SARS Nucleocapsid, 1-49 a.a.

SARS-Associated Coronavirus Nuclecapsid (1-49 a.a.), Recombinant
Shipped with Ice Packs
In Stock

Description

RNA Binding and Genome Packaging

  • The 1-49 a.a. region binds viral RNA via electrostatic interactions, facilitating genome condensation .

  • In vitro studies show this fragment retains binding affinity for single-stranded RNA/DNA despite lacking the full-length protein’s oligomerization capacity .

Immune Evasion and Diagnostic Utility

  • Serological Detection:

    • Acts as an immunodominant epitope, achieving >96% sensitivity and 100% specificity in ELISA for SARS-CoV-2 .

    • IgG responses to this region are detectable earlier than S-protein antibodies, shortening diagnostic windows .

  • Host Interaction:

    • Disrupts STAT1/STAT2 phosphorylation, inhibiting interferon signaling .

Stability and Standardization

  • The NCAP-1 reference material (10.0 ± 1.9 μmol/L) enables harmonized antigen testing across diagnostic platforms .

  • LC-SEC analysis shows the 1-49 a.a. fragment forms tetramers/hexamers in solution, critical for assay reproducibility .

Phase Separation and Viral Assembly

  • Full-length N protein forms biomolecular condensates with viral RNA, but the 1-49 a.a. fragment lacks this capability .

  • Small-molecule disruptors of condensates (e.g., heparin) inhibit viral replication, highlighting the NTD’s role in genome packaging .

Product Specs

Introduction

The SARS Nucleocapsid protein plays a vital structural role in the virus. It binds with the RNA genome, forming a helical nucleocapsid. This protein comprises three structural domains and is associated with three outer structural proteins: membrane (M), envelope (E), and spike (S) proteins.

Description

This recombinant protein is derived from E. coli and encompasses amino acids 1 to 49 of the Nucleocapsid protein, representing immunodominant regions.

Physical Appearance
A clear, sterile-filtered solution.
Formulation

The SARS Nucleocaspid protein solution is supplied in a phosphate-buffered saline (PBS) solution.

Stability

The protein is shipped with ice packs to maintain a low temperature. Upon receiving, store the protein at -20 degrees Celsius.

Purity

SDS-PAGE analysis confirms that the protein purity is greater than 90%.

Source
Escherichia Coli.
Purification Method

Purified by proprietary chromatographic technique.

Specificity

Immunoreactive with sera of SARS-infected individuals.

Q&A

What is the structural composition of the SARS Nucleocapsid protein N-terminal domain (1-49 a.a.)?

The N-terminal domain (1-49 a.a.) of SARS Coronavirus Nucleocapsid protein represents one of the immunodominant regions of the nucleocapsid core protein. This region is part of the larger nucleocapsid protein which forms a helical capsid structure that encapsulates the viral RNA genome. The N-terminal segment contains highly conserved motifs, including a FYYLGTGP sequence that has been identified as evolutionarily conserved across different coronaviruses . This region's structural properties contribute to its ability to bind RNA and perform various regulatory functions during viral replication. When produced recombinantly in expression systems like E. coli, this protein fragment maintains its immunoreactivity with sera from SARS-infected individuals, indicating preservation of key epitopes .

What expression systems are recommended for producing recombinant SARS Nucleocapsid N-terminal fragments?

E. coli expression systems have been successfully employed to produce recombinant SARS Coronavirus Nucleocapsid protein fragments, including the N-terminal domain (1-49 a.a.) . When expressing this protein fragment in E. coli, researchers can typically achieve approximately 90% purity as determined by SDS-PAGE analysis . The bacterial expression system offers advantages of high yield and cost-effectiveness, which are particularly valuable for diagnostic applications. For optimal purification results, researchers should consider incorporating affinity tags that can be subsequently removed if necessary for functional studies. Alternative expression systems such as insect cells or mammalian cells may provide better post-translational modifications when needed for specific applications, though these systems are generally more complex and costly to implement than bacterial systems. Selection of the appropriate expression system should be guided by the intended downstream applications and the requirement for specific structural features.

How can researchers distinguish between true positive serological responses to SARS-CoV-2 N protein (1-49 a.a.) and cross-reactivity with seasonal coronavirus antibodies?

Distinguishing between true SARS-CoV-2 N protein responses and cross-reactivity with seasonal coronaviruses requires a multi-faceted approach. Researchers should implement competitive binding assays using both SARS-CoV-2 N protein fragments and corresponding regions from human coronaviruses (HCoV) like 229E, NL63, HKU1, and OC43. Studies have revealed that despite the N-terminal region containing conserved sequences, the C-terminus region of SARS-CoV-2 N protein shows minimal sequence homology with HCoVs and demonstrates higher specificity for SARS-CoV-2 . Therefore, developing assays that target both regions can help differentiate specific from cross-reactive responses.

Pre-adsorption protocols can be implemented where patient sera are first incubated with HCoV N proteins to remove cross-reactive antibodies before testing against SARS-CoV-2 N protein. Research has identified interesting patterns where crossreactivity appears stronger with alpha-HCoVs (229E, NL63) than beta-HCoVs (HKU1, OC43) despite the latter having greater sequence similarity to SARS-CoV-2 . This suggests conformational epitopes play a crucial role in antibody recognition beyond primary sequence identity.

Analyzing multiple immunoglobulin isotypes (IgG, IgM, IgA) provides additional discrimination power, as cross-reactive responses often show different isotype distributions compared to primary SARS-CoV-2 responses .

What are the critical mutations observed in the N-terminal region (1-49 a.a.) across SARS-CoV-2 variants, and how might they impact diagnostic assays?

Comparative sequence analysis of the SARS-CoV-2 N protein across global isolates has revealed several mutations within the N-terminal region, including positions 6, 13, and 33 . The mutation P13L is among the most frequently observed variations, potentially affecting protein stability and function . These mutations may impact the sensitivity and specificity of diagnostic assays that target this region.

Research comparing the original Wuhan reference sequence with isolates from various regions identified twenty distinct mutations within the N protein, with some clusters appearing in the N-terminal unstructured region . The following table summarizes key mutations identified in the N-terminal domain:

PositionMutationFrequencyPotential Impact on Structure/Function
6VariableLowMinimal predicted structural change
13P13LHighMay affect protein flexibility
33VariableLowMay impact RNA-binding properties

These mutations can affect epitope conformation and antigenicity, potentially compromising the performance of diagnostic tests that utilize the N-terminal region. Assay developers should regularly monitor emerging variants and validate their tests against diverse strain collections. Targeting multiple conserved epitopes simultaneously can increase assay robustness against evolutionary changes .

What methodologies are most effective for analyzing the binding kinetics between SARS Nucleocapsid (1-49 a.a.) and RNA sequences?

For analyzing binding interactions between the SARS Nucleocapsid N-terminal domain (1-49 a.a.) and RNA sequences, several complementary methodologies provide comprehensive kinetic and affinity data. Surface Plasmon Resonance (SPR) offers real-time, label-free measurement of association and dissociation rates. Researchers should immobilize either the protein fragment or RNA sequence on sensor chips, then flow the counterpart analyte at varying concentrations to determine KD values, kon, and koff rates.

Microscale Thermophoresis (MST) provides an alternative approach requiring minimal sample amounts while detecting interactions in solution. Fluorescently labeled RNA or protein allows measurement of thermophoretic mobility changes upon complex formation. For structural characterization of the bound complex, Nuclear Magnetic Resonance (NMR) spectroscopy can map interaction interfaces at atomic resolution by observing chemical shift perturbations upon titration.

Functional studies employing Electrophoretic Mobility Shift Assays (EMSA) with varying salt concentrations and pH conditions help determine the electrostatic contribution to binding. Researchers should systematically vary RNA sequence motifs to identify specificity determinants. The RNA-binding properties of this domain contribute to the selective packaging of viral genomes during virion assembly and may represent potential targets for antiviral interventions .

How do post-translational modifications of the SARS Nucleocapsid (1-49 a.a.) affect its structural properties and interactions?

The SARS Nucleocapsid protein undergoes several post-translational modifications that significantly impact its function, with phosphorylation being the most extensively studied. Analysis using tools like NetPhos3.1 Server has identified multiple potential phosphorylation sites within the N protein, including some in the N-terminal region . These modifications can alter the protein's RNA-binding properties, protein-protein interactions, and localization patterns.

Phosphorylation status affects the N protein's ability to oligomerize and form the ribonucleoprotein complex with viral RNA. Mass spectrometry analysis combined with functional assays reveals that differentially phosphorylated forms of the N protein exhibit varying affinities for RNA and protein partners . Researchers investigating these modifications should employ a combination of site-directed mutagenesis (converting phosphorylation sites to alanine or phosphomimetic residues) and functional assays to determine the impact on RNA binding.

Flow cytometry and confocal microscopy analyses can track how modifications affect subcellular localization patterns, particularly the protein's ability to shuttle between cytoplasm and nucleolus . Additionally, circular dichroism spectroscopy can reveal how phosphorylation alters secondary structure composition, providing insights into structure-function relationships. Understanding these modifications is crucial for developing targeted therapeutics that might disrupt N protein function at specific stages of the viral lifecycle.

What are the optimal conditions for purifying recombinant SARS Nucleocapsid (1-49 a.a.) for structural studies?

Purification of recombinant SARS Nucleocapsid (1-49 a.a.) for high-resolution structural studies requires careful optimization of expression and purification protocols. After expression in E. coli, cells should be lysed in buffers containing 50 mM Tris-HCl (pH 8.0), 300 mM NaCl, and protease inhibitors . For initial capture, immobilized metal affinity chromatography (IMAC) using Ni-NTA resin is effective when the construct includes an N-terminal or C-terminal His-tag.

To achieve higher purity necessary for structural studies (>95%), researchers should implement a multi-step purification strategy. Following IMAC, size-exclusion chromatography using Superdex 75 columns effectively separates the target protein from aggregates and contaminants. For crystallography applications, removal of the affinity tag using specific proteases (TEV or PreScission) is recommended, followed by a reverse IMAC step and a final polishing step using ion-exchange chromatography.

Critical buffer conditions for maintaining stability include 25 mM HEPES (pH 7.5), 150 mM NaCl, and 1 mM DTT. The addition of 5-10% glycerol can improve long-term stability. Researchers should verify final purity by SDS-PAGE and assess homogeneity by dynamic light scattering before proceeding to structural studies . For NMR studies, expression in minimal media supplemented with 15N-labeled ammonium chloride and/or 13C-labeled glucose is necessary for isotopic labeling.

What serological assay designs maximize specificity when targeting the SARS Nucleocapsid (1-49 a.a.) region?

Designing highly specific serological assays targeting the SARS Nucleocapsid (1-49 a.a.) region requires careful consideration of cross-reactivity issues. A one-step antigen capture format employing the recombinant nucleocapsid protein has proven effective in diagnostic applications . For optimal specificity, researchers should implement a dual-antigen approach combining the N-terminal region with the C-terminal domain, which shows minimal sequence homology with seasonal coronaviruses .

Competitive ELISA formats incorporating blocking steps with heterologous coronavirus N proteins can significantly reduce false positives from cross-reactive antibodies. Pre-adsorption of test sera with recombinant N proteins from endemic human coronaviruses (particularly 229E and NL63) effectively removes cross-reactive antibodies before testing against SARS-CoV-2 antigens .

Epitope-specific approaches targeting unique regions within the 1-49 a.a. segment provide greater discrimination. Researchers can employ synthetic peptides corresponding to divergent sequences rather than the entire domain. Multiplex assays measuring responses to several coronavirus N proteins simultaneously allow mathematical algorithms to calculate specificity scores based on differential reactivity patterns .

For clinical validation, assay performance should be assessed using well-characterized sample panels including:

  • Confirmed SARS-CoV-2 cases at different time points post-infection

  • Pre-pandemic samples with documented seasonal coronavirus infections

  • Samples from individuals with other respiratory infections

What bioinformatic approaches can identify conserved functional motifs within the 1-49 a.a. region across coronavirus species?

Identifying conserved functional motifs within the SARS Nucleocapsid 1-49 a.a. region requires sophisticated bioinformatic approaches that integrate sequence, structure, and function data. Multiple sequence alignment (MSA) using tools like Clustal Omega provides the foundation for comparative analysis across coronavirus species . Researchers should compile comprehensive datasets including alpha and beta coronaviruses, with special attention to SARS-CoV, SARS-CoV-2, MERS-CoV, and common human coronaviruses.

Position-specific scoring matrices can detect subtle conservation patterns that simple alignment might miss. The FYYLGTGP motif represents one such conserved element already identified in the N-terminal region . For functional prediction, researchers should employ tools like PROVEAN to assess the functional impact of amino acid substitutions within conserved motifs .

Structure-based approaches complement sequence analysis by identifying spatially clustered residues that may form functional surfaces. Homology modeling using Swiss-Model followed by molecular dynamics simulations can predict how conserved motifs contribute to protein flexibility and interaction capabilities . Machine learning algorithms trained on known RNA-binding proteins can identify potential nucleic acid interaction sites within the 1-49 a.a. region.

Researchers should validate bioinformatic predictions through experimental approaches including mutational analysis and functional assays. Cross-species conservation analysis reveals evolutionary constraints and can highlight potential targets for broad-spectrum antivirals that would remain effective despite viral mutation.

How does antibody response to the SARS Nucleocapsid (1-49 a.a.) correlate with disease severity and duration in COVID-19 patients?

Analysis of antibody responses to SARS Nucleocapsid protein reveals important correlations with clinical outcomes. Studies indicate that antibodies to SARS-CoV-2 N protein are significantly higher in patients experiencing more severe symptoms and longer disease duration . This correlation provides valuable prognostic information when monitoring patient immune responses. Female patients typically demonstrate higher anti-N antibody levels compared to males, suggesting potential sex-based differences in immune response patterns .

The isotype distribution and temporal dynamics of the antibody response also correlate with disease progression. IgG antibodies to the N protein remain relatively stable for at least three months post-infection, while IgA and IgM responses decline more rapidly . This differential persistence has implications for serological testing strategies at various time points after infection.

Quantitative analysis of antibody titers against different regions of the N protein, including the 1-49 a.a. segment, can provide more granular correlation with disease parameters. Higher antibody levels against specific epitopes within this region may indicate particular inflammatory response patterns associated with different clinical manifestations. Comprehensive serological profiling examining responses to multiple viral antigens simultaneously provides the most informative correlation with disease outcomes.

What are the potential advantages and limitations of targeting the SARS Nucleocapsid (1-49 a.a.) for therapeutic interventions?

The SARS Nucleocapsid N-terminal domain (1-49 a.a.) presents several therapeutic targeting opportunities based on its essential functions in viral replication. This domain participates in RNA binding and nucleocapsid assembly, making it an attractive target for small molecule inhibitors that could disrupt these critical processes . The relatively conserved nature of this region across coronavirus species suggests that successful interventions might have broad-spectrum potential.

The following table summarizes potential therapeutic approaches targeting the N-terminal domain:

Therapeutic ApproachMechanismAdvantagesLimitations
Small molecule inhibitorsDisrupt RNA bindingMay block viral replicationRequire cell penetration
Peptide antagonistsInterfere with protein-protein interactionsHigh specificityStability and delivery issues
Nucleic acid aptamersBind to N protein active sitesAdaptable to viral mutationsComplex delivery systems needed
PROTAC technologyInduce N protein degradationComplete protein eliminationRequires extensive optimization

Additionally, targeting this region may face challenges from emerging mutations that could affect inhibitor binding while maintaining protein function . Combination approaches simultaneously targeting multiple viral proteins likely offer the most robust therapeutic strategy.

How can researchers develop quantitative structure-activity relationship (QSAR) models for small molecules targeting the N-terminal domain?

Developing effective QSAR models for small molecules targeting the SARS Nucleocapsid N-terminal domain requires a systematic approach integrating structural data, binding assays, and computational modeling. Researchers should begin by establishing a diverse compound library with known binding affinities or inhibitory activities against the 1-49 a.a. region, determined through biochemical assays such as fluorescence polarization or thermal shift assays.

High-resolution structural characterization of protein-ligand complexes using X-ray crystallography or NMR spectroscopy provides essential data for model development. When experimental structures are unavailable, molecular docking and molecular dynamics simulations can generate binding mode hypotheses. Researchers should extract molecular descriptors that capture physicochemical properties relevant to binding, including electrostatic potential, hydrophobicity patterns, and shape complementarity to binding pockets.

Machine learning algorithms, including random forests, support vector machines, and deep neural networks, can identify relationships between molecular features and biological activity. Cross-validation procedures, including k-fold validation and external test sets, are essential to assess model robustness. The most predictive models can guide iterative optimization of lead compounds through structure-based design.

Given the functional importance of RNA binding by the N-terminal domain, researchers should incorporate RNA-competitive binding assays in their activity measurements to ensure that inhibitors target functionally relevant interactions rather than nonspecific binding sites.

How might structural studies of SARS Nucleocapsid (1-49 a.a.) inform the development of pan-coronavirus vaccines?

Structural studies of the SARS Nucleocapsid N-terminal domain (1-49 a.a.) can substantially advance pan-coronavirus vaccine development through several mechanisms. High-resolution structural characterization using X-ray crystallography, cryo-electron microscopy, and NMR spectroscopy reveals conserved epitopes that could elicit broadly neutralizing antibodies against multiple coronavirus species. The conserved FYYLGTGP motif within this region represents one such target for cross-protective immunity .

Epitope mapping through hydrogen-deuterium exchange mass spectrometry (HDX-MS) in complex with broadly neutralizing antibodies can identify antigenic sites that remain accessible on the assembled nucleocapsid. Comparative structural analysis across different coronavirus N proteins helps identify conserved surface patches that maintain similar conformations despite sequence divergence, potentially serving as targets for cross-protective responses.

Structure-guided immunogen design can create optimized versions of the N-terminal domain with enhanced stability and immunogenicity. Computational approaches like molecular dynamics simulations predict how mutations might affect the presentation of conserved epitopes. While most current vaccine efforts focus on the spike protein, nucleocapsid-based approaches offer complementary protection through T-cell responses, as the N protein is a dominant target for T-cell immunity .

Combined approaches incorporating both S and N protein components may provide more comprehensive protection against emerging coronavirus variants and related species. Structure-based rational design enables the creation of chimeric immunogens presenting multiple protective epitopes from different proteins in optimal conformations.

What novel experimental approaches can elucidate the role of SARS Nucleocapsid (1-49 a.a.) in modulating host immune responses?

Elucidating the immunomodulatory functions of the SARS Nucleocapsid N-terminal domain requires innovative experimental approaches that capture complex protein-host interactions. Single-cell RNA sequencing (scRNA-seq) of immune cells exposed to recombinant N protein fragments can reveal cell type-specific transcriptional responses, identifying pathways modulated by this domain. This approach should be complemented by phosphoproteomics and interactomics to map signaling cascades activated upon N protein exposure.

CRISPR-Cas9 screening in immune cell lines can identify host factors required for N protein-mediated immune modulation. Genome-wide knockout or knockdown libraries allow unbiased discovery of cellular components that interact with the N-terminal domain or mediate its effects. Humanized mouse models expressing the N protein under inducible promoters enable in vivo assessment of immunomodulatory effects in a physiologically relevant context.

Cryo-electron tomography of cells expressing the N protein can visualize its interactions with cellular structures at nanometer resolution. Proximity labeling techniques like BioID or APEX2 can identify proteins that transiently interact with the N-terminal domain in living cells, revealing potential immune signaling components affected by this viral protein.

These approaches should incorporate comparative analyses between seasonal coronavirus and SARS-CoV-2 N proteins to identify unique immunomodulatory mechanisms that might contribute to the distinctive pathogenesis of COVID-19. Understanding these mechanisms could reveal therapeutic opportunities to counteract immune dysregulation in severe disease.

How can integrated multi-omics approaches enhance our understanding of SARS Nucleocapsid interactions during infection?

Integrated multi-omics approaches provide comprehensive insights into SARS Nucleocapsid protein interactions and functions throughout the infection cycle. Researchers should implement parallel analyses combining proteomics, transcriptomics, and interactomics data from infected cells. Time-course experiments capturing different stages of infection are essential for understanding dynamic changes in N protein interactions.

Proximity-dependent biotinylation combined with mass spectrometry can map the N protein interactome in living cells, revealing both stable and transient interaction partners. Cross-linking mass spectrometry (XL-MS) provides structural information about protein complexes formed by the N protein with viral and cellular components. RNA immunoprecipitation sequencing (RIP-seq) identifies specific viral and cellular RNA sequences bound by the N-terminal domain during infection.

Integration of these datasets with phosphoproteomics reveals how post-translational modifications of the N protein respond to changing cellular conditions and affect interaction networks . Computational network analysis identifies key hub proteins and potential vulnerabilities in the interaction network that could be targeted therapeutically.

Spatial transcriptomics and proteomics approaches can map the subcellular distribution of N protein interactions, revealing compartment-specific functions related to its localization in both nucleolus and cytoplasm . Cross-species comparative analyses identify conserved and divergent interaction patterns between different coronavirus N proteins, highlighting evolutionarily important interfaces.

These multi-omics approaches should be applied to both in vitro cell culture systems and clinical samples from COVID-19 patients to validate the physiological relevance of identified interactions and their potential contributions to disease pathogenesis.

Product Science Overview

Introduction

The Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) nucleocapsid protein is a crucial component of the virus’s structure and function. The recombinant form of the nucleocapsid protein, specifically the first 49 amino acids (1-49 a.a.), has been extensively studied for its role in the viral life cycle and its potential applications in diagnostics and therapeutics.

Structure and Function

The nucleocapsid (N) protein of SARS-CoV is a highly conserved structural protein that plays a pivotal role in the packaging of the viral RNA genome into ribonucleoprotein complexes. This protein is essential for the assembly and release of new virions. The N protein is composed of multiple domains, each contributing to its function:

  • N-terminal domain (NTD): This domain, which includes the first 49 amino acids, is responsible for RNA binding and is crucial for the formation of the ribonucleoprotein complex.
  • C-terminal domain (CTD): This domain is involved in dimerization and interaction with other viral and host proteins.
Role in Viral Life Cycle

The N protein is involved in several key processes during the SARS-CoV life cycle:

  1. RNA Binding: The NTD binds to the viral RNA genome, facilitating its encapsidation into ribonucleoprotein complexes.
  2. Virion Assembly: The N protein interacts with the membrane (M) protein to drive the assembly of new virions.
  3. Host Interaction: The N protein can modulate host cell processes, including the immune response, to enhance viral replication and evade host defenses.
Recombinant Nucleocapsid Protein

The recombinant form of the N protein, particularly the first 49 amino acids, has been produced and studied for various applications:

  • Diagnostics: The recombinant N protein can be used as an antigen in serological assays to detect antibodies against SARS-CoV in patient samples. This is crucial for diagnosing past infections and monitoring immune responses.
  • Vaccine Development: The N protein is a target for vaccine development due to its high immunogenicity. Recombinant N protein-based vaccines aim to elicit a strong immune response against the virus.
  • Therapeutics: Understanding the structure and function of the N protein can aid in the development of antiviral drugs that target its interactions with viral RNA and other proteins.
Research and Applications

Recent studies have highlighted the importance of the N protein in the context of the SARS-CoV-2 pandemic. The N protein of SARS-CoV-2 shares significant similarity with that of SARS-CoV, making it a valuable target for cross-reactive diagnostics and therapeutics . Research has focused on the following areas:

  • Structural Analysis: Detailed structural studies of the N protein have provided insights into its RNA-binding mechanisms and interactions with other viral proteins .
  • Functional Studies: Functional analyses have identified key residues and domains critical for the N protein’s role in the viral life cycle .
  • Vaccine and Drug Development: The N protein is being explored as a target for novel vaccines and antiviral drugs aimed at preventing and treating SARS-CoV-2 infections .

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.