ATL75 Antibody

Shipped with Ice Packs
In Stock

Description

Potential Misinterpretation: ATL-Associated Antibodies

Adult T-cell Leukemia/Lymphoma (ATL), caused by human T-cell lymphotropic virus type 1 (HTLV-1), involves antibodies targeting viral or tumor-associated antigens. Key findings include:

Antibody TargetFunction/ApplicationResearch FindingsSources
Anti-ATLA (Adult T-cell Leukemia-associated antigen)Detects HTLV-1 viral particles- Binds surface glycoproteins/structural proteins of ATLV (HTLV-1)
- Used in immunoferritin electron microscopy for ATL diagnosis
Anti-CD25 (IL-2Rα)Therapeutic target in ATL- Expressed in 90% of ATL cases
- Mogamulizumab (anti-CCR4 antibody) shows clinical efficacy
Anti-p75NTR (Neurotrophin Receptor)Investigational biomarker- Overexpressed in ATL cells
- Linked to apoptosis resistance

Potential Misinterpretation: p75NTR-Targeting Antibodies

The Anti-p75NTR antibody (e.g., Alomone Labs #ANT-007) is a well-characterized reagent with applications in oncology and autoimmune research:

Research Data

  • Neuroblastoma Studies:

    • p75NTR expression correlates with MYCN amplification (1.7 μmol/L LC50 in MYCN+ vs. 8.1 μmol/L in MYCN- cells)

    • Xenograft models show 86% tumor regression after p75NTR-targeted therapy

  • Autoimmunity:

    • Anti-p75NTR antibodies reduce autoantibody production in lupus models (p < 0.01 vs. controls)

Analysis of Similar Nomenclature

The alphanumeric sequence "ATL75" may derive from:

  • ATL: Adult T-cell Leukemia

  • 75: Molecular weight (e.g., p75NTR at 75 kDa) or clone designation

No antibody matching this nomenclature exists in current databases. Researchers studying ATL or p75NTR-related pathways typically use:

  • Commercial Antibodies: Anti-p75NTR (#ANT-007), Anti-CD25 (IL-2Rα)

  • Clinical Candidates: ANX005 (anti-C1q antibody in Phase II for Huntington’s)

Recommendations for Further Research

  1. Validate antibody specificity using:

    • Knockout/knockdown controls

    • Blocking peptide competition assays

  2. Explore combination therapies with:

    • CDK inhibitors (e.g., AT7519) in MYCN+ cancers

    • Complement inhibitors (e.g., ANX005) in autoimmune conditions

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
ATL75; At1g49200; F27J15.33; RING-H2 finger protein ATL75; RING-type E3 ubiquitin transferase ATL75
Target Names
ATL75
Uniprot No.

Target Background

Database Links

KEGG: ath:AT1G49200

STRING: 3702.AT1G49200.1

UniGene: At.26144

Protein Families
RING-type zinc finger family, ATL subfamily
Subcellular Location
Membrane; Single-pass membrane protein.

Q&A

What validation methods confirm antibody specificity for targets like LY75?

Antibody specificity validation requires multiple complementary methods to ensure reliable research outcomes. For membrane-bound targets such as LY75, a combination of the following techniques is recommended:

  • Western blotting (WB): Confirms antibody recognition of the target protein at the expected molecular weight. For LY75, this helps verify the antibody binds to the correct protein rather than structurally similar proteins .

  • Immunohistochemistry (IHC): Validates antibody performance in tissue sections, examining expression patterns that should match known biology of the target .

  • Immunocytochemistry/Immunofluorescence (ICC-IF): Confirms subcellular localization patterns consistent with the target protein's known distribution .

  • Knockout/knockdown controls: Essential for definitive validation, using cells where the target gene has been deleted or suppressed .

  • Multiplexed approaches: Recently developed methods challenge antibodies with multiple potential targets simultaneously to assess cross-reactivity, as demonstrated in studies with G protein-coupled receptors (GPCRs) .

A standardized process helps ensure rigorous quality control for polyclonal antibodies such as those against LY75, minimizing batch-to-batch variation in antibody performance .

How do antibody requirements differ when targeting membrane proteins versus soluble proteins?

Targeting membrane-bound proteins like LY75 presents unique challenges compared to soluble proteins:

ParameterMembrane Protein AntibodiesSoluble Protein Antibodies
Epitope accessibilityMust target extracellular domains for live cell applicationsCan access entire protein structure
Sample preparationOften requires special detergents or membrane solubilizationStandard protein extraction methods
Validation complexityHigher due to conformational dependenceGenerally more straightforward
Cross-reactivity riskHigher due to conserved membrane domainsVariable depending on protein family
Application limitationsMay have reduced performance in certain fixed samplesUsually broader application range

For membrane proteins, specialized extraction methods and careful epitope selection are critical. Recent studies have developed multiplexed pipelines to produce and extract membrane receptors like GPCRs to test antibody selectivity against structurally similar family members . These approaches are particularly valuable when working with receptors that share high sequence homology.

What criteria should guide antibody selection for immunohistochemistry applications?

When selecting antibodies for IHC applications, particularly for targets like LY75, researchers should consider:

  • Validation status: Choose antibodies with documented validation in IHC specifically, as performance varies between applications .

  • Tissue specificity: Verify the antibody has been tested in relevant tissue types where your target is expressed.

  • Epitope location: For membrane proteins, antibodies targeting extracellular domains often perform better in IHC.

  • Fixation compatibility: Confirm compatibility with your preferred fixation method, as some epitopes are fixation-sensitive.

  • Concentration optimization: Titrate the antibody to determine optimal working concentration for signal-to-noise ratio.

  • Positive/negative controls: Include tissues with known expression patterns and knockout tissues when possible .

A systematic validation approach is crucial, as demonstrated by the Human Protein Atlas project, which provides extensively validated antibodies for research applications .

How can single-cell sequencing technologies enhance antibody research?

Single-cell sequencing has revolutionized antibody research by enabling precise characterization of immune responses and antibody development:

Key technologies and their applications:

  • 10x Genomics Chromium system: Enables high-throughput analysis of thousands of single cells simultaneously, allowing researchers to dissect major immune populations and accurately retrieve paired antibody chain sequences .

  • Beacon system (Berkeley Lights): Provides a platform for single-cell isolation and functional screening, allowing researchers to correlate antibody sequences with functional properties like binding affinity and specificity .

  • Integrated approaches: Combining functional assays with sequencing enables selection of antigen-specific antibodies directly from antibody-secreting cells (ASCs) .

These technologies offer several advantages over traditional methods:

  • Preservation of native antibody heavy and light chain pairing

  • Direct linking of antibody sequences to cell phenotypes

  • Higher throughput and efficiency compared to hybridoma technology

  • Ability to study rare antibody-producing cells within heterogeneous populations

Modern microfluidic-based assays can compartmentalize single cells in wells or droplets, label RNA with cell-specific barcodes, and trace sequences back to their original cells after sequencing . This approach has dramatically accelerated the discovery of therapeutic antibodies with desired properties.

How do structural prediction tools like AlphaFold 2 support experimental antibody validation?

Computational tools such as AlphaFold 2 provide valuable complementary data to experimental antibody validation:

Integration of computational and experimental approaches:

  • Epitope prediction: AlphaFold 2 can predict protein structures with high accuracy, helping researchers identify accessible epitopes for antibody targeting .

  • Cross-reactivity assessment: Structural modeling can identify regions of structural similarity between related proteins, highlighting potential cross-reactivity issues .

  • Experimental validation support: As demonstrated in the collaboration between SciLifeLab researchers Jochen Schwenk and Arne Elofsson, AlphaFold 2 predictions can help interpret wet lab data on antibody selectivity .

  • Optimization guidance: Structural insights can guide antibody engineering efforts by identifying regions that affect binding affinity or specificity.

The integration of computational and experimental approaches creates a powerful framework for antibody validation. In one notable example, researchers used AlphaFold 2 to support experimental data validating antibodies against G protein-coupled receptors (GPCRs), demonstrating how computational tools can provide new perspectives on antibody-antigen interactions .

What methodologies are most effective for studying human monoclonal antibodies against disease targets?

The study of human monoclonal antibodies against disease targets requires sophisticated methodologies, as exemplified by research on Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) for malaria vaccines:

Key methodological approaches:

  • B-cell isolation: Peripheral blood B cells from vaccinated individuals or patients provide a source of disease-relevant antibodies .

  • Neutralization assays: Functional screening identifies antibodies that neutralize pathogens or block disease-relevant interactions .

  • Epitope mapping: X-ray crystallography and other structural biology techniques determine precise binding sites and interaction mechanisms .

  • Synergy assessment: Testing antibody combinations can reveal synergistic effects, as seen in studies where non-neutralizing antibodies reduced red blood cell invasion speed, potentiating the effect of neutralizing antibodies .

  • Structure-guided analysis: Correlating antibody structure with function informs rational vaccine design .

The research on PfRH5 antibodies demonstrated how isolating monoclonal antibodies from vaccinees can identify distinct binding sites with different functional properties. This approach revealed valuable insights, including the discovery of antibodies that synergize with other malaria invasion protein antibodies, providing a roadmap for structure-guided vaccine development .

How can researchers address non-specific binding in antibody-based experiments?

Non-specific binding is a common challenge in antibody experiments that can lead to misleading results. Addressing this issue requires systematic troubleshooting:

Strategies to minimize non-specific binding:

  • Optimization of blocking conditions:

    • Test different blocking agents (BSA, normal serum, commercial blockers)

    • Extend blocking time for challenging samples

    • Consider using blocking agents that match the species of your secondary antibody

  • Antibody dilution optimization:

    • Perform titration experiments to find optimal concentration

    • Higher antibody concentrations often increase non-specific binding

    • For LY75 antibody, starting with the manufacturer's recommended 0.1 mg/ml concentration and then titrating is advisable

  • Buffer modifications:

    • Include detergents like Tween-20 to reduce hydrophobic interactions

    • Adjust salt concentration to disrupt weak ionic interactions

    • Consider adding carrier proteins to prevent non-specific adsorption

  • Sample preparation improvements:

    • Ensure complete antigen retrieval for fixed samples

    • For membrane proteins like LY75, optimize membrane protein extraction

    • Remove potential interfering substances through additional purification steps

  • Controls:

    • Include isotype controls to identify Fc-mediated binding

    • Use knockout/knockdown samples as negative controls

    • Pre-adsorb antibody with purified antigen to confirm specificity

These approaches should be systematically tested and documented to establish reliable protocols for specific detection of targets like LY75.

What techniques can distinguish functionally active from non-functional antibodies?

Distinguishing functionally active antibodies from non-functional ones is critical for therapeutic development and mechanistic studies. Recent research has developed several approaches to assess antibody functionality:

Functional assessment techniques:

  • Lipid mixing assays: Used to assess membrane fusion activity of antibodies, as demonstrated in studies of atlastin GTPases .

  • Live-cell microscopy: Enables real-time monitoring of antibody effects on cellular processes, as used in studies of erythrocyte invasion by malaria parasites .

  • Neutralization assays: Determine an antibody's ability to prevent pathogen entry or replication .

  • Kinetic analysis: Measuring rate constants can reveal important differences between antibodies:

    • In atlastin studies, initial fusion rates were quantified as percentages of maximal dequenching per second

    • This approach identified disease variants with impaired function (R239C with reduced rate of 0.005/s compared to wild-type 0.02/s)

  • Synergy assessment: Testing antibody combinations can reveal functional interactions:

    • Some antibodies may not be neutralizing alone but potentiate the effects of other antibodies

    • As seen in malaria research, certain antibodies reduced invasion speed, enhancing the effect of all neutralizing antibodies

A comprehensive functional assessment often requires multiple complementary approaches, as different antibodies may exert effects through distinct mechanisms.

How are antibodies employed in the development of antibody-drug conjugates (ADCs)?

Antibody-drug conjugates represent a sophisticated application of antibody technology for targeted therapy. The development process involves multiple critical considerations:

Key components and development considerations:

  • Antibody selection criteria:

    • High affinity and avidity for the target antigen

    • Minimal binding to off-target sites

    • Selective binding leading to accumulation at target site

    • Low immunogenicity and cross-reactivity

    • Appropriate internalization kinetics

  • Structural components of ADCs:

    • Monoclonal antibody: Provides selective targeting

    • Cytotoxic drug payload: Delivers therapeutic effect

    • Linker molecule: Controls drug release within target cells

  • Mechanism of action:

    • ADC binds to cancer cell surface

    • Internalization through receptor-mediated endocytosis

    • Drug release within cancer cell

  • Optimization strategies:

    • Antibody engineering to enhance target binding

    • Linker chemistry modifications for stability

    • Payload selection based on potency and mechanism

ADCs integrate the potency of cytotoxic drugs with the selectivity of monoclonal antibodies, minimizing damage to healthy cells and reducing systemic toxicity . This approach has proven particularly valuable for cancer therapy, where targeted delivery of cytotoxic agents can improve therapeutic index.

What approaches can identify and characterize autoinhibition mechanisms in antibody targets?

Understanding autoinhibition mechanisms in protein targets is crucial for developing effective antibodies. Research on human atlastin GTPases provides insights into approaches for identifying such mechanisms:

Methods to identify autoinhibition:

  • Truncation analysis:

    • Systematic deletion of protein domains can reveal inhibitory regions

    • In atlastin studies, C-terminal truncation activated ATL2, increasing fusion rate by 500-fold (from 0.0006 to 0.3/s)

    • Similar approaches can identify regulatory domains in antibody targets

  • Domain swapping experiments:

    • Exchanging domains between related proteins can pinpoint inhibitory elements

    • Atlastin research demonstrated that swapping C-terminal extensions between ATL1 and ATL2 transferred inhibitory properties

  • Site-directed mutagenesis:

    • Targeted mutations can disrupt autoinhibitory interactions

    • The H258R variant in atlastin doubled fusion rate from 0.02 to 0.05/s

  • Functional reconstitution assays:

    • In vitro reconstitution of protein activity allows quantitative assessment

    • Provides controlled conditions to test hypotheses about regulatory mechanisms

  • Structural analysis:

    • Cryo-EM, X-ray crystallography, or computational modeling can reveal inhibitory conformations

    • Identifies intramolecular interactions that maintain inactive states

These approaches, particularly when combined, can reveal subtle regulatory mechanisms that might be targeted by therapeutic antibodies or considered in experimental design when using antibodies to study protein function.

How are single-cell technologies transforming antibody discovery?

Single-cell technologies are revolutionizing antibody discovery through unprecedented resolution of immune responses:

Transformative approaches:

  • Evolution from traditional methods:

    • Traditional qPCR could measure multiple genes from hundreds of cells but lacked whole-transcriptome analysis

    • FACS sorting provided population-level insights but limited single-cell resolution

    • Modern microfluidic platforms enable compartmentalization of single cells in wells or droplets

  • Technical advantages of new platforms:

    • Cell-specific barcoding allows tracing sequences back to original cells

    • High-throughput processing of thousands of cells simultaneously

    • Accurate recovery of paired antibody chains from individual B cells

    • Integration with functional screening platforms

  • Applications in therapeutic development:

    • Direct isolation and sequencing of antibody-secreting cells

    • Selection of antigen-specific antibodies with desired properties

    • Accelerated development of antibody-based therapeutics

The integration of these technologies with computational analysis pipelines addresses challenges in analyzing single-cell data, enabling researchers to rapidly identify promising antibody candidates for further development.

What novel approaches are advancing the validation of antibodies against membrane-bound receptors?

Recent advances have significantly improved our ability to validate antibodies against challenging membrane-bound receptors:

Innovative validation approaches:

  • Multiplexed pipeline technologies:

    • Researchers at SciLifeLab and Rockefeller University developed methods to produce and extract 215 GPCR receptors

    • These systems enabled testing of over 400 antibodies from the Human Protein Atlas

    • The approach challenges antibodies with receptors from different families to assess specificity

  • Integration of computational and experimental techniques:

    • AlphaFold 2 predictions complement wet lab data on antibody binding

    • Structural modeling helps interpret experimental findings

    • Collaborative approaches between wet lab and computational researchers enhance validation quality

  • Cell-based assays with knockout controls:

    • Generation of cell lines lacking target receptors provides definitive negative controls

    • CRISPR-Cas9 technology enables precise receptor knockout in relevant cell types

  • Receptor extraction technologies:

    • Advanced methods for membrane protein extraction preserve native conformations

    • Improved purification techniques maintain structural integrity for antibody testing

These approaches collectively address the significant challenges in validating antibodies against membrane-bound receptors, which have historically been difficult to study due to their structural complexity and similarity within protein families.

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.