BSA Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Applications in Research and Diagnostics

BSA mAbs are integral to advanced biochemical techniques:

Quantitative ELISA Development

Sandwich ELISA systems employing paired mAbs (e.g., FMU-BSA NO.6 and NO.11) achieve high sensitivity:

  • Detection Limit: 0.38 μg/L (10-fold improvement over polyclonal systems).

  • Quantitative Range: 0.5–40 μg/L, with <10% intra-/inter-assay variability.

  • Use Cases: Vaccine quality control (e.g., MMR, hepatitis vaccines) and BSA quantification in biological fluids.
    Table 1: Performance Metrics of BSA mAb-Based ELISA

ParameterValueSource
Detection Limit0.38 μg/L
Linear Range0.5–40 μg/L
Cross-ValidationComparable to pAb systems

BSA Depletion and Purification

mAbs like AB-6 enable affinity chromatography to remove BSA from culture media, crucial for reducing background interference in cell culture studies.

Hapten Conjugation Strategies

BSA serves as an immunogenic carrier for hapten-specific mAb production. For example, salbutamol (SAL)-BSA conjugates induce mAbs with minimal cross-reactivity to BSA, enabling targeted drug detection.

Conjugation Table (Biotium ALB/398 mAb)

Antibody # PrefixConjugationEx/Em (nm)Detection ChannelFeatures
BNC04CF®405S404/431DAPI (Microscopy)Low fluorescence, high background
BNC88CF®488A490/515FITCHigh brightness, photostability
BNC68CF®568562/583RFPRed fluorescence
BNCBBiotinN/AN/AStreptavidin-based detection
BNUMPurified (BSA-free)N/AN/A1 mg/mL concentration
Note: Blue dyes (e.g., CF®405S) are not recommended for low-abundance targets due to signal-to-noise limitations.

Cross-Reactivity and Specificity

BSA mAbs exhibit strict specificity:

  • Species Selectivity: Minimal binding to human serum albumin or other mammalian albumins.

  • Epitope Conservation: Low-affinity mAbs may recognize conserved epitopes across BSA domains, suggesting structural homology.

Affinity Constants and Binding Kinetics

The Scatchard equation is used to determine equilibrium association constants (Ka):
[BSA]bound[BSA]free=Ka[BSA]bound+Ka[BSA]free\frac{[BSA]_{bound}}{[BSA]_{free}} = Ka \cdot [BSA]_{bound} + Ka \cdot [BSA]_{free}
High-affinity mAbs (Ka ≈ 10⁸ M⁻¹) are critical for detecting trace BSA in biological samples.

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
Our standard lead time for dispatching BSA Monoclonal Antibody is 1-3 business days following receipt of your order. Delivery times may vary depending on the mode of purchase and your location. We recommend contacting your local distributor for specific delivery estimates.

Q&A

What are BSA monoclonal antibodies and how do they differ from polyclonal antibodies?

BSA monoclonal antibodies are immunoglobulins produced by a single B-cell clone that specifically recognize and bind to epitopes on bovine serum albumin molecules. Unlike polyclonal antibodies that are derived from multiple B-cell lineages and recognize various epitopes, monoclonal antibodies originate from a single parent cell and thus bind to a specific epitope on the BSA molecule with consistent affinity and specificity. Research has demonstrated that panels of monoclonal antibodies can be developed against BSA with affinity constants ranging from 10^5 to 10^8 M^-1, indicating varying degrees of binding strength . These monoclonal antibodies can be categorized into distinct groups based on their interaction with specific domains and subdomains of the BSA molecule, allowing for precise targeting of structural features. The homogeneity of monoclonal antibodies makes them particularly valuable for experiments requiring reproducible binding characteristics and minimal batch-to-batch variation.

What are the key structural characteristics of BSA that influence monoclonal antibody recognition?

BSA is a multi-domain protein with a complex tertiary structure containing multiple potential epitopes for antibody recognition. Studies have revealed that BSA possesses similar epitopes distributed across different domains of the molecule, which explains why some low-affinity monoclonal antibodies recognize sites on different portions of the BSA structure . The domain and subdomain specificities of anti-BSA monoclonal antibodies can be determined through binding analysis with purified BSA fragments, providing insights into the structural regions that serve as antigenic determinants. Cross-reactivity patterns with various mammalian albumins further help in delineating the fine specificity of these antibodies. This structural complexity makes BSA an excellent model protein for studying antibody-antigen interactions, as it presents diverse epitope landscapes that can be recognized by different monoclonal antibodies with varying binding properties.

How does the specificity of BSA monoclonal antibodies compare to other protein-targeted antibodies?

BSA monoclonal antibodies typically demonstrate high specificity for bovine serum albumin, but may show varying degrees of cross-reactivity with albumins from other mammalian species depending on epitope conservation. Researchers have assembled BSA monoclonal antibodies into groups of non- or minimally interacting antibodies by analyzing competitive binding experiments, indicating that these antibodies recognize distinct epitopes on the BSA molecule . The specificity profiles of anti-BSA monoclonal antibodies can be further characterized through cross-reactivity studies with albumins from different species, which provides valuable information about the conservation of epitopes across evolutionary related proteins. This cross-reactivity data is particularly useful when selecting antibodies for applications where species specificity is critical, or when broader recognition across species is desired.

What are the most effective methods for producing high-affinity BSA monoclonal antibodies?

Producing high-affinity BSA monoclonal antibodies typically involves immunizing mice with BSA, followed by the fusion of splenocytes from immunized mice with mouse myeloma cells (such as SP2/0) to form hybridomas that secrete antibodies against BSA . The hybridoma technology enables the isolation and propagation of single B-cell clones that produce antibodies with specific binding characteristics. Selection of hybridomas producing high-affinity antibodies requires rigorous screening processes, often involving enzyme-linked immunosorbent assays (ELISA) or other binding assays to identify clones with desired affinity and specificity profiles. Recent advancements include rapid screening methods such as the "Ecobody technology," which employs reverse transcription PCR and Escherichia coli cell-free protein synthesis to evaluate antibodies within two working days, significantly accelerating the traditional hybridoma screening process . This approach allows researchers to quickly isolate B cells that specifically bind an antigen and generate DNA fragments of the VH and VL genes for subsequent protein synthesis and characterization.

How can researchers accurately characterize the affinity and specificity of BSA monoclonal antibodies?

Accurate characterization of BSA monoclonal antibodies involves multiple analytical techniques to assess affinity, specificity, and binding kinetics. Affinity constants can be determined using methods such as surface plasmon resonance (SPR), enzyme-linked immunosorbent assay (ELISA), or isothermal titration calorimetry (ITC), which provide quantitative measures of binding strength . Competitive binding experiments help identify antibodies that target non-overlapping epitopes, while binding studies with purified BSA fragments allow researchers to map the domain specificities of different antibodies. Cross-reactivity testing with albumins from various mammalian species provides additional insights into epitope conservation and antibody specificity. Modern analytical techniques, including chromatographic, electrophoretic, spectroscopic, and electrochemical methods, offer powerful tools for comprehensive characterization of monoclonal antibodies . For instance, reversed-phase liquid chromatography coupled with mass spectrometry (RPLC-MS) can separate antibody subdomains with specific modifications, enabling both qualitative and quantitative assessment of antibody heterogeneity.

What analytical techniques are most valuable for characterizing post-translational modifications in BSA monoclonal antibodies?

Post-translational modifications (PTMs) can significantly impact the functionality of monoclonal antibodies and require sophisticated analytical approaches for thorough characterization. Reversed-Phase Liquid Chromatography (RPLC) has demonstrated excellent resolution in evaluating protein variations arising from different chemical reactions or post-translational modifications . This technique can detect specific alterations including pyroglutamic acid formation, isomerization, deamidation, and oxidation in antibody subdomains. Ion-exchange chromatography (IEX) has been accepted as the standard method for characterizing monoclonal antibody charge variants, which are important quality parameters for stability and process consistency . Capillary electrophoresis (CE) techniques, such as capillary gel electrophoresis (CGE), capillary isoelectric focusing (cIEF), and capillary zone electrophoresis (CZE), are frequently employed for analyzing monoclonal antibodies based on charge and size heterogeneity, glycosylation profiling, and impurity analysis . Additionally, Nuclear Magnetic Resonance (NMR) spectroscopy, particularly two-dimensional NMR, provides highly specific information about the high-ordered structures of monoclonal antibodies at atomic resolution, delivering detailed structural insights.

How can BSA monoclonal antibodies be utilized for targeted drug delivery systems?

BSA monoclonal antibodies have proven valuable in developing targeted drug delivery systems, particularly for cancer therapeutics. Research has demonstrated the effectiveness of coupling anti-HER2 monoclonal antibodies to the surface of drug-loaded BSA nanoparticles for selective delivery to HER2-positive cancer cells . In one study, 1F2, an anti-HER2 monoclonal antibody, was covalently coupled to 5-Fluorouracil (5-FU) loaded BSA nanoparticles using Maleimide-poly (ethylene glycol)-Succinimidyl carbonate (Mal-PEG5000-NHS) as a crosslinker, resulting in a conjugation efficiency of 23 ± 4% . This targeting approach enabled specific interaction with HER2 receptors on the surface of HER2-positive SKBR3 cells, while showing no cellular uptake in HER2-negative MCF7 cells. The in-vitro cytotoxicity evaluation demonstrated lower SKBR3 viability (50.7 ± 9%) after 5 hours of contact with the antibody-coupled drug-loaded nanoparticles compared to control systems, highlighting the efficacy of this targeted approach . These findings emphasize the potential of BSA monoclonal antibody-conjugated delivery systems for enhancing therapeutic efficacy while reducing off-target effects in cancer treatment.

What are the advantages of using BSA as a carrier protein for developing hapten-specific monoclonal antibodies?

BSA serves as an excellent carrier protein for developing hapten-specific monoclonal antibodies due to its well-defined structure, immunogenicity, and multiple conjugation sites. Studies have demonstrated that BSA can effectively present small molecule haptens to the immune system, facilitating the generation of hapten-specific antibodies . Contrary to common misconceptions, research has shown that BSA-hapten conjugates can serve as both immunizing antigens and screening antigens during the preparation of hapten-specific monoclonal antibodies without interference from carrier-specific antibodies . In a study using salbutamol (SAL) as a model hapten, researchers successfully developed six hybridomas secreting monoclonal antibodies specific for free SAL with minimal cross-reactivity with other β-agonists, while no hybridomas secreting anti-BSA antibodies were detected . This selectivity can be attributed to the presence of abundant BSA in fetal bovine serum (FBS) used in hybridoma culture media, which saturates the binding sites of any anti-BSA antibodies produced. The high molar ratio of BSA to monoclonal antibodies in culture supernatants (approximately 5,000:1) ensures that any anti-BSA antibodies are effectively neutralized, allowing for specific screening of hapten-targeted antibodies .

How can researchers optimize the use of BSA monoclonal antibodies in immunoassay development?

Optimizing BSA monoclonal antibodies for immunoassay development requires careful consideration of antibody affinity, specificity, and potential cross-reactivity. Researchers should select antibodies with appropriate affinity constants based on the intended application, with higher affinity antibodies (e.g., 10^7-10^8 M^-1) generally preferred for detecting low-abundance analytes . Understanding the domain and subdomain specificities of the antibodies is crucial, as this knowledge allows for strategic pairing of non-competing antibodies in sandwich immunoassays. Cross-reactivity testing with related proteins helps identify potential sources of false-positive results and enables the selection of antibodies with optimal specificity profiles. The stability of antibody-conjugates should be assessed under various storage conditions, as demonstrated by studies showing that the physicochemical and biological properties of antibody-modified nanoparticles can remain stable after three months of storage at room temperature . Additionally, researchers should evaluate different immobilization strategies and detection systems to maximize assay sensitivity and specificity. Both direct and competitive immunoassay formats should be explored to determine the optimal approach for specific analyte detection, considering factors such as analyte size, concentration range, and sample matrix.

What are common challenges in BSA monoclonal antibody production and how can they be addressed?

Researchers frequently encounter several challenges during BSA monoclonal antibody production, including variable immunogenicity, hybridoma instability, and antibody degradation. The immunogenicity of BSA can vary between individual animals, potentially resulting in suboptimal immune responses and limited antibody diversity . This challenge can be addressed by modifying immunization protocols, such as adjusting adjuvant formulations, implementing prime-boost strategies, or exploring alternative routes of administration to enhance immune responses. Hybridoma stability issues, including chromosome loss and decreased antibody production, may occur during long-term culture. Implementing early-stage screening for stable clones, optimizing culture conditions, and considering cryopreservation of productive hybridomas at early passages can help maintain antibody yield and quality. Post-translational modifications and degradation reactions that occur during synthesis, formulation, and storage can significantly alter antibody structure and functionality . These variations can be minimized through careful control of production conditions, inclusion of appropriate stabilizers in formulations, and implementation of suitable storage protocols. Finally, the presence of BSA in culture media from fetal bovine serum may complicate the screening process, but this can be addressed by adjusting FBS concentrations or using BSA-depleted media during critical screening steps .

How should researchers address batch-to-batch variability in BSA monoclonal antibody production?

Batch-to-batch variability represents a significant challenge in maintaining consistent antibody performance across different production runs. Implementing robust quality control measures is essential for identifying and addressing sources of variability . Researchers should establish a comprehensive characterization panel for each batch, including affinity determination, specificity assessment, and functional validation using standardized assays. Analytical techniques such as capillary electrophoresis, ion-exchange chromatography, and size-exclusion chromatography are valuable for detecting subtle variations in antibody properties between batches . Standardizing production processes, including cell culture conditions, purification protocols, and formulation methods, helps minimize technical variability. The use of reference standards and control samples in characterization assays enables quantitative comparison between batches and facilitates the establishment of acceptance criteria for critical quality attributes. For advanced applications, implementing a quality-by-design approach that identifies critical process parameters and their acceptable ranges can significantly improve batch-to-batch consistency. Additionally, researchers should maintain detailed documentation of production parameters and analytical results to facilitate troubleshooting when variability is observed.

What strategies can be employed to minimize cross-reactivity in BSA monoclonal antibodies?

Cross-reactivity with related proteins or non-specific binding to assay components can compromise the specificity of BSA monoclonal antibodies in research applications. A systematic approach to epitope mapping and specificity testing is essential for selecting antibodies with minimal cross-reactivity . Researchers should perform comprehensive cross-reactivity testing against albumins from various species and other structurally similar proteins to identify potential sources of non-specific binding. Competitive binding experiments can help assemble groups of non- or minimally interacting antibodies, providing insights into their epitope specificity . For applications requiring absolute specificity, affinity maturation techniques or negative selection approaches during the screening process can be employed to eliminate cross-reactive clones. In assay development, optimization of blocking agents, buffer compositions, and washing procedures can significantly reduce non-specific binding. Additionally, incorporating competitive inhibition controls and sample pre-treatment steps can help identify and mitigate cross-reactivity issues. For particularly challenging applications, using combinations of antibodies targeting different epitopes can enhance specificity through the requirement for multiple binding events. Finally, validation in the intended sample matrix is crucial for detecting matrix-specific cross-reactivity that may not be apparent in standard testing conditions.

How do post-translational modifications affect the binding properties of BSA monoclonal antibodies?

Post-translational modifications (PTMs) can significantly alter the structural and functional properties of monoclonal antibodies, impacting their binding characteristics and biological activity. Research has shown that even minimal modifications and low quantities of variants can generate significant structural and biological changes in monoclonal antibodies, potentially diminishing their bioactivity . Common PTMs in monoclonal antibodies include glycosylation, deamidation, oxidation, isomerization, and pyroglutamic acid formation, each affecting antibody properties differently. Glycosylation patterns influence antibody stability, half-life, and effector functions, while deamidation and oxidation can alter the charge distribution and conformation of the antigen-binding regions. Advanced analytical techniques such as reversed-phase LC-MS methodology have been developed to separate antibody subdomains with specific alterations, enabling both qualitative and quantitative assessment of antibody heterogeneity . Researchers investigating the impact of PTMs on BSA monoclonal antibodies should employ multimodal characterization approaches, combining structural analysis with functional binding assays to establish structure-function relationships. Understanding the specific effects of different PTMs can guide the development of production and purification strategies that minimize detrimental modifications and maintain optimal antibody performance.

What are the current limitations in structural characterization of BSA monoclonal antibody-antigen complexes?

Despite significant advances in analytical techniques, the structural characterization of BSA monoclonal antibody-antigen complexes still faces several limitations. X-ray crystallography, while providing atomic-level resolution, requires successful crystallization of the antibody-antigen complex, which can be challenging due to the flexibility and glycosylation of antibodies. Cryo-electron microscopy (cryo-EM) has emerged as a powerful alternative but may still struggle with resolution for smaller epitopes or flexible regions. Nuclear Magnetic Resonance (NMR) spectroscopy offers valuable insights into antibody-antigen interactions in solution, but is limited by protein size constraints and complex spectral interpretation . Hydrogen-deuterium exchange mass spectrometry (HDX-MS) provides information about conformational changes upon antigen binding but offers lower resolution than crystallography or cryo-EM. Current methodologies for epitope mapping often provide incomplete information, particularly for conformational epitopes that depend on the tertiary structure of BSA. Additionally, characterizing transient or weak interactions between antibodies and antigens remains challenging with existing technologies. Researchers are addressing these limitations through integrated approaches combining multiple complementary techniques and developing new methodologies such as site-specific labeling and advanced mass spectrometry approaches. Computational modeling and molecular dynamics simulations are increasingly being utilized to fill gaps in experimental data and predict dynamic aspects of antibody-antigen interactions.

How can researchers leverage BSA monoclonal antibodies for developing novel biomarker detection systems?

BSA monoclonal antibodies can serve as valuable tools in developing innovative biomarker detection systems, particularly when coupled with advanced detection technologies. Researchers can exploit the well-characterized binding properties of BSA monoclonal antibodies to create standardized calibration systems for novel biosensors, ensuring consistent performance across different detection platforms. The wide range of available affinity constants (10^5 to 10^8 M^-1) allows for selection of antibodies with binding characteristics optimized for specific detection requirements . Coupling BSA monoclonal antibodies with nanomaterial-based detection systems, such as gold nanoparticles, quantum dots, or graphene, can enhance sensitivity through signal amplification strategies. The integration of BSA monoclonal antibodies into microfluidic devices enables the development of lab-on-a-chip systems for point-of-care diagnostics with minimal sample volume requirements. For multiplexed detection of multiple biomarkers, arrays of BSA monoclonal antibodies with different specificities can be immobilized on a single platform, allowing simultaneous analysis of multiple analytes. Advanced signal transduction methods, including electrochemical, optical, and mechanical approaches, can be combined with BSA monoclonal antibodies to develop highly sensitive detection systems with improved limit of detection. Finally, machine learning algorithms can be applied to analyze complex patterns in antibody binding data, potentially revealing subtle biomarker signatures that may not be apparent through conventional analysis methods.

What statistical approaches are most appropriate for analyzing BSA monoclonal antibody binding data?

Selection of appropriate statistical methods is crucial for rigorous interpretation of BSA monoclonal antibody binding data. For affinity determinations, non-linear regression analysis using models such as the Langmuir isotherm or Scatchard plot provides reliable estimates of binding parameters, including the equilibrium dissociation constant (KD) and maximum binding capacity (Bmax). When comparing multiple antibodies, statistical tests such as ANOVA followed by post-hoc analysis (e.g., Tukey's HSD) can identify significant differences in binding properties. For data from complex binding experiments, such as competitive binding assays, specialized models like the four-parameter logistic (4PL) model or the competitive binding model offer more accurate interpretations of inhibition curves and IC50 values. Time-course binding experiments should be analyzed using kinetic models that account for association and dissociation phases, providing insights into the dynamics of antibody-antigen interactions. Variability in replicate measurements should be addressed through appropriate error propagation methods and reported as standard deviation, standard error, or confidence intervals depending on the experimental design. For comparison of binding across different experimental conditions, normalization strategies may be necessary, but should be justified based on experimental controls and validation experiments. Finally, researchers should implement robust quality control measures, including outlier detection methods and validation of model assumptions, to ensure reliable interpretation of binding data.

How can researchers accurately compare the performance of different BSA monoclonal antibodies across various analytical platforms?

Comparing BSA monoclonal antibody performance across different analytical platforms requires standardized approaches to ensure valid cross-platform comparisons. Researchers should establish a panel of reference standards and controls that can be used consistently across all platforms, providing a common baseline for performance evaluation. Normalizing results to these standards helps mitigate platform-specific biases and facilitates direct comparison of antibody performance metrics. Key performance indicators (KPIs) should be defined based on the intended application, potentially including affinity, specificity, dynamic range, limit of detection, and reproducibility. These KPIs should be measured using standardized protocols for each platform, with attention to platform-specific factors that might influence results. Statistical methods for cross-platform comparison should account for differences in data structure and variability between platforms, potentially using mixed-effects models or Bayesian approaches that can incorporate platform-specific factors. Validation experiments should be designed to specifically assess platform-independent antibody characteristics, distinguishing intrinsic antibody properties from platform-associated effects. Collaborative studies involving multiple laboratories can provide valuable insights into the robustness of antibody performance across different settings and users. Finally, researchers should clearly report all experimental conditions, analytical parameters, and data processing steps to enable meaningful interpretation of performance comparisons.

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.