CoV-2 S1 (319-541), Sf9

Coronavirus 2019-nCoV Spike Glycoprotein-S1 Receptor Binding Domain Recombinant,SF9
Shipped with Ice Packs
In Stock

Description

Production in Sf9 Cells

Sf9 cells are widely used for high-yield RBD production due to scalability and cost-effectiveness. Key findings include:

  • Optimized yields: Up to 21.1 ± 3.7 mg/L after chromatographic purification .

  • Process parameters:

    • Cell density: Optimal infection at 2×1062 \times 10^6 cells/mL .

    • Multiplicity of infection (MOI): MOI ≥3 ensures maximal protein expression .

  • Purification: His-tag affinity chromatography achieves >95% purity .

ACE2 Binding and Neutralization

  • Sf9-derived RBD retains high ACE2 affinity (Kd1.2nMK_d \approx 1.2 \, \text{nM}) comparable to mammalian systems .

  • Antibodies targeting RBD (e.g., COVA1-16) neutralize SARS-CoV-2 by blocking ACE2 interaction, with IC50_{50} values <1 μg/mL .

Glycosylation Impact

  • Sf9 vs. HEK293:

    Glycosylation FeatureSf9-RBDHEK293-RBD
    N331 sialylation22%80%
    N343 sialylation4%50%
    • Reduced sialylation in Sf9-RBD may alter antibody recognition but does not impair ACE2 binding .

Applications in Research and Therapeutics

  • Vaccine development: Bivalent vaccines combining S1-WT and S1-Mut (variants) elicit cross-neutralizing antibodies against SARS-CoV-2 variants (e.g., B.1.351) .

  • Diagnostics: Sf9-RBD is used in ELISA and lateral flow assays to detect neutralizing antibodies in COVID-19 sera .

  • Antiviral peptides: HR1/HR2-derived peptides inhibit viral fusion by disrupting six-helix bundle formation .

Comparative Analysis with Other Expression Systems

ParameterSf9HEK293
YieldHigh (≥20 mg/L)Moderate (5–10 mg/L)
GlycosylationSimple, paucimannoseComplex, human-like
CostLowHigh
ScalabilityIndustrial-scaleLimited
Functional activityPreserved ACE2 bindingEnhanced immunogenicity .

Challenges and Future Directions

  • Glycoengineering: Modifying Sf9 glycosylation pathways to mimic human-like glycans for improved antibody responses .

  • Yield optimization: Advanced bioreactor designs and fed-batch strategies to enhance productivity .

  • Variant coverage: Developing multivalent RBD vaccines to address emerging variants (e.g., Omicron) .

Product Specs

Introduction

The 2019 novel coronavirus (2019-nCoV), a human-infecting coronavirus causing viral pneumonia, emerged in a Wuhan, Hubei province, China fish market in December 2019. This virus, 87% identical to the 2018 SARS-CoV-2 bat coronavirus found in Zhoushan, eastern China, likely shares a similar receptor-binding domain (RBD) structure despite some differences. This suggests 2019-nCoV may bind to the human ACE2 (angiotensin-converting enzyme 2) receptor. While bats are considered the likely origin, researchers believe an intermediary animal at the seafood market facilitated human transmission. Notably, RSCU analysis indicates 2019-nCoV's spike glycoprotein could be a recombinant of bat coronavirus and an unidentified coronavirus.

Description

This product consists of a single, glycosylated polypeptide chain representing the Receptor Binding Domain (RBD) of the SARS-CoV-2 Spike Glycoprotein-S1. It encompasses amino acids 319-541, resulting in a molecular weight of 26.2 kDa.

A 6 amino acid His-tag is fused to the C-terminus of the CoV-2 S1 (319-541). Purification is achieved using proprietary chromatographic techniques.

Physical Appearance
The product appears as a colorless solution that has undergone sterile filtration.
Formulation

The provided CoV-2 S1 (319-541) solution has a concentration of 0.25mg/ml. It is prepared in a Phosphate-Buffered Saline (pH 7.4) solution containing 10% glycerol.

Stability

For optimal storage, keep the product refrigerated at 4°C if you plan to use the entire vial within 2-4 weeks. For longer storage periods, freezing at -20°C is recommended. Consider adding a carrier protein like 0.1% HSA or BSA for extended storage. Minimize freeze-thaw cycles to maintain product integrity.

Purity

Analysis using SDS-PAGE confirms a purity greater than 95.0%.

Biological Activity

The product's biological activity is evaluated based on its binding affinity to Human ACE-2 in a functional ELISA assay (refer to CAT# enz-1159).

Synonyms

Severe acute respiratory syndrome coronavirus 2, COVID-19, COVID-19 virus, COVID19, HCoV-19, Human coronavirus 2019, SARS-2, SARS-CoV2, SARS2, Wuhan coronavirus, Wuhan seafood market pneumonia virus, SARS-CoV-2 SP RBD, 2019-nCoV SP RBD, 2019-nCoV, 2019-nCoV; Spike RBD Protein.

Source

Sf9, Baculovirus cells.

Amino Acid Sequence

ADPRVQPTES IVRFPNITNL CPFGEVFNAT RFASVYAWNR KRISNCVADY SVLYNSASFS TFKCYGVSPT KLNDLCFTNV YADSFVIRGD EVRQIAPGQT GKIADYNYKL PDDFTGCVIA WNSNNLDSKV GGNYNYLYRL FRKSNLKPFE RDISTEIYQA GSTPCNGVEG FNCYFPLQSY GFQPTNGVGY QPYRVVVLSF ELLHAPATVC GPKKSTNLVK NKCVNFHHHH HH

Q&A

What is the SARS-CoV-2 S1 (319-541) region and why is it significant for research?

The SARS-CoV-2 S1 (319-541) region refers to the receptor-binding domain (RBD) of the viral spike protein, comprising amino acids 319-541 within the S1 subunit. This domain has an approximate molecular weight of 27 kDa and is responsible for the recognition and binding of the virus to the human ACE2 receptor . The RBD constitutes the primary target for neutralizing antibodies and is therefore central to vaccine development, serological testing, and therapeutic intervention strategies . The significance of this region extends beyond its structural role, as it contains epitopes recognized by convalescent sera and houses conserved regions that may confer protection against emerging variants . Research focusing on this protein fragment enables detailed study of virus-host interactions and the development of countermeasures that can effectively target both wild-type and variant SARS-CoV-2 strains .

How do different expression systems affect the characteristics of the recombinant SARS-CoV-2 RBD protein?

Expression systems significantly influence the post-translational modifications, particularly glycosylation patterns, of recombinant SARS-CoV-2 RBD proteins. Mammalian expression systems like CHO and HEK293 cells produce differently glycosylated forms of the RBD, with CHO-derived RBD predominantly displaying core 1 structures with two sialic acids (H1N1S2), while HEK293-derived RBD exhibits more complex glycosylation patterns . E. coli systems, lacking glycosylation machinery, produce non-glycosylated forms but can achieve expression levels of approximately 100 mg/L under IPTG induction . The glycosylation status affects the protein's isoelectric point and potentially its immunogenicity and receptor-binding characteristics . Despite these differences, antibody recognition of RBD expressed in different systems shows high correlation (R²=0.9894 between CHO and HEK293), suggesting that the core epitopes remain accessible regardless of expression system . When selecting an expression system, researchers must consider the trade-offs between yield, glycosylation fidelity, and downstream application requirements.

What are the recommended purification strategies for SARS-CoV-2 S1 (319-541) from different expression systems?

Purification strategies for SARS-CoV-2 S1 (319-541) should be tailored to the expression system and fusion tags employed. For mammalian expression systems, column chromatography is commonly employed for His-tagged or Fc-fusion RBD proteins . For E. coli-expressed fusion proteins, a single affinity binding step using inexpensive cellulose powder has proven effective for CBM9-fused RBD proteins, yielding approximately 0.1 g/L of purified protein . When designing purification protocols, researchers should consider the protein's stability, as some fusion constructs demonstrate greater resistance to protease degradation than others . The choice of buffer conditions, salt concentration, and pH during purification can significantly impact recovery and stability of the final product. Verification of purified protein should include SDS-PAGE analysis to confirm molecular weight and purity , followed by functional assays to ensure that the purification process has not compromised the protein's biological activity.

How can the stability of recombinant SARS-CoV-2 RBD proteins be assessed and improved?

The stability of recombinant SARS-CoV-2 RBD proteins can be assessed through protease resistance assays, thermal shift assays, and long-term storage studies. Research indicates that certain fusion constructs, particularly those involving CBM9-spike protein fragments, demonstrate enhanced resistance to protease degradation . Stability improvements can be achieved through several strategies: (1) Selection of appropriate fusion partners that shield vulnerable protease cleavage sites; (2) Engineering of the protein sequence to remove or modify regions prone to degradation; (3) Optimization of buffer formulations to include stabilizing excipients; and (4) Selection of expression systems that confer favorable post-translational modifications. The stability profile may vary significantly between different constructs – for example, studies show that some CBM9-spike fusion proteins maintained integrity while others were degraded at specific sites within the SARS-CoV-2 protein fragment . When developing RBD-based reagents or vaccine candidates, stability assessments under various storage conditions and temperatures are essential to ensure consistent performance in downstream applications.

What basic validation methods confirm the structural integrity and functionality of expressed SARS-CoV-2 S1 (319-541)?

Validation of expressed SARS-CoV-2 S1 (319-541) should encompass both structural integrity and functional assessments. Structural validation typically begins with SDS-PAGE analysis to confirm the expected molecular weight and purity . For functional validation, several approaches are employed: (1) Antibody binding assays to verify that the recombinant protein is recognized by antibodies directed against the appropriate SARS-CoV-2 antigenic regions ; (2) ACE2 receptor binding assays, which can be measured via flow cytometry, to confirm that the expressed RBD retains its capacity to interact with its cellular receptor ; (3) Inhibition assays to demonstrate that the RBD can block the interaction between the virus and ACE2 . Additionally, glycosylation analysis may be performed on RBD expressed in mammalian or insect cell systems to confirm appropriate post-translational modifications . These validation methods collectively ensure that the expressed RBD maintains both its structural integrity and biological functionality, which is critical for its application in research, diagnostics, or vaccine development.

How do glycosylation patterns of SARS-CoV-2 S1 (319-541) expressed in Sf9 cells compare with those from mammalian expression systems?

Glycosylation patterns of SARS-CoV-2 S1 (319-541) vary significantly across expression systems, with important functional implications. While specific data on Sf9-expressed RBD glycosylation is not directly provided in the search results, comparative studies between CHO and HEK293 expression systems reveal notable differences. CHO-RBD predominantly displays core 1 structures with two sialic acids (H1N1S2), whereas HEK293-RBD exhibits more complex glycosylation patterns . The level of sialylation is particularly variable, with significant differences observed not only between expression systems but also between different constructs of the spike protein. Full-length S protein expressed in HEK293 cells showed only 22% sialylation on N331 and 4% on N343, while the S1 subunit carried 80% and 50% sialylation on these sites, respectively . This demonstrates that glycosylation can change with the length of the expressed protein (S, S1, or RBD) and must be considered when selecting an expression system. Insect cell systems like Sf9 typically produce proteins with simpler, high-mannose glycans compared to the complex glycans of mammalian systems, which may impact immunogenicity and receptor-binding characteristics of the expressed RBD.

What strategies have proven most effective for enhancing the immunogenicity of recombinant SARS-CoV-2 S1 (319-541)?

Several strategies have demonstrated effectiveness in enhancing the immunogenicity of recombinant SARS-CoV-2 S1 (319-541). Fc fusion technology has shown particular promise, with RBD-hFc fusion proteins inducing robust immune responses including increased populations of memory CD4+ and CD8+ T cells, indicating enhanced cellular immunity . The human IgG Fc domain not only improves the half-life of the antigen but also facilitates recognition by immune cells. Adjuvant formulation represents another critical strategy, with studies showing that combination of recombinant S1 proteins with AS03 adjuvant elicits strong neutralizing antibody responses . Bivalent vaccine approaches, incorporating both wild-type and mutant S1 proteins, have demonstrated ideal neutralization properties against multiple variants including B.1.351 . This suggests that presenting multiple epitopes simultaneously can broaden protective immunity against emerging variants. Additionally, carrier protein fusion strategies, such as CBM9-RBD fusions, have been explored to enhance both production efficiency and immunogenicity . The choice among these strategies depends on research goals, with each approach offering distinct advantages for different applications in vaccine development or immunological studies.

What are the comparative advantages of using SARS-CoV-2 S1 (319-541) expressed in Sf9 cells versus other systems for structural studies?

While the search results don't explicitly discuss Sf9-expressed SARS-CoV-2 S1 (319-541) for structural studies, the comparative advantages can be inferred from general principles and the information provided about other expression systems. Insect cell expression systems like Sf9 offer several advantages for structural studies: they provide high expression levels of properly folded proteins with post-translational modifications, albeit with simpler glycosylation patterns than mammalian systems. The unique glycosylation profile of insect cell-expressed proteins can sometimes facilitate crystallization for X-ray crystallography studies. In contrast, mammalian expression systems like HEK293 and CHO cells produce RBD with more complex glycosylation patterns that more closely resemble native viral proteins . This makes them potentially better suited for functional studies but potentially more challenging for some structural techniques. E. coli expression systems, while lacking glycosylation, can produce high yields of protein (approximately 100 mg/L) and may be suitable for structural studies of non-glycosylated protein domains . The choice between expression systems for structural studies ultimately depends on the specific technique (X-ray crystallography, cryo-EM, NMR) and the research question being addressed.

How can SARS-CoV-2 S1 (319-541) be engineered to develop universal vaccines effective against emerging variants?

Engineering SARS-CoV-2 S1 (319-541) for universal vaccine development requires strategic approaches to address antigenic diversity among emerging variants. One promising strategy involves developing bivalent vaccines that incorporate both wild-type S1 protein (S1-WT) and mutant S1 proteins (S1-Mut) containing key mutations (K417N, E484K, N501Y, D614G) . This approach has demonstrated ideal neutralization properties against both wild-type and variant strains, including B.1.351. Another approach focuses on targeting conserved epitopes within or adjacent to the RBD region. Research has identified a conserved region immediately C-terminal to the RBD (amino acids 562-579) that is widely recognized by human convalescent sera and contains a putative protective epitope . Antibodies directed against this region correlate with neutralization activity, and depletion of these antibodies sharply reduces neutralization capacity. Additionally, engineering RBD-Fc fusion proteins enhances immunogenicity while potentially broadening protection against variants . Structure-based design approaches, informed by high-resolution complex analyses of neutralizing antibodies and RBD, can further guide the development of immunogens that present conserved neutralizing epitopes while minimizing exposure of variant-specific regions .

What are the most advanced methods for assessing neutralizing antibody responses against SARS-CoV-2 S1 (319-541) from different expression systems?

Assessment of neutralizing antibody responses against SARS-CoV-2 S1 (319-541) requires sophisticated methodologies that balance throughput with physiological relevance. Pseudovirus neutralization assays represent a robust approach that correlates well with authentic virus neutralization while allowing for safer handling in BSL-2 facilities . For high-throughput screening, RBD-ACE2 binding inhibition assays measure the ability of antibodies to block the interaction between RBD and ACE2, which can be quantified by flow cytometry . This approach provides a surrogate measure of neutralization potential. Assessments of cross-neutralization against variant strains are particularly important, requiring parallel testing against multiple RBD variants to identify broadly neutralizing responses . More comprehensive evaluation includes measurement of antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), especially for Fc-fusion constructs where Fc effector functions may contribute to protection . In animal models, viral challenge studies provide the most definitive assessment of protection, with complete viral clearance following antibody treatment representing the gold standard for efficacy . The integration of these methods provides a comprehensive picture of neutralizing antibody responses against RBD proteins from different expression systems.

What are the optimal transfection and expression conditions for producing SARS-CoV-2 S1 (319-541) in Sf9 cells?

While the search results don't provide specific details about Sf9 expression of SARS-CoV-2 S1 (319-541), general methodological principles can be derived from other expression systems described. For optimal expression in insect cells, the SARS-CoV-2 RBD (319-541) gene sequence should be codon-optimized for Sf9 cells and cloned into a baculovirus transfer vector with an appropriate secretion signal and purification tag (commonly His-tag or Fc-fusion) . Transfection typically involves co-transfection of insect cells with the transfer vector and linearized baculovirus DNA, followed by viral amplification through multiple passages. Expression conditions generally include infection of high-density Sf9 cell cultures (2-3×10^6 cells/ml) with optimized MOI (multiplicity of infection), followed by incubation at 27-28°C for 48-72 hours post-infection. The supernatant containing secreted RBD can be harvested by centrifugation and filtration. For comparison, mammalian expression systems have achieved successful expression using transfection of expression vectors containing His-tagged or Fc-fusion RBD constructs into HEK293 cells , while E. coli systems have achieved expression levels of approximately 100 mg/L using IPTG induction . Scale-up strategies for all systems benefit from optimized fed-batch bioreactor protocols to maximize yield and consistency.

What analytical methods are most effective for characterizing glycosylation patterns of SARS-CoV-2 S1 (319-541)?

Characterization of glycosylation patterns on SARS-CoV-2 S1 (319-541) requires sophisticated analytical techniques to elucidate the complex post-translational modifications that impact protein function. Mass spectrometry-based approaches are particularly valuable, enabling detailed analysis of glycan structures and their attachment sites. Studies comparing CHO and HEK293-expressed RBD revealed significant differences in glycosylation patterns, with CHO-RBD primarily displaying core 1 structures with two sialic acids (H1N1S2) while HEK293-RBD exhibited more complex glycosylation . The analysis demonstrated sialylation levels vary significantly between expression systems and even between different constructs (full S protein versus S1 subunit). Quantitative assessment of site-specific glycosylation revealed dramatic differences in sialylation at N331 (22% in full S protein versus 80% in S1 subunit) and N343 (4% versus 50%) . Functional glycan analysis can be performed by comparing antibody binding properties of native and deglycosylated RBD, which showed correlation coefficients ranging from R²=0.9681 to R²=0.9905, indicating that glycosylation affects but does not completely disrupt antibody recognition . These analytical methods are essential for ensuring consistency in glycosylation between production batches and for understanding how glycosylation impacts immunogenicity and receptor binding.

How can researchers optimize purification protocols for SARS-CoV-2 S1 (319-541) to maximize yield and purity?

Optimizing purification protocols for SARS-CoV-2 S1 (319-541) requires a systematic approach that balances yield, purity, and maintenance of biological activity. For His-tagged constructs, immobilized metal affinity chromatography (IMAC) using Ni-NTA or Co-NTA resins provides a robust initial capture step . For Fc-fusion proteins, Protein A or Protein G affinity chromatography offers highly selective purification . A novel approach using CBM9-fusion proteins enables single-step purification with inexpensive cellulose powder, achieving yields of approximately 0.1 g/L in E. coli expression systems . Following affinity purification, size exclusion chromatography helps remove aggregates and degradation products, which is particularly important as some RBD constructs show susceptibility to proteolytic degradation . The addition of protease inhibitors during early purification stages can help preserve protein integrity. Buffer optimization is crucial, with pH, salt concentration, and stabilizing excipients adjusted to maximize stability of the purified protein. Quality control should include SDS-PAGE analysis to confirm purity and molecular weight , followed by functional assays such as ACE2 binding or antibody recognition to verify that purification has not compromised biological activity . Implementation of these optimized protocols can significantly enhance both yield and quality of the final purified product.

What are the key considerations for designing in vitro assays to evaluate binding affinity between SARS-CoV-2 S1 (319-541) and human ACE2?

Designing robust in vitro assays to evaluate binding affinity between SARS-CoV-2 S1 (319-541) and human ACE2 requires careful consideration of multiple factors. Flow cytometry-based approaches have proven effective for measuring the inhibition of RBD-hFc binding to cell-expressed ACE2, providing quantitative assessment of binding interactions and their inhibition by antibodies or other blocking agents . Surface plasmon resonance (SPR) or bio-layer interferometry (BLI), while not explicitly mentioned in the search results, represent gold-standard techniques for determining binding kinetics and affinity constants. When designing these assays, researchers must consider the orientation and density of immobilized proteins, as these factors can significantly impact measured affinities. The source and quality of recombinant proteins is critical – differences in glycosylation between expression systems may affect binding characteristics, as suggested by the studies on CHO and HEK293-expressed RBD . Assay validation should include appropriate positive and negative controls, and where possible, correlation with functional viral neutralization. When evaluating binding to variant RBDs, parallel testing against wild-type and mutant proteins allows direct comparison of affinities . These considerations ensure that binding assays provide meaningful data that correlate with biologically relevant interactions between the virus and its cellular receptor.

How can researchers design experiments to compare the immunogenicity of SARS-CoV-2 S1 (319-541) from different expression systems?

Designing comprehensive experiments to compare the immunogenicity of SARS-CoV-2 S1 (319-541) from different expression systems requires a multifaceted approach that evaluates both humoral and cellular immune responses. Animal models, such as transgenic mice expressing human ACE2, provide an ideal platform for these comparisons . Immunization protocols should include consistent dosing regimens, typically with two immunizations spaced 2-3 weeks apart, using equivalent protein quantities from each expression system . Adjuvant selection is critical, with AS03 demonstrating effectiveness in enhancing immune responses to S1 proteins . Assessment of humoral immunity should include measurement of total binding antibodies by ELISA and functional neutralizing antibodies using pseudovirus or authentic virus neutralization assays. The inhibition of RBD-ACE2 binding provides a surrogate measure of neutralization potential . For cellular immunity, evaluation should include quantification of IL-4 and IFN-γ secreting cells in splenocytes following stimulation with the recombinant proteins, as well as analysis of memory CD4+ and CD8+ T cell populations by flow cytometry . Cross-neutralization against variant strains is particularly important to assess breadth of protection . These comprehensive immunological analyses enable direct comparison of the quantity, quality, and functionality of immune responses elicited by RBD proteins from different expression systems, informing optimal choices for vaccine development.

How can researchers overcome the challenges of low expression yields of SARS-CoV-2 S1 (319-541) in different systems?

Researchers face significant challenges with low expression yields of SARS-CoV-2 S1 (319-541), particularly given its apparent molecular weight of approximately 27 kDa . Several strategies can address these yield limitations across different expression systems. For E. coli expression, fusion with carrier proteins like CBM9 has achieved yields of approximately 100 mg/L upon IPTG induction . These yields could be further increased through implementation of optimized fed-batch bioreactor protocols rather than standard research growth flasks . For insect and mammalian expression systems, codon optimization for the specific host cell line significantly enhances translation efficiency. Signal sequence optimization improves secretion efficiency, particularly important for Sf9 insect cell expression. For all systems, optimization of culture conditions including temperature, media composition, and induction timing can dramatically impact yields. In mammalian systems like HEK293 and CHO cells, transient expression can be enhanced through co-expression of chaperones to assist protein folding. For stable cell line development, selection of high-producing clones followed by process optimization can establish consistently high yields. Implementation of these strategies, tailored to the specific expression system, can overcome yield limitations and establish robust production platforms for SARS-CoV-2 S1 (319-541) protein for research or vaccine development applications.

What approaches effectively overcome stability issues with recombinant SARS-CoV-2 S1 (319-541) proteins?

Stability challenges with recombinant SARS-CoV-2 S1 (319-541) proteins significantly impact their utility in research and therapeutic applications. Studies have shown variable susceptibility to proteolytic degradation among different constructs, with certain fusion proteins demonstrating enhanced resistance . A systematic approach to overcome stability issues includes several complementary strategies. Fusion partner selection is critical – some CBM9-spike protein fragment fusions have demonstrated substantial resistance to proteolytic degradation compared to other constructs . Human Fc fusion not only enhances immunogenicity but also improves the pharmacokinetic profile through extended half-life . Buffer optimization, including adjustment of pH, ionic strength, and addition of stabilizing excipients like glycerol or sucrose, can significantly enhance storage stability. For long-term storage, lyophilization with appropriate cryoprotectants preserves structure and function. Protein engineering approaches targeting identified degradation-prone regions can substantially improve stability – this requires identification of proteolytic cleavage sites through mass spectrometry analysis of degradation products. The choice of expression system also impacts stability, as post-translational modifications, particularly glycosylation patterns, influence protein solubility and resistance to degradation . Implementation of these complementary approaches can overcome stability challenges and ensure consistent performance of recombinant SARS-CoV-2 S1 (319-541) proteins in downstream applications.

What strategies can resolve inconsistent glycosylation patterns when expressing SARS-CoV-2 S1 (319-541) in different systems?

Glycosylation heterogeneity presents a significant challenge when expressing SARS-CoV-2 S1 (319-541) across different systems, with important implications for structure, function, and immunogenicity. Research has revealed dramatic differences in glycosylation patterns between expression systems, with CHO cells producing predominantly core 1 structures with two sialic acids while HEK293 cells generate more complex glycosylation patterns . Several approaches can address this heterogeneity. System selection forms the foundation – mammalian cells provide complex glycans, insect cells produce simpler high-mannose structures, and yeast systems generate highly mannosylated glycans. Within each system, genetic engineering strategies can enhance consistency, including knockout of certain glycosyltransferases or introduction of additional ones to humanize glycosylation in non-human systems. Process optimization significantly impacts glycosylation – controlling parameters like temperature, pH, dissolved oxygen, and nutrient availability during cultivation. For applications requiring homogeneous glycosylation, enzymatic remodeling of glycans post-purification can generate more uniform structures. Alternatively, removal of N-linked glycans using enzymes like PNGase F produces deglycosylated proteins that maintain core functionality – studies show high correlation between antibody binding to glycosylated and deglycosylated RBD (R²=0.9681-0.9905) . These complementary approaches enable researchers to either control glycosylation heterogeneity or mitigate its effects on experimental outcomes.

How can researchers effectively compare neutralizing activity against both wild-type and variant forms of SARS-CoV-2 using recombinant S1 (319-541)?

Effective comparison of neutralizing activity against both wild-type and variant SARS-CoV-2 using recombinant S1 (319-541) requires carefully designed assays and controls to ensure valid cross-variant comparisons. A comprehensive approach begins with parallel production of multiple recombinant RBD variants, including wild-type and those containing key mutations (K417N, E484K, N501Y, D614G) . Standardization of protein quality and quantity is essential for valid comparisons. Functional assays should include measurement of RBD-ACE2 binding inhibition, which can be quantified using flow cytometry or plate-based assays. Pseudovirus neutralization assays using viruses displaying the corresponding spike variants provide a more physiologically relevant assessment of neutralization potential. For highest confidence, authentic virus neutralization assays with appropriate biocontainment can be performed. The development of bivalent or multivalent reference standards containing both wild-type and variant RBDs enables internal calibration of assays . Animal studies in appropriate models, such as transgenic mice expressing human ACE2, allow assessment of in vivo protection against challenge with wild-type and variant viruses . This multi-tiered approach to neutralization assessment provides comprehensive data on cross-neutralization capacity, guiding development of broadly protective vaccines and therapeutics against current and emerging SARS-CoV-2 variants.

Product Science Overview

Introduction

The Coronavirus 2019-nCoV, also known as SARS-CoV-2, is the virus responsible for the COVID-19 pandemic. One of the critical components of this virus is the spike (S) glycoprotein, which plays a crucial role in the virus’s ability to infect host cells. The spike glycoprotein is composed of two subunits, S1 and S2. The S1 subunit contains the receptor-binding domain (RBD), which is responsible for binding to the host cell receptor, angiotensin-converting enzyme 2 (ACE2). The recombinant form of this protein, expressed in SF9 insect cells, is used extensively in research and vaccine development.

Structure and Function

The spike glycoprotein of SARS-CoV-2 is a large type I transmembrane protein that protrudes from the viral envelope. It is responsible for mediating the entry of the virus into host cells. The S1 subunit of the spike protein contains the receptor-binding domain (RBD), which directly interacts with the ACE2 receptor on the surface of host cells . This interaction is the first step in the viral entry process, leading to the fusion of the viral and cellular membranes.

The spike protein is heavily glycosylated, with numerous N-linked glycans that play roles in protein folding, stability, and immune evasion . The glycosylation patterns can also affect the antigenicity of the spike protein, influencing the immune response.

Recombinant Expression in SF9 Cells

The recombinant form of the SARS-CoV-2 spike glycoprotein S1 receptor-binding domain is often produced using the baculovirus expression system in SF9 insect cells. This system is advantageous due to its ability to produce large quantities of protein with post-translational modifications similar to those in mammalian cells . The recombinant protein can be used for various applications, including structural studies, vaccine development, and serological assays.

Applications in Research and Vaccine Development

The recombinant spike glycoprotein S1 RBD is a critical tool in the development of vaccines and therapeutic antibodies. By studying the structure and function of the RBD, researchers can design vaccines that elicit strong neutralizing antibody responses . Additionally, the recombinant protein can be used in serological assays to detect antibodies in individuals who have been exposed to the virus or vaccinated.

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.