that yields specific signal. For example, when using DR5 monoclonal antibody (D-6) in apoptosis studies, dose-dependent effects should be carefully calibrated .

  • Blocking optimization: Insufficient blocking allows non-specific antibody binding. Test different blocking agents (BSA, serum, commercial blockers) and extend blocking time if necessary.

  • Washing stringency adjustment: Increase wash buffer volumes, duration, or detergent concentration. For CUT&Tag protocols, proper washing after primary and secondary antibody incubations is crucial .

  • Secondary antibody cross-reactivity: Test secondary antibody alone (no primary) to identify non-specific binding. Consider using antibodies pre-adsorbed against potential cross-reactive species.

  • Sample autofluorescence or endogenous enzyme activity: Include appropriate quenching steps—e.g., sodium borohydride for aldehyde-induced autofluorescence or hydrogen peroxide for endogenous peroxidase quenching.

  • Fixation artifact diagnosis: Overfixation can increase background through increased hydrophobicity or tissue autofluorescence. Optimize fixation duration and conditions.

When working with antibody-directed tagmentation methods like CUT&Tag, additional considerations include optimizing digestion time and pA-Tn5 quantity to avoid unspecific tagmentation .

What strategies can overcome poor antibody performance in specific applications?

When antibody performance is suboptimal, consider these intervention strategies:

  • Epitope retrieval modification: For immunohistochemistry, test different antigen retrieval methods (heat-induced versus enzymatic) and buffer compositions (citrate versus EDTA-based).

  • Alternative fixation protocols: If poor performance occurs with standard fixation, evaluate alternative fixatives or fixation durations. For instance, while 10% NBF provides better morphology for the TintoFast Cytokeratin 5 & 6 antibody, acetone fixation may preserve certain epitopes better .

  • Buffer optimization: Adjust salt concentration, pH, or detergent type/concentration. For CUT&Tag assays, increasing NaCl concentration can prevent non-specific Tn5 digestion of accessible chromatin .

  • Antibody format switching: If a directly conjugated antibody performs poorly, try unconjugated primary with separate secondary detection.

  • Clone alternatives: Different clones recognizing different epitopes may perform better in specific applications. For difficult targets, consider testing multiple validated clones.

  • Combined approach methodology: Some targets benefit from enhanced detection through amplification systems or combining complementary techniques. For example, the combination of DR5 monoclonal antibody (D-6) with cisplatin demonstrated enhanced apoptosis induction in ovarian cancer cells compared to single-agent treatment .

Careful documentation of these optimizations builds institutional knowledge and saves time for future experiments targeting the same proteins.

How can antibody-based techniques be adapted for single-cell or low-input samples?

Recent methodological advances have revolutionized antibody applications for limited samples:

The CUT&Tag technique represents a significant advancement for low-input samples, requiring as few as 500 cells compared to the 10⁵-10⁷ cells typically needed for ChIP-Seq . This breakthrough enables:

  • Epigenomic profiling of rare cell populations or clinical samples

  • Analysis of histone modifications and DNA-binding proteins from limited biological material

  • Integration with single-cell sequencing platforms

The workflow adapts traditional techniques by:

  • Using concanavalin A-coated magnetic beads to immobilize permeabilized cells

  • Employing antibody-directed Tn5 transposase (pA-Tn5 adapter transposome) that simultaneously cleaves DNA and inserts sequencing adapters

  • Minimizing amplification cycles (12-14 or fewer) to avoid PCR duplicates

For immunofluorescence applications with limited samples, tyramide signal amplification (TSA) can enhance detection sensitivity by 10-100 fold, enabling visualization of low-abundance targets in rare cells or small tissue biopsies.

These adaptations allow researchers to extract maximum information from precious samples while maintaining signal specificity and experimental rigor.

What are the latest developments in antibody conjugation techniques for enhancing detection sensitivity?

Innovative conjugation approaches have significantly expanded antibody capabilities:

  • Enzyme-based site-specific conjugation:
    The TAM-ChIP method employs antibodies conjugated directly to Tn5 transposase loaded with sequencing adapters, increasing sensitivity and binding site resolution while reducing experimental time . Similar principles apply in newer CUT&Tag implementations.

  • Sortase-mediated conjugation for recombinant antibodies:
    AbFlex® recombinant antibodies containing transpeptidase Sortase recognition sequences on Fc fragments enable site-specific addition of various tags, including Tn5 transposase . This approach allows precise control over conjugation stoichiometry and orientation.

  • Multiplexed detection systems:
    Primary antibody-based tagmentation adapted for multiplexed assays enables simultaneous investigation of multiple histone modifications or transcription factors from the same samples, dramatically increasing experimental efficiency .

  • Application to methylation detection:
    Primary antibody-based tagmentation techniques are being adapted for MeDIP (Methylated DNA Immunoprecipitation) experiments, increasing both sensitivity and specificity by more precisely identifying methylated sequences with higher resolution .

These advanced conjugation methods not only enhance detection sensitivity but also enable novel experimental approaches previously limited by technical constraints.

How can antibodies be utilized in novel therapeutic development workflows?

Antibodies serve as critical tools throughout the therapeutic development pipeline:

In cancer research, therapeutic antibodies targeting death receptors show promising results. The DR5 monoclonal antibody (D-6) demonstrates significant apoptosis-inducing effects on ovarian cancer cells, particularly when combined with cisplatin chemotherapy . This synergistic effect is evidenced by:

  • Increased cell growth inhibition rates in combination treatment

  • Higher apoptosis rates compared to single-agent treatment

  • Reduced expression of caspase-3, 8, and 9 precursors in cells treated with both agents

  • Significant morphological changes indicating enhanced cell death

These findings suggest combination therapies utilizing antibodies may overcome resistance mechanisms in cancer treatment.

For antibody engineering applications, computational approaches like FlowDesign leverage flow matching to enhance antibody complementarity-determining region (CDR) design. This approach offers:

  • Flexible selection of prior distributions

  • Direct matching of discrete amino acid distributions

  • Enhanced computational efficiency for large-scale sampling

When applied to HIV-1 cellular receptor CD4 targeting, this methodology generated antibodies with improved binding affinity and neutralizing potency compared to the state-of-the-art HIV antibody Ibalizumab across multiple HIV mutants , demonstrating how computational antibody design can accelerate therapeutic development.

What considerations are important when designing validation studies for antibodies targeting post-translational modifications?

Validation of antibodies targeting post-translational modifications (PTMs) requires specialized approaches:

  • Modified versus unmodified control samples:

    • Generate samples with and without the modification through:

      • Phosphatase/deacetylase treatment to remove modifications

      • Site-directed mutagenesis (e.g., replacing modifiable residues)

      • In vitro enzymatic modification of recombinant proteins

  • Quantitative validation metrics:

    • Evaluate PTM-specific antibody performance using:

      • Signal-to-noise ratios across modification states

      • Dynamic range of detection across modification levels

      • Cross-reactivity profiles with similar modifications

  • Context-dependent epitope accessibility:

    • PTM accessibility may vary with:

      • Protein conformation changes

      • Interaction with binding partners

      • Subcellular localization

  • Complementary method verification:

    • Confirm antibody specificity using orthogonal approaches:

      • Mass spectrometry to verify modification sites

      • Targeted mutagenesis combined with functional assays

      • Correlation with known modification-dependent functions

  • Stoichiometry considerations:

    • Determine whether the antibody can detect varying levels of modification or only binary (present/absent) states

    • Calibrate detection limits using standards with known modification stoichiometry

When applying novel methods like CUT&Tag to study chromatin modifications, researchers must carefully consider how native conditions affect epitope availability and whether the Tn5 transposase bias toward open chromatin might skew results for certain modifications associated with repressed chromatin .

How might emerging computational approaches improve antibody specificity and application versatility?

Computational methodologies are transforming antibody research through several innovative approaches:

The development of data-driven structural models as informative priors represents a significant advancement in antibody design. As demonstrated in the FlowDesign approach, these models outperform conventional methods across diverse metrics including Amino Acid Recovery (AAR), RMSD, and Rosetta energy calculations . This computational approach enables:

  • Structure-guided optimization:

    • Simultaneous optimization of sequence and structure using flow matching techniques

    • Flexible prior distribution selection that incorporates existing structural knowledge

    • Direct matching of discrete amino acid distributions for improved sequence predictions

  • Enhanced predictive modeling:

    • Better prediction of antibody-antigen interactions through improved complementarity-determining region (CDR) design

    • Reduced experimental iterations through more accurate in silico screening

    • Ability to model complex antibody features beyond binding affinity

  • Application expansion through hybrid approaches:

    • Integration of computational design with high-throughput screening

    • Combination of multiple computational methods to address different aspects of antibody function

    • Platform technologies that bridge computational prediction with experimental validation

These approaches significantly reduce the resources required for antibody development while improving specificity, affinity, and functionality for challenging targets.

What are the methodological frontiers for antibody-based chromatin profiling techniques?

The field of antibody-based chromatin profiling is rapidly evolving beyond traditional ChIP-Seq:

CUT&Tag represents a significant methodological advance that addresses key limitations of ChIP-Seq, including sample input requirements and workflow efficiency . Future directions include:

  • Multimodal chromatin profiling:

    • Integration of CUT&Tag with other genomic assays for simultaneous profiling of multiple chromatin features

    • Development of antibody panels for comprehensive epigenomic characterization

    • Single-cell adaptations enabling cell-type-specific chromatin landscape analysis

  • Enhanced target specificity approaches:

    • Direct conjugation of Tn5 transposase to primary antibodies for improved signal-to-noise ratios

    • Application of recombinant antibody technologies (like AbFlex®) with site-specific transpeptidase Sortase recognition sequences

    • Multiplexed tagmentation systems to investigate multiple factors simultaneously

  • Cross-platform standardization efforts:

    • Development of reference materials and controls to enable data comparison across methodologies

    • Benchmarking against established ENCODE datasets to ensure compatibility with existing resources

    • Protocol optimization for challenging targets such as transcription factors with weak or transient binding

  • Extended applications beyond histone modifications:

    • Adaptation of antibody-directed tagmentation for DNA methylation studies (MeDIP)

    • Integration with chromatin accessibility assays for comprehensive regulatory element characterization

    • Application to non-histone chromosomal proteins and chromatin remodeling complexes

These methodological advances continue to expand our ability to investigate the complex interplay between chromatin structure, epigenetic modifications, and gene regulation with unprecedented resolution and efficiency.

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.