AUF2 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
14-16 weeks (made-to-order)
Synonyms
At1g22220 antibody; F16L1.5 antibody; F-box protein At1g22220 antibody
Target Names
AUF2
Uniprot No.

Target Background

Function
AUF2 is a component of SCF (SKP1-Cullin-F-box protein) E3 ubiquitin ligase complexes. These complexes mediate the ubiquitination and subsequent proteasomal degradation of target proteins.
Gene References Into Functions
  • AUXIN UP-REGULATED F-BOX PROTEIN 2 (AUF2), along with its potential paralog AUF1, has been identified as a key positive regulator of root elongation in Arabidopsis. AUF2 links auxin transport to cytokinin signaling. [PMID: 21653785](https://www.ncbi.nlm.nih.gov/pubmed/21653785)
Database Links

KEGG: ath:AT1G22220

STRING: 3702.AT1G22220.1

UniGene: At.49903

Subcellular Location
Nucleus.

Q&A

What are the primary considerations when selecting antibodies for AAV serotype research?

When selecting antibodies for AAV serotype research, researchers must first consider the specific serotype of interest, as seroprevalence varies significantly between AAV1, AAV2, AAV7, and AAV8 across different geographical populations. The selection should be informed by epidemiological data, which shows that seroprevalences at serum dilutions of >1:20 and >1:80 are typically highest for anti-AAV2 neutralizing antibodies (NAbs) across different global regions including Australia, Europe, Africa, and the United States . Researchers should also consider cross-reactivity between different AAV serotypes when designing experiments, as there can be overlapping immune responses. The sensitivity and specificity of the antibody detection method are crucial, with options including direct ELISA and immune complex (IC) assays, where the latter requires significantly less capsid material and provides intrinsic specificity control . Additionally, researchers should determine whether they need to detect total antibodies or specifically neutralizing antibodies, as this will influence both experimental design and interpretation of results.

How do immune complex (IC) assays compare to direct ELISA for detecting anti-AAV antibodies?

Immune complex (IC) assays offer several advantages over direct ELISA for detecting anti-AAV antibodies, primarily through their significantly reduced capsid material consumption, requiring approximately 10-30 fold less material than conventional methods . The IC assay format is based on the formation of immune complexes in solution and their subsequent detection using a biotinylated anti-AAV2 antibody for capture and an antibody against the study species IgG for detection, making it more resource-efficient for early-stage research . Unlike direct ELISA, the IC assay is intrinsically drug tolerant, capable of detecting both free and AAV2-bound anti-AAV2 antibodies, which provides a more comprehensive picture of the immune response . The IC assay incorporates a built-in specificity control through the use of nonspiked samples as negative controls, enabling clearer discrimination between positive and negative samples without requiring additional confirmatory steps . While the IC assay may exhibit somewhat lower sensitivity in certain contexts, such as in cynomolgus monkey study samples, studies have demonstrated that this difference does not typically lead to qualitatively different interpretations of results compared to direct ELISA methods . The IC assay format can also be easily adapted to different AAV serotypes and species, making it a versatile tool for immunogenicity assessment in gene therapy development.

What are the geographical variations in seroprevalence of neutralizing antibodies to different AAV serotypes?

The seroprevalence of neutralizing antibodies to different AAV serotypes exhibits significant geographical variations, which has important implications for gene therapy research and clinical applications. Across Australia, Europe, Africa, and the United States, the highest seroprevalences at serum dilutions of >1:20 and >1:80 are consistently observed for anti-AAV2 neutralizing antibodies (NAbs) . The second highest seroprevalences were typically observed for anti-AAV1 NAbs in most geographical regions and dilution conditions, with the exception of the United States at >1:80 dilution, where anti-AAV7 NAb seroprevalence was second highest . In Europe, specifically in countries like Belgium, Greece, and Italy, the seroprevalence pattern at >1:20 dilution showed anti-AAV2 NAb exceeding anti-AAV1 NAb, with statistically significant differences observed in Italy and Greece . Interestingly, at higher dilutions of >1:80 in Greece and Italy, this pattern reversed, with anti-AAV1 NAbs exceeding anti-AAV2 NAbs, while Belgium maintained higher anti-AAV2 NAb seroprevalence . Consistently across all countries studied, the seroprevalences of anti-AAV7 and anti-AAV8 NAbs were lower than that of anti-AAV2 NAb at both serum dilutions, suggesting that these serotypes derived from nonhuman primates may offer advantages for human gene therapy applications in populations with high reactivity toward AAV2 and AAV1 .

How do monoclonal antibodies against specific tumor markers demonstrate efficacy in cancer treatment models?

Monoclonal antibodies targeting specific tumor markers demonstrate efficacy through multiple mechanistic pathways that collectively inhibit cancer progression. In the case of anti-AGR2 (Anterior gradient-2) monoclonal antibody mAb18A4, efficacy was established through simultaneous activation of the p53 tumor suppressor pathway and attenuation of the ERK1/2-MAPK pathway, creating a two-pronged approach to cancer inhibition . The antibody's impact on cellular processes was multifaceted, causing attenuated proliferation and colony formation in cancer cell lines, enhanced apoptosis, increased p53 expression, and reduced phosphorylated ERK1/2 expression - all critical factors in controlling tumor growth . In xenograft tumor models, treatment with the monoclonal antibody significantly reduced tumor size, suppressed tumor metastasis, and increased survival rates, demonstrating translation of in vitro efficacy to in vivo therapeutic potential . Additionally, the antibody demonstrated potent suppression of tumor angiogenesis, specifically targeting AGR2-induced blood vessel formation, which is crucial for cutting off the tumor's nutrient supply . Pharmacokinetic and toxicological analyses confirmed the safety profile of the antibody, showing no adverse side effects on major organs and blood parameters in mice, highlighting the advantage of targeted antibody therapies over traditional chemotherapy and radiotherapy approaches which often produce significant off-target effects .

What methodological approaches can overcome the challenges of epitope masking in protein complex analysis with antibodies?

Epitope masking in protein complex analysis presents significant challenges for researchers, as demonstrated in studies where supershift analysis with antibodies like hnRNPI yielded negative results due to epitopes being hidden within protein complexes . Overcoming these challenges requires multiple complementary methodological approaches. One effective strategy involves employing epitope mapping techniques prior to antibody selection, which helps identify accessible regions of the protein when it exists in complexes. This can be accomplished through techniques such as hydrogen-deuterium exchange mass spectrometry (HDX-MS) or cross-linking mass spectrometry (XL-MS) to determine which protein domains remain exposed in complex formations. Researchers should develop antibody panels targeting multiple distinct epitopes on the protein of interest, increasing the probability that at least one epitope will remain accessible in various complex configurations. For particularly challenging targets, engineered antibody fragments like single-chain variable fragments (scFvs) or nanobodies with smaller footprints may access epitopes that would be inaccessible to conventional antibodies. Additionally, modifying experimental conditions through techniques like mild detergent treatment or utilizing different buffer compositions can sometimes partially dissociate complexes enough to expose hidden epitopes without completely disrupting the interactions of interest. When direct antibody detection proves impossible, researchers may resort to indirect approaches such as proximity ligation assays or fluorescence resonance energy transfer (FRET) to verify protein associations without requiring direct epitope binding.

How can researchers optimize immune complex (IC) assay conditions for detecting low-titer antibodies against AAV vectors?

Optimizing immune complex (IC) assay conditions for detecting low-titer antibodies against AAV vectors requires careful consideration of multiple parameters to enhance sensitivity while maintaining specificity. The concentration of capsid particles used for spiking samples is a critical parameter that requires precise optimization, as demonstrated in studies where an optimal rAAV2p spike concentration of 1.65 × 10^9 vp/ml was determined through titration experiments with positive controls and individual serum samples . Researchers should conduct systematic testing of serum dilution factors, as the final serum concentration significantly impacts both sensitivity and background signal; studies have shown that a 1% (v/v) final serum concentration provides an optimal balance for many applications . The capture antibody selection and concentration must be carefully calibrated, with biotinylated monoclonal antibodies like anti-AAV2 antibody A20R at 0.1 μg showing effective performance for immune complex capture onto streptavidin-coated plates . Incubation conditions, including temperature, duration, and buffer composition, should be optimized, with studies indicating that room temperature incubation for 30 minutes in Low Cross Buffer® provides effective complex formation while minimizing non-specific interactions . For enhanced detection of low-titer antibodies, signal amplification strategies can be employed, such as using highly sensitive substrates for horseradish peroxidase detection or implementing multi-layered detection systems. Additionally, establishing an appropriate cut-point methodology is crucial, with some successful approaches defining in-study cut points as two times the mean signal value of nonspiked predose samples, providing reliable discrimination between positive and negative samples even at low antibody titers .

What controls should be included when developing antibody-based assays for AAV immunogenicity testing?

Developing robust antibody-based assays for AAV immunogenicity testing requires a comprehensive suite of controls to ensure reliability and interpretability of results. Positive control antibodies with known binding characteristics, such as the monoclonal antibody A20H used in IC assay development, should be included to validate assay performance and serve as a reference standard for quantification . Negative controls consisting of samples from naive subjects (pre-treatment or from unexposed individuals) are essential for establishing background signal levels and determining assay cut-points; these samples should match the matrix of test samples (e.g., same species serum) to account for matrix effects . Specificity controls must be implemented to confirm that signals are specifically due to anti-AAV antibodies rather than non-specific binding; in the IC assay format, this can be elegantly achieved by running parallel samples without spiked capsid, which eliminates the need for separate confirmatory steps . Dilutional linearity controls, where positive samples are analyzed across a range of dilutions, help verify that the assay responds proportionally to antibody concentration and aids in identifying potential hook effects or interference at certain concentrations . Matrix interference controls containing the therapeutic product but no anti-drug antibodies help identify potential false positives due to matrix components, particularly important when working with diverse sample types or species . Additionally, system suitability controls that monitor critical assay parameters such as binding capacity, signal-to-noise ratio, and day-to-day variability should be incorporated to ensure consistent assay performance across different experimental runs.

How can researchers interpret contradictory antibody data between different detection methods for AAV immunogenicity?

Interpreting contradictory antibody data between different detection methods for AAV immunogenicity requires a systematic analytical approach that considers the fundamental differences between assay technologies. Researchers should first examine the inherent sensitivity differences between methods, as demonstrated in studies comparing IC assays with direct ELISA, where somewhat lower sensitivity was observed in the IC assay for cynomolgus monkey study samples, though this did not lead to qualitatively different interpretations of results . The epitope-binding properties of each assay must be considered, as different methods may detect antibodies binding to different regions of the AAV capsid; neutralizing antibody assays specifically identify antibodies that inhibit transduction, while binding assays like direct ELISA or IC assays detect all antibodies that bind to the capsid regardless of their neutralizing capacity . The detection range and limitations of each assay should be evaluated, with attention to potential signal saturation at high antibody concentrations and floor effects at low concentrations, which may explain discrepancies in samples near the detection limits . Matrix effects and interference factors that affect one assay type more than another should be investigated, particularly when working with complex biological samples that contain components potentially interacting with assay reagents . When faced with contradictory results, researchers should consider conducting orthogonal validation by introducing a third method or modified protocol to resolve discrepancies, and when appropriate, correlate results with functional outcomes such as transgene expression or clinical efficacy to determine which assay better predicts relevant biological effects. Finally, statistical approaches such as Bland-Altman analysis can help quantify the systematic differences between methods and establish conversion factors when combining or comparing data sets generated using different assay platforms.

How might next-generation antibody detection methods improve our understanding of AAV immunity?

Next-generation antibody detection methods hold transformative potential for advancing our understanding of AAV immunity through several key innovations. Single B-cell sequencing technologies, when applied to AAV-responsive B cells, could provide unprecedented insights into the repertoire diversity and affinity maturation processes of anti-AAV antibodies, enabling researchers to track the evolution of immune responses at a molecular level. Mass spectrometry-based approaches like serological proteome analysis might identify the specific epitopes recognized by anti-AAV antibodies with high resolution, potentially revealing previously uncharacterized immunodominant regions that could inform capsid engineering strategies to evade pre-existing immunity . Advanced biosensor technologies including surface plasmon resonance (SPR) and biolayer interferometry (BLI) enable real-time kinetic analysis of antibody-capsid interactions, providing detailed binding parameters that could help differentiate neutralizing from non-neutralizing antibodies based on their binding characteristics rather than functional assays alone . Artificial intelligence and machine learning algorithms applied to large immunogenicity datasets could identify subtle patterns in antibody responses that predict clinical outcomes, potentially allowing for personalized approaches to gene therapy based on individual immune profiles . Multiplex cytokine analysis integrated with antibody profiling could provide a more comprehensive view of the immune response to AAV, capturing both humoral and cellular components simultaneously . Additionally, the development of standardized reference materials and assays through international collaborations would enable more meaningful comparisons across studies and accelerate the field's understanding of factors influencing AAV immunogenicity, ultimately leading to more effective gene therapy approaches with reduced immunological barriers.

What role might modified antibody formats play in overcoming current limitations in therapeutic applications?

Modified antibody formats represent a frontier in overcoming current limitations in therapeutic applications through diverse structural and functional innovations. Bispecific antibodies that simultaneously target tumor markers like AGR2 and components of the immune system could enhance therapeutic efficacy by directly recruiting immune cells to tumor sites, potentially improving upon the already promising results seen with conventional monoclonal antibodies like mAb18A4 . Antibody-drug conjugates (ADCs) that combine the specificity of antibodies with the potency of cytotoxic payloads could deliver highly concentrated therapeutic agents directly to tissues expressing specific targets, minimizing off-target effects while maximizing efficacy at lower doses . Fragment-based approaches utilizing Fab fragments, single-chain variable fragments (scFvs), or nanobodies might enable better tissue penetration compared to full-size antibodies, particularly in solid tumors where poor perfusion often limits therapeutic efficacy . Engineering antibodies with modified Fc regions could tailor their interaction with the immune system, either enhancing effector functions for cancer therapies or reducing them for applications where immune activation is undesirable . Domain antibodies and alternative scaffold proteins represent minimal binding domains that can be produced at lower cost and potentially with improved stability, opening possibilities for novel therapeutic modalities and delivery systems . Additionally, antibody formats engineered for improved central nervous system penetration could address the significant challenge of delivering therapeutics across the blood-brain barrier, potentially enabling antibody-based approaches for neurodegenerative conditions . These innovative formats collectively expand the therapeutic potential of antibodies beyond conventional applications, potentially addressing current limitations in specificity, tissue accessibility, and immune system engagement.

How can researchers best integrate data from antibody studies with other immunological parameters to predict therapeutic outcomes?

Integrating data from antibody studies with other immunological parameters requires sophisticated analytical approaches to develop predictive models for therapeutic outcomes. Multiparametric flow cytometry combined with antibody profiling can create comprehensive immune signatures by simultaneously analyzing multiple cell populations and their activation states alongside antibody responses, providing a more holistic view of the immune landscape . Machine learning algorithms applied to integrated datasets containing antibody titers, neutralizing activity, T-cell responses, cytokine profiles, and clinical outcomes can identify complex patterns and interdependencies that might not be apparent through conventional statistical methods . Systems biology approaches that map the relationships between antibody responses and broader immunological networks through techniques like pathway analysis and network modeling can reveal unexpected connections between seemingly disparate immune parameters . Longitudinal analysis tracking the evolution of both antibody responses and cellular immunity over time, rather than single-point measurements, provides crucial information about the dynamics of immune responses that may better predict long-term outcomes . Spatial transcriptomics and multiplexed immunohistochemistry can place antibody data in the context of tissue microenvironments, revealing local immune interactions that might influence therapeutic responses in ways not captured by systemic measurements . Additionally, developing standardized reporting frameworks for immunological data would facilitate meta-analyses across studies, allowing researchers to identify robust predictive factors from diverse datasets generated using different methodologies . These integrated approaches move beyond simplistic correlations to develop mechanistic models that account for the complex interplay between humoral immunity, cellular responses, and tissue-specific factors, ultimately providing more accurate predictions of therapeutic outcomes in diverse patient populations.

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 2024 Thebiotek. All Rights Reserved.