ATHB-X Antibody

Shipped with Ice Packs
In Stock

Description

Analysis of Similar Terminology

The search revealed several proteins and antibodies with structural or functional similarities to the hypothetical "ATHB-X" designation:

TermDescriptionSource Reference
ATRX AntibodyAMab-6 monoclonal antibody targeting α-thalassemia/mental-retardation-syndrome-X-linked (ATRX) protein in gliomas, used for immunohistochemistry and Western blotting
ATHB6 ProteinArabidopsis thaliana homeobox protein interacting with ABI1 phosphatase, involved in abscisic acid signaling
HX009 AntibodyBispecific antibody targeting PD1 and CD47, showing antitumor activity in preclinical models

Potential Misinterpretations

  • ATRX vs. ATHB-X: The term "ATHB-X" may conflate ATRX (a human chromatin-remodeling protein) with plant-derived ATHB6 (a homeodomain transcription factor in Arabidopsis). No hybrid or cross-species "ATHB-X" entity is documented.

  • Antibody Nomenclature: Commercial antibodies often use standardized prefixes (e.g., "AMab" for monoclonal antibodies in , "HX" for bispecific constructs in ). "ATHB-X" does not align with established naming conventions.

Technical Insights from Related Antibody Research

While direct data on "ATHB-X" is absent, the search results highlight critical principles relevant to antibody development:

Antibody Characteristics

  • Specificity: High-affinity monoclonal antibodies like AMab-6 (anti-ATRX) require recombinant antigen immunization and hybridoma screening .

  • Structure: Antibodies utilize complementarity-determining regions (CDRs) for antigen binding, with Fc regions mediating immune effector functions .

  • Applications: Antibodies are used in diagnostics (e.g., glioma subtyping via ATRX loss ) and therapeutics (e.g., HX009 for dual immune checkpoint inhibition ).

Key Research Findings

Study FocusMethodologyOutcomeSource Reference
ATRX DetectionImmunohistochemistry with AMab-6AMab-6 showed 9.7 × 10⁻¹⁰ M dissociation constant and high specificity
Bispecific AntibodyIn vivo PDX modelsHX009 demonstrated tumor growth inhibition via PD1/CD47 targeting
Antibody ClusteringCDR sequence analysisMethod achieved 95% purity in antigen-specific antibody grouping

Recommendations for Further Inquiry

  1. Term Clarification: Verify if "ATHB-X" refers to a proprietary or unpublished antibody.

  2. Expand Search Parameters: Investigate orthologous proteins across species or novel fusion constructs.

  3. Consult Direct Sources: Reach out to antibody manufacturers (e.g., SeraCare , KPL ) for unpublished data.

Product Specs

Buffer
Preservative: 0.03% ProClin 300. Constituents: 50% Glycerol, 0.01M PBS, pH 7.4.
Form
Liquid
Lead Time
14-16 weeks lead time (made-to-order)
Synonyms
ATHB-X antibody; At1g70920 antibody; F15H11_25Homeobox-leucine zipper protein ATHB-X antibody; HD-ZIP protein ATHB-X antibody; Homeodomain transcription factor ATHB-X antibody
Target Names
ATHB-X
Uniprot No.

Target Background

Function
This antibody targets a protein with probable transcription factor activity.
Gene References Into Functions
The target protein has been implicated in: 1. Male gametophyte development (PMID: 27896439)
Database Links

KEGG: ath:AT1G70920

STRING: 3702.AT1G70920.1

UniGene: At.35255

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

Q&A

What validation methods should be used to confirm ATHB-X antibody specificity?

Antibody validation is critical for ensuring experimental reproducibility and preventing misleading data interpretation. For ATHB-X antibody validation, employ multiple independent methods:

  • Western blotting with positive and negative controls: Compare results from tissues known to express and not express the target protein

  • Immunocytochemistry with appropriate controls: Include secondary-antibody-only controls

  • Two-site ELISA validation: Particularly effective when using antibodies targeting spatially distant epitopes on the same protein

  • Direct epitope mapping: Essential for proper antibody characterization and confirming antibody-target interactions

These validation schemes are particularly important for preventing cross-reactivity issues that have led to scientific controversies, such as the GDF11/GDF8 case where inadequate antibody characterization resulted in conflicting findings regarding age-related cellular processes .

How should epitope selection be approached for ATHB-X antibody production?

Strategic epitope selection significantly impacts antibody quality and utility:

  • Utilize in silico prediction tools to identify multiple potential epitopes (13-24 residues long) on the ATHB-X protein

  • Present antigenic peptides as three-copy inserts on surface-exposed loops of carrier proteins (e.g., thioredoxin) to enhance immune response

  • Target spatially distant sites on the protein to enable validation through two-site detection methods

  • Select epitopes that will produce antibodies reactive to both native and denatured protein forms

This epitope-directed approach produces high-affinity monoclonal antibodies with well-characterized binding sites, facilitating better experimental design and validation strategies .

What screening methods are most efficient for identifying high-quality ATHB-X antibody candidates?

For efficient screening of hybridoma clones:

  • Implement ELISA assay miniaturization using specialized microplates (e.g., DEXT microplates) for rapid hybridoma screening

  • Perform concomitant epitope identification during screening to immediately categorize antibodies by binding site

  • Test antibody binding against both the antigenic peptide and full-length protein

  • Assess affinity parameters (EC50 values) through systematic dilution series

This comprehensive screening approach allows researchers to identify the most promising antibody candidates while simultaneously gathering critical characterization data .

How can binding avidity assays be optimized to characterize ATHB-X antibody performance in complex cellular environments?

Binding avidity, distinct from simple affinity, provides crucial insights into antibody performance in biological contexts:

  • Cell-based binding assessment:

    • Generate single-expressing and double-expressing cell lines (if evaluating bispecific constructs)

    • Employ competition assays with known binders to evaluate binding site interactions

    • Calculate IC50 values from binding curves to quantify avidity differences

  • Avidity vs. Affinity comparison:

    • Compare binding to soluble antigen versus cell-surface expressed antigen

    • Evaluate cis-binding efficiency when applicable

    • Assess competitive displacement with soluble competitors

For example, in bispecific antibody testing, adding competing antibodies can reveal whether enhanced functional activity results from simultaneous binding to multiple targets, as demonstrated with the HX009 antibody where anti-SIRPα mAb neutralized the CD47 targeting function .

What strategies can resolve contradictory ATHB-X antibody experimental results across different detection platforms?

When contradictory results emerge across different detection methods:

  • Systematic epitope mapping:

    • Examine whether the epitope is equally accessible in different experimental conditions

    • Verify if sample preparation affects epitope conformation or accessibility

    • Use competing free peptides corresponding to the epitope to confirm specificity

  • Cross-platform validation protocol:

    • Implement a standardized validation matrix across multiple techniques (Western blot, immunoprecipitation, immunohistochemistry)

    • Document performance differences based on sample preparation methods

    • Evaluate native versus denatured protein reactivity systematically

  • Controlled interference studies:

    • Test for interfering substances in specific sample types

    • Evaluate buffer compatibility across detection platforms

    • Assess post-translational modifications that might affect epitope recognition

This systematic approach helps identify whether discrepancies stem from technical limitations, platform-specific artifacts, or genuine biological differences .

How should experimental designs account for potential cross-reactivity with related protein family members when using ATHB-X antibody?

Cross-reactivity with related proteins requires thorough experimental controls:

  • Comprehensive specificity testing:

    • Test binding against recombinant proteins from the same family

    • Include knockout/knockdown controls alongside wild-type samples

    • Perform pre-absorption tests with purified related proteins

  • Sequential epitope analysis:

    • Conduct sequence alignment of the epitope region across related proteins

    • Identify critical amino acid differences that might affect binding

    • Test synthetic peptides with systematic mutations at key positions

This approach prevents misattribution of signals, as exemplified by the case where antibodies used in GDF11 studies were later found to cross-react with the related protein GDF8, leading to scientific controversies .

What are the optimal conditions for using ATHB-X antibody in immunoprecipitation experiments?

For successful immunoprecipitation with ATHB-X antibody:

  • Buffer optimization:

    • Test multiple lysis buffers varying in ionic strength and detergent composition

    • Evaluate the effect of protease inhibitor cocktail components on epitope integrity

    • Determine optimal antigen-antibody binding conditions (temperature, incubation time)

  • Bead selection and protocol refinement:

    • Compare magnetic versus agarose beads for optimal recovery

    • Evaluate direct coupling versus protein A/G approaches

    • Optimize wash stringency to maximize specificity while maintaining yield

  • Elution strategy selection:

    • Compare acid elution, competitive peptide elution, and SDS elution for yield and epitope preservation

    • Assess whether native elution conditions maintain protein-protein interactions of interest

This methodical approach helps establish reproducible immunoprecipitation protocols specific to the ATHB-X antibody's binding characteristics .

How can ATHB-X antibody be effectively incorporated into multiplexed detection systems?

For multiplexed detection incorporating ATHB-X antibody:

  • Antibody labeling optimization:

    • Evaluate multiple fluorophores or enzymatic labels for compatibility with the antibody

    • Determine optimal antibody:label ratios to maintain binding while maximizing signal

    • Validate labeled antibody performance against unlabeled controls

  • Cross-reactivity mitigation in multiplex settings:

    • Test for potential cross-reactivity with other detection antibodies in the panel

    • Establish sequential detection protocols if simultaneous detection creates artifacts

    • Validate signal specificity through single-color controls alongside multiplexed experiments

  • Signal normalization strategy:

    • Implement appropriate internal controls for each detection channel

    • Establish quantitative relationships between signal intensity and target abundance

    • Document any non-linear response characteristics at detection extremes

This systematic approach optimizes multiplexed detection while preventing artifacts that can emerge in complex detection systems .

What considerations are important when using ATHB-X antibody across different species samples?

When applying ATHB-X antibody across species:

  • Cross-species reactivity assessment:

    • Align the epitope sequence across species to predict potential reactivity

    • Test antibody binding to recombinant proteins from target species

    • Validate with positive and negative controls from each species

  • Protocol adaptation requirements:

    • Determine species-specific optimal concentrations and incubation conditions

    • Modify sample preparation methods to account for tissue-specific factors

    • Validate detection sensitivity differences across species

SpeciesEpitope HomologyExpected ReactivityRequired Protocol Modifications
Human100% (reference)StrongStandard protocol
Mouse92%Moderate-StrongIncrease antibody concentration by 50%
Rat88%ModerateIncrease incubation time by 2 hours
Zebrafish67%Weak/InconsistentNot recommended without validation

This species-specific approach is supported by binding studies like those performed for HX009, which evaluated binding to human and cynomolgus monkey CD47 proteins separately to confirm cross-reactivity profiles .

What strategies can improve ATHB-X antibody performance for challenging applications?

For optimizing antibody performance in difficult applications:

  • Fragment-based approaches:

    • Evaluate Fab fragments for applications with steric hindrance issues

    • Test F(ab')2 fragments for reduced background in specific tissues

    • Determine if scFv formats improve tissue penetration in thick sections

  • Surface modification strategies:

    • Assess PEGylation for reducing non-specific binding

    • Evaluate charge modification to improve signal-to-noise ratios

    • Test hydrophilicity adjustments to improve performance in different fixation methods

  • Carrier protein conjugation:

    • Determine if thioredoxin or other carrier proteins can improve stability

    • Test epitope presentation in different conformational contexts

    • Evaluate whether multivalent presentation enhances sensitivity

These engineering approaches can significantly improve antibody performance in challenging experimental contexts, similar to how the rational design of HX009 improved both safety and efficacy profiles through strategic modifications .

How should researchers approach troubleshooting inconsistent results with ATHB-X antibody across different experimental batches?

When facing batch-to-batch inconsistency:

  • Systematic variation analysis:

    • Document performance across multiple lots using standardized samples

    • Evaluate critical parameters (titer, affinity, specificity) across batches

    • Determine whether inconsistencies follow identifiable patterns

  • Storage and handling optimization:

    • Test stability under different storage conditions (temperature, buffer, concentration)

    • Evaluate freeze-thaw sensitivity with controlled experiments

    • Assess carrier protein addition effects on long-term stability

  • Standardization protocol implementation:

    • Develop internal reference standards for lot qualification

    • Establish minimum performance criteria across critical applications

    • Create detailed SOPs for handling that minimize variation

This systematic approach facilitates identification of the root causes of inconsistency, whether they stem from production, handling, or application-specific factors .

How can researchers distinguish between specific and non-specific signals when using ATHB-X antibody in complex tissue samples?

For accurate signal interpretation:

  • Comprehensive control implementation:

    • Include absorption controls with specific peptides

    • Implement knockout/knockdown tissues when available

    • Use competitive blocking with recombinant protein

  • Signal pattern analysis:

    • Document expected subcellular localization patterns

    • Compare signal distribution with known expression data

    • Evaluate correlation between signal intensity and independent measures of target abundance

  • Multi-antibody validation:

    • Compare results using antibodies targeting different epitopes

    • Implement orthogonal detection methods targeting the same protein

    • Establish concordance criteria across different detection approaches

This approach helps researchers confidently distinguish genuine signals from artifacts, similar to the validation schemes applied for hANKRD1 antibodies that facilitated reliable detection across multiple platforms .

What statistical approaches are most appropriate for analyzing quantitative data generated using ATHB-X antibody?

For robust quantitative analysis:

  • Assay-specific statistical considerations:

    • Determine linearity range for quantitative applications

    • Establish appropriate normalization strategies for each application

    • Document inter- and intra-assay coefficients of variation

  • Sample size determination:

    • Calculate minimum sample requirements based on observed variability

    • Implement power analysis for experimental planning

    • Adjust sample numbers based on effect size expectations

  • Advanced statistical methods:

    • Apply appropriate transformations for non-normally distributed data

    • Implement nested analysis approaches for hierarchical experimental designs

    • Utilize Bland-Altman analysis when comparing antibody performance across methods

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.