AUL1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to AUA1 Antibody

AUA1 is a murine monoclonal antibody (MoAb) first characterized for its reactivity with tumor-associated antigens on epithelial cells. It has been extensively studied in urothelial carcinomas and other epithelial malignancies. Notably, AUA1 demonstrates preferential binding to high-grade tumors, making it a tool for diagnostic and research applications .

Target Antigen and Epitope

The AUA1 antigen is a glycoprotein expressed on normal and neoplastic epithelial cells. Key features include:

  • Epitope Localization: Recognizes a surface-exposed region in the N-terminal domain of the antigen .

  • Pathological Relevance: Overexpressed in high/intermediate-grade transitional cell carcinomas (TCCs) .

  • Heterogeneity: Antigen density varies between tumors of the same grade and within individual tumors .

Key Use Cases

  • Cancer Diagnostics:

    • AUA1 reactivity correlates with tumor aggressiveness in bladder cancer .

    • Used in immunohistochemistry (IHC) to distinguish low-grade vs. high-grade TCCs .

  • Antigen Isolation:

    • Employed in affinity chromatography to purify glycoproteins from human colonic mucosa .

Limitations

  • Cross-reactivity with a subset of normal urothelial cells limits specificity .

Clinical Significance in Oncology

Study FocusFindingsSource
Transitional Cell CarcinomaAUA1 binds 85% of high-grade TCCs vs. 12% of low-grade tumors
Antigenic HeterogeneityVariable expression patterns observed within tumors, suggesting clonal evolution
Prognostic ValueHigher AUA1 reactivity associated with poorer differentiation and advanced-stage disease

Study 1: Antigen Expression in Urothelial Carcinomas

  • Methods: IHC staining of 86 TCC samples using AUA1.

  • Results:

    • 73% of high-grade TCCs showed strong AUA1 reactivity.

    • Antigen density inversely correlated with tumor differentiation status.

Study 2: Molecular Characterization

  • Methods: Affinity purification and N-terminal sequencing of the AUA1 antigen.

  • Results:

    • Identified a 17-amino acid sequence (DTYRYI-like motif).

    • Confirmed chromosomal localization to 2p13.2-p13.3 via PCR mapping.

Comparative Analysis with Similar Antibodies

AntibodyTargetClinical UtilityKey Difference from AUA1
AU1 TagEngineered epitope (BPV-1)Protein tagging in researchSynthetic epitope vs. endogenous antigen
Anti-D1 β2GPIDomain 1 of β2-glycoprotein IThrombosis/APS diagnosticsTargets phospholipid-binding protein

Future Directions

  • Therapeutic Potential: Engineering AUA1 into antibody-drug conjugates (ADCs) for targeted therapy in high-grade TCCs.

  • Biomarker Validation: Large-scale studies to standardize AUA1 IHC scoring for clinical prognostication.

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
AUL1 antibody; At1g75310 antibody; F1B16.14 antibody; F22H5.15 antibody; Auxilin-like protein 1 antibody
Target Names
AUL1
Uniprot No.

Q&A

What is the AU1 epitope tag and how does it function in protein detection?

The AU1 epitope tag represents the amino acid sequence DTYRYI, which is derived from the major capsid protein of bovine papillomavirus-1 (BPV-1) . This short peptide sequence can be genetically engineered at either the N- or C-terminus of a recombinant protein of interest, providing a universal detection point that does not typically interfere with protein function. The primary advantage of the AU1 tag lies in its small size and relative uniqueness, which minimizes the probability of cross-reactivity with endogenous cellular proteins . When researchers incorporate the AU1 tag into their protein constructs, they can reliably detect and analyze the tagged protein regardless of whether antibodies against the native protein exist or function effectively in experimental applications.

The fundamental mechanism of AU1 antibody detection relies on the high-affinity recognition between the anti-AU1 antibody and the DTYRYI epitope sequence. This interaction forms the basis for various detection methods including western blotting, immunoprecipitation, immunocytochemistry, and ELISA assays . The specificity of this interaction provides researchers with a reliable tool for tracking protein expression, localization, and interaction studies without developing custom antibodies against each protein of interest.

How does AU1 antibody compare with other common epitope tag antibodies in research applications?

When selecting an epitope tagging system for research applications, it's important to understand how AU1 compares with other commonly used epitope tags such as HA, FLAG, c-Myc, and His-tag. The AU1 tag offers distinct advantages in certain experimental contexts where small tag size and minimal interference with protein function are priorities . Unlike larger tags that might affect protein folding or function, the six-amino acid AU1 tag represents a minimal addition to the protein structure.

In terms of detection sensitivity, AU1 antibodies demonstrate high specificity when properly optimized, though they may not reach the exceptional sensitivity levels achieved by some commercial anti-FLAG or anti-HA detection systems that have undergone decades of optimization . The choice between AU1 and other tags often depends on the specific experimental requirements, including the cellular localization of the protein, the need for multiple different tags in the same experiment, and the availability of compatible detection reagents. For multi-protein tracking experiments, AU1 can be effectively combined with other epitope tags to enable simultaneous detection of different proteins within the same sample.

What are the optimal conditions for using AU1 antibody in western blotting applications?

For western blotting applications, the AU1 antibody demonstrates exceptional versatility across a wide range of dilutions, typically performing optimally between 1:1,000 to 1:20,000 dilutions . This broad working range allows researchers to adjust antibody concentration based on expression levels of the target protein and detection system sensitivity. When working with AU1-tagged proteins expressed in bacterial systems, such as E. coli, researchers have successfully detected as little as 50 ng of whole cell lysate expressing the tagged fusion protein . This level of sensitivity makes the AU1 antibody suitable for detecting both high and low abundance proteins.

The optimal blocking solution for AU1 antibody western blots typically involves 5% non-fat dry milk or 3-5% BSA in TBST (Tris-buffered saline with 0.1% Tween-20), with overnight incubation at 4°C often yielding the best signal-to-noise ratio for low-abundance proteins . When developing the blot, chemiluminescence detection methods have demonstrated excellent sensitivity, with exposure times as short as 3 seconds for strongly expressed proteins . For quantitative western blot applications, researchers should establish a standard curve using known quantities of purified AU1-tagged protein to ensure measurements fall within the linear range of detection.

What are the key considerations for successful immunoprecipitation using AU1 antibody?

When designing an immunoprecipitation experiment with AU1 antibody, researchers should consider several methodological factors. First, the choice of lysis buffer significantly impacts epitope accessibility—RIPA buffers provide stringent conditions that may disrupt some protein-protein interactions, while gentler NP-40 or digitonin-based buffers better preserve complexes but may yield higher background . Pre-clearing the lysate with protein A/G beads before adding the AU1 antibody reduces non-specific binding. Additionally, the inclusion of appropriate controls is essential: (1) a non-immune IgG control to assess non-specific binding; (2) lysate from cells not expressing the AU1-tagged protein; and (3) a known AU1-tagged protein as a positive control. For challenging applications, crosslinking the antibody to beads using dimethyl pimelimidate (DMP) can prevent antibody co-elution and improve specificity of the precipitated material.

How should AU1 antibody be utilized in immunocytochemistry and immunofluorescence applications?

Permeabilization represents another critical parameter for AU1 detection in cellular compartments. For cytoplasmic and nuclear proteins, 0.1-0.5% Triton X-100 or 0.1-0.2% saponin in PBS typically provides adequate permeabilization without disrupting cellular structures . When conducting double or triple immunofluorescence staining, researchers should carefully consider antibody species compatibility and fluorophore selection to avoid cross-reactivity and spectral overlap. Additionally, including a blocking step with normal serum (5-10%) from the same species as the secondary antibody reduces non-specific binding. For quantitative analysis of subcellular localization, Z-stack confocal microscopy with appropriate controls for bleed-through and co-localization algorithms provides more reliable data than single-plane epifluorescence imaging.

What strategies can resolve weak or absent signals when using AU1 antibody?

When researchers encounter weak or absent signals in AU1 antibody applications, several systematic troubleshooting strategies can help identify and resolve the underlying issues. First, verify protein expression using alternative detection methods—for instance, if western blotting yields no signal, confirm expression using RT-PCR or a different antibody against another domain of the fusion protein . Second, examine epitope accessibility, as the AU1 tag may be sterically hindered in certain protein conformations or membrane-embedded contexts. Addressing this might require repositioning the tag from N- to C-terminus (or vice versa) or introducing a flexible linker sequence between the tag and the protein of interest.

For western blotting specifically, insufficient transfer of high molecular weight proteins can be addressed by extending transfer time or using specialized buffers for large proteins. Using freshly prepared samples and avoiding multiple freeze-thaw cycles helps preserve epitope integrity. Additionally, modifying the antibody incubation conditions—such as extending primary antibody incubation to overnight at 4°C or increasing the concentration for challenging samples—often improves detection . For particularly difficult samples, signal amplification systems like biotin-streptavidin enhancement or highly sensitive chemiluminescent substrates can significantly improve detection limits. If all these approaches fail, consider whether post-translational modifications or proteolytic processing might be affecting the epitope region.

How can cross-reactivity issues with AU1 antibody be identified and mitigated?

Cross-reactivity represents a significant challenge in antibody-based detection systems, including those utilizing AU1 antibody. To systematically identify and address potential cross-reactivity issues, researchers should first perform careful control experiments. Using lysates or tissues from systems not expressing the AU1 tag helps identify any endogenous proteins that might cross-react with the antibody . Peptide competition assays, where the antibody is pre-incubated with excess AU1 peptide, can distinguish specific from non-specific signals—true AU1-specific signals should be eliminated in this competition experiment.

Several approaches can mitigate identified cross-reactivity. First, increasing the stringency of washing steps by adding higher concentrations of detergent (0.1-0.5% Tween-20) or salt (up to 500 mM NaCl) to wash buffers often reduces non-specific binding . Second, optimizing blocking conditions—testing different blocking agents like BSA, non-fat milk, normal serum, or commercial blocking reagents—can significantly impact specificity. Third, employing alternative detection methods that rely on different principles, such as direct fluorophore-conjugated primary antibodies instead of secondary antibody systems, sometimes eliminates cross-reactivity issues. Finally, for critical applications with persistent cross-reactivity problems, researchers should consider using epitope-tag antibodies from alternative suppliers or different clones, as the specific binding characteristics vary between antibody preparations.

What are the best practices for long-term storage and handling of AU1 antibody to maintain performance?

Proper storage and handling practices significantly impact AU1 antibody performance and longevity. The AU1 antibody demonstrates optimal stability when stored at 2-8°C for periods of at least one year, according to manufacturer specifications . For longer-term storage, aliquoting the antibody into smaller volumes and storing at -20°C or -80°C prevents repeated freeze-thaw cycles, which can cause antibody denaturation and reduced activity. When preparing working dilutions, using high-quality, sterile buffers and adding preservatives such as sodium azide (0.09%) helps prevent microbial contamination and extend shelf-life of diluted antibody preparations .

Proper handling during experimental procedures also impacts antibody performance. Researchers should avoid prolonged exposure to room temperature and direct light, particularly for fluorophore-conjugated antibodies. When diluting the antibody, use the same buffer composition as the original formulation when possible, as changes in pH or salt concentration can affect binding characteristics . For critical applications requiring maximum sensitivity, freshly thawed aliquots typically provide superior performance compared to antibodies subjected to multiple freeze-thaw cycles. Researchers should maintain detailed records of antibody lot numbers, storage conditions, and performance metrics to monitor potential degradation over time. If performance decline is observed, validation experiments comparing the current antibody preparation with a fresh lot can determine whether replacement is necessary.

How can AU1 antibody be effectively implemented in multi-protein tracking experiments?

Advanced research frequently requires simultaneous tracking of multiple proteins within the same experimental system. The AU1 epitope tag offers distinctive advantages in these complex experimental designs, particularly when used in combination with other common epitope tags such as HA, FLAG, or Myc. When designing multi-protein tracking experiments, researchers should carefully consider potential steric hindrance between tags in protein complexes and select tag positions that minimize interference with protein-protein interactions or enzymatic activity . Additionally, validating that each tagged protein retains its native function and localization is essential before proceeding with complex experimental designs.

For immunofluorescence applications involving multiple proteins, the selection of primary antibodies from different host species (e.g., rabbit anti-AU1 combined with mouse anti-HA and goat anti-FLAG) enables simultaneous detection using species-specific secondary antibodies with distinct fluorophores . When same-species antibodies cannot be avoided, sequential immunostaining with intermediate blocking steps or directly conjugated primary antibodies provides alternatives. For biochemical approaches like co-immunoprecipitation of differentially tagged proteins, tandem purification strategies can be implemented where sequential purification steps target different epitope tags (e.g., AU1 followed by FLAG), dramatically increasing the specificity of complex isolation. These advanced applications benefit from careful optimization of antibody concentrations to achieve balanced signal intensities across different channels and comprehensive controls to validate specificity of each detection component.

What considerations are important when using AU1-tagged proteins in structural biology applications?

While epitope tags like AU1 have revolutionized protein detection and purification, their implementation in structural biology applications requires specialized considerations. The placement of the AU1 tag can significantly impact protein folding, crystallization properties, and structural determination outcomes. When designing constructs for structural studies, researchers should evaluate multiple tag positions (N-terminal, C-terminal, or internal loops) and incorporate cleavable linkers containing protease recognition sites (e.g., TEV or PreScission) to enable tag removal after purification . This approach preserves the utility of the tag for purification while minimizing its impact on structural determinations.

How does AU1 antibody perform in challenging applications such as chromatin immunoprecipitation (ChIP) and FACS-based cell sorting?

For FACS applications, surface-expressed AU1-tagged proteins can be detected using indirect staining with primary AU1 antibody followed by fluorophore-conjugated secondary antibodies. This approach typically offers higher sensitivity compared to direct conjugates due to signal amplification . For intracellular AU1-tagged proteins, fixation with 2-4% paraformaldehyde followed by permeabilization with 0.1-0.5% saponin typically preserves both cellular morphology and epitope accessibility. When developing AU1 antibody-based cell sorting protocols, titriation experiments determining optimal antibody concentration are essential, as excessive antibody can increase background while insufficient amounts reduce separation between positive and negative populations. For both ChIP and FACS applications, including appropriate isotype controls and samples without AU1 expression is crucial for establishing specific signal thresholds and accurate data interpretation.

How should researchers interpret variations in AU1 antibody signal intensity across different experimental systems?

Variations in signal intensity when using AU1 antibody across different experimental systems require careful interpretation to distinguish biological significance from technical artifacts. First, researchers should recognize that epitope accessibility can vary significantly depending on protein conformation, subcellular localization, and interactions with other molecules. For instance, the same AU1-tagged protein might yield different signal intensities in western blotting versus immunoprecipitation due to differences in protein denaturation state . Similarly, fixation methods for immunocytochemistry can differentially expose or mask the AU1 epitope, leading to apparent differences in expression that actually reflect detection efficiency variations.

When comparing signal intensities between experiments, several quantitative approaches improve reliability. Implementing internal loading controls and normalization procedures addresses variations in total protein content or transfer efficiency. Using standard curves with purified AU1-tagged proteins helps establish the linear detection range and absolute quantification limits of the system . For microscopy applications, measuring relative signal intensities rather than absolute values, and reporting the ratio between the AU1 signal and a reference channel (like total protein or a housekeeping gene) improves quantitative comparisons. Additionally, researchers should maintain consistent image acquisition settings across samples when quantifying immunofluorescence data, and employ unbiased automated analysis workflows when possible to minimize subjective interpretation of signal intensities.

What statistical approaches are most appropriate for analyzing AU1 antibody-based quantitative data?

Quantitative analysis of AU1 antibody-derived data requires appropriate statistical approaches tailored to the specific experimental design and data characteristics. For western blot densitometry, researchers should first confirm that measurements fall within the linear dynamic range of detection through standard curve analysis . Log transformation of densitometry data often improves normality for parametric statistical testing. When comparing multiple experimental conditions, analysis of variance (ANOVA) followed by appropriate post-hoc tests (e.g., Tukey's HSD for all pairwise comparisons or Dunnett's test for comparisons against a control) provides more statistical power than multiple t-tests while controlling for family-wise error rates.

For immunofluorescence quantification, the nested nature of cellular data (multiple cells within multiple fields within multiple experiments) necessitates hierarchical statistical approaches or mixed-effects models rather than simple t-tests or ANOVAs. When analyzing co-localization data between AU1-tagged proteins and cellular markers, quantitative metrics such as Pearson's correlation coefficient, Manders' overlap coefficient, or object-based co-localization provide more objective assessments than visual inspection alone . For flow cytometry data using AU1 antibodies, appropriate gating strategies and compensation controls must precede statistical analysis, and measurements of median fluorescence intensity typically provide more robust metrics than mean values when distributions are skewed. Across all quantitative applications, researchers should report both effect sizes and statistical significance, as large sample sizes can make biologically insignificant differences appear statistically significant.

How is AU1 antibody technology being integrated with advanced imaging techniques?

The integration of AU1 antibody technology with advanced imaging approaches represents a frontier in molecular visualization research. Super-resolution microscopy techniques—including structured illumination microscopy (SIM), stimulated emission depletion (STED), and single-molecule localization methods like PALM and STORM—overcome the diffraction limit of conventional microscopy, enabling visualization of AU1-tagged proteins with nanometer-scale resolution . These approaches reveal previously inaccessible details about protein organization within subcellular structures. For optimal performance in super-resolution applications, researchers often employ directly-conjugated primary antibodies or smaller detection reagents like nanobodies to minimize the displacement between the fluorophore and the actual protein location, which becomes crucial at nanoscale resolution.

Emerging live-cell imaging applications include the development of cell-permeable anti-AU1 antibody fragments and complementary systems where split fluorescent proteins complement when an AU1-tagged protein interacts with an AU1-antibody fragment fusion . These approaches enable real-time tracking of protein dynamics without fixation artifacts. Additionally, correlative light and electron microscopy (CLEM) techniques combining AU1 immunofluorescence with electron microscopy provide unprecedented context by correlating protein localization with ultrastructural features. For quantitative mapping of protein-protein interactions in intact cells, proximity ligation assays (PLA) using AU1 antibody in combination with antibodies against potential interaction partners generate fluorescent signals only when proteins are within 40nm of each other, offering spatial resolution beyond conventional co-immunoprecipitation approaches.

What are the latest methodological advances for enhancing AU1 antibody specificity and sensitivity?

Recent methodological advances have significantly enhanced both the specificity and sensitivity of AU1 antibody applications in complex biological systems. Recombinant antibody technology has enabled the development of engineered AU1-binding proteins with improved properties compared to conventional antibodies. Single-chain variable fragments (scFvs) and nanobodies derived from AU1 antibodies offer smaller detection tools with superior tissue penetration for imaging applications and reduced steric hindrance for detecting protein complexes . These smaller binding reagents also enable more precise protein localization in super-resolution microscopy by reducing the displacement between the fluorophore and the actual protein position.

Signal amplification technologies have dramatically improved detection sensitivity for low-abundance AU1-tagged proteins. Tyramide signal amplification (TSA) systems, in which horseradish peroxidase-conjugated secondary antibodies catalyze the deposition of fluorescent tyramide molecules, can enhance signal intensity by 10-100 fold compared to conventional detection . Similarly, branched DNA technology and rolling circle amplification provide exponential signal enhancement for detecting scarce AU1-tagged molecules. For multiplexed detection applications, mass cytometry (CyTOF) using metal-conjugated AU1 antibodies enables simultaneous quantification of dozens of parameters without the spectral overlap limitations of fluorescence-based methods. Finally, the development of AU1 antibodies with conditional binding properties—such as pH-dependent or photoactivatable antibodies—allows for spatiotemporally controlled detection that can reveal dynamic aspects of protein behavior previously inaccessible with conventional fixed-time-point analysis.

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.