CRRSP19 Antibody

Shipped with Ice Packs
In Stock

Description

Potential Contextual Clues

  • Cusabio’s antibody catalog (Result ), which lists antibodies for proteins like CSP41B, CP29B, and CYP19-4 in Arabidopsis thaliana.

  • COVID-19-related studies (Results , , , , , , ), which focus on spike protein-targeting antibodies (e.g., SC27, bispecific antibodies).

  • General antibody structure (Results , ), which describe Fab/Fc regions, CDRs, and effector functions.

Hypothetical Antibody Profile

If "CRRSP19 Antibody" were a theoretical or emerging compound, its characteristics might align with:

ParameterHypothetical Attributes
Target AntigenUndetermined (speculative: CRISPR-related protein, viral spike component, or plant protein)
ApplicationResearch, therapeutic (e.g., cancer, autoimmune diseases, infectious diseases)
StructureConventional IgG/IgM format with Fab (antigen-binding) and Fc (effector) domains
GlycosylationN-linked glycans in Fc region, affecting stability and immune interaction
Cross-ReactivityPotential for conserved epitopes (e.g., SARS-CoV-2 spike cryptic sites )

Research Gaps and Recommendations

Given the lack of data:

  • Verify Nomenclature: Confirm spelling and context (e.g., "CRISPR-SpCas9" vs. "CRRSP19").

  • Expand Sources: Include specialized databases (e.g., Antibody Registry, UniProt) or preprint platforms.

  • Alternative Approaches:

    • Bispecific Antibodies: Explore dual-targeting strategies for enhanced efficacy .

    • Neutralization Assays: Reference methods for evaluating antibody potency (e.g., pseudovirus assays ).

Limitations

The provided materials focus on:

  • SARS-CoV-2 antibodies (Results , , , , , , ).

  • Plant-specific antibodies (Result ).

  • General antibody biology (Results , , ).

No references to "CRRSP19" exist in these domains.

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
CRRSP19 antibody; At3g21930 antibody; MZN24.5Putative cysteine-rich repeat secretory protein 19 antibody
Target Names
CRRSP19
Uniprot No.

Target Background

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

Q&A

What are the primary antibodies produced in response to SARS-CoV-2 infection?

The human immune system produces several types of antibodies in response to SARS-CoV-2 infection, with IgM and IgG being the most significant for diagnostic and research purposes. IgM antibodies are typically the first to appear, developing within 1-3 weeks after symptom onset and generally remaining in the blood for approximately 3-8 weeks. IgG antibodies develop slightly later but persist longer and may contribute to long-term immunity . Research from Canada has demonstrated that IgM has critical neutralizing capabilities against SARS-CoV-2, which was an unexpected finding since IgM is not typically associated with strong virus neutralizing activity .

How do researchers distinguish between antibodies produced through natural infection versus vaccination?

Methodologically, researchers analyze antibody specificity patterns. Naturally infected individuals typically develop antibodies against multiple viral proteins including the nucleocapsid protein, while vaccine-induced immunity generally produces antibodies only against the spike protein. Quantitative analysis of antibody titers against these different proteins allows researchers to differentiate between these two immune response origins. Additionally, temporal analysis of antibody development patterns and isotype distribution can provide further distinguishing characteristics .

What methodological approaches are used to determine antibody neutralizing capacity?

Researchers employ several laboratory techniques to assess neutralizing capacity:

  • Pseudovirus neutralization assays: Using engineered viruses expressing the SARS-CoV-2 spike protein

  • Plaque reduction neutralization tests (PRNT): Measuring an antibody's ability to prevent viral infection in cell culture

  • Competitive binding assays: Determining if antibodies can block the interaction between the viral spike protein and ACE2 receptors

  • Flow cytometry-based assays: Measuring antibody binding to cells expressing viral proteins

These approaches help determine both qualitative (whether neutralization occurs) and quantitative (the potency of neutralization) aspects of antibody function, with sensitivity and specificity values typically exceeding 95% when properly validated .

How can single B cell isolation techniques be optimized for novel antibody discovery?

Single B cell isolation represents a powerful approach for discovering novel neutralizing antibodies. One effective methodology involves incubating peripheral blood mononuclear cells with biotinylated target antigens (such as hepatitis B surface antigen or SARS-CoV-2 spike protein), followed by single-cell flow cytometry-based sorting of live, CD19+ IgG+ antigen-positive cells. After isolation, researchers amplify and sequence immunoglobulin genes from these single memory B cells to identify variable heavy and light chain sequences .

This approach has been successfully implemented to isolate human monoclonal antibodies with broad reactivity and neutralization capacity against multiple viral variants. The technique's efficiency is demonstrated by its ability to identify pathogen-specific monoclonal human antibodies from relatively small donor cell numbers, making it particularly valuable during pandemic situations .

What are the experimental challenges in developing chimeric antigen receptor (CAR) constructs from neutralizing antibodies?

Developing CAR constructs from neutralizing antibodies presents several methodological challenges:

  • Epitope selection: Researchers must determine whether to target conformational versus linear epitopes, which significantly impacts background activation levels

  • Signaling domain design: Engineering appropriate co-stimulatory domains (such as CD28) alongside primary signaling domains (CD3zeta)

  • Functional avidity assessment: Measuring the binding strength and activation threshold of the CAR construct

  • Polyfunctionality evaluation: Assessing multiple effector functions including cytokine secretion and target cell killing capacity

Research demonstrates that CARs recognizing linear epitopes (like the 4D08 construct) consistently demonstrate lower background activation compared to those targeting conformational epitopes (like 4D06), suggesting important considerations for construct design .

How do researchers assess antibody cross-reactivity with endemic coronaviruses?

Methodologically, researchers employ multiple approaches to assess cross-reactivity:

  • Enzyme-linked immunosorbent assays (ELISAs) using purified proteins from different coronavirus strains

  • Peptide arrays containing overlapping sequences from multiple coronaviruses

  • Competitive binding assays to determine epitope sharing

  • Absorption studies where sera are pre-incubated with one virus strain before testing against another

Recent findings suggest that pre-existing antibodies against common cold coronaviruses may influence the immune response to SARS-CoV-2. Some studies have observed higher levels of common cold coronavirus antibodies in convalescent plasma from recovered COVID-19 patients, suggesting potential cross-protective effects that warrant further investigation .

What cell line development strategies can accelerate antibody production for research purposes?

Based on experiences from COVID-19 monoclonal antibody development, several strategies can accelerate cell line development:

StrategyTraditional TimelineAccelerated TimelineKey Considerations
Use high productivity host cells3-4 months1-2 monthsConsider targeted integration approaches
Skip pool screening4-6 weeksEliminatedMay impact final clone selection quality
Abbreviated cell line stability testing2-3 months2-4 weeksFocus on critical quality attributes only
Concurrent master cell bank testingSequential (3-4 weeks)ParallelRequires risk mitigation planning

These approaches enabled several companies to initiate COVID-19 clinical trials in 50-70 days, which is substantially faster than traditional monoclonal antibody development timelines .

What are the critical parameters for assessing antibody sensitivity and specificity in diagnostic applications?

When designing antibody-based diagnostic tests, researchers must carefully evaluate:

  • Time-dependency factors: The sensitivity of antibody tests varies significantly based on the time since symptom onset. For example, Abbott's IgG antibody test demonstrates 100% sensitivity at ≥14 days post-symptom onset, while their IgM test shows 95% sensitivity at ≥15 days post-symptom onset .

  • Cross-reactivity assessment: Comprehensive testing against other viral antibodies is essential to ensure specificity. The Abbott IgG test demonstrates 99.63% specificity, meaning there is minimal cross-reactivity with antibodies developed against other viruses .

  • Population demographics influence: Age, comorbidities, and immunocompromised status can all affect antibody development patterns and must be accounted for in validation studies.

  • Antigen selection: Tests targeting different viral proteins (spike vs. nucleocapsid) may show varying performance characteristics depending on the research question being addressed.

How should researchers design experiments to evaluate antibody persistence and durability?

A methodologically sound approach to studying antibody persistence requires:

  • Longitudinal sampling: Collecting specimens at multiple timepoints (baseline, 1 month, 3 months, 6 months, and 12+ months)

  • Isotype-specific analysis: Measuring multiple antibody isotypes (IgG, IgM, IgA) and subclasses to understand the evolution of the immune response

  • Functional assays: Complementing quantitative measurements with neutralization assays to assess functional persistence, not merely presence

  • Memory B cell assessment: Evaluating the persistence of memory B cells capable of rapid antibody production upon re-exposure

  • Standardized controls: Including specimens from pre-pandemic samples and vaccinated individuals for comparative analysis

Recent research demonstrates that IgM antibodies, though traditionally considered short-lived, play a critical role in neutralizing SARS-CoV-2 and may contribute to long-lasting immunity, highlighting the importance of comprehensive isotype analysis .

How can apparent contradictions in antibody neutralization data across different studies be reconciled?

Methodological differences that contribute to data contradictions include:

  • Assay platform variations: Different neutralization assay formats (pseudovirus vs. live virus) produce systematically different results

  • Viral variant considerations: Studies using different viral variants or isolates may show divergent neutralization profiles, particularly as new variants emerge

  • Antibody source variability: Monoclonal antibodies versus polyclonal sera demonstrate fundamentally different neutralization characteristics

  • Endpoint definition inconsistencies: Various studies define neutralization differently (50% vs. 80% inhibition)

To reconcile these differences, researchers should:

  • Standardize assay platforms across laboratories

  • Include reference standards and control antibodies in all experiments

  • Clearly report methodological details including viral strain, cell type, and incubation conditions

  • Perform head-to-head comparisons when possible rather than relying on historical controls

What statistical approaches are most appropriate for analyzing antibody titer data in longitudinal studies?

Longitudinal antibody titer data presents unique statistical challenges requiring specialized approaches:

  • Mixed effects modeling: Accounts for both fixed effects (treatment, age, sex) and random effects (individual variation)

  • Time-to-event analysis: Particularly useful for studying the duration until antibody levels fall below a protective threshold

  • Area under the curve (AUC) analysis: Captures the cumulative antibody response over time rather than at discrete timepoints

  • Baseline normalization techniques: Critical for accounting for pre-existing cross-reactive antibodies that may influence the response to new antigens

  • Non-parametric methods: Often more appropriate than parametric approaches due to non-normal distribution of antibody titer data

How should researchers interpret the relationship between in vitro neutralization and in vivo protection?

The relationship between laboratory neutralization results and actual clinical protection is complex and requires careful interpretation:

  • Neutralization titers represent a correlate of protection but not the only determinant of immunity

  • Complementary immune mechanisms including T cell responses and non-neutralizing antibody functions (such as antibody-dependent cellular cytotoxicity) contribute significantly to protection

  • Threshold effects exist where protection may require a minimum neutralization titer, above which additional increases may not provide proportional benefit

  • Tissue-specific considerations are important as circulating antibody levels may not reflect mucosal immunity at respiratory surfaces

Research with monoclonal antibody therapies for COVID-19 provides insights into this relationship, demonstrating that neutralizing potency in vitro generally correlates with clinical efficacy, but with important exceptions that highlight the complexity of immune protection .

What methodological approaches can identify broadly neutralizing antibodies effective against multiple variants?

Cutting-edge approaches for identifying broadly neutralizing antibodies include:

  • Structure-guided antibody engineering: Using structural biology data to identify conserved epitopes and engineer antibodies targeting these regions

  • Deep mutational scanning: Systematically testing antibody binding against thousands of viral variants to identify broadly reactive candidates

  • B cell repertoire deep sequencing: Analyzing the complete antibody repertoire of convalescent patients to identify naturally occurring broadly neutralizing clones

  • Heterologous prime-boost immunization strategies: Exposing animal models to different viral variants sequentially to elicit broad responses

  • Computational epitope prediction: Using machine learning algorithms to predict conserved epitopes likely to generate broad neutralization

Recent research achieving accelerated monoclonal antibody development (75% reduction in development time) demonstrates the feasibility of rapidly adapting these approaches to emerging variants .

How might antibody-based therapeutics be optimized for next-generation respiratory virus treatments?

Next-generation optimization strategies for antibody therapeutics include:

  • Half-life extension technologies: Fc engineering to extend circulatory half-life from weeks to months, potentially enabling single-dose prophylaxis

  • Bispecific and multispecific formats: Targeting multiple epitopes simultaneously to prevent escape through mutation

  • Antibody cocktail formulation: Combining antibodies with complementary properties to enhance breadth and potency

  • Alternative delivery methods: Exploring inhalational or intranasal delivery for direct action at respiratory surfaces

  • Fc effector function enhancement: Engineering antibodies with optimized effector functions beyond simple neutralization

Experience from COVID-19 monoclonal antibody development, where clinical trials began within 50-70 days after project initiation, demonstrates the feasibility of rapidly implementing these optimization strategies .

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.