ATL78 belongs to the Arabidopsis Tóxicos en Levadura (ATL) family of RING-H2 E3 ubiquitin ligases. These proteins regulate stress adaptation by tagging target substrates for proteasomal degradation. Key features include:
Structure: Contains a transmembrane domain, RING-H2 zinc finger motif, and variable C-terminal regions .
Function: Modulates responses to cold, drought, and hormonal signaling pathways .
Role: AtATL78 acts as a negative regulator of cold tolerance.
Role: AtATL78 is a positive regulator of drought tolerance.
While no direct ATL78 antibody is described, studies on plant antibodies highlight methodologies relevant to ATL78 research:
Success Rate: ~55% of recombinant protein-derived antibodies detect targets reliably .
Affinity Purification: Critical for improving signal-to-noise ratios .
Studies using antibodies against ATL homologs (e.g., ATL2, β-III tubulin) reveal mechanisms applicable to ATL78:
ATL2: Cysteine-138 in the RING domain is essential for E3 ligase activity. Mutations (C138A) abolished antifungal resistance in transgenic plants .
β-III Tubulin: Antibody [2G10] (ab78078) demonstrated specificity across species (human, rat, chicken) and applications (IHC, flow cytometry) .
ATL78 interacts with:
RAV2: Binds the ShATL78L promoter to regulate stress-induced transcription .
CSN5B: A COP9 signalosome component that modulates ubiquitination dynamics .
ATL78 (AtATL78) is an Arabidopsis RING E3 ubiquitin ligase localized at the plasma membrane that plays opposing roles in stress responses. It functions as a negative regulator of cold stress response and a positive regulator of drought stress response in Arabidopsis. This dual regulatory role makes it a significant target for understanding plant adaptation mechanisms to environmental stresses . The protein is particularly interesting because suppression of AtATL78 increases tolerance to cold stress while decreasing tolerance to drought, highlighting its potential importance in agricultural applications focused on enhancing crop resilience .
When designing antibodies against ATL78, researchers should consider that it is a membrane-localized RING E3 ubiquitin ligase with specific domains that may affect epitope accessibility. The protein contains a RING finger domain essential for its E3 ligase activity, a transmembrane domain for plasma membrane localization, and regions involved in substrate recognition . Antibodies should ideally target unique, accessible epitopes that do not interfere with functional domains if the goal is to detect the native protein without affecting its activity. Structural modeling and sequence analysis should be employed to identify amino acid residues that may affect antibody affinity, stability, and specificity .
ATL78 expression is differentially regulated by various environmental stresses. RT-PCR and promoter-GUS assays have revealed that AtATL78 is up-regulated by cold stress but down-regulated by drought conditions . This opposing expression pattern correlates with its contrasting functions in these stress responses. During evolutionary development in Brassicaceae lineages, ATL78 acquired enhanced expression through the insertion of a TATA box within the core promoter region after a short tandem duplication event . Understanding these expression patterns is crucial when designing experiments using ATL78 antibodies, as protein levels will vary significantly depending on the stress conditions applied to the plant samples.
For developing highly specific antibodies against ATL78, researchers should employ a multi-faceted approach combining in silico analysis with experimental validation. Begin with computational sequence analysis to identify unique peptide regions that distinguish ATL78 from other ATL family members, particularly AtATL81, its close homolog . Consider using a combination of monoclonal antibodies targeting different epitopes to enhance specificity. Apply phylogenetic and structural modeling to identify amino acid residues that may affect affinity, stability, and expression yield . For optimal results, employ systematic design of experiment (DoE) technology to explore sequence-function relationships through testing 48-400 antibody variants over 1-4 iterations to identify the most specific and high-performing antibody candidates .
Optimizing purification of ATL78 antibodies requires a systematic approach addressing expression, purification, and formulation challenges. Initially, optimize codons, vectors, and expression systems to generate high-producing stable pools, particularly important if dealing with poorly expressing antibodies . For purification, apply a sequential process including affinity chromatography (using Protein A/G columns), followed by ion-exchange chromatography to remove contaminants, and size-exclusion chromatography to eliminate aggregates. Characterize the purified antibody using analytics for quantity (A280 reading, Bradford assay), purity (SDS-PAGE, reverse phase chromatography), identity (Western Blot, mass spectrometry), and functional potency (ELISA, Octet-based assays) . Finally, optimize formulation to allow concentration without aggregation, which is particularly important for concentrated antibody applications requiring >100 mg/ml solutions .
Validating ATL78 antibody specificity in plant tissues requires multiple complementary approaches to ensure accurate results. First, perform Western blot analysis comparing wild-type Arabidopsis with atl78 knockout/knockdown lines, expecting absence or reduction of signal in the mutant samples . Include cross-reactivity tests against related ATL family proteins, particularly AtATL81, to confirm specificity . Second, conduct immunoprecipitation followed by mass spectrometry to verify that the antibody captures the intended target. Third, perform immunolocalization studies to confirm that the antibody detects ATL78 at its known plasma membrane location . Finally, use the antibody in plants exposed to cold and drought conditions to verify that detected protein levels correspond with known expression patterns (increased under cold, decreased under drought) . Document all validation data comprehensively, including positive and negative controls, as these will be essential for interpreting experimental results.
ATL78 antibodies can serve as powerful tools to investigate the opposing roles of ATL78 in cold and drought stress responses through several methodological approaches. First, use immunoblotting to quantify ATL78 protein levels at different time points during cold and drought stress to establish temporal expression profiles that can be correlated with physiological responses . Second, employ co-immunoprecipitation with ATL78 antibodies followed by mass spectrometry to identify interaction partners that may differ between cold and drought conditions, potentially revealing divergent signaling pathways. Third, combine with chromatin immunoprecipitation (ChIP) studies of transcription factors known to regulate stress responses to determine whether ATL78 influences their DNA binding or stability. Fourth, perform immunolocalization studies under different stress conditions to determine if ATL78 subcellular distribution changes in response to cold versus drought . These approaches will help elucidate how a single protein can function as both a negative regulator of cold stress and a positive regulator of drought responses .
When designing experiments to study ubiquitination targets of ATL78 using antibodies, several critical considerations must be addressed. First, implement a proteasome inhibitor treatment (e.g., MG132) prior to protein extraction to prevent degradation of ubiquitinated targets, as these are often rapidly processed by the 26S proteasome. Second, use denaturing conditions during cell lysis to disrupt protein-protein interactions and inactivate deubiquitinating enzymes that might remove ubiquitin from targets. Third, employ a dual immunoprecipitation approach: first with ATL78 antibodies to pull down the E3 ligase complex, then with anti-ubiquitin antibodies to enrich for ubiquitinated proteins within this complex . Fourth, include appropriate controls, including atl78 knockout/knockdown plants and plants subjected to opposing stress conditions (cold vs. drought), to identify stress-specific ubiquitination targets . Finally, validate potential targets using in vitro ubiquitination assays with recombinant ATL78 and candidate proteins to confirm direct enzyme-substrate relationships.
ATL78 antibodies can provide unique insights into evolutionary adaptations of plant stress responses through comparative studies across species. First, utilize the antibodies in cross-species Western blot analyses to examine conservation and expression levels of ATL78 homologs across different Brassicaceae species, correlating findings with each species' natural habitat conditions . Second, perform immunoprecipitation studies comparing ATL78 and its homologs (particularly ATL81) across species to identify evolutionary shifts in protein-protein interaction networks that may reflect adaptations to specific environmental niches . Third, combine antibody-based protein quantification with promoter analyses to investigate how changes in promoter architecture (such as the TATA box insertion observed in ATL78) correlate with protein expression levels and stress tolerance phenotypes across the Brassicaceae lineage . Fourth, use immunohistochemistry to compare tissue-specific localization patterns of ATL78 across species, potentially revealing evolutionary shifts in where and when this regulatory protein functions in response to environmental challenges.
Detecting ATL78 in plant samples presents several challenges that require specific troubleshooting approaches. First, the membrane localization of ATL78 necessitates effective membrane protein extraction protocols; use detergent-based buffers (e.g., 1% Triton X-100 or 0.5% SDS) to solubilize membrane proteins effectively, and consider subcellular fractionation to enrich plasma membrane proteins . Second, the differential expression under stress conditions may lead to very low protein levels under certain conditions (particularly drought); in such cases, employ sample concentration techniques and highly sensitive detection methods like chemiluminescence or fluorescence-based Western blotting . Third, potential cross-reactivity with related ATL family members might occur; overcome this by using highly specific monoclonal antibodies and including appropriate controls like atl78 mutant plants . Fourth, post-translational modifications might mask epitopes; address this by using multiple antibodies targeting different regions of ATL78 and/or including phosphatase treatment during sample preparation if phosphorylation is suspected to interfere with antibody binding.
When faced with discrepancies between ATL78 protein levels (detected via antibodies) and mRNA levels (from RT-PCR or transcriptome analysis), researchers should systematically analyze potential explanations through several approaches. First, consider that post-transcriptional and post-translational regulation may cause protein and mRNA levels to diverge; examine possible miRNA regulation of ATL78 transcripts or protein stability factors like deubiquitinating enzymes . Second, verify antibody specificity using appropriate controls to rule out false positive/negative signals; include wild-type and atl78 mutant samples in parallel . Third, assess temporal dynamics, as protein levels often lag behind transcriptional changes; conduct time-course experiments measuring both mRNA and protein levels at multiple time points after stress application . Fourth, examine compartmentalization, as transcripts measured in whole-tissue extracts may not reflect protein levels in specific subcellular locations; combine cell fractionation with immunoblotting to determine if localization changes affect detected protein levels . Finally, quantitatively analyze the relationship between transcript and protein data using statistical methods to determine if the discrepancy represents a biologically relevant regulatory mechanism rather than technical variation.
For robust analysis of quantitative data from ATL78 antibody-based experiments, researchers should implement a multi-layered statistical framework. First, perform normalization of immunoblot data using appropriate housekeeping proteins that remain stable under the tested stress conditions (actin may not be ideal as cytoskeletal proteins can change under stress; consider membrane proteins like H+-ATPase for normalization when studying membrane-localized ATL78) . Second, apply linear mixed-effects models to account for both fixed effects (e.g., treatment, genotype) and random effects (e.g., biological replicates, technical variations) when comparing ATL78 protein levels across conditions. Third, conduct power analysis before experiments to determine the minimum sample size needed to detect biologically relevant differences in ATL78 levels, particularly important given its differential expression under stress conditions . Fourth, employ non-parametric tests when data do not meet normality assumptions, which is common with protein quantification data. Finally, use multivariate analyses (e.g., principal component analysis) when examining relationships between ATL78 levels and multiple physiological parameters or when comparing results across different antibody-based techniques (Western blot, ELISA, immunoprecipitation).
To investigate the mechanistic basis of ATL78's opposing functions in stress responses, researchers should implement a comprehensive antibody-based experimental strategy. First, perform stress-specific interactome analysis using co-immunoprecipitation with ATL78 antibodies under cold and drought conditions, followed by mass spectrometry to identify condition-specific protein interactions that may explain the divergent regulatory roles . Second, employ proximity-labeling techniques (BioID or APEX) fused to ATL78 under different stress conditions to capture transient interactions that might be missed by traditional co-IP approaches. Third, use phospho-specific antibodies to determine if post-translational modifications of ATL78 differ between cold and drought conditions, potentially switching its function from negative to positive regulation . Fourth, combine ChIP-seq of stress-responsive transcription factors with ATL78 immunoprecipitation data to establish whether ATL78 differentially regulates key transcription factors under opposing stress conditions. Finally, implement quantitative ubiquitination profiling using tandem ubiquitin-binding entities (TUBEs) in combination with ATL78 antibodies to compare ubiquitination targets under cold versus drought stress, potentially revealing stress-specific proteolytic regulation .
Developing phospho-specific antibodies for ATL78 requires careful consideration of several factors to ensure specificity and utility in regulatory studies. First, conduct in silico analysis of ATL78 sequence to identify potential phosphorylation sites, focusing on serine, threonine, and tyrosine residues in regions that might influence E3 ligase activity or protein-protein interactions . Second, prioritize sites based on conservation analysis across plant species and predictive algorithms for kinase recognition motifs that might respond to stress signaling pathways. Third, employ mass spectrometry analysis of immunoprecipitated ATL78 from plants under various stress conditions to experimentally verify phosphorylation sites before antibody development . Fourth, when designing phospho-specific antibodies, ensure that peptide antigens include 5-10 amino acids flanking the phosphorylated residue and consider using multiple rabbits for immunization to increase chances of obtaining high-affinity antibodies . Fifth, implement rigorous validation protocols including testing against phosphatase-treated samples and phospho-mimetic/phospho-dead ATL78 mutants. Finally, characterize antibody performance across different technical applications (Western blot, immunoprecipitation, immunofluorescence) to determine optimal conditions for each use case in studying stress-dependent phosphorylation of ATL78.
ATL78 antibodies can be powerful tools for investigating the interplay between ubiquitination and other post-translational modifications (PTMs) through several sophisticated approaches. First, employ sequential immunoprecipitation protocols where samples are first immunoprecipitated with ATL78 antibodies, then with antibodies against specific PTMs (phosphorylation, SUMOylation, acetylation) to identify modified forms of ATL78 or its substrates . Second, develop a PTM-specific interactome analysis by comparing protein interaction partners of ATL78 before and after inducing specific modifications (e.g., using kinase activators or inhibitors) to determine how these modifications influence E3 ligase complex assembly or substrate recognition under different stress conditions . Third, utilize proximity ligation assays (PLA) with ATL78 antibodies paired with PTM-specific antibodies to visualize and quantify co-occurrence of modifications in situ at subcellular resolution. Fourth, combine ATL78 immunoprecipitation with targeted mass spectrometry approaches like parallel reaction monitoring (PRM) to quantitatively profile changes in modification patterns under cold versus drought stress . Finally, implement CRISPR-based gene editing to introduce tag sequences at the endogenous ATL78 locus, allowing affinity purification under native conditions to preserve physiologically relevant modifications for comprehensive PTM mapping.
To resolve contradictions in published data on ATL78 function across different plant species, researchers should implement a systematic comparative methodology using antibody-based approaches. First, develop a panel of antibodies with verified cross-reactivity across species of interest, potentially targeting highly conserved epitopes in the RING domain while confirming specificity against close homologs like ATL81 . Second, perform side-by-side analysis of protein expression, localization, and interaction partners across species under identical experimental conditions, controlling for developmental stage, tissue type, and stress parameters . Third, conduct comparative ubiquitination assays using immunoprecipitated ATL78 from different species to determine if substrate specificity varies across evolutionary lineages, potentially explaining functional divergence . Fourth, implement reciprocal genetic complementation experiments where ATL78 from different species is expressed in atl78 mutant Arabidopsis, followed by antibody-based confirmation of protein expression and functional analysis of stress tolerance phenotypes . Finally, use quantitative proteomics with species-specific ATL78 antibodies to compare absolute protein levels across species, as differences in expression levels rather than intrinsic protein function might explain some contradictory findings in stress response studies.