CHX11 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CHX11 antibody; At3g44920 antibody; F28D10_110Cation/H(+) antiporter 11 antibody; Protein CATION/H+ EXCHANGER 11 antibody; AtCHX11 antibody
Target Names
CHX11
Uniprot No.

Target Background

Function
This antibody may function as a cation/H(+) antiporter.
Database Links
Protein Families
Monovalent cation:proton antiporter 2 (CPA2) transporter (TC 2.A.37) family, CHX (TC 2.A.37.4) subfamily
Subcellular Location
Membrane; Multi-pass membrane protein.
Tissue Specificity
Specifically expressed in pollen.

Q&A

What are the fundamental principles of antibody specificity and validation?

Antibody specificity is a critical consideration in any research application. Validation should include multiple complementary techniques to confirm target binding. Key validation methods include:

  • Western blotting to confirm molecular weight specificity

  • Immunohistochemistry with appropriate positive and negative controls

  • Flow cytometry using cells with known expression levels

  • Competitive binding assays with purified antigens

How can I determine the appropriate antibody concentration for my experiments?

Determining optimal antibody concentration requires titration experiments across different application methods:

  • For flow cytometry: Prepare a dilution series (typically 0.1-10 μg/ml) and identify the concentration that provides maximum signal-to-noise ratio

  • For Western blotting: Test concentrations from 0.1-5 μg/ml to find optimal signal with minimal background

  • For immunoassays: Create a standard curve with recombinant protein to establish detection limits

The optimal concentration will vary based on:

  • Target abundance in your sample

  • Sample preparation methods

  • Detection system sensitivity

  • Binding affinity of the antibody

Data from studies with other antibodies show that concentrations yielding proper neutralization in TZM-bl assays can vary significantly, with some requiring as little as 0.02 μg/ml while others require higher concentrations .

What factors influence antibody binding affinity?

Antibody binding affinity is influenced by multiple factors that researchers should account for in experimental design:

  • Epitope accessibility in the target protein conformation

  • Buffer conditions (pH, salt concentration, detergents)

  • Temperature during incubation periods

  • Post-translational modifications of the target

  • Presence of competitive binding molecules

Research has demonstrated that some antibodies can bind to both active and inactive conformations of target proteins without inducing cellular signaling, as observed with anti-CD11d-2 antibodies . This highlights the importance of understanding conformational specificity when selecting antibodies for specific applications.

What are the optimal storage conditions for maintaining antibody functionality?

Proper storage is crucial for preserving antibody function over time:

Storage ParameterRecommended ConditionNotes
Temperature-20°C to -80°C for long-termAvoid repeated freeze-thaw cycles
Working stock4°C for up to 2 weeksAdd preservative for longer storage
Preservatives0.02-0.05% sodium azideNot compatible with HRP applications
Protein stabilizers1-5% BSA or gelatinPrevents adsorption to container
Aliquoting10-50 μl volumesMinimizes freeze-thaw damage

Research indicates that antibody stability is highly dependent on proper storage. Samples stored improperly may show decreased binding affinity and increased non-specific binding, compromising experimental results.

How should I prepare cells or tissues to maximize antibody binding while preserving epitope integrity?

Sample preparation significantly impacts antibody binding efficiency:

For cell samples:

  • Gentle fixation protocols using 2-4% paraformaldehyde preserve most epitopes

  • Permeabilization should be optimized based on the cellular location of the target

  • Blocking with appropriate sera (5-10%) reduces non-specific binding

For tissue samples:

  • Fresh frozen tissues often retain better epitope integrity than formalin-fixed samples

  • Antigen retrieval methods should be optimized based on fixation time and target location

  • Endogenous peroxidase or phosphatase activity should be blocked if using enzymatic detection

Studies with primary human leukocytes have shown that preparation methods can significantly affect detection of surface proteins like CD11d, with differences observed between total and surface-level expression analyses .

How can I design experiments to comprehensively evaluate antibody specificity across different applications?

A robust experimental design for antibody validation should include:

  • Multiple detection methods (flow cytometry, Western blot, ELISA)

  • Positive and negative control samples with known expression levels

  • Competitive inhibition with purified antigen

  • siRNA knockdown of target protein to confirm specificity

  • Comparison with alternative antibody clones targeting different epitopes

Research demonstrates that combining multiple validation methods strengthens confidence in antibody specificity. For example, in the development of humanized anti-CD11d antibodies, researchers combined flow cytometry, western blotting, immunohistochemistry, and functional assays to validate target binding .

What controls should be included when using antibodies in flow cytometry experiments?

Essential controls for flow cytometry experiments include:

  • Unstained cells to establish autofluorescence baseline

  • Isotype control matching the antibody class, species, and fluorophore

  • FMO (Fluorescence Minus One) controls for multicolor panels

  • Positive control samples with known target expression

  • Negative control samples lacking target expression

  • Single-color controls for compensation when using multiple fluorophores

Flow cytometry studies have successfully used these controls to demonstrate antibody binding to specific cell populations. For instance, humanized anti-CD11d clones were validated by showing binding to both human monocytes and neutrophils using appropriate controls .

How can antibody engineering and data mining approaches improve antibody performance for research applications?

Advanced antibody engineering utilizes computational and experimental approaches to optimize performance:

  • Sequence-based optimization: Mining public databases like Observed Antibody Space (OAS) containing over half a billion antibody sequences to identify naturally occurring antibody variations that may improve properties

  • High-throughput mutational studies: Systematically evaluate property changes upon introducing specific modifications to strengthen computational sequence-activity models

  • Hit expansion screens: Leverage promising lead antibodies to drive related optimization through:

    • Synthesis of variant panels with mutations predicted to improve developability

    • Comparative analysis of biophysical properties relative to original hits

    • Bioinformatic mining of antibody repertoires to discover related natural antibody clones with better profiles

  • Repertoire mining: Express and assay diverse naïve or synthetic repertoires related to current leads to map sequence-activity relationships and uncover shared motifs that improve key properties

These approaches can significantly accelerate the development of antibodies with improved specificity, affinity, and reduced cross-reactivity.

What methodologies can be employed to isolate novel antibodies with unique binding properties?

Advanced antibody isolation methodologies include:

  • Combinatory antibody-antigen microarray screening:

    • Enables direct screening of multiple single-chain fragment variable (scFv) clones

    • Integrates solid-phase peptide synthesis with phage display libraries

    • Allows primary screening of soluble single antibody clones followed by biophysical analysis

  • Memory B cell isolation from subjects with unique immune responses:

    • Can yield antibodies with exceptional properties, as demonstrated in HIV research

    • From one chronically infected individual (CH0219), researchers isolated antibodies from distinct clonal lineages with complementary neutralization properties

    • The combined antibodies (CH01 and VRC-CH31) achieved broad neutralization (92% of tested virus panel) representing different epitope specificities

  • Humanization of research antibodies:

    • Conversion of murine antibodies to humanized versions while maintaining specificity

    • Involves grafting complementarity-determining regions (CDRs) onto human frameworks

    • Critical for therapeutic development while providing valuable research tools

How can I investigate potential discrepancies between antibody binding and target protein functionality?

Addressing discrepancies between antibody binding and target functionality requires sophisticated experimental approaches:

  • Conformational specificity analysis:

    • Determine whether antibodies bind active, inactive, or both conformations of the target

    • Example: The humanized anti-CD11d-2 clone was shown to bind both active and inactive CD11d/CD18 conformations without inducing inflammatory cell signaling

  • Investigation of downstream signaling effects:

    • Western blotting for phosphorylation of downstream targets

    • Analysis of transcriptional responses

    • Functional cellular assays (migration, adhesion, proliferation)

  • Comparison of surface vs. total protein expression:

    • Research with CD11d/CD18 uncovered mismatches between total and surface-level expression

    • Protein kinase inhibition studies (e.g., CK2 inhibition) can help understand regulatory mechanisms

  • Epitope mapping to identify binding regions:

    • Peptide arrays or mutagenesis studies to pinpoint exact binding sites

    • Competition binding with ligands or other antibodies with known epitopes

This comprehensive analysis can help resolve seemingly contradictory results and provide deeper insights into protein biology.

What strategies can address non-specific binding in immunoassays?

Non-specific binding can significantly impact experimental outcomes. Address this issue with:

  • Optimize blocking conditions:

    • Test different blocking agents (BSA, milk, normal serum)

    • Increase blocking time or concentration

    • Use blocking agents from species different from antibody source

  • Modify washing procedures:

    • Increase number of wash steps

    • Add low concentrations of detergents (0.05-0.1% Tween-20)

    • Extend washing times

  • Adjust antibody concentration:

    • Titrate to find optimal concentration that maximizes signal-to-noise ratio

    • Consider using antibody fragments (Fab) to reduce Fc-mediated binding

  • Pre-absorb antibodies:

    • Incubate with irrelevant tissues/cells to remove cross-reactive antibodies

    • Use antigen-conjugated beads to purify specific antibodies

How can I determine if conflicting experimental results stem from antibody limitations or true biological variance?

Resolving conflicting results requires systematic investigation:

  • Antibody validation with orthogonal methods:

    • Confirm target binding using multiple techniques (Western blot, flow cytometry, immunoprecipitation)

    • Use alternative antibodies targeting different epitopes of the same protein

    • Validate with genetic approaches (knockout/knockdown controls)

  • Biological sample validation:

    • Verify cell/tissue identity and condition

    • Check for potential contamination or phenotypic drift

    • Confirm expression levels of target protein in samples

  • Experimental condition analysis:

    • Document exact buffer compositions, temperatures, and incubation times

    • Account for lot-to-lot variability in reagents

    • Consider potential interference from sample components

  • Statistical approaches:

    • Increase sample size to determine if differences are statistically significant

    • Perform power analysis to ensure adequate statistical power

    • Use appropriate statistical tests for data type

How might emerging technologies enhance antibody characterization and applications?

Emerging technologies offer new opportunities for antibody research:

  • Single-cell antibody sequencing:

    • Enables direct linking of antibody genotype with phenotype

    • Allows for high-throughput screening of natural antibody repertoires

    • Can identify rare antibodies with unique properties

  • Cryo-electron microscopy:

    • Provides atomic-level resolution of antibody-antigen complexes

    • Enables visualization of conformational epitopes

    • Facilitates structure-based antibody engineering

  • AI-driven antibody design:

    • Predicts antibody properties based on sequence

    • Identifies optimal mutations for improved affinity or specificity

    • Accelerates antibody optimization process

  • Multiplex antibody screening platforms:

    • Similar to the combinatory antibody-antigen microarray approach

    • Allows simultaneous testing of multiple antibodies against various targets

    • Increases throughput of antibody characterization

What considerations are important when developing antibodies for both research and potential therapeutic applications?

Developing dual-purpose antibodies requires balancing research utility with therapeutic potential:

  • Species cross-reactivity:

    • Evaluate binding to orthologs from multiple species for preclinical studies

    • Consider humanization strategies while maintaining research functionality

  • Epitope selection:

    • Target functionally relevant epitopes that are conserved across species

    • Ensure epitope accessibility in native protein conformation

  • Affinity optimization:

    • Optimize affinity for research applications while considering potential avidity effects in vivo

    • Evaluate on-rate and off-rate kinetics separately

  • Format flexibility:

    • Design constructs amenable to different formatting (full IgG, Fab, single-chain, etc.)

    • Consider stability in various buffer conditions relevant to both research and therapeutic use

The development of humanized anti-CD11d monoclonal antibodies demonstrates this dual-purpose approach, creating tools suitable for both basic research and therapeutic applications in conditions like neurotrauma .

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.