APG3 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to APG3 Antibody

The APG3 antibody targets the autophagy-related protein 3 (ATG3/APG3), a ubiquitin-like-conjugating enzyme critical for autophagosome formation during autophagy. This conserved process mediates the degradation and recycling of cellular components, playing roles in development, cancer, and neurodegenerative diseases . APG3 antibodies are widely used in research to study autophagy mechanisms, protein localization, and disease associations.

Protein Overview

  • Molecular Weight: ~36 kDa (calculated), though observed bands often appear at 36–40 kDa due to post-translational modifications .

  • Domains: Contains a catalytic cysteine residue critical for its E2-like activity in conjugating ATG8 (LC3) to phosphatidylethanolamine (PE) .

  • Function: Essential for autophagosome membrane expansion by mediating ATG8 lipidation, a step required for substrate engulfment .

Antibody Specifics

FeatureDetails
ImmunogenRecombinant human APG3 (e.g., amino acids M1–T244) or synthetic peptides .
ClonalityPolyclonal (rabbit) or monoclonal (rabbit) .
Cross-ReactivityHuman, mouse, rat, monkey . No cross-reactivity with unrelated proteins .
ApplicationsWestern blot (WB), immunohistochemistry (IHC), immunofluorescence (IF), ELISA .

Key Uses in Experimental Models

  • Autophagy Regulation: Detecting ATG3 expression under nutrient deprivation or pharmacological treatments (e.g., rapamycin) .

  • Disease Studies: Investigating roles in cancer (e.g., tumor suppression via autophagy) and neurodegenerative disorders .

  • Subcellular Localization: Visualizing ATG3 distribution in cytoplasmic compartments during autophagosome formation .

Recommended Assay Conditions

ApplicationDilution RangeSample Types Validated
WB1:500–1:5000Human HeLa, mouse NIH/3T3, rat C6 lysates .
IHC/IF1:50–1:200Tissue sections and fixed cells .
ELISACustomizable*Serum, cell culture supernatants .

Validation Data

  • Western Blot: Distinct bands at ~36–40 kDa in human (K562, HeLa), mouse (NIH/3T3), and rat (C6) lysates .

  • Immunofluorescence: Cytoplasmic staining consistent with ATG3’s role in autophagosome assembly .

  • Specificity Controls: No off-target binding in knockout cell lines or blocking peptide experiments .

Key Research Findings Using APG3 Antibodies

  1. Mechanistic Insights: ATG3’s catalytic cysteine (Cys264) is essential for LC3-PE conjugation, confirmed via mutagenesis and immunoblotting .

  2. Therapeutic Targets: Elevated ATG3 expression correlates with chemoresistance in cancers, suggesting autophagy inhibition as a treatment strategy .

  3. Neurodegeneration: ATG3 knockout models exhibit accumulated protein aggregates, linking autophagy defects to Alzheimer’s and Parkinson’s diseases .

Challenges and Considerations

  • Post-Translational Modifications: Observed molecular weight discrepancies (e.g., 40 kDa vs. 36 kDa predicted) may reflect phosphorylation or ubiquitination .

  • Species Specificity: While most antibodies cross-react with rodents, validation in non-model organisms requires additional controls .

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
APG3 antibody; RF1 antibody; At3g62910 antibody; T20O10.10Peptide chain release factor APG3 antibody; chloroplastic antibody; Chloroplast ribosome release factor 1 antibody; AtcpRF1 antibody; Protein ALBINO AND PALE GREEN 3 antibody
Target Names
APG3
Uniprot No.

Target Background

Function
This antibody may direct the termination of translation in response to the peptide chain termination codon UAG. It is essential for normal chloroplast development and plays critical roles in the termination of translation within plastids.
Gene References Into Functions
  1. The APG3 gene encodes Chloroplast ribosome release factor 1, which is crucial for the chloroplast translational machinery. [Chloroplast ribosome release factor 1] [AtcpRF1] PMID: 17450416
Database Links

KEGG: ath:AT3G62910

STRING: 3702.AT3G62910.1

UniGene: At.28015

Protein Families
Prokaryotic/mitochondrial release factor family
Subcellular Location
Plastid, chloroplast.
Tissue Specificity
Expressed in roots, leaves and stems.

Q&A

What is the difference between APG3 and ATG3 antibodies?

APG3 and ATG3 refer to the same protein, with ATG3 (Autophagy Related 3 homolog) being the current standard nomenclature while APG3 (Autophagy-related protein 3) is an older designation. These terms are used interchangeably in the literature and commercial antibody descriptions . When searching for relevant antibodies, researchers should query both names to ensure comprehensive results. The protein serves as an E2 conjugating enzyme that catalyzes the conjugation of ATG8-like proteins to phosphatidylethanolamine, a critical step in autophagosome formation during autophagy . All antibodies labeled as either APG3 or ATG3 target the same protein and its associated functions in autophagy pathways.

Which applications are validated for APG3/ATG3 antibodies?

APG3/ATG3 antibodies have been validated for numerous experimental applications, with specificity depending on the particular antibody clone and format. Based on the technical information available, the following applications have been verified:

Antibody TypeValidated ApplicationsDilution Recommendations
Monoclonal (OTI3H2)Western Blot, Flow Cytometry, Immunohistochemistry, Immunocytochemistry/Immunofluorescence, CyTOFWB: 1:500-2000, FC: 1:100, IHC: 1:150, ICC/IF: 1:100
Polyclonal (Rabbit)Immunohistochemistry, Immunohistochemistry-ParaffinIHC: 1:200-1:500, IHC-P: 1:200-1:500
Polyclonal (Rabbit)Western BlotAs specified in product documentation

Researchers should note that pre-experimental optimization is still recommended despite these suggested dilutions, as factors like tissue type, fixation methods, and detection systems can influence optimal antibody concentration .

How should APG3/ATG3 antibodies be stored for maximum stability?

Proper storage of APG3/ATG3 antibodies is critical for maintaining their specificity and activity. Most APG3/ATG3 antibodies are supplied in lyophilized format or in solution with stabilizers. For long-term storage, the following guidelines should be observed:

  • Lyophilized antibodies should be stored at -20°C until reconstitution .

  • After reconstitution with appropriate buffer (typically PBS pH 7.3), short-term storage at 4°C is acceptable for up to one month .

  • For extended storage after reconstitution, aliquoting and freezing at -20°C is recommended to avoid repeated freeze-thaw cycles that can damage antibody structure and function .

  • Some formulations contain stabilizers like trehalose (4 mg per vial) and are supplemented with salts like NaCl (0.9 mg) and Na2HPO4 (0.2 mg) to maintain antibody integrity .

Researchers should strictly avoid repeated freeze-thaw cycles as this significantly reduces antibody performance in downstream applications .

How can I optimize ATG3/APG3 antibody performance for detecting autophagy in different cell types?

Optimizing ATG3/APG3 antibody performance for autophagy detection across diverse cell types requires careful consideration of several experimental parameters:

  • Cell-type specific expression levels: ATG3/APG3 expression varies between tissues, with human testis showing high expression and skeletal muscle exhibiting relatively low expression . This requires adjustment of antibody concentration based on the expected protein abundance in your specific cell type.

  • Fixation and antigen retrieval optimization: For immunohistochemistry applications, heat-induced epitope retrieval (HIER) at pH 6 is specifically recommended for paraffin-embedded tissues . Different cell types may require modified fixation protocols to preserve ATG3 epitopes.

  • Detection system sensitivity adjustment: In cells with lower ATG3 expression, amplification systems like tyramide signal amplification may be necessary to visualize protein localization.

  • Validation with knockout controls: Include ATG3 knockout or knockdown samples as negative controls to confirm antibody specificity in your experimental system, especially when working with novel cell types.

  • Co-localization studies: For comprehensive autophagy analysis, combine ATG3 detection with other autophagy markers (LC3, p62) to distinguish between different stages of the autophagic process in specialized cell types.

For western blotting applications, particular attention should be paid to sample preparation, as ATG3's role in membrane dynamics means its extraction efficiency can vary between cell types with different membrane compositions .

What are the critical considerations when using APG3/ATG3 antibodies for studying the relationship between autophagy and cancer?

When investigating autophagy-cancer connections using APG3/ATG3 antibodies, researchers must address several complex methodological considerations:

ATG3/APG3 has been specifically indicated in research areas focused on both autophagy and cancer , reflecting the protein's potential significance in cancer biology and as a possible therapeutic target. When designing experiments, researchers should consider the specific cancer context and potential differences in autophagy regulation between cancer subtypes.

How can I effectively discriminate between autophagy induction and inhibition when monitoring ATG3/APG3 in experimental systems?

Distinguishing between autophagy induction and inhibition requires sophisticated experimental approaches beyond simple ATG3 level measurement:

  • Temporal dynamics assessment: Monitor ATG3 levels at multiple timepoints following experimental manipulation. Autophagy induction typically shows transient ATG3 recruitment to autophagosomal membranes, while inhibition may cause persistent accumulation.

  • Protein complex formation analysis: ATG3 functions through interactions with ATG7 and the ATG12-ATG5-ATG16L1 complex. Co-immunoprecipitation experiments using ATG3 antibodies can reveal whether these functional complexes are forming (induction) or disrupted (inhibition).

  • Lipidation assay integration: Since ATG3 catalyzes LC3/GABARAP lipidation, parallel monitoring of LC3-I to LC3-II conversion provides context for interpreting ATG3 levels. The ratio between these activities indicates whether autophagy proceeds normally.

  • Membrane fraction enrichment: ATG3 redistributes to membrane fractions during active autophagy. Cell fractionation followed by western blotting with ATG3 antibodies in cytosolic versus membrane fractions can reveal functional engagement in the autophagy pathway.

  • Orthogonal validation: Combine ATG3 antibody-based detection with orthogonal autophagy monitoring techniques, such as transmission electron microscopy or tandem fluorescent-tagged LC3 reporters, to comprehensively assess autophagic status.

This multi-parametric approach provides more reliable interpretation than relying solely on ATG3 levels, which may not change dramatically despite significant shifts in autophagy dynamics .

What are the recommended approaches for validating APG3/ATG3 antibody specificity?

Rigorous validation of APG3/ATG3 antibody specificity is essential for generating reliable research data. The following comprehensive validation protocol is recommended:

  • Molecular weight verification: Confirm detection of the expected 35.7 kDa band corresponding to ATG3/APG3 in western blot applications . Any additional bands should be carefully investigated for potential splice variants or post-translational modifications.

  • Genetic knockout/knockdown controls: Test antibody reactivity in samples with CRISPR-Cas9 mediated ATG3 knockout or siRNA-mediated knockdown. Complete elimination or substantial reduction of signal confirms specificity.

  • Peptide competition assay: Pre-incubate the antibody with excess immunizing peptide (when available) before application to samples. Specific signals should be substantially reduced or eliminated.

  • Cross-species reactivity assessment: Evaluate antibody performance across species using samples from human, mouse, rat, and other relevant model organisms. This verifies the conservation of the recognized epitope and confirms reported species reactivity claims .

  • Recombinant protein controls: Include purified recombinant ATG3 protein as a positive control to establish detection sensitivity and linearity range of the antibody.

  • Cross-validation with multiple antibodies: Compare results using antibodies recognizing different epitopes of ATG3/APG3 (e.g., N-terminal vs. C-terminal regions) to ensure consistent detection patterns.

The immunogen information provided for specific antibodies can guide validation approaches. For instance, the NBP2-37894 antibody was developed against a recombinant protein corresponding to a specific amino acid sequence of human APG3, which shares 96% sequence identity with mouse and rat APG3 .

How should I design experiments to study the role of ATG3/APG3 in specialized autophagy processes like mitophagy?

Designing experiments to investigate ATG3/APG3 in specialized autophagy processes requires careful planning and execution:

  • Targeted subcellular localization analysis: Use immunofluorescence with the appropriate ATG3/APG3 antibody dilution (typically 1:100) in combination with mitochondrial markers (TOM20, COXIV) to assess ATG3 recruitment to damaged mitochondria during mitophagy .

  • Mitophagy induction protocol standardization: Establish consistent mitophagy triggers (CCCP, Antimycin A/Oligomycin, or hypoxia) with standardized duration and concentration to ensure reproducible ATG3 recruitment patterns.

  • Temporal resolution studies: Design time-course experiments (0-24 hours post-induction) with ATG3 immunostaining to capture the dynamic recruitment and dissociation phases during mitophagy progression.

  • Genetic modification approach: Implement CRISPR-Cas9 editing to introduce mutations in ATG3's membrane-sensing domain or catalytic site, followed by immunodetection with validated antibodies to correlate structural features with functional roles in mitophagy.

  • Mechanistic pathway dissection: Use immunoprecipitation with ATG3 antibodies to identify mitophagy-specific interaction partners that may differ from general autophagy processes.

  • Quantitative assessment: Develop quantitative image analysis protocols to measure the percentage of mitochondria co-localized with ATG3 and subsequent clearance rates under various experimental conditions.

When studying these specialized processes, researchers should note that ATG3 functions as both an enzyme and a membrane curvature sensor, facilitating LC3/GABARAP lipidation by detecting local membrane stress during autophagosome formation .

What factors influence antibody performance in detecting the cleaved form of APG3/ATG3?

Detecting cleaved forms of APG3/ATG3 presents unique challenges requiring specific methodological considerations:

  • Epitope-specific antibody selection: Use antibodies specifically designed to recognize cleaved epitopes, such as the cleaved-specific APG3 antibody described in source , which targets the 90-104 amino acid region exposed after proteolytic processing.

  • Sample preparation optimization: Proteolytic fragments are often unstable or present at low abundance. Incorporate protease inhibitors during sample preparation and minimize time between sample collection and processing to preserve cleaved forms.

  • Denaturing conditions adjustment: Modified SDS-PAGE conditions may be necessary, as cleaved fragments can behave differently under standard conditions. Consider using gradient gels (4-20%) to effectively resolve both full-length (35.7 kDa) and cleaved fragments.

  • Detection system sensitivity enhancement: Use high-sensitivity detection methods (enhanced chemiluminescence or fluorescent secondary antibodies) to visualize potentially low-abundance cleaved forms.

  • Positive control inclusion: Generate positive controls by treating samples with known inducers of APG3 cleavage to confirm the antibody's ability to detect the cleaved form. This provides a reference band pattern for comparison with experimental samples.

  • Cross-reactivity elimination: Validate the absence of cross-reactivity with other autophagy-related proteins that might undergo similar cleavage events, particularly in stress conditions that trigger multiple proteolytic cascades.

The biochemical context is important to consider - APG3/ATG3 cleavage may occur under specific stress conditions or during particular phases of autophagy, so experimental timing is critical for successful detection .

How should researchers interpret quantitative changes in ATG3/APG3 levels during autophagy modulation experiments?

Accurate interpretation of ATG3/APG3 level changes requires comprehensive analytical approaches:

  • Baseline normalization: Establish cell-type specific baseline levels of ATG3/APG3 before intervention, as expression varies significantly between tissues. Human testis shows high expression while skeletal muscle exhibits low expression .

  • Context-dependent analysis: Interpret ATG3/APG3 changes in the context of other autophagy markers. Increased ATG3 without corresponding changes in LC3-II or autophagic flux may indicate compensatory upregulation rather than enhanced autophagy.

  • Temporal dynamics consideration: Analyze ATG3/APG3 levels across multiple timepoints, as transient changes may be missed in single-timepoint experiments. ATG3 involvement in autophagy typically shows phase-dependent patterns corresponding to autophagosome formation dynamics.

  • Subcellular fraction analysis: Differentiate between total cellular ATG3 levels and its membrane-associated fraction, as functional autophagy induction often involves redistribution rather than expression changes.

  • Statistical robustness: Apply appropriate statistical analyses for time-series data, such as repeated measures ANOVA with post-hoc tests, when analyzing ATG3 level changes over time .

This analytical framework helps distinguish between changes in ATG3/APG3 levels that reflect functional autophagy modulation versus compensatory or non-specific responses to experimental conditions.

What approaches should be used to analyze ATG3/APG3 activity in relation to disease states and therapeutic interventions?

Analyzing ATG3/APG3 in disease contexts requires specialized approaches beyond standard expression analysis:

  • Disease-specific baseline establishment: Compare ATG3/APG3 levels and localization between affected and control tissues using immunohistochemistry with optimized antibody dilutions (1:200-1:500) . Distinguish between changes in expression versus activity.

  • Functional complex analysis: Assess ATG3's incorporation into active autophagy complexes using co-immunoprecipitation with ATG7 and ATG12-ATG5-ATG16L1 components, which reflects its functional engagement rather than mere presence.

  • Post-translational modification profiling: Evaluate disease-associated modifications of ATG3 (phosphorylation, ubiquitination) that may alter its activity without changing expression levels, using modification-specific antibodies when available.

  • Therapeutic response monitoring: Implement time-course analyses following therapeutic intervention, applying mathematical modeling approaches similar to antibody response models to characterize ATG3 activity dynamics.

  • Multivariate pattern analysis: Apply multivariate statistical methods to correlate ATG3 patterns with disease progression markers, particularly in chronic conditions where autophagy dysregulation develops gradually.

  • Patient stratification approach: Analyze ATG3 patterns across patient subgroups to identify potential responder/non-responder signatures for autophagy-modulating therapeutics.

For therapeutic interventions specifically targeting autophagy, researchers should implement comprehensive ATG3 functional assays rather than relying solely on expression levels to accurately assess treatment efficacy.

What emerging methodologies might improve ATG3/APG3 detection and functional analysis?

Several cutting-edge approaches are poised to enhance ATG3/APG3 research:

  • Proximity labeling technologies: Implementing BioID or APEX2 fusions with ATG3 would allow identification of transient interaction partners during specific autophagy phases, providing deeper functional insights than conventional co-immunoprecipitation with ATG3 antibodies.

  • Super-resolution microscopy integration: Combining highly specific ATG3 antibodies with techniques like STORM or PALM could reveal previously undetectable spatial organization of ATG3 during autophagosome formation at nanoscale resolution.

  • Single-cell ATG3 dynamics: Adapting antibody-based detection for single-cell analysis would address the heterogeneity in autophagy responses that is often masked in population-level studies, similar to the heterogeneity observed in antibody responses .

  • Multiplex protein modification analysis: Developing approaches to simultaneously detect multiple post-translational modifications of ATG3 would provide integrated understanding of its regulation under various conditions.

  • In situ proximity ligation assays: Implementing PLA with ATG3 antibodies and antibodies against interaction partners would allow visualization of specific protein complexes in their native cellular context, overcoming limitations of biochemical fractionation.

  • CRISPR-based endogenous tagging: Generating knock-in cell lines with minimal epitope tags on endogenous ATG3 would enable live-cell imaging of physiological ATG3 dynamics without overexpression artifacts.

These methodologies promise to address current limitations in understanding ATG3's spatiotemporal dynamics and context-specific functions in different autophagy pathways.

How can researchers effectively integrate ATG3/APG3 antibody-based studies with other autophagy research approaches?

Effective integration of ATG3/APG3 antibody data with broader autophagy research requires systematic methodological coordination:

  • Multidimensional experimental design: Combine antibody-based detection of ATG3 with complementary approaches like transcriptomics and metabolomics to create integrated autophagy profiles across experimental conditions.

  • Standardized positive controls: Incorporate well-characterized autophagy inducers (rapamycin, starvation) and inhibitors (bafilomycin A1, chloroquine) alongside experimental treatments to establish reference ATG3 response patterns.

  • Systems biology framework application: Position ATG3 antibody data within computational models of autophagy pathways to predict system-wide effects of observed ATG3 changes.

  • Correlative microscopy approaches: Link antibody-based localization of ATG3 at the light microscopy level with ultrastructural analysis of autophagosome formation using correlative light and electron microscopy (CLEM).

  • Cross-platform validation protocol: Establish validation workflows where ATG3 antibody findings are systematically confirmed using orthogonal methods like mass spectrometry-based proteomics or CRISPR screening.

  • Data integration repositories: Contribute standardized ATG3 antibody data to community resources and databases to facilitate meta-analyses across different experimental systems and conditions.

This integrated approach places ATG3 antibody-based research within the broader context of autophagy biology, enhancing the mechanistic insights derived from individual experiments and contributing to a more comprehensive understanding of autophagy regulation.

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.