ARR13 Antibody

Shipped with Ice Packs
In Stock

Description

Contextualizing ARR13 in Biological Systems

ARR13 is part of the type-B response regulators in plants, such as Arabidopsis thaliana, which mediate cytokinin signaling. These proteins typically lack kinase activity and function as transcription factors to regulate gene expression downstream of cytokinin perception . Key findings include:

  • Functional Studies: ARR13, ARR19, and ARR20 failed to rescue the arr1 arr12 mutant phenotype in root growth assays, indicating distinct roles compared to ARR1 and ARR12 .

  • Expression Patterns: Type-B ARRs like ARR13 exhibit tissue-specific expression, with roles in modulating cytokinin responses such as shoot regeneration and stress adaptation .

Cross-Referencing with ARR3 Antibody Research

Given the absence of ARR13-specific antibody data, insights can be drawn from ARR3 (Arrestin 3) antibody studies, which highlight methodologies and challenges in developing antibodies for similar proteins.

Challenges in Developing Antibodies for ARR13

The lack of ARR13-specific antibodies in commercial catalogs or peer-reviewed studies may stem from:

  1. Low Research Priority: ARR13’s role in plant signaling contrasts with the therapeutic focus on human proteins like ARR3.

  2. Technical Barriers:

    • Antigen Stability: ARR13’s intracellular localization and potential instability may hinder epitope accessibility.

    • Cross-Reactivity: Homology with other type-B ARRs (e.g., ARR1, ARR12) could lead to non-specific binding.

  3. Validation Gaps: Antibody characterization crises, as noted in , emphasize the need for rigorous validation (e.g., Western blot, immunoprecipitation) to ensure specificity.

Recommendations for Future Research

To advance ARR13 antibody development, researchers should:

  1. Optimize Immunogen Design: Use synthetic peptides or recombinant ARR13 protein for immunization, focusing on unique regions to minimize cross-reactivity.

  2. Employ Bispecific or Chimeric Approaches: Inspired by bispecific antibody platforms (e.g., ART-Ig) , engineer ARR13-specific antibodies with enhanced binding affinity or dual functionality.

  3. Leverage High-Throughput Screening: Utilize antibody arrays or DNA-based scaffolds to identify high-affinity binders efficiently.

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
ARR13 antibody; At2g27070 antibody; T20P8.12Putative two-component response regulator ARR13 antibody
Target Names
ARR13
Uniprot No.

Target Background

Function
ARR13 is a putative transcriptional activator protein that exhibits specific binding affinity for the DNA sequence 5'-[AG]GATT-3'. It functions as a response regulator within a His-to-Asp phosphorelay signal transduction system. Phosphorylation of the aspartic acid residue in its receiver domain activates the protein, enabling it to promote the transcription of target genes. ARR13 may directly activate certain type-A response regulators in response to cytokinin signaling.
Database Links
Protein Families
ARR family, Type-B subfamily
Subcellular Location
Nucleus.

Q&A

What is ARR3 antibody and what cellular functions does the target protein regulate?

ARR3 (Arrestin 3, Retinal X-Arrestin) antibodies recognize the protein product of the ARR3 gene, which functions primarily in retinal cells. This protein belongs to the arrestin family that regulates G protein-coupled receptor (GPCR) signaling. The C-terminus-specific antibodies, such as ABIN6260057, detect endogenous levels of total Arrestin C . The ARR3 protein plays a crucial role in photoreceptor function, particularly in cone cells, where it participates in the desensitization of activated photopigments during visual signal transduction.

Unlike its better-known relatives (β-arrestin-1 and β-arrestin-2), ARR3 has a more specialized function in visual processes rather than broad GPCR regulation. When designing experiments targeting ARR3, researchers should consider its tissue-specific expression patterns, primarily in retinal tissues, to establish appropriate controls and experimental conditions.

How do polyclonal and monoclonal anti-ARR3 antibodies differ in research applications?

The choice between polyclonal and monoclonal anti-ARR3 antibodies significantly impacts experimental outcomes. Polyclonal antibodies, such as ABIN6260057 (rabbit host), recognize multiple epitopes on the ARR3 protein, offering advantages in signal amplification and detection of proteins in various conformational states . These antibodies are particularly effective in applications like Western blotting and immunohistochemistry where high sensitivity is required.

In contrast, monoclonal antibodies (e.g., mouse monoclonal 2D7) target a single epitope with high specificity . This property makes them superior for distinguishing between closely related proteins, such as different arrestin family members, and for applications requiring consistent lot-to-lot performance. Monoclonal antibodies typically produce cleaner results in immunoprecipitation studies and protein interaction analyses.

When designing experiments, researchers should consider these differences and select the appropriate antibody format based on the specific research question. For preliminary studies or when protein expression is low, polyclonal antibodies often provide better detection. For highly specific applications or when distinguishing between closely related protein isoforms, monoclonal antibodies are preferred.

What validation methods ensure reliable results with ARR3 antibodies?

Proper antibody validation is essential for experimental reproducibility. For ARR3 antibodies, multiple validation approaches should be employed:

  • Western blot validation: Confirm antibody specificity by detecting a protein of the expected molecular weight (ARR3 is approximately 40-45 kDa). Compare results with positive and negative control tissues (retinal tissue as positive; non-expressing tissues as negative).

  • Knockout/knockdown controls: If possible, validate using ARR3 knockout models or siRNA-mediated knockdown samples to confirm signal specificity.

  • Peptide competition: Pre-incubate the antibody with the immunizing peptide to demonstrate that this blocks specific binding, as done during the development of antibodies like ABIN6260057, which uses a synthesized peptide from the C-terminal region of human Arrestin C .

  • Cross-reactivity assessment: Test the antibody against related arrestin family members to ensure specificity, particularly important when studying ARR3 in systems that express multiple arrestin proteins.

  • Multiple antibody concordance: Compare results using different antibodies targeting distinct epitopes of ARR3 to verify consistent detection patterns.

Researchers should document these validation steps in their methods sections to strengthen the reliability of their findings and facilitate reproducibility.

How should ARR3 antibodies be optimized for Western blotting protocols?

Optimizing Western blotting protocols for ARR3 detection requires attention to several critical factors:

  • Sample preparation: For retinal samples (primary source of ARR3), use RIPA buffer supplemented with protease inhibitors to prevent degradation. Similar to the protocol used for ARPC1A antibody testing, load approximately 35 μg of protein per lane .

  • Antibody dilution: Begin with a 1:500 to 1:1000 dilution for polyclonal ARR3 antibodies like ABIN6260057, and adjust based on signal intensity. For reference, other antibodies in similar applications use concentrations around 0.1-0.3 μg/mL .

  • Blocking strategy: Use 5% non-fat dry milk in TBST for standard blocking, but consider 5% BSA if background issues persist.

  • Incubation conditions: Optimal primary antibody incubation should be conducted at 4°C overnight, though some antibodies may perform well with shorter incubations (1 hour) at room temperature .

  • Detection system: For ARR3, which may have moderate expression levels in some tissues, enhanced chemiluminescence (ECL) systems provide suitable sensitivity.

  • Size verification: Confirm detection at the expected molecular weight (ARR3 has a predicted molecular weight of approximately 40-45 kDa), being aware that post-translational modifications may affect migration patterns.

The inclusion of positive controls (retinal tissue lysate) and negative controls (tissues not expressing ARR3) is essential for proper interpretation of results.

What are the critical considerations for immunofluorescence applications with ARR3 antibodies?

Successful immunofluorescence (IF) studies with ARR3 antibodies require careful attention to fixation methods, permeabilization, and signal amplification:

  • Fixation: For ARR3 detection, 4% paraformaldehyde (PFA) for 15-20 minutes at room temperature preserves epitope accessibility while maintaining cellular structure. Avoid methanol fixation which can destroy some epitopes.

  • Permeabilization: Use 0.1-0.3% Triton X-100 to permeabilize membranes, allowing antibody access to intracellular ARR3.

  • Blocking: Implement a robust blocking step (5-10% normal serum from the secondary antibody host species) to minimize non-specific binding.

  • Antibody concentration: For polyclonal ARR3 antibodies like ABIN6260057 (which is validated for IF), start with a 1:100 to 1:200 dilution and optimize through titration experiments .

  • Controls: Include peptide competition controls where the antibody is pre-incubated with the immunizing peptide to confirm specificity of staining patterns.

  • Co-localization studies: Consider dual-labeling with markers of subcellular compartments (e.g., membrane markers for receptor-associated ARR3) to confirm expected localization patterns.

  • Signal amplification: For tissues with lower ARR3 expression, tyramide signal amplification systems may improve detection sensitivity while maintaining specificity.

When interpreting results, expect ARR3 staining primarily in retinal cone cells, with potential localization differences depending on light adaptation state of the tissue.

How can researchers effectively use ARR3 antibodies in multiplex detection systems?

Multiplex detection involving ARR3 requires strategic planning to prevent cross-reactivity and signal interference:

  • Antibody selection: Choose ARR3 antibodies raised in host species that differ from other target antibodies in your multiplex panel. For example, if using rabbit polyclonal ARR3 antibodies like ABIN6260057, pair with mouse or goat antibodies for other targets .

  • Sequential staining: For challenging combinations, implement sequential staining protocols with complete blocking steps between each antibody application.

  • Fluorophore selection: When designing multiplex immunofluorescence, select fluorophores with minimal spectral overlap and consider the tissue's autofluorescence characteristics (particularly important in retinal tissue, which can have significant autofluorescence).

  • Cross-reactivity testing: Before full experimental implementation, test each antibody individually and in combination to identify any cross-reactivity issues.

  • Antibody array technology: For broader protein detection panels, consider antibody array approaches similar to the Human Inflammation Antibody Array (ab134003), which allows simultaneous detection of multiple targets . These systems could be adapted for neurological protein panels that include ARR3.

  • Validation with single-plex controls: Always run single-antibody controls in parallel with multiplex experiments to ensure detection specificity is maintained in the multiplex setting.

The optimization of multiplex protocols should be systematic, with careful documentation of all parameters to ensure reproducibility.

How can ARR3 antibodies be utilized to investigate protein-protein interactions?

ARR3 interactions with other proteins can be studied using several antibody-based approaches:

  • Co-immunoprecipitation (Co-IP): ARR3 antibodies targeting the C-terminal region, such as ABIN6260057, can be used to pull down ARR3 and its binding partners . Importantly, epitope location should be considered, as antibodies binding to interaction interfaces may disrupt protein complexes.

  • Proximity ligation assay (PLA): This technique can detect ARR3 interactions with other proteins within a 40 nm radius in fixed cells or tissues. It requires antibodies from different host species—the rabbit polyclonal ARR3 antibody could be paired with mouse antibodies against potential interaction partners.

  • FRET/BRET analysis: While not directly using antibodies during the assay, ARR3 antibodies are valuable for validating the expression and localization of tagged ARR3 constructs used in these energy transfer techniques.

  • Cross-linking mass spectrometry: ARR3 antibodies can help validate crosslinked complexes identified through mass spectrometry approaches, confirming the presence of ARR3 in protein complexes.

  • Pull-down assays with recombinant proteins: Validate interactions identified through other methods by using purified components and ARR3-specific antibodies for detection.

When designing interaction studies, researchers should consider ARR3's known binding partners (such as phosphorylated opsins) and the potential regulatory effects of phosphorylation on these interactions.

What approaches enable differentiation between ARR3 and other arrestin family members?

Distinguishing ARR3 from other arrestin family proteins requires strategic experimental design:

  • Epitope selection: Utilize antibodies targeting regions of maximum divergence between arrestins. The C-terminal region of ARR3 (targeted by ABIN6260057) offers greater sequence uniqueness compared to conserved central domains .

  • Western blot discrimination: Arrestin family members have different molecular weights (ARR3: ~40 kDa, β-arrestin-1: ~48 kDa, β-arrestin-2: ~46 kDa, SAG/arrestin-1: ~45 kDa), allowing discrimination on well-resolved Western blots.

  • Isoform-specific knockdown: Validate antibody specificity using siRNA knockdown of specific arrestin isoforms, confirming that ARR3 antibody signal decreases only when ARR3 is depleted.

  • Tissue specificity exploitation: Leverage ARR3's enriched expression in retinal cone photoreceptors compared to other arrestins, which have broader expression patterns.

  • Differential extraction: Some arrestins show different solubility in various extraction buffers, which can be exploited for partial biochemical separation before antibody detection.

  • Mass spectrometry validation: For definitive identification, combine immunoprecipitation using ARR3 antibodies with mass spectrometry analysis to confirm identity based on unique peptide sequences.

Researchers should always validate their system by testing antibody reactivity against recombinant versions of all arrestin family members when possible.

How do post-translational modifications affect ARR3 antibody recognition?

Post-translational modifications (PTMs) can significantly impact antibody-epitope interactions:

  • Phosphorylation effects: ARR3 undergoes regulatory phosphorylation, which may mask epitopes or change protein conformation. Antibodies like ABIN6260057 that target the C-terminal region may have altered binding efficiency depending on the phosphorylation state .

  • Conformation-specific detection: Some antibodies may preferentially recognize specific conformational states of ARR3 (active vs. inactive). This property should be characterized during validation to understand potential binding biases.

  • PTM-specific antibodies: For studying ARR3 regulation, consider using antibodies that specifically detect phosphorylated forms of ARR3, though these would need to be separately validated.

  • Sample preparation considerations: Phosphatase inhibitors should be included in lysis buffers when studying phosphorylated forms of ARR3. Conversely, dephosphorylation treatment may be necessary when using antibodies that preferentially bind non-phosphorylated epitopes.

  • Multiple antibody approach: Using antibodies targeting different regions of ARR3 can help create a more complete picture of the protein's modification state and ensure detection regardless of modification status.

When publishing results, researchers should clearly specify which ARR3 epitope was targeted and discuss potential impacts of known PTMs on detection.

What strategies can resolve non-specific binding in Western blots with ARR3 antibodies?

Non-specific binding is a common challenge when working with antibodies. For ARR3 antibodies, consider these approaches:

  • Blocking optimization: Test different blocking agents beyond standard milk or BSA, such as fish gelatin or commercial blocking solutions, which may more effectively prevent non-specific interactions.

  • Antibody dilution adjustment: Increase the dilution of primary ARR3 antibody, as concentrated antibody solutions can contribute to non-specific binding. For polyclonal antibodies like ABIN6260057, testing a range from 1:500 to 1:5000 may help identify optimal specificity .

  • Washing stringency: Increase the number and duration of washing steps, and consider adding low concentrations (0.05-0.1%) of SDS to TBST wash buffer to disrupt weak, non-specific interactions.

  • Pre-adsorption: Pre-incubate the ARR3 antibody with proteins from non-expressing tissues to remove antibodies that bind to proteins other than ARR3.

  • Selective extraction: Given that Arrestin C is often studied in retinal tissue, which contains many specialized proteins, optimizing extraction protocols to enrich for soluble proteins and reduce membrane components may improve specificity.

  • Secondary antibody selection: Ensure the secondary antibody is highly cross-adsorbed against proteins from the species being studied to minimize non-specific interactions.

  • Sample preparation refinement: Consider size-exclusion or ion-exchange chromatography to partially purify samples before Western blotting, particularly when working with complex tissue lysates.

How can researchers address inconsistent results between different experimental platforms?

When ARR3 antibody results vary between techniques (e.g., Western blot vs. immunofluorescence), systematic troubleshooting is required:

  • Epitope accessibility assessment: Different preparation methods may affect epitope exposure. For ARR3 antibodies targeting the C-terminus like ABIN6260057, denaturation in Western blotting may expose epitopes hidden in native conformations used in other techniques .

  • Fixation method comparison: Test multiple fixation protocols for immunohistochemistry/immunofluorescence, as some fixatives may better preserve ARR3 epitopes (e.g., PFA vs. methanol).

  • Buffer system evaluation: Different buffers used across techniques may affect antibody binding. Standardize or systematically test buffer components (pH, salt concentration, detergents) to identify optimal conditions.

  • Sample processing effects: Heat, reducing agents, and detergents used in Western blotting may alter epitope structure differently than milder conditions used in other techniques. Consider native gel electrophoresis as an intermediary approach.

  • Cross-validation with multiple antibodies: Use several ARR3 antibodies targeting different epitopes across techniques to build a more complete understanding of detection patterns.

  • Quantitative validation: Implement quantitative standards (e.g., known quantities of recombinant ARR3) to calibrate detection across platforms and establish detection limits for each technique.

Documentation of these optimization steps in research protocols facilitates reproducibility and allows other researchers to adapt methods to their specific experimental systems.

How are AI-based approaches enhancing antibody development for targets like ARR3?

Artificial intelligence is revolutionizing antibody research through several mechanisms:

These AI approaches promise to accelerate development of next-generation ARR3 antibodies with superior specificity, affinity, and production characteristics, potentially democratizing access to high-quality reagents as projected in the VUMC initiative .

What alternative antibody formats might improve ARR3 detection in challenging experimental contexts?

Novel antibody formats offer potential advantages for difficult applications:

  • Fab H3 fragments: This alternative antibody fragment format replaces constant domains with engineered IgG1 CH3 domains, potentially offering "higher soluble yields than its Fab counterpart and a comparable binding affinity against the target antigen" . For ARR3 studies in complex matrices, such formats might improve penetration and reduce non-specific binding.

  • Single-domain antibodies (nanobodies): Derived from camelid antibodies, nanobodies' small size (~15 kDa vs. ~150 kDa for conventional antibodies) enables access to sterically hindered epitopes on ARR3, potentially revealing functional regions obscured in traditional assays.

  • Aptamer alternatives: DNA/RNA aptamers selected against ARR3 might offer advantages in certain applications, including reversible binding under mild conditions and compatibility with live-cell imaging.

  • Recombinant antibody fragments: Using techniques similar to those described for Fab H3, expressing ARR3-targeting antibody fragments in E. coli cytoplasm using the CyDisCo system could yield "natively folded" detection reagents with "comparable affinity to its Fab counterpart" .

  • Bispecific formats: Antibodies engineered to simultaneously bind ARR3 and another protein of interest could enable direct detection of protein interactions or localization relative to subcellular markers.

These alternative formats may help overcome the "bottlenecks associated with the folding and production of Fabs" noted in recent research, potentially making ARR3 detection more accessible and reliable across diverse experimental contexts .

How can antibody arrays be leveraged for studying ARR3 in complex physiological contexts?

Antibody array technologies offer powerful approaches for contextualizing ARR3 function:

  • Custom neurological signaling arrays: Based on platforms like the Human Inflammation Antibody Array that can detect "40 Human Inflammatory Factors," custom arrays could simultaneously measure ARR3 alongside related signaling molecules in visual transduction pathways .

  • Tissue microenvironment profiling: Arrays detecting ARR3 alongside tissue-specific markers could map its expression and activation state across development or disease progression in retinal tissues.

  • Post-translational modification arrays: Specialized arrays with antibodies recognizing different phosphorylated forms of ARR3 and related proteins could profile arrestin pathway activation states in response to various stimuli.

  • Localized signalosome mapping: Proximity-based antibody arrays could detect proteins physically associated with ARR3 in different cellular compartments, revealing context-specific interaction networks.

  • Reverse-phase protein arrays (RPPA): This technique allows quantitative measurement of ARR3 expression across many samples simultaneously, enabling large-scale studies of expression correlation with physiological or pathological states.

The advantages of array technologies include "simultaneous detection" of multiple factors, making them valuable for systems biology approaches to understanding ARR3 function in complex biological contexts .

How does ARR3 function differ from other arrestin family members in experimental systems?

Understanding the functional specialization of ARR3 requires comparative experimental approaches:

  • Receptor coupling specificity: Unlike β-arrestins that regulate hundreds of GPCRs, ARR3 shows preferential binding to visual opsins, particularly cone opsins. Experimental designs should account for this specialization when studying arrestin family function.

  • Kinetic differences: ARR3 typically exhibits faster binding and dissociation kinetics than other arrestins, which influences experimental design parameters such as time points for measuring responses.

  • Expression pattern distinctions: While β-arrestins are ubiquitously expressed, ARR3 shows highly restricted expression primarily in retinal cone photoreceptors. This tissue specificity should inform sample selection and control tissues.

  • Regulatory mechanisms: ARR3 regulation differs from other arrestins, with distinct phosphorylation sites and binding partners. Phospho-specific antibodies targeting ARR3-specific sites would be valuable for distinguishing its regulation from other family members.

  • Subcellular trafficking patterns: ARR3 shows distinctive translocation patterns in response to light compared to rod arrestin (SAG/arrestin-1), requiring specialized imaging conditions for proper characterization.

When designing experiments to study ARR3, researchers should carefully consider these functional differences and validate findings with isoform-specific approaches, particularly when using antibodies like ABIN6260057 that target specific regions of the protein .

What considerations are important when designing cross-species studies using ARR3 antibodies?

Cross-species ARR3 research presents both challenges and opportunities:

  • Epitope conservation analysis: Before applying antibodies across species, analyze sequence conservation at the epitope region. For ARR3 antibodies targeting the C-terminus like ABIN6260057, this region shows moderate conservation across mammals but may diverge in non-mammalian vertebrates .

  • Validation hierarchy: Establish a validation hierarchy, starting with species with confirmed reactivity (Human, Mouse for ABIN6260057) before extending to predicted reactive species (Pig, Bovine, Horse, Sheep, Rabbit, Dog, Chicken, Xenopus) .

  • Control selection: Include species-appropriate positive controls (retinal tissue) and negative controls (non-retinal tissues) to verify specificity in each new species.

  • Dilution optimization: Optimization of antibody dilutions may differ across species due to subtle epitope variations. Perform titration experiments for each new species rather than transferring protocols directly.

  • Fixation adaptation: Different species may require modified fixation protocols to preserve ARR3 epitopes effectively, particularly for immunohistochemistry applications.

  • Evolutionary context interpretation: When comparing ARR3 expression or function across species, consider evolutionary adaptations in visual systems (e.g., nocturnal vs. diurnal species, rod vs. cone dominance) that may influence interpretation.

Researchers should document cross-species validation efforts in publications to build a community resource of verified applications across experimental models.

How can researchers integrate antibody-based detection with genomic and transcriptomic data for comprehensive ARR3 analysis?

Multi-omics integration enhances understanding of ARR3 biology:

  • Expression correlation verification: Use ARR3 antibodies to confirm protein-level expression patterns observed in transcriptomic datasets, addressing potential post-transcriptional regulation.

  • Splice variant detection: Design epitope-specific antibodies to distinguish ARR3 splice variants identified through RNA-sequencing, particularly important in specialized tissues like retina.

  • Genetic variant impact assessment: For genetic variants affecting ARR3 (identified through genomic studies), use antibody-based approaches to assess impacts on protein expression, localization, and interaction patterns.

  • Cell-type resolved expression mapping: Combine single-cell RNA-sequencing data with immunohistochemistry using ARR3 antibodies to validate cell type-specific expression patterns at protein level.

  • Functional genomics integration: Use ARR3 antibodies to assess protein-level outcomes of CRISPR-based gene editing or RNAi experiments, providing functional validation of genomic interventions.

  • Quantitative proteomics calibration: Use ARR3 antibodies as standards for calibrating mass spectrometry-based proteomic quantification, enabling more accurate protein quantification.

This integrative approach leverages the strengths of antibody-based detection (protein-level, spatial information) with the comprehensive nature of -omics approaches to build complete models of ARR3 function in visual 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 2025 TheBiotek. All Rights Reserved.