At4g39550 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (Made-to-order)
Synonyms
At4g39550 antibody; F23K16.180F-box/kelch-repeat protein At4g39550 antibody
Target Names
At4g39550
Uniprot No.

Target Background

Function
This antibody targets At4g39550, a component of SCF (SKP1-cullin-F-box) E3 ubiquitin ligase complexes. These complexes mediate the ubiquitination and subsequent proteasomal degradation of target proteins.
Database Links

KEGG: ath:AT4G39550

UniGene: At.31086

Q&A

What is At4g39550 and why is it significant in Arabidopsis research?

At4g39550 is a gene locus in Arabidopsis thaliana that encodes a protein belonging to the F-box family. F-box proteins in Arabidopsis are characterized by an N-terminal F-box motif followed by other domains such as kelch repeats that mediate protein-protein interactions . The significance of At4g39550 lies in its potential role in the ubiquitin-proteasome pathway, which is crucial for controlled protein degradation in plants. F-box proteins specifically recruit target proteins for ubiquitination through their protein-protein interaction domains . Understanding At4g39550's function can provide insights into regulatory mechanisms controlling various cellular processes in plants, including development, stress responses, and hormone signaling.

What types of antibodies are commonly used for At4g39550 protein detection?

For At4g39550 protein detection, researchers typically employ either polyclonal or monoclonal antibodies, each with distinct advantages depending on the experimental context. Polyclonal antibodies recognize multiple epitopes on the At4g39550 protein, providing robust detection but potentially lower specificity. These are generated by immunizing animals (commonly rabbits) with either the full-length recombinant At4g39550 protein or synthetic peptides corresponding to unique regions of the protein . Monoclonal antibodies, while more challenging to produce, offer higher specificity by recognizing a single epitope, making them valuable for distinguishing At4g39550 from other closely related F-box family members in Arabidopsis . For detecting native protein conformations in techniques like immunoprecipitation, antibodies raised against the full protein are preferable, while peptide antibodies often perform better in applications like Western blotting where proteins are denatured.

How can I validate the specificity of an At4g39550 antibody?

Validating antibody specificity for At4g39550 requires a multi-faceted approach. First, perform Western blot analysis comparing wild-type Arabidopsis tissue with At4g39550 knockout or knockdown lines – a specific antibody should show reduced or absent signal in the mutant lines . Second, conduct peptide competition assays where pre-incubation of the antibody with the immunizing peptide should eliminate signal if the antibody is specific . Third, express tagged recombinant At4g39550 in a heterologous system and verify that both the antibody against At4g39550 and an antibody against the tag detect the same protein band. Fourth, immunoprecipitate At4g39550 followed by mass spectrometry to confirm the identity of the precipitated protein. Finally, evaluate cross-reactivity with other F-box proteins, particularly those with high sequence similarity, to ensure the antibody distinguishes between family members with similar structural motifs .

What are the optimal sample preparation methods for At4g39550 antibody applications?

Optimal sample preparation for At4g39550 antibody applications must account for the protein's characteristics within the F-box protein family. For protein extraction, use a buffer containing 50mM Tris-HCl (pH 7.5), 150mM NaCl, 1% Triton X-100, supplemented with protease inhibitors and 1-5mM of reducing agents like DTT or β-mercaptoethanol to preserve protein structure . Include phosphatase inhibitors if investigating phosphorylation states. For immunoprecipitation and co-immunoprecipitation experiments, gentler non-ionic detergents (0.1-0.5% NP-40 or Digitonin) better preserve protein-protein interactions, particularly important for capturing the association between At4g39550 and its target proteins or ASK1 (Arabidopsis SKP1-like protein) through the F-box domain . For immunohistochemistry, tissue fixation with 4% paraformaldehyde for 2-4 hours followed by careful permeabilization is recommended. To reduce background signal in Arabidopsis tissues with high autofluorescence, include 0.1% sodium borohydride treatment and use longer blocking times (2-3 hours) with 5% BSA or normal serum.

How can At4g39550 antibodies be used to identify protein interaction partners?

Identifying protein interaction partners of At4g39550 requires sophisticated immunoprecipitation strategies leveraging antibody specificity. Begin with co-immunoprecipitation using anti-At4g39550 antibodies in mild lysis conditions (150mM NaCl, 0.5% NP-40) to preserve native protein complexes . This approach can capture both stable and transient interactions with potential target proteins and components of the SCF (Skp1-Cullin-F-box) complex. For detecting weaker or transient interactions, implement proximity-based labeling techniques by generating transgenic Arabidopsis expressing At4g39550 fused to enzymes like BioID or APEX2, followed by immunoprecipitation with specific antibodies . Alternatively, use dual-epitope approaches where reciprocal co-immunoprecipitation with antibodies against both At4g39550 and suspected partner proteins provides stronger evidence for genuine interactions. Mass spectrometry analysis of immunoprecipitated complexes allows unbiased identification of partners, while subsequent validation via yeast two-hybrid or bimolecular fluorescence complementation confirms direct interactions . Importantly, conducting experiments under various physiological conditions can reveal condition-specific interactions relevant to At4g39550's biological functions.

What advanced imaging techniques can be used with At4g39550 antibodies for subcellular localization studies?

Advanced imaging techniques with At4g39550 antibodies can provide high-resolution insights into subcellular localization and dynamics. Super-resolution microscopy methods like Structured Illumination Microscopy (SIM) or Stochastic Optical Reconstruction Microscopy (STORM) overcome the diffraction limit of conventional confocal microscopy, achieving 20-50nm resolution to precisely map At4g39550 localization relative to cellular compartments . For examining dynamic protein behavior, implement Fluorescence Recovery After Photobleaching (FRAP) using fluorescently-tagged secondary antibodies against primary At4g39550 antibodies in permeabilized cells or use live-cell immunofluorescence with membrane-permeable nanobodies. Expansion microscopy physically enlarges fixed specimens after immunolabeling, providing enhanced spatial resolution with standard confocal microscopes. For studying temporal changes in localization, use synchronized Arabidopsis cell cultures with fixed timepoint immunostaining. Combine immunogold labeling with transmission electron microscopy for ultrastructural localization at nanometer resolution. When investigating co-localization with other proteins, apply spectral unmixing algorithms to eliminate bleed-through and quantify overlap using Pearson's or Manders' coefficients for statistical robustness in co-localization claims.

How can we develop a computational model to predict antigen-specific antibodies for At4g39550 protein?

Developing a computational model for predicting antigen-specific antibodies against At4g39550 requires integration of sequence analysis, structural modeling, and machine learning approaches. First, conduct epitope mapping of the At4g39550 protein sequence to identify regions with high antigenicity, hydrophilicity, and surface accessibility, particularly focusing on unique sequences not conserved in other F-box proteins to ensure specificity . Next, employ protein structure prediction tools like AlphaFold2 to generate a three-dimensional model of At4g39550, highlighting potential conformational epitopes . Implement machine learning models such as MAGE (Monoclonal Antibody GEnerator) that can generate paired variable heavy and light chain antibody sequences against the target antigen sequence . These models should be trained on diverse antibody-antigen interaction datasets and validated using binding affinity prediction algorithms. For improved accuracy, implement active learning strategies where the model iteratively identifies the most informative antibody-antigen pairs for experimental testing, thereby enhancing prediction performance with minimal experimental validation . Finally, validate computational predictions through wet-lab experiments testing antibody binding affinity, specificity, and cross-reactivity with related F-box proteins.

What are the most effective approaches for troubleshooting non-specific binding with At4g39550 antibodies?

Troubleshooting non-specific binding with At4g39550 antibodies requires systematic optimization of multiple parameters. First, identify the source of background by testing the antibody against knockout plant tissues – persistent signal indicates non-specificity . For cross-reactivity with related F-box family members, implement epitope mapping to redesign antibodies against unique regions of At4g39550. Optimize blocking conditions by testing different blocking agents (5% BSA, 5% non-fat milk, commercial blocking buffers) and extending blocking times to 2-3 hours at room temperature. Titrate primary antibody concentrations (typically testing 1:500 to 1:5000 dilutions) and reduce incubation temperature to 4°C overnight to enhance specificity . Increase stringency of wash steps by adding 0.1-0.5% Tween-20 or 0.1% Triton X-100 to wash buffers and extending wash durations. For particularly problematic samples, pre-adsorb antibodies against knockout plant lysates immobilized on nitrocellulose to deplete cross-reactive antibodies. If Western blots show multiple bands, add denaturing agents like 6M urea to sample buffers to ensure complete protein denaturation, or use antibody affinity purification against immobilized target epitopes to isolate the most specific antibody population from polyclonal sera.

How should I design experiments to study At4g39550 protein expression patterns during plant development?

Designing experiments to study At4g39550 expression patterns during plant development requires a comprehensive temporal and spatial approach. Establish a developmental timeline sampling Arabidopsis tissues at key stages: seed germination, seedling development (3, 5, 7, 10 days), vegetative growth, floral transition, reproductive development, and senescence . For each stage, dissect and separately analyze different organs (roots, shoots, leaves, flowers, siliques) to create a tissue-specific expression map. Implement multiple detection methods: Western blotting with At4g39550 antibodies for protein quantification, immunohistochemistry for cellular localization, and RT-qPCR for transcript levels to correlate with protein abundance . Include appropriate controls: positive controls (tissues with known high expression), negative controls (At4g39550 knockout lines), and loading controls (anti-actin or anti-tubulin). For immunohistochemistry, perform both longitudinal and cross-sectional tissue preparations to capture cell-type specific expression patterns. To understand regulation, subject plants to relevant hormonal treatments (auxin, gibberellin, cytokinin, abscisic acid) and environmental stresses (drought, salt, heat, cold), then measure changes in At4g39550 expression using the same antibody-based detection methods. Quantify results using image analysis software for immunohistochemistry and densitometry for Western blots to generate statistically robust expression profiles.

What controls should be included when using At4g39550 antibodies in immunoprecipitation experiments?

Rigorous immunoprecipitation experiments with At4g39550 antibodies require multiple controls to ensure data validity. First, include a negative control using pre-immune serum or isotype-matched control antibodies to establish baseline non-specific binding . Second, perform parallel immunoprecipitations with lysates from At4g39550 knockout or knockdown plants to identify non-specific proteins that co-precipitate even in the absence of the target protein. Third, include a positive control using antibodies against known interacting partners of At4g39550, such as ASK1 (Arabidopsis SKP1-like protein) which interacts with the F-box domain . Fourth, implement technical controls by retaining samples from each experimental step (input, unbound, washes, and eluate) to track protein enrichment efficiency. Fifth, perform reciprocal co-immunoprecipitations where suspected interaction partners are immunoprecipitated and probed for At4g39550, confirming bidirectional interaction. Sixth, include competition controls where excess antigen peptide is added to antibody before immunoprecipitation, which should abolish specific target capture. Finally, prepare experimental replicates with varying detergent stringencies (0.1%, 0.5%, and 1% NP-40 or Triton X-100) to distinguish between stable and transient protein interactions, providing insight into the strength of molecular associations within At4g39550 complexes.

How can I integrate antibody-based detection methods with transcriptomic data for a comprehensive analysis of At4g39550 function?

Integrating antibody-based detection with transcriptomic data for comprehensive analysis of At4g39550 function requires a multi-modal data integration approach. Begin by establishing temporal correlation between protein levels (detected via quantitative Western blotting with At4g39550 antibodies) and mRNA expression (from RNA-seq or microarray data) across developmental stages or stress conditions . It's critical to note that concordance between mRNA and protein levels occurs in only approximately 20% of cases, making this integrated approach essential for accurate functional assessment . Next, perform chromatin immunoprecipitation sequencing (ChIP-seq) or chromatin immunoprecipitation followed by mass spectrometry (ChIP-MS) using antibodies against transcription factors predicted to regulate At4g39550 expression based on promoter analysis. To connect At4g39550 to downstream pathways, conduct RNA-seq in wild-type versus At4g39550 knockout/knockdown lines, then validate differential expression of key genes at the protein level using targeted antibodies. For post-translational regulation studies, combine phospho-specific or ubiquitin-specific antibodies with immunoprecipitation followed by mass spectrometry to identify modifications of At4g39550 and correlate these with transcriptional changes. Finally, implement network analysis tools to integrate protein interaction data (from co-immunoprecipitation with At4g39550 antibodies) with differentially expressed genes, creating a systems-level map of At4g39550's functional context and regulatory relationships.

What approaches can resolve contradictory results between antibody-based detection and gene expression data for At4g39550?

Resolving contradictory results between antibody-based detection and gene expression data for At4g39550 requires systematic investigation of multiple biological and technical factors. First, examine post-transcriptional regulation by quantifying At4g39550 mRNA stability through actinomycin D time-course experiments and polysome profiling to assess translation efficiency . Second, investigate post-translational modifications and protein stability by treating samples with proteasome inhibitors (MG132) or performing cycloheximide chase assays with At4g39550 antibody detection to determine protein half-life. Third, assess potential technical artifacts by using multiple antibodies targeting different epitopes of At4g39550 and comparing results across different detection methods (Western blot, ELISA, immunofluorescence) . Fourth, evaluate antibody specificity through knockout validation and peptide competition assays to rule out cross-reactivity with related F-box proteins . Fifth, consider tissue heterogeneity by performing single-cell RNA-seq paired with immunohistochemistry on serial sections to identify cell-type specific differences that might be masked in bulk tissue analyses. Sixth, examine temporal dynamics through fine-grained time-course sampling, as mRNA levels often change before corresponding protein changes become detectable. Finally, investigate transcriptional regulation using reporter constructs with the At4g39550 promoter to identify conditions affecting transcription, while using antibodies to monitor concurrent protein levels. This comprehensive approach can identify the specific biological mechanisms responsible for the observed discrepancies.

How can mass spectrometry complement antibody-based approaches for studying At4g39550 protein modifications?

Mass spectrometry (MS) provides powerful complementary approaches to antibody-based detection for studying At4g39550 modifications with site-specific resolution. Begin with immunoprecipitation using validated At4g39550 antibodies to enrich the target protein, followed by MS analysis to identify post-translational modifications (PTMs) including phosphorylation, ubiquitination, SUMOylation, and glycosylation . Implementation of multiple reaction monitoring (MRM) or parallel reaction monitoring (PRM) allows targeted quantification of specific modified peptides across experimental conditions. To enhance detection of low-abundance modifications, employ enrichment strategies before MS analysis: immobilized metal affinity chromatography (IMAC) for phosphopeptides, ubiquitin remnant antibodies for ubiquitinated sites, or lectin affinity chromatography for glycosylated residues. Cross-linking mass spectrometry (XL-MS) can capture transient protein-protein interactions involving At4g39550, particularly with components of the SCF complex and substrate proteins. For studying the dynamics of modifications, implement stable isotope labeling with amino acids in cell culture (SILAC) or tandem mass tag (TMT) labeling to compare modification states across developmental stages or stress conditions. Native mass spectrometry can determine the stoichiometry of modifications and characterize intact protein complexes containing At4g39550. These MS approaches overcome limitations of antibody-based methods, which may lack specificity for particular modification sites or fail to detect novel modifications that could be critical to At4g39550 function.

What bioinformatic approaches can predict epitopes for generating highly specific At4g39550 antibodies?

Advanced bioinformatic approaches for predicting optimal epitopes for At4g39550-specific antibodies integrate structural information with sequence analysis. Begin with comprehensive sequence alignment of the entire F-box protein family in Arabidopsis to identify regions unique to At4g39550, particularly outside the conserved F-box and kelch repeat domains . Apply epitope prediction algorithms that evaluate factors including antigenicity (Kolaskar-Tongaonkar), hydrophilicity (Parker), surface accessibility (Emini), and flexibility (Karplus-Schulz) to prioritize candidate regions. Incorporate protein secondary structure predictions to select peptides that are likely exposed in the native protein conformation, avoiding buried regions within β-propeller structures formed by kelch repeats . Implement B-cell epitope prediction tools like BepiPred-2.0 or SEPPA that use machine learning approaches trained on known antibody-antigen complexes. For higher specificity, generate a three-dimensional model of At4g39550 using AlphaFold2 or similar tools to identify conformational epitopes that may not be apparent from sequence analysis alone . Calculate epitope conservation scores across plant species to select regions that are either highly conserved (for cross-species reactivity) or species-specific (for Arabidopsis-specific detection). Finally, apply molecular docking simulations to assess potential interactions between candidate epitopes and modeled antibody paratopes, prioritizing those with favorable binding energies . These predictions should guide synthesis of multiple peptide antigens for experimental antibody production, followed by validation for specificity and sensitivity.

How can At4g39550 antibodies be used in high-throughput protein interaction screening approaches?

High-throughput protein interaction screening with At4g39550 antibodies can systematically map the interactome of this F-box protein using several complementary approaches. First, implement protein microarray screening where purified Arabidopsis proteome arrays are probed with fluorescently labeled At4g39550 antibodies to identify direct binding partners, particularly potential substrate proteins for the SCF complex . Second, develop an automated co-immunoprecipitation platform where At4g39550 antibodies are immobilized on magnetic beads in a 96-well format, followed by incubation with plant lysates from different tissues or conditions, and identification of co-precipitated proteins by mass spectrometry . Third, establish a proximity-dependent biotin identification (BioID) system where At4g39550 is fused to a biotin ligase, expressed in plants, and nearby biotinylated proteins are captured using streptavidin and identified by mass spectrometry, with validation using specific antibodies. Fourth, implement split-reporter systems like split-luciferase complementation where potential interaction partners are systematically screened against At4g39550, with positive interactions reconstituting luciferase activity for detection. Fifth, develop a cross-linking immunoprecipitation (CLIP) approach where UV cross-linking is followed by At4g39550 immunoprecipitation to identify transient or weak interactions that might be missed by conventional methods. These approaches can be scaled to test interactions under various stress conditions, hormone treatments, or developmental stages to construct a dynamic interaction network that reveals the context-specific functions of At4g39550.

What targeted proteomics approaches can quantify At4g39550 protein abundance in complex plant samples?

Targeted proteomics approaches offer precise quantification of At4g39550 protein abundance in complex plant samples with high sensitivity and specificity. Implement parallel reaction monitoring (PRM) or selected reaction monitoring (SRM) mass spectrometry using heavy isotope-labeled synthetic peptides as internal standards corresponding to unique regions of At4g39550 . These approaches can detect femtomole quantities of target protein and provide absolute quantification in complex backgrounds. For enhanced sensitivity, combine immunoaffinity enrichment using At4g39550 antibodies with targeted mass spectrometry in an immunoSRM workflow. This hybrid approach first enriches the target protein before MS analysis, lowering detection limits by several orders of magnitude. Alternatively, develop a custom multiplex immunoassay platform using techniques like Luminex or proximity extension assays (PEA) where antibodies against At4g39550 and other proteins of interest are conjugated to distinct beads or DNA barcodes, enabling simultaneous quantification of multiple proteins from a single sample. For spatial resolution, implement digital spatial profiling using in situ At4g39550 antibody staining combined with spatial transcriptomics to correlate protein abundance with gene expression profiles across tissue regions. To measure At4g39550 turnover rates, combine pulse-chase stable isotope labeling with immunoprecipitation and targeted MS to determine protein half-life and synthesis rates. These approaches provide complementary information on At4g39550 dynamics that cannot be obtained from standard immunoblotting techniques, offering deeper insights into its regulatory mechanisms in response to developmental cues or environmental signals.

What are the critical considerations for designing antibodies specific to different domains of the At4g39550 protein?

Designing domain-specific antibodies for At4g39550 requires careful consideration of several critical factors. First, perform comprehensive sequence analysis to distinguish between the N-terminal F-box domain, the central kelch repeat region, and any C-terminal domains, as each presents different challenges for antibody design . For the F-box domain, identify unique sequence variations that differentiate At4g39550 from other F-box proteins in Arabidopsis, as this domain is highly conserved across the family . When targeting kelch repeats, avoid designing antibodies against individual repeats, as their structural similarity may cause cross-reactivity; instead, focus on junctions between repeats or unique loop regions within the β-propeller structure . Consider the structural accessibility of epitopes by modeling the three-dimensional conformation of each domain using AlphaFold2 or similar tools, selecting surface-exposed regions for antibody recognition . For applications requiring native protein detection, avoid epitopes that may be occluded by protein-protein interactions, particularly within the F-box domain which interacts with ASK1 . Evaluate the biochemical properties of potential epitopes, selecting those with balanced hydrophilicity/hydrophobicity and minimal post-translational modification sites that could interfere with antibody binding. Design multiple antibodies targeting different domains to provide complementary information – antibodies against conserved domains for pan-recognition of related proteins, and antibodies against unique regions for At4g39550-specific detection . Finally, consider the intended application when selecting epitopes: linear epitopes for Western blotting versus conformational epitopes for immunoprecipitation or immunohistochemistry.

How can machine learning models be applied to improve At4g39550 antibody design?

Machine learning models can revolutionize At4g39550 antibody design by optimizing epitope selection and predicting antibody-antigen binding with higher accuracy. Implement protein language models like MAGE (Monoclonal Antibody GEnerator) that can generate paired variable heavy and light chain sequences specifically targeting At4g39550 without requiring pre-existing antibody templates . These models, trained on extensive antibody-antigen interaction datasets, can predict binding affinities and cross-reactivity profiles before experimental validation . Apply deep learning frameworks to analyze the three-dimensional structure of At4g39550, identifying conformational epitopes that may not be apparent from sequence analysis alone. Utilize active learning strategies that iteratively refine predictions by identifying the most informative antibody-antigen pairs for experimental testing, thereby enhancing model performance with minimal laboratory validation . Develop ensemble models that integrate multiple predictors (epitope accessibility, antigenicity, hydrophilicity) to rank potential epitopes based on their likelihood of generating specific antibodies. Implement generative adversarial networks (GANs) to explore novel antibody sequence spaces that optimize both binding affinity and specificity for At4g39550 while minimizing cross-reactivity with related F-box proteins. For validation strategies, use convolutional neural networks to analyze immunohistochemistry or Western blot images, providing quantitative assessments of antibody performance across different experimental conditions. These machine learning approaches can significantly reduce the time and resources required for antibody development while improving the quality and specificity of the resulting antibodies for At4g39550 research.

What approaches can generate monoclonal antibodies with the highest specificity for At4g39550?

Generating monoclonal antibodies with exceptional specificity for At4g39550 requires advanced immunization and screening strategies. Begin with careful antigen design, using computational approaches to identify unique peptide regions of At4g39550 that have minimal sequence similarity to other F-box proteins in Arabidopsis . Implement a subtractive immunization strategy where animals are first tolerized to common F-box protein epitopes before immunization with At4g39550-specific antigens, thereby focusing the immune response on unique regions. During hybridoma screening, implement a multi-stage selection process: initial ELISA screening against the immunizing antigen, followed by competitive ELISAs with related F-box proteins to eliminate cross-reactive clones, and finally validation by Western blotting against plant lysates from both wild-type and At4g39550 knockout lines . For enhanced specificity, apply phage display technology to select antibody fragments with optimal binding characteristics from diverse antibody libraries, followed by affinity maturation through iterative rounds of mutation and selection. Alternatively, implement next-generation sequencing of B-cell receptors from immunized animals to identify expanded clones responding to At4g39550, then express and screen these naturally selected antibodies. Consider using transgenic animals carrying human immunoglobulin genes for generating antibodies with reduced immunogenicity in certain applications. For validation, perform epitope binning using surface plasmon resonance or bio-layer interferometry to characterize the precise binding sites and ensure antibodies recognize distinct epitopes. Finally, test selected antibodies across multiple applications (Western blotting, immunoprecipitation, immunohistochemistry) to identify those with consistent performance and specificity across different experimental contexts.

How can recombinant antibody technologies improve research tools for At4g39550 studies?

Recombinant antibody technologies offer significant advantages for developing advanced research tools for At4g39550 studies. Engineer single-chain variable fragments (scFvs) or antigen-binding fragments (Fabs) derived from validated anti-At4g39550 monoclonal antibodies for applications requiring smaller binding molecules with better tissue penetration . Develop bispecific antibody formats with one binding site targeting At4g39550 and another recognizing a common tag or reporter protein, enabling versatile detection without direct labeling of primary antibodies. For tracking At4g39550 in live cells, generate fluorescent protein fusions with antibody fragments (e.g., scFv-GFP) that can function as intrabodies, allowing real-time visualization of protein dynamics without fixation artifacts . Implement antibody engineering to create variants with site-specific conjugation sites for attaching functional moieties like biotin, enzymes, or photocrosslinkers without compromising antigen binding. Develop nanobodies (single-domain antibodies derived from camelid heavy-chain antibodies) against At4g39550, which offer advantages including small size (~15 kDa), high stability, and the ability to recognize epitopes inaccessible to conventional antibodies . For degradation studies, engineer proteolysis-targeting chimeras (PROTACs) by fusing At4g39550-specific antibody fragments with degrons, creating tools to induce targeted protein degradation and observe resulting phenotypes. Implement yeast or mammalian surface display for directed evolution of antibody affinity and specificity, generating variants with optimized properties for specific applications. These recombinant antibody approaches provide researchers with precisely tailored tools for studying At4g39550 function, localization, and interactions with unprecedented flexibility and specificity compared to conventional antibody formats.

How can antibodies be used to study evolutionary conservation of At4g39550 across plant species?

Antibodies provide powerful tools for studying evolutionary conservation of At4g39550 across diverse plant species through comparative immunological approaches. Design antibodies targeting both conserved domains (F-box motif, kelch repeats) and species-specific regions of At4g39550 to distinguish between general conservation and lineage-specific adaptations . Perform Western blot analysis of protein extracts from phylogenetically diverse plant species (spanning monocots, dicots, gymnosperms, and early land plants) using these antibodies to create an immunological phylogenetic profile. Complement protein detection with immunoprecipitation followed by mass spectrometry to identify and compare interacting partners across species, revealing conservation of functional networks. Conduct immunohistochemistry across diverse plant species to compare tissue-specific expression patterns and subcellular localization, providing insights into potential functional divergence. For species where direct antibody cross-reactivity is limited, employ epitope-tagging approaches by transforming heterologous species with tagged At4g39550 orthologs, enabling detection with tag-specific antibodies while maintaining the protein's native regulation. Implement quantitative immunoblotting to compare expression levels across species under standardized growth conditions and various stresses, revealing differential regulation of conserved proteins. For deeper evolutionary analysis, combine antibody-based detection with computational genomics approaches that analyze selective pressure on different protein domains, correlating molecular evolution rates with immunologically detected structural conservation. This integrated approach provides a comprehensive view of At4g39550 evolution across plant lineages, connecting sequence divergence with functional conservation or innovation at the protein level.

What are the methodological differences when using At4g39550 antibodies in different plant species models?

Using At4g39550 antibodies across different plant species requires methodological adaptations to account for biological and technical variables. First, optimize protein extraction protocols for each species based on tissue composition – woody tissues may require stronger lysis buffers (higher detergent concentrations, mechanical disruption), while mucilage-rich tissues benefit from additional washing steps to remove interfering compounds . Adjust antibody concentrations based on cross-reactivity strength with orthologs; typically higher concentrations (2-5x) are needed for distantly related species compared to Arabidopsis. Modify blocking solutions to account for species-specific backgrounds; milk-based blockers may introduce artifacts in some species, necessitating BSA or commercial alternatives. For immunohistochemistry, optimize fixation protocols according to tissue characteristics – thicker tissues require longer fixation times or vacuum infiltration of fixatives, while highly vacuolated cells may need modified permeabilization procedures. Implement comparative Western blotting with known quantities of recombinant At4g39550 protein to calibrate signal intensities across species, accounting for epitope variation in orthologs. For immunoprecipitation, adjust salt and detergent concentrations to maintain similar stringency despite species differences in protein complex stability. When examining species with genome duplications (e.g., many crops), use additional controls to distinguish between paralogs, including competing peptides specific to each paralog. Finally, validate antibody specificity in each species using RNAi lines or mutants when available, or heterologous expression of the corresponding ortholog in a system where the endogenous protein is absent. These adaptations ensure meaningful cross-species comparisons while minimizing artifacts arising from methodological inconsistencies.

How can epitope conservation analysis guide antibody selection for comparative studies of F-box proteins?

Epitope conservation analysis provides a systematic framework for selecting antibodies in comparative studies of F-box proteins across species. Begin with multiple sequence alignment of At4g39550 orthologs and paralogs from diverse plant lineages, calculating conservation scores for overlapping peptide windows to identify regions of high, moderate, and low conservation . Generate conservation heat maps highlighting species-specific variations within potential epitope regions to guide antibody selection for different experimental goals: highly conserved epitopes for broad cross-species reactivity versus variable regions for species-specific detection. For the F-box domain, which shows higher conservation, focus epitope design on subtle sequence variations while avoiding regions that interact with SKP1 proteins, as these interfaces are typically highly conserved and may be inaccessible in native protein complexes . For kelch repeat regions, which often show greater sequence divergence between species, identify repeat-specific variations that can serve as species-discriminating epitopes . Implement structural alignment of predicted three-dimensional models to identify conformationally conserved regions that may maintain similar epitope structures despite sequence divergence. Calculate epitope conservation indices that weight amino acid substitutions based on physicochemical properties rather than simple identity, providing more nuanced predictions of antibody cross-reactivity. Design multi-epitope antibody strategies where antibodies targeting differentially conserved regions are used in combination to create species-specific detection profiles, enabling fine discrimination between closely related F-box proteins. Finally, validate predicted cross-reactivity experimentally using recombinant protein arrays expressing orthologous F-box proteins from multiple species, measuring binding affinities to confirm conservation predictions and establish detection thresholds for comparative studies.

What insights can be gained from studying post-translational modifications of At4g39550 across different plant species?

Studying post-translational modifications (PTMs) of At4g39550 orthologs across plant species yields profound insights into evolutionary conservation of regulatory mechanisms. Employ phospho-specific antibodies targeting conserved modification sites to track phosphorylation patterns across species, revealing preserved regulatory nodes despite sequence divergence . Implement immunoprecipitation with general At4g39550 antibodies followed by mass spectrometry analysis to create comprehensive PTM maps for each species, identifying conserved and species-specific modification landscapes. Compare ubiquitination patterns using antibodies against ubiquitin after At4g39550 immunoprecipitation, providing insights into conserved degradation mechanisms and potentially divergent turnover rates across species. Correlate PTM differences with species-specific phenotypes, environmental adaptations, or life history traits to identify modifications potentially involved in evolutionary specialization. Analyze conservation of kinase recognition motifs surrounding phosphorylation sites to determine whether the same kinase families regulate At4g39550 orthologs across diverse plant lineages. Investigate co-evolution between At4g39550 modification sites and their enzymatic writers (kinases, ubiquitin ligases) or readers (phospho-binding domains, ubiquitin-binding proteins) across phylogenetic distances. Implement comparative phosphoproteomic approaches under standardized stress conditions to determine whether stress-responsive PTMs are conserved, suggesting fundamental importance to plant survival. For functional validation, develop transgenic complementation systems where modified forms of At4g39550 (phospho-mimetic or phospho-deficient mutants) from different species are expressed in Arabidopsis knockout backgrounds, assessing the functional consequences of species-specific modification patterns. This multi-faceted approach connects molecular modifications with protein function and species adaptation, providing evolutionary context for At4g39550 regulation across plant diversity.

How can CRISPR-based technologies enhance antibody validation for At4g39550 research?

CRISPR-based technologies provide powerful approaches for rigorous antibody validation in At4g39550 research. Generate precise knockout lines by targeting the At4g39550 gene using CRISPR-Cas9, creating definitive negative controls for antibody specificity testing across applications – a complete loss of signal in these lines confirms antibody specificity . Implement CRISPR interference (CRISPRi) to achieve tunable repression of At4g39550 expression, creating a gradient of protein abundance for calibrating antibody sensitivity and dynamic range. Develop epitope-tagged knock-in lines using CRISPR-mediated homology-directed repair to insert small epitope tags at the endogenous At4g39550 locus, enabling antibody validation through dual detection with both At4g39550-specific and epitope-tag antibodies. For paralogs or highly similar F-box family members, employ multiplexed CRISPR editing to create combinatorial knockout lines, allowing systematic assessment of antibody cross-reactivity across related proteins . Generate domain-specific deletions or modifications using precise CRISPR editing to map the exact binding regions of different antibodies within the At4g39550 protein structure. Implement CRISPR activation (CRISPRa) to overexpress At4g39550 in tissues where it is normally absent or at low levels, providing positive controls for antibody validation in diverse cellular contexts. For temporal validation, combine CRISPR with inducible promoter systems to create conditional At4g39550 knockouts, enabling time-course validation of antibody specificity during protein depletion. Additionally, use CRISPR base editing to introduce specific amino acid changes at predicted epitope sites, allowing fine-mapping of the precise residues required for antibody recognition. These CRISPR-based validation strategies establish a gold standard for antibody specificity, significantly enhancing confidence in immunological studies of At4g39550 and related proteins.

What single-cell approaches can be combined with At4g39550 antibodies for higher-resolution studies?

Single-cell approaches combined with At4g39550 antibodies enable unprecedented resolution of protein expression and function at the cellular level. Implement imaging mass cytometry (IMC) using metal-conjugated At4g39550 antibodies to simultaneously visualize protein expression alongside dozens of other cellular markers in tissue sections, revealing cell type-specific expression patterns with spatial context . Apply cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) by labeling dissociated plant protoplasts with oligo-tagged At4g39550 antibodies, allowing simultaneous protein detection and transcriptome analysis from the same cells to correlate protein abundance with gene expression profiles. Develop single-cell Western blotting techniques for plant protoplasts using microfluidic devices where individual cells are lysed in nanoliter chambers, followed by electrophoretic separation and on-chip immunoprobing with At4g39550 antibodies to quantify protein levels in individual cells. Implement proximity ligation assays (PLA) at single-cell resolution to detect protein-protein interactions involving At4g39550 in intact tissues, generating fluorescent signals only when two proteins are within 40nm proximity. Combine laser capture microdissection with highly sensitive immunoassays to analyze At4g39550 expression in specific cell types isolated from complex tissues. For dynamic studies, implement microfluidic live cell arrays where individual protoplasts expressing fluorescently tagged proteins are captured in microwells and monitored over time, with periodic immunostaining using membrane-permeable antibody fragments to track At4g39550 dynamics. These single-cell approaches provide critical insights into cell-to-cell variability in At4g39550 expression, localization, and interaction patterns that would be masked in conventional bulk analyses, revealing how heterogeneity at the cellular level contributes to tissue-level and organism-level functions.

How will antibody engineering technologies impact future research on At4g39550 and related proteins?

Emerging antibody engineering technologies will revolutionize research on At4g39550 and related F-box proteins through innovations in specificity, functionality, and application versatility. Rational paratope design using computational protein engineering will create antibodies with unprecedented specificity for distinguishing between highly similar F-box family members, overcoming traditional cross-reactivity limitations . Light-controllable antibodies incorporating photoswitchable domains will enable spatiotemporal control of At4g39550 detection or manipulation, allowing researchers to selectively visualize or perturb protein function in specific cells at defined time points. Cell-permeable mini-antibodies engineered through cyclic peptide scaffolds will facilitate live-cell imaging and perturbation of At4g39550 without the need for genetic modification, expanding the range of applicable experimental systems. Multiplexed detection systems using DNA-barcoded antibodies will enable simultaneous profiling of dozens of F-box proteins in single samples, providing comprehensive views of family-wide expression patterns. Antibody-based proximity labeling tools where At4g39550-specific antibodies are conjugated to engineered enzymes like TurboID will map protein neighborhoods in native contexts without overexpression artifacts. For functional studies, antibody-drug conjugates designed to induce selective degradation of At4g39550 will provide precise temporal control over protein depletion, complementing genetic approaches. Advanced humanized plant-specific antibodies will reduce background and increase specificity in plant tissues while maintaining compatibility with mammalian secondary detection systems. Computationally designed antibody cocktails targeting multiple epitopes on At4g39550 will enhance detection sensitivity and reliability across applications. These innovations will transform At4g39550 research by expanding the experimental toolkit beyond traditional limitations, enabling more precise interrogation of protein function in complex biological contexts.

What role will artificial intelligence play in advancing antibody development for plant proteins like At4g39550?

Artificial intelligence will fundamentally transform antibody development for plant proteins like At4g39550 through innovations spanning design, production, and application. Deep learning models like MAGE (Monoclonal Antibody GEnerator) will design paired heavy-light chain antibody sequences specifically targeting At4g39550 without requiring pre-existing templates, accelerating the development of novel antibodies with optimized properties . Reinforcement learning algorithms will guide antibody optimization by iteratively refining designs based on experimental validation data, creating a feedback loop that continuously improves specificity and affinity for At4g39550. Natural language processing models trained on scientific literature will extract contextual knowledge about At4g39550 function and structure, informing epitope selection strategies that target biologically relevant regions. Generative adversarial networks will explore novel antibody sequence spaces beyond traditional frameworks, potentially discovering unconventional binding modalities with enhanced performance for challenging targets. Graph neural networks will analyze protein interaction networks to identify optimal antibody binding sites that minimize interference with At4g39550's biological functions while maximizing detection sensitivity. Computer vision algorithms will standardize and automate antibody validation processes, objectively evaluating staining patterns in immunohistochemistry or band specificity in Western blots across multiple experimental conditions . Advanced active learning strategies will dramatically reduce experimental validation requirements by identifying the most informative subset of antibody candidates for testing, maximizing information gain while minimizing resource expenditure . Federated learning approaches will enable collaborative training of antibody design models across research institutions without sharing sensitive data, accelerating progress through distributed expertise. Multimodal AI systems integrating structural, sequence, and functional data will create comprehensive antibody development platforms that consider multiple design constraints simultaneously, balancing specificity, stability, production efficiency, and application performance for At4g39550 and other challenging plant protein targets.

What are the most significant limitations of current antibody-based approaches for At4g39550 research?

Current antibody-based approaches for At4g39550 research face several significant limitations that constrain experimental reliability and interpretation. First, distinguishing At4g39550 from other F-box family members remains challenging due to the high sequence conservation within the F-box domain and structural similarities in kelch repeat regions, often leading to cross-reactivity that confounds specific detection . Second, validating antibody specificity is hampered by limited availability of proper genetic controls like knockout lines or the presence of functionally redundant paralogs that may compensate for At4g39550 loss. Third, detecting native levels of At4g39550 protein is difficult due to potentially low abundance, tissue-specific expression, or conditional regulation, requiring highly sensitive detection methods that may introduce artifacts. Fourth, antibodies often fail to distinguish between post-translationally modified forms of At4g39550, missing critical regulatory mechanisms involving phosphorylation, ubiquitination, or other modifications that modulate protein function . Fifth, available antibodies may recognize only a limited subset of conformational states of At4g39550, potentially missing functionally relevant structural changes associated with protein-protein interactions or substrate binding. Sixth, batch-to-batch variability in polyclonal antibodies creates reproducibility challenges across studies and laboratories, complicating data comparison and meta-analysis. Seventh, current antibodies may not capture the full range of species-specific variations in At4g39550 orthologs, limiting comparative studies across evolutionary distances. Finally, current approaches lack temporal resolution for studying dynamic processes, providing static snapshots rather than capturing the rapid changes in protein localization, modification, or degradation that may be central to At4g39550 function. Addressing these limitations requires integrating multiple complementary approaches alongside antibody-based detection to build a comprehensive understanding of At4g39550 biology.

What combination of techniques provides the most comprehensive analysis of At4g39550 function?

A comprehensive analysis of At4g39550 function requires integrating multiple complementary techniques that overcome the limitations of individual approaches. Begin with genetic characterization using CRISPR-generated knockout lines, RNAi knockdowns, and complementation studies with tagged or mutated variants to establish phenotypes associated with At4g39550 dysfunction . Combine this with protein-level analysis using validated domain-specific antibodies for immunoblotting, immunoprecipitation, and immunolocalization to detect native protein levels, interactions, and subcellular distribution across tissues and conditions . Implement proteomics approaches including immunoprecipitation followed by mass spectrometry to identify interaction partners and post-translational modifications, complemented by targeted approaches like phospho-specific antibodies to track specific modifications . For transcriptional context, employ RNA-seq in wild-type versus mutant backgrounds under various conditions to identify genes and pathways affected by At4g39550 function. Incorporate structural biology through AlphaFold2 prediction or experimental structure determination of At4g39550 alone and in complex with partners to understand molecular mechanisms . For in vivo dynamics, use live-cell imaging with fluorescently tagged proteins under native promoters, validated by antibody detection of endogenous protein. Apply biochemical approaches to characterize enzymatic activities of SCF complexes containing At4g39550, identifying substrates and quantifying ubiquitination activities. Finally, implement systems biology modeling to integrate these diverse datasets into coherent functional networks, generating testable hypotheses about At4g39550's role in plant development and stress responses. This multi-faceted approach compensates for the limitations of individual techniques, providing multiple lines of evidence that converge on a comprehensive understanding of At4g39550 function and regulation.

What are the emerging trends in antibody-based plant protein research that will impact At4g39550 studies?

Emerging trends in antibody-based plant protein research will substantially impact future At4g39550 studies through methodological innovations and conceptual advances. Nanobody and single-domain antibody technologies derived from camelid immune systems are gaining prominence, offering smaller binding molecules with enhanced tissue penetration and epitope accessibility for studying At4g39550 in intact plant tissues . Recombinant antibody engineering is moving toward modular formats where binding domains can be rapidly swapped or combined, enabling quick adaptation of validated antibodies for new applications like proximity labeling or controlled degradation of At4g39550 . Computational antibody design using machine learning is increasingly replacing traditional immunization and selection approaches, accelerating development of highly specific antibodies against challenging epitopes of plant F-box proteins . Multiplexed detection systems based on DNA-barcoded antibodies or mass cytometry are enabling simultaneous analysis of dozens of proteins including At4g39550 and its interaction partners in single samples, providing systems-level insights. Antibody-enabled proximity proteomics methods like BioID and APEX are becoming standard for mapping protein neighborhoods in their native environments, revealing contextual interactions of At4g39550 in different cellular compartments. Single-cell antibody-based technologies are increasingly being adapted for plant systems, offering unprecedented resolution of cell-type specific variation in At4g39550 expression and function. Environmentally responsive antibody-based biosensors are emerging for tracking protein modifications or conformational changes in response to stimuli, potentially revealing dynamic aspects of At4g39550 regulation. Finally, synthetic biology approaches are combining antibody specificity with programmable outputs like fluorescence, enzymatic activity, or transcriptional regulation, transforming antibodies from passive detection tools into active research instruments for manipulating and monitoring At4g39550 function in complex plant systems.

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 2024 Thebiotek. All Rights Reserved.