His Tag antibodies are integral to multiple laboratory techniques:
His Tag antibodies undergo rigorous testing to ensure reliability:
Reduced Cross-Reactivity: No binding to non-transfected cell lysates or untagged proteins .
Linearity and Precision: ELISA assays achieve r² > 0.99 and CV < 15% .
Multiplex Compatibility: Compatible with co-staining using target-specific antibodies without epitope interference .
Example Validation Data (R&D Systems MAB050):
Western Blot: Detected His-tagged human CIQ4 (32 kDa), MYCOC (65 kDa), and BAI-1 (120 kDa) .
Flow Cytometry: Distinct PE signal in His-tagged IGFBP6-expressing HEK293 cells vs. controls .
Non-Specific Bands: Observed in Western blots may indicate protein degradation. Mitigation includes protease inhibitors or chromatography purification .
Low Signal: Optimize permeabilization (e.g., 0.25% Triton X-100) and blocking (5% BSA) for intracellular targets .
Recent advancements include:
Monoclonal antibodies are generated by immunizing mice with a synthesized 6xHis polypeptide. Purification of this antibody is achieved through protein-A affinity chromatography.
Provided as a 1mg/ml solution in PBS (pH 7.4) containing 0.02% Sodium Azide and 10% Glycerol.
Suitable applications include ELISA and Western Blot (W.B.). Researchers are advised to determine the optimal dilutions for their specific experiments.
PAT1D6AT.
Kappa IgG1.
His tag antibodies are monoclonal or polyclonal antibodies specifically designed to recognize sequences of histidine residues (typically 6-10) fused to recombinant proteins. These antibodies function by binding to the His tag epitope regardless of the protein to which it is attached, allowing for universal detection of His-tagged fusion proteins.
The antibody's paratope specifically recognizes the spatial arrangement of histidine residues, binding with high affinity and specificity. This interaction forms the basis for various detection methods including Western blot, ELISA, immunofluorescence, and flow cytometry. Unlike protein-specific antibodies that target unique epitopes on individual proteins, His tag antibodies provide a dependable method for detecting any protein engineered to contain a His tag . This versatility makes them invaluable for confirming expression, tracking purification, and analyzing the localization of recombinant proteins.
Yes, properly designed His tag antibodies can recognize His tags regardless of their position within the fusion protein. According to experimental evidence, antibodies like THETM His Tag Antibody (mAb, Mouse) effectively recognize His tags localized at the N-terminal, C-terminal, and even internal regions of fusion proteins .
Western blot analyses specifically demonstrate this versatility:
| Tag Position | Detection Efficiency | Notes | 
|---|---|---|
| N-terminal | Excellent | Clear bands at expected molecular weight | 
| C-terminal | Excellent | Comparable signal to N-terminal tags | 
| Internal | Good | May have slightly reduced accessibility | 
This positional flexibility is confirmed through multiple experimental methods including Western blot analysis, where both N-terminal and C-terminal His-tagged fusion proteins showed clear detection as demonstrated in Figure 6 of the GenScript antibody validation studies . This characteristic makes His tag antibodies highly versatile for different construct designs.
His tags offer several distinct advantages that make them one of the most widely used epitope tags in molecular biology research:
The primary advantage of His tags is their small size (only 1 kDa for a 6×His tag), which minimizes interference with protein structure and function . Unlike larger tags such as GST (26 kDa) or MBP (42 kDa), His tags rarely affect protein folding or activity, making them suitable for functional studies.
His tags also demonstrate remarkable stability under harsh conditions, allowing for protein purification and detection under denaturing conditions . This makes them particularly valuable when working with insoluble proteins or inclusion bodies that require solubilization with strong denaturants.
Additionally, His-tagged proteins can be purified using inexpensive and widely available nickel or cobalt resins through immobilized metal affinity chromatography (IMAC) . This practical advantage contributes to the widespread adoption of His tags in research laboratories worldwide.
The optimal detection method for His-tagged proteins depends on the specific experimental context, sample complexity, and research question. Here are the most effective methods for different scenarios:
For protein purification monitoring and expression confirmation, Western blot analysis using anti-His tag antibodies provides excellent sensitivity and specificity. Research has demonstrated successful detection of His-tagged proteins in complex lysates with clear discrimination between specific signals and background . The reported sensitivity is comparable to standard SDS-PAGE methods with the advantage of specific target identification.
For rapid screening of multiple fractions (such as during chromatographic purification), newer FRET-based immunoassays offer substantial advantages. These assays use a lanthanide dye-labeled His-peptide and an acceptor-labeled anti-His tag antibody to create a mix-and-measure format with results in under two minutes . This rapid approach is particularly valuable when processing numerous samples during protein purification workflows.
For cellular localization studies, immunofluorescence using His tag antibodies has been validated for both fixed and permeabilized cells. Flow cytometry analysis using His tag antibodies has successfully detected His-tagged proteins in transfected HEK-293 cells, with clear discrimination between transfected and untransfected populations .
Optimizing Western blot protocols for His tag detection requires attention to several key parameters:
For antibody selection and dilution, use well-characterized antibodies at manufacturer-recommended concentrations. Validation studies show optimal results with antibody concentrations around 1 μg/mL for Western blot applications . Significant lot-to-lot consistency has been demonstrated for quality antibodies like THETM His Antibody (GenScript, A00186), ensuring reproducible results across experiments .
When selecting secondary antibodies, options include HRP-conjugated antibodies for chemiluminescent detection or IRDye™ 800 conjugated antibodies for fluorescent detection systems. For enhanced sensitivity, consider using rabbit monoclonal secondary antibodies like MonoRab™ Anti-Mouse IgG .
For challenging samples or low abundance proteins, incorporate these optimization strategies:
Use PVDF membranes instead of nitrocellulose for better protein retention
Extend primary antibody incubation time (overnight at 4°C)
Include 0.1% Tween-20 in blocking buffer to reduce background
Consider signal enhancement systems for low abundance targets
Blocking conditions significantly impact specificity and signal-to-noise ratio. For His tag detection, 5% BSA in TBS-T typically provides better results than milk-based blocking agents, which can contain endogenous biotin and cause background issues .
Designing robust controls is essential for reliable interpretation of His tag antibody experiments:
Positive controls should include a well-characterized recombinant protein with a His tag of the same length and position (N-terminal or C-terminal) as your protein of interest. Commercial positive controls like Multiple Tag Cell Lysate (GenScript, M0100) can provide standardized reference samples . These controls confirm antibody functionality and establish expected signal intensity.
Negative controls should include:
Untransfected/untransformed cells or lysates containing no His-tagged proteins
Samples expressing untagged versions of your protein of interest
Isotype control antibody reactions to identify non-specific binding
For immunoprecipitation experiments, parallel reactions using isotype control antibodies (e.g., A01007) are critical to distinguish specific from non-specific interactions . Flow cytometry applications benefit from non-transfected cell populations as internal negative controls, as demonstrated in validation studies using CHO cells .
When evaluating novel detection methods, direct comparison with established techniques (like Western blot) provides validation. The FRET-based immunoassay for His-tagged proteins demonstrated correlation with detectable bands in Western blot applications, confirming its reliability .
Recent innovations have expanded the toolkit for detecting His-tagged proteins beyond traditional Western blotting:
The UVHis-PAGE approach using Ni²⁺-trisNTAAlexa405 represents a significant advance in direct gel imaging. This method demonstrates high specificity for His-tagged proteins in complex samples, with no additional unspecific bands visualized even in complex mixture samples . The approach eliminates the need for transfer and immunostaining steps, significantly reducing processing time.
FRET-based immunoassays have emerged as rapid alternatives for His-tagged protein detection. These assays utilize a lanthanide dye-labeled low-affinity His-peptide and an acceptor-labeled anti-His tag antibody. When a His-tagged protein is present, it displaces the donor-labeled peptide, generating a concentration-dependent time-resolved fluorescence signal . With total assay times under two minutes including sample preparation, this approach substantially reduces workload for identifying His-tag-protein-containing fractions during purification processes.
Multiplex detection systems now allow simultaneous detection of His tags alongside other epitope tags or protein modifications. These systems enable more comprehensive characterization of recombinant proteins while conserving precious samples and reducing experimental variation.
Biolayer interferometry (BLI) binding affinity measurements provide quantitative assessments of His tag antibody interactions with tagged proteins. Validation studies demonstrate that antibodies like THETM His Tag Antibody can recognize His tags at different positions within proteins with measurable binding kinetics . This approach offers advantages for quantitative assessments of antibody performance.
Non-specific binding is a common challenge in His tag antibody applications that can complicate data interpretation. Several methodological approaches can address this issue:
For Western blot applications, optimize blocking conditions based on systematic testing. BSA-based blockers (3-5%) typically outperform milk-based blockers for His tag detection. Include 0.1-0.3% Tween-20 in washing buffers to reduce hydrophobic interactions that contribute to background.
When working with cell lysates, pre-clearing samples can significantly reduce non-specific binding. Incubate lysates with Protein A/G beads or an isotype control antibody before adding the His tag antibody. This step removes proteins that bind non-specifically to antibodies or beads.
For metal-binding proteins that might cross-react with Ni-NTA detection systems, include imidazole (10-20 mM) in binding buffers to reduce non-specific interactions. This approach is particularly important when using Ni²⁺-trisNTAAlexa405 detection methods .
Validation studies demonstrate that quality His tag antibodies like THETM His Tag Antibody show high specificity even in complex samples. When conducting immunoprecipitation from cell lysates containing His-tagged fusion proteins, properly validated antibodies show clear discrimination between specific signals and background .
The appearance of multiple bands in Western blots using His tag antibodies requires careful interpretation:
Proteolytic fragmentation is a common cause of multiple bands. His-tagged proteins may undergo partial degradation during expression or sample preparation, resulting in multiple fragments that retain the His tag. To address this, add protease inhibitors during lysis and maintain samples at 4°C throughout processing.
Post-translational modifications can alter protein mobility. Phosphorylation, glycosylation, or ubiquitination can shift the apparent molecular weight of His-tagged proteins. Compare results with phosphatase-treated samples or deglycosylation enzymes to identify modification-dependent shifts.
Alternative start codons or premature termination may generate truncated products. Examine your construct sequence for potential internal translation initiation sites. Using different His tag positions (N-terminal versus C-terminal) can help identify such events.
Protein aggregation or dimers resistant to denaturation can appear as higher molecular weight bands. Try stronger reducing conditions (increased DTT concentration) or extending the boiling time for SDS-PAGE samples.
Validation data from antibody manufacturers often include Western blot images showing expected banding patterns. For example, THETM His Tag Antibody Western blot results show consistent detection of His-tagged fusion proteins at the predicted 52 kDa marker . Comparing your results with these reference patterns can aid interpretation.
Quantitative assessment of His-tagged protein expression requires standardized approaches and appropriate controls:
For Western blot-based quantification, implement these methodological improvements:
Include serial dilutions of a purified His-tagged reference protein
Use fluorescently-labeled secondary antibodies for wider linear dynamic range
Analyze band intensity using software like ImageJ with background subtraction
Normalize to total protein (stain-free gels or housekeeping proteins)
Flow cytometry provides robust quantification of His-tagged proteins in intact cells. Studies have successfully used this approach to detect His-tagged proteins in transfected cells with clear discrimination from non-transfected controls . For absolute quantification, use calibration beads with known quantities of fluorophores.
ELISA-based methods offer high sensitivity for quantifying secreted His-tagged proteins. Design a sandwich ELISA using a capture antibody against your protein of interest and a His tag antibody for detection, or vice versa.
The rapid FRET-based immunoassay provides semiquantitative detection of His-tagged proteins with sensitivity comparable to Western blot approaches . This method is particularly valuable for rapid screening of multiple samples during chromatographic purification.
Strategic design of His-tagged constructs significantly impacts detection success:
Tag position selection should consider protein structure and function. Both N-terminal and C-terminal His tags are detectable by quality antibodies , but accessibility may differ. For membrane proteins, place the tag on the side predicted to be cytoplasmic or extracellular based on your detection needs.
Linker sequences between the His tag and protein of interest improve tag accessibility. Include a flexible linker (such as GSGSGS) to reduce steric hindrance. Validation studies show successful detection of His tags in various positions, but optimal linker design can further enhance sensitivity .
Tag length optimization balances detection sensitivity with minimal impact on protein function. While 6×His is most common, some challenging applications benefit from 8×His or 10×His tags. Longer tags provide stronger binding to both antibodies and purification resins.
Codon optimization for your expression system improves protein yields. Avoid rare codons, especially for histidine in the tag region, to prevent premature translation termination or poor expression.
Protease cleavage sites can be included between the tag and protein for tag removal after purification. Common options include TEV, PreScission, or enterokinase recognition sequences. This design allows purification and detection via the tag, followed by its removal for functional studies.
Fixation and permeabilization protocols significantly impact His tag antibody performance in immunofluorescence applications:
For cross-linking fixatives like paraformaldehyde (PFA), short fixation times (10-15 minutes) with 4% PFA minimize epitope masking while maintaining cellular structure. Validation studies demonstrate successful detection of His-tagged proteins in cells fixed with 70% ethanol for 10 minutes, suggesting alcohol-based fixatives preserve His tag epitopes effectively .
Permeabilization conditions must balance membrane disruption with epitope preservation. Gentle detergents like 0.25% Triton X-100 for 20 minutes provide effective permeabilization while preserving antibody binding sites . Saponin (0.1-0.2%) offers a milder alternative for sensitive epitopes.
Blocking conditions significantly impact signal-to-noise ratios. Extended blocking (1 hour at room temperature) with 5% BSA effectively reduces background signal in His tag immunofluorescence applications . This approach provides superior results compared to shorter blocking protocols.
The antibody incubation protocol can be optimized based on antibody properties. While some protocols recommend 3-hour incubations at room temperature , overnight incubation at 4°C may provide enhanced sensitivity for low-abundance targets.
Signal amplification systems should be considered for low expression levels. Tyramide signal amplification or biotin-streptavidin systems can enhance detection limits when expression levels are marginal.
Reconciling contradictory results between detection methods requires systematic troubleshooting and method-specific considerations:
Method sensitivity differences are common causes of discrepancies. Western blotting typically offers higher sensitivity than direct in-gel detection methods. The FRET-based immunoassay has demonstrated detection limits comparable to Western blot , while Ni²⁺-trisNTAAlexa405 in UVHis-PAGE shows high specificity but potentially different sensitivity thresholds .
Epitope accessibility variations between native and denatured conditions may cause contradictory results. His tags may be partially obscured in native proteins but fully exposed after denaturation, explaining why purification (often performed under native conditions) and detection (often performed under denaturing conditions) can yield different results.
Cross-reactivity profiles differ between antibody-based and metal affinity-based detection. While antibodies recognize the specific amino acid sequence, metal-based detection relies on coordination chemistry with histidine residues. Histidine-rich endogenous proteins may cause background in metal-based detection but not in antibody-based methods.
To systematically reconcile contradictions:
Compare detection protocols side-by-side using identical samples
Include positive and negative controls for each method
Consider protein concentration effects on detection
Evaluate sample preparation differences between methods
Assess whether contradictions represent true methodological differences or technical artifacts
Validation studies using multiple detection methods provide valuable reference points. For example, research has demonstrated correlation between FRET-based immunoassay results and Western blot detection of His-tagged proteins , establishing expected concordance between these methods.
Several emerging technologies show promise for enhancing His-tagged protein research:
Single-molecule detection methods are pushing sensitivity limits, potentially allowing visualization of individual His-tagged proteins within cellular contexts. These approaches may reveal heterogeneity in protein distribution that bulk methods overlook.
Automated high-throughput screening platforms incorporating His tag detection will accelerate protein engineering efforts. These systems can rapidly assess expression, solubility, and functionality of multiple protein variants in parallel.
Machine learning algorithms for image analysis are improving the quantification and classification of immunofluorescence data from His-tagged proteins. These computational approaches enhance reproducibility and extract more information from existing data.
Nanobody-based detection reagents specifically targeting His tags offer advantages of smaller size, better tissue penetration, and potentially improved access to sterically hindered epitopes compared to conventional antibodies.
Multiplexed detection systems allow simultaneous tracking of His-tagged proteins alongside other cellular markers. This capability enhances contextual understanding of protein function within complex biological systems.
The continued development of faster, more sensitive assays like the FRET-based immunoassay and direct detection methods like UVHis-PAGE represents an important trend toward more efficient research workflows. These approaches significantly reduce the workload associated with recombinant protein detection and characterization.
Computational approaches increasingly support His-tagged protein research at multiple levels:
Structure prediction algorithms can evaluate how His tag placement might affect protein folding and function. These tools help researchers select optimal tag positions that minimize interference with protein structure.
Expression optimization software analyzes construct sequences to identify potential expression bottlenecks, including rare codons, secondary structure in mRNA, and cryptic splice sites. These analyses improve expression yields through rational construct design.
Image analysis pipelines for immunofluorescence and Western blot quantification enhance reproducibility and sensitivity. Machine learning-based approaches can distinguish specific signals from background more effectively than traditional threshold-based methods.
Data integration platforms combine information from multiple detection methods (Western blot, mass spectrometry, immunofluorescence) to provide comprehensive characterization of His-tagged proteins. These systems help reconcile apparently contradictory results by identifying method-specific biases.
Purification simulation tools predict chromatographic behavior of His-tagged proteins based on sequence and structural features. These simulations help optimize purification protocols before experimental implementation, reducing time and resource expenditure.
As these computational resources continue to develop, they will increasingly complement experimental approaches, improving efficiency and reproducibility in His-tagged protein research.
A polyhistidine tag, commonly referred to as a His-tag, is an amino acid sequence consisting of at least six histidine residues. This tag is often used in protein purification processes due to its strong affinity for metal ions, particularly nickel. The His-tag can be added to either the N- or C-terminus of a protein, facilitating its isolation and purification through immobilized metal ion affinity chromatography (IMAC) .
The principle behind the polyhistidine tag lies in the ability of histidine residues to coordinate with metal ions. When a His-tagged protein is introduced to a carrier with immobilized metal ions, such as nickel, the histidine residues chelate the metal ions, binding the protein to the carrier. This allows for the selective isolation of the His-tagged protein, as other proteins either do not bind or bind weakly and can be washed away. The tagged protein can then be eluted from the carrier using an appropriate buffer .
Polyhistidine tags are widely used in various applications, including:
Mouse antibodies are immunoglobulins produced by mice in response to antigens. These antibodies are often used in research and therapeutic applications due to their specificity and ability to target particular antigens. However, the use of mouse antibodies in humans can lead to the development of human anti-mouse antibodies (HAMA), which can reduce the effectiveness of the treatment and cause adverse reactions .
When a polyhistidine tag is added to a mouse antibody, it allows for the efficient purification and detection of the antibody. This combination is particularly useful in research settings where the isolation of specific antibodies is required for further study or application. The His-tagged mouse antibodies can be purified using IMAC and detected using anti-His antibodies, streamlining the experimental process.