SARS-CoV-2 N Antibody Pair 2

Shipped with Ice Packs
In Stock

Description

Definition and Composition

The SARS-CoV-2 N Antibody Pair 2 typically consists of two mAbs that bind non-overlapping epitopes on the N protein. These antibodies are optimized for high sensitivity and specificity in sandwich-based assays, where one antibody captures the antigen and the other provides a detection signal (e.g., gold nanoparticles in LFAs) .

AntibodyClonalityEC50 (pM)Cross-Reactivity
GTX635685Recombinant9SARS-CoV-2 (Delta variant)
GTX635688Recombinant6SARS-CoV-2 (Delta variant)

EC50 values indicate the concentration of antibody required to achieve half-maximal binding .

Research and Development

  • Epitope Mapping: The antibodies in Pair 2 target distinct regions of the N protein, ensuring robust detection even with viral mutations. For example, GTX635685 binds to the N-terminal domain (NTD), while GTX635688 recognizes the C-terminal domain (CTD) .

  • Validation: Studies confirm the pair’s efficacy in detecting recombinant N protein, virus-infected cell lysates, and clinical nasal swab specimens. Sensitivity exceeds 95% for samples with viral loads ≥ 10^4 copies/mL .

Rapid Antigen Tests (RATs)

  • Lateral Flow Assays: The pair is integrated into point-of-care devices for self-testing, offering results within 15–20 minutes .

  • Cross-Reactivity: GTX635685 and GTX635688 show minimal cross-reactivity with other coronaviruses (e.g., SARS-CoV-1, MERS-CoV), reducing false positives .

Diagnostic Research

  • ELISA and Sandwich Assays: Pair 2 is used in high-throughput screening to validate N protein expression in patient samples .

  • Variant Detection: Both antibodies retain binding affinity for N proteins of SARS-CoV-2 variants (e.g., Alpha, Beta, Delta), making them suitable for monitoring evolving strains .

Key Research Findings

  • Therapeutic Potential: While primarily diagnostic, N-targeting antibodies like those in Pair 2 have shown promise in inhibiting complement hyperactivation induced by the N protein, a mechanism linked to COVID-19 severity .

  • Longitudinal Studies: Anti-N IgG antibodies persist for up to 6 months post-infection, supporting their use in serological assays .

Challenges and Future Directions

  • Variant Escape: Emerging variants with N protein mutations (e.g., Omicron) may reduce assay sensitivity, necessitating regular updates to antibody pairs .

  • Standardization: Harmonization of antibody pairs across manufacturers is critical to ensure consistent diagnostic performance .

References

  1. Nature: "A SARS-CoV-2 antibody curbs viral nucleocapsid protein-induced complement hyperactivation" (2021) .

  2. PMC: "High-throughput isolation of SARS-CoV-2 nucleocapsid antibodies" (2023) .

  3. Nature: "Evolution of antibody immunity to SARS-CoV-2" (2021) .

  4. GeneTex: "SARS-CoV-2 (COVID-19) Antibodies and Reagents" (2021) .

  5. PLOS ONE: "Monoclonal antibody pairs against SARS-CoV-2 for rapid antigen detection" (2022) .

Product Specs

Buffer
**Capture Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
**Detection Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for specific delivery timeframes.
Notes
We recommend using the capture antibody at a concentration of 1 µg/mL and the detection antibody at a concentration of 0.42 µg/mL. Optimal dilutions should be determined experimentally by the researcher.
Synonyms
N; Nucleoprotein; N; Nucleocapsid protein; NC; Protein N
Uniprot No.

Target Background

Function
The SARS-CoV-2 N protein packages the positive strand viral genome RNA into a helical ribonucleocapsid (RNP). It plays a critical role during virion assembly through its interactions with the viral genome and membrane protein M. Furthermore, the N protein significantly enhances the efficiency of subgenomic viral RNA transcription and viral replication. It may also modulate transforming growth factor-beta signaling by binding host smad3.
Protein Families
Betacoronavirus nucleocapsid protein family
Subcellular Location
Virion. Host cytoplasm.

Q&A

What are SARS-CoV-2 N protein antibody pairs and why are they important in research?

SARS-CoV-2 N protein antibody pairs consist of two monoclonal antibodies (mAbs) that recognize different epitopes on the viral nucleocapsid protein. These pairs are critically important in research because they enable the development of sandwich-based detection methods like double sandwich ELISA (dsELISA), lateral flow assays, and other immunodiagnostic platforms that require two antibodies binding simultaneously to the target antigen. The N protein is particularly valuable as a target because it is abundantly expressed during infection and highly conserved across variants, making these antibody pairs useful for both fundamental research and clinical applications .

How do N protein antibodies differ from spike (S) protein antibodies in SARS-CoV-2 research?

N protein antibodies and S protein antibodies target different structural components of SARS-CoV-2, resulting in distinct research applications:

This distinction is especially important in serological studies where researchers need to differentiate between vaccine-induced and infection-induced immunity, as described in studies using paired RBD and N protein-specific assays .

What are the key criteria for selecting effective N protein antibody pairs for diagnostic assays?

When selecting N protein antibody pairs for diagnostic assays, researchers should consider:

  • Epitope complementarity: The antibodies should bind to non-overlapping epitopes to avoid competitive binding, allowing simultaneous attachment to the N protein.

  • Affinity and specificity: High binding affinity to SARS-CoV-2 N protein without cross-reactivity to other coronaviruses is essential. For example, studies have shown mAbs that do not cross-react with MERS-CoV or HCoV-229E, though some may cross-react with SARS-CoV-1 due to genetic similarity .

  • Functional compatibility: One antibody should function effectively as a capture antibody (coating) while the other serves as a detection antibody (often biotinylated or directly labeled) .

  • Sensitivity threshold: The pair should achieve clinically relevant detection limits. For instance, the 2G11/bio-1C7 mAbs combination demonstrated sensitivity as low as 15 pg/well, comparable to commercial ELISA kits .

  • Performance in intended assay format: Not all antibody pairs perform equally across different platforms (ELISA, lateral flow, etc.) .

How can researchers optimize antibody pair screening methodologies for SARS-CoV-2 N protein detection?

Optimizing antibody pair screening for N protein detection requires a systematic approach:

  • Initial antibody generation: Generate diverse mAbs against recombinant SARS-CoV-2 N protein using hybridoma technology or phage display libraries from convalescent COVID-19 patients .

  • Individual antibody characterization: Screen each mAb individually for binding to the N protein using direct ELISA, and characterize their binding domains using epitope mapping techniques .

  • Cross-reactivity testing: Evaluate cross-reactivity against other coronavirus N proteins, particularly SARS-CoV-1, MERS-CoV, and common human coronaviruses .

  • Pair-wise screening: Test all possible antibody combinations in a matrix format using a sandwich ELISA where one antibody is coated on the plate and another is biotinylated or otherwise labeled . For example, researchers tested combinations of mAbs 1C7, 4F10, and 2G11 coated on plates with biotinylated versions of the same antibodies to identify the optimal 2G11/bio-1C7 pairing .

  • Sensitivity determination: Determine the limit of detection for each promising pair using serial dilutions of recombinant N protein .

  • Validation with clinical specimens: Finally, validate the best-performing pairs with real patient samples that have been confirmed by RT-PCR .

What are the best methods to evaluate the specificity and sensitivity of N protein antibody pairs?

Evaluating the specificity and sensitivity of N protein antibody pairs requires rigorous testing protocols:

  • Specificity testing:

    • Test against recombinant N proteins from other coronaviruses (SARS-CoV-1, MERS-CoV, HCoV-229E)

    • Include unrelated proteins as negative controls (e.g., recombinant prion protein was used in one study)

    • Evaluate with confirmed negative clinical samples

    • Assess potential interference from common sample matrix components

  • Sensitivity assessment:

    • Determine the limit of detection (LOD) using purified recombinant N protein in standard curves

    • Calculate signal-to-noise ratios at different antigen concentrations

    • Compare sensitivity to reference methods or commercial kits (e.g., the 2G11/bio-1C7 mAbs combination detected SARS-CoV-2 N protein as low as 15 pg/well, comparable to commercial kits)

    • Correlate with viral loads determined by RT-PCR in clinical samples

  • Clinical validation:

    • Test with confirmed positive and negative patient samples

    • Calculate clinical sensitivity, specificity, positive predictive value, and negative predictive value

    • Assess correlation between test signal intensity and RT-PCR cycle threshold (Ct) values

How can researchers effectively map epitopes recognized by N protein monoclonal antibodies?

Effective epitope mapping of N protein monoclonal antibodies involves several complementary approaches:

  • Domain-level mapping: Express different domains of the N protein (N-terminal domain, RNA-binding domain, C-terminal domain) as separate recombinant proteins and test antibody binding to identify the recognized domain .

  • Linear epitope mapping: Synthesize overlapping peptides spanning the entire N protein sequence and test antibody binding to each peptide to identify linear epitopes. This approach has helped elucidate SARS-CoV-2 S and N interactions in lateral flow chromatography .

  • Competition assays: Determine if antibodies compete for binding to the N protein, suggesting they recognize the same or overlapping epitopes.

  • Structural analysis: Use X-ray crystallography or cryo-electron microscopy to determine the structure of the antibody-antigen complex at atomic resolution. For example, the complex structure of the N protein RNA binding domain with a high-affinity mAb (nCoV396) revealed changes in epitopes and antigen's allosteric regulation .

  • Mutagenesis studies: Create point mutations in the N protein and assess their impact on antibody binding to identify critical residues in the epitope.

  • Cross-reactivity analysis: Test antibody binding to N proteins from other coronaviruses to identify conserved versus specific epitopes .

How can N protein antibody pairs be utilized to study SARS-CoV-2 complement activation and immune pathology?

N protein antibody pairs offer valuable tools for investigating SARS-CoV-2 complement activation and immune pathology:

  • Monitoring N protein-induced complement activation: Specific mAbs like nCoV396 have been shown to compromise N protein-induced complement hyperactivation, which is a risk factor for morbidity and mortality in COVID-19 patients. Using these antibodies in experimental models can help elucidate the mechanisms of complement-mediated pathology .

  • Studying complement pathway activation: Antibody pairs can be used in assays to measure the deposition of complement components (C3, C4, C5b-9) in response to N protein exposure, helping to understand which pathways (classical, alternative, or lectin) are primarily involved .

  • In vitro complement activation models: Researchers can develop cell-based assays using N protein antibody pairs to measure complement-mediated cell damage in the presence of N protein and serum, with specific antibodies potentially inhibiting this process .

  • Tissue studies: Immunohistochemistry with N protein antibody pairs can detect both the viral protein and complement components in tissue samples from COVID-19 patients, revealing their co-localization and potential causal relationships .

  • Therapeutic potential assessment: Testing whether N protein-specific antibodies can modulate complement activation provides insights into potential therapeutic approaches, as complement-mediated thrombotic microvascular injury contributes to the atypical ARDS features of COVID-19 .

What are the current limitations in using N protein antibody pairs for detecting SARS-CoV-2 variants and how can these be addressed?

Current limitations and potential solutions for N protein antibody pairs in variant detection include:

  • Epitope conservation across variants: While the N protein is more conserved than the spike protein, it still accumulates mutations that might affect antibody binding. Researchers should map epitopes recognized by their antibody pairs and monitor emerging variants for mutations in these regions .

  • Sensitivity variations: Different variants may express varying levels of N protein or present conformational changes affecting antibody binding. Researchers can address this by:

    • Testing antibody pairs against recombinant N proteins from major variants

    • Validating performance with clinical samples from patients infected with different variants

    • Developing multiplex assays with antibodies targeting different N protein epitopes

  • Cross-reactivity challenges: Some N protein antibody pairs may cross-react with SARS-CoV-1 due to high sequence similarity but not with MERS-CoV or seasonal coronaviruses. For variant-specific detection, researchers should:

    • Screen for antibodies binding to variant-specific epitopes

    • Develop competitive assays that can distinguish between variants based on differential binding

  • Validation methodology: When validating for new variants, researchers should:

    • Use both recombinant proteins and well-characterized clinical samples

    • Compare results with genomic sequencing data

    • Continuously monitor performance as new variants emerge

How can researchers integrate N protein antibody pairs with other biomarkers for improved COVID-19 diagnostics and prognostics?

Integrating N protein antibody-based detection with other biomarkers can significantly enhance COVID-19 diagnostics and prognostics:

  • Multi-target antigen detection: Combine N protein detection with spike protein or other viral antigens for increased sensitivity and specificity. This approach helps overcome limitations of single-target assays, particularly with emerging variants .

  • Antibody avidity measurement: Novel anti-N antibody avidity methods can identify SARS-CoV-2 reinfections with higher specificity (85%; 95% CI, 80%–90%) compared to anti-N antibody levels alone (72%; 95% CI, 66%–79%) in vaccinated cohorts. This approach can be integrated with other serological markers for more comprehensive infection history profiling .

  • Combined RNA and antigen detection: Developing multiplexed platforms that simultaneously detect viral RNA (via RT-PCR or LAMP) and N protein can increase diagnostic sensitivity across different stages of infection .

  • Inflammatory biomarker correlation: Correlating N protein levels with inflammatory markers (e.g., C-reactive protein, IL-6, ferritin) can improve prognostic accuracy. Research shows complement hyperactivation, which can be induced by the N protein, is associated with severe COVID-19 .

  • Longitudinal monitoring protocols: Designing testing algorithms that track N protein and antibody levels over time, along with other clinical parameters, can provide more actionable information for patient management .

How do N protein antibody-based lateral flow assays compare with ELISA methods in research applications?

N protein antibody-based lateral flow assays and ELISA methods have distinct advantages and limitations for research applications:

ParameterLateral Flow AssaysELISA Methods
SensitivityTypically 10^3-10^4 copies/mL, less sensitive than ELISACan detect as little as 15 pg/well of N protein
Specificity85-99% depending on antibody pair quality95-100% with optimized antibody pairs
Time to result10-30 minutes2-4 hours
Equipment requiredNone (visual readout) or simple readerPlate reader, washers, incubators
Sample processingMinimal, direct applicationMultiple washing and incubation steps
Quantification capabilitySemi-quantitative with readersFully quantitative with standard curves
Multiplexing potentialLimitedHigh with multi-well formats
Application in field settingsExcellentLimited by equipment needs
Correlation with RT-PCRModerate, better with high viral loadsStrong, inverse correlation with Ct values

For research requiring rapid field testing or point-of-care applications, lateral flow assays using N protein antibody pairs offer practical advantages despite lower sensitivity. Conversely, laboratory research benefiting from precise quantification and higher sensitivity should utilize ELISA methods with optimized antibody pairs like 2G11/bio-1C7 .

What are the experimental considerations when using N protein antibody pairs versus spike protein antibody pairs for distinguishing infection from vaccination?

When using antibody pairs to distinguish SARS-CoV-2 infection from vaccination, researchers should consider these experimental factors:

  • Assay design considerations:

    • N protein antibody assays detect only infection-induced responses since approved vaccines target the spike protein

    • Spike/RBD antibody assays detect both infection and vaccination responses

    • Combined testing provides the most accurate classification

  • Timing factors:

    • N protein antibodies typically appear 7-14 days post-infection

    • N antibody responses may be weaker in vaccinated individuals who experience breakthrough infections

    • Anti-N responses after breakthrough infection are typically lower than after primary infection in unvaccinated individuals

  • Quantitative thresholds:

    • Establishing appropriate cutoff values for each assay is critical

    • Different thresholds may be needed for different populations (e.g., immunocompromised, elderly)

    • Consider using ratio metrics between S and N antibody levels rather than absolute values

  • Antibody avidity measurement:

    • High-avidity anti-N antibodies can detect reinfections after a single infection with higher specificity (85%) compared to anti-N antibody levels alone (72%)

    • This method is particularly valuable for retroactive investigation of reinfection epidemiology in vaccinated cohorts

  • Cross-reactivity controls:

    • Include pre-pandemic samples as negative controls

    • Test for cross-reactivity with other human coronaviruses

    • Validate with confirmed infection and vaccination history samples

How can researchers enhance the sensitivity of N protein antibody pair-based detection systems for low viral load samples?

Enhancing sensitivity for N protein antibody pair-based detection in low viral load samples requires multifaceted approaches:

  • Antibody engineering strategies:

    • Use affinity maturation techniques to improve antibody binding constants

    • Develop recombinant antibody fragments (scFv) against the N protein from convalescent patients, which has shown subnanometer IC values against SARS-CoV-2

    • Consider antibody humanization to improve performance in diagnostic applications

  • Signal amplification methods:

    • Employ enzyme-based amplification (HRP, alkaline phosphatase) with sensitive substrates

    • Utilize chemiluminescent or fluorescent detection instead of colorimetric methods

    • Implement tyramide signal amplification or poly-HRP systems

    • Consider digital ELISA approaches (single molecule arrays) for ultimate sensitivity

  • Sample preparation optimization:

    • Develop concentration methods for N protein from clinical samples

    • Optimize lysis buffers to maximize release of N protein from viral particles

    • Remove interfering substances that might mask N protein detection

  • Advanced readout technologies:

    • Implement image-based analysis of lateral flow tests using mobile phones with specialized apps

    • Calculate normalized signal pixel intensities, which have been shown to inversely correlate with RT-PCR cycle threshold (Ct) values

    • Use machine learning algorithms to enhance signal detection from weak positives

  • Novel assay formats:

    • Develop ultrasensitive detection platforms like digital immunoassays

    • Consider microfluidic immunoassay designs with improved kinetics

    • Implement isothermal amplification methods coupled with antibody detection

What are the prospects for using N protein antibody pairs in developing universal SARS-CoV-2 variant detection systems?

The prospects for N protein antibody pairs in universal variant detection are promising for several reasons:

  • Evolutionary conservation advantage: The nucleocapsid protein shows higher conservation across SARS-CoV-2 variants compared to the spike protein, making it an excellent target for universal detection systems. Unlike the spike protein that evolves rapidly under immune pressure, the N protein maintains functional constraints that limit its mutational escape .

  • Multi-epitope targeting strategies: Researchers can develop cocktails of antibody pairs targeting different conserved epitopes on the N protein. This redundant recognition approach creates robust detection systems less vulnerable to single mutations in any one epitope .

  • Computational epitope prediction: Advanced bioinformatic approaches can predict conserved epitopes across known and predicted future variants, guiding the development of antibody pairs with broader detection capabilities .

  • Structure-guided antibody engineering: Cryo-EM structures of antibodies complexed with the N protein, like those determined for spike-binding antibodies, can guide rational design of broadly reactive antibodies. For instance, structurally identified footprints of antibodies can illuminate mechanisms of escape mutations and inform design of antibodies targeting highly conserved regions .

  • Integrated nucleic acid and protein detection: Future systems may combine N protein antibody detection with targeted nucleic acid sensing of variant-specific sequences, creating comprehensive diagnostic platforms capable of both detecting SARS-CoV-2 and identifying specific variants .

How might N protein antibody research contribute to understanding long COVID and persistent viral reservoirs?

N protein antibody research offers several promising avenues for investigating long COVID and viral persistence:

  • Biomarker discovery: N protein persistence in tissues may serve as a biomarker for viral reservoirs. Highly sensitive antibody pairs could detect low levels of N protein in different tissue compartments where the virus might persist after acute infection .

  • Tracking antibody dynamics: Monitoring anti-N antibody levels, isotypes, and avidity over time can provide insights into prolonged antigen exposure. The novel anti-N antibody avidity method for identifying reinfections could potentially be adapted to distinguish persistent infection from reinfection in long COVID patients .

  • Immune complex detection: N protein-antibody immune complexes might contribute to inflammation in long COVID. Specialized assays using N protein antibody pairs could detect these complexes in circulation or tissues .

  • Complement activation studies: N protein-induced complement hyperactivation has been linked to COVID-19 pathology. Continued monitoring of this phenomenon using functional antibodies like nCoV396 might explain persistent inflammation in long COVID .

  • Tissue-specific detection methods: Developing immunohistochemistry or in situ hybridization methods using N protein antibody pairs could help identify viral reservoirs in biopsies from long COVID patients, providing direct evidence of viral persistence .

  • Therapeutic exploration: Understanding how certain N protein antibodies like nCoV396 specifically compromise N protein-induced complement hyperactivation could lead to novel therapeutic approaches for managing long COVID symptoms related to dysregulated inflammation .

What are the challenges and opportunities in developing therapeutic applications for N protein-targeting antibodies?

Developing therapeutic applications for N protein-targeting antibodies presents unique challenges and opportunities:

  • Challenges:

    • Intracellular target: Unlike spike protein, the N protein is primarily intracellular, making it less accessible to conventional antibody therapeutics

    • Functional redundancy: Neutralizing the N protein may not directly prevent viral entry or replication

    • Timing considerations: By the time N protein is abundantly expressed, infection is already established

    • Limited precedent: Few successful therapeutics have targeted internal viral proteins

  • Opportunities:

    • Complement modulation: Some N protein-specific antibodies like nCoV396 can compromise N protein-induced complement hyperactivation, potentially reducing immunopathology in severe COVID-19

    • Immune complex clearance: Therapeutic antibodies could facilitate clearance of extracellular N protein, potentially reducing inflammatory triggers

    • Antibody engineering: Developing cell-penetrating antibodies or antibody derivatives could access intracellular N protein

    • Combination approaches: N protein antibodies could complement spike-targeting therapeutics for multi-mechanism treatment strategies

  • Emerging research directions:

    • Fc engineering: Modifying the Fc region of N protein antibodies to enhance effector functions like antibody-dependent cellular cytotoxicity against infected cells

    • Bispecific antibodies: Developing bispecific antibodies that target both the spike and N proteins could improve therapeutic efficacy

    • Intrabodies: Engineering antibodies expressed inside cells to target the N protein during viral replication

    • Complement intervention: Further exploration of antibodies like nCoV396 that specifically compromise N protein-induced complement hyperactivation, which is a risk factor for morbidity and mortality

The research on functional anti-N protein monoclonal antibodies like nCoV396 lays the foundation for identifying therapeutic applications beyond the conventional neutralization mechanisms associated with anti-spike antibodies .

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 2024 Thebiotek. All Rights Reserved.