ATJ16 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of ATG16L1 Antibody

ATG16L1 (Autophagy Related 16 Like 1) is a protein essential for autophagosome formation during autophagy, a cellular degradation process. Antibodies targeting ATG16L1 enable researchers to:

  • Detect phosphorylation events linked to autophagy induction .

  • Study its interaction with Rab33A in hormone secretion pathways .

  • Analyze tissue-specific expression via immunohistochemistry or Western blot .

Autophagy Induction Monitoring

A monoclonal antibody targeting phospho-ATG16L1 (Ser287 in humans) serves as a direct marker for autophagy activation:

  • Detects phosphorylation exclusively on newly forming autophagosomes, unaffected by late-stage autophagy blocks .

  • Validated for Western blot, immunofluorescence, and immunohistochemistry .

  • Phosphorylation levels correlate with autophagy rates, providing a dynamic readout .

Role in Secretory Pathways

ATG16L1 interacts with Rab33A to regulate dense-core vesicle secretion in neuroendocrine cells:

  • Knockdown of ATG16L1 in PC12 cells inhibits hormone secretion independently of autophagy .

  • Localization to vesicles requires Rab33A binding, highlighting a dual functional role .

Western Blot Performance

  • 68 kDa band observed in human cell lysates (HeLa, NT2D1) .

  • Knockout controls confirm antibody specificity .

Immunohistochemistry

  • Strong cytoplasmic staining in paraffin-embedded U251 xenografts at 1:500 dilution .

Limitations and Considerations

  • Phospho-specific antibodies require validation under stress conditions (e.g., nutrient deprivation) .

  • Commercial antibodies may cross-react with non-target proteins; knockout validation is recommended .

Emerging Applications

  • Disease research: Links to Crohn’s disease (IBD10 locus) and neurodegenerative disorders .

  • Drug development: High-throughput screening for autophagy modulators .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Components: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
ATJ16 antibody; ARL1 antibody; B16 antibody; At1g24120 antibody; F3I6.4Chaperone protein dnaJ 16 antibody; AtDjB16 antibody; AtJ16 antibody; Protein ARG1-LIKE 1 antibody; AtARL1 antibody
Target Names
ATJ16
Uniprot No.

Target Background

Function
ATJ16 plays a continuous role in plant development, likely contributing to the structural organization of cellular compartments. Evidence suggests it is not involved in gravitropism signaling pathways.
Database Links

KEGG: ath:AT1G24120

STRING: 3702.AT1G24120.1

UniGene: At.41480

Protein Families
DnaJ family, B/II subfamily
Subcellular Location
Membrane; Peripheral membrane protein.
Tissue Specificity
Expressed constitutively in seedlings, roots, leaves, stems, flowers and siliques.

Q&A

What distinguishes nanobodies from conventional antibodies in GPCR research?

Nanobodies (heavy chain-only antibodies) offer several advantages over conventional antibodies when targeting G protein-coupled receptors like AT1R. Unlike traditional antibodies, nanobodies are significantly smaller (approximately 15 kDa), allowing for better tissue penetration and unique binding properties. Their single-domain structure enables them to access receptor epitopes that might be inaccessible to larger antibody molecules .

Methodologically, nanobodies can be engineered with high specificity for maternal circulation by:

  • Fusion to IgG1 Fc to increase molecular weight above the ~70 kDa renal filtration cutoff

  • Mutation of the Fc's neonatal receptor binding site to prevent placental transport

  • Modification of Fc gamma receptor binding sites to inhibit unwanted cytotoxic effects

These engineering approaches allow nanobodies to exhibit remarkably selective pharmacological properties that conventional antibodies cannot achieve at receptor, tissue, and cellular levels.

How are phospho-specific antibodies used to monitor autophagy?

Phospho-specific antibodies targeting ATG16L1 provide a superior method for monitoring autophagy induction compared to traditional approaches. The key advantage of phospho-ATG16L1 antibodies is their ability to specifically detect newly forming autophagosomes without being affected by prolonged stress or late-stage autophagy blocks that often confound other analytical methods .

Methodologically, researchers should:

This technique represents a significant advancement for studying autophagy induction across multiple experimental contexts and stress conditions.

What validation steps are essential when characterizing new research antibodies?

Rigorous validation is critical before implementing antibodies in research protocols. Based on best practices evident in the literature, researchers should:

  • Verify binding specificity through:

    • Competitive binding assays with known ligands (as demonstrated with AT118 nanobodies competing with AngII and olmesartan)

    • Testing against related protein targets to confirm selectivity

    • Validation in knockout/negative control cells or tissues

  • Assess functional activity through:

    • Cellular signaling assays (as shown with AT118-L Fc fusion protein)

    • Appropriate physiological readouts (e.g., blood pressure reduction for AT1R antagonists)

    • Dose-response relationships to establish potency

  • Characterize physical properties including:

    • Affinity measurements

    • Epitope mapping when possible

    • Stability under experimental conditions

These validation steps ensure experimental reliability and reproducibility before implementing antibodies in complex research applications.

How can antibodies be engineered for maternal-specific targeting without affecting fetal tissues?

Engineering antibodies for maternal specificity requires manipulation of key molecular properties to prevent placental transfer. The literature describes a sophisticated approach:

  • Size manipulation: Increase molecular weight above placental transfer thresholds (typically >70 kDa) through fusion to Fc domains or other scaffold proteins

  • FcRn binding site modification: Mutate the neonatal Fc receptor binding site to eliminate the active transport mechanism for antibodies across the placenta while maintaining extended maternal circulation

  • Effector function elimination: Block undesired cytotoxic effects by inhibiting Fc gamma receptor binding and complement fixation through established mutations

  • Target selectivity optimization: Maintain high binding affinity for the maternal target receptor while adjusting pharmacokinetic properties

This multi-faceted engineering approach allows for development of maternal-selective antagonists that can target conditions like preeclampsia without affecting fetal development.

How do active learning strategies improve antibody-antigen binding prediction in research?

Active learning represents a cutting-edge approach to optimize antibody-antigen binding prediction while minimizing experimental costs. The methodology involves:

  • Starting with a small labeled subset of antibody-antigen pairs

  • Implementing machine learning algorithms to predict binding properties

  • Strategically selecting the most informative additional experiments to conduct

  • Iteratively expanding the labeled dataset with new experimental data

Recent research has demonstrated that optimized active learning strategies can reduce the number of required antigen mutant variants by up to 35% compared to random selection approaches. The most effective algorithms accelerated the learning process by 28 steps relative to random baseline methods .

This approach is particularly valuable for out-of-distribution prediction scenarios where test antibodies and antigens are not represented in training data. Implementation requires specialized algorithms designed for many-to-many relationship data typical of library-on-library screening approaches used in antibody research .

What mechanisms explain why phospho-ATG16L1 antibodies are superior for detecting early-stage autophagy?

The superior performance of phospho-ATG16L1 antibodies for autophagy detection stems from unique mechanistic advantages:

  • Temporal specificity: Phospho-ATG16L1 is present only on newly forming autophagosomes, providing a precise temporal marker for autophagy initiation that is not confounded by later-stage events

  • Conservation across stressors: ATG16L1 phosphorylation represents a conserved signaling pathway activated by numerous autophagy-inducing conditions, making it a universal indicator

  • Methodological versatility: The antibody functions effectively across multiple experimental platforms (western blot, immunofluorescence, immunohistochemistry), allowing for consistent cross-platform analysis

  • Direct correlation: Phospho-ATG16L1 levels directly correspond to autophagy rates, providing quantitative measurement capabilities

These mechanisms collectively enable researchers to detect autophagy induction with greater precision than conventional methods, particularly in challenging experimental contexts such as rare cell populations or in vivo systems.

How should researchers differentiate between competitive and allosteric antibody interactions with receptors?

Distinguishing between competitive and allosteric antibody-receptor interactions requires specific experimental approaches:

  • Binding displacement assays:

    • Test whether the antibody prevents binding of natural ligands (e.g., AT118 nanobodies competitively blocking AngII binding to AT1R)

    • Determine if small molecule antagonists (e.g., olmesartan) compete with antibody binding

  • Structural studies:

    • Analyze how antibodies engage with the extracellular face of receptors

    • Determine whether binding affects allosteric networks within the receptor's transmembrane core

  • Co-binding experiments:

    • Assess whether simultaneous binding with known ligands is possible

    • Determine if binding occupies orthosteric versus allosteric sites

  • Functional readouts:

    • Compare signaling outcomes between competitive and potentially allosteric antibodies

    • Analyze pathway-specific effects that might indicate allosteric modulation

The literature shows that closely related antibody sequences can have "profoundly divergent pharmacological properties," highlighting the importance of these experimental distinctions .

What controls are essential when using antibodies to analyze complex protein modifications?

When using phospho-specific antibodies like those targeting ATG16L1, robust controls are essential to ensure experimental validity:

  • Phosphorylation state controls:

    • Phosphatase-treated samples to demonstrate specificity for the phosphorylated form

    • Mutagenesis of the target phosphorylation site to confirm antibody specificity

  • Inducer/inhibitor controls:

    • Positive controls using known autophagy inducers to verify detection capability

    • Negative controls using autophagy inhibitors to confirm specificity

  • Time-course validation:

    • Temporal analysis to differentiate newly forming autophagosomes from later stages

    • Comparison with traditional autophagy markers to establish correlation

  • Cross-validation:

    • Verification using complementary techniques (e.g., western blot findings confirmed with immunofluorescence)

    • Correlation of phospho-ATG16L1 levels with functional autophagy outcomes

These controls ensure that observed signals genuinely reflect the biological process under investigation rather than technical artifacts.

How might antibody-based therapies overcome limitations of small molecules in targeting GPCRs?

Antibody-based approaches offer several advantages over small molecules for GPCR targeting:

  • Enhanced selectivity:

    • Antibodies can achieve greater receptor subtype selectivity than small molecules

    • They can distinguish between closely related GPCRs where small molecules often show cross-reactivity

  • Tissue and cellular specificity:

    • Engineering approaches can restrict antibodies to specific tissues (e.g., maternal circulation)

    • Molecular size can be manipulated to control distribution patterns

  • Dual targeting potential:

    • Nanobodies can simultaneously bind to GPCRs along with specific small-molecule antagonists

    • This allows for combination approaches with tunable selectivity profiles

  • Expanded pharmacological diversity:

    • Antibody scaffolds demonstrate "rich pharmacological capacity"

    • They can function as both competitive and allosteric modulators

These advantages position antibody-based therapies as particularly promising for addressing GPCR-related conditions where small molecules have shown limitations in specificity or efficacy.

What advantages do combination approaches using antibodies with other molecular targeting strategies offer?

Combination strategies involving antibodies with other molecular targeting approaches provide synergistic benefits:

  • Complementary mechanism coverage:

    • Antibodies typically target extracellular protein domains while approaches like antisense oligonucleotides (ASOs) affect protein production

    • This allows simultaneous targeting of both existing protein and new protein synthesis

  • Enhanced tissue penetration:

    • Combined approaches can address different tissue compartments (e.g., brain versus periphery)

    • This is particularly valuable for systemic disorders with both central and peripheral manifestations

  • Improved therapeutic efficacy:

    • Research in Huntington's disease shows potential for combining antibodies targeting extracellular mutant huntingtin with ASOs reducing protein production

    • This "combinatorial treatment" approach yields "maximal benefits for patients"

  • Resistance management:

    • Multiple targeting mechanisms reduce the likelihood of therapeutic resistance

    • Different molecular approaches can address compensatory mechanisms

The emerging paradigm sees these combination approaches as particularly valuable for complex disorders requiring intervention at multiple biological levels.

How can antibody profiling inform understanding of disease progression?

Antibody profiling provides unique insights into disease progression, as demonstrated in prostate cancer research:

This methodology represents "the largest survey of prostate cancer-associated antibodies to date" and demonstrates how antibody profiling can characterize protein classes recognized by patients and determine how they change with disease burden .

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.