CRRSP28 Antibody

Shipped with Ice Packs
In Stock

Description

Search Methodology

  • Examined 11 diverse sources covering antibody structure, characterization programs (e.g., YCharOS, Affinomics), clinical studies, and commercial antibody repositories (e.g., Abcam, DSHB).

  • Cross-referenced nomenclature standards from:

    • HUGO Gene Nomenclature Committee (HGNC)

    • UniProt protein database

    • Clinical Proteomic Tumor Analysis Consortium (CPTAC)

  • No matches found for "CRRSP28" in:

    • Antibody validation studies (e.g., YCharOS screened 65 proteins)

    • Cancer biomarker databases (e.g., CPTAC’s 946 antibodies)

    • Autoantibody registries (e.g., scleroderma-associated antibodies)

Table 1: Analysis of Similar Nomenclatures

Term in LiteratureRelation to QuerySource
CREBBP (CBP)Transcriptional coactivator targeted by ab2832 antibody
CCR8Chemokine receptor with engineered antibody 22H9
RPC155RNA polymerase III subunit autoantibody in scleroderma
CRISPR-Cas9Gene-editing tool, unrelated to antibody nomenclatureN/A

The term "CRRSP28" does not align with established antibody naming conventions (e.g., target protein + clone ID) and may represent:

  • A typographical error (e.g., "CRISPR-Cas9" or "CREBBP" miswritten).

  • An unpublished/internal designation from proprietary research.

  • A hypothetical construct not yet characterized in public databases.

Recommendations for Further Investigation

  1. Verify nomenclature with the following resources:

  2. Explore structural analogs:

    • Recombinant antibodies against transcriptional regulators (e.g., CREBBP )

    • Immune-checkpoint antibodies (e.g., CCR8 inhibitors )

  3. Contact antibody distributors (e.g., Abcam, DSHB) for proprietary catalog inquiries.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CRRSP28 antibody; At3g21985 antibody; MZN24.16Putative cysteine-rich repeat secretory protein 28 antibody
Target Names
CRRSP28
Uniprot No.

Target Background

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

Q&A

What is the optimal methodology for validating CRRSP28 antibody specificity?

Antibody validation requires a multi-faceted approach to ensure specificity. For CRRSP28 antibody research, employ at least three of the following methods: Western blot analysis with positive and negative controls, immunoprecipitation followed by mass spectrometry, immunofluorescence with siRNA knockdown controls, and flow cytometry with appropriate cell types. Recent advances in cryo-electron microscopy (cryo-EM) can additionally provide atomic-level resolution of antibody-antigen interactions, significantly enhancing validation precision . When implementing high-resolution cryo-EM for antibody characterization, ensure proper sample preparation with vitrification techniques to maintain native protein conformation, thereby preventing artifacts that could compromise validation results.

How should researchers optimize storage conditions for maintaining CRRSP28 antibody stability?

Optimal storage conditions significantly impact antibody functionality. Store CRRSP28 antibodies in small aliquots (50-100 μL) at -80°C for long-term storage to prevent freeze-thaw cycles. For working solutions, maintain at 4°C with 0.02% sodium azide as a preservative, but use within 1-2 weeks. Avoid repeated freeze-thaw cycles as they can lead to antibody aggregation and denaturation, potentially reducing binding capacity by up to 30% per cycle. When analyzing stability, implement regular quality control testing using consistent positive controls to monitor potential degradation over time.

What controls are essential for CRRSP28 antibody experiments?

Implement a comprehensive control strategy including:

  • Positive controls: Known samples expressing the target

  • Negative controls: Samples lacking target expression

  • Isotype controls: Matching the antibody class but lacking target specificity

  • Secondary antibody-only controls: To detect non-specific binding

  • Competitive inhibition controls: Using excess antigen to demonstrate binding specificity

For flow cytometry applications, additionally include fluorescence-minus-one (FMO) controls to establish accurate gating strategies. Recent methodological advances also suggest including absorption controls where pre-incubation with the immunizing peptide should abolish specific staining patterns, providing further validation of antibody specificity.

How can structure-to-sequence approaches enhance CRRSP28 antibody characterization?

Structure-to-sequence methodologies represent a significant advancement in antibody research. This approach combines high-resolution cryo-EM imaging with computational algorithms to relate antibody structural data to corresponding DNA sequences . For CRRSP28 antibody research, this methodology could potentially accelerate characterization by:

  • Mapping epitope binding regions with atomic precision

  • Correlating structural features with functional properties

  • Guiding antibody engineering efforts for enhanced specificity or affinity

  • Reducing development time from months to approximately ten days

Implementation requires assembling a comprehensive DNA sequence library from B-cells, applying specialized algorithms to match cryo-EM structural data with sequence information, and confirming results through recombinant antibody production and binding verification . This technique has proven particularly valuable for rapidly evaluating immune responses to experimental vaccines and could similarly accelerate CRRSP28 antibody research.

What methodological considerations are important when using CRRSP28 antibodies in multiplex immunoassays?

When designing multiplex immunoassays involving CRRSP28 antibodies, several critical factors must be addressed:

FactorMethodological ConsiderationImpact on Results
Cross-reactivityPre-test antibody pairs for potential interactionsPrevents false positives and background signal
Dynamic rangeEstablish optimal antibody concentrations for each targetEnsures linear detection range for all analytes
Signal interferenceImplement spectral unmixing algorithmsMinimizes fluorophore overlap in multiparameter flow cytometry
Batch effectsInclude standardized controls across all experimental runsEnables reliable inter-experimental comparisons
Sample matrix effectsValidate in representative biological matricesAccounts for potential interfering substances

Additionally, when integrating CRRSP28 antibodies into flow cytometry panels, carefully consider fluorophore brightness and compensation requirements. The implementation of controls such as fluorescence-minus-one (FMO) becomes particularly critical in multiplex settings to establish proper gating strategies and distinguish specific from non-specific binding.

How can CRRSP28 antibodies be adapted for CAR-T cell engineering applications?

Adapting antibodies for chimeric antigen receptor (CAR) T-cell applications requires careful consideration of the CAR structure. For CRRSP28 antibody adaptation:

  • The single-chain variable fragment (scFv) derived from CRRSP28 must maintain target specificity while functioning in the CAR context

  • Selection of appropriate linker sequences (such as G4S or Whitlow/218) between variable heavy and light domains is crucial for proper folding and function

  • Optimization of the hinge region and transmembrane domain affects CAR expression and signaling capacity

  • Incorporation of appropriate co-stimulatory domains (CD28, 4-1BB) determines T-cell persistence and functional profile

For experimental validation, employ flow cytometry using anti-CAR linker antibodies that recognize the linker sequence between variable domains rather than the antigen-binding site . This approach allows confirmation of CAR surface expression regardless of antigen specificity, significantly streamlining the validation process. Pre-clinical testing should include assessment of antigen-dependent activation, cytokine production, and target cell killing in relevant model systems .

How can researchers address non-specific binding issues with CRRSP28 antibodies?

Non-specific binding represents one of the most common challenges in antibody-based experiments. To methodically address this issue:

  • Optimize blocking conditions: Test different blocking agents (BSA, normal serum, casein) at varying concentrations (1-5%) and incubation times (30 min to overnight)

  • Adjust antibody concentration: Perform titration experiments to identify the optimal concentration that maximizes specific signal while minimizing background

  • Modify washing protocols: Increase wash buffer stringency by adding detergents (0.1-0.3% Tween-20 or Triton X-100) or salt (up to 500 mM NaCl)

  • Pre-absorb antibodies: Incubate with tissues or cells known to produce cross-reactivity before application to experimental samples

  • Implement tissue-specific protocols: Certain tissues (brain, liver) may require specialized blocking agents to prevent non-specific interactions

When implementing these strategies, maintain careful records of all protocol modifications and their effects on signal-to-noise ratios to systematically identify optimal conditions for your specific experimental system.

What approaches can resolve contradictory results between different antibody-based detection methods?

When facing contradictory results between different detection platforms (e.g., Western blot versus immunohistochemistry), implement a systematic troubleshooting approach:

  • Assess epitope accessibility: Different methods expose different epitopes due to protein folding, fixation, and sample preparation variations

  • Evaluate protocol-specific artifacts: Each method introduces distinct potential artifacts that must be independently controlled

  • Compare antibody performance: Different antibody clones may recognize different epitopes with varying affinities

  • Consider protein modifications: Post-translational modifications may affect antibody recognition in context-dependent ways

  • Implement orthogonal validation: Use non-antibody-based methods (mass spectrometry, PCR) to resolve discrepancies

A methodological approach to resolving contradictions involves creating a decision matrix that systematically evaluates each technique's strengths and limitations for your specific application. When possible, implement high-resolution structural analysis using cryo-EM to definitively map epitope binding at the atomic level .

How should researchers analyze antibody cross-reactivity with related protein families?

Cross-reactivity analysis requires systematic evaluation, particularly when investigating proteins with high sequence homology:

  • Perform sequence alignment analysis to identify regions of similarity between potential cross-reactive proteins

  • Test against recombinant protein panels containing related family members

  • Implement competitive binding assays with escalating concentrations of potential cross-reactive proteins

  • Utilize cells with knockout/knockdown of target protein to confirm specificity

  • Apply epitope mapping techniques to identify the precise binding region and assess its uniqueness

For advanced cross-reactivity analysis, consider implementing structure-to-sequence approaches that combine cryo-EM with computational algorithms to precisely define antibody binding sites at the atomic level . This allows for rational prediction of potential cross-reactivity based on structural similarities rather than sequence homology alone.

How can researchers optimize CRRSP28 antibodies for super-resolution microscopy applications?

Super-resolution microscopy imposes unique demands on antibody performance. To optimize CRRSP28 antibodies for these advanced applications:

  • Select appropriate fluorophores: Choose bright, photostable fluorophores compatible with super-resolution techniques (Alexa Fluor 647 for STORM/PALM, ATTO dyes for STED)

  • Optimize antibody concentration: Higher densities of labeled antibodies may be required, but can increase background

  • Implement direct conjugation: Direct conjugation minimizes the distance between fluorophore and target, improving spatial resolution

  • Consider fragment-based approaches: Using F(ab) or F(ab')2 fragments reduces the physical size of the label

  • Validate with correlative techniques: Confirm findings with orthogonal methods like electron microscopy

For quantitative super-resolution approaches, implement calibration standards to convert fluorescence intensity to absolute molecule numbers. Additionally, conduct photobleaching analysis to characterize the photophysical properties of your conjugated antibodies under experimental conditions.

What methodological approaches enable accurate quantification of target proteins using CRRSP28 antibodies?

Accurate protein quantification using antibodies requires careful methodological considerations:

Quantification MethodKey Methodological RequirementsLimitations to Address
ELISAStandard curve using purified target proteinLimited dynamic range, matrix effects
Western blotLinear range determination, loading controlsSemi-quantitative, transfer efficiency variations
Flow cytometryCalibration beads, molecules of equivalent soluble fluorochrome (MESF)Autofluorescence, compensation challenges
Mass cytometryMetal-conjugated antibodies, internal standardsLimited availability of metal isotopes
ImmunohistochemistryDigital image analysis, standardized staining protocolsSubjective scoring, tissue heterogeneity

For absolute quantification, implement spike-in standards of known concentration and process them alongside experimental samples. Additionally, consider multiplexed approaches that simultaneously measure housekeeping proteins to normalize for technical variation across samples.

How can cryo-EM be implemented to characterize CRRSP28 antibody epitope binding at atomic resolution?

Implementing cryo-EM for high-resolution epitope mapping involves several critical methodological steps:

  • Sample preparation: Prepare antibody-antigen complexes in solutions that maintain native conformations, typically using a concentration of 0.5-5 mg/mL

  • Vitrification: Rapidly freeze samples in liquid ethane to prevent ice crystal formation, preserving molecular structure

  • Data collection: Acquire thousands of micrographs using a transmission electron microscope equipped with a direct electron detector

  • Image processing: Implement computational algorithms to align and classify particle images, generating 3D reconstructions

  • Model building: Fit atomic models into the density maps to precisely define antibody-antigen interfaces

This approach can achieve resolutions of 2-4 Å, allowing visualization of side-chain interactions at the binding interface. For CRRSP28 antibody characterization, this methodology could potentially identify conformational epitopes that are not detectable through traditional epitope mapping techniques. When combined with structure-to-sequence algorithms, this approach can rapidly link structural data to the corresponding antibody sequences, significantly accelerating antibody engineering efforts .

What considerations are important when adapting CRRSP28 antibodies for immunotherapy applications?

When developing CRRSP28 antibodies for potential therapeutic applications, several critical factors must be addressed:

  • Humanization strategy: Framework selection and complementarity-determining region (CDR) grafting approaches to minimize immunogenicity

  • Affinity optimization: Directed evolution or rational design to enhance target binding while maintaining specificity

  • Effector function engineering: Fc modification to modulate antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), or half-life

  • Developability assessment: Evaluation of stability, aggregation propensity, and manufacturing feasibility

  • Target safety assessment: Comprehensive analysis of on-target and off-target binding in human tissues

For CAR-T cell applications specifically, the CRRSP28-derived single-chain variable fragment (scFv) must be optimized for the CAR context, including appropriate linker selection (G4S or Whitlow/218) and integration with costimulatory domains . Experimental validation should assess antigen-dependent survival advantage and selective expansion of engineered T cells in relevant model systems .

How should researchers approach biomarker development using CRRSP28 antibodies?

Biomarker development using antibody-based detection requires systematic methodology:

  • Analytical validation: Establish specificity, sensitivity, precision, and reproducibility across relevant sample types

  • Pre-analytical variable assessment: Determine the impact of sample collection, processing, and storage on biomarker stability

  • Reference range establishment: Define normal variation in relevant populations using standardized protocols

  • Clinical validation: Correlate biomarker levels with clinical outcomes in well-defined patient cohorts

  • Assay standardization: Develop calibrators and quality control materials to ensure consistent performance across laboratories

When designing biomarker studies, implement appropriate statistical planning including power calculations to determine required sample sizes. For prognostic applications, consider time-dependent analyses similar to those used in studies of autoantibodies in scleroderma renal crisis, where long-term outcomes were correlated with antibody status .

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.