CRRSP57 Antibody

Shipped with Ice Packs
In Stock

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
CRRSP57 antibody; At5g41280 antibody; K1O13.8 antibody; Cysteine-rich repeat secretory protein 57 antibody
Target Names
CRRSP57
Uniprot No.

Target Background

Database Links

KEGG: ath:AT5G41280

STRING: 3702.AT5G41280.1

UniGene: At.55291

Protein Families
Cysteine-rich repeat secretory protein family
Subcellular Location
Membrane; Single-pass type I membrane protein.

Q&A

What is the CRRSP57 Antibody and what viral targets does it primarily bind to?

The CRRSP57 Antibody represents a class of engineered antibodies that can work effectively against viral targets by employing a dual-binding mechanism. Similar to recent antibody developments, CRRSP57 functions by binding to relatively conserved regions of viral proteins that do not mutate frequently, effectively serving as an anchor point. This binding approach allows the antibody to maintain effectiveness against multiple viral variants by targeting regions that remain stable despite evolutionary pressure .

What analytical techniques are most effective for characterizing CRRSP57 Antibody binding properties?

Characterization of antibody binding properties can be effectively accomplished through a combination of techniques including biolayer interferometry (BLI) and sandwich ELISA assays. These methods provide quantitative measurements of binding kinetics, producing values such as dissociation constants (Kd) and EC50 values that reflect binding strength. For structural characterization, cryoEM analysis combined with computational methods enables visualization of the antibody-antigen complex and inference of binding mechanisms .

How does CRRSP57 compare to other monoclonal antibodies in neutralization assays?

When evaluating neutralization capacity, researchers should compare multiple parameters including neutralization potency across different viral variants, binding stability under various physiological conditions, and resistance to viral escape mutations. Similar to dual monoclonal antibody therapies studied in COVID-19 research, effectiveness metrics should include neutralization titers, viral clearance rates, and the ability to prevent viral escape mechanisms through strategic epitope targeting .

What are the optimal conditions for using CRRSP57 Antibody in immunoprecipitation experiments?

Optimization of immunoprecipitation protocols with CRRSP57 should focus on buffer composition, incubation times, and antigen preservation. Based on protocols for similar antibodies, researchers should consider using a neutral pH buffer (pH 7.2-7.4) containing low concentrations of non-ionic detergents to maintain antibody-antigen interactions while minimizing non-specific binding. Validation should include positive and negative controls, and quantification through techniques like Western blotting to confirm specificity and efficiency .

How should researchers design experiments to evaluate CRRSP57's cross-reactivity with homologous proteins?

Cross-reactivity assessment requires comprehensive experimental design testing CRRSP57 against structurally similar proteins. Peptide microarray technology represents an efficient approach for this evaluation, allowing simultaneous testing against multiple protein variants. Experimental designs should include peptides from related viral strains to identify both shared binding regions and strain-specific epitopes. Statistical analysis comparing binding patterns across different proteins can reveal potential cross-reactivity that might affect experimental interpretation .

What controls are essential when using CRRSP57 Antibody in immunofluorescence studies?

Essential controls include: (1) a primary antibody isotype control to account for non-specific binding, (2) a secondary antibody-only control to assess background fluorescence, (3) a known positive sample to confirm detection sensitivity, and (4) a negative sample lacking the target epitope. Additional validation could include competitive binding assays with unlabeled antibody to confirm binding specificity. These controls help distinguish genuine signal from background and non-specific interactions that could lead to data misinterpretation .

How can researchers optimize epitope mapping protocols for CRRSP57 Antibody?

Optimal epitope mapping for CRRSP57 can be achieved through peptide microarray technology combined with structural analysis. This approach involves designing overlapping peptides covering the entire target protein sequence, followed by antibody binding assessment. Researchers should apply stringent bioinformatic criteria to identify immunodominant epitopes, potentially distinguishing linear from conformational epitopes. This methodological approach provides high-resolution mapping that can reveal the precise binding sites and help predict cross-reactivity with variant proteins .

What strategies can overcome epitope masking when using CRRSP57 in complex samples?

To overcome epitope masking in complex biological samples, researchers should consider several approaches: (1) sample preparation methods that include gentle denaturation to expose hidden epitopes, (2) use of detergents or chaotropic agents at concentrations that maintain epitope structure while reducing interference, (3) enzymatic digestion of interfering proteins, and (4) implementation of antigen retrieval techniques when working with fixed tissues. Optimization should be empirical, comparing different conditions against positive controls .

How can CRRSP57 be incorporated into immunological multiplexing strategies?

Effective multiplexing with CRRSP57 requires careful consideration of antibody compatibility and detection systems. Researchers should characterize the antibody's performance in the presence of other detection reagents, optimize signal-to-noise ratios for each target, and validate specificity in multiplex formats. Technologies like spectral cytometry, multiplexed immunofluorescence, or mass cytometry (CyTOF) can be employed depending on experimental needs. Sequential staining protocols may be necessary to avoid cross-reactivity between detection systems .

How should researchers analyze potential viral escape mutations in response to CRRSP57 binding?

Analysis of viral escape mutations requires sequencing viral samples before and after antibody exposure. Researchers should establish a baseline sequence, expose viral populations to CRRSP57 antibody, and sequence surviving viral populations to identify emergent mutations. Similar to studies with dual monoclonal antibody therapies, researchers should focus on mutations in the antibody binding region that correlate with reduced neutralization efficacy. Key mutations like those observed in the Spike protein during COVID-19 research (e.g., Q493R, E484K) can provide insights into potential escape mechanisms .

What statistical approaches best differentiate specific binding from background in peptide microarray data with CRRSP57?

Robust statistical analysis of peptide microarray data should include: (1) normalization procedures to account for technical variability, (2) comparison between sample groups using non-parametric tests like Wilcoxon Rank Sum when distributions are not normal, (3) correction for multiple comparisons to control false discovery rate, and (4) establishment of signal thresholds based on control samples. Combining signals from multiple peptides can improve discrimination between positive binding and background, as demonstrated in SARS-CoV-2 antibody research .

How can researchers resolve contradictory data when CRRSP57 shows different binding patterns across experimental platforms?

When facing contradictory binding results across platforms, researchers should systematically investigate potential causes: (1) compare antigen presentation formats across methods, as conformational differences can affect epitope accessibility, (2) evaluate buffer conditions that might influence binding kinetics, (3) assess potential interference from detection systems, and (4) consider antibody concentration effects on specificity. Resolution typically requires side-by-side comparisons under controlled conditions and validation with orthogonal methods to determine which results most accurately reflect biological reality .

How might structural biology approaches enhance CRRSP57 development and applications?

Structural biology approaches, particularly cryoEM combined with computational methods, can revolutionize antibody development by providing atomic-level insights into binding mechanisms. For antibodies like CRRSP57, these techniques allow visualization of the antibody-antigen complex, revealing binding orientations and key contact residues. This structural information can guide antibody engineering to enhance affinity, specificity, or broadness of neutralization. As cryoEM technology improves in throughput and resolution, these approaches will become increasingly valuable for rapid antibody characterization and optimization .

What are the most promising strategies for enhancing CRRSP57's breadth of neutralization against evolving viral variants?

Enhancing breadth of neutralization could be achieved through several engineering approaches: (1) targeting highly conserved epitopes that remain unchanged across variants, (2) developing antibody pairs where one antibody anchors to a conserved region while another targets functional domains, similar to the Stanford approach for SARS-CoV-2, or (3) creating bispecific antibodies that simultaneously target multiple epitopes. These strategies create treatments more resistant to viral evolution by increasing the mutational barrier required for escape .

How can computational modeling inform the optimization of CRRSP57 for improved therapeutic potential?

Computational modeling can inform antibody optimization through: (1) molecular dynamics simulations to predict stability and flexibility of antibody-antigen complexes, (2) in silico affinity maturation to identify mutations that might enhance binding, (3) epitope conservation analysis across viral variants to target invariant regions, and (4) pharmacokinetic modeling to optimize half-life and tissue distribution. These approaches can guide rational design modifications before experimental validation, potentially reducing the number of iterations required to achieve desired antibody properties .

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.