AVT3A Antibody

Shipped with Ice Packs
In Stock

Description

Expression Profile

ATAD3A is significantly upregulated in BCa tissues compared to normal adjacent tissues. Key findings include:

Tissue TypeATAD3A ExpressionSource
Bladder CancerHigh (491 cases)
Normal BladderLow (110 cases)

Mechanistic Insights

  • ER stress and survival: ATAD3A knockdown increases ER stress and reduces calreticulin exposure, enhancing immunogenic cell death .

  • Mitochondrial function: ATAD3A interacts with Tom40 and Tim23, regulating PINK1-mediated autophagy and mitochondrial fission .

  • Prognostic relevance: High ATAD3A levels correlate with poor outcomes in BCa, suggesting its utility as a diagnostic biomarker .

4. ATAD3A Antibody Specifications
The following data are derived from commercial antibody sources (Abcam):

ParameterValueApplicationSource
Target ProteinATAD3A (71 kDa)IHC, Western blot
HostMouse
ClonalityMonoclonal
ImmunogenRecombinant human ATAD3A
Cross-reactivityHuman

Clinical and Research Applications

  • Diagnostic biomarker: Detects ATAD3A overexpression in BCa, aiding in early diagnosis and prognosis .

  • Therapeutic target: Silencing ATAD3A may enhance cancer cell apoptosis and immune infiltration, offering potential for targeted therapies .

  • Mitochondrial studies: Used to investigate mitochondrial dynamics and ER-mitochondria crosstalk in cancer .

Challenges and Future Directions

  • Specificity concerns: ATAD3A antibodies may cross-react with similar mitochondrial proteins, requiring validation in diverse tumor types.

  • Therapeutic translation: Further studies are needed to explore ATAD3A inhibition strategies and their impact on cancer progression.

References Antibody structure and function (general overview). ATAD3A in bladder cancer. ATAD3A antibody specifications.

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
AVT3A antibody; At5g65990 antibody; K2A18.5Amino acid transporter AVT3A antibody; AtAvt3A antibody; Aromatic and neutral amino acid transporter-like protein 3 antibody
Target Names
AVT3A
Uniprot No.

Target Background

Function
This antibody targets AVT3A, a protein that facilitates the translocation of neutral amino acids, and to a lesser extent, aromatic amino acids, from the vacuole to the cytoplasm. This process requires ATP for proper functioning.
Gene References Into Functions
  1. The AtAVT3 family represents the long-sought-for vacuolar amino acid exporters in plants. [AtAVT3A] PMID: 27925655
Database Links

KEGG: ath:AT5G65990

STRING: 3702.AT5G65990.1

UniGene: At.49231

Protein Families
Amino acid/polyamine transporter 2 family, Amino acid/auxin permease (AAAP) (TC 2.A.18.8) subfamily
Subcellular Location
Vacuole membrane; Multi-pass membrane protein.
Tissue Specificity
Ubiquitous.

Q&A

What is the mechanism of action for AVT3A antibody in viral neutralization?

AVT3A belongs to the family of broadly neutralizing antibodies (NAbs) that target conserved epitopes on viral envelope proteins. Similar to the well-characterized AR3A antibody, AVT3A likely functions by binding to key structural components that are essential for viral entry into host cells . The antibody's neutralizing activity involves preventing viral attachment to host receptors or disrupting post-binding conformational changes required for membrane fusion. This mechanism is critical for researchers to consider when designing experiments that evaluate antiviral efficacy or escape mutations.

How do I evaluate the neutralization potency of AVT3A in vitro?

To effectively evaluate neutralization potency, researchers should employ cell culture-based neutralization assays using chimeric recombinant viruses. For instance, studies with AR3A utilized chimeric cell culture-infectious HCV recombinants (HCVcc) to assess neutralization potency . A standardized methodology includes:

  • Prepare serial dilutions of purified AVT3A antibody

  • Pre-incubate with a standardized viral inoculum (1-2 hours at 37°C)

  • Add the antibody-virus mixture to susceptible cell lines (e.g., Huh7.5 for HCV)

  • Incubate for 48-72 hours

  • Assess infection rates using reporter systems or immunostaining

  • Calculate IC50 values to quantify neutralization potency

This methodological approach enables systematic evaluation of neutralization efficiency across different viral strains or mutants.

What cell lines are most appropriate for studying AVT3A antibody function?

The selection of appropriate cell lines is critical for studying antibody function. For viral neutralization studies, researchers should choose cell lines that express the relevant viral receptors and support viral replication. Based on similar antibody research, Huh7.5 cells are commonly used for hepatitis virus studies . For functional studies of agonist antibodies similar to AVT3A, reporter cell lines that can measure downstream signaling are essential . When designing experiments:

  • Verify receptor expression levels in your chosen cell line

  • Consider the impact of cell passage number on receptor expression

  • Include appropriate control cell lines that lack the target receptor

  • Validate that the cell line supports the full viral life cycle if studying neutralization

Selection of appropriate cell lines ensures that results accurately reflect antibody function in relevant biological contexts.

How can I design experiments to identify viral escape mutations against AVT3A?

Designing robust experiments to identify viral escape mutations requires a systematic approach similar to that used for AR3A antibody. A comprehensive methodology includes:

  • Culture susceptible cells infected with the target virus in the presence of sub-neutralizing concentrations of AVT3A

  • Gradually increase antibody concentration over multiple passages

  • Sequence viral isolates after each passage to identify emerging mutations

  • Introduce identified mutations into infectious molecular clones using site-directed mutagenesis

  • Evaluate the impact of individual mutations on antibody binding and neutralization sensitivity

This approach has successfully identified resistance substitutions in HCV studies, such as M345T, L438S, and F442Y against AR3A . When analyzing escape mutants, researchers should consider both the impact on antibody binding and potential fitness costs to the virus, as some mutations (e.g., G523A, G530A, and D535A in HCV) significantly reduce viral fitness .

What are the best methodologies for epitope mapping of AVT3A?

Advanced epitope mapping requires multiple complementary approaches to achieve high-resolution understanding of antibody-antigen interactions. A comprehensive strategy includes:

MethodologyApplicationResolutionAdvantages
Alanine scanning mutagenesisIdentifying critical binding residuesAmino acid levelIdentifies functional hotspots
X-ray crystallography3D structure of antibody-antigen complexAtomic levelReveals precise binding interactions
Cryo-electron microscopyStructure of antibody bound to intact virionsMedium to highCaptures binding in native context
Hydrogen-deuterium exchangeDynamic binding interactionsPeptide levelReveals conformational changes
Phage display librariesBinding motif identificationPeptide levelHigh-throughput epitope profiling

For targets similar to AVT3A, researchers have successfully employed alanine substitutions to identify key residues (e.g., positions 424, 525, and 540) that affect antibody binding . When interpreting data, consider that critical binding residues may have different impacts on antibody binding versus viral fitness, requiring careful differentiation between these effects.

How can structural data be leveraged to improve AVT3A activity?

Structural insights can dramatically enhance antibody engineering efforts, as demonstrated by recent advances in agonist antibody development. To leverage structural data for optimizing AVT3A:

  • Obtain high-resolution structures of AVT3A in complex with its target antigen

  • Identify key interaction interfaces, particularly within the CDR regions

  • Use computational approaches to predict mutations that might enhance binding affinity or functional activity

  • Generate focused libraries of variants with mutations in the binding interface

  • Screen these libraries for improved neutralization potency or breadth

This approach has been successfully used to convert antagonistic single-domain antibodies into agonists through rational mutation of CDR3 residues that interact with the receptor binding pocket, without disrupting binding affinity . When applying structure-guided design, focus modifications on CDR regions, particularly CDR3, which often makes the most extensive contacts with the antigen.

What are the primary mechanisms by which viruses develop resistance to AVT3A?

Viruses can develop resistance to neutralizing antibodies through multiple mechanisms. Based on studies with similar antibodies like AR3A, key resistance mechanisms include:

  • Direct epitope mutations: Substitutions that directly interfere with antibody binding while maintaining viral function

  • Conformational masking: Mutations that alter protein conformation to shield the epitope

  • Glycan shielding: Addition of glycosylation sites that sterically hinder antibody access

  • Cooperative escape: Multiple partial resistance mutations that together confer high-level resistance

Research with AR3A identified specific resistance substitutions such as M345T in H77/JFH1, L438S and F442Y in H77/JFH1ΔHVR1, and D431G in J6/JFH1ΔHVR1 . Notably, the impact of these mutations can be context-dependent, with some substitutions (L438S and F442Y) conferring high-level resistance in one viral background while abrogating infectivity in another . This complex interplay between resistance and viral fitness highlights the importance of studying escape mutations in multiple viral contexts.

How does viral genetic diversity affect AVT3A neutralization efficacy?

Viral genetic diversity significantly impacts neutralizing antibody efficacy through multiple mechanisms:

Diversity FactorImpact on NeutralizationResearch Implications
Hypervariable regionsCan shield conserved epitopesStudy antibody efficacy with/without HVR1 regions
Genotype variationAffects epitope conservationTest neutralization across genotypes
Quasispecies diversityPre-existing resistant variantsDeep sequencing to identify minor resistant populations
Compensatory mutationsRestore fitness to resistant variantsLong-term passage experiments to identify co-evolution

Studies with AR3A demonstrated that the presence of hypervariable region 1 (HVR1) significantly affected the viability and resistance profile of various escape mutations . For example, D431G conferred resistance to J6/JFH1ΔHVR1 but not to J6/JFH1, highlighting the complex role of HVR1 in virus escape mechanisms . When designing AVT3A studies, researchers should test multiple viral isolates and consider the impact of regions like HVR1 on neutralization outcomes.

What strategies can improve the neutralization breadth of AVT3A antibody?

Enhancing neutralization breadth is crucial for developing antibodies with therapeutic potential against diverse viral variants. Advanced engineering approaches include:

  • Affinity maturation: Using directed evolution to enhance binding to the primary epitope

  • Epitope grafting: Incorporating complementary epitopes from other neutralizing antibodies

  • Fc engineering: Modifying the Fc region to enhance effector functions

  • Bispecific formats: Creating bispecific antibodies that target multiple epitopes

  • Valency optimization: Developing multivalent antibody formats to increase avidity

Research on agonist antibodies has demonstrated that tetravalent biepitopic antibodies can show superior bioactivity compared to bivalent formats . When designing optimization strategies, consider that different modifications may have synergistic or antagonistic effects. For example, increasing valency might enhance neutralization potency but could negatively impact tissue penetration in vivo.

How can I optimize AVT3A valency and specificity for enhanced activity?

Optimizing antibody valency and specificity can dramatically improve functional activity, particularly for antibodies that act via receptor clustering. Based on research with agonist antibodies, consider:

  • Create bivalent and tetravalent antibody constructs to compare activity

  • Develop both monoepitopic (targeting one epitope) and biepitopic (targeting two non-overlapping epitopes) formats

  • Test various molecular architectures, including:

    • Dual variable domain formats (DVD)

    • ScFv-Fc fusions

    • Diabody formats

Experimental evidence indicates that tetravalent biepitopic variants often show superior activity in vitro compared to simpler constructs . When evaluating these formats, assess both functional activity and pharmaceutical properties such as stability, expression levels, and pharmacokinetics. Importantly, tetravalent biepitopic antibodies have demonstrated improved pharmacodynamic profiles in vivo while maintaining pharmacokinetic properties similar to standard IgG formats .

How should I analyze conflicting neutralization data with AVT3A?

Conflicting neutralization data can arise from methodological variations or biological factors. A systematic approach to resolving such conflicts includes:

  • Standardize experimental conditions:

    • Viral stock preparation and quantification

    • Cell culture conditions and passage number

    • Incubation times and temperatures

    • Readout methodology

  • Consider biological variables:

    • Presence of hypervariable regions (e.g., HVR1) that can affect resistance patterns

    • Viral coreceptor dependency, which may change with mutations

    • Conformational states of the antigen ("envelope breathing")

    • Cell type-specific factors that influence neutralization

  • Employ statistical approaches:

    • Perform experiments in at least triplicate

    • Calculate 95% confidence intervals for neutralization curves

    • Use appropriate statistical tests to evaluate significance of differences

When interpreting conflicting data, remember that context-dependent effects are common in antibody-virus interactions. For example, the D431G mutation affected neutralization sensitivity differently in J6/JFH1 versus J6/JFH1ΔHVR1, possibly due to altered coreceptor dependency .

What controls are essential for validating AVT3A specificity and function?

Rigorous control experiments are crucial for validating antibody specificity and function. Essential controls include:

Control TypePurposeImplementation
Isotype controlControls for non-specific effectsUse matched isotype antibody at equivalent concentration
Target knockoutConfirms target specificityTest antibody on cells lacking target expression
Binding competitionValidates epitope specificityPre-incubate with soluble antigen or competing antibodies
Positive control antibodyBenchmarks activityInclude well-characterized antibody against same target
Functional readout controlsValidates assay performanceInclude known activators/inhibitors of the pathway

For neutralization assays, additional controls should include antibodies with known neutralizing activity against the same virus and non-neutralizing antibodies that bind the same target. When studying antibody-mediated immune responses, consider including controls to assess background T cell activation, such as unstimulated cells and non-specific stimuli .

How can I assess AVT3A's ability to enhance T cell responses against viral infections?

Assessing antibody-mediated enhancement of T cell responses requires comprehensive immunological analyses. Based on studies of anti-HIV-1 antibody therapy , a methodological approach includes:

  • Measure antigen-specific T cell responses using:

    • Intracellular cytokine staining (ICS) for IFN-γ, TNF-α, MIP1-β, and CD107A

    • Activation-induced marker (AIM) assays to identify antigen-specific T cells

    • Proliferation assays using CFSE or similar dyes

  • Evaluate T cell functionality through:

    • Polyfunctionality analysis (cells producing multiple cytokines)

    • Cytotoxicity assays against infected target cells

    • Assessment of exhaustion markers (PD-1, HLA-DR, CD38)

  • Monitor kinetics of responses:

    • Establish baseline measurements before antibody administration

    • Assess responses at peak antibody concentration

    • Follow long-term persistence of enhanced T cell responses

Research with anti-HIV-1 antibodies demonstrated significant increases in antigen-specific CD8+ T cells expressing IFN-γ, TNF-α, MIP1-β, and/or CD107A during antibody therapy . These responses peaked at 6-7 weeks but remained significantly elevated for weeks after antibody administration, suggesting that neutralizing antibodies may have immunomodulatory effects beyond direct viral neutralization .

What is the significance of epitope targeting for AVT3A's barrier to resistance?

The specific epitope targeted by neutralizing antibodies directly influences the genetic barrier to viral resistance. Understanding this relationship is critical for therapeutic development:

  • Conserved functional epitopes typically present a higher barrier to resistance

  • Residues essential for viral fitness cannot easily mutate without functional cost

  • Structural constraints limit viable escape pathways

Studies with AR3A demonstrated a high barrier to resistance, with initial escape mutations conferring only low-level resistance . This characteristic is particularly important for therapeutic applications, as a high genetic barrier to resistance prolongs clinical efficacy. When evaluating AVT3A's potential as a therapeutic, researchers should systematically assess:

  • The conservation of the target epitope across viral variants

  • The functional significance of epitope residues for viral entry or replication

  • The fitness cost of potential resistance mutations

  • The combined effect of AVT3A with other antibodies targeting different epitopes

Combination antibody therapy targeting multiple non-overlapping epitopes can further increase the genetic barrier to resistance, similar to combination antiretroviral therapy for HIV .

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.