CoV-2 S1 (16-685), Fc is produced under stringent conditions:
Expression: HEK293 or CHO cells ensure proper glycosylation and folding .
Purification: Affinity chromatography (e.g., Protein A/G for Fc tags) followed by endotoxin removal (<1.0 EU/µg) .
Validation:
Antibody-Dependent Cellular Cytotoxicity (ADCC): CoV-2 S1 (16-685), Fc elicits antibodies that activate NK cells via FcγRIIIa (CD16), though hybrid immunity (infection + vaccination) enhances ADCC potency compared to vaccination alone .
Neutralizing Antibody Titers: Immunization with S1-Fc fusion proteins induces 5-fold higher affinity antibodies than S2-targeted antigens, correlating with viral neutralization efficacy .
Broad-Spectrum Epitopes: The S1-Fc protein identifies conserved epitopes across SARS-CoV-2 variants (Alpha, Beta, Delta) .
Monoclonal Antibody Screening: Phage display libraries using S1-Fc have yielded high-affinity neutralizing antibodies, including cross-reactive VHH-Fc constructs .
Neuroinflammation: S1-Fc activates TLR2/TLR4 signaling in rodent models, inducing social avoidance and inflammatory gene expression in brain regions .
Post-Vaccine Symptoms: S1 protein persists in CD16+ monocytes for months post-vaccination, potentially driving cytokine dysregulation (e.g., elevated IL-6, IL-8) .
Glycosylation Variability: Batch-to-batch glycosylation differences may impact antibody recognition .
Variant Compatibility: Ongoing research assesses S1-Fc efficacy against emerging variants (e.g., Omicron) .
Therapeutic Optimization: Engineering Fc domains to enhance effector functions (e.g., ADCC, phagocytosis) is under exploration .
The 2019 novel coronavirus (2019-nCoV), a human-infecting coronavirus responsible for causing viral pneumonia, emerged in December 2019 in a fish market in Wuhan, Hubei province, China.
Genetic analysis reveals that 2019-nCoV shares 87% of its identity with the bat-derived severe acute respiratory syndrome coronavirus 2018 (SARS-CoV-2) discovered in Zhoushan, eastern China. Despite some structural differences, the receptor-binding domain (RBD) of 2019-nCoV is similar to that of 2018 SARS-CoV, suggesting a potential binding affinity for the human angiotensin-converting enzyme 2 (ACE2) receptor protein.
While bats are considered the likely reservoir host of 2019-nCoV, researchers hypothesize that an intermediary animal host, potentially from the seafood market, played a role in its transmission to humans. Recombination analysis indicates that the spike glycoprotein of 2019-nCoV is a product of recombination between a bat coronavirus and an unidentified coronavirus.
This recombinant protein, expressed in HEK293 cells, encompasses amino acids 16 to 685 of the Spike Glycoprotein S1 subunit from the Wuhan-Hu-1 strain of Coronavirus 2019 (CoV-2). It is fused with an Fc tag at the C-terminus.
The CoV-2 S1 protein is supplied in a buffer containing sodium chloride, glycine, arginine, adjusted to a pH of 7.5, and supplemented with 5% trehalose.
Lyophilized CoV-2 Spike S1 Glycoprotein exhibits stability at room temperature for up to 3 weeks; however, it is recommended to store it desiccated at a temperature below -18°C. Once reconstituted, the CoV2 Spike protein can be stored at 4°C for 2 to 7 days. For extended storage, it is advisable to store it below -18°C. Adding a carrier protein, such as 0.1% HSA or BSA, is recommended for long-term storage. Repeated freeze-thaw cycles should be avoided.
The purity of the protein exceeds 95%, as determined by SDS-PAGE analysis.
HEK293 Cells.
Purified by Protein-G chromatographic technique.
The SARS-CoV-2 S1 (16-685) domain encompasses amino acids 16-685 of the Spike protein's S1 subunit, which contains the receptor-binding domain (RBD) essential for viral entry. This region is particularly significant as it contains a "C-end rule" (CendR) motif (682RRAR685) not conserved in SARS and MERS coronaviruses . This motif significantly potentiates viral entry into cells and allows interaction with neuropilin-1 (NRP-1), potentially affecting pain signaling pathways through competition with vascular endothelial growth factor-A (VEGF-A) . When fused to an Fc domain, the resulting S1-Fc construct offers extended half-life, enhanced stability, and simplified purification through protein A/G affinity chromatography, making it valuable for studying antibody responses, receptor binding, and protein-protein interactions.
The S1 (16-685) domain primarily mediates interaction with the ACE2 receptor through its RBD. Additionally, research has revealed that this domain interacts with neuropilin-1 (NRP-1) via its CendR motif. Investigations using VEGF-A, a physiological ligand for the b1b2 pocket in NRP-1, have shown that the Spike protein can compete with VEGF-A for NRP-1 binding, potentially affecting VEGF-A/NRP-1 signaling and associated pain behaviors . This dual-receptor binding capability makes S1 constructs particularly valuable for studying viral entry mechanisms and developing therapeutic interventions targeting these interactions.
S1-Fc fusion proteins typically combine the S1 domain (aa 16-685) of the SARS-CoV-2 spike protein with an immunoglobulin Fc region. This construct retains the receptor-binding domain (RBD) and the full binding capabilities of the S1 subunit while gaining the advantages of the Fc domain. The Fc component enhances stability and half-life, facilitates purification, and enables detection with anti-Fc antibodies. The structure includes the CendR motif (682RRAR685) that enables interaction with neuropilin-1 . When used in immunization studies, these S1-Fc constructs can induce antibody responses with binding characteristics that differ from those elicited by other spike protein domains, as demonstrated in comparative immunization experiments .
Detection of S1 protein in biological samples employs several methodological approaches:
Flow cytometry: Used to detect S1 protein in specific cell populations such as CD16+ monocytes .
Mass spectrometry: Provides definitive identification of S1, S1 mutant, and S2 peptide sequences in cellular samples .
ELISA-based serological assays: Quantify both serum IgG and IgA against the S1 protein .
Surface Plasmon Resonance (SPR): Measures binding kinetics and affinity of antibodies against S1 proteins .
For example, in studies of post-vaccination individuals with PASC-like symptoms, researchers combined flow cytometry and mass spectrometry to detect persistent S1 protein in CD16+ monocytes, enabling correlation with clinical symptoms and inflammatory markers .
The expression and purification of high-quality S1-Fc fusion proteins typically follows this optimized protocol:
Vector Construction:
Clone the S1 (16-685) sequence into a mammalian expression vector containing an in-frame Fc domain
Include a signal peptide for secretion and appropriate restriction sites
Verify the construct by sequencing
Expression System:
Transfect HEK293T cells for proper glycosylation and folding
Culture in serum-free media to simplify downstream purification
Harvest cell supernatant after 72-96 hours
Purification Steps:
Clarify supernatant by centrifugation (5000×g, 15 minutes) and filtration (0.45μm)
Perform Protein A/G affinity chromatography (binding at pH 7.4, elution at pH 2.8)
Immediately neutralize eluted fractions with 1M Tris-HCl pH 9.0
Conduct size exclusion chromatography to remove aggregates
Concentrate using 30 kDa MWCO concentrators
Quality Control:
This protocol maximizes yield while preserving the critical structural features of the S1 domain, including the CendR motif that interacts with the NRP-1 receptor .
Differentiating between S1-binding antibodies and functionally neutralizing antibodies requires a multi-assay approach:
Binding Assays:
ELISA to quantify total binding antibodies against S1
Surface Plasmon Resonance (SPR) to determine antibody affinity and off-rate constants
Flow cytometry to assess binding to cell-surface expressed S1
Functional Neutralization Assays:
Correlation Analysis:
Calculate Spearman's rank correlation between binding titers and neutralization activity
Generate scatter plots of ELISA/SPR results versus neutralization to identify thresholds
Determine positive predictive value of binding assays for neutralization
Research has shown that antibodies generated against different spike protein domains (S1+S2, S1, RBD, S2) have different neutralization capacities despite showing binding in ELISA assays . For example, anti-RBD antibodies demonstrated 5-fold higher affinity and strong neutralization, while anti-S2 antibodies showed binding but minimal neutralization . Therefore, binding assays alone are insufficient to characterize the functional protective capacity of antibodies.
Detection of persistent S1 protein in monocyte subsets requires careful attention to several technical factors:
Sample Preparation:
Isolation of PBMCs using density gradient centrifugation
Immediate processing or proper cryopreservation to maintain cell viability
Careful handling to avoid ex vivo activation of monocytes
Flow Cytometric Analysis:
Multi-parameter approach including CD14 and CD16 to identify monocyte subsets (classical, intermediate, and non-classical)
Fixation and permeabilization optimization to enable intracellular S1 detection without destroying epitopes
Inclusion of multiple control samples (healthy unexposed, COVID-19 recovered, vaccinated asymptomatic)
Use of specific anti-S1 antibodies with validation using recombinant S1 protein
Mass Spectrometry Confirmation:
Data Analysis:
Research using these approaches has identified persistent S1 protein in CD16+ monocytes up to 245 days post-vaccination, correlating with pro-inflammatory cytokine production and symptoms resembling long COVID .
S1-Fc constructs provide valuable tools to investigate the interaction between SARS-CoV-2 spike protein and neuropilin-1 (NRP-1) signaling, which appears to involve the CendR motif (682RRAR685) . Researchers can employ the following methodological approaches:
Competition Binding Assays:
Pre-incubate cells expressing NRP-1 with varying concentrations of S1-Fc
Add fluorescently-labeled VEGF-A (a physiological ligand for NRP-1)
Measure displacement of VEGF-A binding using flow cytometry or confocal microscopy
Calculate IC50 values to quantify binding affinity
Signaling Pathway Analysis:
Treat NRP-1-expressing cells with S1-Fc, VEGF-A, or both
Measure downstream signaling events (phosphorylation of specific targets, calcium flux)
Use selective inhibitors to dissect pathway components
Compare signaling responses between neuronal and vascular endothelial cells
Functional Outcome Assessment:
In neuronal models, measure pain-related signaling responses
In vascular models, assess permeability and endothelial activation
Employ both in vitro cellular systems and ex vivo tissue preparations
Compare wild-type S1-Fc with constructs containing mutated CendR motifs
This approach has already revealed that SARS-CoV-2 spike protein can co-opt the VEGF-A/NRP-1 pathway, potentially explaining some neurological symptoms observed in COVID-19. Research has shown elevated VEGF-A levels in bronchial alveolar lavage fluid from COVID-19 patients, with lower levels in asymptomatic compared to symptomatic individuals , suggesting this pathway may contribute to symptom manifestation.
The Coronavirus 2019 Spike Glycoprotein-S1 (16-685 a.a.), Fc Recombinant, is a crucial component in the study of SARS-CoV-2, the virus responsible for the COVID-19 pandemic. This recombinant protein is a fragment of the spike (S) glycoprotein, specifically the S1 subunit, which plays a vital role in the virus’s ability to infect host cells.
The spike glycoprotein of SARS-CoV-2 is a large, trimeric protein that protrudes from the viral surface. It is composed of two subunits: S1 and S2. The S1 subunit, which spans amino acids 16 to 685, contains the receptor-binding domain (RBD) that directly interacts with the host cell receptor, angiotensin-converting enzyme 2 (ACE2). This interaction is critical for viral entry into host cells .
The Fc region in the recombinant protein refers to the fragment crystallizable region of an antibody, which is often fused to other proteins to enhance their stability and facilitate purification. In this context, the Fc fusion helps in the production and purification of the spike glycoprotein-S1 fragment.
The recombinant spike glycoprotein-S1 is extensively used in research to understand the mechanisms of viral entry, immune response, and vaccine development. Studies have shown that the spike S1 domain can act as a TLR4 agonist, upregulating ACE2 expression and inducing a pro-inflammatory response . This makes it a valuable tool for studying the pathogenesis of COVID-19 and the body’s immune response to the virus.