HA-33 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to HA-33 Antibody

HA-33 Antibody refers to immunoglobulins targeting the hemagglutinin-33 (HA-33) protein, a key component of botulinum neurotoxin (BoNT) complexes. HA-33 is critical for protecting BoNT from gastrointestinal proteolysis, enhancing toxin stability, and facilitating interactions with host cells . Antibodies against HA-33 are researched for diagnostic applications, vaccine development, and toxin neutralization strategies.

Structure and Function of HA-33

HA-33 adopts a β-trefoil fold, a structure shared with other neurotoxin-associated proteins, enabling carbohydrate-binding activity . Key functional roles include:

  • Protection of BoNT: Resists proteolysis in acidic environments, preserving neurotoxin integrity during gastrointestinal transit .

  • Adjuvant Properties: Enhances immune responses, making it a candidate for vaccine formulations .

  • Cellular Binding: Interacts with sialic acid-containing glycolipids/glycoproteins on intestinal epithelial cells, aiding toxin internalization .

Table 1: Key Properties of HA-33

PropertyDescriptionSource
Molecular Weight~33 kDa
Structural Foldβ-Trefoil with carbohydrate-binding domain
Immunogenic RoleElicits high antibody titers; major immunogenic component of BoNT complexes

Recombinant Production and Antigenicity

HA-33 antibodies are generated via recombinant expression systems:

  • Expression in E. coli: pET28a vectors yield ~33 kDa HA-33 with >85% purity (SDS-PAGE) .

  • Antigenicity: Recombinant HA-33 induces high antibody titers (1:128,000 dilution) in mice, validated via ELISA .

Antibody Engineering and Affinity Optimization

Thermostabilization strategies improve antibody durability:

  • Disulfide Bonds: Introducing L12C/K104C in IgG1 heavy chains enhances CH2 domain stability .

  • Affinity Maturation: Mutations (e.g., H35N, A53L, Q64R in heavy chain; N50D, G66E in light chain) reduce equilibrium dissociation constant (K<sub>D</sub>) to 30–45 pM .

Table 2: Engineered HA-33 Antibody Variants

Antibody VariantKey MutationsK<sub>D</sub> (pM)T<sub>m</sub> (°C)ΔT<sub>m</sub>Source
APE1553H35N, A53L, Q64R (HC); N50D, G66E (LC)3088.2−3.9
APE1854G66E removed from APE15534592.0+3.9

Immunoassays for Toxin Detection

  • Sandwich ELISA: Recombinant α-HA33 antibodies detect both HA-33 and BoNT/A toxoid with high sensitivity .

  • Western Blotting: Polyclonal antibodies confirm HA-33 expression in E. coli lysates .

Therapeutic Potential

While not yet clinical, HA-33 antibodies may:

  • Neutralize Toxins: Prevent HA-33-mediated toxin stabilization or cellular binding .

  • Enhance Vaccines: Serve as adjuvants in oral vaccines targeting BoNT .

Challenges and Future Directions

  • Cross-Reactivity: Limited data on antibody specificity across BoNT serotypes.

  • Scalability: Recombinant HA-33 production requires optimization for high-yield fermentation .

  • Thermostability: Further engineering to enhance antibody durability for field applications .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders for HA-33 Antibody within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. For specific delivery time estimates, please consult your local distributor.
Target Names
HA-33
Uniprot No.

Q&A

What is HA-33 and what is its role in the Botulinum neurotoxin complex?

HA-33 (Hemagglutinin-33) is a non-toxic protein component of the Botulinum neurotoxin (BoNT) complex, specifically associated with BoNT type A (BoNT/A). This approximately 33 kDa protein serves multiple critical functions within the toxin complex. HA-33 contributes significantly to the toxin complex's resistance to proteases and acidic pH conditions, effectively protecting the neurotoxin during passage through the harsh gastrointestinal environment. Beyond its protective role, HA-33 also facilitates the absorption of the toxin via intestinal epithelial cells and may enhance the endopeptidase activity of the toxin itself. Research has additionally suggested that HA-33 may function as an adjuvant in vaccinology applications, further expanding its research significance .

What structural and functional characteristics make HA-33 a valuable research target?

HA-33 possesses several distinctive properties that make it particularly valuable for research. The protein exhibits remarkable resistance to proteolytic degradation and pH extremes, making it stable under experimental conditions that would denature many other proteins. This stability appears to be conferred through its structural configuration, which enables it to protect the neurotoxin component of the BoNT complex from degradation in the gastrointestinal tract. Functionally, HA-33 contributes to the toxin's ability to cross epithelial barriers, a mechanism critical for botulinum pathogenesis. These characteristics make HA-33 a compelling target for both basic research into botulinum toxicity and applied research in vaccinology, where its adjuvant properties and role in oral vaccine formulations are being explored .

How can researchers distinguish between antibodies targeting HA-33 and other components of the botulinum toxin complex?

Distinguishing antibodies that specifically target HA-33 from those targeting other components of the botulinum toxin complex requires careful experimental design incorporating multiple validation techniques. The most effective approach employs immunoblotting against purified recombinant HA-33 protein alongside other isolated components of the BoNT complex. Researchers should conduct competition assays in which antibodies are evaluated for their ability to recognize HA-33 in the presence of other purified components of the toxin complex. Additionally, researchers can employ epitope mapping techniques to precisely identify the binding regions of antibodies, confirming HA-33 specificity. Cross-reactivity testing against other hemagglutinin components (such as HA-17, HA-55, and HA-70) is essential to establish specificity, as these proteins share some structural similarities with HA-33 .

What expression systems are most effective for recombinant production of HA-33?

For recombinant HA-33 production, prokaryotic expression systems utilizing E. coli have demonstrated considerable success. Specifically, the pET28a(+) vector system with T7 promoter in E. coli BL21 (DE3) strain has proven highly effective for high-yield expression. This system benefits from strong promoter control and has been optimized for HA-33 expression using parameters of 1 mM IPTG induction, 37°C incubation temperature, and 5-hour induction time with shaking at 150 rpm. When preparing expression constructs, researchers should consider codon optimization, particularly when expressing Clostridium genes (which are AT-rich) in E. coli hosts. This optimization process involves adjusting the codon usage pattern, GC content, and Codon Adaptation Index to remove rare codons, significantly improving expression efficiency. The incorporation of affinity tags, such as His-tags, greatly facilitates subsequent purification processes .

What purification strategies yield the highest purity HA-33 protein for antibody development?

Purification of recombinant HA-33 protein for antibody development requires strategies that maximize both yield and purity. For His-tagged recombinant HA-33, nickel nitrilotriacetic acid (Ni-NTA) agarose affinity chromatography has proven highly effective. The optimal purification protocol begins with bacterial lysis by sonication (typically 4 cycles of 10 seconds with 30-second intervals at 75% amplitude), followed by centrifugation at 18,000 ×g at 4°C for 20 minutes to remove insoluble debris. For purification buffers, researchers should employ a gradient elution strategy with increasing imidazole concentrations. Maximum elution of high-purity HA-33 typically occurs with 250 mM imidazole in MES buffer. For especially challenging purifications, researchers may consider a two-phase approach combining Ni-NTA affinity chromatography with size exclusion or ion-exchange chromatography to achieve exceptionally high purity. Quality assessment by SDS-PAGE should reveal a distinct ~33 kDa band corresponding to the target protein, with confirmatory Western blotting using anti-BoNT/A serum providing validation of identity .

How can researchers optimize recombinant HA-33 expression to achieve maximum yield?

Optimizing recombinant HA-33 expression requires systematic evaluation of multiple parameters to achieve maximum yield while maintaining protein functionality. Critical factors include induction timing, inducer concentration, expression temperature, and incubation duration. Experimental data indicates that optimal conditions for E. coli BL21(DE3) with pET28a(+) vector include induction at OD595 of 0.5, using 1 mM IPTG with incubation at 37°C for 5 hours under constant shaking at 150 rpm. Temperature optimization is particularly important; while higher temperatures (37°C) can increase expression rates, they may also lead to inclusion body formation. Researchers should evaluate expression at different temperatures (25°C, 30°C, and 37°C) and determine the optimal balance between yield and solubility. Additionally, media composition can significantly impact expression levels, with supplemented media often yielding higher protein concentrations. Following optimization, researchers can expect yields of approximately 10 mg/L of purified HA-33 protein from culture suspensions .

What methodologies are most effective for assessing anti-HA33 antibody titers?

Enzyme-Linked Immunosorbent Assay (ELISA) remains the gold standard for quantitatively assessing anti-HA33 antibody titers. For optimal results, researchers should coat ELISA plates with 3.5 μg of purified recombinant HA-33 protein per well in carbonate-bicarbonate buffer (15 mM Na₂CO₃ and 36 mM NaHCO₃, pH 9.8) and allow overnight adsorption at 4°C. Blocking should be performed with 5% skimmed milk solution to minimize non-specific binding. Serial dilutions of test sera should begin at 1:1,000 and extend to at least 1:128,000 to accurately capture the full range of antibody responses. For detection, anti-species IgG-HRP conjugates at 1:2,000 dilution with orthophenyl-enediamine and H₂O₂ as substrate provide reliable colorimetric results. Experimental evidence shows that properly immunized subjects can produce detectable antibody responses at dilutions exceeding 1:128,000, indicating the high immunogenicity of recombinant HA-33. To ensure assay accuracy, researchers should include appropriate controls, including pre-immune sera and sera from subjects immunized with adjuvant only .

What approaches can be used to develop thermostable anti-HA33 antibodies?

Development of thermostable anti-HA33 antibodies can be achieved through a multifaceted engineering approach combining complementarity-determining region (CDR) grafting, mammalian cell display, and in vitro somatic hypermutation (SHM). The process begins with identifying a thermostable human IgG framework that can serve as a stable scaffold. CDR regions from existing anti-HA33 antibodies are then grafted onto this framework, preserving the specificity of the original antibody while enhancing stability. The mammalian cell display platform allows for screening of variant antibodies under stringent thermostability conditions, identifying clones that maintain structural integrity and binding capacity at elevated temperatures. In vitro somatic hypermutation can be employed to further enhance both affinity and thermostability through directed evolution. This combined approach has demonstrated success in improving the thermostability of anti-HA33 antibodies by approximately 10°C while simultaneously enhancing binding affinity by approximately 300-fold over the original antibody .

How should researchers evaluate the functional activity of anti-HA33 antibodies?

Comprehensive evaluation of anti-HA33 antibody functional activity requires multiple complementary assays addressing different aspects of antibody performance. Beyond simple binding assays, researchers should assess the ability of antibodies to neutralize the protective effect of HA-33 on botulinum neurotoxin in simulated gastrointestinal conditions. This can be accomplished by exposing BoNT/A complex (with and without anti-HA33 antibody pre-treatment) to acidic conditions and proteolytic enzymes, followed by assessment of remaining toxin activity. Additionally, epithelial cell translocation assays using polarized Caco-2 monolayers provide valuable insights into whether anti-HA33 antibodies can block the toxin-facilitating function of HA-33 at the intestinal barrier. For antibodies intended for therapeutic applications, researchers should evaluate dose-dependent protection in mouse botulism models, comparing pre- and post-exposure efficacy. Combination studies with anti-neurotoxin antibodies can reveal potential synergistic protective effects, which may have significant therapeutic implications .

How can researchers effectively study the adjuvant properties of HA-33?

Investigating the adjuvant properties of HA-33 requires systematic experimental design and multiple immunological readouts. Researchers should begin by comparing immune responses to model antigens with and without co-administration of purified recombinant HA-33. A comprehensive immunological assessment includes measuring antibody titers through ELISA, analyzing antibody isotype distribution to evaluate Th1/Th2 balance, and characterizing memory B-cell responses through ELISPOT assays. The adjuvant effect on cellular immunity should be assessed by evaluating antigen-specific T-cell responses via flow cytometry and cytokine profiling. Dose-response studies are essential to establish the optimal HA-33 concentration for maximum adjuvant effect while maintaining safety. For mechanistic understanding, researchers should investigate the interaction of HA-33 with antigen-presenting cells, particularly dendritic cells, examining changes in surface markers, cytokine production, and antigen presentation efficiency. Additionally, structural modifications of HA-33 through site-directed mutagenesis can help identify specific domains responsible for the adjuvant activity .

What are the challenges and solutions in isolating high-purity HA-33 from botulinum toxin complexes versus recombinant production?

Isolating high-purity HA-33 directly from botulinum toxin complexes presents significant challenges compared to recombinant production approaches. Direct isolation from toxin complexes typically requires multiple purification steps including ammonium sulfate precipitation, various chromatography techniques (often employing mild denaturants such as guanidine hydrochloride or urea/lithium chloride), and gel filtration columns. Despite these extensive procedures, many attempts have failed to achieve complete separation of individual HA components, particularly HA-17, which tends to irreversibly precipitate during purification. Furthermore, direct isolation necessitates large-scale cultivation of Clostridium botulinum, introducing significant biosafety concerns.

In contrast, recombinant production offers several advantages, including higher yield, greater purity, and elimination of biosafety risks associated with toxin-producing bacteria. The recombinant approach using optimized expression systems with affinity tags enables straightforward single-step purification protocols. Additionally, recombinant production allows for site-directed mutagenesis to study structure-function relationships and create modified variants with enhanced properties. Given these considerations, recombinant production is generally preferred for research applications requiring high-purity HA-33, particularly when structural or immunological studies demand homogeneous protein preparations .

What methodologies are appropriate for studying HA-33's role in epithelial cell translocation of botulinum toxin?

Investigating HA-33's role in epithelial cell translocation of botulinum toxin requires sophisticated experimental approaches that model the intestinal barrier while allowing quantitative assessment of toxin transport. Polarized intestinal epithelial cell monolayers, particularly Caco-2 cells grown on permeable Transwell inserts, provide an excellent in vitro model system. Researchers should confirm monolayer integrity through transepithelial electrical resistance (TEER) measurements before conducting translocation experiments. The experimental design should compare translocation of purified neurotoxin alone versus neurotoxin complexed with HA-33, with both apical-to-basolateral and basolateral-to-apical transport assessed to determine directionality.

For mechanistic insights, researchers can employ pharmacological inhibitors targeting different cellular uptake pathways (e.g., clathrin-dependent endocytosis, lipid raft-mediated uptake) to identify the specific mechanisms facilitated by HA-33. Confocal microscopy with fluorescently labeled toxin components can visualize the trafficking pathway, while co-immunoprecipitation and proximity ligation assays can identify potential cellular receptors interacting with HA-33. To further validate findings, researchers should develop HA-33 knockout or mutant variants of the toxin complex and assess their translocation efficiency compared to wild-type complexes. These combined approaches can comprehensively characterize the molecular mechanisms underlying HA-33's role in toxin absorption across the intestinal epithelium .

How should researchers design experiments to evaluate HA-33 antibody cross-reactivity with other hemagglutinin components?

Designing robust experiments to evaluate cross-reactivity of HA-33 antibodies with other hemagglutinin components requires comprehensive antigen panels and multiple analytical techniques. Researchers should prepare a complete panel of purified recombinant hemagglutinin proteins, including HA-33, HA-17, HA-55, and HA-70 from the same and different botulinum serotypes. Initial screening should employ ELISA with equivalent coating concentrations of each protein to quantitatively assess relative binding affinities. Western blotting under both reducing and non-reducing conditions provides important information about epitope nature (conformational versus linear) and potential cross-reactivity patterns.

For more detailed characterization, researchers should perform competition assays in which unlabeled hemagglutinin proteins are used to inhibit antibody binding to immobilized HA-33. Surface plasmon resonance (SPR) or bio-layer interferometry (BLI) enable precise determination of binding kinetics and affinities for each potential cross-reactant. To establish biological relevance of any observed cross-reactivity, functional assays measuring the antibody's ability to neutralize the specific activities of different hemagglutinin components should be conducted. Finally, epitope mapping through techniques such as hydrogen-deuterium exchange mass spectrometry or peptide arrays can definitively identify the specific binding regions and explain observed cross-reactivity patterns based on sequence or structural homology between hemagglutinin components .

What statistical approaches are most appropriate for analyzing anti-HA33 antibody response data?

Statistical analysis of anti-HA33 antibody response data requires carefully selected approaches that appropriately address the typical distribution characteristics and experimental designs in immunological studies. For antibody titer data, which typically follows a log-normal distribution, log-transformation should be applied before parametric statistical tests. When comparing antibody responses between different treatment groups or time points, repeated measures ANOVA with appropriate post-hoc tests (such as Tukey's or Bonferroni) should be employed to account for multiple comparisons while controlling the familywise error rate.

For dose-response studies, nonlinear regression models should be applied to determine EC50 values and maximum response parameters. When evaluating the correlation between antibody titers and protective efficacy in challenge models, researchers should consider both Pearson's correlation (for linear relationships) and Spearman's rank correlation (for monotonic but potentially non-linear relationships). Survival data from protection studies requires Kaplan-Meier analysis with log-rank tests for comparing groups. For complex experimental designs incorporating multiple variables, mixed-effects models provide robust analysis while accounting for both fixed effects (e.g., dose, adjuvant presence) and random effects (e.g., individual variation, litter effects). Sample size calculations for immunization studies should be based on preliminary data, with power analysis ensuring sufficient statistical power (typically 0.8 or greater) to detect biologically meaningful differences .

How can researchers integrate computational approaches with experimental data to optimize anti-HA33 antibody design?

Integrating computational approaches with experimental data represents a powerful strategy for optimizing anti-HA33 antibody design. The process should begin with computational structural analysis of HA-33 to identify potential epitopes based on surface accessibility, hydrophilicity, and predicted antigenicity. Homology modeling can generate structural models of antibody candidates, which can then undergo molecular docking simulations with HA-33 to predict binding modes and interaction energies. Molecular dynamics simulations offer insights into the stability of antibody-antigen complexes under various conditions, including elevated temperatures for thermostability assessment.

Machine learning algorithms can analyze experimental data from antibody panels to identify sequence and structural features associated with desirable properties such as high affinity, specificity, and stability. These insights can guide the design of focused antibody libraries for experimental screening. For affinity maturation, computational design of targeted mutations in the complementarity-determining regions (CDRs) based on energy calculations can significantly accelerate the optimization process. Importantly, computational predictions should be iteratively validated through experimental testing, with the resulting data used to refine computational models in a continuous improvement cycle.

This integrated approach has demonstrated success in antibody engineering, as evidenced by achievements in thermostabilization of anti-HA33 antibodies while simultaneously enhancing binding affinity. By combining computational design with experimental validation, researchers can navigate the vast sequence space more efficiently, dramatically reducing the time and resources required to develop optimized anti-HA33 antibodies for research and potential therapeutic applications .

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.