Abrin-c Antibody

Shipped with Ice Packs
In Stock

Description

Abrin-c Antibody: Definition and Context

Abrin-c antibodies are immunoglobulins designed to neutralize abrin-c by binding to its structural or functional domains, thereby preventing its toxic effects. These antibodies typically target either the A-chain (enzymatic subunit) or B-chain (lectin subunit) of abrin-c, mirroring strategies used for other abrin isoforms .

Key Features of Abrin-c Antibodies

FeatureDescription
Target SubunitA-chain (N-glycosidase activity) or B-chain (galactose-binding lectin)
MechanismNeutralizes enzymatic activity or blocks receptor binding
Cross-reactivityPotential to neutralize other abrin isoforms (a, b, d) due to shared epitopes

Antibody Development Strategies

Research on abrin antibodies has focused on isolating high-affinity neutralizers, often using hybridoma technology or humanization techniques. Below are key findings from studies relevant to abrin-c antibodies:

2.1. Humanized Antibodies

  • S008 Antibody: A humanized anti-abrin A-chain antibody derived from a mouse monoclonal antibody (10D8). It demonstrated high affinity (KD = 0.2095 nM) and neutralized abrin in vitro and in vivo. While not explicitly tested against abrin-c, its specificity for the A-chain suggests potential cross-reactivity .

  • Mechanism: S008 inhibits the enzymatic activity of the A-chain without blocking toxin internalization, as shown by fluorescence microscopy .

2.2. Monoclonal Antibodies

  • D6F10 Antibody: A mouse monoclonal antibody targeting the A-chain of abrin. It neutralized abrin toxicity in cell cultures and protected mice from lethal doses. Cross-reactivity with abrin-c is inferred due to conserved A-chain epitopes .

  • 10D8 Antibody: Exhibited sub-picomolar affinity (KD = 4.9 pM) and neutralized abrin-a by inhibiting ribosomal RNA depurination. Structural studies suggest epitopes on the A-chain are conserved across isoforms .

Epitope Mapping and Cross-reactivity

Studies mapping immunodominant epitopes on abrin subunits reveal that antibodies targeting the A-chain often recognize conserved regions critical for enzymatic activity. For abrin-c, which shares structural homology with other isoforms, these antibodies may retain neutralizing capacity .

Epitope RegionSubunitFunctionRelevance to Abrin-c
T82, G83, H85A-chainEnzymatic activity (N-glycosidase)Critical for toxin neutralization
D103, H105A-chainRibosomal RNA binding and depurinationLikely conserved across isoforms

Neutralization Assays and Efficacy

Antibodies against abrin-c (or its conserved epitopes) have shown varying degrees of success in preclinical models:

AntibodyTargetNeutralization EfficacyTherapeutic Window
S008A-chainIC50 = 0.01 μg/mL (in vitro) Up to 6 hours post-exposure
10D8A-chain54% reduction in cytotoxicity (in vitro) Pre- and post-exposure
D6F10A-chainFull neutralization in cell culture Not specified

Challenges and Future Directions

  • Isoform Specificity: No antibodies are explicitly reported to target abrin-c exclusively. Most studies focus on abrin-a/d due to their higher toxicity .

  • Cross-reactivity: Antibodies against abrin-a may neutralize abrin-c due to shared epitopes, but this requires validation .

  • Therapeutic Potential: Humanized antibodies like S008 offer a safer profile for clinical use but may face accelerated immune clearance in vivo .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Synonyms
Abrin-c [Cleaved into: Abrin-c A chain (EC 3.2.2.22) (rRNA N-glycosidase); Linker peptide; Abrin-c B chain]
Target Names
Abrin-c
Uniprot No.

Target Background

Function
Abrin-c Antibody targets the A chain of abrin, which is responsible for inhibiting protein synthesis. This occurs through the catalytic inactivation of 60S ribosomal subunits by removing adenine from position 4,324 of 28S rRNA. Abrin-a is known to be more toxic than ricin. The B chain of abrin acts as a galactose-specific lectin, facilitating the binding of abrin to the cell membrane, which precedes endocytosis.
Protein Families
Ribosome-inactivating protein family, Type 2 RIP subfamily

Q&A

What is Abrin-c and how does it differ from other abrin variants?

Abrin-c is one of several subclass toxins found in Abrus precatorius (rosary pea) seeds, with lower toxicity compared to principal variants like Abrin-a. Abrin exists as a heterogeneous mixture in natural plant extracts, containing several subclasses including Abrin-a, Abrin-b, and Abrin-c. These variants differ in their toxic potency, with Abrin-c demonstrating reduced lethality compared to Abrin-a, though all function as type-II ribosome inactivating proteins . The toxicity variation is attributed to structural differences in the binding domains, affecting cellular internalization efficiency rather than differences in the catalytic mechanisms. Researchers should note that natural abrin extracts often contain a mixture of these variants, which can influence experimental outcomes when developing neutralizing antibodies against specific subclasses .

What methodologies are available for detecting Abrin-c in experimental samples?

Several methodologies have been developed for detecting Abrin-c in experimental samples, with ELISA and electrochemiluminescence (ECL) being the most sensitive and widely used techniques. The ECL assay has demonstrated limits of detection (LODs) of 0.1 to 0.5 ng/ml for abrin isozymes in buffer, while maintaining similar sensitivity even in food samples diluted 5-fold . ELISA techniques, though slightly less sensitive in complex matrices, can detect abrin variants at LODs of 0.1 to 0.5 ng/ml in buffer solutions . For antibody-based detection, immunoassays utilizing rabbit polyclonal antibodies and mouse monoclonal antibodies have been developed against abrin isozymes. Additionally, Bio-Layer Interferometry (BLI) serves as a quantitative analysis method for assessing antibody-toxin binding interactions, using anti-human IgG Fc capture biosensors with program settings that involve washing for 60 seconds, 180 seconds association, and 300 seconds dissociation .

How can researchers evaluate antibody affinity against Abrin-c?

Researchers can evaluate antibody affinity against Abrin-c using multiple complementary techniques. Bio-Layer Interferometry (BLI) provides quantitative analysis of binding kinetics through real-time measurements of molecular interactions. The protocol involves immobilizing humanized antibodies using anti-human IgG Fc capture biosensors or murine antibodies using anti-mouse IgG Fc capture biosensors, followed by exposure to gradient concentrations of the abrin toxin (typically 200 to 3.13 nmol/L) . Additionally, Enzyme-Linked Immunosorbent Assay (ELISA) is valuable for comparative binding studies, involving coating plates with abrin (2 μg/mL), exposing them to gradient-diluted antibodies (10 μg/mL to 100 pg/mL), and detecting binding using HRP-conjugated secondary antibodies . Surface plasmon resonance offers another approach, with studies demonstrating an LOD of 35 ng/ml using human monoclonal antibody Fab E12 . Researchers should consider conducting these assays in parallel to obtain comprehensive affinity profiles before progressing to neutralization studies.

What cell-based assays are appropriate for testing anti-Abrin-c antibody neutralization capacity?

Cell-based assays using Vero and Jurkat cell lines have proven effective for evaluating anti-Abrin-c antibody neutralization capacity. The experimental approach involves co-incubating cells (5×10^5 cells/mL) with diluted abrin (using 7 gradients of 3-fold dilution starting at 100 ng/mL for Vero cells or 10 ng/mL for Jurkat cells) for 48 hours . Cellular viability is then assessed using CCK-8 solution with absorbance measured at 450 nm. For neutralization studies, researchers should first determine the toxic concentration of abrin that significantly impacts cell viability (typically 1 ng/mL for Jurkat cells and 3 ng/mL for Vero cells), then assess neutralization by incubating cells with abrin in the presence of antibodies at decreasing concentrations (from 50 μg/mL to 0) . This methodology allows for determination of EC50 values and comparative analysis of neutralization efficacy between different antibody candidates. Researchers should maintain appropriate controls, including cells without toxin exposure and cells with toxin but no antibody protection.

What are the molecular mechanisms behind anti-Abrin-c antibody neutralization?

The molecular mechanisms of anti-Abrin-c antibody neutralization are complex and do not necessarily involve preventing cellular entry of the toxin. Research with the S008 antibody demonstrated that neutralization occurs despite toxin internalization, suggesting intracellular blockade mechanisms . FACS analysis and fluorescence microscopy have confirmed that anti-abrin A chain antibodies like S008 do not prevent toxin entry but likely interfere with post-internalization events . Three primary mechanisms have been proposed: (1) binding to toxic sites on the A chain to directly inhibit ribosomal inactivation activity, (2) preventing intracellular separation of the A and B chains required for toxicity, or (3) inducing lysosomal degradation of abrin-antibody complexes . Researchers investigating these mechanisms should employ rabbit reticulocyte lysate systems containing T7 Polymerase and DNA templates to assess the inhibition of protein synthesis. By measuring luciferase activity after exposure to Abrin A chain (20 ng/mL) with or without antibodies (10 to 1 ng/mL), the IC50 values for protein synthesis inhibition can be determined .

What approaches are most effective for humanizing mouse-derived anti-Abrin-c antibodies?

Effective humanization of mouse-derived anti-Abrin-c antibodies requires sophisticated computer-aided design approaches that balance reduced immunogenicity with maintained specificity and affinity. The development of S008, a successful humanized anti-abrin antibody, illustrates a methodology involving homology modeling based on the 3-dimensional structure of the original mouse hybridoma (10D8) . This process focuses on replacing mouse-derived codon bias to achieve higher humanization scores while preserving structural stability and antigen recognition. Unlike conventional approaches that simply graft complementarity-determining regions (CDRs), comprehensive humanization addresses the framework regions to minimize human anti-mouse antibody (HAMA) reactions that otherwise lead to rapid clearance and reduced half-life . Modern bioinformatics tools allow for structure-guided modifications that retain critical binding residues while substituting non-essential regions with human equivalents. Researchers should verify humanization success through immunogenicity assessment, binding affinity comparisons (using ELISA and Bio-Layer Interferometry), and functional neutralization assays both in vitro and in vivo to ensure therapeutic potential is maintained .

How can researchers optimize in vivo models for evaluating anti-Abrin-c antibody efficacy?

Optimizing in vivo models for evaluating anti-Abrin-c antibody efficacy requires careful consideration of multiple parameters to ensure reliable and clinically relevant outcomes. Researchers should establish accurate LD50 values for the specific abrin preparation being tested, as natural variations between plant strains significantly impact toxicity . Mouse models are preferred, with toxin administration typically performed via intraperitoneal injection followed by antibody delivery through intravenous routes. To assess therapeutic potential rather than just preventative capacity, the experimental design should include delayed antibody administration (e.g., 6 hours post-toxin exposure) to simulate real-world treatment scenarios . Survival rate monitoring should extend beyond the acute phase (7-14 days) to capture delayed toxicity effects. Additionally, comprehensive organ toxicity assessment should include histopathological examination of liver, kidney, spleen, and lungs to document tissue damage and recovery patterns. Researchers must also evaluate dose-dependency by testing multiple antibody concentrations to establish minimum effective doses and therapeutic windows. Control groups should include both toxin-only exposures and administration of non-specific antibodies of the same isotype to differentiate specific neutralization from non-specific protective effects .

What cell-free systems can evaluate the inhibition of Abrin-c ribosome-inactivating activity by antibodies?

Cell-free systems provide valuable platforms for directly assessing how antibodies inhibit the ribosome-inactivating activity of Abrin-c without cellular complexity confounding results. The most effective methodology utilizes rabbit reticulocyte lysate systems supplemented with T7 RNA polymerase, reaction buffer, amino acids, RNasin ribonuclease inhibitor, and luciferase T7 DNA template . In this approach, researchers should first establish the dose-response curve for protein synthesis inhibition by adding different concentrations of purified Abrin A chain (from 1 μg/mL to 0.1 ng/mL) to the reaction system and incubating for 90 minutes at 30°C. For antibody evaluation, Abrin A chain (typically at 20 ng/mL) is pre-incubated with varying concentrations of antibodies (10 to 1 ng/mL) before addition to the lysate system . Protein synthesis is quantified by measuring luciferase activity, with positive controls containing no toxin or antibodies and negative controls containing toxin without antibodies. IC50 values determined from these assays provide direct measurement of an antibody's capacity to neutralize the catalytic activity of Abrin-c. This system is particularly valuable for testing antibodies against the A chain, as it bypasses cellular entry mechanisms and focuses exclusively on ribosomal protection .

How can researchers reconcile differences between in vitro and in vivo neutralization efficacy of anti-Abrin-c antibodies?

Reconciling differences between in vitro and in vivo neutralization efficacy of anti-Abrin-c antibodies requires systematic investigation of several potential underlying factors. The experience with the S008 antibody demonstrated that despite comparable or superior in vitro neutralizing capacity compared to its parent mouse antibody (10D8), it showed slightly weaker in vivo protection . This discrepancy likely stems from pharmacokinetic differences rather than intrinsic neutralizing ability. Researchers should conduct comparative half-life studies of antibody variants in the target organism, as the human Fc region of humanized antibodies may interact differently with mouse clearance mechanisms, potentially accelerating elimination . Tissue distribution studies using fluorescently labeled antibodies can reveal whether access to sites of toxin accumulation differs between antibody variants. Additionally, antibody-dependent cellular cytotoxicity (ADCC) potential should be evaluated, as the Fc region modifications during humanization may alter recruitment of immune effector cells that contribute to toxin clearance in vivo . These investigations should be complemented with time-course studies examining antibody concentration in serum and tissues relative to protective efficacy. Researchers should also consider developing mathematical models integrating pharmacokinetic parameters with in vitro neutralization data to better predict in vivo outcomes and optimize dosing regimens for maximal therapeutic benefit.

What are the critical quality control parameters when preparing anti-Abrin-c antibodies for research?

Critical quality control parameters for anti-Abrin-c antibody preparation include extensive characterization of purity, specificity, affinity, and functionality. Researchers must verify antibody homogeneity through SDS-PAGE under reducing and non-reducing conditions, with expected bands at approximately 150 kDa (intact antibody), 50 kDa (heavy chain), and 25 kDa (light chain) . Size exclusion chromatography should confirm monomer percentage exceeds 95% to avoid aggregation-related artifacts. Specificity assessment requires Western blot analysis against purified Abrin-c and cross-reactivity testing against other abrin variants (Abrin-a, Abrin-b) and related toxins like ricin to determine selectivity profiles. Affinity determination through Bio-Layer Interferometry should establish association (ka) and dissociation (kd) rates with expected KD values in the nanomolar to sub-nanomolar range for high-affinity antibodies . Functional validation through neutralization assays in both cell-based systems (using Vero and Jurkat cells) and cell-free rabbit reticulocyte lysate systems must demonstrate dose-dependent protection against toxin-induced inhibition of protein synthesis . For humanized antibodies, immunogenicity prediction using in silico tools should identify potential T-cell epitopes, while stability assessment through accelerated stress testing (temperature, pH, agitation) should verify structural integrity under various conditions.

How should researchers design experiments to determine the epitope specificity of anti-Abrin-c antibodies?

Designing experiments to determine epitope specificity of anti-Abrin-c antibodies requires a multi-faceted approach combining structural, biochemical, and functional analyses. Researchers should begin with competitive binding assays using a panel of characterized antibodies with known epitopes to establish whether the novel antibody competes for the same binding regions . Epitope mapping through hydrogen-deuterium exchange mass spectrometry (HDX-MS) can identify specific peptide regions protected from deuterium incorporation upon antibody binding. For higher resolution mapping, alanine scanning mutagenesis of the Abrin-c A chain should systematically replace surface-exposed residues with alanine to identify critical binding determinants when tested against the antibody . X-ray crystallography of the antibody-toxin complex provides the most definitive structural information but requires significant protein quantities and crystallization optimization. Functional correlations between epitope location and neutralization mechanisms can be established by comparing antibodies binding to different regions and their relative ability to inhibit protein synthesis in cell-free systems or prevent cytotoxicity in cellular assays. Additionally, researchers should analyze epitope conservation across abrin variants to determine whether the antibody might cross-neutralize related toxins, which has implications for broad-spectrum therapeutic potential .

What strategies can improve the therapeutic window of anti-Abrin-c antibodies for post-exposure treatment?

Improving the therapeutic window of anti-Abrin-c antibodies requires strategic modifications targeting pharmacokinetics, tissue penetration, and neutralization efficiency. The S008 antibody demonstrated effectiveness when administered up to 6 hours post-exposure, but extending this window further demands innovative approaches . Researchers should consider Fc engineering to extend half-life through increased FcRn binding, potentially incorporating the YTE or LS mutations that have demonstrated 3-4 fold half-life extensions in other therapeutic antibodies. Antibody fragment development (Fab, F(ab')2, or scFv) may improve tissue penetration and cellular entry, potentially reaching intracellular toxin more effectively, though at the cost of reduced half-life . Affinity maturation through directed evolution or structure-guided design can enhance binding kinetics, particularly reducing off-rates to maintain toxin neutralization over extended periods. Combination therapy approaches pairing antibodies targeting different epitopes may provide synergistic protection by simultaneously blocking multiple toxin functional domains. Researchers should also explore antibody-drug conjugate strategies that directly target toxin-containing cells for elimination. Formulation optimization with subcutaneous or intramuscular administration rather than intravenous delivery might provide more sustained antibody release, extending protection duration. Each approach requires systematic evaluation in appropriate animal models with delayed administration protocols that realistically simulate human exposure scenarios and treatment delays .

How should researchers interpret contradictory results between different neutralization assay formats?

Interpreting contradictory results between different neutralization assay formats requires systematic analysis of methodological differences and their biological implications. When cell-free systems indicate strong neutralization but cell-based assays show limited protection (or vice versa), researchers should consider that these assays evaluate fundamentally different aspects of toxin-antibody interactions . Cell-free systems using rabbit reticulocyte lysates directly assess inhibition of ribosome inactivation but bypass cellular entry, trafficking, and processing variables. Researchers should analyze antibody binding to different toxin domains (A vs. B chain) to determine whether neutralization occurs by preventing cellular entry (B chain targeting) or intracellular activity (A chain targeting) . Additionally, differences in antibody concentration, toxin:antibody ratios, incubation times, and temperature between assay formats can significantly impact outcomes. Standardization using reference antibodies with known neutralization profiles across all assay platforms provides essential benchmarking. Researchers should also consider performing modified assays that bridge the gap between formats, such as pre-treating cells with toxin before adding antibodies to distinguish entry-prevention from post-entry neutralization mechanisms . Finally, in vivo results should be given precedence when contradictions persist, as they integrate all aspects of toxin exposure, antibody distribution, and neutralization in the complex physiological environment most relevant to therapeutic applications.

What novel approaches could enhance the intracellular neutralization capabilities of anti-Abrin-c antibodies?

Enhancing intracellular neutralization capabilities of anti-Abrin-c antibodies requires innovative approaches targeting antibody delivery and intracellular functionality. Research with antibody S008 revealed that effective neutralization can occur without preventing toxin entry, suggesting intracellular blockade mechanisms . Future research should explore cell-penetrating peptide (CPP) conjugation to antibodies, utilizing sequences like TAT, penetratin, or transportan to facilitate cytoplasmic delivery. Engineered bispecific antibodies combining anti-Abrin-c specificity with intracellular target recognition (such as endosomal proteins) could redirect toxin trafficking away from its site of action. Researchers might also develop pH-sensitive antibodies that maintain binding in the acidic endosomal environment where toxin translocation occurs . Another promising direction involves intrabodies—antibodies expressed intracellularly through gene therapy approaches—which would position neutralizing capacity directly at the toxin's site of action. Additionally, antibody engineering to target specific intracellular compartments through localization signals could concentrate neutralizing capacity where needed most. These approaches should be evaluated systematically in cellular models with fluorescently labeled toxins to visualize trafficking patterns and neutralization sites, followed by in vivo testing to confirm therapeutic potential in physiologically relevant contexts .

How might computational approaches improve the design of next-generation anti-Abrin-c antibodies?

Computational approaches offer powerful tools for designing next-generation anti-Abrin-c antibodies with enhanced properties. Structure-based antibody design utilizing the crystal structure of abrin complexed with existing antibodies can guide rational engineering of complementarity-determining regions (CDRs) to optimize binding interfaces . Molecular dynamics simulations can predict stability changes from mutations and identify flexible regions that might benefit from rigidification to improve binding entropy. Machine learning algorithms trained on antibody-antigen interaction datasets can predict binding affinity changes from sequence modifications, accelerating the identification of promising variants without exhaustive experimental testing . Immunogenicity prediction tools that analyze T-cell epitope content can guide humanization strategies that minimize potential immune responses while maintaining neutralization efficacy. Network pharmacology approaches can model system-wide effects of antibody modifications on pharmacokinetics, tissue distribution, and toxin neutralization. For epitope optimization, computational alanine scanning and hydrogen bond network analysis can identify critical interaction residues for preservation during affinity maturation . These computational approaches should be integrated into iterative design-build-test cycles where in silico predictions guide experimental validation, with feedback loops refining models based on experimental outcomes. This integrated approach promises to accelerate development of antibodies with improved therapeutic windows, reduced immunogenicity, and enhanced intracellular neutralization capabilities.

What research is needed to develop combination antibody therapies targeting multiple abrin variants simultaneously?

Developing combination antibody therapies targeting multiple abrin variants simultaneously requires comprehensive research into epitope conservation, neutralization synergy, and formulation stability. Initial efforts should focus on comprehensive structural comparison of Abrin-a, Abrin-b, and Abrin-c to identify both conserved and variant-specific regions that could serve as antibody targets . High-throughput screening of antibody panels against all variants would identify candidates with either broad or complementary specificities. Epitope binning experiments using techniques like Bio-Layer Interferometry would categorize antibodies into groups based on competitive binding, ensuring selected combinations target non-overlapping epitopes . Synergy testing through neutralization assays with antibody combinations at varying ratios would quantify potential additive or synergistic effects, ideally identifying pairs that provide greater-than-additive protection . Researchers should also investigate potential interference between antibodies through competitive binding studies. Pharmacokinetic analysis of antibody cocktails is essential to determine whether co-administration alters clearance rates compared to individual administration. Formulation studies must address stability challenges of antibody mixtures, including potential aggregation, precipitation, or activity loss during storage. Finally, in vivo efficacy studies should test protection against purified variants and natural abrin mixtures in appropriate animal models, with comprehensive toxicokinetic monitoring to understand how antibody combinations affect toxin distribution, organ accumulation, and elimination .

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.