ATHB-22 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Context of ATHB-22

ATHB-22 refers to Arabidopsis thaliana Homeobox-leucine zipper protein ATHB-22, a plant-specific transcription factor involved in developmental regulation. It is listed as a recombinant protein product (UniProt ID: Q4PSR7) with the following characteristics:

  • Product Code: CSB-EP686902DOA-B

  • Storage: Stable for 6–12 months at -20°C/-80°C in lyophilized or liquid form .

This protein is unrelated to antibodies and instead belongs to the homeodomain transcription factor family, which regulates gene expression in plants.

Clarification of Terminology

The term "ATHB-22 Antibody" does not appear in peer-reviewed literature or immunological databases within the provided sources. Notably:

  • CD22-targeted antibodies (e.g., epratuzumab, SM03) are well-documented in B-cell immunology .

  • Antibody structure (e.g., Fab, Fc domains, CDRs) is extensively described in general antibody research .

Potential Misinterpretation

If the query intends to refer to anti-CD22 antibodies (a validated therapeutic target in B-cell malignancies and autoimmune diseases), key findings from the search results include:

Table 1: Anti-CD22 Antibody Mechanisms

Antibody NameTargetMechanism of ActionClinical Relevance
EpratuzumabCD22 (B cells)Induces phosphorylation of CD22 ITIM motifs, inhibits BCR signaling SLE, RA trials
SM03CD22 (B cells)Converts cis- to trans-binding of CD22, enhances SHP-1 activity, reduces NF-κB Phase III RA trials

Table 2: Antibody Engineering Features

FeatureDescription
Fab DomainContains variable regions (CDRs) for antigen binding; determines specificity
Fc DomainMediates effector functions (e.g., complement activation, phagocytosis)
GlycosylationImpacts stability and effector functions; varies by isotype

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
ATHB-22 antibody; At2g36610 antibody; F13K3.1 antibody; F1O11.24 antibody; Homeobox-leucine zipper protein ATHB-22 antibody; HD-ZIP protein ATHB-22 antibody; Homeodomain transcription factor ATHB-22 antibody
Target Names
ATHB-22
Uniprot No.

Target Background

Function
Putative transcription factor.
Database Links

KEGG: ath:AT2G36610

STRING: 3702.AT2G36610.1

UniGene: At.53052

Protein Families
HD-ZIP homeobox family, Class I subfamily
Subcellular Location
Nucleus.
Tissue Specificity
Expressed in siliques.

Q&A

How should researchers assess antibody specificity for targets like ATHB-22?

Antibody specificity assessment requires a multi-faceted approach. Researchers should implement at least two of the "five pillars" of antibody characterization: genetic strategies, orthogonal strategies, multiple antibody strategies, recombinant strategies, and immunocapture MS strategies . For genetic strategies, comparing signals between wildtype samples and those where the target gene (like ATHB-22) has been knocked out provides strong evidence of specificity. Western blots using lysates from both sample types should show band presence in wildtype and absence in knockout samples. Orthogonal strategies involve comparing antibody-based detection with antibody-independent methods like mass spectrometry or RNA-seq to confirm target expression patterns match across methodologies .

What information should be recorded when validating antibodies for research applications?

When validating antibodies, researchers must document: (1) evidence that the antibody binds to the intended target (like ATHB-22); (2) confirmation that binding occurs in complex protein mixtures (cell lysates or tissue sections); (3) evidence that the antibody does not cross-react with other proteins; and (4) verification that the antibody performs as expected under the specific experimental conditions used . This documentation should include positive and negative controls, validation method details, and the exact experimental conditions used. Studies indicate that approximately 50% of commercial antibodies fail to meet basic characterization standards, resulting in significant financial losses and potentially unreliable research results .

How does the generation method of an antibody affect its research applications?

The generation method significantly impacts antibody performance and reliability. Monoclonal antibodies offer higher specificity and reproducibility compared to polyclonal antibodies, as demonstrated by comparative characterization studies using knockout cell lines . Recombinant antibodies provide even greater consistency between batches and experiments. For research applications requiring high specificity, such as detecting low-abundance proteins like transcription factors, recombinant antibodies generated through bacterial expression of antigens and high-throughput screening for affinity provide superior performance . The Protein Capture Reagents Program (PCRP) has generated 1,406 monoclonal antibodies using these approaches, targeting 737 human proteins, including many transcription factors .

What strategies can researchers employ when working with antibodies showing batch-to-batch variability?

Batch-to-batch variability presents a significant challenge in antibody-based research. To address this issue, researchers should implement a systematic approach:

  • Pre-experiment validation: For each new batch, perform side-by-side comparison with previous batches using identical samples and protocols

  • Internal standards: Include consistent positive and negative controls in each experiment

  • Recombinant alternatives: Consider switching to recombinant antibodies, which demonstrate significantly less batch-to-batch variability than traditional polyclonal antibodies

  • Multiple antibody approach: Use multiple antibodies targeting different epitopes of the same protein to confirm results

  • Documentation: Maintain detailed records of batch numbers and validation results to track performance variations over time

For critical experiments, researchers should consider performing antibody characterization using knockout controls even when vendors provide their own validation data, as the specific experimental conditions may affect antibody performance .

How can computational approaches and databases enhance antibody selection and characterization?

Computational approaches have revolutionized antibody research through several mechanisms:

Computational ResourceApplication in Antibody ResearchBenefit to Researchers
Structural Antibody Database (SAbDab)Provides structural information for antibody-antigen complexesAids epitope prediction and antibody engineering
Therapeutic Structural Antibody Database (Thera-SAbDab)Tracks antibody therapeutics with corresponding structuresEnables structure-based design of research antibodies
Deep generative modelsDe novo design of antibodies against specific targetsCreates diverse binders with unique sequences and conformations
Recombinant Antibody NetworkRepository of well-characterized recombinant antibodiesProvides access to renewable antibody resources

Researchers can leverage these resources by: (1) using structural databases to identify antibodies with similar binding properties to their target; (2) employing sequence analysis to predict cross-reactivity; and (3) utilizing computational design tools to enhance specificity and affinity. For example, deep generative models have successfully designed antibodies with nanomolar affinities against specific targets like HER2 , suggesting similar approaches could be applied to other targets.

What methodological approaches are recommended when studying low-abundance proteins with antibodies?

Low-abundance proteins present significant detection challenges. Recommended methodological approaches include:

  • Signal amplification techniques: Implement tyramide signal amplification or proximity ligation assays to enhance detection sensitivity

  • Sample enrichment: Use immunoprecipitation to concentrate the target protein before analysis

  • Specific cell types: Identify and isolate cell populations with higher target expression

  • Recombinant expression strategies: Employ in vitro expression systems to validate antibody binding against the purified target protein

  • Multiple detection methods: Combine antibody-based detection with mass spectrometry to confirm results

When selecting antibodies for low-abundance targets, prioritize those validated using knockout controls and showing specificity in complex protein mixtures. The Antibody Characterization Laboratory (ACL) has developed nearly 1,000 antibodies targeting 570 antigens, many of which are suitable for detecting low-abundance proteins .

How should researchers address immunogenicity concerns when using antibodies in long-term experiments?

Immunogenicity (the production of antibodies against the research antibody) can compromise experimental results, particularly in longitudinal studies. To address this concern:

  • Monitor anti-drug antibody (ADA) development: Regularly test for antibodies that bind to the research antibody

  • Document baseline reactivity: Establish pre-existing antibody levels before initiating experiments

  • Consider antibody engineering: Use humanized antibodies or fragments to reduce immunogenicity

  • Analyze impact on results: Assess whether ADAs correlate with changes in experimental outcomes

Clinical studies of therapeutic antibodies provide valuable insights into immunogenicity patterns. For example, in Vyvgart (efgartigimod alfa) trials, pre-existing antibodies were detected in 15% of patients, while treatment-induced antibodies developed in 21% of treated individuals . Importantly, neutralizing antibodies were detected in 7% of patients . These patterns suggest researchers should anticipate similar issues in experimental systems and implement appropriate controls.

What strategies can resolve contradictory results when different antibodies targeting the same protein yield inconsistent findings?

Contradictory results from different antibodies are a common challenge. Systematic resolution involves:

  • Epitope mapping: Determine which regions of the target protein each antibody recognizes

  • Protein isoform analysis: Investigate whether different antibodies detect different isoforms or post-translational modifications

  • Validation hierarchy: Establish a validation hierarchy using genetic approaches as the gold standard:

    • Results confirmed with knockout/knockdown controls

    • Results obtained with multiple independent antibodies

    • Results verified with orthogonal (non-antibody) methods

  • Context-dependent specificity: Evaluate whether inconsistencies arise from context-dependent binding properties of the antibodies

When different antibodies yield conflicting results, researchers should not simply select the antibody that confirms their hypothesis. Instead, they should systematically investigate the underlying causes of the discrepancy and report all findings transparently in publications.

How can researchers optimize antibody-based protocols for challenging sample types or fixation conditions?

Optimizing antibody-based protocols for challenging samples requires systematic methodology:

  • Epitope accessibility assessment: Different fixation methods can mask epitopes. Compare multiple fixation protocols (PFA, methanol, acetone) to determine optimal epitope preservation

  • Antigen retrieval optimization: For fixed tissues, test various antigen retrieval methods (heat-induced, enzymatic, pH variations) systematically

  • Signal-to-noise optimization matrix:

ParameterVariable RangeOptimization Approach
Antibody concentration0.1-10 μg/mlTitration series with consistent incubation time
Incubation time1 hour to overnightTime course with consistent antibody concentration
Blocking reagentsBSA, serum, caseinCompare different blockers at 1-5% concentration
Detergent concentration0.05-0.3% Triton/TweenAssess permeabilization efficacy vs. background
  • Sample-specific controls: Include tissue-matched or cell-type-matched negative controls (ideally knockout samples) to establish background levels

Researchers should recognize that antibody characterization is context-dependent, and validation performed in one experimental system may not translate to others . Therefore, optimization must be performed for each specific application and sample type.

What minimum information should researchers include in publications to ensure antibody-based experiments are reproducible?

To ensure reproducibility, publications should include:

  • Antibody identifier information:

    • Research Resource Identifier (RRID) for each antibody

    • Vendor name and catalog number

    • Clone designation for monoclonal antibodies

    • Lot number (particularly important for polyclonal antibodies)

  • Validation evidence:

    • Methods used to validate specificity (e.g., knockout controls, multiple antibodies)

    • Whether validation was performed in-house or by vendor

    • How validation conditions relate to experimental conditions

  • Detailed methodology:

    • Precise antibody concentration

    • Incubation conditions (time, temperature, buffer composition)

    • Detection system specifications

    • Image acquisition parameters

  • Control experiments:

    • Positive and negative controls used

    • Technical and biological replication details

Including this information addresses the "antibody characterization crisis" that contributes to irreproducible research costing billions of dollars annually .

How can researchers effectively transition between different antibody formats while maintaining consistent results?

Transitioning between antibody formats (e.g., from research-grade to therapeutic-grade, or between different fragment types) requires systematic comparison:

  • Sequential characterization protocol:

    • Begin with side-by-side binding assays (ELISA, SPR) to compare affinities

    • Progress to functional assays relevant to research question

    • Conduct epitope binning to confirm similar binding sites

    • Perform cross-competition assays to assess epitope overlap

  • Format-specific considerations:

    • When transitioning from full antibodies to fragments (Fab, scFv), expect potential avidity effects

    • For recombinant reformatting, verify sequence identity between formats

    • When moving between species variants (e.g., mouse to humanized), assess potential epitope shifts

  • Bridging studies design:

    • Include both formats in parallel experiments during transition period

    • Establish conversion factors for quantitative applications

    • Document any systematic differences in sensitivity or specificity

These approaches mirror strategies used in therapeutic antibody development, where reformatting from discovery platforms to therapeutic formats requires careful characterization .

What approaches can researchers use to manage antibody degradation and maintain long-term consistency in experiments?

Antibody degradation can compromise experimental consistency over time. Evidence-based management strategies include:

  • Storage optimization based on comparative stability studies:

    • Store antibodies in small aliquots to minimize freeze-thaw cycles

    • For long-term storage, maintain at -80°C rather than -20°C

    • For working solutions, add stabilizing proteins (0.1-1% BSA)

    • Consider preservatives for refrigerated storage (0.02% sodium azide)

  • Quality control program:

    • Implement regular testing against reference standards

    • Use functional assays rather than just concentration measurements

    • Document batch performance over time to identify degradation patterns

    • Establish acceptance criteria for continued use

  • Stability indicators:

    • Monitor for aggregation using dynamic light scattering

    • Assess fragmentation by SDS-PAGE

    • Evaluate binding activity using consistent positive controls

    • Check for precipitation or color changes before use

Implementing these practices creates a systematic approach to maintaining antibody quality throughout the research lifecycle, supporting experimental consistency and reproducibility.

How are artificial intelligence and machine learning transforming antibody development and characterization?

Artificial intelligence and machine learning are revolutionizing antibody research through several innovative approaches:

  • De novo design capabilities: Deep generative models can now design novel antibody complementarity-determining regions (CDRs) with specific binding properties. For example, IgMPNN and MaskedDesign models have successfully generated antibodies with nanomolar affinities against targets like HER2 .

  • Structure prediction advancements: AI models can predict antibody-antigen complex structures with increasing accuracy, facilitating rational design approaches and epitope prediction.

  • Screening optimization: Machine learning algorithms can identify optimal candidates from large antibody libraries, reducing experimental burden while increasing success rates.

  • Characterization automation: AI-powered image analysis can standardize and accelerate antibody validation in techniques like immunohistochemistry and immunofluorescence.

These technologies are enabling zero-shot generation of antibodies—creating novel binders without requiring existing antibodies against the target as design templates . As these approaches mature, they will likely reduce reliance on traditional immunization methods while expanding the diversity and specificity of available research antibodies.

What emerging standards and initiatives are addressing the "antibody characterization crisis" in scientific research?

The scientific community has recognized the "antibody characterization crisis" and is developing standards and initiatives to address it:

  • YCharOS initiative: This organization performs independent characterization of antibodies using knockout cell lines, providing unbiased validation data to the research community .

  • International Working Group for Antibody Validation: This group established the "five pillars" framework for antibody characterization, providing methodological guidance for validation efforts .

  • Research Resource Identifier (RRID) program: This initiative creates unique identifiers for antibodies, enhancing traceability and supporting reproducibility .

  • Journal reporting requirements: Leading journals are implementing increasingly stringent requirements for antibody characterization information in manuscripts.

  • Antibody registry databases: Resources like Antibody Registry, Antibodypedia, and SAbDab provide centralized information on antibody characteristics and applications .

These initiatives collectively work to establish standards for antibody characterization, improve documentation practices, and enhance the reliability of antibody-based research across scientific disciplines.

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.