ATHB-9 Antibody

Shipped with Ice Packs
In Stock

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
ATHB-9 antibody; HB9 antibody; PHV antibody; At1g30490 antibody; F26G16.11Homeobox-leucine zipper protein ATHB-9 antibody; HD-ZIP protein ATHB-9 antibody; Homeodomain transcription factor ATHB-9 antibody; Protein PHAVOLUTA antibody
Target Names
ATHB-9
Uniprot No.

Target Background

Function
ATHB-9 is a probable transcription factor that plays a crucial role in establishing adaxial-abaxial polarity within ovule primordia. It specifically designates adaxial leaf fates and binds to the DNA sequence 5'-GTAAT[GC]ATTAC-3'.
Gene References Into Functions
  1. A yeast two-hybrid screen using DRN, followed by co-immunoprecipitation and bimolecular fluorescence complementation, identified PHAVOLUTA (PHV) as a protein interaction partner in planta. PMID: 17376809
  2. ATHB-9 exhibits sensitivity to redox conditions. PMID: 17900520
  3. Class III HD-Zip and KANADI genes, including PHAVOLUTA (PHV), are essential for root development. PMID: 15286295
Database Links

KEGG: ath:AT1G30490

STRING: 3702.AT1G30490.1

UniGene: At.27961

Protein Families
HD-ZIP homeobox family, Class III subfamily
Subcellular Location
Nucleus.

Q&A

What is ATHB-9 and why is it significant in plant research?

ATHB-9 (also known as PHV or PHAVOLUTA) is a member of the class III homeodomain-leucine zipper (HD-ZIP III) family of transcription factors in Arabidopsis thaliana. These proteins play crucial roles in plant development, particularly in regulating adaxial-abaxial patterning in lateral organs and meristem formation. Antibodies against ATHB-9 are valuable tools for studying plant developmental biology, specifically for investigating leaf polarity, vascular development, and meristem function. The significance of ATHB-9 lies in its involvement in fundamental developmental processes, making it an important target for researchers studying plant morphogenesis and adaptation mechanisms .

How do ATHB-9 antibodies compare to other plant transcription factor antibodies in terms of specificity?

The specificity of ATHB-9 antibodies must be carefully evaluated in the context of the HD-ZIP III family's high sequence homology. ATHB-9 shares significant structural similarity with other family members (ATHB-8, ATHB-14/PHB, REV, and ATHB-15/CNA), particularly in the homeodomain region. When developing or selecting ATHB-9 antibodies, researchers should target unique peptide sequences, typically from the N or C-terminal regions, to minimize cross-reactivity.

Compared to antibodies against other plant transcription factors, ATHB-9 antibodies often require more rigorous validation due to:

  • High conservation within the HD-ZIP III family

  • Relatively low expression levels in most tissues

  • Potential protein-protein interactions that might mask epitopes

Validation should include western blot comparison using wild-type and athb-9 mutant tissues, along with testing against recombinant ATHB-8 and ATHB-14 proteins to confirm specificity .

What are the optimal sample preparation methods for ATHB-9 antibody applications?

For optimal ATHB-9 protein detection in plant tissues, sample preparation should follow these methodological guidelines:

  • Tissue harvest: Collect young, actively growing tissues (shoot apices, young leaves, developing embryos) where ATHB-9 expression is highest. Flash-freeze samples immediately in liquid nitrogen.

  • Protein extraction buffer optimization:

    • Use a buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% Triton X-100

    • Include protease inhibitor cocktail to prevent degradation

    • Add 10 mM DTT or β-mercaptoethanol as reducing agents

    • Include phosphatase inhibitors if studying phosphorylation states

    • Add 5-10% glycerol to stabilize proteins

  • Nuclear protein enrichment: As ATHB-9 is a transcription factor, nuclear protein extraction protocols yield better results than total protein extractions. This typically involves tissue homogenization followed by filtered centrifugation steps to isolate nuclei before extraction.

  • Fixation for immunohistochemistry: For tissue localization studies, 4% paraformaldehyde fixation followed by careful permeabilization is recommended, with antigen retrieval steps often necessary to expose nuclear epitopes.

These preparation methods significantly enhance detection sensitivity while preserving the native conformation of ATHB-9 protein, critical for antibody recognition and experimental reproducibility.

How should researchers design validation experiments for new ATHB-9 antibodies?

When validating new ATHB-9 antibodies, researchers should implement a comprehensive multi-step approach that addresses the particular challenges of plant transcription factor antibodies. A robust validation protocol should include:

  • Western blot analysis: Compare wild-type, athb-9 knockout mutants, and ATHB-9 overexpression lines to confirm specific detection at the expected molecular weight (approximately 94 kDa). Include recombinant ATHB-9 protein as a positive control.

  • Cross-reactivity assessment: Test against other HD-ZIP III proteins (ATHB-8, ATHB-14) to determine specificity within this closely related family. This is critical because TLR9 signaling has been shown to affect antibody affinity maturation, which could impact the development of highly specific antibodies .

  • Immunoprecipitation validation: Perform IP-MS (immunoprecipitation followed by mass spectrometry) to confirm the antibody captures ATHB-9 and identify any non-specific interactions.

  • Immunolocalization studies: Compare immunohistochemistry or immunofluorescence results with previously published ATHB-9 mRNA expression patterns and GFP-fusion protein localization studies.

  • Peptide competition assay: Pre-incubate the antibody with the immunizing peptide to confirm signal elimination in subsequent applications.

This systematic validation approach ensures that experimental results using ATHB-9 antibodies are reliable and reproducible, particularly important when studying proteins with high homology to related family members .

What are the most effective immunohistochemistry protocols for localizing ATHB-9 in plant tissues?

For optimal ATHB-9 immunolocalization in plant tissues, researchers should follow this specialized protocol that addresses the unique challenges of plant material:

  • Tissue fixation and embedding:

    • Fix tissue in 4% paraformaldehyde in PBS (pH 7.4) for 12-16 hours at 4°C

    • Dehydrate through an ethanol series (30%, 50%, 70%, 85%, 95%, 100%)

    • Clear with a xylene substitute

    • Embed in paraffin or similar medium

    • Section at 6-8 μm thickness

  • Antigen retrieval (critical for nuclear transcription factors):

    • Deparaffinize and rehydrate sections

    • Heat-induced epitope retrieval using 10 mM sodium citrate buffer (pH 6.0) at 95°C for 20-30 minutes

    • Allow slow cooling to room temperature

  • Immunodetection procedure:

    • Block with 2-3% BSA in PBS containing 0.1% Triton X-100 for 1-2 hours

    • Incubate with primary ATHB-9 antibody (1:100-1:500 dilution) overnight at 4°C

    • Wash extensively with PBS (at least 3×15 minutes)

    • Apply fluorescently-labeled or HRP-conjugated secondary antibody (1:200-1:1000) for 1-2 hours

    • For chromogenic detection, develop with DAB substrate

    • Counterstain nuclei with DAPI for fluorescent detection

  • Controls (essential for validation):

    • Include athb-9 mutant tissue sections as negative controls

    • Use pre-immune serum or isotype controls

    • Perform peptide competition assays

This protocol maximizes specific signal while minimizing background, which is particularly important for nuclear transcription factors that may have relatively low expression levels .

What approaches improve ATHB-9 antibody sensitivity in western blot applications?

Enhancing ATHB-9 antibody sensitivity in western blot applications requires several specialized techniques that address the challenges of detecting plant transcription factors:

  • Protein extraction optimization:

    • Implement nuclear extraction protocols to enrich for ATHB-9

    • Include 10 mM N-ethylmaleimide to prevent post-lysis deubiquitination

    • Maintain samples at 4°C throughout the extraction process

  • Electrophoresis considerations:

    • Use gradient gels (4-12% or 4-15%) for improved resolution

    • Reduce sample heating time (65°C for 5 minutes instead of 95°C for 10 minutes)

    • Add 0.5% SDS to the sample buffer to enhance denaturation

  • Transfer parameters:

    • Implement semi-dry transfer at lower amperage over longer periods (6-8 hours)

    • Use PVDF membranes (0.45 μm pore size) for improved protein retention

    • Include 10% methanol in transfer buffer to enhance binding

  • Detection enhancement:

    • Extend primary antibody incubation to overnight at 4°C

    • Apply signal enhancement systems such as biotin-streptavidin amplification

    • Use highly sensitive ECL substrates with longer exposure times

    • Consider tyramide signal amplification for extremely low abundance targets

  • Background reduction:

    • Implement extended blocking (5% milk or BSA for 2+ hours)

    • Add 0.05% Tween-20 to all antibody dilutions

    • Increase wash durations between antibody incubations

These methodological improvements can increase detection sensitivity by 5-10 fold, critical for capturing ATHB-9 expression in tissues where it occurs at low abundance .

How can chromatin immunoprecipitation (ChIP) be optimized for ATHB-9 antibodies?

Optimizing chromatin immunoprecipitation for ATHB-9 requires addressing specific challenges associated with plant transcription factor ChIP experiments:

  • Tissue preparation and crosslinking:

    • Harvest 1-2 grams of young, actively growing tissue

    • Vacuum infiltrate with 1% formaldehyde for precisely 10 minutes

    • Quench with 125 mM glycine for 5 minutes

    • Flash freeze and grind to fine powder in liquid nitrogen

  • Chromatin extraction and sonication:

    • Extract nuclei using a buffer containing 0.25 M sucrose, 10 mM Tris-HCl (pH 8.0), 10 mM MgCl₂, 1% Triton X-100

    • Resuspend in nuclear lysis buffer (50 mM Tris-HCl pH 8.0, 10 mM EDTA, 1% SDS)

    • Sonicate to generate 200-500 bp fragments (typically 15-20 cycles of 30 seconds on/30 seconds off)

    • Verify fragment size by agarose gel electrophoresis

  • Immunoprecipitation optimization:

    • Pre-clear chromatin with protein A/G beads and non-immune IgG

    • Use 5-10 μg of ATHB-9 antibody per sample

    • Extend incubation to 12-16 hours at 4°C with gentle rotation

    • Include 0.1% SDS and 1% Triton X-100 in IP buffer to reduce background

  • Washing and elution:

    • Implement stringent wash steps with increasing salt concentrations

    • Elute at 65°C with fresh elution buffer (1% SDS, 0.1 M NaHCO₃)

    • Reverse crosslinks for 6-8 hours at 65°C

  • Controls and validation:

    • Include input chromatin (non-immunoprecipitated)

    • Perform parallel IgG control immunoprecipitation

    • Use known ATHB-9 binding sites as positive controls

    • Include negative control regions (non-bound genomic regions)

This optimized protocol significantly improves signal-to-noise ratio for ATHB-9 ChIP experiments, enabling reliable identification of direct target genes .

How does antibody affinity maturation impact ATHB-9 antibody development strategies?

Antibody affinity maturation plays a critical role in developing high-quality ATHB-9 antibodies, with recent research providing important insights for optimization strategies:

  • Adjuvant selection considerations:

    • While CpG-based adjuvants (TLR9 agonists) increase antibody titers, they may reduce affinity maturation

    • For ATHB-9 antibodies, developers should consider using alum or other non-TLR9 activating adjuvants when affinity is prioritized over titer

    • Alternatively, a modified immunization schedule using CpG initially followed by non-CpG boosters may balance titer and affinity

  • Immunization strategies:

    • Extending intervals between immunizations to 4-6 weeks rather than 2-3 weeks allows more complete germinal center reactions

    • Lower antigen doses (10-25 μg rather than 50-100 μg) can promote better affinity maturation

    • Multiple small booster immunizations rather than fewer larger ones enhance selection for high-affinity clones

  • B cell screening approaches:

    • Single B cell sorting techniques to isolate ATHB-9-specific B cells

    • Competitive elution strategies during screening to select higher affinity antibodies

    • Sequential screening with decreasing antigen concentrations to identify high-affinity binders

Understanding these mechanisms allows researchers to make informed decisions when developing new ATHB-9 antibodies, balancing between high titer and high affinity based on the intended application .

What are the challenges and solutions for monitoring ATHB-9 protein-protein interactions in vivo?

Investigating ATHB-9 protein-protein interactions in plant systems presents several specific challenges that require specialized methodological approaches:

  • Key technical challenges:

    • Low endogenous expression levels of ATHB-9

    • Transient nature of many transcription factor interactions

    • Nuclear localization complicating extraction conditions

    • Potential artifacts from overexpression systems

    • Limited compatibility of plant systems with mammalian interaction detection methods

  • Proximity-based labeling approaches:

    • BioID or TurboID fusion with ATHB-9 expressed under native promoter

    • Optimization of biotin pulse timing (2-12 hours) for different tissue types

    • Nuclear-targeted controls to filter out common nuclear protein contaminants

    • Streptavidin pull-down followed by mass spectrometry analysis

  • Split-reporter complementation systems:

    • Split-luciferase complementation assays optimized for plant tissues

    • Multicolor BiFC (Bimolecular Fluorescence Complementation) to visualize multiple interaction partners

    • Careful selection of fusion protein orientation to minimize steric hindrance

    • Validation using reversed fusion protein configurations

  • Co-immunoprecipitation optimization:

    • Use of gentle, non-ionic detergents (0.5% NP-40 or Digitonin)

    • Inclusion of protein-protein interaction stabilizers (e.g., disuccinimidyl suberate)

    • Two-step immunoprecipitation with different epitope tags

    • Native gel electrophoresis followed by western blotting to preserve complexes

  • Advanced mass spectrometry approaches:

    • Crosslinking mass spectrometry (XL-MS) to capture transient interactions

    • SILAC or TMT labeling for quantitative interaction analysis

    • Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces

These methodological approaches collectively overcome the challenges of studying ATHB-9 interactions in plant systems, providing complementary data to build comprehensive interaction networks .

How can researchers address non-specific binding issues with ATHB-9 antibodies?

Non-specific binding represents a significant challenge when working with ATHB-9 antibodies in plant systems. Researchers can implement these methodological interventions to improve specificity:

  • Buffer optimization strategies:

    • Increase blocking agent concentration to 5-7% (milk or BSA)

    • Add 0.1-0.3% non-ionic detergents to reduce hydrophobic interactions

    • Include 5-10% normal serum from the secondary antibody host species

    • Add 0.1-0.2 M glycine to reduce ionic interactions

    • Consider adding 1-5% polyethylene glycol to reduce non-specific binding

  • Antibody preparation approaches:

    • Pre-adsorb antibody against plant tissue from athb-9 knockout plants

    • Affinity purify antibody using recombinant ATHB-9 protein

    • Implement IgG purification from crude antisera

    • If polyclonal, consider depleting cross-reactive antibodies using recombinant ATHB-8 and ATHB-14

  • Experimental design modifications:

    • Always include appropriate negative controls (pre-immune serum, isotype controls)

    • Implement peptide competition assays to confirm specificity

    • Use graduated antibody dilutions to determine optimal signal-to-noise ratio

    • Reduce primary antibody incubation time if background is excessive

  • Sample preparation refinements:

    • Extend blocking time to 2-4 hours at room temperature

    • Implement more stringent washing procedures (increased time, detergent concentration)

    • For fixed tissues, extend permeabilization time to ensure complete antibody access

    • Consider alternative fixatives if current protocols yield high background

These approaches systematically address different mechanisms of non-specific binding, from hydrophobic interactions to ionic binding and steric trapping, significantly improving the signal-to-noise ratio in ATHB-9 antibody applications .

What quality control metrics should be applied when evaluating commercial ATHB-9 antibodies?

Researchers should apply rigorous quality control metrics when selecting and validating commercial ATHB-9 antibodies, with an emphasis on documentation, reproducibility, and application-specific performance:

  • Essential documentation verification:

    • Immunogen sequence information (verify uniqueness to ATHB-9 vs. other HD-ZIP III proteins)

    • Host species and antibody type (monoclonal/polyclonal)

    • Validation data against Arabidopsis tissues and recombinant proteins

    • Lot-to-lot consistency reporting

    • Cross-reactivity assessment with ATHB-8 and ATHB-14

  • Basic validation experiments:

    • Western blot showing single band at correct molecular weight (~94 kDa)

    • Signal presence in wild-type and absence in athb-9 knockout plants

    • Peptide competition assay showing signal elimination

    • Immunoprecipitation efficiency quantification (>70% target depletion)

  • Application-specific quality metrics:

    • For immunohistochemistry: signal-to-noise ratio >3:1, expression pattern matching mRNA localization

    • For ChIP applications: enrichment >8-fold at known target loci

    • For co-IP: minimal non-specific binding in IgG controls

    • For ELISA: standard curve linearity (R² > 0.98) and detection limit

  • Advanced quality assessments:

    • Epitope mapping confirmation

    • Affinity determination (ideally K​D < 10 nM for most applications)

    • Specificity index (ratio of specific:non-specific signal)

    • Stability testing under different storage conditions

This comprehensive quality control framework ensures that only antibodies meeting stringent performance criteria are used in ATHB-9 research, improving reproducibility and reliability of experimental results .

How should researchers interpret contradictory results between different ATHB-9 antibody detection methods?

When faced with contradictory results between different ATHB-9 antibody detection methods, researchers should implement a systematic analytical framework:

  • Methodological factors assessment:

    • Epitope accessibility differences between applications (native vs. denatured vs. fixed)

    • Buffer compatibility issues affecting antibody-epitope interactions

    • Sample preparation differences altering protein conformation or epitope exposure

    • Detection sensitivity thresholds varying between methods

  • Antibody characteristics evaluation:

    • Epitope location (N-terminal, C-terminal, internal) and how processing may affect detection

    • Monoclonal antibodies may recognize single epitopes that can be masked in certain applications

    • Polyclonal antibodies may detect multiple epitopes with different accessibilities

    • Clone-specific performance variability in different applications

  • Biological context considerations:

    • Post-translational modifications potentially masking epitopes

    • Protein-protein interactions affecting epitope availability

    • Alternative splicing creating isoform-specific detection patterns

    • Developmental or tissue-specific differences in protein conformation

  • Resolution strategy implementation:

    • Design validation experiments using multiple antibodies targeting different epitopes

    • Employ epitope-tagged ATHB-9 constructs as parallel controls

    • Implement orthogonal detection methods (mass spectrometry, activity assays)

    • Sequence verification of the ATHB-9 gene in study material to confirm conservation of epitopes

These analytical approaches help researchers determine whether discrepancies reflect true biological phenomena or technical artifacts, guiding appropriate experimental modifications and interpretations .

How can computational antibody design approaches be applied to develop next-generation ATHB-9 antibodies?

Computational antibody design represents a promising frontier for developing highly specific ATHB-9 antibodies with enhanced properties:

  • Epitope selection optimization:

    • Computational analysis of ATHB-9 structure identifies optimal epitopes that maximize:

      • Accessibility in native protein conformations

      • Sequence divergence from ATHB-8 and ATHB-14 homologs

      • Stability across various experimental conditions

      • Low probability of post-translational modifications

    • Machine learning algorithms predict epitope immunogenicity and antigenicity

  • Complementarity determining region (CDR) design:

    • De novo CDR design algorithms can create antibody binding regions specifically targeting ATHB-9 epitopes

    • The OptCDR approach selects canonical structure backbones for each CDR from known antibody structures

    • Mixed-integer linear programming optimization selects optimal canonical structures

    • Computational interaction energy calculations guide affinity maturation

  • Framework selection for stability:

    • Germline framework selection based on stability, expression level, and compatibility with designed CDRs

    • Energy minimization to refine structure and maximize predicted interactions with target epitopes

    • Computational screening for potential aggregation hotspots

  • Validation before synthesis:

    • Molecular dynamics simulations to assess binding stability and specificity

    • In silico affinity maturation to optimize binding kinetics

    • Cross-reactivity prediction against related proteins (ATHB-8, ATHB-14)

This computational approach has shown promise in designing antibodies with predefined specificities, as demonstrated by the successful design of antibodies against the FLAG peptide . Applied to ATHB-9, these approaches could yield antibodies with superior specificity, affinity, and application versatility compared to traditional immunization methods.

What new insights into ATHB-9 function could be gained through hybrid immunity approaches?

Hybrid immunity approaches—combining natural infection and vaccination principles—can provide novel strategies for ATHB-9 antibody development and functional characterization:

  • Dual-stimulus immunization strategies:

    • Initial immunization with recombinant ATHB-9 protein followed by DNA vaccine encoding ATHB-9

    • Alternating between different ATHB-9 epitopes to create broader recognition profiles

    • Combining different adjuvants to stimulate complementary immune responses

    • This approach mirrors findings from SARS-CoV-2 research showing superior antibody responses from hybrid immunity

  • Advanced epitope targeting:

    • Immunizing with multiple distinct domains of ATHB-9 sequentially

    • Targeting conserved functional domains and unique regions simultaneously

    • Creating antibody panels that collectively recognize different ATHB-9 conformational states

    • Developing antibodies specific to ATHB-9 post-translational modifications

  • Functional blocking antibody development:

    • Targeting ATHB-9 DNA-binding domain to block transcriptional activity

    • Developing antibodies against protein-protein interaction interfaces

    • Creating conditional antibodies activated only under specific cellular conditions

    • Designing bispecific antibodies that recognize ATHB-9 and interacting partners simultaneously

  • Dynamic cellular analysis applications:

    • Intrabody development for in vivo tracking of ATHB-9 in living plant cells

    • Antibody-based biosensors detecting ATHB-9 conformational changes

    • FRET-paired antibodies for studying ATHB-9 protein interactions in real-time

    • Nanobody-based approaches for improved intracellular detection

These hybrid approaches could overcome limitations of traditional antibody development methods, leading to more versatile tools for studying ATHB-9 function in diverse experimental contexts. The application of these strategies could reveal new aspects of ATHB-9's role in plant development and stress responses .

How might single-cell techniques with ATHB-9 antibodies revolutionize plant developmental biology research?

The integration of ATHB-9 antibodies with emerging single-cell technologies promises to transform our understanding of plant development at unprecedented resolution:

  • Single-cell proteomics applications:

    • ATHB-9 antibody-based CyTOF (mass cytometry) for simultaneous detection of multiple proteins

    • Spatial proteomics using ATHB-9 antibodies for protein localization at subcellular resolution

    • Correlation of ATHB-9 protein levels with other transcription factors at single-cell level

    • These approaches could reveal cell-type specific ATHB-9 expression patterns previously masked in whole-tissue analyses

  • Spatial transcriptomics integration:

    • Combined ATHB-9 protein immunodetection with spatial transcriptomics

    • Correlation between ATHB-9 protein localization and downstream target gene expression

    • Identification of cell-specific ATHB-9 regulatory networks

    • Mapping developmental trajectories based on ATHB-9 expression dynamics

  • Live-cell imaging innovations:

    • Development of non-perturbing ATHB-9 antibody fragments for live imaging

    • Single-molecule tracking of ATHB-9 dynamics during cell division and differentiation

    • FRAP (Fluorescence Recovery After Photobleaching) analysis of ATHB-9 mobility in different cell types

    • Optogenetic control of ATHB-9 function using antibody-based targeting

  • Microfluidic applications:

    • Droplet-based single-cell sorting using ATHB-9 antibodies

    • Microfluidic antibody capture for quantifying ATHB-9 secretion

    • Organ-on-chip models for studying ATHB-9 function in controlled microenvironments

    • High-throughput screening of compounds affecting ATHB-9 expression or activity

These integrative approaches would enable researchers to map the spatiotemporal dynamics of ATHB-9 function at single-cell resolution, potentially revealing new mechanisms in cell fate determination, tissue patterning, and plant organ development .

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.