ARR2 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Overview of ARR2 Antibody

The term "ARR2 Antibody" refers to antibodies targeting the Beta Arrestin 2 protein (ARRB2), a member of the arrestin family involved in G protein-coupled receptor (GPCR) signaling regulation. ARRB2 plays critical roles in receptor desensitization, internalization, and intracellular signaling cascades. Antibodies against ARRB2 are widely used in research to study its expression, localization, and interaction partners in cellular pathways .

Target Protein Structure

  • ARRB2: A 46 kDa cytoplasmic protein composed of 409 amino acids, featuring an N-terminal arrestin domain and a C-terminal phosphoinositide-binding domain .

  • Key Domains:

    • Arrestin-N domain: Mediates receptor recognition.

    • Arrestin-C domain: Facilitates clathrin and AP2 binding during receptor internalization .

Antibody Characteristics

  • Clone 3G1 (ab54790): A mouse monoclonal antibody validated for Western blot (WB), immunocytochemistry (ICC), and immunofluorescence (IF).

    • Epitope: Linear epitope within residues 200–300 of human ARRB2 .

    • Specificity: Confirmed using ARRB2 knockout cell lines (e.g., HepG2 and A549) .

Expression Profiling

Cell LineARRB2 Expression (WB)Knockout Validation
Wild-type HepG246 kDa band observedNo band in ARRB2 KO
Wild-type A54945 kDa band observedNo band in ARRB2 KO
K562Weak expressionN/A
Data derived from abcam validation experiments .

Functional Studies

  • GPCR Regulation: ARRB2 antibodies have been used to demonstrate its role in β2-adrenergic receptor desensitization and MAPK pathway activation .

  • Disease Associations: Elevated ARRB2 levels correlate with cancer progression and neurological disorders, as shown in hippocampal neuron studies .

Autoimmunity and Inflammation

  • Autoantibodies against ARRB2 have been detected in systemic lupus erythematosus (SLE) and severe COVID-19, suggesting a link to immune dysregulation .

  • COVID-19: ARRB2 antibodies may modulate ACE2-related inflammatory responses, potentially exacerbating disease severity .

Technical Applications

  • Western Blot: Optimized at 1:500–1:1000 dilution for detecting endogenous ARRB2 .

  • Immunocytochemistry: Localizes ARRB2 to cytoplasmic vesicles in neuronal cells .

Challenges and Future Directions

  • Specificity Issues: Cross-reactivity with ARRB1 remains a concern; rigorous validation using knockout controls is essential .

  • Therapeutic Potential: Targeting ARRB2 signaling pathways may offer novel strategies for cancer and inflammatory diseases .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
ARR2 antibody; ARP5 antibody; At4g16110 antibody; dl4095w antibody; FCAALL.297Two-component response regulator ARR2 antibody; Receiver-like protein 5 antibody
Target Names
ARR2
Uniprot No.

Target Background

Function
ARR2 is a transcriptional activator that specifically binds to the DNA sequence 5'-[AG]GATT-3'. It functions as a response regulator involved in the His-to-Asp phosphorelay signal transduction system. Phosphorylation of the Asp residue in the receiver domain activates the protein, enabling it to promote the transcription of target genes. ARR2 can directly activate certain type-A response regulators in response to cytokinins. It plays a role in the expression of nuclear genes for components of mitochondrial complex I, promotes cytokinin-mediated leaf longevity, and is involved in both the ethylene signaling pathway (in an ETR1-dependent manner) and the cytokinin signaling pathway.
Gene References Into Functions
  1. Cytokinin-facilitated degradation of ARR2 is a crucial post-translational event regulating the intensity of cytokinin signaling. PMID: 22751310
  2. Cytokinin promotes the degradation of ARR2 protein. PMID: 22050482
  3. Research indicates that cytokinins modulate salicylic acid signaling to enhance resistance against Pst, a process in which the interaction between NPR1/TGA3 and ARR2 is essential. PMID: 20708590
  4. The Arabidopsis response regulator 2 (ARR2) has been identified as a signaling component functioning downstream of ETR1 in ethylene signal transduction. PMID: 15282545
  5. Transgenic overexpression of wild-type ARR2, but not an unphosphorylatable mutant ARR2, resulted in delayed leaf senescence. PMID: 16407152

Show More

Hide All

Database Links

KEGG: ath:AT4G16110

STRING: 3702.AT4G16110.1

UniGene: At.21955

Protein Families
ARR family, Type-B subfamily
Subcellular Location
Nucleus.
Tissue Specificity
Detected in the whole plant. Predominantly expressed in pollen.

Q&A

What is ARR2 and what are its key biological functions?

ARR2 has distinct contexts and functions depending on the research system. In plant biology, ARR2 (Arabidopsis Response Regulator 2) is a transcription factor containing a GARP DNA binding domain that plays a crucial role in cytokinin signaling. It specifically binds to promoter regions of target genes like ARR5 to regulate gene expression in response to cytokinin .

In mammalian studies, ARR2 often refers to β-arrestin-2 (Arr2), which functions as an anti-apoptotic factor affecting cell proliferation and signal transduction. In endometrial carcinoma, Arr2 has been shown to up-regulate Toll-like receptor 2 (TLR2) signaling and inhibit apoptosis, promoting resistance to paclitaxel treatment .

When studying ARR2, researchers should clearly specify which protein they are investigating, as the abbreviation can refer to different proteins depending on the research context and model system.

What detection methods are most effective for ARR2 expression studies?

Several complementary immunochemical techniques have proven effective for detecting ARR2 expression:

  • Immunohistochemistry (IHC): Provides spatial information about ARR2 localization within tissues and cells through fixed tissue sections using labeled antibodies. This technique is particularly valuable for determining subcellular localization and expression patterns across different cell types .

  • Western Blotting (Immunoblotting): Detects ARR2 based on molecular weight after separation by SDS-PAGE and transfer to a membrane. Using 10% gels generally achieves good separation of proteins in the 10,000-200,000 Da range, allowing for identification of ARR2 and assessment of antibody specificity .

  • Enzyme-Linked Immunosorbent Assay (ELISA): Enables quantitative measurement of ARR2 levels in tissue homogenates or biological fluids, offering higher throughput than western blotting .

  • Immunofluorescence microscopy: Provides detailed subcellular localization of ARR2 with superior resolution compared to IHC, especially when combined with confocal microscopy.

Each method offers distinct advantages and limitations. For comprehensive characterization, researchers should employ multiple techniques and include appropriate controls to validate findings.

How does ARR2 participate in cellular signaling pathways?

ARR2 plays distinct roles in signaling pathways depending on biological context:

In plant systems (Arabidopsis Response Regulator 2):
ARR2 functions as a key transcription factor in the cytokinin signaling cascade. Upon cytokinin perception, a phosphorelay system activates ARR2 through phosphorylation, enabling it to bind to target gene promoters via its GARP DNA binding domain. This binding occurs specifically at cytokinin response motifs (CRMs) with the consensus sequence 5′-(A/G)GAT(T/C)-3′ and extended cytokinin response motifs (ECRMs) . The interaction occurs primarily through the α3-helix and N-terminal arm of the GARP domain, activating transcription of target genes like ARR5 .

In mammalian systems (β-arrestin-2):
Arr2 functions as a multifaceted signaling protein that initially was characterized for mediating desensitization of G protein-coupled receptors but is now recognized as a scaffold for various signaling pathways. In endometrial carcinoma, Arr2 has been demonstrated to up-regulate TLR2 signaling, subsequently activating NF-κB p56 and promoting the release of inflammatory cytokines including IL-6, IL-8, and TNF-α . This signaling cascade ultimately promotes tumor cell proliferation and inhibits apoptosis, contributing to paclitaxel resistance in cancer cells .

Understanding these distinct signaling mechanisms is essential for developing targeted experimental approaches and interpreting research findings appropriately.

What distinguishes ARR2 from other related proteins?

ARR2 possesses several distinctive features that differentiate it from related proteins:

In plant systems:

  • ARR2 belongs to the B-type ARR family of transcription factors characterized by the GARP DNA binding domain. Unlike A-type ARRs (such as ARR5), which function primarily as cytokinin response genes, B-type ARRs like ARR2 act as transcriptional activators .

  • The GARP domain of ARR2 (GARP2) forms a helix-turn-helix (HTH) motif that specifically recognizes cytokinin response motifs. This DNA binding specificity distinguishes it from other transcription factors .

  • ARR2 demonstrates distinct target gene specificity compared to other B-type ARRs, as evidenced by its differential binding to regions of the ARR5 promoter and response to cytokinin .

In mammalian systems:

  • β-arrestin-2 (Arr2) is one of two β-arrestin isoforms (β-arrestin-1 and β-arrestin-2) that regulate G protein-coupled receptor signaling. While structurally similar, they exhibit different tissue distribution patterns and receptor binding affinities.

  • Arr2 has been specifically implicated in promoting resistance to paclitaxel treatment in endometrial carcinoma through up-regulation of TLR2 signaling and inflammatory cytokine production, a function not shared by β-arrestin-1 .

These structural and functional differences highlight the importance of precise targeting when developing antibodies for specific research applications focused on ARR2.

What experimental controls are essential when working with ARR2 antibodies?

When working with ARR2 antibodies, implementing rigorous controls is critical for ensuring reliable and interpretable results:

  • Specificity Controls:

    • Positive control: Include samples with confirmed ARR2 expression

    • Negative control: Utilize samples from knockout/knockdown models or tissues known not to express ARR2

    • Peptide competition/pre-absorption control: Pre-incubate antibody with purified ARR2 protein/peptide to demonstrate signal elimination

    • Isotype control: Use matched isotype antibodies to detect non-specific binding

  • Technical Controls:

    • Secondary antibody only: Omit primary antibody to assess background from secondary antibody

    • Loading controls: Include housekeeping proteins or total protein staining for Western blots

    • Multiple antibodies: Use antibodies targeting different epitopes of ARR2 to confirm specificity

  • Validation Across Methods:

    • Cross-validate findings using multiple detection techniques (Western blot, IHC, ELISA)

    • Correlate protein expression with mRNA levels

    • Confirm binding specificity with recombinant ARR2 protein

  • Experimental Design Controls:

    • Include dose-response elements when studying ARR2 overexpression or knockdown

    • Establish appropriate timepoints for dynamic processes

    • Use multiple cell lines or tissue types to ensure reproducibility

These controls help distinguish genuine ARR2 signals from artifacts, validate antibody specificity, and ensure experimental reproducibility across different research contexts.

How can researchers accurately measure ARR2 binding affinity to DNA?

Accurate measurement of ARR2 binding affinity to DNA requires sophisticated biomolecular interaction techniques. Based on current research methodologies, the following approaches provide complementary data:

  • Quantitative DNA-Protein Interaction ELISA (qDPI-ELISA): This technique immobilizes DNA fragments containing target sequences (e.g., ARR5 promoter regions) on microtiter plates and measures purified ARR2 GARP domain binding. Detection occurs through specific anti-ARR2 antibodies and enzymatic reactions, allowing high-throughput screening of binding sites and comparative affinity analysis .

  • Fluorescence Correlation Spectroscopy (FCS): This biophysical method measures fluctuations in fluorescence intensity as fluorescently-labeled molecules diffuse through a small observation volume. FCS can determine binding constants (KD) by detecting changes in diffusion times when protein-DNA complexes form, providing insights into binding dynamics in solution .

  • Microscale Thermophoresis (MST): MST detects changes in molecular movement along microscopic temperature gradients upon binding. By titrating fluorescently-labeled ARR5 promoter fragments with increasing concentrations of purified ARR2 protein, researchers can determine dissociation constants (KD) with high precision under native conditions .

  • In planta transactivation assays: To validate binding in biologically relevant contexts, researchers can employ protoplast-based reporter gene assays. Fusing different fragments of target promoters (e.g., ARR5) to luciferase reporters and co-expressing ARR2 allows assessment of functional binding significance under various conditions, including cytokinin treatment .

For comprehensive characterization, employing multiple complementary methods is recommended to overcome limitations of individual techniques and account for variables influencing binding measurements.

What approaches ensure optimal validation of ARR2 antibody specificity?

Rigorous validation of ARR2 antibody specificity requires a multi-faceted approach:

  • Western Blotting Validation Strategy:

    • Test against recombinant ARR2 protein as positive control

    • Include knockout/knockdown samples as negative controls

    • Verify single band at expected molecular weight

    • Demonstrate signal reduction following ARR2 depletion

    • Compare multiple antibodies targeting different epitopes

  • Cross-Platform Validation:

    • Demonstrate consistent results across immunohistochemistry, Western blotting, ELISA, and immunofluorescence

    • Correlate antibody-based detection with orthogonal methods (mRNA expression, mass spectrometry)

    • Verify expected subcellular localization patterns in imaging applications

  • Specificity Confirmation Tests:

    • Peptide competition/pre-absorption assays showing signal elimination when antibody is pre-incubated with antigen

    • Testing for cross-reactivity with related proteins (other ARRs in plants, β-arrestin-1 in mammals)

    • Immunoprecipitation followed by mass spectrometry to confirm target identity

  • Application-Specific Validation:

    • Optimize fixation conditions for IHC/IF to maintain epitope accessibility

    • Test different blocking agents to minimize background

    • Validate across relevant species if cross-reactivity is claimed

    • Confirm specificity under intended experimental conditions

  • Documentation and Transparency:

    • Record complete validation data including positive and negative results

    • Document antibody source, catalog number, lot, dilution, and protocols

    • Share validation data with research community when publishing

This comprehensive validation approach significantly increases confidence in experimental results and facilitates reproducibility across different research groups.

How does ARR2 overexpression affect cellular pathways and what experimental designs best capture these effects?

ARR2 overexpression significantly impacts cellular pathways with distinct experimental approaches required to capture these effects:

For β-arrestin-2 (Arr2) in mammalian systems:
Arr2 overexpression has been demonstrated to:

  • Promote resistance to apoptosis-inducing therapeutic agents

  • Up-regulate TLR2 signaling pathways

  • Increase inflammatory cytokine expression (TNF-α, IL-6, IL-8)

  • Enhance NF-κB p56 activation

  • Promote tumor growth in cancer models

Optimal experimental designs include:

  • In vivo tumor models:

    • Generate stable cell lines overexpressing ARR2 through plasmid transfection

    • Implant these cells in animal models (e.g., nude mice) to form tumors

    • Compare tumor growth dynamics between ARR2-overexpressing and control tumors

    • Measure response to therapeutic agents across groups

    • This approach provides physiologically relevant context for ARR2's impact on tumor progression

  • Dose-response experimental design:

    • Subject ARR2-overexpressing and control cells/tumors to increasing doses of therapeutic agents

    • Measure endpoints including tumor volume, weight, and necrosis index

    • Calculate treatment/control (T/C) ratios to quantify resistance

    • In a recent study, tumors overexpressing Arr2 showed T/C values of 64.86% and 54.06% with 10mg/kg and 20mg/kg paclitaxel respectively, versus 43.14% and 33.38% in control groups

  • Pathway analysis integration:

    • Examine multiple pathway components simultaneously (e.g., TLR2, NF-κB, inflammatory cytokines)

    • Utilize Western blot for protein expression and real-time PCR for mRNA levels

    • Compare signaling activation patterns between ARR2-overexpressing and control samples

    • This approach elucidates the molecular mechanisms underlying observed phenotypes

These experimental designs collectively provide robust evidence of ARR2's effects on cellular pathways and potential mechanisms for therapeutic targeting.

What methodological considerations are critical when studying ARR2 across different model systems?

When investigating ARR2 across different model systems, several critical methodological considerations must be addressed:

  • Protein Expression and Purification Strategies:

    • For plant ARR2 studies, expression of the GARP domain versus full-length protein affects binding properties

    • Expression system selection (bacterial, insect, mammalian) impacts folding and post-translational modifications

    • Purification methods must maintain protein stability and native conformation

    • Consider using affinity tags positioned to avoid interference with ARR2 function

  • Experimental Condition Standardization:

    • Buffer composition significantly affects ARR2-DNA binding interactions

    • Temperature, pH, and salt concentration require optimization and standardization

    • For plant studies, cytokinin treatment conditions (concentration, duration, type) must be carefully controlled

    • For β-arrestin-2 research, cell confluence and passage number affect expression and signaling pathway activity

  • Model System Selection Rationale:

    • Plant studies: Consider Arabidopsis protoplasts for transactivation assays versus whole plants for physiological relevance

    • Mammalian studies: Cell line selection affects baseline expression of ARR2 and interacting partners

    • Animal models: Immunocompetent versus immunocompromised models influence tumor microenvironment factors

  • Control Selection Strategy:

    • Include negative controls with mutated binding sites for DNA-binding studies

    • Use multiple positive controls with established behavior

    • For overexpression studies, include both empty vector controls and dose-dependent expression models

  • Detection Method Adaptation:

    • Validate antibodies specifically for each model system

    • Address species-specific differences in epitope recognition

    • Optimize detection protocols for tissue-specific or species-specific variables

    • Establish detection sensitivity thresholds for each system

By systematically addressing these considerations, researchers can develop robust experimental designs that generate reliable and comparable data across different model systems, enhancing the translational value of ARR2 research.

How can computational modeling enhance understanding of ARR2-DNA interactions?

Computational modeling provides valuable insights into ARR2-DNA interactions that complement experimental approaches:

  • Homology-Based Structural Modeling Applications:

    • Leverages known structures of related proteins to predict ARR2 structure

    • Has identified that the GARP domain of ARR2 contains a helix-turn-helix (HTH) motif crucial for DNA binding

    • Revealed the α3-helix and N-terminal arm as key interaction regions

    • Guides experimental design for site-directed mutagenesis studies

  • Protein-DNA Docking Simulation Benefits:

    • Enables virtual screening of ARR2 GARP domain binding to various DNA sequences

    • Predicts binding energy and specific nucleotide-amino acid interactions

    • Identifies key residues involved in DNA recognition that can be experimentally validated

    • Helps prioritize mutations for functional testing

  • Integrated Structure-Function Analysis:

    • Correlates structural predictions with functional assay results

    • Research has demonstrated that mutations in the α3-helix and N-terminal arm of the GARP domain hinder ARR2's ability to activate transcription

    • These findings validate structural models predicting these regions as critical for DNA interaction

  • Comparative Analysis Applications:

    • Comparing the ARR2 GARP domain with other B-type ARRs reveals structural differences explaining functional specificity

    • Identifies conserved and variable regions that influence binding preferences to different promoter sequences

    • Informs the design of specific antibodies targeting unique regions

A successful application of this approach is demonstrated in recent research where a structural 3D model of the GARP2 protein-DNA complex was created and validated through transactivation assays. This integrated approach identified specific amino acids in the α3-helix and N-terminal arm as critical for protein-DNA interaction, directly linking structural features to functional outcomes .

What technical challenges exist in developing highly specific ARR2 antibodies?

Developing highly specific ARR2 antibodies presents several significant technical challenges:

  • Epitope Selection Complexities:

    • ARR2 shares sequence homology with other B-type ARRs in plants, complicating unique epitope identification

    • In mammalian systems, β-arrestin-2 shares approximately 78% amino acid identity with β-arrestin-1

    • Structural accessibility constraints: Selecting epitopes that remain accessible in native protein conformations

    • Functional domain considerations: Avoiding epitopes in key functional regions that might be masked by protein interactions

  • Application-Specific Performance Variability:

    • Antibodies successful in Western blotting may fail in immunohistochemistry due to fixation-induced epitope alterations

    • Native versus denatured protein recognition differences require different antibody characteristics

    • Cross-species reactivity varies based on epitope conservation

    • Quantitative applications demand different performance parameters than qualitative detection

  • Reproducibility Challenges:

    • Lot-to-lot variability in polyclonal antibody production affects consistency

    • Hybridoma stability issues for monoclonal antibodies over time

    • Storage and handling conditions significantly impact antibody performance

    • Standardization difficulties across different research groups and applications

  • Technical Detection Limitations:

    • Post-translational modifications can mask or alter epitope recognition

    • Protein-protein interactions may block antibody binding sites

    • Variable expression levels require different antibody sensitivities

    • Background signal in complex tissues reduces specificity

  • Validation Burden Across Applications:

    • Each application (Western blot, IHC, ELISA, IP, ChIP) requires separate validation

    • Tissue-specific background patterns necessitate application-specific optimization

    • Fixation protocols affect epitope preservation differently across applications

Addressing these challenges requires comprehensive validation strategies, the use of multiple antibodies targeting different epitopes, detailed documentation of validation results, and continuing refinement of antibody development techniques.

What systematic approach resolves inconsistent Western blot results with ARR2 antibodies?

Resolving inconsistent Western blot results with ARR2 antibodies requires a systematic troubleshooting approach:

  • Sample Preparation Optimization:

    • Evaluate different lysis buffers to optimize protein extraction and epitope preservation

    • Implement stringent protocols to prevent protein degradation (fresh samples, protease inhibitors)

    • Standardize protein denaturation conditions (temperature, duration, reducing agents)

    • Ensure consistent protein quantification methodology across experiments

  • Gel Electrophoresis Parameter Adjustment:

    • Select appropriate gel percentage (10% gels typically provide optimal separation for proteins between 10-200 kDa)

    • Consider using commercial pre-cast gels to eliminate variability in gel preparation

    • Standardize running conditions (voltage, time, buffer composition)

    • Optimize protein transfer parameters based on ARR2 molecular weight

  • Antibody Condition Refinement:

    • Perform systematic antibody titration to determine optimal working concentration

    • Test different incubation conditions (temperature, duration, buffer composition)

    • Evaluate multiple blocking agents (BSA, milk, commercial blockers) for signal-to-noise optimization

    • Validate antibody lot consistency when receiving new shipments

  • Detection System Optimization:

    • Match detection system sensitivity to expected ARR2 expression level

    • Standardize exposure times or implement objective exposure determination

    • Compare different detection modalities (chemiluminescence, fluorescence)

    • Use positive controls to calibrate detection parameters

  • Systematic Control Implementation:

    • Include molecular weight markers positioned adjacent to samples

    • Run positive control samples with known ARR2 expression

    • Include negative controls (null or knockdown samples)

    • Use purified recombinant ARR2 as reference standard

Troubleshooting Decision Matrix:
For no signal detection:

  • Verify primary/secondary antibody compatibility and activity

  • Confirm successful protein transfer with reversible membrane staining

  • Test higher antibody concentration or extended incubation

  • Evaluate alternative epitope antibodies

For non-specific bands:

  • Increase blocking stringency and washing steps

  • Implement higher antibody dilution

  • Add detergents to reduce non-specific binding

  • Perform antibody pre-absorption with non-specific proteins

For variable results between experiments:

  • Create standardized protocols with detailed documentation

  • Prepare fresh working solutions for each experiment

  • Implement total protein normalization rather than single housekeeping proteins

  • Control for environmental variables (temperature, incubation times)

This systematic approach enables researchers to identify and address specific factors contributing to inconsistent Western blot results with ARR2 antibodies.

What advanced techniques enhance ARR2 protein-protein interaction studies?

Cutting-edge techniques for studying ARR2 protein-protein interactions combine traditional approaches with innovative technologies to provide comprehensive insights:

  • Proximity-Based Labeling Methods:

    • BioID: Fusion of ARR2 with a biotin ligase (BirA*) that biotinylates proteins within approximately 10nm radius

    • APEX2: ARR2-APEX2 fusion that catalyzes biotinylation of proximal proteins upon H₂O₂ exposure

    • TurboID: Enhanced biotin ligase variant offering faster labeling kinetics

    • These approaches capture transient and weak interactions in living cells, identifying the proximal proteome surrounding ARR2

  • Advanced Microscopy Applications:

    • Förster Resonance Energy Transfer (FRET): Measures protein proximity (1-10nm) using fluorescently tagged ARR2 and potential partners

    • Fluorescence Lifetime Imaging Microscopy (FLIM): Provides FRET measurements with higher sensitivity and reduced artifacts

    • Single-molecule tracking: Monitors ARR2 dynamics and co-localization with other proteins in real-time

    • Super-resolution microscopy: Techniques like STORM or PALM deliver nanometer-scale resolution of protein co-localization

  • Quantitative Interaction Proteomics:

    • SILAC (Stable Isotope Labeling with Amino acids in Cell culture): Differentiates specific from non-specific interactions through isotope labeling

    • TMT (Tandem Mass Tag) labeling: Enables multiplexed comparison of interaction partners across experimental conditions

    • Parallel Reaction Monitoring (PRM): Provides targeted proteomics for validating specific interactions with high sensitivity

  • Protein Complementation Assays:

    • NanoBiT: Split luciferase system with exceptional sensitivity for detecting protein-protein interactions

    • Split GFP: Fluorescence-based detection suitable for visualization of interaction location

    • These systems can be adapted for high-throughput screening of ARR2 interaction partners

  • Integrated Computational-Experimental Approaches:

    • Molecular docking simulations to predict potential interaction interfaces

    • Machine learning algorithms to identify potential interaction partners

    • Network analysis to map ARR2 within broader signaling pathways

    • Experimental validation of computationally predicted interactions

The integration of multiple complementary techniques provides a more complete understanding of ARR2's interaction network and its functional significance in different biological contexts, overcoming limitations inherent to individual methods.

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.