AUXI1 Antibody

Shipped with Ice Packs
In Stock

Description

AUX1 Protein Overview

AUX1 is a transmembrane transporter responsible for auxin (indole-3-acetic acid, IAA) uptake into plant cells. It belongs to the AUX/LAX family and facilitates IAA⁻ transport at mildly acidic apoplastic pH levels (pH 5.2–5.5) with a binding affinity (KdK_d) comparable to its transport KmK_m (~800 nM) . AUX1 is predominantly localized to the plasma membrane (PM) , though minor ER-associated pools have been observed .

Immunogen Design

  • The N-terminal 19-amino-acid sequence of AUX1 was selected for antibody production due to its divergence from other AUX/LAX family members .

  • Expressed as an HA-tagged construct (HA-AUX1) in Spodoptera frugiperda (Sf9) insect cells via baculovirus infection .

Validation

  • Specificity: Anti-AUX1 antibodies show no cross-reactivity with AUX/LAX paralogs (e.g., LAX1, LAX2, LAX3) .

  • Western Blot: Detects HA-AUX1 at ~60 kDa (full-length) and a truncated form at ~40 kDa .

  • Negative Controls: No signal in aux1-100 T-DNA null mutants .

Subcellular Localization

FractionMarker ProteinAUX1 Signal IntensityCitation
MicrosomalPM H⁺-ATPaseHigh
ERCalnexinLow
Golgiγ-TIPAbsent

Protein-Protein Interactions

  • AXR4 Interaction: Co-immunoprecipitation (Co-IP) in insect cells confirms AUX1 binds directly to AXR4, an ER accessory protein critical for AUX1 trafficking .

  • Control Experiments: No interaction observed between AXR4 and the mammalian ABCB1 transporter, ruling out nonspecific binding .

Auxin Transport Mechanism

  • AUX1 mediates IAA⁻ uptake with a KmK_m of 800 nM in Xenopus oocytes , resolving discrepancies from earlier cell culture studies (1–5 μM) .

  • Competitive binding assays show auxin analogues (e.g., 2,4-D, NPA) exhibit distinct affinities for AUX1, correlating with their in vivo efficacy .

Role in Cytokinin Signaling

  • AUX1 regulates cytokinin-mediated root cell elongation but not proliferation. aux1 mutants show insensitivity to cytokinin-induced growth inhibition .

Limitations and Future Directions

  • Cross-Reactivity: No data yet confirm reactivity with AUX1 orthologs in crops like cucumber .

  • Structural Insights: Computational modeling of antibody-antigen interfaces could optimize AUX1 antibody affinity for low-abundance targets.

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
AUXI1 antibody; At4g12780 antibody; T20K18.130Auxilin-related protein 1 antibody; Auxilin-like protein 1 antibody
Target Names
AUXI1
Uniprot No.

Target Background

Function
This antibody promotes the uncoating of clathrin-coated vesicles. It may interact directly with clathrin.
Database Links

KEGG: ath:AT4G12780

STRING: 3702.AT4G12780.1

UniGene: At.21973

Subcellular Location
Cell membrane; Peripheral membrane protein. Golgi apparatus, trans-Golgi network. Endoplasmic reticulum. Cytoplasmic vesicle.

Q&A

What is the AUXI1 protein and why are antibodies against it valuable for research?

AUXI1 (also known as AUX1) functions as an auxin influx carrier in plants, particularly in Arabidopsis thaliana. This transmembrane protein plays a crucial role in auxin transport, which regulates various aspects of plant growth and development. Antibodies against AUXI1 are valuable research tools that enable detection, localization, and functional analysis of this protein in plant tissues. The AUXI1 protein belongs to the AUX/LAX family, with the 19-amino-acid N-terminal sequence representing the most divergent segment among family members, making it an ideal target for specific antibody production . These antibodies facilitate studies on auxin transport mechanisms, which are fundamental to understanding plant development, tropisms, and responses to environmental stimuli.

How is the AUXI1 antibody typically generated for research applications?

AUXI1 antibodies are commonly generated using the 19-amino-acid N-terminal sequence of the protein as the peptide antigen for antibody production. This region is selected because it represents the most divergent segment of protein coding sequence among the four members of the Arabidopsis AUX/LAX family, enhancing antibody specificity . The standard approaches for antibody generation include:

  • Peptide immunization: Synthesizing the N-terminal peptide sequence, conjugating it to a carrier protein, and immunizing animals (typically rabbits for polyclonal or mice for monoclonal antibodies)

  • Recombinant protein expression: Expressing portions of the AUXI1 protein in bacterial systems for immunization

  • Hybridoma technology: For monoclonal antibody production following immunization

  • Recombinant antibody technology: Using phage display or similar methods to generate antibodies with defined sequences

The specificity of the resulting antibody can be confirmed by testing against wild-type samples and aux1 null mutants (such as aux1-100), where a cross-reacting band should be detectable in wild-type samples but absent in the mutant .

What controls should be included when using AUXI1 antibodies in experiments?

When using AUXI1 antibodies, several essential controls should be incorporated to ensure experimental validity:

  • Genetic controls: Include samples from aux1 null mutants (e.g., T-DNA–tagged aux1-100) alongside wild-type samples to confirm antibody specificity. The absence of signal in the null mutant validates that the antibody is detecting the intended target .

  • Preimmune serum control: Use serum collected before immunization to identify any non-specific background signals.

  • Competing peptide control: Pre-incubate the antibody with excess antigenic peptide to demonstrate signal specificity.

  • Cross-reactivity tests: Test against other AUX/LAX family members to confirm specificity for AUXI1.

  • Multiple antibody validation: When possible, use independent antibodies raised against different epitopes of AUXI1 to confirm results, following the "multiple antibody strategy" pillar of antibody validation .

  • Subcellular fractionation controls: When performing localization studies, include appropriate markers for different cellular compartments (e.g., plasma membrane, ER, Golgi, tonoplast) to accurately interpret the distribution of AUXI1 .

Implementing these controls enhances the reliability of results and addresses the common concern of antibody specificity in the research community.

What are the recommended methods for validating the specificity of an AUXI1 antibody?

Validating AUXI1 antibody specificity should follow multiple complementary approaches aligned with the "five pillars of antibody characterization" :

  • Genetic strategies: Testing against aux1 knockout or knockdown plant lines is the gold standard. For example, comparing signal between wild-type Arabidopsis and the T-DNA–tagged aux1-100 null mutant can confirm specificity .

  • Orthogonal strategies: Compare antibody-dependent detection methods with antibody-independent techniques (such as mass spectrometry or RNA-seq) to correlate protein detection with gene expression levels.

  • Multiple antibody strategy: Use different antibodies targeting distinct epitopes of AUXI1 to verify consistent localization or expression patterns.

  • Recombinant expression: Test the antibody against samples with artificially increased AUXI1 expression to confirm signal enhancement.

  • Immunocapture followed by mass spectrometry: Use the antibody for immunoprecipitation followed by mass spectrometry to confirm the identity of the captured protein as AUXI1.

Additionally, Western blotting against total protein, soluble, and microsomal fractions can verify the expected membrane localization pattern, where AUXI1 should be absent from the soluble fraction but enriched in the microsomal fraction . Proper documentation of these validation efforts is essential, as it has been estimated that approximately 50% of commercial antibodies fail to meet basic characterization standards .

How do I determine the optimal working dilution and conditions for an AUXI1 antibody in different applications?

Determining optimal working conditions for AUXI1 antibodies requires systematic titration across different applications:

For Western blotting:

  • Perform a dilution series (e.g., 1:500, 1:1000, 1:2000, 1:5000) using consistent sample loading

  • Test different blocking agents (BSA, milk, commercial blockers) to minimize background

  • Optimize incubation times and temperatures (typically 1-2 hours at room temperature or overnight at 4°C)

  • Test different detection systems (HRP-conjugated secondary antibodies, fluorescent secondaries)

For immunolocalization:

  • Test fixation protocols (paraformaldehyde, glutaraldehyde, or combinations)

  • Evaluate permeabilization methods (detergents like Triton X-100 or saponin)

  • Try antigen retrieval methods if initial attempts show weak signal

  • Test antibody dilutions starting higher than for Western blots (1:100 to 1:500)

For immunoprecipitation:

  • Determine optimal antibody-to-protein ratio

  • Test different lysis buffers to maintain AUXI1 in its native conformation

  • Consider cross-linking strategies for membrane proteins

Document the outcome of each condition using consistent positive and negative controls. Follow the example of resources like NeuroMab, which emphasizes transparency by providing detailed protocols and highlighting the need to optimize conditions in each laboratory and assay employed .

What characterization data should researchers report when publishing studies using AUXI1 antibodies?

When publishing studies utilizing AUXI1 antibodies, researchers should report comprehensive characterization data to ensure reproducibility and reliability:

  • Antibody source and identifier:

    • Commercial source with catalog and lot numbers

    • For custom antibodies: detailed immunization protocol, antigen sequence, and production method

    • RRID (Research Resource Identifier) when available

  • Validation experiments:

    • Evidence of specificity (testing on aux1 mutants)

    • Blot images showing appropriate molecular weight

    • Subcellular fractionation data confirming expected localization patterns

    • Cross-reactivity tests with other AUX/LAX family members

  • Experimental conditions:

    • Complete protocols with dilutions and incubation times

    • Sample preparation methods (extraction buffers, fixation protocols)

    • Blocking conditions and washing steps

    • Detection methods and image acquisition parameters

  • Controls included:

    • Genetic controls (wild-type vs. mutant)

    • Technical controls (preimmune serum, peptide competition)

    • Subcellular marker controls in localization studies

This detailed reporting aligns with efforts to address the "antibody characterization crisis" that has resulted in estimated financial losses of $0.4–1.8 billion per year in the United States alone due to poorly characterized antibodies . Transparent reporting enables other researchers to properly evaluate and build upon published findings.

How can AUXI1 antibodies be used to study subcellular localization of the protein?

AUXI1 antibodies are valuable tools for determining the subcellular localization of the protein through several complementary approaches:

Biochemical fractionation and immunoblotting:

  • Prepare total, soluble, and microsomal fractions from plant tissues (e.g., Arabidopsis root cultures)

  • Perform protein gel blotting with anti-AUXI1 antibodies

  • Analyze the distribution pattern, which should show enrichment in the microsomal fraction and absence from the soluble fraction, consistent with membrane localization

  • Further resolve subcellular localization using sucrose density-based organelle fractionation (15-50% continuous gradient)

  • Compare AUXI1 signal distribution with established markers for different compartments (plasma membrane H+-ATPase, ER BiP, Golgi, and tonoplast markers)

Immunohistochemistry and immunofluorescence microscopy:

  • Fix plant tissues using optimized protocols that preserve AUXI1 epitopes

  • Perform immunolabeling with AUXI1 antibodies

  • Counterstain with markers for cellular compartments

  • Use confocal microscopy for high-resolution imaging

  • Apply image analysis to quantify colocalization with membrane markers

In previous studies, these approaches demonstrated that AUXI1 predominantly localizes to the plasma membrane in Arabidopsis root cells, with signal comigrating with plasma membrane H+-ATPase marker in sucrose gradient fractions . This plasma membrane localization is consistent with AUXI1's proposed function as an auxin influx carrier.

How can flow cytometry be used with AUXI1 antibodies for plant cell analysis?

While flow cytometry with AUXI1 antibodies is less common than other techniques in plant research, it can provide valuable quantitative data on protein expression across cell populations:

  • Sample preparation:

    • Isolate protoplasts from plant tissues using enzymatic digestion

    • Fix and permeabilize cells to enable antibody access to intracellular AUXI1

    • Block nonspecific binding sites with appropriate agents

  • Antibody staining:

    • Incubate with optimized concentration of AUXI1 primary antibody

    • Wash to remove unbound antibody

    • Label with fluorochrome-conjugated secondary antibody

    • Include appropriate controls (unstained, secondary-only, isotype controls)

  • Data acquisition and analysis:

    • Collect multiparameter data including forward scatter, side scatter, and fluorescence

    • Analyze using sequential gating strategies to identify AUXI1-positive populations

    • Compare expression levels across different cell types or treatment conditions

  • Advanced applications:

    • Combine with cell sorting to isolate specific cell populations

    • Perform multicolor analysis with additional markers for cell identity or other proteins

    • Quantify relative AUXI1 expression levels across cell types

Flow cytometry offers the advantage of analyzing thousands of individual cells rapidly, providing statistical power for detecting subtle changes in AUXI1 expression or localization that might be missed by other techniques .

What approaches can be used to study AUXI1 protein interactions using antibodies?

Studying AUXI1 protein interactions requires specialized approaches for membrane proteins:

  • Co-immunoprecipitation (Co-IP):

    • Solubilize membrane proteins using gentle detergents that maintain protein-protein interactions

    • Immunoprecipitate AUXI1 using validated antibodies

    • Analyze co-precipitated proteins by Western blotting or mass spectrometry

    • Verify interactions through reciprocal Co-IP experiments

    • Include appropriate controls (IgG control, aux1 mutant extracts)

  • Proximity-based labeling:

    • Generate fusion proteins combining AUXI1 with enzymes like BioID or APEX2

    • Express in plant systems to label proteins in close proximity to AUXI1

    • Use streptavidin pulldown to capture biotinylated proteins

    • Identify interacting proteins by mass spectrometry

    • Confirm specific interactions using AUXI1 antibodies

  • In situ proximity ligation assay (PLA):

    • Use AUXI1 antibody together with antibodies against suspected interaction partners

    • Apply secondary antibodies with attached DNA oligonucleotides

    • When proteins are in close proximity, oligonucleotides can interact

    • Amplify and detect the signal by addition of fluorescent probes

    • Visualize interactions at their subcellular locations

  • Antibody-based FRET or BRET analysis:

    • Label AUXI1 antibody and partner protein antibody with FRET-compatible fluorophores

    • Measure energy transfer as indication of protein proximity

    • Compare results in wild-type and aux1 mutant tissues as controls

These approaches can help identify components of auxin transport complexes and regulatory partners of AUXI1, advancing our understanding of auxin transport mechanisms in plant development.

What are common issues when using AUXI1 antibodies and how can they be resolved?

When working with AUXI1 antibodies, researchers may encounter several challenges that require systematic troubleshooting:

IssuePossible CausesSolutions
No signal in Western blot- Degraded protein sample
- Inefficient transfer
- Antibody denaturation
- Epitope inaccessibility
- Add protease inhibitors to extraction buffer
- Optimize transfer conditions for membrane proteins
- Try different extraction buffers
- Reduce SDS concentration in sample buffer
High background- Insufficient blocking
- Antibody concentration too high
- Cross-reactivity
- Secondary antibody issues
- Increase blocking time/concentration
- Use different blocking agent (BSA vs. milk)
- Dilute primary antibody further
- Increase wash steps duration/frequency
- Try a different secondary antibody
Unexpected band size- Post-translational modifications
- Protein degradation
- Splice variants
- Cross-reactivity
- Include deglycosylation controls
- Use fresh samples with protease inhibitors
- Compare with recombinant AUXI1 standard
- Confirm with mass spectrometry
Poor reproducibility- Lot-to-lot antibody variation
- Inconsistent sample prep
- Protocol variations
- Use recombinant antibodies when available
- Standardize sample preparation
- Document detailed protocols
- Include consistent positive controls
Weak signal in IHC/IF- Insufficient fixation
- Overfixation masking epitopes
- Inadequate permeabilization
- Antibody concentration too low
- Optimize fixation time
- Try antigen retrieval methods
- Increase permeabilization
- Increase antibody concentration or incubation time

It's important to note that antibody characterization is "context-dependent," requiring validation by end users for each specific application . When troubleshooting, systematic variation of one parameter at a time while keeping others constant will help identify the source of the problem.

How should researchers analyze and interpret AUXI1 localization data from immunofluorescence studies?

Analyzing and interpreting AUXI1 localization data requires careful consideration of several factors:

  • Image acquisition considerations:

    • Use consistent exposure settings across samples

    • Acquire z-stacks to capture the full 3D distribution

    • Include multi-channel imaging for colocalization studies

    • Collect sufficient biological and technical replicates

  • Quantitative analysis approaches:

    • Measure signal intensity along cell membranes vs. cytoplasm

    • Calculate Pearson's or Mander's coefficients for colocalization with membrane markers

    • Analyze polar distribution patterns in cells with directional auxin transport

    • Use intensity profile plots across cellular regions

  • Interpretation guidelines:

    • Compare patterns with known subcellular markers (e.g., plasma membrane H+-ATPase)

    • Consider the biological context (cell type, developmental stage)

    • Interpret in light of aux1 mutant phenotypes and auxin transport models

    • Account for potential fixation artifacts

  • Validation strategies:

    • Confirm key findings with complementary approaches (e.g., biochemical fractionation)

    • Use fluorescently-tagged AUXI1 constructs as alternative localization method

    • Correlate localization patterns with functional assays

Previous studies have shown that AUXI1 predominantly localizes to the plasma membrane, consistent with its role as an auxin influx carrier . When analyzing novel patterns or distribution changes, researchers should consider how these relate to the protein's functional role in auxin transport and plant development.

How can researchers quantitatively analyze AUXI1 protein levels across different samples or conditions?

Quantitative analysis of AUXI1 protein levels requires careful experimental design and appropriate analytical methods:

  • Western blot quantification:

    • Use a dilution series of samples to ensure measurements fall within the linear range of detection

    • Include loading controls appropriate for membrane proteins (avoid using soluble proteins like GAPDH)

    • Apply image analysis software to measure band intensities

    • Normalize AUXI1 signal to loading controls

    • Include an internal standard across blots for inter-blot normalization

  • ELISA-based quantification:

    • Develop a sandwich ELISA using captured AUXI1 antibody and a detection antibody

    • Generate a standard curve using recombinant AUXI1 protein

    • Process samples and standards in parallel

    • Calculate absolute AUXI1 concentrations based on the standard curve

  • Flow cytometry quantification:

    • Use calibrated beads to convert fluorescence intensity to molecules of equivalent soluble fluorochrome

    • Apply consistent gating strategies across samples

    • Report median fluorescence intensity (MFI) rather than mean values

    • Use histogram overlays to visualize shifts in expression levels

  • Statistical analysis considerations:

    • Apply appropriate statistical tests based on data distribution

    • Account for biological and technical replicates in the analysis

    • Use appropriate visualization (box plots, violin plots) to show data distribution

    • Report effect sizes alongside p-values

When comparing AUXI1 levels across genotypes or conditions, researchers should be mindful that changes in protein levels may reflect alterations in expression, stability, or localization, each with distinct biological implications for auxin transport.

How can AUXI1 antibodies be used to study protein-structure function relationships?

AUXI1 antibodies offer powerful approaches for investigating structure-function relationships when combined with analysis of aux1 allelic variants:

  • Epitope mapping with missense mutations:

    • Use the extensive aux1 allelic series (which includes 15 missense mutations) to identify critical functional domains

    • Test antibody binding to different mutant proteins to map epitope locations

    • Correlate antibody binding patterns with functional defects in mutants

    • Focus on the central permease region (amino acids 201-300) where mutations cluster significantly

  • Conformational antibodies for transport studies:

    • Generate antibodies that recognize specific conformational states of AUXI1

    • Use these to track transport-associated conformational changes

    • Apply conformation-specific antibodies in the presence/absence of auxin

    • Correlate conformational dynamics with transport activity

  • Topology analysis:

    • Generate antibodies against different domains of AUXI1

    • Use protease protection assays with these antibodies to map membrane topology

    • Compare topology with related amino acid transporters like NAT2/AAP1

    • Identify cytoplasmic loops and extracellular domains

  • Co-immunoprecipitation with mutant variants:

    • Use antibodies to immunoprecipitate wild-type and mutant AUXI1 proteins

    • Analyze differential protein interactions in various mutants

    • Connect specific domains to protein-protein interaction functions

    • Map the transport complex architecture

This approach leverages the wealth of genetic resources available in Arabidopsis, where aux1 allelic series offers a powerful experimental approach to identify important residues and domains .

What strategies can be employed to develop more specific next-generation antibodies against AUXI1?

Developing more specific next-generation AUXI1 antibodies requires leveraging modern antibody engineering technologies:

  • Recombinant antibody approaches:

    • Convert existing high-quality monoclonal antibodies to recombinant format

    • Sequence the variable regions of effective hybridomas

    • Express as single-chain variable fragments (scFvs) or full IgG molecules

    • Make sequences publicly available to enhance reproducibility

  • Domain-specific targeting:

    • Design antibodies against the most divergent regions of AUXI1 compared to other AUX/LAX family members

    • Target regions outside the highly conserved central permease domain

    • Create epitope-tagged versions for highly specific detection

  • Enhanced validation strategies:

    • Implement all five pillars of antibody validation (genetic, orthogonal, multiple antibodies, recombinant expression, MS)

    • Characterize in multiple assays under various conditions

    • Test across different plant species to ensure cross-reactivity where desired

  • Advanced affinity maturation:

    • Apply directed evolution to enhance specificity and affinity

    • Use yeast or phage display to screen for improved variants

    • Optimize binding kinetics for specific applications

The development of next-generation antibodies should follow the examples of programs like NeuroMab, which emphasizes transparency by providing detailed protocols and making antibody sequences publicly available . This approach supports reproducibility while addressing the limitations of traditional antibodies.

How can researchers integrate AUXI1 antibody data with other omics approaches for comprehensive auxin transport studies?

Integrating AUXI1 antibody data with other omics approaches enables systems-level understanding of auxin transport:

  • Multi-omics integration strategies:

    • Correlate AUXI1 protein levels/localization (from antibody studies) with transcriptomics data

    • Compare proteomics and antibody-based results to validate expression patterns

    • Integrate metabolomics data on auxin distribution with AUXI1 localization

    • Use phosphoproteomics to identify regulatory post-translational modifications of AUXI1

  • Spatial resolution approaches:

    • Combine cell-type specific transcriptomics with immunolocalization

    • Apply spatial proteomics techniques alongside antibody-based imaging

    • Correlate AUXI1 distribution with auxin response reporter activity

    • Map protein interactions across different cell types and developmental contexts

  • Temporal dynamics analysis:

    • Track AUXI1 levels/localization changes during development

    • Correlate with time-series transcriptomics/proteomics

    • Monitor responses to environmental stimuli over time

    • Develop mathematical models integrating temporal protein dynamics

  • Data integration tools and visualization:

    • Apply machine learning approaches to identify patterns across datasets

    • Use network analysis to position AUXI1 in the broader auxin signaling network

    • Develop interactive visualization tools that combine protein localization with omics data

    • Create predictive models of auxin transport based on integrated datasets

This integrative approach enables researchers to connect AUXI1 function to whole-plant phenotypes and developmental processes, providing a more comprehensive understanding of auxin transport mechanisms than would be possible with antibody studies alone.

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.