AUG4 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to AUG4 Antibody

AUG4 antibody targets the AUG4 antigen, a high-prevalence glycoprotein component of the Augustine blood group system (ISBT System 36). This system involves four antigens (AUG1–AUG4) expressed on equilibrative nucleoside transporter 1 (ENT1), a transmembrane protein critical for nucleoside transport in erythroid cells . AUG4 antibodies are clinically significant in transfusion medicine and obstetrics due to their potential to trigger hemolytic reactions .

Antigen Characteristics

AntigenPrevalenceAllele VariantKey Features
AUG1HighSplice siteMost common
AUG2HighGlu391LysAlso termed At<sup>a</sup>
AUG3LowThr387ProRare variant
AUG4HighAsn81SerAssociated with transfusion reactions

The AUG:1,2,4 phenotype (co-expressing AUG1, AUG2, and AUG4) is predominant in most populations .

Clinical Significance of AUG4 Antibodies

AUG4 antibodies are immunoglobulin G (IgG) class antibodies that develop following exposure to incompatible RBCs via transfusion or pregnancy . Key clinical implications include:

  • Hemolytic Transfusion Reactions: AUG4 antibodies can cause acute hemolysis upon re-exposure to AUG4-positive blood .

  • Hemolytic Disease of the Fetus and Newborn (HDFN): Maternal AUG4 antibodies may cross the placenta, leading to fetal anemia .

  • Null Phenotype Risks: Individuals lacking ENT1 (AUG-null) exhibit ectopic calcification and abnormal RBCs due to disrupted adenosine transport .

Research Findings and Case Reports

  • First Identification: The first AUG4 antibody was reported in 1995 in a Caucasian woman with prior pregnancy and transfusion history .

  • Functional Insights: ENT1, the carrier protein for AUG4, facilitates adenosine uptake during RBC differentiation. Dysregulation of this transporter in AUG-null individuals underscores its role in cellular metabolism .

  • Prevalence Data: AUG4 antibodies are rare but clinically critical; screening for Augustine antigens is recommended in transfusion-dependent patients .

Diagnostic and Therapeutic Considerations

  • Antibody Detection: AUG4 antibodies are identified via indirect antiglobulin testing (IAT) using antigen-negative RBC panels .

  • Transfusion Management: AUG4-negative blood is required for sensitized individuals to prevent hemolysis .

  • Research Gaps: Further studies are needed to elucidate the structural epitopes of AUG4 and optimize compatibility protocols .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
AUG4 antibody; At1g50710 antibody; F17J6.23 antibody; F4M15.6 antibody; AUGMIN subunit 4 antibody
Target Names
AUG4
Uniprot No.

Target Background

Function
Plays a critical role in microtubule reorganization during spindle and phragmoplast development.
Database Links

KEGG: ath:AT1G50710

STRING: 3702.AT1G50710.1

UniGene: At.37974

Protein Families
HAUS4 family
Subcellular Location
Cytoplasm, cytoskeleton, spindle. Cytoplasm, cytoskeleton, phragmoplast.

Q&A

What factors determine antibody specificity in experimental applications?

Antibody specificity is determined by both molecular characteristics of the immunoglobulin and those of the antigen, particularly the epitope's folding state. Specificity can be compromised when the epitope becomes masked or altered due to post-translational modifications or changes introduced during sample preparation. Therefore, antibody-antigen interactions are strictly context-dependent, varying across experimental conditions .

Methodologically, researchers should:

  • Verify epitope accessibility in your experimental conditions

  • Consider potential post-translational modifications that might affect recognition

  • Evaluate antibody performance in the specific assay format you intend to use

  • Test antibodies against both positive and negative controls to confirm specificity

How can researchers identify duplicate antibody products across different vendor catalogs?

Antibodies from original manufacturers are frequently rebranded and may appear in several catalogs, making it challenging to identify identical products marketed under different names. This issue is compounded when vendors add their own data or customer data and alter the product name, creating multiple identities for the same antibody .

To address this challenge:

  • Cross-reference antibody clone numbers and immunogen sequences

  • Compare product datasheets for similarities in validation data and images

  • Use resources like CiteAb or Antibodypedia that track antibody relationships

  • Contact vendors directly to inquire about the original manufacturer

  • Maintain detailed records of antibody performance in your specific applications

What is the significance of antibody validation in research reproducibility?

The most frequent mistake in antibody research is failure to experimentally verify specificity before use. Many researchers assume vendor reputation is sufficient assurance of performance, leading to widespread use of cross-reactive antibodies, inaccurate data, wasted resources, and delayed scientific progress .

A robust validation approach includes:

Validation MethodPurposeAppropriate Controls
Western blottingConfirm molecular weight and specificityPositive and negative cell/tissue lysates
ImmunoprecipitationVerify ability to bind native proteinIsotype control antibodies
ImmunohistochemistryAssess tissue distribution patternsKnown positive/negative tissues
Knockout/knockdownDefinitive specificity testCells with target gene deleted/silenced
Orthogonal targetingCorrelation with independent methodsAlternative detection methods

The impact of poorly validated antibodies extends beyond individual experiments to invalidate costly clinical trials when such reagents are used as biomarker detection tools .

How do structural design considerations impact antibody developability profiles?

Antibody developability profiles are determined by a combination of physicochemical properties that can be assessed through high-throughput biophysical assays. Analysis of 152 human or humanized monoclonal antibodies has established correlations between different properties and downstream process parameters .

Key structural considerations include:

Early assessment of these properties allows for engineering modifications without affecting program timelines, enabling elimination of antibodies with suboptimal characteristics early in the selection process .

What computational approaches are most effective for designing self-assembling protein nanoparticles for antibody presentation?

De novo design of protein nanoparticles offers a systematic approach to investigate how antigen presentation geometry influences immune response. Computational approaches have successfully created trimers tailored for antigen fusion, featuring N-terminal helices positioned to match C-termini of viral glycoproteins .

The methodology involves:

  • Designing trimeric protein scaffolds using Rosetta software

  • Optimizing interface residues to promote specific assembly geometries

  • Incorporating flexible linkers between the scaffold and antigen

  • Simulating assembled structures using molecular dynamics

  • Evaluating energetic stability of the designed assemblies

This approach has been validated for presenting viral glycoproteins including HIV-1 Env, influenza hemagglutinin, and RSV F trimers in predicted geometries. Electron microscopy confirmed that these designed nanoparticles presented antigenically intact prefusion viral glycoproteins .

How does the Assisted Design of Antibody and Protein Therapeutics (ADAPT) platform enhance single-domain antibody efficacy?

The ADAPT platform interleaves computational predictions with experimental testing to guide affinity maturation. Originally validated for monoclonal antibodies, this approach has been extended to single-domain antibodies (sdAbs), demonstrating significant improvements in binding affinity and neutralization capacity .

In the case study of camelid sdAb A26.8 targeting Clostridium difficile toxin A:

  • ADAPT-guided mutations improved binding affinity by one order of magnitude (reaching 2 nM KD)

  • The best binding mutants maintained or improved stability relative to the parent sdAb

  • Affinity improvement generated a 6-fold enhancement in toxin neutralization efficacy

  • The designed mutations introduced novel electrostatic interactions with the antigen

  • Near-complete additivity of mutation effects was observed, except for adjacent positively charged residues

This methodology demonstrates how computational design can guide the improvement of therapeutic antibody fragments through rational mutation strategies.

What high-throughput developability workflows can predict antibody manufacturability challenges?

An integrated high-throughput developability workflow can accelerate candidate selection and reduce risks in development by ensuring only robust antibody molecules progress to development activities. The workflow involves analyzing key biophysical properties that correlate with downstream process parameters .

The recommended methodology includes:

  • In silico analysis of antibody sequences for liability motifs

  • High-throughput biophysical characterization assays:

    • Thermal stability (Tm, Tagg)

    • Colloidal stability (kD, zeta potential)

    • Chemical stability (oxidation, deamidation susceptibility)

    • Self-interaction propensity (cross-interaction chromatography)

    • Viscosity prediction at high concentrations

  • Data management systems to track and correlate properties

  • Iterative engineering to address identified issues

This approach enables the identification of candidates with optimal properties for downstream processing, including storage stability, viral inactivation susceptibility, chromatographic yield, and performance in high-concentration preparations .

How can researchers ensure antibody validation across different experimental contexts?

Antibody validation must be context-specific, as performance can vary dramatically across different applications. The specificity of an antibody in one experimental setting does not guarantee its specificity in another .

A comprehensive validation approach includes:

  • Application-specific validation:

    • For Western blotting: Verify band size, test knockout controls

    • For immunohistochemistry: Compare staining with known expression patterns

    • For flow cytometry: Compare with alternative antibody clones

    • For ELISA: Test for cross-reactivity with structurally similar proteins

  • Sample preparation validation:

    • Test effect of different fixation methods on epitope recognition

    • Evaluate impact of denaturing vs. native conditions

    • Assess influence of blocking reagents on background

  • Concentration optimization:

    • Perform titration experiments to determine optimal working concentration

    • Evaluate signal-to-noise ratio across concentration range

  • Batch-to-batch validation:

    • Compare new lots with previously validated lots

    • Document performance metrics for reference

Proper validation contributes to research reproducibility and prevents the propagation of misleading findings in scientific literature .

What are the mechanistic principles behind combining immune checkpoint inhibitors with anti-angiogenic antibodies?

The combination of immune checkpoint inhibitors (such as PD-1/CTLA-4 bi-specific antibodies) with anti-angiogenic therapies (such as VEGFR-2 monoclonal antibodies) is based on the interplay between immune cells and tumor angiogenesis. This combination approach targets multiple aspects of the tumor microenvironment simultaneously .

The underlying mechanisms include:

  • Normalization of tumor vasculature by anti-angiogenic therapy, improving immune cell infiltration

  • Reduction of immunosuppressive cells (regulatory T cells, myeloid-derived suppressor cells) in the tumor microenvironment

  • Enhancement of antigen presentation through improved dendritic cell function

  • Unblocking immunosuppression at different cycles of the acquired immune pathway

  • Inhibition of VEGF-mediated immunosuppression

Clinical trials investigating this approach focus on patients with advanced solid tumors who have failed standard treatments, potentially offering new therapeutic options. The PD-1/CTLA-4 bi-specific antibody Cadonilimab combined with VEGFR-2 monoclonal antibody exemplifies this strategy in current clinical evaluation .

How should pharmacokinetic and immunogenicity data be integrated into antibody therapeutic development?

Pharmacokinetic (PK) and immunogenicity data are critical parameters in antibody therapeutic development. Clinical studies assess these properties alongside safety and tolerability profiles to inform dosing strategies and risk assessment .

A methodological approach to integrating this data includes:

  • Establishing baseline PK parameters:

    • Determine elimination half-life

    • Assess volume of distribution

    • Evaluate clearance mechanisms

    • Characterize dose-proportionality

  • Immunogenicity assessment:

    • Measure anti-drug antibody (ADA) development

    • Correlate ADA titers with PK alterations

    • Assess neutralizing capacity of ADAs

    • Evaluate impact on safety and efficacy

  • Integration with preclinical data:

    • Compare human PK with animal models

    • Assess predictive value of preclinical immunogenicity

    • Adjust therapeutic index calculations

  • Population PK modeling:

    • Incorporate patient variables (age, weight, disease state)

    • Model impact of immunogenicity on exposure

    • Simulate dosing regimens to maintain therapeutic levels

This integrated approach enables rational dose selection and administration schedule optimization while accounting for individual variability factors .

How can researchers optimize their antibody research findings to appear in Google's "People Also Ask" section?

To populate in Google's "People Also Ask" (PAA) section with antibody research findings, researchers must structure their content to align with information-seeking queries. The PAA section appears in approximately 30% of all Google queries and represents valuable first-page visibility .

Strategic approaches include:

  • Identify popular questions in your research domain:

    • Use keyword research tools to identify common questions

    • Review existing PAAs for related antibody research topics

    • Analyze question patterns in scientific forums

  • Structure content in question-answer format:

    • Create FAQ sections on lab websites and publications

    • Implement FAQ schema markup to help Google identify Q&A content

    • Ensure answers are concise yet comprehensive (typically 40-60 words)

  • Focus on informational queries:

    • Address methodology questions (how to validate antibodies)

    • Explain technical concepts (differences between antibody formats)

    • Clarify experimental approaches (selecting appropriate controls)

  • Create dedicated blog posts or knowledge base articles:

    • Use question-based headlines that match search intent

    • Provide detailed, authoritative answers

    • Include supporting visuals and data tables

This approach increases the likelihood of research findings appearing in PAAs, improving visibility and citation potential for antibody research .

What strategies help researchers track antibody batch variability and availability issues?

Tracking antibody batch variability and addressing availability issues are significant challenges in research continuity. When well-validated antibodies (especially polyclonal antibodies generated in finite amounts) are sold out or replaced, research reproducibility is threatened .

Effective management strategies include:

  • Proactive inventory monitoring:

    • Maintain detailed records of antibody sources, catalog numbers, and lot numbers

    • Document performance metrics for each batch

    • Purchase larger quantities of critical antibodies when possible

  • Lot testing and comparison:

    • Test new lots alongside previous lots before depleting old stock

    • Generate reference data sets for comparison

    • Document any performance differences

  • Alternative validation approaches:

    • Identify and validate backup antibodies targeting different epitopes

    • Develop orthogonal detection methods as contingencies

    • Consider generating in-house antibodies for critical applications

  • Vendor communication:

    • Establish relationships with technical support representatives

    • Request advance notification of product discontinuation

    • Inquire about antibody production methods and QC parameters

  • Community database participation:

    • Contribute to antibody validation repositories

    • Share batch performance data in community resources

    • Advocate for improved reporting of reagent details in publications

These practices help mitigate the risks associated with antibody discontinuation and batch-to-batch variability that can undermine experimental reproducibility .

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.