ABIL1 Antibody

Shipped with Ice Packs
In Stock

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
ABIL1 antibody; At2g46225 antibody; T3F17Protein ABIL1 antibody; Abl interactor-like protein 1 antibody; AtABIL1 antibody
Target Names
ABIL1
Uniprot No.

Target Background

Function
ABIL1 Antibody is involved in the regulation of actin and microtubule organization. It is part of a WAVE complex that activates the Arp2/3 complex.
Database Links

KEGG: ath:AT2G46225

UniGene: At.28094

Protein Families
ABI family
Subcellular Location
Cytoplasm, cytoskeleton.
Tissue Specificity
Expressed in seedlings, roots, hypocotyls, cotyledons, leaves, stems, and flowers.

Q&A

What is ABI1 and what are its primary biological functions?

ABI1 (Abl-Interactor 1) is a multifunctional adaptor protein that plays critical roles in various cellular processes. It acts in the negative regulation of cell growth and transformation by interacting with nonreceptor tyrosine kinases ABL1 and/or ABL2. ABI1 is involved in cytoskeletal reorganization and EGFR signaling, participating in the transduction of signals from Ras to Rac through formation of a trimeric complex with EPS8 and SOS1 that exhibits Rac-specific guanine nucleotide exchange factor (GEF) activity. This protein regulates ABL1/c-Abl-mediated phosphorylation of ENAH and recruits WASF1 to lamellipodia, where it appears to regulate WASF1 protein levels. In neural tissues, ABI1 regulates dendritic outgrowth and branching, influencing the shape and number of synaptic contacts in developing neurons .

What types of ABI1 antibodies are available for research applications?

Researchers can access various ABI1 antibodies with different specificities and applications. Most commercially available ABI1 antibodies are rabbit polyclonal antibodies, though they differ in their binding specificity to different regions of the ABI1 protein. These include antibodies targeting the N-terminal region (amino acids 1-200, 81-108, 87-117, or 151-175), the central domain (amino acids 180-230 or 300-400), and the C-terminal region (amino acids 483-494). Some antibodies recognize full-length recombinant ABI1 protein. These antibodies are primarily available in unconjugated forms, though conjugated versions may be available for specialized applications .

What are the typical applications for ABI1 antibodies in research?

ABI1 antibodies support multiple experimental approaches in cellular and molecular biology research. Common applications include Western blotting (WB) for protein detection and quantification, immunohistochemistry on paraffin-embedded sections (IHC-P) for visualization of ABI1 in tissue contexts, immunoprecipitation (IP) for protein complex isolation, immunocytochemistry (ICC) for cellular localization studies, immunofluorescence (IF) for high-resolution localization, and flow cytometry (FACS) for quantitative analysis in cell populations. These antibodies have demonstrated reactivity with human, mouse, rat, and pig samples, making them versatile tools for comparative studies across species .

How should I select the appropriate ABI1 antibody for my specific research question?

Selection of the appropriate ABI1 antibody requires careful consideration of several factors. First, determine which region of ABI1 is most relevant to your research question—N-terminal antibodies may be preferable for studying interactions with N-WASP, while C-terminal antibodies might be better for examining interactions with ABL kinases. Second, ensure compatibility with your experimental system by checking species reactivity (human, mouse, rat). Third, verify that the antibody has been validated for your intended application (WB, IHC-P, IP, etc.). Fourth, consider whether you need to detect specific isoforms or post-translational modifications of ABI1, which may require specialized antibodies. Finally, review literature using these antibodies to assess their performance in contexts similar to your planned experiments .

What are the optimal conditions for Western blotting with ABI1 antibodies?

For optimal Western blotting with ABI1 antibodies, several technical parameters require careful optimization. Sample preparation should include efficient cell lysis with a buffer containing appropriate protease inhibitors to prevent ABI1 degradation. Protein separation is typically achieved on 8-12% SDS-PAGE gels, with 10% being most common for the ~65 kDa ABI1 protein. Transfer to nitrocellulose or PVDF membranes should be performed at 100V for 60-90 minutes in standard transfer buffer with 20% methanol. Blocking with 5% non-fat milk or BSA in TBS-T for 1 hour at room temperature is recommended. Primary ABI1 antibody should be diluted according to manufacturer specifications (typically 1:500 to 1:2000) and incubated overnight at 4°C. Following thorough washing, appropriate HRP-conjugated secondary antibodies should be applied at 1:5000 to 1:10000 dilution for 1 hour at room temperature. Signal detection using enhanced chemiluminescence should reveal specific bands corresponding to ABI1 without significant background .

How can I optimize co-immunoprecipitation protocols using ABI1 antibodies?

Optimizing co-immunoprecipitation (co-IP) protocols with ABI1 antibodies requires attention to preserving protein-protein interactions. Use gentle cell lysis buffers containing 0.5-1% NP-40 or Triton X-100, 150 mM NaCl, 50 mM Tris-HCl (pH 7.4), and protease/phosphatase inhibitors. Pre-clear lysates with protein A/G beads for 1 hour at 4°C to reduce non-specific binding. For the immunoprecipitation step, incubate cleared lysates with 2-5 μg of ABI1 antibody overnight at 4°C with gentle rotation, then add protein A/G beads for 2-4 hours. Wash precipitates carefully 4-5 times with buffer containing reduced detergent concentration. When studying stimulus-dependent interactions (such as ACh-induced ABI1/N-WASP interaction), ensure consistent timing between stimulation and lysis. For verification, perform reverse co-IP (e.g., precipitate with N-WASP antibody and probe for ABI1), as demonstrated in studies examining the ACh-enhanced interaction between ABI1 and N-WASP in smooth muscle cells .

What controls should be included when using ABI1 antibodies in experimental procedures?

Rigorous experimental design with ABI1 antibodies requires multiple controls to ensure result validity. For Western blotting and immunohistochemistry, include positive controls (tissues or cell lines known to express ABI1) and negative controls (tissues or knockdown cells with minimal ABI1 expression). For antibody specificity validation, pre-absorption controls using the immunizing peptide help confirm signal specificity. When performing immunoprecipitation, include antibody-only controls (no lysate) and non-specific IgG controls to identify non-specific binding. For functional studies, use genetic approaches (ABI1 knockdown or knockout models) as complementary validation. Knockdown controls are particularly valuable, as demonstrated in studies where ABI1 knockdown cells showed absence of ABI1-N-WASP interaction in immunoprecipitation experiments, confirming assay specificity. Loading controls (β-actin, GAPDH) are essential for quantitative Western blotting to normalize ABI1 expression across samples .

How can I validate the specificity of my ABI1 antibody?

Validating ABI1 antibody specificity requires a multi-faceted approach. Begin with Western blotting to confirm the antibody detects bands of the expected molecular weight (~65 kDa for full-length ABI1), with additional bands potentially representing isoforms or post-translationally modified versions. Compare results across multiple antibodies targeting different ABI1 epitopes to establish consensus detection patterns. Perform peptide competition assays by pre-incubating the antibody with excess immunizing peptide before application; specific signals should be substantially reduced. Employ genetic validation through ABI1 knockdown or knockout models—signal intensity should decrease proportionally to reduction in ABI1 expression. Cross-reactivity testing with related proteins (e.g., ABI2) helps establish specificity within the protein family. For antibodies recognizing post-translational modifications, validate with appropriate controls (phosphatase treatment for phospho-specific antibodies). Recombinant protein expression systems can provide additional validation by correlating signal with known expression levels .

How can ABI1 antibodies be used to investigate protein-protein interactions in signaling pathways?

ABI1 antibodies provide powerful tools for dissecting complex signaling networks through multiple complementary approaches. Co-immunoprecipitation with ABI1 antibodies followed by mass spectrometry can identify novel interaction partners in an unbiased manner. For targeted studies of known or suspected interactions (such as with N-WASP, ABL kinases, or EPS8), reciprocal co-immunoprecipitation provides strong evidence of direct or indirect association. Proximity ligation assays using ABI1 antibodies in combination with antibodies against potential interaction partners enable visualization of protein-protein interactions within intact cells with nanometer resolution. For stimulus-dependent interactions, time-course studies following cellular stimulation (e.g., with ACh or EGF) reveal dynamic assembly and disassembly of protein complexes. Research has demonstrated that ACh stimulation enhances the interaction between ABI1 and N-WASP in both cultured cells and intact tissue, highlighting the physiological relevance of such stimulus-responsive interactions. These approaches collectively enable mapping of ABI1's role in cytoskeletal reorganization and signal transduction pathways .

What strategies can be employed to study ABI1's role in cytoskeletal dynamics?

Investigating ABI1's function in cytoskeletal regulation requires integrating antibody-based techniques with dynamic cellular assays. Immunofluorescence microscopy using ABI1 antibodies combined with phalloidin staining for F-actin reveals colocalization patterns during cytoskeletal remodeling. Live-cell imaging with fluorescently-tagged ABI1 complemented by fixed-cell immunostaining with ABI1 antibodies provides temporal information about recruitment to cellular structures like lamellipodia. For functional studies, combine ABI1 knockdown or overexpression with quantitative metrics of cytoskeletal dynamics (lamellipodial persistence, actin polymerization rates, cell migration velocity). Phospho-specific antibodies against ABI1 can track activation states during cytoskeletal remodeling. Biochemical fractionation followed by Western blotting with ABI1 antibodies distinguishes cytosolic versus cytoskeleton-associated pools. Super-resolution microscopy using ABI1 antibodies enables nanoscale visualization of ABI1's association with actin structures. These approaches have demonstrated ABI1's critical role in recruiting WASF1 to lamellipodia and regulating dendritic spine morphology in neurons, processes fundamentally dependent on actin cytoskeleton remodeling .

How do I design experiments to analyze ABI1's involvement in cell growth regulation and oncogenic transformation?

Experimental design for studying ABI1's role in growth regulation and transformation requires multi-level analysis of both expression and function. Begin with quantitative assessment of ABI1 expression levels across normal versus transformed cell lines using validated ABI1 antibodies in Western blotting and immunohistochemistry. Manipulate ABI1 expression through knockdown, knockout, or overexpression approaches, then measure proliferation rates, colony formation, and anchorage-independent growth. Analyze the phosphorylation status of ABL kinase substrates following ABI1 modulation, as ABI1 regulates ABL-mediated phosphorylation events. Examine ABI1's impact on EGF-induced ERK pathway activation through phospho-specific antibodies against ERK1/2. For mechanistic studies, use co-immunoprecipitation with ABI1 antibodies to track interaction with growth-regulatory partners (ABL kinases, EPS8) under normal and oncogenic conditions. In vivo studies using xenograft models with ABI1-modulated cells, followed by immunohistochemical analysis of tumor sections with ABI1 antibodies, provide translational relevance. These approaches leverage ABI1's known role in negative regulation of cell growth through interaction with nonreceptor tyrosine kinases ABL1/ABL2 .

What methodological approaches can reveal ABI1's functions in neuronal development?

Investigating ABI1's neuronal functions requires specialized techniques combining antibody-based detection with neurobiological methods. Primary neuronal cultures transfected with ABI1 constructs or treated with ABI1 siRNA provide cellular models for studying dendritic morphogenesis and synaptogenesis. Immunocytochemistry with ABI1 antibodies, combined with markers for pre- and post-synaptic structures, reveals localization patterns during development. Quantitative analysis of dendritic complexity (Sholl analysis) and spine morphology following ABI1 manipulation elucidates functional impacts. Time-lapse imaging of neurons expressing fluorescent ABI1, complemented by fixed timepoint immunostaining with ABI1 antibodies, captures dynamic aspects of ABI1 localization during developmental processes. In brain sections, layer-specific and developmental stage-specific immunohistochemistry with ABI1 antibodies maps expression patterns relevant to circuit formation. Synaptosomal fractionation followed by Western blotting with ABI1 antibodies determines synaptic enrichment. Electrophysiological recordings combined with molecular manipulation of ABI1 establish functional consequences for synaptic transmission. These approaches build upon findings that ABI1 regulates dendritic outgrowth, branching, and synaptic contact formation in developing neurons .

Why might I observe multiple bands in Western blots using ABI1 antibodies?

Multiple bands in ABI1 Western blots can arise from several biological and technical factors. Biologically, ABI1 exists in multiple splice variants, including the full-length 65 kDa form and shorter isoforms, which genuine ABI1 antibodies will detect as distinct bands. Post-translational modifications, particularly phosphorylation, can cause molecular weight shifts, resulting in ladder-like patterns. Proteolytic processing during sample preparation may generate fragments that retain the antibody epitope. Technically, insufficient blocking or high antibody concentration can increase non-specific binding. Cross-reactivity with related proteins (e.g., ABI2) may occur, especially with polyclonal antibodies. Sample degradation due to improper handling can produce breakdown products. To distinguish these possibilities, compare patterns across multiple ABI1 antibodies targeting different epitopes, perform peptide competition assays, verify with ABI1 knockdown samples, and optimize sample preparation protocols to minimize degradation. Comparing observed band patterns with literature reports helps identify which bands represent legitimate ABI1 isoforms versus artifacts .

How can I address variability in ABI1 detection between different experimental systems?

Addressing system-dependent variability in ABI1 detection requires systematic optimization and standardization. Begin by validating antibody performance in each specific experimental system (cell line, tissue type, species) through Western blotting before proceeding to more complex applications. Optimize protein extraction protocols for each system, as ABI1 solubility may vary between tissues or cell types. Standardize protein quantification methods and loading controls appropriate for each system. For cross-system comparisons, consider using multiple ABI1 antibodies targeting different epitopes to ensure consistent detection. When comparing human versus rodent samples, select antibodies with validated cross-species reactivity, noting that antibodies targeting amino acids 151-175 have demonstrated reactivity with human, mouse, and rat ABI1. For phosphorylation-dependent studies, validate phospho-specific antibodies separately in each system, as phosphorylation patterns may differ. Maintain consistent sample handling procedures, as ABI1 stability may vary between sample types. These approaches minimize technical variability, allowing reliable detection of true biological differences in ABI1 expression or modification across experimental systems .

What considerations are important when analyzing ABI1 interactions that change upon cellular stimulation?

Analysis of stimulus-dependent ABI1 interactions requires careful experimental design with attention to temporal dynamics and physiological relevance. Establish appropriate stimulation conditions (concentration, duration) based on literature or preliminary dose-response experiments. For interactions that rapidly form and dissolve, such as the ACh-enhanced interaction between ABI1 and N-WASP, perform time-course experiments with multiple sampling points to capture peak association. Ensure consistent timing between stimulation and cell lysis/fixation across experimental replicates to minimize variability. Include unstimulated controls processed identically to stimulated samples. For co-immunoprecipitation of stimulus-dependent interactions, use cell lysis buffers that preserve transient protein associations (avoid harsh detergents). Perform reciprocal co-immunoprecipitation (e.g., precipitate with both ABI1 and interaction partner antibodies) to confirm results. When possible, validate in cell culture models and intact tissues, as has been done for the ABI1-N-WASP interaction in both cultured smooth muscle cells and bronchial rings. Quantify interaction strength through densitometry of Western blots, normalizing to the amount of immunoprecipitated protein to control for precipitation efficiency differences .

How should I optimize ABI1 antibody concentrations for different experimental techniques?

Optimizing ABI1 antibody concentrations requires technique-specific titration approaches to balance signal intensity with specificity. For Western blotting, perform a dilution series (typically 1:500 to 1:5000) using positive control samples, selecting the lowest concentration that produces clear bands with minimal background. For immunohistochemistry and immunofluorescence, test dilutions from 1:100 to 1:1000 on known positive tissues, optimizing for specific signal localization without diffuse background staining. Include appropriate negative controls at each concentration. For immunoprecipitation, determine the minimum antibody amount (typically 1-5 μg) required for efficient target protein capture, as excess antibody can increase non-specific binding. For flow cytometry, establish optimal concentrations through comparison of positive versus negative cell populations, calculating signal-to-noise ratios. When using multiple detection methods in parallel (e.g., Western blotting and immunofluorescence), optimize separately for each application rather than assuming a uniform optimal concentration. Document optimal conditions (including incubation times and temperatures) for each antibody and application to ensure reproducibility across experiments. Regular validation using new antibody lots is essential, as performance may vary between manufacturing batches .

How can I use ABI1 antibodies in combination with antibody engineering technologies for improved specificity?

Integrating ABI1 antibodies with advanced antibody engineering approaches enables development of research tools with enhanced specificity profiles. Current computational models combining biophysics-informed modeling with extensive selection experiments can predict antibody binding properties and guide the design of variants with custom specificity profiles. These models optimize energy functions to generate either cross-specific sequences (interacting with multiple distinct ligands) or highly specific sequences (interacting exclusively with desired targets while excluding others). For ABI1 research, this approach could develop antibodies that specifically recognize individual ABI1 isoforms or phosphorylation states, discriminating between highly similar epitopes. Implementation requires iterative cycles of computational prediction, experimental validation through phage display selection, and refinement based on outcomes. This strategy has broad applicability beyond antibodies and offers powerful tools for designing proteins with precisely tuned physical properties, addressing a fundamental challenge in protein engineering: discrimination between very similar ligands .

What strategies can I employ to quantitatively analyze ABI1 expression and activation patterns in complex tissue samples?

Quantitative analysis of ABI1 in complex tissues requires integration of multiple complementary approaches. Multiplex immunofluorescence with ABI1 antibodies combined with cell-type-specific markers enables quantification of expression across distinct cell populations within heterogeneous tissues. For phosphorylation-dependent activation, phospho-specific ABI1 antibodies paired with total ABI1 antibodies provide activation ratios at single-cell resolution. Digital pathology platforms with automated image analysis algorithms can quantify nuclear versus cytoplasmic ABI1 localization across thousands of cells. Laser capture microdissection of specific tissue regions followed by Western blotting with ABI1 antibodies provides region-specific quantification. For absolute quantification, develop standard curves using recombinant ABI1 protein. Single-cell Western blotting or mass cytometry with ABI1 antibodies captures cell-to-cell variability masked in whole-tissue analyses. Three-dimensional tissue clearing combined with immunostaining for ABI1 and confocal microscopy enables volumetric quantification of expression patterns. These approaches collectively provide multi-parameter data on ABI1 expression, localization, and activation state across diverse cell types within intact tissue architecture .

How can I integrate ABI1 antibody-based detection with functional genomics approaches for comprehensive mechanistic studies?

Integration of antibody-based detection with functional genomics creates powerful synergies for mechanistic investigation of ABI1 biology. Combine CRISPR-Cas9 genome editing of ABI1 (knockout, knock-in of tagged versions, or specific domain deletions) with antibody-based detection of interaction partners to define domain-specific functions. Following RNA-seq of ABI1-modulated cells, validate expression changes of key targets at the protein level using specific antibodies. ChIP-seq using transcription factor antibodies on ABI1-modulated cells can identify ABI1-dependent transcriptional programs. For high-throughput screening, use automated immunofluorescence with ABI1 antibodies following siRNA or CRISPR library treatments to identify genes affecting ABI1 localization or expression. Proximity-dependent biotinylation (BioID or TurboID) with ABI1 fusion proteins followed by mass spectrometry identifies the ABI1 interactome, which can be validated using co-immunoprecipitation with ABI1 antibodies. These integrated approaches have revealed ABI1's role in regulating N-WASP and WASF1 localization and function, demonstrating how combined genetic and antibody-based methods provide mechanistic insights beyond what either approach alone could achieve .

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.