ARP2 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
ARP2 antibody; WRM antibody; At3g27000 antibody; MOJ10.7 antibody; Actin-related protein 2 antibody; Protein WURM antibody
Target Names
ARP2
Uniprot No.

Target Background

Function
ARP2 antibody functions as the ATP-binding component of the Arp2/3 complex. This complex is involved in regulating actin polymerization and, in conjunction with an activating nucleation-promoting factor (NPF), mediates the formation of branched actin networks. ARP2 appears to contact the pointed end of the daughter actin filament. The Arp2/3 complex plays a critical role in controlling cell morphogenesis through modulating cell polarity development. It is involved in regulating cell morphogenesis in leaf epidermal pavement cells, root hairs, hypocotyls epidermal cells, and trichomes, particularly during rapid cell expansion. ARP2 regulates the directionality of cell expansion by controlling actin organization, thereby influencing microtubule distribution and the fusion of small vacuoles.
Gene References Into Functions
  1. The Arp2/3 complex regulates mitochondrial-dependent Ca(2+) signaling in response to salt stress. Arp2 is required for mitochondrial distribution. PMID: 24280386
  2. Research indicates that regulating actin reassembly through ARP2/3 complex activity is crucial for stomatal regulation. PMID: 22570440
  3. The rapid growth of roots in the light requires a functional ARP2/3-SCAR complex. PMID: 21972261
  4. Differences among SCARs in mRNA levels and the biochemical efficiency of ARP2/3 activation may explain the functional contributions of individual genes PMID: 18424615
Database Links

KEGG: ath:AT3G27000

STRING: 3702.AT3G27000.1

UniGene: At.37122

Protein Families
Actin family, ARP2 subfamily
Subcellular Location
Cytoplasm, cytoskeleton.
Tissue Specificity
Expressed at low levels in roots, seedlings, stems, leaves, flowers, pollen, siliques and at a higher level in inflorescences. Specifically localized in cells adjacent to mature xylem and in developing xylem vessels.

Q&A

What is ARP2 and what cellular functions does it mediate?

ARP2 (Actin-Related Protein 2) is a key component of the ARP2/3 complex that facilitates nucleation of new actin filaments and promotes formation of branched actin networks. This protein plays crucial roles in numerous cellular functions including chemotaxis, cell migration, cell adhesion, and platelet activation . ARP2 contributes to maintaining cellular structure and enables dynamic changes in cell shape and movement. The protein has a calculated molecular weight of 45 kDa, with observed molecular weights of 43-45 kDa in experimental settings . The ARP2/3 complex binds to sides of existing actin filaments or pointed ends, catalyzing the growth of new filaments in the barbed direction. This process is essential for the formation of cellular structures such as lamellipodia and filopodia .

What are the differences between monoclonal and polyclonal ARP2 antibodies?

Monoclonal ARP2 antibodies, such as the E-12 antibody (mouse monoclonal IgG1 kappa light chain), recognize specific epitopes on the ARP2 protein and provide consistent lot-to-lot reproducibility . These antibodies offer high specificity for a single epitope but may be more susceptible to epitope masking during sample preparation. In contrast, polyclonal ARP2 antibodies like the rabbit polyclonal antibody (10922-1-AP) recognize multiple epitopes on the target protein, providing stronger signal through multiple binding sites but potentially with increased background . Polyclonal antibodies are often generated using ARP2 fusion proteins as immunogens and undergo antigen affinity purification to improve specificity . The choice between these antibody types depends on the experimental requirements for specificity, sensitivity, and application compatibility.

Which experimental applications are most validated for ARP2 antibodies?

ARP2 antibodies have been extensively validated across multiple applications, with varying degrees of published validation. The most commonly validated applications include:

ApplicationValidation LevelTypical Dilution RangeCitations
Western Blot (WB)Highly validated1:1000-1:400013 publications
Immunofluorescence (IF/ICC)Well validated1:50-1:5003 publications
Immunohistochemistry (IHC)Validated1:200-1:8001 publication
KD/KO experimentsValidatedExperiment-dependent2 publications
Flow Cytometry (FC)Validated0.40 μg per 10^6 cellsLimited publications

Western blot applications are the most robustly validated, with positive detection in multiple cell lines including HeLa, Jurkat, MCF-7, and SH-SY5Y cells, as well as in mouse kidney tissue . Immunohistochemistry applications have been validated with human prostate cancer tissue using various antigen retrieval methods . For immunofluorescence, positive detection has been confirmed in U-251 cells, while flow cytometry applications have been validated with HEK-293 cells .

How should I optimize ARP2 antibody dilutions for different applications?

Optimizing ARP2 antibody dilutions requires systematic titration specific to each application. For Western blot analysis, the recommended starting range is 1:1000-1:4000, with many researchers finding optimal results at 1:2000 dilution . When performing immunohistochemistry, begin with a 1:200-1:800 dilution range, noting that antigen retrieval methods significantly impact signal intensity . For immunofluorescence and immunocytochemistry applications, a wider range of 1:50-1:500 is recommended as a starting point, with lower dilutions often required compared to WB applications . Flow cytometry applications typically require precise antibody amounts, with a starting recommendation of 0.40 μg per 10^6 cells in a 100 μl suspension . For any application, perform preliminary experiments with a dilution series to determine the optimal antibody concentration that maximizes specific signal while minimizing background. Sample-dependent variables including protein expression level, fixation method, and detection system sensitivity will necessitate experiment-specific optimization.

What positive controls should I include when validating a new ARP2 antibody?

Including appropriate positive controls is essential when validating a new ARP2 antibody. Based on validated samples from the literature, the following positive controls are recommended:

ApplicationRecommended Positive ControlsNotes
Western BlotHeLa cells, Jurkat cells, MCF-7 cellsThese cell lines consistently express detectable levels of ARP2
IHCHuman prostate cancer tissueRequires appropriate antigen retrieval (TE buffer pH 9.0 or citrate buffer pH 6.0)
IF/ICCU-251 cellsShows reliable subcellular localization pattern
Flow CytometryHEK-293 cellsDemonstrated consistent intracellular staining

For knockout/knockdown validation, consider using CRISPR/Cas9-mediated ARP2 knockout cells or siRNA-mediated knockdown samples as negative controls to conclusively demonstrate antibody specificity. When possible, use recombinant ARP2 protein as a positive control for Western blot to confirm the antibody detects the correct molecular weight (43-45 kDa observed, 45 kDa calculated) . For tissue samples, mouse kidney tissue has been validated as a positive control for ARP2 detection .

What are the key considerations for sample preparation when using ARP2 antibodies?

Sample preparation significantly impacts ARP2 antibody performance across different applications. For Western blot, complete protein denaturation is crucial, requiring standard SDS-PAGE sample preparation with β-mercaptoethanol and heating to 95°C for 5 minutes. For immunohistochemistry applications, antigen retrieval methodology is critical, with two validated options: TE buffer (pH 9.0) or citrate buffer (pH 6.0) . The higher pH TE buffer is suggested as the primary method, with citrate buffer as an alternative when signal is suboptimal . For immunofluorescence, cellular fixation method impacts epitope availability—paraformaldehyde (4%) fixation for 15 minutes at room temperature typically preserves ARP2 antigenicity while maintaining cellular architecture. When preparing samples for flow cytometry, effective permeabilization is essential as ARP2 is an intracellular protein, requiring detergents like 0.1% Triton X-100 or saponin-based permeabilization solutions. For all applications, using freshly prepared samples improves detection sensitivity, as prolonged storage may reduce antigen availability through protein degradation or epitope masking.

Why might I observe multiple bands when using ARP2 antibodies in Western blot?

Multiple bands in ARP2 Western blots can occur for several methodologically relevant reasons. The primary ARP2 band should appear at 43-45 kDa, which corresponds to the expected molecular weight of the protein . Additional bands may represent:

  • Post-translational modifications: ARP2 undergoes phosphorylation and potentially other modifications that can alter migration patterns.

  • Protein degradation: Incomplete protease inhibition during sample preparation may result in ARP2 degradation products appearing as lower molecular weight bands.

  • Cross-reactivity: Some antibodies may exhibit cross-reactivity with structurally similar proteins, particularly other actin-related proteins in the same family.

  • Alternative splice variants: Though not extensively documented for ARP2, splice variants could potentially manifest as bands of different molecular weights.

To address these issues, implement methodological improvements including: using fresh protease inhibitors during sample preparation; testing different sample preparation buffers; validating results with an alternative ARP2 antibody that recognizes a different epitope; and including knockout/knockdown controls to confirm band specificity . Additionally, consider using reducing agents like DTT instead of β-mercaptoethanol if disulfide bond reduction efficiency is suspected to be an issue.

How can I reduce background staining when using ARP2 antibodies in immunohistochemistry?

Reducing background staining in ARP2 immunohistochemistry requires systematic optimization of multiple parameters. First, optimize antibody dilution by testing a wider range (1:100-1:1000) to determine the concentration that maximizes signal-to-noise ratio . Second, implement effective blocking procedures using 5-10% normal serum from the same species as the secondary antibody for 1-2 hours at room temperature. Third, modify antigen retrieval procedures, comparing TE buffer (pH 9.0) with citrate buffer (pH 6.0) to determine which provides optimal epitope accessibility with minimal non-specific binding . Fourth, incorporate additional blocking steps for endogenous peroxidase (3% H₂O₂ for 10 minutes) and biotin (avidin/biotin blocking kit) if using peroxidase or biotin-based detection systems. Fifth, optimize incubation conditions by testing both overnight incubation at 4°C versus 1-2 hours at room temperature to determine which produces cleaner results. Finally, include additional washing steps (5-6 washes of 5 minutes each) with PBS containing 0.1% Tween-20 to remove unbound antibody more effectively. Implementing multiple controls, including a no-primary-antibody control and isotype control, will help distinguish specific from non-specific staining patterns.

What strategies can help resolve inconsistent results between different ARP2 antibody applications?

Inconsistent results across applications require methodological troubleshooting addressing application-specific variables. First, verify antibody compatibility with each application, as antibodies may perform well in Western blot but poorly in IHC or IF due to epitope accessibility differences in native versus denatured states . Second, optimize fixation and sample preparation methods for each application independently—protocols that work for one technique may not be optimal for others. Third, validate antibody performance using known positive and negative controls for each specific application . Fourth, compare results using multiple ARP2 antibodies targeting different epitopes to confirm findings, as some epitopes may be masked in certain applications. Fifth, ensure appropriate secondary antibody selection with minimal cross-reactivity to the experimental system. Sixth, standardize protein extraction methods for Western blot applications, and implement consistent antigen retrieval methods for IHC applications . Finally, maintain detailed records of lot numbers, as antibody performance can vary between production batches. If inconsistencies persist, consider application-specific antibodies that have been extensively validated for your particular experimental approach .

How can ARP2 antibodies be used to study actin cytoskeleton dynamics?

ARP2 antibodies enable sophisticated analysis of actin cytoskeleton dynamics through multiple methodological approaches. For live cell imaging, fluorescently conjugated ARP2 antibodies can be microinjected or introduced via cell-penetrating peptides to visualize dynamic ARP2/3 complex localization during cellular processes like migration and membrane ruffling . For fixed cell analysis, co-localization studies combining ARP2 antibodies with phalloidin staining of F-actin filaments can identify active sites of actin branching and nucleation at high resolution . When studying the temporal dynamics of ARP2 recruitment, researchers can implement a synchronized stimulation approach (e.g., with growth factors or chemotactic agents) followed by fixation at defined time points and immunofluorescence analysis. Super-resolution microscopy techniques like STORM or PALM combined with ARP2 antibodies can resolve the nanoscale organization of branched actin networks beyond the diffraction limit. To study ARP2 functionality in specific cellular compartments, combining ARP2 immunofluorescence with compartment-specific markers enables quantification of ARP2 recruitment to structures such as lamellipodia, filopodia, and endocytic vesicles . Additionally, FRAP (Fluorescence Recovery After Photobleaching) experiments using fluorescently-tagged ARP2 antibody fragments can measure the turnover rates of the ARP2/3 complex at specific cellular structures.

What co-immunoprecipitation strategies work best with ARP2 antibodies?

Optimizing co-immunoprecipitation (co-IP) with ARP2 antibodies requires careful consideration of several methodological factors to preserve protein-protein interactions while maintaining specificity. The most effective lysis buffers contain mild, non-denaturing detergents (0.5-1% NP-40 or 0.5% Triton X-100) with physiological salt concentrations (150mM NaCl) to preserve the integrity of the ARP2/3 complex and its associated proteins . Pre-clearing the lysate with appropriate control IgG and protein A/G beads for 1 hour at 4°C significantly reduces non-specific binding. When selecting antibodies for pull-down, those validated for immunoprecipitation applications show superior performance, with rabbit polyclonal antibodies often yielding better results due to their recognition of multiple epitopes . Key methodological improvements include:

  • Using reversible cross-linking agents like DSP (dithiobis(succinimidyl propionate)) to stabilize transient protein interactions

  • Maintaining samples at 4°C throughout the procedure to prevent complex dissociation

  • Including phosphatase inhibitors (sodium fluoride, sodium orthovanadate) and protease inhibitors in all buffers

  • Performing sequential co-IP to identify components of multi-protein complexes

  • Validating results with reciprocal co-IP experiments using antibodies against known ARP2 interacting partners

When analyzing co-IP results, mass spectrometry provides comprehensive identification of ARP2-associated proteins, while Western blotting with specific antibodies confirms individual interactions with proteins like ARPC1-5, WASP family proteins, and cortactin .

How can ARP2 antibodies be applied in super-resolution microscopy studies?

Applying ARP2 antibodies in super-resolution microscopy requires specific methodological adaptations to achieve optimal resolution and signal quality. For STORM (Stochastic Optical Reconstruction Microscopy) applications, directly conjugated ARP2 antibodies with photoswitchable fluorophores like Alexa Fluor 647 provide superior localization precision . Sample preparation requires careful optimization with aldehyde-based fixatives (3% paraformaldehyde with 0.1% glutaraldehyde) to preserve nanoscale architecture while maintaining epitope accessibility. For multi-color STORM imaging of the ARP2/3 complex and associated proteins, sequential labeling with activator-reporter dye pairs minimizes chromatic aberration effects. When implementing STED (Stimulated Emission Depletion) microscopy, ARP2 antibodies conjugated to dyes with appropriate depletion characteristics (ATTO647N, ATTO594) yield optimal results, with concentration optimization critical to achieve single-molecule detection density. For DNA-PAINT approaches, DNA-conjugated secondary antibodies targeting ARP2 primary antibodies enable extended imaging with minimal photobleaching. Key methodological considerations include:

  • Using small (Fab fragments) or nanobody-based detection systems to minimize the distance between fluorophore and target

  • Implementing drift correction using fiducial markers (gold nanoparticles)

  • Optimizing labeling density to achieve Nyquist sampling criteria while avoiding overcrowding

  • Employing two-step immunolabeling with primary ARP2 antibodies followed by secondary antibodies with appropriate fluorophores

  • Validating super-resolution findings with complementary techniques like electron microscopy

These approaches have revealed the precise organization of ARP2/3 complex at branching points within actin networks and its co-localization with regulatory proteins at nanometer resolution .

How should I quantify ARP2 protein levels in Western blot experiments?

Quantifying ARP2 protein levels in Western blot experiments requires rigorous methodological approaches to ensure accuracy and reproducibility. Densitometric analysis should be performed using specialized software (ImageJ, Image Studio, etc.) with the following protocol: First, normalize ARP2 band intensity to appropriate loading controls—β-actin is not recommended due to potential functional relationships with ARP2; instead, use GAPDH, tubulin, or total protein stains like Ponceau S . Second, implement a standard curve using recombinant ARP2 protein at known concentrations (typically 10-100 ng range) to establish detection linearity. Third, ensure sample loading falls within the linear detection range by testing multiple loading amounts (typically 10-50 μg total protein). Fourth, perform technical replicates (minimum of three) and biological replicates (minimum of three independent experiments) to establish statistical significance. Fifth, when comparing ARP2 levels between experimental conditions, analyze samples on the same blot whenever possible to eliminate inter-blot variability. For statistical analysis, implement appropriate tests based on data distribution (parametric vs. non-parametric) and experimental design (t-test for two groups, ANOVA for multiple groups). Report results as fold-change relative to control conditions with error bars representing standard deviation or standard error of the mean, and p-values to indicate statistical significance.

What approaches are most effective for analyzing ARP2 localization in cellular compartments?

Analyzing ARP2 subcellular localization requires quantitative image analysis methodologies that can objectively measure spatial distribution patterns. For co-localization studies, calculate Pearson's correlation coefficient and Mander's overlap coefficient between ARP2 and markers for specific cellular structures (e.g., phalloidin for F-actin, cortactin for lamellipodia) . When measuring ARP2 enrichment at specific cellular structures, implement line scan analysis across features of interest (e.g., leading edge to cell body) to generate intensity profiles that quantify relative distribution. For temporal analyses of ARP2 recruitment, establish baseline fluorescence intensity in resting cells, then measure fold-change in intensity at regions of interest following stimulation at defined time points. To analyze ARP2 distribution in 3D cellular environments, employ z-stack acquisition followed by volume rendering and 3D intensity mapping. For population-level analysis, perform automated high-content imaging of hundreds of cells followed by machine learning-based classification of ARP2 localization patterns. Statistical analysis should include:

Analysis TypeRecommended Statistical ApproachSample Size Guidelines
Co-localizationPearson's r with Fisher's z transformationMinimum 20-30 cells per condition
Enrichment ratioPaired t-test or Wilcoxon signed-rank testMinimum 30 cells per condition
Temporal dynamicsRepeated measures ANOVA with post-hoc tests10-15 cells across 5-7 time points
Pattern classificationChi-square test for distribution differences100+ cells per condition

These quantitative approaches provide objective metrics for ARP2 localization that can be statistically evaluated across experimental conditions .

How can I distinguish between specific ARP2 signal and non-specific background?

Distinguishing specific ARP2 signal from non-specific background requires implementing rigorous experimental controls and quantitative analysis methods. First, include biological negative controls such as ARP2 knockdown or knockout samples prepared and processed identically to experimental samples . Second, implement technical negative controls including secondary antibody-only controls and isotype controls matching the primary antibody host species and isotype. Third, conduct peptide competition assays where the antibody is pre-incubated with excess immunizing peptide before application to samples—specific signal should be substantially reduced while non-specific binding remains unchanged. Fourth, compare staining patterns from multiple ARP2 antibodies targeting different epitopes—convergent patterns strongly suggest specific detection. Fifth, validate subcellular localization patterns against published literature describing ARP2 distribution in similar cell types or tissues . Sixth, analyze signal distribution quantitatively—specific ARP2 staining typically shows defined subcellular localization (particularly at sites of actin polymerization like lamellipodia and membrane ruffles) while nonspecific background presents as diffuse cytoplasmic staining or non-biological patterns . Finally, establish signal-to-noise ratios by measuring intensity in regions with expected positive signal versus regions expected to lack ARP2, with ratios below 3:1 suggesting predominant non-specific binding.

How can ARP2 antibodies be utilized to study disease mechanisms?

ARP2 antibodies offer powerful methodological approaches for investigating disease mechanisms across multiple pathological contexts. In cancer research, multiplex immunofluorescence combining ARP2 antibodies with markers of invasiveness can quantitatively assess correlations between ARP2/3 complex activity and metastatic potential across tumor types and stages . For neurodegenerative diseases, brain tissue immunohistochemistry with ARP2 antibodies can evaluate cytoskeletal abnormalities in affected regions, with quantitative analysis of branched actin networks in dendritic spines . In cardiovascular pathologies, ARP2 immunostaining of atherosclerotic plaques can assess macrophage migration and foam cell formation mechanisms. For immunological disorders, flow cytometric analysis using ARP2 antibodies can quantify defects in immune cell migration and phagocytosis . Methodologically, these disease studies benefit from combining traditional antibody applications with emerging techniques:

  • Tissue microarray analysis with ARP2 antibodies to screen large patient cohorts

  • Microfluidic migration assays with real-time ARP2 imaging to assess cell motility defects

  • Patient-derived organoid immunostaining to evaluate ARP2 function in 3D tissue models

  • Single-cell proteomics to correlate ARP2 levels with disease-relevant phenotypes

  • CRISPR-mediated genome editing combined with rescue experiments using ARP2 variants identified in patient populations

These approaches leverage ARP2 antibodies to connect cytoskeletal abnormalities with specific disease mechanisms, potentially identifying novel therapeutic targets targeting actin cytoskeletal regulation .

What emerging technologies might enhance ARP2 antibody applications?

Emerging technologies are poised to revolutionize ARP2 antibody applications with enhanced resolution, sensitivity, and throughput. Expansion microscopy combined with ARP2 immunofluorescence enables physical enlargement of samples, providing super-resolution imaging on conventional microscopes while preserving relative protein localization . For dynamic visualization, engineered ARP2 antibody fragments conjugated to cell-permeable peptides allow live-cell imaging of endogenous ARP2 without genetic manipulation. Proximity labeling approaches using ARP2 antibodies conjugated to enzymes like APEX2 or TurboID enable spatially-resolved proteomic mapping of the ARP2 interactome in specific cellular compartments. Microfluidic antibody delivery systems provide precise temporal control of ARP2 inhibition in living cells through controlled introduction of function-blocking antibodies. For high-throughput screening applications, automated microwell platforms combining ARP2 immunofluorescence with machine learning analysis enable rapid phenotypic screening of cytoskeletal modulators across thousands of conditions. Digital spatial profiling combining ARP2 antibodies with barcoded detection systems allows multiplexed analysis of cytoskeletal regulation in the context of complex tissues. Looking forward, nascent technologies including:

  • Quantum dot-conjugated ARP2 antibodies for extended live-cell imaging

  • DNA-origami scaffolds presenting multiple ARP2 antibodies at defined nanoscale distances

  • Antibody-mediated targeted degradation of ARP2 through PROTAC conjugation

  • Cryo-electron tomography with immunogold-labeled ARP2 antibodies for structural analysis

  • Single-molecule tracking of ARP2 dynamics using photoswitchable antibody conjugates

These technologies will continue expanding the utility of ARP2 antibodies in research applications requiring increased sensitivity, resolution, and specificity .

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.