The ATL12 antibody targets the Arabidopsis Toxicos en Levadura 12 (ATL12) protein, a RING-H2 zinc finger ubiquitin E3 ligase implicated in plant stress responses. ATL12 regulates pathways associated with salt tolerance, reactive oxygen species (ROS) signaling, and abscisic acid (ABA)-mediated stress responses in Arabidopsis thaliana . While commercial ATL12-specific antibodies are not explicitly detailed in available literature, research-grade antibodies are critical for detecting ATL12 protein expression in genetic and molecular studies, such as Western blotting and immunofluorescence .
ATL12 enhances salt tolerance by modulating ROS production and ion extrusion mechanisms. Overexpression of ATL12 in Arabidopsis increases the expression of salt-responsive genes (e.g., SOS1, SOS2) and ABA-dependent pathways (e.g., RD29B), while atl12 mutants exhibit reduced ROS generation under NaCl stress .
| Parameter | Wild-Type (Col-0) | atl12 Mutant | ATL12 Overexpression Line |
|---|---|---|---|
| ROS production (150 mM NaCl) | Moderate | Reduced | Elevated |
| RD29B expression | Baseline | Downregulated | Upregulated |
| Salt tolerance | Normal | Reduced | Enhanced |
ATL12 negatively regulates ABA-dependent responses. Mutants show delayed germination, reduced root elongation, and altered stomatal aperture under ABA treatment, while overexpression lines exhibit heightened sensitivity to ABA .
Antibodies against ATL12 are essential for:
Protein Localization: Confirming plasma membrane localization via immunofluorescence .
Expression Analysis: Validating ATL12 levels in transgenic lines (e.g., overexpression or knockout mutants) using Western blot .
ROS Quantification: Correlating ATL12 expression with ROS dynamics in stress assays .
Western Blot:
Overexpression Lines: Show 5x higher ATL12 transcript levels vs. wild-type, linked to increased ROS and salt tolerance .
Mutants (atl12): Exhibit 50% reduction in CERK1 expression, impairing chitin-induced defense responses .
ATL12 integrates hormonal (ABA) and pathogen-responsive (chitin) signaling. Chitin treatment induces ATL12 expression by 3-fold within 2 hours, enhancing ROS bursts via NADPH oxidases .
KEGG: ath:AT2G20030
STRING: 3702.AT2G20030.1
ATG12 (Autophagy-related protein 12) is a ubiquitin-like protein with a molecular weight of approximately 15kDa that plays a critical role in autophagy vesicle formation. This protein functions through conjugation with ATG5 via a ubiquitin-like conjugating system that also involves ATG7 (acting as an E1-like activating enzyme) and ATG10 (functioning as an E2-like conjugating enzyme). The resulting ATG12-ATG5 conjugate operates as an E3-like enzyme, which is essential for the lipidation of ATG8 family proteins and their subsequent association with vesicle membranes. This mechanism makes ATG12 indispensable for researchers studying autophagy pathways, cellular stress responses, and related pathologies .
Polyclonal ATG12 antibodies (such as rabbit polyclonal variants) recognize multiple epitopes on the ATG12 protein, providing stronger signals but potentially lower specificity. These antibodies are particularly valuable in applications where protein detection might be challenging due to low expression levels. In contrast, monoclonal ATG12 antibodies (like the mouse monoclonal IgG2b variant) recognize a single epitope, offering higher specificity but potentially lower sensitivity. Monoclonal antibodies (e.g., clone 43CT73.3.5.5.4) are preferred for experiments requiring consistent lot-to-lot reproducibility and when studying specific conformational states of ATG12. For differential experimental requirements, polyclonal antibodies typically work well in multiple applications including ELISA and Western blotting, while monoclonal antibodies may provide more reliable results in applications like immunohistochemistry where background reduction is critical .
Validation of ATG12 antibody specificity requires a multi-step approach. Initially, Western blotting should be performed with both positive controls (cell lines known to express ATG12, such as human cell lines for human-reactive antibodies) and negative controls (ATG12 knockout cells or tissues). Researchers should verify detection of the expected 15kDa band (free ATG12) and ~55kDa band (ATG12-ATG5 conjugate). For additional validation, immunoprecipitation followed by mass spectrometry can confirm that the antibody is pulling down ATG12 and its binding partners. RNA interference experiments (siRNA or shRNA against ATG12) should show reduced antibody signal. Finally, cross-reactivity testing against similar proteins in the ATG family is essential to ensure specificity. With certain antibodies showing cross-reactivity with yeast samples, researchers must carefully evaluate reactivity across species when designing experiments with evolutionarily conserved autophagy pathways .
For investigating autophagy in viral infection models using ATG12 antibodies, researchers should implement a systematic approach. Begin by establishing baseline ATG12-ATG5 conjugate levels in uninfected cells through quantitative Western blotting (1:4000 dilution recommended). Upon viral infection (particularly with hepatitis C virus or vesicular stomatitis virus), monitor temporal changes in ATG12 expression and conjugation at strategic time points (0, 2, 6, 12, 24 hours post-infection). Crucially, implement co-immunoprecipitation studies using ATG12 antibodies to capture interaction partners during infection progression, as ATG12 has been demonstrated to function as a proviral factor in HCV infection by facilitating the translation of incoming viral RNA during initial infection stages. Additionally, perform confocal microscopy with fluorescently-labeled ATG12 antibodies to track the subcellular redistribution of ATG12-ATG5 complexes during infection. This approach is particularly important given that ATG12, in association with ATG5, has been shown to negatively regulate innate antiviral immune responses by impairing type I interferon production pathways during vesicular stomatitis virus infection .
When studying autophagy in cancer progression using ATG12 antibodies, researchers must first address several methodological challenges. Begin by selecting antibodies validated in cancer tissues of interest, as ATG12 expression varies significantly across cancer types. Implement multiplexed immunofluorescence protocols combining ATG12 antibodies with markers for cancer progression (e.g., Ki-67) and other autophagy proteins (ATG5, LC3) to correlate autophagy dynamics with malignant transformation. Critical considerations include: (1) using phospho-specific antibodies to distinguish activated forms of ATG12-ATG5 complexes; (2) comparing conjugated versus free ATG12 ratios across tumor grades; (3) carefully controlling for fixation artifacts, as autophagy proteins are sensitive to fixation conditions; and (4) validating findings across multiple methodologies (Western blot, immunohistochemistry, and flow cytometry). Since autophagy can function both as a tumor suppressor and promoter depending on context, researchers should include appropriate controls representing both early and late-stage cancers when analyzing ATG12 expression patterns. Consider correlating findings with clinical parameters such as those observed in Adult T-cell leukemia/lymphoma (ATL) patients, where autophagy pathway alterations may influence disease progression .
Interpreting ATG12 antibody signals in relation to ATG5 conjugation presents several technical challenges. First, researchers must distinguish between free ATG12 (~15kDa) and the ATG12-ATG5 conjugate (~55kDa) using appropriate gel separation techniques (12-15% gels recommended). The antibody's epitope location is critical - antibodies targeting regions involved in the conjugation interface may preferentially detect free ATG12 over conjugated forms, leading to misinterpretation of conjugation efficiency. Additionally, the stoichiometric relationship between ATG12 and ATG5 can be altered under different cellular stresses, requiring quantitative analysis rather than qualitative assessment. For accurate interpretation, implement dual-color Western blotting with both ATG12 and ATG5 antibodies, and calculate conjugation efficiency as the ratio of conjugated to free forms of both proteins. Importantly, protein extraction methods significantly impact conjugate stability, with RIPA buffers potentially disrupting weak protein interactions. Use gentler lysis conditions (NP-40 based buffers) and avoid repeated freeze-thaw cycles of samples. Finally, researchers should consider that ATG12-ATG5 conjugates localize differently than free ATG12, requiring distinct protocols for cytoplasmic versus membrane-associated fraction analysis .
Designing proper controls for autophagy flux assays with ATG12 antibodies requires a multi-layered approach. First, include technical controls: (1) primary antibody-only and secondary antibody-only controls to assess non-specific binding; (2) loading controls using housekeeping proteins not affected by autophagy manipulation (β-actin is preferred over GAPDH, which can be affected by autophagy). For biological controls, researchers must include: (1) ATG12 knockout or knockdown samples as negative controls; (2) samples treated with autophagy inducers (rapamycin or starvation) as positive controls for increased ATG12-ATG5 conjugation; (3) samples treated with bafilomycin A1 to block autophagosome-lysosome fusion for accumulation of autophagy components. Critically, time-course experiments should be conducted to distinguish between increased autophagy initiation versus blockade of autophagy completion, as both can result in elevated ATG12-ATG5 conjugate levels. For flux assays specifically, compare samples with and without lysosomal inhibitors to determine the rate of autophagy progress rather than static levels of ATG12. Finally, include comparative controls with other autophagy markers (LC3-II, p62/SQSTM1) to comprehensively assess autophagy dynamics .
Preserving ATG12 epitopes for immunodetection requires careful consideration of sample preparation methods. For cell lysates, use phosphate-buffered extraction with non-denaturing detergents (0.5% NP-40 or 1% Triton X-100) rather than harsh RIPA buffers that may disrupt protein conformation. Include protease inhibitors (PMSF, leupeptin, aprotinin) and phosphatase inhibitors (sodium fluoride, sodium orthovanadate) to prevent degradation and maintain post-translational modifications. For tissue samples, flash-freezing in liquid nitrogen followed by mechanical homogenization preserves protein integrity better than chemical homogenization methods. When performing immunohistochemistry or immunofluorescence, use paraformaldehyde fixation (4%, maximum 10 minutes) followed by gentle permeabilization (0.1% Triton X-100, 5 minutes) to maintain epitope accessibility. Antigen retrieval methods must be optimized - for ATG12 detection, citrate buffer (pH 6.0) with heat-induced epitope retrieval typically yields superior results compared to EDTA buffer systems. For long-term storage, maintain purified antibodies at -20°C in small aliquots to prevent freeze-thaw cycles, as recommended for the monoclonal ATG12 antibody produced against recombinant APG12L protein .
Accurate quantification of ATG12-ATG5 conjugate versus free ATG12 requires a systematic analytical approach. First, optimize protein separation using gradient gels (4-20%) to clearly resolve the ~15kDa free ATG12 band from the ~55kDa ATG12-ATG5 conjugate. For Western blotting quantification, use fluorescently-labeled secondary antibodies rather than chemiluminescence detection to ensure signal linearity across a wider dynamic range. Crucial for accuracy is the inclusion of recombinant ATG12 protein standards at known concentrations (5-100ng range) to generate a calibration curve for absolute quantification. For relative quantification, calculate the conjugation ratio (CR) using the formula: CR = [ATG12-ATG5]/([ATG12-ATG5] + [free ATG12]), which provides a normalized measure of conjugation efficiency independent of total ATG12 expression levels. When analyzing tissue samples, implement subcellular fractionation to separately assess cytosolic versus membrane-associated conjugates, as their distributions provide insight into autophagy progression. For immunoprecipitation-based quantification, use antibodies against ATG5 to pull down the conjugate followed by ATG12 detection to prevent bias from free ATG12 pools. Finally, consider using proximity ligation assays (PLA) in intact cells to visualize and quantify the spatial distribution of ATG12-ATG5 interactions with greater sensitivity than conventional co-localization studies .
For investigating viral infection and autophagy interactions, researchers should implement a multi-modal approach using ATG12 antibodies. Begin with time-course experiments examining ATG12-ATG5 conjugate levels following infection with viruses known to manipulate autophagy (e.g., hepatitis C virus, which requires ATG12 for initial viral RNA translation). Use immunofluorescence microscopy with co-staining of viral proteins and ATG12 to track the spatial reorganization of autophagy machinery during infection progression. Critically, implement RNA silencing experiments targeting ATG12 to assess its proviral versus antiviral functions in different infection systems - this is particularly important as ATG12 has been shown to negatively regulate the innate antiviral immune response by impairing type I interferon production during vesicular stomatitis virus infection. For mechanistic studies, use co-immunoprecipitation with ATG12 antibodies followed by mass spectrometry to identify novel viral protein interactions with the autophagy machinery. Additionally, researchers should examine post-translational modifications of ATG12 during infection using phospho-specific antibodies, as viral manipulation often occurs through altered phosphorylation states of autophagy proteins. When studying chronic viral infections like HTLV-1 (associated with Adult T-cell leukemia), correlate ATG12 expression patterns with viral protein expression and disease progression markers to elucidate long-term consequences of autophagy manipulation .
When performing immunohistochemistry with ATG12 antibodies on pathological tissues, researchers must address several critical issues. First, tissue fixation protocols significantly impact epitope accessibility - formalin-fixed paraffin-embedded (FFPE) tissues require optimized antigen retrieval (heat-mediated citrate buffer at pH 6.0 is generally effective for ATG12). For frozen sections, brief fixation (2-4% PFA for 10 minutes) maintains optimal epitope preservation. Tissue-specific considerations are essential, as baseline ATG12 expression varies dramatically across tissues - liver and neuronal tissues typically show higher baseline autophagy than connective tissues. When examining diseased tissues (particularly cancers or neurodegenerative conditions), include adjacent normal tissue as internal controls within the same section to normalize for staining variability. For multiplex staining, carefully select antibody combinations to avoid cross-reactivity - pair ATG12 antibodies with markers of specific cell types (CD3 for T-cells in lymphoma samples, for example) and other autophagy markers (LC3, p62) to comprehensively assess autophagy status. Quantification requires standardized approaches - implement digital pathology analysis using consistent thresholding parameters across samples, and report both staining intensity and percentage of positive cells. Finally, validate key findings with orthogonal methods (Western blotting of tissue lysates) to confirm specificity, particularly in tissues with high autofluorescence or endogenous peroxidase activity that may confound interpretation .
Analyzing ATG12 involvement in neurodegenerative disease-related autophagy dysregulation requires specialized methodological approaches. Begin with comparative quantification of ATG12-ATG5 conjugate levels in affected versus unaffected brain regions using carefully controlled Western blotting (at 1:4000 dilution for optimal signal-to-noise ratio). For neurodegenerative disease models, implement immunofluorescence co-localization studies examining the spatial relationship between ATG12 and disease-specific protein aggregates (tau, amyloid-β, α-synuclein). Analysis should include both biochemical fractionation to separate soluble from insoluble protein fractions and proximity ligation assays to detect direct interactions between ATG12 and neurodegenerative disease-associated proteins. When working with human post-mortem brain tissues, account for post-mortem interval effects on autophagy proteins by implementing standardized tissue collection protocols and documenting intervals. Time-course studies in disease models are essential for determining whether ATG12 alterations represent cause or consequence of pathology progression. Additionally, researchers should examine ATG12 in the context of the broader autophagy pathway by simultaneously assessing multiple autophagy markers across disease stages. Since the ATG12-ATG5 conjugate is essential for proper autophagosome formation, defects in this conjugation system may represent a critical point of autophagy dysfunction in neurodegenerative conditions characterized by protein aggregation and impaired clearance mechanisms .
When faced with conflicting ATG12 antibody results across experimental platforms, researchers should implement a systematic troubleshooting approach. First, thoroughly evaluate antibody characteristics - epitope location significantly influences detection patterns, as antibodies targeting regions involved in protein interactions may show differential accessibility in native versus denatured states. This explains discrepancies between techniques that utilize denatured proteins (Western blot) versus native conformations (immunoprecipitation). Second, verify that observed molecular weights are consistent with expectations (15kDa for free ATG12; 55kDa for the ATG12-ATG5 conjugate); anomalous bands may indicate cross-reactivity, proteolytic fragments, or post-translationally modified variants. Third, analyze buffer compatibility, as certain detergents or salt concentrations may disrupt epitope recognition in specific applications. Fourth, consider cell/tissue-specific ATG12 expression patterns and isoforms that may be preferentially detected by different antibodies. To systematically resolve discrepancies, researchers should: (1) perform side-by-side comparison using multiple antibodies against different ATG12 epitopes; (2) validate results using genetic approaches (siRNA knockdown) to confirm specificity; (3) implement recombinant protein controls expressing tagged ATG12 variants; and (4) compare results across multiple cell types to identify context-dependent patterns. Importantly, researchers should recognize that certain experimental conditions (nutrient status, stress levels) dynamically alter ATG12 conjugation states, potentially explaining temporal inconsistencies in results .
Common sources of false positives when using ATG12 antibodies include: (1) cross-reactivity with other ATG family members that share structural homology with ATG12; (2) non-specific binding to denatured proteins exposing similar epitopes; (3) excessive antibody concentration leading to background signal; (4) inappropriate secondary antibody selection causing cross-species reactivity; and (5) endogenous peroxidase or phosphatase activity in tissue samples confounding enzymatic detection methods. False negatives frequently result from: (1) epitope masking due to protein-protein interactions or post-translational modifications; (2) insufficient antigen retrieval in fixed tissues; (3) degradation of ATG12 during sample preparation; (4) excessively stringent washing conditions disrupting antibody-antigen binding; and (5) sample buffer incompatibility affecting epitope conformation. For preventing false results, researchers should: (1) validate antibodies using positive and negative controls including ATG12 knockout samples; (2) optimize antibody concentration through titration experiments; (3) include blocking peptides to confirm specificity; (4) implement multiple detection methods to corroborate findings; and (5) carefully control experimental conditions that affect autophagy (serum starvation, cell confluence) which can dramatically alter ATG12 expression and conjugation states. Additionally, researchers should be aware that certain fixation methods can generate artifacts that resemble autophagy structures, necessitating careful interpretation of morphological data .
Distinguishing between altered ATG12 expression and changed conjugation efficiency requires specialized analytical approaches. Implement quantitative PCR to measure ATG12 mRNA levels in parallel with protein analysis to determine if changes originate at the transcriptional level. For protein analysis, use Western blotting with careful sample preparation to preserve both free and conjugated forms, and calculate the conjugation ratio (CR = conjugated ATG12/total ATG12) to normalize for expression differences. Critical controls include cycloheximide-treated samples to inhibit new protein synthesis (revealing conjugation/deconjugation dynamics) and proteasome inhibitors to block protein degradation (revealing turnover rates). For accurate interpretation, perform pulse-chase experiments with metabolic labeling to track newly synthesized ATG12 incorporation into conjugates versus degradation of free ATG12. Additionally, use proximity ligation assays to visualize and quantify ATG12-ATG5 interactions in situ, allowing spatial analysis of conjugation events in relation to autophagosome formation sites. Importantly, researchers should compare results across multiple time points, as autophagy is highly dynamic, with rapid fluctuations in both expression and conjugation rates occurring in response to cellular stressors. When analyzing patient samples (such as from ATL patients), consider that chronic disease states may show compensatory changes in both expression and conjugation efficiency, requiring careful interpretation in relation to disease progression markers .
ATG12 antibodies are increasingly utilized to investigate non-canonical functions of autophagy proteins beyond classical macroautophagy. Researchers should implement co-immunoprecipitation experiments with ATG12 antibodies followed by mass spectrometry to identify novel interaction partners in non-autophagy contexts. Of particular interest is the role of ATG12 in viral infection responses, where the ATG12-ATG5 conjugate has been demonstrated to negatively regulate innate antiviral immunity by suppressing type I interferon production during vesicular stomatitis virus infection. For these studies, researchers should perform chromatin immunoprecipitation using ATG12 antibodies to identify potential transcriptional regulatory roles in immune response genes. Additionally, subcellular fractionation followed by immunoblotting is essential to track the redistribution of ATG12 to non-autophagic compartments (mitochondria, endoplasmic reticulum, nucleus) under different cellular conditions. For investigating ATG12's role in cell death pathways, implement live-cell imaging with fluorescently-tagged ATG12 antibody fragments to monitor real-time localization during apoptosis induction. When examining potential roles in cancer biology, correlate ATG12 expression and localization patterns with markers of cell cycle regulation, as emerging evidence suggests interconnections between autophagy proteins and cell proliferation control. Importantly, researchers should validate antibody specificity in each subcellular compartment, as conformational changes or post-translational modifications may alter epitope accessibility in different cellular environments .
Recent methodological advancements have significantly enhanced ATG12 detection in complex samples. Proximity extension assays (PEA) combine antibody specificity with nucleic acid amplification, allowing ultrasensitive ATG12 quantification in minimal sample volumes (useful for cerebrospinal fluid or biopsy specimens). Single-molecule array (Simoa) technology enables detection of ATG12 at femtomolar concentrations, facilitating analysis in samples where autophagy proteins are minimally expressed. For improved specificity, researchers are implementing multiple-epitope targeting approaches using antibody pairs recognizing distinct ATG12 regions to reduce false positives. Advances in mass spectrometry-based approaches include selective reaction monitoring (SRM) for targeted ATG12 peptide quantification without antibody dependence, particularly valuable for modified forms not recognized by available antibodies. For spatial analysis, multiplexed ion beam imaging (MIBI) and imaging mass cytometry (IMC) combine antibody specificity with mass spectrometry resolution, enabling simultaneous visualization of multiple autophagy proteins in tissue sections with subcellular resolution. When implementing these advanced methods, researchers should validate results against conventional approaches like Western blotting, while recognizing that each technique reveals distinct aspects of ATG12 biology. For example, antibody-based methods detect specific epitopes that may be masked in certain protein conformations, while mass spectrometry approaches identify post-translational modifications potentially missed by antibody recognition. Ultimately, complementary methodologies provide the most comprehensive understanding of ATG12 dynamics in complex biological systems .