HA-Tag Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Structure and Epitope Design

The HA-tag is a 9-amino acid peptide (YPYDVPDYA) derived from the influenza hemagglutinin protein. Its small size (Mr ~1 kDa) ensures minimal interference with protein function, making it ideal for tagging recombinant proteins. The tag lacks intrinsic immunogenicity, requiring conjugation to carrier proteins (e.g., OVA or BSA) during antibody production .

Antibody Synthesis and Specificity

The monoclonal antibody is generated via hybridoma technology:

  • Immunization: BALB/c mice are immunized with HA-tag-OVA conjugates synthesized using carbodiimide chemistry .

  • Cloning: Hybridoma cells secreting HA-specific antibodies are screened via indirect ELISA and limiting dilution assays .

  • Purification: Ascites fluid is purified using octyl-ammonium sulfate precipitation .

The antibody exhibits high specificity for HA-tagged proteins, with no cross-reactivity to other tags (e.g., FLAG, Myc) .

Applications in Molecular Biology

ApplicationDilution RangeKey Features
Western Blotting (WB)1:1000–1:3000Detects HA-tagged proteins in lysates or transfected cells .
Immunoprecipitation (IP)1:50–1:200Efficiently pulls down tagged proteins for downstream analysis .
Immunofluorescence (IF/ICC)1:200–1:500Localizes HA-tagged proteins in fixed cells .
Flow Cytometry1:500–1:1000Quantifies surface or intracellular HA-tagged proteins .

Research Findings and Validation

  • Western Blotting: Detects HA-tagged JNK-1 in transfected cell lysates (Thermofisher) and distinguishes N-terminal/C-terminal tags (GenScript) .

  • Immunohistochemistry: Visualizes HA-tagged proteins in transfected COS/293T cells (patent) and HEK293 cells (GenScript) .

  • Flow Cytometry: Differentiates HA-transfected CHO cells from non-transfected controls (GenScript) .

Product Specs

Buffer
Phosphate Buffered Saline (PBS), pH 7.4, containing 0.02% sodium azide as a preservative and 50% glycerol.
Form
Liquid
Lead Time
Typically, we are able to ship orders within 1-3 business days of receipt. Delivery times may vary based on the purchasing method or location. For specific delivery timelines, please consult your local distributor.
Target Names
HA-Tag

Q&A

What is the HA tag and why is it commonly used in protein research?

The HA tag (hemagglutinin) is a short peptide sequence (YPYDVPDYA) derived from the human influenza hemagglutinin surface glycoprotein, corresponding to amino acids 98-106. It serves as an epitope tag that provides a method to localize gene products, study protein topology, identify associated proteins, and characterize newly identified proteins when protein-specific antibodies are unavailable .

HA tag is particularly valuable because:

  • It can be positioned at the N-terminus, C-terminus, or within the internal sequence of a protein

  • It enables consistent detection across different experimental systems

  • It facilitates protein purification and localization experiments

  • It allows for identification of low abundance or poorly immunogenic proteins

How should I choose between different HA-Tag antibody clones for my experiment?

The selection of an appropriate HA-Tag antibody clone depends on your specific application, experimental conditions, and target protein characteristics:

CloneAdvantagesBest ApplicationsSpecial Considerations
HA.C5Most widely used clone on the market, cited in >265 publicationsWB, ICC/IFExcellent for general purpose detection
12CA5Original "gold standard" cloneMultiple applicationsContext-dependent affinities reported
16B12Recognizes HA epitopes in all positions (N/C-terminus, internal)WB, ICC/IF, IP, FlowExcellent substitute for 12CA5
5B1D10Specific for both N and C-terminal tagsWBValidated with JNK-1 transfected lysates
2-2.2.14Recombinant monoclonal with better lot-to-lot consistencyMultiple applicationsRecommended for critical reproducibility

For critical experiments requiring maximum reproducibility, consider using recombinant antibodies like clone 2-2.2.14, which offer better specificity and consistency between lots .

What are the optimal conditions for using HA-Tag antibodies in Western blot applications?

For optimal Western blot results with HA-Tag monoclonal antibodies:

  • Sample preparation:

    • Use 20-50 μg of total protein lysate from transfected cells

    • Include appropriate controls (untransfected cells, empty vector)

  • Antibody dilution range:

    • Primary antibody: 1:2000-1:100000 (optimize for each clone)

    • Example: HA.C5 clone performs well at 1:2000 dilution

  • Incubation conditions:

    • Primary antibody: 4°C overnight or 1-2 hours at room temperature

    • Secondary antibody: 1 hour at room temperature

  • Detection system:

    • HRP-conjugated secondary antibodies work well with ECL detection

    • Optimize exposure time (typically 10s-1min for standard expression)

  • Troubleshooting tips:

    • For weak signals, increase antibody concentration or protein amount

    • For high background, increase blocking time or washing steps

How can I optimize immunoprecipitation of low-abundance HA-tagged proteins?

Immunoprecipitation of low-abundance HA-tagged proteins requires careful optimization:

  • Lysate preparation:

    • Use mild lysis buffers containing 1% NP-40 or Triton X-100

    • Include protease inhibitors and phosphatase inhibitors if studying phosphorylated proteins

    • For nuclear proteins, include DNase I treatment

  • Binding optimization:

    • Pre-clear lysates with protein A/G beads to reduce non-specific binding

    • For very low abundance proteins, increase incubation time (4-16 hours)

    • Use 0.5-4.0 μg antibody per 1-3 mg of total protein lysate

  • Washing conditions:

    • For stringent washing: Use buffers containing 300-500 mM NaCl

    • For gentle washing: Use buffers with 150 mM NaCl

    • Perform 4-5 washes to minimize background

  • Elution strategies:

    • Competitive elution with HA peptide (0.5-1 mg/ml) preserves antibody and native protein

    • SDS sample buffer at 95°C provides maximum recovery but denatures proteins

  • Validation approaches:

    • Confirm specificity with Western blot of input, unbound, and eluted fractions

    • Include untransfected or empty vector controls

What strategies can mitigate context-dependent recognition issues with HA-Tag antibodies?

Context-dependent recognition can occur when the antibody's access to the HA tag is hindered by protein folding or adjacent sequences. To address this issue:

  • Tag positioning strategies:

    • Test both N and C-terminal tag positions

    • Include flexible linker sequences (e.g., Gly-Ser repeats) between the tag and protein

    • For difficult proteins, consider internal tagging at exposed loops identified by structural analysis

  • Multiple tag approaches:

    • Use multimeric HA tags (e.g., 3×HA) which significantly improve affinity

      • Example: Clone 3F10 shows Kd of 0.38 nM for single HA but 0.067 nM for HAx3

    • Consider dual tagging with both HA and another tag system

  • Validation with multiple antibody clones:

    • Test recognition with different clones (e.g., 12CA5, 16B12)

    • Clone 16B12 has demonstrated ability to recognize tags in multiple contexts

  • Denaturation strategies:

    • For applications allowing denaturation, SDS treatment may improve epitope exposure

    • Consider chemical crosslinking before lysis for interacting proteins

  • Buffer optimization:

    • Test different detergents and salt concentrations

    • Addition of 0.1% SDS can improve accessibility in some contexts

How can I troubleshoot high background or non-specific binding with HA-Tag antibodies?

High background or non-specific binding can significantly impact experimental results. Methodological approaches to troubleshoot these issues include:

  • Antibody validation:

    • Always include negative controls (untransfected cells, non-tagged proteins)

    • Validate antibody specificity with known positive controls

    • Consider using highly specific recombinant antibodies

  • Blocking optimization:

    • Extend blocking time (1-2 hours at room temperature)

    • Test different blocking agents (5% BSA, 5% milk, commercial blockers)

    • For ICC/IF, include 0.1-0.3% Triton X-100 in blocking buffer

  • Washing optimization:

    • Increase number of washes (5-6 washes of 5-10 minutes each)

    • Include detergents in wash buffers (0.05-0.1% Tween-20 or 0.1% Triton X-100)

    • For flow cytometry, use excess wash buffer and multiple centrifugation steps

  • Antibody dilution and incubation:

    • Further dilute primary antibody (up to 1:100000 for some applications)

    • Reduce incubation temperature (4°C instead of room temperature)

    • Pre-adsorb antibody with cell lysates from untransfected cells

  • Secondary antibody considerations:

    • Use highly cross-adsorbed secondary antibodies

    • Ensure secondary antibody is compatible with host species of primary antibody

    • Keep secondary antibody concentrations low (typically 1:2000-1:5000)

What are the best practices for validating HA-Tag antibody specificity?

Rigorous validation of HA-Tag antibody specificity is essential for reliable research outcomes:

  • Essential controls:

    • Untransfected cells (negative control)

    • Cells transfected with empty vector (negative control)

    • Cells expressing validated HA-tagged proteins (positive control)

    • Competitive inhibition with excess HA peptide

  • Multi-technique validation:

    • Confirm specificity across multiple applications (WB, IF, IP)

    • Compare results with different HA-tag antibody clones

    • For critical experiments, validate with orthogonal approaches (e.g., MS confirmation)

  • Quantitative assessment:

    • Determine signal-to-noise ratio across antibody dilution series

    • Establish limits of detection using titrated amounts of HA-tagged protein

    • Document batch-to-batch variation when using new antibody lots

  • Systematic analyses:

    • Test for cross-reactivity with endogenous proteins in your experimental system

    • Evaluate performance across different cell types and fixation methods

    • For recombinant systems, confirm tag sequence integrity by sequencing

  • Reproducibility assessment:

    • Document protocol parameters that affect specificity and sensitivity

    • Maintain detailed records of antibody lot numbers and performance

    • Consider recombinant antibodies for improved reproducibility

How can HA-Tag antibodies be effectively used in multi-protein complex isolation?

Isolating intact protein complexes containing HA-tagged proteins requires specialized approaches:

  • Chemical crosslinking strategies:

    • Mild formaldehyde crosslinking (0.1-1%) can stabilize transient interactions

    • DSS or BS3 (membrane-permeable crosslinkers) can preserve cytoplasmic complexes

    • Photo-activatable crosslinkers offer temporal control

    • Optimize crosslinking time and concentration for your specific complex

  • Two-step purification approaches:

    • Combine HA-tag IP with size exclusion chromatography

    • Implement tandem affinity purification using dual-tagged constructs

    • Use density gradient centrifugation to separate complexes by size

  • Buffer optimization for complex stability:

    • Reduce detergent concentrations to minimum required for solubilization

    • Include stabilizing agents (glycerol 10%, reducing agents)

    • Adjust salt concentration to maintain ionic interactions

    • Consider specialized detergents for membrane protein complexes

  • Native elution methods:

    • Use competitive elution with HA peptide (0.5-1 mg/ml)

    • Optimize elution conditions (time, temperature, buffer composition)

    • For sensitive complexes, elute in multiple small fractions

  • Validation of complex integrity:

    • Confirm presence of known interaction partners by Western blot

    • Use native PAGE to verify complex size and composition

    • Combine with mass spectrometry for comprehensive interactome analysis

What are the considerations for using HA-Tag antibodies in live cell imaging applications?

Live cell imaging with HA-Tag antibodies presents unique challenges and opportunities:

  • Antibody format selection:

    • Use fluorophore-conjugated HA-Tag antibodies (e.g., Alexa Fluor® 488, PE)

    • Consider using recombinant single-chain antibody fragments (scFv) for better penetration

    • Fab fragments have reduced size compared to full IgG

  • Cell delivery methods:

    • Microinjection provides precise delivery but is low-throughput

    • Cell-penetrating peptide conjugation can enhance membrane permeability

    • Electroporation works for many cell types but requires optimization

    • Specialized commercial delivery reagents may improve efficiency

  • Surface vs. intracellular applications:

    • For surface proteins, antibodies can be added directly to media

    • For intracellular targets, membrane permeabilization is required

    • Some detergents (0.05% saponin) allow antibody entry while maintaining cell viability

  • Imaging parameters:

    • Use minimal illumination intensity to reduce phototoxicity

    • Consider oxygen scavengers in imaging media to reduce photobleaching

    • Optimize acquisition settings for temporal resolution vs. signal intensity

  • Controls and validation:

    • Include competing HA peptide controls to verify binding specificity

    • Compare to fixed cell imaging results to confirm pattern accuracy

    • Test for antibody-induced clustering or alterations in protein function

How can HA-Tag antibodies be optimized for multiplexed detection in single-cell analysis?

Advanced single-cell analysis increasingly requires simultaneous detection of multiple proteins:

  • Antibody conjugation strategies:

    • Use spectrally distinct fluorophores for flow cytometry or imaging

    • Employ metal-conjugated antibodies for mass cytometry (CyTOF)

    • Consider oligonucleotide-conjugated antibodies for CITE-seq approaches (e.g., TotalSeq™-C1131)

  • Optimization for sequential staining:

    • Establish antibody stripping/elution protocols that preserve sample integrity

    • Test different fixation methods for compatibility with multiple rounds of staining

    • Optimize order of antibody application to minimize steric hindrance

  • Panel design considerations:

    • Address spectral overlap by careful fluorophore selection

    • Test for antibody cross-reactivity in multiplexed panels

    • Include isotype controls for each conjugated antibody

  • Signal amplification methods:

    • Tyramide signal amplification for immunofluorescence

    • Branched DNA approaches for RNA-protein co-detection

    • Proximity ligation assays for validating protein interactions

  • Data analysis approaches:

    • Implement compensation matrices for spectral overlap correction

    • Use dimensionality reduction techniques (t-SNE, UMAP) for visualizing complex datasets

    • Apply clustering algorithms to identify cell populations based on marker expression patterns

What methodological adaptations are needed for using HA-Tag antibodies in organoid and tissue section analysis?

Working with complex 3D structures requires specialized approaches:

  • Tissue penetration optimization:

    • Extended incubation times (24-48 hours at 4°C)

    • Use of detergents (0.2-0.5% Triton X-100) or lipid-clearing techniques

    • Consider specialized clearing protocols (CLARITY, iDISCO) for thick specimens

    • Mechanical sectioning to optimal thickness (50-100 μm for organoids)

  • Antigen retrieval methods:

    • Heat-induced epitope retrieval (HIER) with citrate or EDTA buffers

    • Enzymatic digestion (proteinase K, trypsin) for heavily fixed samples

    • Extended washing steps to remove fixatives

  • Signal detection strategies:

    • Use of signal amplification (TSA, HRP-polymer systems)

    • Consider long-wavelength fluorophores to minimize autofluorescence interference

    • Confocal or light-sheet microscopy for 3D reconstruction

  • Quantification approaches:

    • 3D image analysis algorithms for volumetric assessment

    • Distance mapping from reference structures (e.g., vasculature, basement membrane)

    • Machine learning-based segmentation for identifying cellular compartments

  • Validation methods:

    • Parallel analysis with multiple antibody clones

    • Correlation with genetic reporters when possible

    • Systematic assessment of detection limits within thick specimens

How do HA-Tag antibodies compare with other tag systems for protein interaction studies?

Different tag systems offer distinct advantages and limitations for protein interaction studies:

Tag SystemSizeAdvantagesLimitationsBest Applications
HA Tag9 aaSmall size, high specificity antibodies, works in multiple contextsSingle epitope may have limited sensitivityGeneral purpose, widely compatible
FLAG Tag8 aaHigh specificity, calcium-dependent elution possibleSome cross-reactivity issues reportedGentle purification needs
Myc Tag10 aaSmall size, highly specific antibodies availableSome context-dependent recognitionMultiple applications
His Tag6-10 aaMetal affinity purification, no antibody needed for some applicationsPotential metal ion interference, background in some tissuesProtein purification
GST Tag211 aaHigh solubility, affinity purificationLarge size may affect protein functionImproving solubility of partners

Integration strategies:

  • Use HA tag in combination with orthogonal tags for tandem purification

  • Consider dual tagging approaches for validation across different detection systems

  • Select tag system based on experimental requirements (size constraints, elution conditions, antibody compatibility)

  • Validate that tag does not interfere with protein function or interactions

What are the methodological considerations when integrating HA-Tag antibodies with mass spectrometry-based proteomics?

Successful integration of immunoprecipitation with mass spectrometry requires careful attention to potential artifacts:

  • Sample preparation optimization:

    • Use MS-compatible detergents (e.g., RapiGest, ProteaseMAX) or ensure complete removal

    • Consider on-bead digestion to minimize sample loss

    • Implement stringent washing to reduce contaminants

    • Use filter-aided sample preparation (FASP) for sensitive samples

  • Antibody-related considerations:

    • Account for antibody-derived peptides in analysis (heavy chain ~50 kDa, light chain ~25 kDa)

    • Consider crosslinking antibody to beads to minimize contamination

    • Evaluate non-specific binders using mock immunoprecipitation controls

  • Quantitative approaches:

    • Implement SILAC, TMT, or label-free quantification for distinguishing specific interactors

    • Use intensity-based absolute quantification (iBAQ) for stoichiometry assessment

    • Compare enrichment against matched IgG controls or untransfected cells

  • Validation strategies:

    • Confirm key interactions by reciprocal IP or orthogonal methods

    • Use probability-based scoring systems (e.g., SAINT, CompPASS) to filter contaminants

    • Compare results against known interactors in public databases

  • Technical innovations:

    • Consider proximity labeling approaches (BioID, APEX) as complementary strategies

    • Implement crosslinking mass spectrometry (XL-MS) for detailed interaction sites

    • Use ion mobility separation for enhanced peptide identification in complex samples

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.