CRRSP43 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
14-16 week lead time (made-to-order)
Synonyms
CRRSP43 antibody; At4g20640 antibody; F9F13.290Cysteine-rich repeat secretory protein 43 antibody
Target Names
CRRSP43
Uniprot No.

Target Background

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

Q&A

What is the binding specificity of CRRSP43 antibody compared to other research antibodies?

The binding specificity of antibodies is a critical parameter that determines their utility in research applications. When evaluating antibody specificity, researchers should employ multiple validation techniques, including western blotting, immunoprecipitation, and flow cytometry with appropriate positive and negative controls. Based on current antibody research methodologies, specificity should be tested using cells or tissues with differential expression of the target protein. Similar to epitope tag antibodies that recognize specific amino acid sequences, novel antibodies require thorough cross-reactivity testing against structurally similar proteins to establish binding specificity profiles . For viral-targeting antibodies, techniques similar to those used in SARS-CoV-2 research involve testing against both target and non-target viral proteins to ensure specificity .

What are the recommended storage conditions and stability parameters for maintaining CRRSP43 antibody activity?

Antibody stability is critical for reproducible experimental results. Most research antibodies should be stored at -20°C for long-term preservation and at 4°C for short-term use (typically 1-2 weeks). For novel antibodies, stability testing should include periodic activity assessment using standardized assays. Based on established practices for research-grade antibodies, aliquoting is strongly recommended to prevent freeze-thaw cycles, which can significantly degrade antibody function. The stability profile can be determined through accelerated degradation studies at various temperatures (4°C, 25°C, 37°C) with activity measurements at regular intervals. For antibodies developed for therapeutic applications, as seen in Stanford's SARS-CoV-2 research, extended stability testing under various buffer conditions may be necessary to ensure maintained neutralizing capacity .

What control experiments should be performed when first using CRRSP43 antibody in a new experimental system?

When implementing a novel antibody in research, comprehensive control experiments are essential. These should include:

  • Positive controls: Testing the antibody against known positive samples or recombinant proteins

  • Negative controls: Using isotype-matched irrelevant antibodies to assess non-specific binding

  • Concentration titration: Determining optimal antibody concentration for signal-to-noise ratio

  • Blocking peptide controls: Confirming specificity by pre-incubating with the target peptide/protein

Researchers should also include appropriate secondary antibody-only controls to evaluate background staining. As demonstrated in the PD-1 antibody research, cross-blocking experiments with existing antibodies can provide valuable information about epitope recognition . Such experiments help establish the spatial relationship between binding sites and functional relevance of the antibody, particularly important when working with novel antibodies targeting important biological pathways.

How should CRRSP43 antibody be validated for use in immunoprecipitation experiments?

Validating antibodies for immunoprecipitation requires a systematic approach to ensure reproducible results. The validation procedure should include:

  • Initial testing with recombinant protein or overexpression systems

  • Optimization of antibody concentration and incubation parameters

  • Comparison with established antibodies targeting the same protein (if available)

  • Analysis of pull-down efficiency using quantitative western blotting

For novel antibodies like CRRSP43, researchers should first determine optimal binding conditions including buffer composition, salt concentration, and detergent types. Based on methodologies from epitope tag antibody studies, pre-clearing lysates with protein A/G beads can reduce non-specific binding . The immunoprecipitation efficiency should be quantified by comparing the amount of target protein in input vs. immunoprecipitated fractions, with at least 30-50% recovery considered acceptable for most applications. Additionally, mass spectrometry analysis of immunoprecipitated samples can provide comprehensive validation of specificity by identifying co-immunoprecipitated proteins.

What are the optimal conditions for using CRRSP43 antibody in flow cytometry applications?

Optimization of antibody use in flow cytometry requires careful consideration of multiple parameters:

ParameterRecommended Testing RangeNotes
Antibody concentration0.1-10 μg/mlTitrate to determine optimal signal-to-noise ratio
Incubation temperature4°C vs. room temperatureLower temperatures typically reduce non-specific binding
Incubation time15-60 minutesLonger times may increase signal but also background
Buffer compositionPBS vs. specialized buffersInclude protein (0.5-2% BSA) to block non-specific binding
Fixation compatibilityPre- vs. post-fixation stainingTest with different fixatives (PFA, methanol)

Drawing from methodologies used in PD-1 antibody research, researchers should determine if the epitope recognized by CRRSP43 is sensitive to fixation or permeabilization procedures . For intracellular epitopes, optimizing permeabilization conditions is crucial to maintain both epitope accessibility and cellular morphology. Additionally, compensation controls must be included when performing multicolor flow cytometry to account for spectral overlap. Validation should include comparison of staining patterns with known biology of the target protein and appropriate isotype controls to establish specificity.

How can CRRSP43 antibody be conjugated to fluorophores or enzymes while preserving its activity?

Antibody conjugation requires careful consideration of both chemistry and biology to maintain binding activity. The recommended conjugation approach involves:

  • Determining the optimal antibody-to-label ratio through small-scale pilot experiments

  • Selecting conjugation chemistry based on available reactive groups (typically primary amines on lysine residues)

  • Using gentle buffer conditions to preserve antibody structure during conjugation

  • Implementing purification steps to remove unconjugated label

Similar to the approaches used for epitope tag antibodies, researchers should first evaluate antibody concentration and purity, as contaminants can interfere with conjugation efficiency . For novel antibodies, small-scale test conjugations comparing different fluorophores or enzymes (HRP, AP) help identify optimal conditions. Post-conjugation validation should include activity comparison with the unconjugated antibody using identical experimental conditions. For sensitive applications, site-specific conjugation technologies (such as sortase-mediated approaches) may better preserve binding activity by targeting the constant region rather than potentially interfering with the variable regions responsible for antigen recognition.

How does the epitope recognition of CRRSP43 antibody compare with other antibodies targeting similar domains?

Understanding epitope recognition is fundamental for predicting antibody function and potential cross-reactivity. For novel antibodies, epitope mapping techniques include:

  • Peptide array analysis using overlapping peptides covering the target protein

  • Hydrogen-deuterium exchange mass spectrometry to identify protected regions

  • Site-directed mutagenesis of suspected binding residues

  • Competition binding assays with antibodies of known epitope specificity

Drawing from methodologies in the PD-1 antibody research, cross-blocking experiments can provide valuable information about the spatial relationship between binding sites . The study demonstrated how different antibodies targeting the same protein (PD-1) exhibited variable blocking patterns, with some antibodies (like 29F.1A12) completely preventing detection with other clones while others (like RMP1-14) did not interfere with detection. For antibodies targeting viral proteins, such as those in SARS-CoV-2 research, determining whether the epitope is conserved across variants is crucial for predicting broad neutralization potential .

What approaches can resolve contradictory results when using CRRSP43 antibody across different experimental platforms?

Contradictory results across experimental platforms represent a common challenge in antibody-based research. Systematic troubleshooting approaches include:

  • Evaluating epitope accessibility under different experimental conditions

  • Assessing protein conformation changes during sample preparation

  • Comparing binding kinetics across different assay formats

  • Implementing orthogonal detection methods to validate findings

When facing contradictory results, researchers should carefully examine differences in sample preparation protocols, buffer conditions, and detection methods. Drawing from PD-1 antibody research, some antibodies may have dramatically different performance characteristics depending on application, with variable staining intensity observed across clones even when targeting the same protein . For complex samples, pre-absorbing the antibody with potential cross-reactive components can help eliminate non-specific interactions. Additionally, validating results with alternative antibodies or methodologies that do not rely on antibody recognition (such as mass spectrometry or nucleic acid-based detection) can help resolve discrepancies and confirm true biological effects.

How can CRRSP43 antibody be used in combination with other antibodies for multiplex analysis systems?

Multiplex antibody applications require careful optimization to maintain specificity while enabling simultaneous detection of multiple targets. Key considerations include:

  • Spectral compatibility of detection systems (fluorophores, enzyme substrates)

  • Cross-reactivity assessment between primary and secondary antibodies

  • Sequential staining protocols for sterically hindered epitopes

  • Optimizing signal amplification strategies for balanced detection sensitivity

Based on methodologies from epitope tag research, testing antibodies individually before combining them helps establish baseline performance metrics . For immunofluorescence applications, implementing linear unmixing algorithms can resolve spectral overlap issues. When using multiple antibodies from the same species, implementing tyramide signal amplification or directly conjugated primary antibodies can circumvent cross-reactivity of secondary antibodies. For complex tissues or cells with variable target expression, optimization of detection sensitivity for each antibody independently before multiplexing helps ensure balanced signal detection across all targets.

What are the critical variables affecting reproducibility when using CRRSP43 antibody in different laboratory settings?

Reproducibility challenges in antibody-based experiments stem from multiple sources that must be systematically controlled:

VariableImpact on ReproducibilityStandardization Approach
Antibody lot variationDifferent binding characteristicsPurchase sufficient quantity of single lot for entire study
Sample preparationEpitope accessibility differencesDetailed SOP for sample handling and processing
Instrument calibrationDetection sensitivity variationsRegular calibration with standardized beads/controls
Image acquisitionSubjective analysis parametersAutomated acquisition with defined settings
Data analysis methodsInconsistent quantificationPre-established analysis pipelines with objective thresholds

Drawing from PD-1 antibody research methods, implementing detailed protocols for cell preparation, staining procedures, and instrument settings is essential for reproducible results . For novel antibodies, researchers should validate performance across different experimental batches, days, and operators to establish variability parameters. Incorporating quantitative standards (such as recombinant proteins of known concentration) in each experiment provides internal calibration points. For collaborative studies, sharing detailed protocols, reagent sources, and raw data enables more effective troubleshooting of reproducibility issues.

How should researchers interpret unexpected cross-reactivity patterns observed with CRRSP43 antibody?

Unexpected cross-reactivity requires systematic investigation to distinguish technical artifacts from biologically meaningful interactions. The recommended approach includes:

  • Confirming cross-reactivity using multiple detection methods

  • Performing immunoprecipitation followed by mass spectrometry to identify cross-reactive proteins

  • Evaluating sequence and structural homology between the intended target and cross-reactive proteins

  • Conducting epitope mapping to identify the specific recognition motif

From the PD-1 antibody research, we learn that cross-reactivity patterns can provide valuable insights into structural relationships between proteins . Unexpected cross-reactivity might reveal previously unknown protein isoforms, post-translational modifications, or structurally related family members. Researchers should distinguish between specific cross-reactivity (binding to related epitopes) and non-specific binding (interaction with unrelated proteins through Fc regions or hydrophobic interactions). In some cases, apparent cross-reactivity may actually represent biologically relevant interactions between the primary target and its binding partners, which can be verified through proximity ligation assays or co-immunoprecipitation studies.

What statistical approaches are most appropriate for analyzing quantitative data generated using CRRSP43 antibody?

Quantitative analysis of antibody-based experiments requires statistical approaches tailored to the specific application and data distribution:

  • For flow cytometry data: Non-parametric tests (Mann-Whitney U, Kruskal-Wallis) are often more appropriate than parametric tests due to non-normal distribution of fluorescence intensity

  • For immunohistochemistry quantification: Mixed effects models that account for both biological and technical variation

  • For western blot densitometry: ANCOVA to adjust for loading control variations

  • For high-dimensional data (CyTOF, multiplexed imaging): Dimensionality reduction techniques (tSNE, UMAP) followed by clustering analysis

Similar to the approach used in PD-1 antibody research, calculating percent inhibition on a log scale rather than linear scale may be more appropriate for certain applications . Researchers should include sufficient biological and technical replicates to power statistical analyses, typically minimum n=3 for preliminary studies and n≥5 for definitive experiments. For longitudinal studies, repeated measures ANOVA or linear mixed models better account for within-subject correlations. Regardless of the statistical approach, researchers should report both effect sizes and p-values, and consider multiple testing corrections when performing numerous comparisons.

How can CRRSP43 antibody be engineered to improve its performance characteristics for specific research applications?

Antibody engineering offers multiple approaches to enhance performance for specific applications:

  • Affinity maturation through directed evolution or site-directed mutagenesis

  • Fragment generation (Fab, scFv) to improve tissue penetration or reduce non-specific binding

  • Fc engineering to modulate effector functions or extend half-life

  • Site-specific conjugation to improve homogeneity of labeled antibodies

Drawing from the Stanford antibody research against SARS-CoV-2, engineering bispecific antibodies that combine complementary functions represents a powerful approach to enhance therapeutic potential . For research applications, humanization of antibody sequences can reduce immunogenicity in human samples or animal models. Introducing specific mutations in the framework regions can improve stability under harsh experimental conditions. For applications requiring intracellular delivery, engineering cell-penetrating peptide fusions or utilizing electroporation techniques can facilitate antibody entry. Additionally, developing recombinant antibody formats with standardized production methods can improve batch-to-batch consistency compared to traditional hybridoma approaches.

What emerging technologies could enhance the utility of CRRSP43 antibody in single-cell analysis platforms?

Emerging technologies are revolutionizing antibody applications at the single-cell level:

  • Photocleavable antibody-DNA conjugates for spatial transcriptomics integration

  • Split-pool barcoding strategies for high-throughput screening applications

  • Proximity labeling approaches for identifying molecular interactions in situ

  • Multiplexed ion beam imaging for simultaneous detection of 40+ antibodies

These technologies enable integration of antibody-based protein detection with other 'omics approaches. For example, CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) combines antibody detection with single-cell RNA sequencing, while CO-Detection by indEXing (CODEX) enables highly multiplexed imaging using DNA-barcoded antibodies. When applying these technologies with novel antibodies, researchers should validate performance in the specific platform, as conjugation chemistry and detection modalities may affect binding characteristics. Additionally, computational approaches for integrating protein expression data with other single-cell parameters are essential for extracting biological insights from these complex datasets.

How can CRRSP43 antibody be evaluated for potential therapeutic applications while focusing on research parameters?

Evaluating antibodies for therapeutic potential within a research context involves assessing several critical parameters:

  • Target specificity and affinity under physiological conditions

  • Functional effects in relevant cellular and animal models

  • Cross-reactivity with related proteins and orthologous targets in model organisms

  • Stability and activity in biological matrices (serum, interstitial fluid)

Drawing from the Stanford SARS-CoV-2 antibody research, a comprehensive evaluation should include testing in multiple experimental systems that recapitulate disease biology . For example, researchers identified antibodies that could neutralize all SARS-CoV-2 variants by targeting conserved epitopes, demonstrating the importance of epitope selection in developing broadly effective antibodies. The research showed how pairing antibodies with complementary functions (one serving as an anchor by binding to conserved regions, another inhibiting viral infection) can overcome viral evolution, a principle that could be applied to other therapeutic antibody development efforts.

The evaluation process should focus on mechanistic understanding rather than commercial development parameters, including detailed characterization of:

  • Mode of action through structure-function studies

  • Epitope mapping and conservation analysis

  • Off-target effects using proteome-wide binding assays

  • Pharmacodynamic biomarkers for assessing target engagement

This research-oriented approach provides fundamental insights that can guide later-stage therapeutic development while maintaining academic research focus.

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.