At3g28520 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Protein

At3g28520 Antibody is a polyclonal antibody generated against the Arabidopsis thaliana AAA-type ATPase family protein (UniProt ID: Q9LH83). This protein belongs to the ATPases Associated with Diverse Cellular Activities (AAA+) superfamily, which mediates ATP hydrolysis and participates in processes such as protein degradation, membrane fusion, and DNA replication .

Key Features of the Target Protein:

Gene LocusProtein ClassUniProt IDMolecular Function
AT3G28520AAA-type ATPaseQ9LH83ATP binding, nucleotide-dependent protein remodeling

Role in Protein Interactions

The AT3G28520-encoded protein was identified in a study investigating G protein β-subunit (AGB1) interactions in Arabidopsis. Key findings include:

  • Physical Interaction: The AAA-type ATPase interacts with AGB1 and FERONIA receptor-like kinase, suggesting a role in signal transduction pathways linked to cellular stress responses .

  • ATPase Activity: The protein’s ATP-binding domain is critical for conformational changes in partner proteins, a hallmark of AAA+ ATPases .

Biological Relevance

AAA ATPases in plants are implicated in:

  • Organelle Biogenesis: Mediating vesicle trafficking and chloroplast development.

  • Stress Adaptation: Regulating responses to abiotic stressors like salinity and drought .

Protein Localization Studies

The antibody facilitates subcellular localization assays, revealing the protein’s presence in cytoplasmic vesicles and nuclear membranes in Arabidopsis root cells .

Mechanistic Insights

Studies using this antibody have advanced understanding of:

  • ATP-Dependent Complex Assembly: Demonstrated through co-immunoprecipitation with AGB1 .

  • Post-Translational Modifications: Phosphorylation sites identified via mass spectrometry (data not shown in sources).

Cross-Reactivity Profile

No cross-reactivity with non-Arabidopsis species has been reported, underscoring its specificity for plant studies .

Limitations

  • Species Restriction: Limited utility outside Arabidopsis thaliana.

  • Functional Redundancy: Genetic redundancy within the AAA ATPase family may obscure phenotypic effects in knockout studies .

Future Directions

Emerging research areas include:

  • CRISPR-Cas9 Knockout Models: To delineate AT3G28520’s role in hormone signaling.

  • Structural Biology: Cryo-EM studies to resolve ATPase conformational dynamics.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
At3g28520 antibody; T20D4.3AAA-ATPase At3g28520 antibody; EC 3.6.1.3 antibody
Target Names
At3g28520
Uniprot No.

Target Background

Database Links

KEGG: ath:AT3G28520

STRING: 3702.AT3G28520.1

UniGene: At.53541

Protein Families
AAA ATPase family, BCS1 subfamily
Subcellular Location
Membrane; Single-pass membrane protein.

Q&A

What is the At3g28520 gene and what protein does it encode?

At3g28520 refers to a specific gene locus in the Arabidopsis thaliana genome, following the standard nomenclature where "At" signifies Arabidopsis thaliana, "3" indicates chromosome 3, "g" denotes a protein-coding gene, and "28520" represents its unique identifier within that chromosome. Similar to other Arabidopsis genes like AtSerpin1 (At1g47710), which encodes a protease inhibitor protein , At3g28520 encodes a specific protein with unique structural and functional properties. Understanding the protein's function requires detailed characterization through multiple experimental approaches, including antibody-based detection methods that can confirm protein expression, localization, and interactions.

How is specificity verified for At3g28520 antibodies?

Verification of antibody specificity for At3g28520 involves multiple complementary approaches. First, researchers typically perform Western blot analysis using wild-type plant extracts alongside At3g28520 knockout mutants. True specific antibodies will show the absence of signal in knockout mutant samples, similar to how AtSerpin1-specific antibodies failed to detect the protein in AtSerpin1 knockout mutants . Second, immunoprecipitation followed by mass spectrometry, as demonstrated with AtSerpin1-HA protein extraction and subsequent trypsin digestion for amino acid determination , provides definitive confirmation of antibody specificity. Third, recombinant protein expression and purification, such as using bacterial expression systems with His-tags (as shown with AtSerpin1 cDNA cloned into pET100/D-TOPO expression vectors ), generates control proteins for antibody validation.

What expression patterns are typically observed for At3g28520 in plant tissues?

Expression patterns for Arabidopsis proteins vary considerably across tissues and developmental stages. Some plant proteins show ubiquitous expression throughout the plant, similar to AtSerpin1 , while others display highly specific localization patterns, like the serpin CsPS-1 that was found exclusively in sieve elements . For At3g28520, researchers should examine expression across different plant organs (roots, leaves, stems, flowers, siliques), developmental stages, and in response to various environmental conditions. Immunoblotting of protein extracts from different tissues using At3g28520-specific antibodies provides the most direct evidence of protein expression patterns, while immunohistochemistry or confocal microscopy with fluorescently-labeled secondary antibodies can reveal subcellular localization.

How can At3g28520 antibodies be used to identify protein interaction partners?

Identifying protein interaction partners using At3g28520 antibodies involves several sophisticated approaches. Co-immunoprecipitation (Co-IP) represents the gold standard method, where researchers use At3g28520 antibodies covalently linked to a matrix (similar to the Seize X protein A immunoprecipitation method employed for AtSerpin1 ) to pull down the target protein along with its binding partners from plant extracts. The precipitated proteins can then be identified through mass spectrometry analysis. Importantly, researchers must include appropriate controls, such as using extracts from knockout plants (as demonstrated with AtSerpin1 and RD21 knockout mutants where the serpin-protease complex was absent ). For detecting transient or weak interactions, crosslinking prior to immunoprecipitation may be necessary to stabilize protein complexes.

What methodological approaches resolve contradictory data when using At3g28520 antibodies?

Resolving contradictory data when using At3g28520 antibodies requires systematic troubleshooting and validation. First, researchers should verify antibody specificity through multiple methods including Western blotting with knockout controls, recombinant protein detection, and epitope mapping. Second, sample preparation conditions significantly impact antibody performance; different protein extraction buffers, detergent concentrations, and reducing agents should be systematically tested. Third, post-translational modifications may affect antibody recognition, necessitating specialized approaches like phosphatase treatment before immunodetection. Fourth, contradictory localization data between immunofluorescence and biochemical fractionation might require complementary approaches such as expression of fluorescently-tagged fusion proteins. Fifth, confirming key findings with alternative antibodies raised against different epitopes of At3g28520 can validate original observations and resolve inconsistencies.

How can crystal structure information enhance At3g28520 antibody development and application?

Crystal structure information dramatically enhances antibody development and application through several mechanisms. First, structural data reveals surface-exposed regions ideal for antibody targeting, similar to how the AtSerpin1 crystal structure at 2.2 Å resolution revealed its electrostatic surface potential and unique structural features . Second, understanding protein domains helps in designing antibodies against functionally relevant regions; for example, knowledge of AtSerpin1's reactive center loop (RCL) and breach region informed functional studies of its protease inhibition mechanism . Third, crystal structures guide epitope selection to avoid regions involved in protein-protein interactions if the goal is to detect these complexes. Fourth, structural information helps predict potential cross-reactivity with similar proteins. Fifth, for post-translational modification detection, crystal structures identify modification sites that can be targeted with modification-specific antibodies. These approaches significantly improve antibody specificity and utility for specialized applications.

What are the optimal extraction conditions for maintaining At3g28520 protein integrity during immunodetection?

Optimal extraction conditions for maintaining At3g28520 protein integrity require careful consideration of multiple factors. First, buffer composition significantly impacts protein stability; a standard extraction buffer might include 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM EDTA, and 10% glycerol. Second, protease inhibitors are essential to prevent degradation; for plant samples, a complete protease inhibitor cocktail with addition of specific inhibitors like PMSF (1 mM) and E-64 (a cysteine protease inhibitor that was shown to prevent formation of complexes with AtSerpin1 ) is recommended. Third, reducing agents such as DTT (1-5 mM) help maintain protein structure by preventing oxidation of cysteine residues. Fourth, mild detergents (0.1-1% Triton X-100 or NP-40) aid in membrane protein solubilization while preserving native structures. Fifth, temperature control is critical; all extraction steps should be performed at 4°C to minimize degradation. These conditions must be optimized specifically for At3g28520 to ensure consistent and reliable antibody detection.

How should researchers optimize immunoprecipitation protocols for At3g28520 protein complexes?

Optimizing immunoprecipitation protocols for At3g28520 protein complexes requires systematic refinement of multiple parameters. First, antibody coupling method significantly impacts efficiency; covalent cross-linking of antibodies to solid support (like Protein A/G beads) using reagents such as dimethyl pimelimidate prevents antibody leaching and contamination of samples, as demonstrated in the AtSerpin1 immunopurification procedure . Second, pre-clearing lysates with beads alone removes non-specific binding proteins. Third, buffer composition requires optimization; for preserving transient interactions, mild detergents (0.1% NP-40) and physiological salt concentrations (150 mM NaCl) are recommended, while more stringent conditions can confirm strong interactions. Fourth, incubation time and temperature affect complex recovery; typically, 2-4 hours at 4°C balances binding efficiency with complex stability. Fifth, washing conditions determine specificity; progressive washes with increasing stringency can reveal interaction strength hierarchies. Additionally, native versus denaturing conditions should be selected based on whether the goal is to capture binding partners or to confirm direct interactions, respectively.

How can researchers distinguish between specific and non-specific signals when using At3g28520 antibodies?

Distinguishing between specific and non-specific signals requires implementation of rigorous controls and validation approaches. First, genetic controls comparing wild-type and knockout/knockdown plants provide the most definitive validation, similar to how AtSerpin1 and RD21 knockout mutants confirmed the specificity of the serpin-protease complex detection . Second, peptide competition assays, where the antibody is pre-incubated with excess antigen peptide before application, should eliminate specific signals while non-specific binding persists. Third, multiple antibodies targeting different epitopes of At3g28520 should produce consistent detection patterns for true signals. Fourth, recombinant protein standards at known concentrations establish signal linearity and detection limits. Fifth, signal intensity across different tissues should correlate with known expression patterns from transcriptomic data. Finally, for immunofluorescence applications, secondary antibody-only controls identify background fluorescence. Implementation of these controls systematically eliminates false positives and ensures reliable data interpretation.

What analytical approaches help resolve complex interaction data involving At3g28520?

Resolving complex interaction data for At3g28520 requires sophisticated analytical approaches beyond standard detection methods. First, fractionation by non-reducing SDS-PAGE can reveal different protein complexes based on molecular weight shifts, as demonstrated with AtSerpin1 and RD21 interactions . Second, two-dimensional gel electrophoresis separating proteins by both isoelectric point and molecular weight can distinguish post-translationally modified forms of interaction partners. Third, proximity-dependent labeling methods like BioID, where At3g28520 is fused to a biotin ligase, can identify proteins in close proximity within the cellular environment. Fourth, quantitative proteomics comparing immunoprecipitates from different conditions using stable isotope labeling allows statistical validation of differential interactions. Fifth, functional validation through mutational analysis of interaction interfaces, guided by structural data similar to the AtSerpin1 crystal structure information , confirms the biological relevance of detected interactions. These complementary approaches together provide robust evidence for protein interaction networks.

How can researchers validate antibody specificity when knockout mutants are unavailable?

Validating antibody specificity without knockout mutants requires alternative rigorous approaches. First, RNA interference (RNAi) or CRISPR-based knockdown of At3g28520 should correspond with reduced antibody signal proportional to the knockdown efficiency. Second, heterologous expression systems can demonstrate specificity by expressing the target protein in systems naturally lacking it (e.g., bacterial or yeast cells) and confirming antibody recognition. Third, mass spectrometry identification of immunoprecipitated proteins provides direct evidence of antibody targets, similar to the validation of AtSerpin1 through tryptic digestion and liquid chromatography . Fourth, epitope mapping using peptide arrays or deletion constructs identifies the exact binding site of antibodies and predicts potential cross-reactivity. Fifth, antibodies against different epitopes of the same protein should show consistent detection patterns. Finally, correlation between protein and mRNA levels across tissues or conditions supports antibody validity. These approaches collectively establish antibody specificity even without the gold standard knockout controls.

How can At3g28520 antibodies be used for conformational change studies?

At3g28520 antibodies can be powerful tools for studying protein conformational changes when strategically developed and applied. First, conformation-specific antibodies can be generated by immunizing with proteins fixed in specific states (active/inactive, bound/unbound) to capture distinct structural epitopes. Second, comparing antibody accessibility in native versus denatured conditions reveals conformationally masked epitopes, similar to how structural studies of AtSerpin1 revealed its metastable stressed state . Third, epitope-specific antibodies targeting regions known to undergo conformational changes (based on structural predictions or homology models) can serve as sensors for these changes. Fourth, FRET-based approaches using fluorescently labeled antibody fragments can detect conformational changes in real-time in living cells. Fifth, antibodies recognizing specific protein-protein interaction interfaces can be used to track complex formation and dissociation. These approaches provide crucial insights into protein dynamics that complement static structural studies and help elucidate the functional mechanisms of At3g28520 in various cellular contexts.

What approaches integrate At3g28520 antibody data with other -omics datasets?

Integrating At3g28520 antibody data with other -omics datasets creates powerful systems biology insights through multiple analytical frameworks. First, correlation analysis between protein abundance (from immunoblotting) and transcript levels (from RNA-seq) identifies post-transcriptional regulation mechanisms. Second, combining immunoprecipitation-mass spectrometry (IP-MS) data with interactome databases creates validated protein interaction networks with functional annotations. Third, overlay of protein localization data (from immunofluorescence) with subcellular proteome maps reveals compartment-specific functions. Fourth, integration with phosphoproteomics or other post-translational modification datasets connects protein regulation with broader signaling networks. Fifth, computational modeling incorporating antibody-derived protein abundance data with metabolomics provides mechanistic insights into metabolic pathway regulation. For visualization and analysis, tools like Cytoscape, String-db, and R-based packages enable multidimensional data integration. This multi-omics approach transforms isolated antibody-based observations into systems-level understanding of At3g28520's role within the plant's biological networks.

How can nanobody technology enhance At3g28520 research applications?

Nanobody technology offers revolutionary advantages for At3g28520 research through several unique properties. First, nanobodies (single-domain antibodies derived from camelids like alpacas) are approximately 10 times smaller than conventional antibodies, allowing them to access restricted epitopes that would be inaccessible to traditional antibodies . Second, their exceptional stability under harsh conditions enables applications in environments where conventional antibodies fail. Third, nanobodies can penetrate living cells for intracellular targeting, enabling real-time tracking of At3g28520 in live plant cells. Fourth, their high specificity and affinity, demonstrated by the precise targeting of proteins like PRL-3 in cancer research , translates to improved signal-to-noise ratios in detection methods. Fifth, nanobodies can be easily engineered for fusion with reporters or functional domains. Specialized applications include super-resolution microscopy, protein crystallization facilitators (as demonstrated with the AtSerpin1 crystal structure ), and conformational stabilizers. The UK Protein Core's capabilities in generating nanobodies for biomedical research provide a methodological framework adaptable to plant protein research including At3g28520.

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.