At1g58055 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 week lead time (made-to-order)
Synonyms
At1g58055 antibody; T15M6 antibody; Defensin-like protein 117 antibody
Target Names
At1g58055
Uniprot No.

Target Background

Database Links

KEGG: ath:AT1G58055

STRING: 3702.AT1G58055.1

UniGene: At.64952

Protein Families
DEFL family
Subcellular Location
Secreted.

Q&A

What validation methods should be employed to confirm At1g58055 antibody specificity?

Robust antibody validation requires multiple complementary approaches to establish specificity. Begin with ELISA and western blot analysis against purified target proteins and cell lysates. For definitive validation, employ knockout models as negative controls, similar to the AT1Ra/b knockout mice used to validate AT1R antibody binding in vivo . Flow cytometry validation should include appropriate isotype controls and competitive binding assays. For instance, researchers studying receptor-targeting antibodies have demonstrated specificity by showing reduced binding in receptor-deficient systems and through competition assays with natural ligands . Additional validation can include immunohistochemistry of tissues with known expression patterns and mass spectrometry confirmation of immunoprecipitated targets.

How do monoclonal and polyclonal antibodies differ in their research applications for studying plant proteins like At1g58055?

Monoclonal antibodies offer superior specificity for discrete epitopes, making them ideal for distinguishing between highly homologous plant proteins that may share significant sequence similarity. This specificity is crucial when investigating protein isoforms or closely related family members. For example, monoclonal antibodies can be generated through hybridoma techniques to target specific domains of membrane proteins . Polyclonal antibodies recognize multiple epitopes, providing stronger signals through epitope redundancy and greater tolerance to protein denaturation or conformational changes, but with potential cross-reactivity challenges. The choice depends on experimental goals: use monoclonals for highly specific detection of single epitopes and polyclonals when signal amplification and detection across various experimental conditions is prioritized.

What expression systems are optimal for producing recombinant proteins for At1g58055 antibody generation?

The selection of expression systems must align with the structural characteristics of the target protein. For plant membrane proteins like potential At1g58055-encoded products, mammalian expression systems (such as HEK293 cells) preserve complex post-translational modifications and proper protein folding . These systems have been successfully used to express membrane receptors like AT1R for subsequent antibody generation and validation. For soluble plant proteins, bacterial systems like E. coli offer cost-effective production but lack post-translational modifications. Insect cell systems represent an intermediate option, balancing proper folding with reasonable yields. Always validate the recombinant protein's structural integrity through circular dichroism or limited proteolysis before immunization to ensure antibodies recognize native epitopes.

What immunization strategies yield the most specific antibodies against plant transmembrane proteins?

Generating antibodies against transmembrane proteins requires specialized approaches that preserve native conformations. Research demonstrates that immunization with membrane-embedded proteins yields antibodies recognizing conformational epitopes critical for functional studies . For plant transmembrane proteins, employ membrane extracts from cells overexpressing the target protein rather than synthetic peptides alone. When designing the immunization protocol, include both membrane-embedded full-length protein and peptides corresponding to extracellular domains to generate complementary antibody populations. A strategic approach involves immunizing with membrane extracts containing human AT1R, which successfully produced functional antibodies recognizing conformational epitopes . Additionally, incorporate T-cell epitope mapping to identify immunodominant regions that enhance antibody production, as demonstrated with the AT1R 149-172 peptide immunization strategy .

How can phage display technology be optimized for selecting high-affinity antibodies against plant proteins?

Phage display optimization for plant protein targets requires careful consideration of selection conditions and screening strategies. Implement a staged selection approach with increasing stringency through multiple rounds, as demonstrated in antibody development research . Begin with pre-selection steps that deplete cross-reactive antibodies by incubating the phage library with related plant proteins or cellular components that might generate non-specific binders. For transmembrane proteins, perform selections against membrane extracts containing the overexpressed target. Modern phage display protocols incorporate high-throughput sequencing at each selection stage to monitor antibody population dynamics and identify enriched sequences . This approach allows computational analysis to distinguish specific binding modes and predict antibody variants with customized specificity profiles, even for closely related epitopes .

What experimental controls are essential when evaluating At1g58055 antibody function in cellular assays?

Rigorous experimental controls are critical for interpreting antibody-based cellular assays. Always include isotype-matched control antibodies at equivalent concentrations to distinguish specific effects from non-specific binding or Fc-mediated functions . For target validation, incorporate genetic approaches such as CRISPR-knockout cell lines or siRNA-mediated knockdown to confirm antibody specificity. When investigating receptor-targeting antibodies, include receptor-deficient systems as negative controls, similar to the AT1Ra/b knockout mice used to validate AT1R antibody functions . Dose-response experiments are essential to distinguish specific effects from off-target activity at high concentrations. For functional studies, include both positive controls (known modulators of the pathway) and negative controls (structurally similar compounds without expected activity) to validate assay performance and antibody specificity.

How can computational modeling enhance the design of antibodies with tailored specificity profiles for plant protein research?

Computational modeling has revolutionized antibody engineering by enabling rational design of specificity profiles beyond what can be achieved through experimental selection alone . For plant protein applications, biophysics-informed models can be trained on phage display selection data to identify distinct binding modes associated with specific epitopes . This approach can disentangle binding preferences even between chemically similar ligands, allowing researchers to predict and design antibodies with customized specificity profiles not present in the original library . The model identifies sequence-function relationships by associating amino acid patterns in CDRs with specific binding modes. Researchers can leverage these models to design antibodies that either specifically target a single epitope or display cross-reactivity with multiple related targets, depending on research needs . This computational approach is particularly valuable for plant proteins that share high sequence homology with family members but require specific targeting.

What high-throughput approaches can characterize the epitope specificity of At1g58055 antibodies?

Modern epitope mapping combines multiple high-throughput technologies to comprehensively characterize antibody binding sites. Peptide microarrays displaying overlapping sequences can rapidly identify linear epitopes, while hydrogen-deuterium exchange mass spectrometry (HDX-MS) identifies conformational epitopes by measuring changes in protein dynamics upon antibody binding. X-ray crystallography and cryo-electron microscopy provide atomic-resolution structures of antibody-antigen complexes, definitively identifying binding interfaces. For antibodies targeting transmembrane proteins like potential At1g58055 products, label-free biosensing technologies such as dynamic mass redistribution (DMR) can assess binding to membrane-embedded targets in their native conformation . Computational analysis of selection data from phage display experiments can identify sequence patterns associated with specific binding modes, enabling prediction of epitope specificity for novel antibody variants .

How should researchers interpret antibody cross-reactivity data when studying plant protein families?

Cross-reactivity analysis requires systematic characterization and contextual interpretation. First, conduct comprehensive cross-reactivity testing against related family members using multiple methods (ELISA, western blot, immunoprecipitation) with recombinant proteins expressed in identical systems. Quantify relative binding affinities to distinguish primary targets from cross-reactive species. Epitope mapping can identify whether cross-reactivity stems from conserved regions, informing antibody applications. Importantly, cross-reactivity should be interpreted in the biological context—antibodies recognizing conserved functional domains across protein families may provide valuable insights into shared mechanisms . When absolute specificity is required, employ biophysics-informed computational models to identify amino acid substitutions that enhance selectivity for the primary target . Present cross-reactivity data as quantitative binding profiles rather than binary designations, allowing researchers to assess antibody suitability for specific applications.

What strategies can resolve inconsistent immunodetection results when studying low-abundance plant proteins?

Inconsistent detection of low-abundance proteins requires systematic optimization of multiple parameters. Begin by evaluating antibody concentration through titration experiments to identify the optimal signal-to-noise ratio. Enhance epitope accessibility through different sample preparation methods—test multiple fixation protocols for immunohistochemistry or membrane transfer conditions for western blotting. For plant proteins with complex post-translational modifications, evaluate different extraction buffers that preserve protein conformation while maximizing yield. Signal amplification methods such as tyramide signal amplification or polymer-based detection systems can significantly improve sensitivity without increasing background . Compare multiple antibodies targeting different epitopes on the same protein to differentiate between technical issues and biological variation. Importantly, validate positive and negative controls under identical conditions to establish assay performance benchmarks and implement standardized protocols to minimize experimental variation.

How can researchers distinguish between true positive signals and artifacts when using At1g58055 antibodies in immunolocalization studies?

Distinguishing true signals from artifacts in immunolocalization requires multiple validation approaches. Always include appropriate negative controls: (1) omission of primary antibody, (2) isotype control antibodies, and (3) pre-adsorption of antibody with purified antigen. Validate specificity through comparative staining in tissues with known expression patterns versus those without target expression. For definitive validation, use genetic approaches such as comparing wild-type samples with knockout/knockdown specimens . When studying plant subcellular localization, co-stain with established compartment markers to confirm expected distribution patterns. Additionally, validate immunolocalization findings with complementary approaches such as fluorescent protein fusions or in situ hybridization for mRNA localization. Carefully control image acquisition settings across all samples and controls, and implement blinded analysis to prevent confirmation bias in signal interpretation.

What approaches can optimize At1g58055 antibody performance in chromatin immunoprecipitation (ChIP) experiments?

Optimizing antibodies for ChIP applications requires addressing several technical challenges. First, evaluate different fixation conditions (formaldehyde concentration and crosslinking duration) to balance epitope preservation with efficient DNA recovery. Test multiple chromatin fragmentation methods (sonication parameters or enzymatic digestion) to identify conditions that preserve epitope integrity while generating appropriately sized DNA fragments. For plant chromatin, which often contains interfering compounds, implement additional purification steps such as PVPP treatment or gradient centrifugation. Optimize antibody concentration and incubation conditions through pilot experiments, comparing enrichment of known target regions versus negative control regions. To enhance specificity, include pre-clearing steps with protein A/G beads and non-specific IgG. Sequential ChIP (re-ChIP) approaches can confirm co-localization with other factors at specific genomic loci. Finally, validate ChIP-seq peaks through independent methods such as reporter gene assays or targeted mutagenesis of binding sites.

How should quantitative data from At1g58055 antibody-based assays be normalized for comparative analysis?

Proper normalization is essential for meaningful comparisons across experimental conditions. For western blot analysis, normalize target protein signals to stable reference proteins (validated housekeeping genes appropriate for the specific tissue and treatment conditions) rather than total protein content, which may vary significantly between samples. In immunohistochemistry quantification, normalize signal intensity to tissue area or cell number, and establish consistent thresholding parameters based on negative controls. For flow cytometry, report median fluorescence intensity ratios between specific antibody and isotype control signals rather than raw values. In high-throughput binding assays, implement internal calibration standards across plates to account for inter-assay variation. Statistical analysis should include appropriate tests for normality and employ non-parametric methods when distributions deviate from normality. Report both biological and technical replication, and clearly distinguish between them in statistical analysis .

What approaches can integrate antibody-based measurements with transcriptomic and proteomic data for systems-level analysis?

Integrative multi-omics analysis requires careful consideration of each data type's characteristics. When combining antibody-based protein measurements with transcriptomic data, account for temporal differences between mRNA and protein expression changes. Normalize each data type independently before integration, and consider transformation methods that make data distributions comparable across platforms. Network analysis approaches can identify functional modules where protein-protein interactions (detected by co-immunoprecipitation with At1g58055 antibodies) correlate with co-expression patterns in transcriptomic data. Pathway enrichment analysis should incorporate both transcriptomic and proteomic evidence, weighted by measurement confidence. For discordant findings between antibody-based protein detection and mRNA levels, investigate potential post-transcriptional regulation through targeted studies of protein stability or translation efficiency. Visualization tools should highlight areas of agreement and divergence between different data types, facilitating hypothesis generation about regulatory mechanisms .

How can structural biology approaches complement antibody studies to elucidate At1g58055 protein function?

Integrating structural biology with antibody studies provides comprehensive insights into protein function. X-ray crystallography or cryo-electron microscopy of antibody-antigen complexes reveals binding interfaces at atomic resolution, informing structure-function relationships. Epitope mapping through hydrogen-deuterium exchange mass spectrometry identifies conformational changes upon antibody binding, potentially revealing allosteric mechanisms. For membrane proteins, antibodies can be used as crystallization chaperones, stabilizing specific conformations to facilitate structure determination . Label-free biosensing technologies, such as dynamic mass redistribution, can detect antibody-induced conformational changes in membrane-embedded targets, providing functional insights complementary to structural data . Molecular dynamics simulations incorporating antibody binding data can predict conformational changes and allosteric effects relevant to protein function. By mapping functional epitopes through antibody competition studies, researchers can identify critical interaction surfaces for protein-protein or protein-ligand interactions, informing targeted mutagenesis studies to validate functional predictions.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.