APRR2 Antibody

Shipped with Ice Packs
In Stock

Description

Molecular Function of APRR2

APRR2 belongs to the pseudo-response regulator family and contains a Myb-like DNA-binding domain (GARP domain). It localizes to the nucleus and regulates:

  • Chlorophyll and carotenoid accumulation: APRR2 modulates chloroplast development by influencing plastid number and pigment content in fruits and leaves .

  • Salicylic acid (SA)-mediated immunity: APRR2 enhances SA biosynthesis and PR1 protein accumulation during Pseudomonas syringae infection in Arabidopsis .

  • Fruit ripening: In cucurbits (melon, watermelon, cucumber), APRR2 determines immature rind color and mature fruit carotenoid levels .

Table 1: APRR2 Functional Insights Across Species

SpeciesRole of APRR2Key MechanismReference
ArabidopsisEnhances SA signaling, camalexin production, and pathogen resistanceUpregulates ICS1, PAD3, and PR1 during bacterial infection
MelonDetermines dark vs. light green rind; regulates chlorophyll and carotenoidsPremature stop codons (e.g., G→T in exon 8) disrupt protein function
WatermelonControls fruit pigmentationAllelic variation in APRR2 correlates with β-carotene content
Bitter gourdGoverns pericarp color via chlorophyll/carotenoid balancePromoter and exonic SNPs (e.g., Indel 4422) linked to pigment content

Table 2: APRR2 Mutational Impact in Melon

Mutation TypeEffect on ProteinPhenotypic Outcome
Exon 8 SNP (G→T)Premature stop codon (292 vs. 527 aa)Light rind, reduced chlorophyll
Exon 9 13-bp insertionFrameshift, truncated protein (430 aa)Impaired chloroplast development
Non-synonymous SNPsAltered DNA-binding domainVariable pigment accumulation

Antibody Applications in APRR2 Research

While no studies explicitly describe "APRR2 antibodies," related methodologies include:

  • RNA immunoprecipitation (RIP): Used to study RNA-binding proteins like PRC2 (e.g., SUZ12 antibody in RIP-Western assays) .

  • Western blotting: Validates protein expression in transgenic lines (e.g., PR1 accumulation in Arabidopsis) .

  • GWAS and allelism tests: Identify APRR2 haplotypes linked to phenotypic variation .

Engineering and Agricultural Relevance

  • Biofortification: APRR2 is a prime target for enhancing carotenoid levels in cucurbits .

  • Disease resistance: Overexpression of APRR2 in Arabidopsis boosts camalexin and SA-dependent defenses .

Unresolved Questions

  • Does APRR2 directly bind DNA or interact with other transcriptional regulators?

  • How do post-translational modifications (e.g., phosphorylation) influence its activity?

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
APRR2 antibody; TOC2 antibody; At4g18020 antibody; T6K21.200Two-component response regulator-like APRR2 antibody; Pseudo-response regulator 2 antibody; TOC2 protein antibody
Target Names
APRR2
Uniprot No.

Target Background

Function
This antibody targets APRR2, a transcriptional activator that binds specifically to the DNA sequence 5'-[AG]GATT-3'.
Database Links

KEGG: ath:AT4G18020

STRING: 3702.AT4G18020.1

UniGene: At.25394

Protein Families
ARR-like family
Subcellular Location
Nucleus.

Q&A

What is AP-2 protein and why is it important in research?

AP-2 (also known as Transcription Factor AP-2 Alpha or TFAP2A) is a critical nuclear transcription factor that regulates gene expression in various developmental and physiological processes. It belongs to the AP-2 family of transcription factors characterized by their DNA-binding domains and dimerization regions. AP-2 is particularly important in embryonic development, cell growth, and differentiation.

In research, AP-2 is studied for its roles in cancer progression, neural crest development, and epithelial biology. The protein has several known synonyms including "Activating enhancer-binding protein 2-alpha," "AP2-alpha," "AP2TF," and "Transcription factor AP-2-alpha" . Detection and quantification of AP-2 using specific antibodies allows researchers to investigate its expression patterns and functional roles across different experimental conditions.

How do I select the appropriate AP-2 antibody for my specific research application?

When selecting an AP-2 antibody, several factors should be considered:

  • Application compatibility: Confirm the antibody has been validated for your intended application (e.g., Western blotting, immunohistochemistry). Commercial AP-2 antibodies like the rabbit polyclonal A38588 are typically validated for specific applications such as Western blot (WB) and immunohistochemistry (IHC) .

  • Species reactivity: Ensure the antibody recognizes AP-2 from your experimental species. Some antibodies, like A38588, are validated for human, mouse, and rat samples .

  • Epitope information: Consider which region of AP-2 the antibody targets, particularly if you're interested in specific isoforms or domains. Some antibodies are generated against synthesized peptides derived from human AP-2 .

  • Validation data: Review the available scientific validation data, including Western blot images and immunohistochemistry results on relevant tissues. Look for clear, specific bands at the expected molecular weight and appropriate staining patterns in tissues known to express AP-2 .

  • Contrastive analysis: Consider computational approaches that predict epitope relationships, as these can help ensure your selected antibody targets the desired epitope region .

What are the standard positive controls for AP-2 antibody validation?

For rigorous AP-2 antibody validation, appropriate positive controls should include:

  • Cell lines: COLO205 cells have been demonstrated as suitable positive controls for AP-2 antibody validation by Western blot . These cells show detectable levels of endogenous AP-2 protein.

  • Tissue samples: Human breast carcinoma tissue is an established positive control for AP-2 antibody validation in immunohistochemistry applications . This tissue type exhibits consistent expression of AP-2, making it ideal for assessing antibody performance.

  • Recombinant protein: Purified recombinant AP-2 protein can serve as a defined positive control, especially when establishing detection limits or assessing batch-to-batch consistency.

  • Overexpression systems: Cells transfected with AP-2 expression constructs provide a robust positive control, particularly useful when testing antibodies in systems where endogenous expression may be low.

When validating AP-2 antibodies, these controls should demonstrate clear, specific signals at the expected molecular weight (approximately 50-52 kDa) in Western blotting or appropriate nuclear localization in immunostaining applications.

What are the optimal conditions for using AP-2 antibodies in Western blotting?

For optimal Western blotting results with AP-2 antibodies, consider the following protocol adaptations:

  • Sample preparation:

    • Lyse cells in a buffer containing phosphatase and protease inhibitors

    • Include appropriate detergents (0.1-1% Triton X-100 or NP-40) to extract nuclear proteins

    • Heat samples at 95°C for 5 minutes in Laemmli buffer with reducing agent

  • Gel electrophoresis and transfer:

    • Use 10-12% polyacrylamide gels for optimal resolution around 50 kDa (AP-2's molecular weight)

    • Transfer to PVDF membranes (rather than nitrocellulose) for better protein retention

    • Use semi-dry transfer at 15-25V for 30-45 minutes or wet transfer at 100V for 60-90 minutes

  • Blocking and antibody incubation:

    • Block membranes with 3-5% BSA or non-fat milk in TBST for 1 hour at room temperature

    • Dilute primary AP-2 antibody (typically 1:500-1:1000) in blocking buffer

    • Incubate with primary antibody overnight at 4°C with gentle agitation

    • Wash 3-5 times with TBST before applying appropriate HRP-conjugated secondary antibody

  • Detection optimization:

    • For enhanced chemiluminescence (ECL), expose for 30 seconds initially, then adjust as needed

    • Consider using high-sensitivity substrates if signal is weak

This protocol has been successfully employed with commercial AP-2 antibodies for detecting endogenous AP-2 protein in various cell lines, including COLO205 cells .

How should I optimize immunohistochemistry protocols for AP-2 antibody staining?

For optimal immunohistochemical detection of AP-2 protein in tissue samples:

  • Tissue preparation and antigen retrieval:

    • Use formalin-fixed, paraffin-embedded (FFPE) sections cut at 4-6 μm thickness

    • Perform heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)

    • Heat in a pressure cooker or microwave for 15-20 minutes, then cool gradually to room temperature

  • Blocking and antibody incubation:

    • Block endogenous peroxidase with 3% hydrogen peroxide for 10 minutes

    • Apply protein block (5% normal goat serum) for 30 minutes at room temperature

    • Dilute primary AP-2 antibody (typically 1:100-1:200) in antibody diluent

    • Incubate overnight at 4°C in a humidified chamber

  • Detection and counterstaining:

    • Use a polymer-based detection system for enhanced sensitivity

    • Develop with DAB chromogen for 3-5 minutes under microscopic control

    • Counterstain nuclei with hematoxylin for 30-60 seconds

    • Dehydrate, clear, and mount with permanent mounting medium

This protocol has been validated for paraffin-embedded human breast carcinoma tissue using commercially available AP-2 antibodies, resulting in clear nuclear staining consistent with AP-2's function as a transcription factor .

How can I determine cross-reactivity between AP-2 antibodies and other AP-2 family members?

To assess potential cross-reactivity with other AP-2 family members (TFAP2B, TFAP2C, TFAP2D, and TFAP2E):

  • Sequence alignment analysis:

    • Perform computational alignment of the immunogen sequence with other AP-2 family members

    • Identify regions of high homology that might lead to cross-reactivity

  • Knockout/knockdown validation:

    • Use CRISPR/Cas9 or siRNA to create AP-2α-deficient cells

    • Test antibody reactivity in Western blot and immunostaining

    • Persistent signal in knockout cells suggests cross-reactivity

  • Overexpression systems:

    • Express each AP-2 family member individually in a heterologous system

    • Perform parallel Western blots with the antibody in question

    • Quantify relative signal intensity across family members

  • Epitope mapping:

    • Apply computational epitope prediction approaches similar to those developed for other antibody families

    • Use peptide arrays to identify the exact binding epitope of the antibody

    • Compare binding profiles against synthetic peptides from different AP-2 family members

For antibodies like the commercial A38588, manufacturers typically validate specificity for AP-2α (TFAP2A) through affinity purification using epitope-specific immunogens , but independent validation using the methods above provides added confidence in antibody specificity.

How can computational epitope prediction improve AP-2 antibody selection?

Recent advances in computational epitope prediction offer powerful new approaches for AP-2 antibody selection:

  • Sequence-based prediction thresholds:

    • When antibody pairs share both heavy and light chain V-genes and have >70% CDRH3 amino acid identity, they consistently bind overlapping epitopes

    • This threshold can help identify antibodies likely to recognize similar epitopes on AP-2 protein

  • Supervised contrastive learning approaches:

    • Language model embeddings enhanced with epitope-specificity information can predict epitope relationships with up to 74.4% accuracy

    • These models generate unified sequence embeddings through a dual-stream transformer network followed by multi-layer perceptron processing

  • Generalized models across protein families:

    • Models like AbLang-PDB extend epitope prediction capabilities across diverse protein families

    • Such approaches achieve five-fold increases in average precision for overlapping-epitope prediction compared to sequence-based methods

  • Confidence scoring:

    • Cosine similarity metrics provide reliable confidence scores for overlapping epitope antibodies

    • High-confidence predictions (cosine similarity >0.5) show strong correlation (ρ = 0.811) with actual epitope overlap

Applying these computational approaches to AP-2 antibody selection can significantly improve experimental outcomes by identifying antibodies that target unique or overlapping epitopes based on specific research needs.

What strategies can enhance AP-2 detection in challenging samples with low expression?

For detecting AP-2 in samples with low expression levels:

  • Signal amplification systems:

    • Implement tyramide signal amplification (TSA) to enhance chromogenic or fluorescent detection

    • Use polymer-based detection systems with multiple HRP molecules per antibody

  • Sample enrichment:

    • Perform nuclear fractionation to concentrate AP-2 protein before Western blotting

    • Use immunoprecipitation to enrich AP-2 before detection

  • Optimized antibody combinations:

    • Employ a cocktail of non-competing AP-2 antibodies targeting different epitopes

    • Combine with phospho-specific antibodies if studying activated forms of AP-2

  • Enhanced imaging and analysis:

    • Use computational deconvolution for improved signal-to-noise ratio in fluorescence imaging

    • Apply digital pathology tools for quantitative analysis of weak IHC signals

  • Epitope retrieval optimization:

    • Test multiple antigen retrieval methods (heat, enzymatic, pH variations)

    • Extend retrieval times for challenging FFPE tissues

These strategies have been successfully applied to detect low levels of nuclear transcription factors in various research contexts and can be adapted for AP-2 detection using commercially available antibodies that detect endogenous levels of total AP-2 protein .

How can I design experiments to study AP-2 protein-protein interactions using antibody-based approaches?

To investigate AP-2 protein-protein interactions:

  • Co-immunoprecipitation (Co-IP) optimization:

    • Select AP-2 antibodies validated for immunoprecipitation applications

    • Use gentle lysis conditions to preserve protein complexes (150-300 mM NaCl, 0.1-0.5% NP-40)

    • Pre-clear lysates with protein A/G beads to reduce non-specific binding

    • Include appropriate negative controls (isotype-matched antibodies, IgG)

  • Proximity ligation assay (PLA):

    • Combine AP-2 antibody with antibodies against suspected interaction partners

    • Optimize antibody dilutions and incubation conditions for maximum signal-to-noise ratio

    • Include proper controls for antibody specificity and PLA reagent performance

  • Bimolecular fluorescence complementation (BiFC):

    • Design constructs expressing AP-2 fused to one fragment of a fluorescent protein

    • Express potential interaction partners fused to complementary fragments

    • Monitor fluorescence reconstitution as evidence of protein-protein interaction

  • FRET-based approaches:

    • Use antibodies conjugated with appropriate FRET donor/acceptor fluorophores

    • Measure energy transfer as indication of protein proximity

    • Calculate FRET efficiency to estimate interaction strength

When designing these experiments, consider the antibody's specific epitope on AP-2 to ensure that antibody binding doesn't disrupt or artificially enhance the protein-protein interactions being studied. For commercial antibodies like A38588, knowing that they target synthetic peptide derivatives of human AP-2 can help assess potential interference with interaction domains.

What are common causes of false positive or false negative results when using AP-2 antibodies?

Understanding potential pitfalls in AP-2 antibody experiments is crucial for accurate interpretation:

False Positive Causes:

  • Cross-reactivity: AP-2 family members share structural similarities; antibodies may recognize multiple family members unless specifically validated

  • Non-specific binding: Insufficient blocking or inappropriate antibody dilution can lead to background signal

  • Detection system artifacts: Endogenous peroxidase activity or biotin can generate signal independent of specific antibody binding

  • Sample contamination: Inadvertent introduction of AP-2-expressing cells into negative samples

False Negative Causes:

  • Epitope masking: Post-translational modifications or protein-protein interactions may block antibody access to the target epitope

  • Inadequate antigen retrieval: Insufficient unmasking of epitopes in fixed tissues

  • Protein degradation: Poor sample handling leading to proteolysis of the AP-2 protein

  • Suboptimal detection sensitivity: Using inappropriate secondary antibodies or detection reagents

Mitigation Strategies:

  • Multiple antibody validation: Use at least two antibodies targeting different AP-2 epitopes

  • Comprehensive controls: Include positive controls (known AP-2 expressors like COLO205 cells) , negative controls (AP-2 knockout/knockdown samples), and technical controls (omitting primary antibody)

  • Quantitative assessment: Use digital image analysis to establish clear thresholds for positive signals

  • Orthogonal validation: Confirm antibody results with non-antibody methods (RT-PCR, CRISPR screens)

By systematically addressing these factors, researchers can significantly improve the reliability of AP-2 antibody-based experiments.

How should I evaluate batch-to-batch variability in AP-2 antibody performance?

To systematically assess batch-to-batch variability in AP-2 antibodies:

  • Standard sample panel testing:

    • Create a reference panel of positive and negative controls (e.g., COLO205 cells, breast carcinoma tissue)

    • Preserve samples as frozen aliquots or FFPE blocks to ensure consistency

    • Test each new antibody batch against this standard panel

  • Quantitative metrics:

    • Establish quantifiable parameters: signal-to-noise ratio, EC50 values, staining intensity

    • Use digital image analysis for objective comparison

    • Implement statistical thresholds for acceptable performance variation

  • Comparative experimental design:

    • Run parallel experiments with previous and new antibody batches

    • Use titration series to identify potential shifts in optimal concentration

    • Document lot-specific optimization requirements

  • Long-term monitoring:

    • Maintain a database of performance metrics across batches

    • Track trending changes that might indicate manufacturing drift

    • Implement control charts to visualize performance over time

  • Computational approaches:

    • Consider applying epitope prediction models to assess potential manufacturing variations

    • Computational models that predict overlapping-epitope antibodies can help identify antibodies that should theoretically perform similarly

This systematic approach allows researchers to maintain experimental consistency despite the inherent variability in antibody production, particularly important for long-term studies of AP-2 expression or function.

How are contrastive learning approaches transforming antibody design for transcription factors like AP-2?

Contrastive learning is revolutionizing antibody design for transcription factor detection:

  • Epitope-specificity encoding:

    • Supervised contrastive learning frameworks can encode epitope-specificity information directly into antibody sequence embeddings

    • These approaches utilize dual-stream transformer networks followed by multi-layer perceptrons to generate unified sequence embeddings

    • Modified normalized temperature-scaled cross-entropy (NT-Xent) loss functions process multiple positive examples simultaneously

  • Embedding space optimization:

    • This technique concurrently attracts all antibodies sharing an epitope within a training batch while repelling those binding distinct epitopes

    • The resulting embedding space captures nuanced epitope relationships beyond what traditional sequence analysis can achieve

    • Models learn antibody sequence patterns indicative of shared epitope binding that are missed by V-gene and CDRH3 thresholds

  • Cross-antigen applications:

    • Generalized models like AbLang-PDB extend epitope prediction capabilities across diverse protein families

    • Such approaches could be applied to develop AP-2 antibodies with precisely defined epitope characteristics

    • This would allow rational selection of antibody pairs for sandwich assays or multiplexed detection

  • Practical validation:

    • These computational approaches have demonstrated practical success in identifying antibodies sharing epitope overlap with therapeutically relevant antibodies like 8ANC195 for HIV-1

    • Similar approaches could identify or design optimal AP-2 antibodies for specific research applications

These advances suggest that future AP-2 antibody development will increasingly incorporate computational design elements, potentially leading to antibodies with enhanced specificity, defined epitope targeting, and improved performance characteristics.

What new methodological approaches are enhancing the specificity of nuclear transcription factor detection?

Innovative methodologies are improving nuclear transcription factor detection specificity:

  • Proximity-based detection systems:

    • Proximity extension assays (PEA) using oligonucleotide-coupled antibodies provide improved specificity

    • Only when both antibodies bind their target is a signal generated, dramatically reducing background

  • Single-molecule detection platforms:

    • Digital ELISA approaches can detect individual protein molecules

    • These ultra-sensitive methods improve detection of low-abundance transcription factors like AP-2

  • Spatially resolved antibody-based assays:

    • Multiplexed ion beam imaging (MIBI) and imaging mass cytometry combine antibody specificity with spatial resolution

    • These approaches allow visualization of AP-2 in the context of other cellular markers

  • Antibody engineering approaches:

    • Recombinant antibody fragments (Fabs, scFvs) offer improved tissue penetration

    • Engineered scaffolds based on clinical antibodies with replicated natural complementarity-determining regions provide enhanced specificity

  • Computational validation pipelines:

    • Integration of experimental data with computational epitope prediction

    • Models that predict overlapping-epitope antibodies (like AbLang-PDB) show five-fold improvement in average precision compared to sequence-based methods

These emerging approaches are particularly valuable for nuclear transcription factors like AP-2, where distinguishing specific signal from background in the nuclear compartment has traditionally been challenging.

How might machine learning integration improve AP-2 antibody applications in multi-omics research?

Machine learning is transforming antibody applications in multi-omics research contexts:

  • Integrated data analysis:

    • Neural networks can correlate AP-2 antibody-based detection with transcriptomic and epigenomic data

    • This integration improves interpretation of AP-2's functional role in complex cellular processes

  • Automated image analysis:

    • Convolutional neural networks analyze immunohistochemistry or immunofluorescence images

    • These systems can quantify nuclear AP-2 localization, intensity, and heterogeneity across tissue samples

  • Predictive epitope mapping:

    • Supervised contrastive learning frameworks encode epitope-specificity information

    • These approaches enable selection of antibodies targeting specific AP-2 domains relevant to particular research questions

  • Improved experimental design:

    • Machine learning models predict optimal antibody combinations for multiplexed detection

    • Algorithms identify potential cross-reactivity before experimental implementation

  • Validation frameworks:

    • Automated systems assess antibody performance metrics across multiple experiments

    • These frameworks establish confidence scores for antibody-generated data

The incorporation of these machine learning approaches is particularly valuable for transcription factor research, where integrating protein-level data (from antibody-based detection) with genome-wide binding data (from ChIP-seq) and expression analysis (from RNA-seq) provides a comprehensive understanding of factors like AP-2.

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.