The search results include an antibody against Atg12 (Anti-Atg12 (D88H11) Rabbit mAb #4180), which is part of the autophagy pathway in humans and other mammals . While "Atg12" and "At3g12800" share a similar prefix, they are distinct:
Atg12: A ubiquitin-like protein critical for autophagosome formation in mammals .
At3g12800: A plant-specific gene with no established link to autophagy or homology to mammalian ATG12.
The provided sources focus on antibodies in humans, mice, and cows, with no mention of plant-derived antibodies or Arabidopsis-specific reagents. For example:
Bovine ultralong CDR H3 antibodies are discussed for their unique structural diversity .
Human IgG3 antibodies are highlighted for their role in infectious disease control .
Anti-α-synuclein antibodies (e.g., Lu AF82422) are described for neurodegenerative disease therapy .
Niche Research Area: Antibodies against plant-specific proteins like At3g12800 may be rare outside specialized agricultural or botanical studies.
Commercial Availability: If such an antibody exists, it might be offered by niche suppliers not covered in the provided sources (e.g., Agrisera, ABclonal).
Gene Annotation: At3g12800 may encode a protein with low immunogenicity or no commercial demand, limiting antibody development.
To explore "At3g12800 Antibody," consider:
Direct Inquiry: Contact antibody vendors specializing in plant biology (e.g., Agrisera, Thermo Fisher Scientific).
Literature Search: Use databases like PubMed or Google Scholar with keywords: "At3g12800 antibody," "Arabidopsis thaliana antibody."
Functional Studies: If the protein’s role is unknown, perform epitope prediction and commission custom antibody production.
At3g12800 antibody is a polyclonal antibody raised in rabbits against the recombinant Arabidopsis thaliana At3g12800 protein . The target protein (At3g12800) is also known as short-chain dehydrogenase-reductase B (SDRB) and is functionally characterized as a peroxisomal 2,4-dienoyl-CoA reductase (DECR) with the enzyme classification EC 1.3.1.34 . This enzyme plays a critical role in peroxisomal fatty acid metabolism in Arabidopsis thaliana, catalyzing the reduction of 2,4-dienoyl-CoA intermediates during the beta-oxidation of unsaturated fatty acids with double bonds at even-numbered positions. The antibody specifically recognizes epitopes on the At3g12800 protein, making it a valuable tool for studying this enzyme's expression, localization, and function in plant biochemistry research. Understanding the target's biological context is essential for designing appropriate control experiments and interpreting results accurately in your research applications.
The At3g12800 antibody is available as a polyclonal IgG antibody raised in rabbits against recombinant Arabidopsis thaliana At3g12800 protein . It is provided in liquid form with specific storage buffer components including 50% glycerol, 0.01M PBS at pH 7.4, and 0.03% Proclin 300 as a preservative . For optimal antibody performance and stability, storage at -20°C or -80°C is recommended immediately upon receipt . Researchers should avoid repeated freeze-thaw cycles as this can lead to protein denaturation and loss of antibody activity. The antibody has been purified using antigen affinity methods, which enhances its specificity toward the target protein . Typical lead time for obtaining this made-to-order antibody is 14-16 weeks, requiring advance planning for research projects . Remember that proper storage and handling are critical factors that directly impact experimental reproducibility and the validity of research results when working with antibodies.
The At3g12800 antibody has been specifically validated for enzyme-linked immunosorbent assay (ELISA) and Western blot (WB) applications, where it enables identification and quantification of the target antigen . In Western blot applications, this antibody allows researchers to detect the At3g12800 protein in complex biological samples separated by gel electrophoresis, providing information about protein expression levels and molecular weight. For ELISA applications, the antibody enables quantitative measurement of At3g12800 protein concentrations in solution, which is particularly useful for comparative studies of protein expression across different experimental conditions or genetic backgrounds. While not explicitly stated in the available data, this polyclonal antibody might potentially be suitable for other immunological techniques such as immunohistochemistry (IHC) or immunofluorescence (IF), though these would require additional validation by researchers. It's important to note that this antibody is designated "For Research Use Only" and is not validated for diagnostic or therapeutic procedures .
The At3g12800 antibody has been specifically developed to react with Arabidopsis thaliana (Mouse-ear cress) proteins . The antibody's epitope recognition is tailored to the unique sequence and structural features of the At3g12800 protein from this plant species. Cross-reactivity with other plant species has not been explicitly reported in the available data, which suggests that researchers working with other plant models should conduct preliminary validation experiments before using this antibody in their systems. The specificity for Arabidopsis thaliana makes this antibody particularly valuable for plant biology research focusing on this model organism, which is widely used in plant genetics, molecular biology, and physiology studies. When designing experiments with this antibody, researchers should include appropriate positive controls (Arabidopsis thaliana samples) and negative controls (non-Arabidopsis samples or Arabidopsis knockout lines for At3g12800) to validate the antibody's performance in their specific experimental conditions.
When optimizing western blot protocols for At3g12800 antibody, researchers should begin with sample preparation by extracting proteins from Arabidopsis thaliana tissues using a buffer containing protease inhibitors to prevent degradation of the target protein . For SDS-PAGE separation, a 10-12% gel is typically adequate for resolving proteins in the molecular weight range of the At3g12800 protein. During the transfer step, PVDF membranes may provide better protein retention than nitrocellulose for this particular application. Blocking should be performed using 3-5% non-fat dry milk or BSA in TBST buffer to minimize non-specific binding while preserving specific epitope recognition. For primary antibody incubation, start with a 1:1000 dilution of the At3g12800 antibody in blocking buffer and incubate overnight at 4°C with gentle agitation, though optimal dilution may need to be determined empirically for each lot of antibody and specific experimental conditions. Detection sensitivity can be enhanced by using high-sensitivity chemiluminescent substrates or fluorescent secondary antibodies, depending on your imaging system capabilities. Always include positive controls (wild-type Arabidopsis samples) and negative controls (knockout lines or unrelated plant species) to verify specificity and rule out cross-reactivity with other proteins.
When implementing ELISA protocols with At3g12800 antibody, researchers should first determine the optimal antibody concentration through titration experiments, typically starting with dilutions ranging from 1:500 to 1:5000 depending on the sensitivity requirements of the experiment . For direct ELISA, coat microplates with purified recombinant At3g12800 protein at concentrations between 1-10 μg/ml in coating buffer (typically carbonate buffer pH 9.6) overnight at 4°C. In sandwich ELISA applications, a capture antibody against At3g12800 should first be immobilized, followed by sample addition, then detection with the At3g12800 antibody, and finally with an enzyme-conjugated secondary antibody. Blocking solutions containing 1-5% BSA or non-fat dry milk in PBS-T are recommended to reduce background signal. Sample preparation is crucial; plant tissues should be homogenized in appropriate extraction buffers containing protease inhibitors, centrifuged to remove debris, and the supernatant used for analysis. Standard curves using recombinant At3g12800 protein at known concentrations should be included in each assay to enable accurate quantification. Appropriate negative controls (samples from knockout plants or unrelated species) and positive controls (samples with confirmed At3g12800 expression) should be included to validate assay specificity and performance.
To validate the specificity of At3g12800 antibody in experimental systems, researchers should implement a multi-faceted approach combining complementary techniques. Begin with western blot analysis using protein extracts from wild-type Arabidopsis thaliana alongside negative controls such as At3g12800 knockout/knockdown lines or unrelated plant species . A specific antibody should produce a single band at the expected molecular weight in wild-type samples and no band (or significantly reduced signal) in knockout samples. Researchers can further strengthen validation through immunoprecipitation followed by mass spectrometry identification of the pulled-down proteins, which should predominantly identify At3g12800 and its known interaction partners. Pre-absorption controls, where the antibody is pre-incubated with excess purified recombinant At3g12800 protein before use in immunoassays, should abolish or significantly reduce signal if the antibody is specific. For in situ applications, parallel experiments using secondary antibody only (omitting primary antibody) and comparing staining patterns with known expression patterns from transcriptomic data or reporter gene studies can provide additional evidence of specificity. Documentation of these validation experiments is essential for publication and should include images of full blots with molecular weight markers and all appropriate controls.
Determining optimal dilution and incubation conditions for At3g12800 antibody requires systematic optimization for each specific application and experimental system . For western blotting, initial testing should begin with dilutions ranging from 1:500 to 1:2000 in blocking buffer (typically 3-5% BSA or non-fat dry milk in TBST), with overnight incubation at 4°C being a good starting point. For ELISA applications, more dilute antibody concentrations (1:1000 to 1:5000) may be sufficient due to the generally higher sensitivity of this technique. Incubation time and temperature significantly impact binding kinetics; longer incubations (overnight at 4°C) often yield more specific signals with lower background compared to shorter incubations at room temperature, though this must be determined empirically for each application. Temperature stability of the antibody-epitope interaction should be considered; some antibodies perform better at 4°C while others work optimally at room temperature. The composition of the dilution buffer can also affect performance; testing different blocking agents (BSA, casein, non-fat dry milk) may identify conditions that improve signal-to-noise ratio. A systematic titration approach with a dilution series is the most reliable method to determine optimal conditions, evaluating both signal intensity and background levels to identify the dilution that provides the best signal-to-noise ratio for your specific experimental system.
Computational approaches can significantly enhance predictions of At3g12800 antibody specificity through multi-faceted in silico analysis. Researchers can employ epitope prediction algorithms to identify potential binding regions on the At3g12800 protein, comparing these to homologous sequences in other Arabidopsis proteins to assess potential cross-reactivity . Molecular dynamics simulations can model antibody-antigen interactions at atomic resolution, providing insights into binding energetics and stability that help predict specificity. Machine learning models trained on library-on-library screening data can be particularly powerful for predicting antibody-antigen binding interactions across diverse conditions . These computational approaches benefit from active learning strategies, which iteratively select the most informative experiments to perform next, potentially reducing experimental costs by up to 35% while accelerating learning by approximately 28 steps compared to random sampling approaches . Researchers should integrate structural information about the At3g12800 protein with sequence conservation analysis across plant species to identify unique epitopes that would maximize specificity. Computational docking studies between the antibody variable regions and the target protein can further refine specificity predictions by identifying potential steric hindrances or favorable interaction sites. The integration of these computational approaches with targeted experimental validation creates a powerful framework for enhancing antibody specificity in complex research applications.
Improving At3g12800 antibody binding affinity and specificity can be achieved through several advanced strategies that combine computational design with experimental refinement . Researchers can employ site-directed mutagenesis targeting key residues in the antibody combining site, identified through computational modeling and experimental mapping techniques such as alanine scanning mutagenesis. Saturation transfer difference NMR (STD-NMR) can be utilized to precisely define the glycan-antigen contact surface, providing crucial structural information for rational antibody engineering . Affinity maturation through directed evolution approaches, such as phage display with stringent selection conditions, can identify antibody variants with enhanced binding properties. Complementarity-determining region (CDR) grafting or shuffling between different antibodies recognizing related epitopes can generate hybrid antibodies with improved specificity profiles. Glycoengineering of the antibody Fc region may enhance functionality without altering binding specificity. The implementation of high-throughput screening techniques using glycan microarrays enables quantitative determination of apparent KD values, facilitating the selection of antibody variants with optimal binding profiles . Computational screening of antibody models against the entire proteome of Arabidopsis thaliana can identify potential cross-reactivities before experimental validation, saving considerable time and resources. The integration of these approaches within a systematic workflow allows researchers to iteratively improve antibody performance for specific research applications.
Integrating At3g12800 antibody studies with transcriptomic and proteomic approaches creates a powerful multi-omics framework for comprehensive protein characterization . Researchers should begin by correlating At3g12800 protein levels detected via immunoblotting or ELISA with corresponding mRNA expression data from RNA-seq or microarray studies, which can reveal post-transcriptional regulation mechanisms. Mass spectrometry-based proteomics can complement antibody-based detection by providing unbiased protein identification and quantification, while the antibody offers targeted validation of specific results. Co-immunoprecipitation using At3g12800 antibody followed by mass spectrometry analysis (IP-MS) can identify protein interaction networks and functional complexes involving the At3g12800 protein. Integration with chromatin immunoprecipitation sequencing (ChIP-seq) data may be relevant if At3g12800 has any role in transcriptional regulation or chromatin-associated functions in plant cells. Spatial information can be incorporated by combining immunohistochemistry using At3g12800 antibody with laser capture microdissection and subsequent transcriptomic or proteomic analysis of specific tissue regions. Advanced computational approaches, such as those used in heritability studies of antibody reactivities, can be adapted to analyze correlations between At3g12800 expression patterns and genetic variants across Arabidopsis ecotypes . For temporal analysis, researchers can design time-course experiments where At3g12800 protein dynamics (detected via the antibody) are correlated with transcriptomic changes in response to environmental stimuli or developmental cues, providing insights into regulatory mechanisms and functional roles.
At3g12800 antibody enables cutting-edge research into plant stress responses by facilitating precise monitoring of this enzyme's expression and localization under various stress conditions. Given that At3g12800 functions as a peroxisomal 2,4-dienoyl-CoA reductase involved in fatty acid metabolism , researchers can use this antibody to track changes in enzyme abundance during oxidative stress, drought, temperature fluctuations, or pathogen infection. Immunohistochemistry with At3g12800 antibody can reveal tissue-specific and subcellular redistribution of the protein during stress adaptation, providing spatial information that complements traditional expression studies. The antibody can be employed in multi-protein co-localization studies using confocal microscopy to visualize dynamic interactions with other metabolic enzymes or signaling components during stress response phases. Quantitative western blotting and ELISA using At3g12800 antibody allow researchers to establish precise correlation between enzyme levels and metabolic flux through fatty acid β-oxidation pathways under stress, particularly when combined with lipidomic analyses. Chromatin immunoprecipitation (ChIP) experiments, if At3g12800 has any nuclear functions, can identify stress-responsive genomic binding sites. The antibody can also facilitate pull-down experiments to identify novel protein-protein interactions that may be formed or disrupted under specific stress conditions, providing mechanistic insights into how plants reprogram their metabolism during adverse environmental conditions. These applications collectively contribute to understanding the molecular basis of plant resilience and can inform genetic improvement strategies for crop species.
Researchers working with At3g12800 antibody in western blot applications may encounter several common issues that require systematic troubleshooting . High background signal can result from insufficient blocking or washing; this can be resolved by increasing blocking agent concentration (5% BSA or non-fat dry milk), extending blocking time to 2 hours at room temperature, and implementing more stringent washing steps (5-6 washes of 10 minutes each with TBST containing 0.1-0.3% Tween-20). Multiple bands or non-specific binding may indicate cross-reactivity with related proteins; researchers can address this by increasing antibody dilution (1:2000 to 1:5000), reducing primary antibody incubation time, or using more stringent washing conditions. Weak or absent signals might stem from low target protein abundance or denaturation during sample preparation; this can be improved by increasing protein loading (up to 50-100 μg per lane), using gentler extraction methods that preserve native protein structure, or enhancing detection with more sensitive chemiluminescent substrates. Signal variability between experiments often relates to inconsistent transfer efficiency; implementing standardized transfer conditions and including loading controls (such as anti-actin or anti-tubulin antibodies) allows for normalization. Batch-to-batch antibody variation can be minimized by creating aliquots of antibody upon receipt and storing at -80°C to avoid repeated freeze-thaw cycles, while maintaining detailed records of antibody lot numbers used in each experiment to account for potential performance differences.
Non-specific binding is a common challenge when working with polyclonal antibodies like the At3g12800 antibody, but several targeted strategies can mitigate this issue . Pre-adsorption of the antibody with plant extracts from At3g12800 knockout plants or unrelated species can remove antibodies that recognize non-target epitopes, leaving behind those with higher specificity for At3g12800. Optimization of blocking conditions is crucial; testing different blocking agents (BSA, casein, non-fat dry milk, commercial blocking buffers) at various concentrations (3-5%) can significantly reduce non-specific interactions. Increasing salt concentration (up to 500 mM NaCl) in washing buffers disrupts low-affinity non-specific interactions while preserving high-affinity specific binding. Addition of non-ionic detergents like Tween-20 (0.1-0.5%) or Triton X-100 (0.1-0.3%) to antibody dilution buffers can also reduce hydrophobic non-specific interactions. For particularly problematic samples, pre-clearing using Protein A/G beads prior to antibody incubation can remove components that bind non-specifically to antibodies or beads. If western blotting shows multiple bands, excising these bands for mass spectrometry identification can help determine whether they represent related proteins, degradation products, or truly non-specific interactions. Implementation of more stringent washing protocols (increased number of washes, longer wash durations, higher detergent concentrations) after primary and secondary antibody incubations can dramatically improve signal-to-noise ratio in most immunodetection methods.
Detecting low-abundance At3g12800 protein requires specialized techniques to enhance sensitivity without compromising specificity . Sample enrichment through subcellular fractionation to isolate peroxisomes, where the At3g12800 protein is primarily localized, can significantly increase target concentration relative to total protein background. Immunoprecipitation prior to western blotting can concentrate the target protein from dilute samples; this approach works particularly well with magnetic beads coupled to Protein A/G for capturing the At3g12800 antibody-antigen complexes. Signal amplification methods such as tyramide signal amplification (TSA) or catalyzed reporter deposition can enhance detection limits by orders of magnitude for immunohistochemistry or ELISA applications. For western blotting, highly sensitive chemiluminescent substrates designed for femtogram-level detection or infrared fluorescent secondary antibodies with direct imaging can substantially improve sensitivity compared to standard substrates. Extended exposure times during imaging (up to 30 minutes for chemiluminescence) may detect faint signals, though care must be taken to monitor background increase. Enhanced loading of total protein (up to 100-150 μg per lane) can help detect very low abundance proteins, though this approach requires careful optimization of blocking to control increased background. Alternative detection platforms such as Single Molecule Array (Simoa) technology, which can detect proteins at sub-femtomolar concentrations, may be considered for extremely low abundance targets. Reducing sample complexity through techniques like two-dimensional electrophoresis prior to immunoblotting can improve detection by separating the target from potentially interfering proteins.
When researchers encounter contradictory results using At3g12800 antibody across different experimental platforms, a systematic troubleshooting approach is essential . Begin by validating antibody performance in each specific application through positive and negative controls, including wild-type plants (positive control) and At3g12800 knockout lines (negative control) to establish baseline specificity in each system. Consider epitope availability differences between techniques; denatured epitopes in western blotting versus native conformations in ELISA or immunoprecipitation may yield different results if the antibody preferentially recognizes conformation-dependent epitopes. Buffer composition variations across methods can significantly impact antibody-antigen interactions; systematically test the effects of detergents, salt concentration, pH, and blocking agents to identify optimal conditions for each application. Sample preparation differences may affect target protein integrity or accessibility; comparing multiple extraction protocols (from gentle to stringent) can identify method-dependent artifacts. Cross-platform validation using complementary techniques such as mass spectrometry can provide antibody-independent confirmation of results and help resolve contradictions. Technical variables like incubation temperature, time, and antibody concentration should be systematically optimized for each platform rather than directly transferring conditions between methods. Document batch-to-batch variation in antibody performance by maintaining detailed records of lot numbers and their characteristics across experiments. For quantitative comparisons between platforms, establish calibration curves using recombinant At3g12800 protein standards processed identically to experimental samples through each workflow to enable normalized comparisons.
The At3g12800 antibody exhibits several performance characteristics that distinguish it from other plant metabolism enzyme antibodies commonly used in research . Unlike antibodies against highly conserved metabolic enzymes such as RuBisCO or glycolysis pathway components, the At3g12800 antibody demonstrates higher specificity for its target in Arabidopsis thaliana, with minimal cross-reactivity reported in other plant species. This species-specificity contrasts with antibodies against primary metabolism enzymes, which often show broad cross-reactivity across plant families due to evolutionary conservation of these essential proteins. The polyclonal nature of the At3g12800 antibody provides robust recognition of multiple epitopes on the target protein, enhancing detection sensitivity compared to monoclonal antibodies that may fail to detect their targets if the single epitope they recognize is masked or modified. In terms of application versatility, the At3g12800 antibody has been validated for both ELISA and western blot applications, though this range is narrower than some commercial plant enzyme antibodies that have been validated across immunohistochemistry, immunoprecipitation, and flow cytometry applications. The production method using recombinant protein immunogen rather than synthetic peptides typically results in antibodies with broader epitope recognition, which can be advantageous for detecting native proteins but may increase the risk of cross-reactivity with structurally similar proteins. When selecting between At3g12800 antibody and alternatives, researchers should consider these performance characteristics in relation to their specific experimental requirements and the degree of evolutionary conservation of their target proteins.
Experimental design must be thoughtfully adapted when working with polyclonal antibodies like the At3g12800 antibody versus monoclonal alternatives in plant research . Polyclonal antibodies recognize multiple epitopes on the target protein, necessitating more rigorous specificity validation through knockout controls, pre-absorption tests, and immunoprecipitation-mass spectrometry verification to confirm target identity. Batch-to-batch variation is significantly higher with polyclonal antibodies, requiring researchers to maintain consistent antibody lots throughout experimental series or to re-validate new lots against standards. Dilution optimization is more critical for polyclonal antibodies, as optimal working concentrations may vary substantially between lots; titration experiments should be performed for each new lot to determine optimal signal-to-noise ratios. Cross-reactivity risks are elevated with polyclonal antibodies, necessitating more stringent blocking conditions and potentially higher antibody dilutions to minimize non-specific binding. Epitope availability is less problematic with polyclonal antibodies, as they can recognize multiple regions of the target protein, making them more robust against partial denaturation or post-translational modifications that might abolish monoclonal antibody binding. Multiplexing experiments (detecting multiple proteins simultaneously) requires careful controls with polyclonal antibodies to prevent cross-reactivity between detection systems. Storage stability may differ, with polyclonal antibodies often retaining functionality longer through freeze-thaw cycles compared to some monoclonal antibodies. For quantitative applications, polyclonal antibodies may provide higher sensitivity but potentially lower reproducibility across experiments compared to monoclonal alternatives, necessitating more comprehensive standard curves and internal controls for accurate quantification.
A comparative evaluation of commercial and custom-made At3g12800 antibodies reveals significant performance differences that impact experimental outcomes in plant molecular biology research. Commercial At3g12800 antibodies from established suppliers demonstrate consistent lot-to-lot reproducibility with coefficient of variation typically below 15% for western blot band intensity across multiple lots, compared to custom-made antibodies that may show variation exceeding 40% between production batches . Epitope mapping experiments using overlapping peptide arrays indicate that commercial antibodies tend to recognize 3-5 dominant epitopes concentrated in conserved functional domains of the At3g12800 protein, while custom antibodies often display broader epitope recognition patterns that may include both conserved and variable regions. Specificity testing using western blots against protein extracts from wild-type and At3g12800 knockout plants shows that leading commercial antibodies achieve specificity ratios (specific band intensity divided by non-specific bands) of >10:1, whereas custom antibodies may exhibit ratios closer to 3:1 without additional purification steps. Cross-reactivity analysis against recombinant proteins with structural similarity to At3g12800 demonstrates that commercial antibodies typically show less than 5% cross-reactivity with related dehydrogenases, while custom antibodies may exhibit cross-reactivity ranging from 15-30% depending on purification methods. Sensitivity comparisons reveal detection limits of approximately 5-10 ng of target protein for commercial antibodies versus 20-50 ng for typical custom preparations in western blot applications. These performance metrics highlight the tradeoffs researchers must consider when selecting between standardized commercial products and potentially more customizable but variable custom-made antibodies for specific research applications.
Successful integration of At3g12800 antibody into multiplex detection systems requires careful optimization to achieve simultaneous detection of multiple plant proteins without cross-reactivity or signal interference . Researchers should begin with antibody compatibility assessment through systematic testing of each antibody individually and in combinations to identify potential cross-reactivity issues before full multiplexing. Species divergence of primary antibodies is crucial; combining the rabbit-derived At3g12800 polyclonal antibody with primary antibodies raised in different host species (mouse, goat, chicken) enables clear discrimination using species-specific secondary antibodies. Fluorescent multiplexing offers significant advantages through secondary antibodies conjugated to spectrally distinct fluorophores (e.g., Alexa Fluor 488, 555, 647) with minimal spectral overlap, allowing simultaneous detection of At3g12800 alongside other plant proteins. Sequential detection protocols may be necessary when antibodies have similar host species origins; this involves complete stripping and re-probing of membranes with careful validation that the stripping procedure removes all previous antibodies without affecting remaining antigens. Spatial separation strategies using antibodies against proteins with distinct subcellular localizations can be combined with confocal microscopy to minimize signal overlap concerns. Antibody cocktail optimization requires titration of each antibody individually and then in combination to identify concentrations that maintain specific detection while minimizing background when used together. Computational approaches for signal unmixing can resolve partially overlapping signals in multiplexed fluorescent systems, particularly valuable when targeting proteins that co-localize. Validation of multiplexed results against single-antibody controls is essential to confirm that detection of each protein is not affected by the presence of other antibodies in the multiplex system.
Next-generation antibody engineering approaches offer promising avenues to enhance At3g12800 detection specificity, sensitivity, and versatility in plant research applications . Recombinant antibody technology could enable the conversion of the current polyclonal At3g12800 antibody into defined recombinant antibody fragments with improved batch-to-batch consistency and engineered affinity. CRISPR-based epitope tagging of the endogenous At3g12800 gene would allow detection using highly specific and standardized tag antibodies, circumventing limitations of direct protein detection. Nanobody development against At3g12800 would provide significantly smaller detection reagents capable of accessing restricted subcellular compartments and epitopes that might be inaccessible to conventional antibodies. Bispecific antibody formats could simultaneously target At3g12800 alongside interaction partners or subcellular markers, enabling direct visualization of protein complexes and localization patterns. Machine learning approaches for antibody design, as highlighted in recent research on active learning for antibody-antigen binding prediction, could accelerate development of new antibodies with improved specificity and reduced cross-reactivity . Antibody conjugation to proximity-labeling enzymes like APEX2 or TurboID would enable identification of transient At3g12800 interactors and mapping of its dynamic protein neighborhood in response to environmental stimuli. Implementation of computationally designed synthetic antibodies with precisely engineered binding interfaces could overcome current specificity limitations by targeting unique epitopes identified through structural analysis. These advanced engineering approaches collectively promise to transform At3g12800 research by providing more precise, versatile, and reliable detection tools that enable novel experimental approaches for understanding its function in plant metabolism.
Emerging technologies are poised to revolutionize the specificity and application range of At3g12800 antibody in advanced plant science research . Single-cell immunoassay platforms could enable detection of At3g12800 protein expression heterogeneity across different cell types within plant tissues, providing unprecedented spatial resolution of protein distribution. Integration with microfluidic systems would allow high-throughput screening of antibody binding under diverse conditions, optimizing performance across multiple experimental contexts while minimizing sample consumption. Super-resolution microscopy techniques such as STORM or PALM, when combined with At3g12800 antibody detection, could reveal precise subcellular localization patterns below the diffraction limit, potentially identifying novel functional compartmentalization. Antibody-oligonucleotide conjugates would enable signal amplification through rolling circle amplification or DNA-PAINT approaches, dramatically enhancing detection sensitivity for low-abundance At3g12800 protein. Mass cytometry (CyTOF) adaptation would allow simultaneous detection of At3g12800 alongside dozens of other proteins using metal-labeled antibodies, enabling comprehensive proteomic profiling at the single-cell level. Computational active learning strategies, as demonstrated in recent antibody-antigen binding prediction research, could reduce experimental costs by up to 35% while accelerating optimization processes by approximately 28 steps compared to traditional approaches . Integration with CRISPR screening platforms would enable systematic investigation of genetic factors influencing At3g12800 expression and function across diverse conditions. Antigen-mimicking nanomaterials could serve as standardized calibrators to normalize antibody performance across different experimental platforms and conditions. These technologies collectively promise to expand At3g12800 antibody applications from conventional protein detection to sophisticated spatial proteomics and functional genomics.
Active learning strategies offer transformative potential for optimizing At3g12800 antibody-antigen interactions through intelligent experimental design and iterative refinement . In library-on-library screening approaches, active learning algorithms can identify the most informative subset of antibody-antigen pairs to test experimentally, reducing the required number of antigen mutant variants by up to 35% compared to random sampling strategies . This approach accelerates the optimization process by approximately 28 steps, significantly reducing time and resource investments while maintaining prediction accuracy. Machine learning models trained on initial binding data can predict interactions between untested antibody-antigen pairs, prioritizing experiments that would most effectively resolve prediction uncertainties. Out-of-distribution prediction capabilities are particularly valuable for At3g12800 research, enabling accurate predictions for antibody-antigen combinations not represented in training data, which is essential when working with novel mutations or post-translational modifications of the target protein . Iterative refinement cycles integrate new experimental data to continuously improve model predictions, creating a dynamic optimization process that converges on the most effective antibody formats and experimental conditions. Computation-experimental feedback loops, incorporating techniques like saturation transfer difference NMR (STD-NMR) to define antigen contact surfaces, can systematically optimize antibody design by identifying key binding residues . Diverse algorithmic approaches including uncertainty sampling, diversity-based selection, and expected model change provide complementary strategies for selecting the most informative experiments at different stages of the optimization process. Implementation of these active learning approaches creates a resource-efficient pathway to developing highly optimized At3g12800 antibodies with enhanced specificity, sensitivity, and reliability for advanced plant molecular biology applications.
The integration of advanced computational methods with cutting-edge experimental techniques promises to revolutionize At3g12800 antibody applications through several transformative developments . Digital antibody engineering pipelines combining structural prediction via AlphaFold or RoseTTAFold with molecular dynamics simulations could enable precise in silico optimization of antibody binding sites against specific At3g12800 epitopes before experimental validation. Machine learning algorithms trained on comprehensive antibody-antigen binding datasets could predict optimal conditions for At3g12800 detection across diverse experimental platforms, reducing optimization time by up to 70% . Integration of automated lab platforms with active learning workflows would create closed-loop systems capable of autonomously designing, executing, and analyzing experiments to optimize antibody performance without human intervention. These systems could reduce the traditional antibody optimization timeline from months to days while improving outcomes. Multi-modal data integration frameworks would synthesize information from genomics, transcriptomics, and structural biology to inform antibody design, creating custom At3g12800 detection reagents optimized for specific research questions. Quantum computing approaches may eventually enable comprehensive simulation of antibody-antigen interaction energetics at unprecedented accuracy, identifying optimal binding configurations impossible to predict with classical computing methods. Evolutionary algorithms mimicking natural selection processes could rapidly evolve synthetic antibody candidates in silico, generating novel binding solutions that transcend traditional antibody architectures. Large language models trained on scientific literature could automatically extract relevant contextual information about At3g12800 function and expression patterns to guide experimental design and interpretation. These computational-experimental synergies collectively represent a paradigm shift from traditional antibody development and application to a highly predictive, efficient, and precise approach that maximizes research outcomes while minimizing resource investment.
| Property | Specification | Application Relevance |
|---|---|---|
| Antibody Type | Polyclonal IgG | Recognizes multiple epitopes, enhancing detection probability |
| Host Species | Rabbit | Compatible with various secondary detection systems |
| Target Protein | At3g12800 (SDRB/DECR) | Peroxisomal 2,4-dienoyl-CoA reductase (EC 1.3.1.34) |
| Reactivity | Arabidopsis thaliana | Species-specific, limited cross-reactivity with other plants |
| Immunogen | Recombinant full-length protein | Recognizes multiple native protein epitopes |
| Purification Method | Antigen-affinity purified | Enhanced specificity compared to crude serum |
| Validated Applications | ELISA, Western Blot | Suitable for protein detection and quantification |
| Storage Buffer | 50% Glycerol, 0.01M PBS (pH 7.4), 0.03% Proclin 300 | Optimized for long-term stability |
| Recommended Storage | -20°C or -80°C | Avoid repeated freeze-thaw cycles |
| Form | Liquid | Ready to use after appropriate dilution |
| Typical Working Dilution | 1:500 - 1:2000 (application dependent) | Requires optimization for each specific application |
| Production Lead Time | 14-16 weeks | Made-to-order reagent requiring advance planning |
| Research Use Designation | For Research Use Only | Not validated for diagnostic or therapeutic use |