CRRSP26 Antibody

Shipped with Ice Packs
In Stock

Description

Absence of Primary References

None of the provided sources ( ) mention "CRRSP26 Antibody." The antibodies discussed in these studies include:

  • CR6261: Broadly neutralizing influenza antibody ( )

  • CR3022: SARS-CoV-2 antibody targeting a conserved cryptic epitope ( )

  • C105/C121: SARS-CoV-2 neutralizing antibodies with distinct epitope classes ( )

  • CC25.36: Group 1 RBD antibody with cross-reactive binding ( )

These antibodies are structurally and functionally characterized, but no nomenclature overlap with "CRRSP26" exists.

Naming Convention Issues

  • Antibody names typically follow standardized formats (e.g., CR for coronavirus-related, C for COVID-19, or CC for Caltech-Crohn’s antibodies).

  • The "CRRSP26" designation does not align with established naming systems in virology or immunology literature.

Typographical Errors

  • Possible misspellings (e.g., CR6261 vs. CRRSP26) or misinterpretation of alphanumeric codes.

Unpublished or Proprietary Research

  • The antibody might be part of ongoing, non-public research not yet indexed in academic databases.

Recommendations for Further Inquiry

To resolve this ambiguity:

  1. Verify the antibody name with original sources or collaborators.

  2. Explore recent preprints on platforms like bioRxiv or medRxiv for unpublished data.

  3. Consult structural databases:

    • PDB (Protein Data Bank) for crystallographic data

    • SAbDab (Structural Antibody Database) for annotated antibody structures

Comparative Analysis of Similar Antibodies

For reference, below is a table of structurally characterized antibodies from the provided sources that share functional or naming similarities:

AntibodyTarget PathogenEpitope ClassNeutralization MechanismKey Reference
CR6261Influenza (H1N1/H5N1)HA stemBlocks membrane fusion
CR3022SARS-CoV-2Cryptic RBDBinds silent face
C144SARS-CoV-2ACE2-competing RBDCross-links spike trimers
CC25.36SARS-CoV-2/OmicronConserved RBDYYDML motif binding

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CRRSP26 antibody; At3g21970 antibody; MZN24.14Cysteine-rich repeat secretory protein 26 antibody
Target Names
CRRSP26
Uniprot No.

Target Background

Database Links

KEGG: ath:AT3G21970

STRING: 3702.AT3G21970.1

UniGene: At.6142

Protein Families
Cysteine-rich repeat secretory protein family
Subcellular Location
Secreted.

Q&A

What methods should be used for initial characterization of CRRSP26 antibody?

Initial characterization of CRRSP26 antibody should employ multiple complementary techniques. Begin with enzyme-linked immunosorbent assay (ELISA) to confirm binding to the target antigen. Follow with western blotting to verify recognition of the target protein under denatured conditions. For higher resolution analysis, employ surface plasmon resonance (SPR) to determine binding kinetics.

Affinity measurements by SPR have proven highly effective for antibody characterization. For instance, studies of therapeutic antibodies have shown sub-nanomolar KD values for high-affinity antibodies, with measurements of both association (Kon) and dissociation (Koff) rates providing critical insights into binding behavior . Consider the following parameters as benchmarks:

MeasurementHigh-Quality Antibody RangeExample Values (from therapeutic antibodies)
KD (M)10^-9 to 10^-123.46E-11 to 5.34E-12
Kon (/Ms)10^5 to 10^78.66E+05 to 1.58E+06
Koff (/s)10^-3 to 10^-63.00E-05 to 8.44E-06

Finally, immunofluorescence or immunohistochemistry should be performed to confirm the ability to detect the target in its native cellular environment .

How can I assess CRRSP26 antibody specificity and cross-reactivity?

Assessment of CRRSP26 antibody specificity requires a multi-tiered approach. Begin with competitive binding assays using known ligands or substrates of the target protein. For thorough cross-reactivity assessment, test the antibody against structural homologs or related proteins within the same family.

Implementing mutation analysis is particularly valuable for defining epitope specificity. This approach involves creating point mutations in key amino acid residues of the target protein and measuring how these alterations affect antibody binding. Recent studies with therapeutic antibodies have demonstrated that cell-based mutation assays can effectively map epitope regions and predict cross-reactivity with variant proteins .

For example, in studies of SARS-CoV-2 neutralizing antibodies, researchers systematically tested antibody binding against spike proteins with mutations at positions W406, K417, F456, T478, E484, F486, F490, and Q493, revealing which amino acid positions constituted major epitopes for human humoral immunity .

What structural characterization techniques provide the most insightful data about CRRSP26 antibody binding mechanisms?

Cryo-electron microscopy (cryo-EM) represents the gold standard for structural characterization of antibody-antigen complexes. This technique enables visualization of the antibody-antigen interface at near-atomic resolution, revealing critical binding interactions.

The methodology involves:

  • Complex formation between purified antibody Fab fragments and target protein

  • Vitrification of samples on specialized grids

  • Image acquisition under cryogenic conditions

  • Single-particle analysis and 3D reconstruction

  • Local refinement to improve density for the Fab-antigen interface

  • Molecular modeling of variable domains bound to the target

When resolution limitations exist due to molecular flexibility, implement local refinement protocols focusing specifically on the antibody-antigen interface. For example, in studies of antibodies binding to SARS-CoV-2 spike protein, local refinement improved the density for each Fab-RBD portion, enabling more precise modeling of the variable domain interactions .

This approach allows classification of binding modes (such as the three major binding classes observed for RBD-targeting antibodies) and identification of specific contact residues that constitute the epitope .

How should I design neutralization assays to evaluate CRRSP26 antibody functionality?

Neutralization assays for CRRSP26 antibody should employ a tiered approach with increasing complexity and physiological relevance. Begin with cell-based inhibition assays measuring disruption of protein-protein interactions mediated by your target. Follow with functional assays that assess inhibition of specific cellular processes.

A successful neutralization assessment strategy includes:

  • Cell-based binding inhibition assays: Measure the antibody's capacity to block interactions between your target protein and its binding partners. For example, in SARS-CoV-2 research, Spike-ACE2 inhibition assays were used as an initial screening method .

  • Cell fusion assays: If your target mediates membrane fusion or cellular interactions, evaluate the antibody's ability to inhibit these processes. Such assays correlate well with binding inhibition assays while providing functional information .

  • Authentic system neutralization: Test the antibody in authentic biological systems, such as with native protein complexes or infectious agents. For viral targets, end-point micro-neutralization assays determine the minimum antibody concentration required for complete neutralization .

  • Validation across variants: Assess neutralization efficacy against known variants of your target protein to determine the breadth of protection and identify resistance mutations. This approach identified that E484K mutations affected multiple therapeutic antibodies, suggesting this position as a critical epitope for human immunity .

The correlation between different assay types should be evaluated to ensure robust characterization. Studies have demonstrated good correlation between cell-based inhibition assays and authentic virus neutralization, supporting the validity of the screening approach .

What modifications to the CRRSP26 antibody structure might improve its therapeutic potential?

Strategic modifications to the CRRSP26 antibody structure can significantly enhance its therapeutic potential by altering effector functions, extending half-life, or preventing adverse effects. Consider the following modifications based on therapeutic antibody development research:

  • Fc receptor engagement modifications:

    • N297A mutation: Almost eliminates binding to Fc receptors, potentially preventing antibody-dependent enhancement (ADE) of disease. This modification has been successfully implemented in therapeutic antibodies to eliminate Fc-mediated cellular uptake .

    • LALA modification: Reduces binding to Fc receptors, similar to the TM modification used in etesevimab .

    • YTE modification: Alters Fc receptor binding profiles, as implemented in AZD7442 .

  • Half-life extension strategies:

    • LS modification: Increases binding to FcRn, as used in sotrovimab to extend serum half-life .

    • PEGylation: Addition of polyethylene glycol moieties to increase molecular size and reduce renal clearance.

  • Combinatorial approaches:

    • Antibody cocktails: Combining CRRSP26 with complementary antibodies targeting different epitopes can broaden protection against variant forms of the target. This approach has demonstrated improved efficacy in macaque models compared to monotherapy .

When selecting modifications, consider the trade-offs. For example, while eliminating Fc receptor binding may prevent ADE, some studies suggest this could decrease therapeutic efficacy, though this remains controversial with contradictory findings in different models .

What animal models are most appropriate for evaluating CRRSP26 antibody efficacy?

Selection of appropriate animal models for CRRSP26 antibody evaluation should follow a progressive approach from small to larger models with increasing translational relevance. Consider the following model systems based on therapeutic antibody research experience:

  • Rodent models (mice/hamsters):

    • Advantages: Cost-effective, genetically manipulable, suitable for preliminary efficacy assessment

    • Implementation: Administer antibody (typically 50 mg/kg) intraperitoneally after establishing disease model

    • Measurements: Assess target engagement in tissues, measure disease-specific parameters, and quantify antibody levels in serum

    • Timeline: Short experimental duration (3-7 days) allows for rapid assessment

  • Non-human primate models:

    • Advantages: Higher translational value, similar physiology to humans, allows for longer studies

    • Implementation: Use lower doses (5-7 mg/kg) to better approximate human dosing

    • Measurements: Collect longitudinal samples (e.g., nasal swabs on days 1, 3, 5, 7) and terminal tissue samples

    • Special considerations: These models may show spontaneous disease resolution within approximately one week, requiring careful timing of assessments

  • Combination therapy assessment:

    • For highest translational value, evaluate the CRRSP26 antibody both as monotherapy and in combination with other treatment modalities

    • In non-human primate studies, antibody cocktails (combining equal amounts of complementary antibodies) have demonstrated superior efficacy compared to single antibodies

When designing these studies, ensure you include rigorous controls, including isotype-matched control antibodies, and collect comprehensive pharmacokinetic data including serum antibody levels and tissue distribution .

How can I determine if CRRSP26 antibody has potential as a diagnostic biomarker?

To evaluate CRRSP26 antibody as a diagnostic biomarker, implement a systematic approach comparing its performance to established biomarkers in well-characterized patient cohorts. The assessment should follow these methodological steps:

  • Cohort selection:

    • Include treatment-naïve early-stage patients to avoid therapy-induced confounding factors

    • Ensure proper classification of patients using established diagnostic criteria (e.g., American College of Rheumatology criteria for rheumatic diseases)

    • Include appropriate control groups with similar demographic characteristics

  • Isotype profiling:

    • Measure multiple antibody isotypes (IgG, IgA, IgM) as they may exhibit different diagnostic values

    • Recent studies of autoantibodies have identified distinct isotype signatures in different patient groups, with some conditions showing elevations in all three isotypes

  • Comparative analysis with established biomarkers:

    • Evaluate concordance and discordance with standard biomarkers

    • Determine the percentage of standard biomarker-negative cases that are positive for your novel marker

    • For instance, studies of Anti-CD26 autoantibodies found that 51.3% of patients who were negative for conventional markers (ACPA/RF) were positive for the novel autoantibody

  • Association with disease parameters:

    • Assess correlations with established disease activity markers (ESR, CRP)

    • Evaluate relationships with clinical disease activity scores

    • Note that weak associations may still indicate complementary diagnostic value rather than redundancy with existing markers

  • Longitudinal validation:

    • Follow patients longitudinally to determine the predictive value for disease development

    • For example, patients with unresolved undifferentiated arthritis who were negative for conventional markers but positive for novel autoantibodies may represent an important diagnostic subset

The potential value of CRRSP26 antibody as a biomarker would be particularly high if it identifies patients who are negative for conventional markers but still develop disease, addressing an important diagnostic gap .

How can I address inconsistent results in CRRSP26 antibody neutralization assays?

Inconsistent neutralization results with CRRSP26 antibody may stem from multiple sources requiring systematic troubleshooting. Implement the following methodological approaches to identify and resolve variability:

  • Validate antibody integrity:

    • Perform quality control testing of each antibody lot using ELISA and SDS-PAGE

    • Verify binding affinity using SPR to ensure consistency between experiments

    • Consider checking for aggregation using dynamic light scattering or size-exclusion chromatography

  • Standardize target protein preparation:

    • Ensure consistent expression and purification of the target protein

    • For membrane proteins, standardize detergent or reconstitution methods

    • Verify protein folding and conformation using circular dichroism or thermal shift assays

  • Examine assay-specific variables:

    • Cell-based assays: Control for passage number, confluence, and expression levels

    • For functional assays, implement positive controls with known neutralizing activity

    • Include internal normalization standards in each experiment

  • Address target heterogeneity:

    • Test against defined variants or mutants of your target protein

    • Research has shown that single amino acid mutations (e.g., E484K in SARS-CoV-2 spike) can dramatically alter neutralization potency of otherwise effective antibodies

    • Create a systematic mutation panel to identify specific sensitivity determinants

  • Employ complementary assay formats:

    • Validate results across multiple assay types (e.g., binding inhibition, cell fusion, and authentic system neutralization)

    • Studies have demonstrated good correlation between different neutralization assay formats when properly executed

Remember that true biological variability in antibody performance may reflect important insights about the target protein rather than technical issues. Systematic documentation of conditions associated with different neutralization outcomes can yield valuable mechanistic information about antibody-target interactions .

What considerations are important when developing CRRSP26 antibody therapeutics to prevent resistance development?

Development of CRRSP26 antibody therapeutics must incorporate strategies to mitigate resistance development. Implement these evidence-based approaches to create robust therapeutic candidates:

  • Epitope diversity strategy:

    • Characterize the complete epitope landscape of your target protein

    • Identify conserved epitopes less prone to mutation due to functional constraints

    • Target multiple distinct epitopes simultaneously through antibody cocktails

    • Research with therapeutic antibodies has demonstrated that combining antibodies with different neutralization profiles provides broader protection against emerging variants

  • Mutation impact prediction:

    • Perform comprehensive mutagenesis studies to identify resistance-associated mutations

    • Studies have identified that mutations at positions E484, K417, and F486 affect multiple therapeutic antibodies, highlighting these as potential resistance hotspots

    • Use cell-based assays with mutated protein variants to evaluate susceptibility profiles of candidate antibodies

  • Functional modification strategies:

    • Consider Fc modifications that optimize therapeutic effect while minimizing selective pressure

    • N297A modification has been implemented to prevent antibody-dependent enhancement while maintaining therapeutic efficacy

    • Balance improvement of antibody properties with maintenance of natural immune response mechanisms

  • Combination therapy approach:

    • Develop antibody cocktails targeting non-overlapping epitopes

    • In vivo studies in macaque models have demonstrated superior efficacy of antibody cocktails compared to monotherapy

    • Consider combining antibody therapy with complementary treatment modalities targeting different viral/pathogen lifecycle stages

  • Ongoing surveillance:

    • Implement systematic monitoring for emerging variants that might affect antibody binding

    • Maintain antibody development pipelines that can rapidly address emerging resistance

    • Research groups maintaining collections of peripheral blood samples from patients have successfully developed effective antibodies against emerging variants

The experience with therapeutic antibodies against pathogens like SARS-CoV-2 demonstrates that resistance development should be anticipated and systematically addressed through these complementary strategies .

How might advanced engineering techniques enhance CRRSP26 antibody performance?

Advanced engineering approaches can significantly enhance CRRSP26 antibody performance beyond conventional optimization. Consider these cutting-edge methodologies:

  • Structure-guided affinity maturation:

    • Utilize cryo-EM structural data to identify specific interaction sites

    • Implement computational design to predict mutations that enhance binding without disrupting framework stability

    • Focus on complementarity-determining regions (CDRs) while maintaining germline framework residues for stability

    • Research has demonstrated that antibodies with KD values in the picomolar range (10^-12 M) can be achieved through systematic engineering

  • Domain fusion strategies:

    • Create bispecific antibodies by linking CRRSP26 binding domains with complementary specificities

    • Develop antibody-cytokine fusions for targeted immune modulation

    • Consider antibody-drug conjugates for targeted delivery of therapeutic payloads

    • These approaches have shown promise in extending the therapeutic range of single antibodies

  • Fc engineering beyond conventional modifications:

    • Apply glycoengineering to fine-tune Fc receptor engagement profiles

    • Consider hexamerization-enhancing modifications for complement activation

    • Implement pH-dependent binding modifications for improved recycling and tissue penetration

    • Research has demonstrated that strategic Fc modifications can significantly alter therapeutic profiles without affecting antigen binding

  • Stability enhancement techniques:

    • Implement machine learning algorithms to identify destabilizing residues

    • Introduce stabilizing disulfide bonds without affecting binding interface

    • Apply camelization or humanization techniques to reduce immunogenicity while maintaining function

    • These approaches can extend shelf-life and in vivo half-life of therapeutic antibodies

  • Alternative scaffold integration:

    • Consider incorporating CRRSP26-binding domains into alternative binding scaffolds

    • Explore nanobody or single-domain antibody formats for enhanced tissue penetration

    • Investigate non-immunoglobulin scaffolds with similar binding properties but different biophysical characteristics

By implementing these advanced engineering approaches, CRRSP26 antibody can be optimized beyond conventional parameters to address specific therapeutic or diagnostic challenges .

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.