CoV-2 S1 (1-681)

Coronavirus 2019-nCoV Spike Glycoprotein-S1 (1-681), Recombinant
Shipped with Ice Packs
In Stock

Description

Immune Activation Mechanisms

Immobilized S1 (1-681) activates human monocytes to produce cytokines central to COVID-19 cytokine release syndrome (CRS):

Cytokine production in S1-stimulated monocytes :

CytokineFold Increase vs ControlClinical Relevance
IL-64.8×Drives acute-phase inflammation
TNF-α3.2×Promotes vascular permeability
IL-1β2.9×Pyrogen and tissue remodeler

This activation requires the NTD's galectin-fold but not ACE2-binding regions (S1-CTD/RBD), suggesting a lectin-like signaling mechanism .

Viral Evolution and Functional Mutations

The S1/S2 furin cleavage site (PRRAR<sup>681-685</sup>) underwent critical evolutionary changes:

Impact of key mutations:

MutationVariantEffect on S1 Function
D614GGlobalStabilizes S1-S2 interaction
P681RDeltaEnhances furin cleavage by 2.7×
N501YAlpha/BetaIncreases ACE2 binding affinity 4.5×

These mutations demonstrate evolutionary pressure to optimize S1's dual roles in receptor engagement and immune evasion.

Therapeutic Applications

Vaccine development:

  • A VSV-vectored S1 vaccine (ConVac) induced neutralizing antibodies (ID<sub>50</sub> 1:512) and protected hamsters from lung viral replication (3.7-log reduction vs control) .

Antiviral targets:

TargetStrategyEfficacy
Galectin-foldCompetitive inhibitorsReduced IL-6 by 68% in vitro
RBD-ACE2 interfaceMonoclonal antibodiesNeutralization IC<sub>50</sub> 0.8 μg/mL

Biochemical Properties

Physical characteristics:

  • Molecular weight: ~130 kDa (glycosylated)

  • Isoelectric point: 6.2-6.8 (variation due to glycosylation)

  • Expression systems: HEK293 (95% purity), insect cells (82% purity)

Receptor binding kinetics :

ParameterValue
ACE2 K<sub>D</sub>14.7 ± 1.3 nM
Association rate (k<sub>on</sub>)2.1×10<sup>5</sup> M<sup>-1</sup>s<sup>-1</sup>
Dissociation rate (k<sub>off</sub>)3.0×10<sup>-3</sup> s<sup>-1</sup>

Product Specs

Introduction

Initially identified in Wuhan, China, in December 2019, the 2019 novel coronavirus (2019-nCoV) is a human coronavirus responsible for causing viral pneumonia. This virus shares significant genetic similarities with the bat-derived SARS-CoV-2, with 87% similarity to a strain discovered in Zhoushan, eastern China. The 2019-nCoV possesses a receptor-binding domain (RBD) structure analogous to that of the 2018 SARS-CoV, suggesting a potential to bind to the human ACE2 receptor (angiotensin-converting enzyme 2). Although bats are considered the likely reservoir of 2019-nCoV, an intermediary animal host, possibly from seafood sold at Wuhan's Huanan Seafood Wholesale Market, is suspected. Research indicates that the 2019-nCoV's spike glycoprotein may be a product of recombination between a bat coronavirus and an unknown coronavirus.

Description

This recombinant protein, derived from HEK293 cells, consists of the S1 subunit of the Coronavirus 2019-nCoV Spike Glycoprotein (Wuhan-Hu-1 strain). It encompasses amino acids 1 to 681 and is fused with an Fc tag at the C-terminus, resulting in a molecular weight of 76 kDa.

Physical Appearance

The product is a clear solution that has undergone sterile filtration.

Formulation

The CoV-2 S1 protein solution is provided in phosphate-buffered saline (PBS) with a pH of 7.4.

Stability

The protein is shipped using ice packs to maintain a low temperature. Upon receiving the product, it should be stored at -20°C.

Purity

SDS-PAGE analysis confirms that the protein purity is greater than 95%.

Source

HEK293.

Purification Method

Purified by Protein-G chromatography technique.

Q&A

What is the significance of the S1/S2 furin cleavage site in SARS-CoV-2, particularly at position 681?

The S1/S2 furin cleavage site in SARS-CoV-2 plays a crucial role in virus transmissibility and infection properties. The site includes position 681, which is particularly significant for proteolytic processing. In the original SARS-CoV-2 sequence, the amino acid sequence at this site is 681-P-R-R-A-R|S-686, with proteolytic cleavage occurring between the arginine (R) and serine (S) residues. The cellular protease furin cleaves at this multibasic motif, requiring at minimum an R-x-x-R motif with a preference for an additional basic residue at P2 position (R-x-B-R). Mutations at position 681, such as P681R, provide an additional basic residue at the P5 position that may enhance S1/S2 cleavability by furin, potentially altering viral infection dynamics .

The presence of this furin cleavage site distinguishes SARS-CoV-2 from many other betacoronaviruses and enhances virus transmissibility. When studying the functional effects of modifications at this site, researchers should consider that successful viral variants typically combine mutations at this site with other less well-identified changes as part of natural selection processes .

How do SARS-CoV-2 S1-based serological assays compare to nucleocapsid (N)-based assays for research applications?

S1 and N-based serological assays both demonstrate high sensitivity and specificity for detecting SARS-CoV-2 antibodies, but each has distinct advantages for research applications. S1-based ELISAs showed an area under curve (AUC) of 0.938 ± 0.027 (95% CI 0.886–0.990) for IgG and 0.953 ± 0.021 (95% CI 0.911–0.995) for IgM detection. N-based ELISAs demonstrated an AUC of 0.977 ± 0.015 (95% CI 0.948–1.000) for IgG and 0.886 ± 0.037 (95% CI 0.812–0.959) for IgM .

The cut-off values for these assays were determined to be:

  • S1 IgG-ELISA: 0.17

  • S1 IgM-ELISA: 0.30

  • N IgG-ELISA: 0.40

  • N IgM-ELISA: 0.55

Importantly, both assays are complementary, and researchers have found significant correlation between IgG responses against S1 and N proteins. While S1 serves as the primary target for neutralizing antibodies, the N protein is less prone to mutations due to lower selective pressure, offering more stable detection over time. For comprehensive studies, researchers should consider using both S1 and N-based assays to capture the maximum number of SARS-CoV-2 positive cases .

What methods are available for producing recombinant SARS-CoV-2 S1 (1-681) protein for research purposes?

When producing recombinant SARS-CoV-2 S1 (1-681) for research applications, researchers must consider several methodological approaches based on the specific experimental requirements:

  • Expression Systems:

    • Mammalian expression systems: Preferred for producing properly folded and glycosylated S1 protein, critical for structural and functional studies. HEK293 and CHO cells are commonly used.

    • Insect cell expression: Offers a middle-ground between prokaryotic simplicity and mammalian glycosylation patterns.

    • Prokaryotic expression: While the S1 protein is naturally glycosylated, E. coli-based systems may be used for applications where glycosylation is not critical, or when analyzing specific peptide regions.

  • Purification Strategies:

    • Immobilized metal affinity chromatography (IMAC) using histidine tags

    • Affinity chromatography with antibodies or ACE2-Fc fusion proteins

    • Size exclusion chromatography for final polishing steps

  • Quality Control:

    • Western blot verification with anti-S1 antibodies

    • Mass spectrometry to confirm sequence integrity

    • Glycan profiling for mammalian-expressed proteins

    • Functional binding assays with ACE2 receptors

When designing experiments, researchers should note that unlike the nucleocapsid protein, the S1 subunit contains multiple glycosylation sites, making it more challenging to produce in prokaryotic systems but essential for maintaining native conformational epitopes in immunological studies .

How does the P681R mutation affect the structural and functional properties of the SARS-CoV-2 spike protein?

The P681R mutation, first identified in the A.23.1 variant in Uganda and later appearing in the widely transmitted Delta variant, introduces an additional basic residue at the P5 position of the S1/S2 cleavage site. This mutation significantly impacts the spike protein's processing and function in several ways:

For researchers investigating this mutation, it's essential to consider the entire sequence context rather than studying the mutation in isolation. The efficacy of the P681R mutation depends on complementary changes elsewhere in the spike protein or viral genome, highlighting the complex evolutionary dynamics of SARS-CoV-2 .

What neurological effects have been observed with SARS-CoV-2 S1 protein in experimental models?

Experimental studies involving direct administration of SARS-CoV-2 S1 protein to the mouse brain have revealed significant neurological impacts, providing potential mechanisms for the cognitive symptoms reported by COVID-19 patients. When S1 protein was introduced into the dorsal hippocampus of mice, the following effects were observed:

  • Cognitive Deficits: S1 protein-injected mice exhibited reduced discrimination capacity in novel object recognition and novel location tests compared to vehicle-injected controls, indicating impaired cognitive function .

  • Anxiety-like Behavior: In both elevated plus maze and open field tests, S1 protein-injected mice spent significantly less time in center areas and more time in closed/peripheral areas, demonstrating increased anxiety-like behavior .

  • Neuronal Death: Histological analysis revealed markedly reduced neuronal cell density in the CA1 and DG areas of both dorsal and ventral hippocampus. Specifically, S1 protein injection reduced dorsal hippocampal neurons by approximately 35% in both the CA1 and DG regions, while ventral hippocampal neurons decreased by about 20% .

  • Glial Cell Activation: Following S1 protein administration, GFAP-immunoreactive astrocytes increased notably, with fluorescence intensity rising by 59-63% in both dorsal and ventral hippocampus. Additionally, significant increases in Iba-1 immunoreactivity indicated microglial activation, with these cells displaying morphological changes characteristic of reactive microglia (more circular cell bodies and shorter branch lengths) .

These findings align with clinical observations of "brain fog" and cognitive impairments in COVID-19 patients and suggest that the S1 protein itself, even in the absence of active viral replication, may contribute to neurological manifestations of COVID-19 .

What is the antibody response kinetics against SARS-CoV-2 S1 protein following infection?

The antibody response against SARS-CoV-2 S1 protein follows a distinct temporal pattern after infection. Studies using S1-specific serological assays have characterized this kinetic profile:

  • Early Response: IgM and IgG antibodies specific to the S1 protein can be detected as early as the first week after symptom onset in RT-PCR confirmed COVID-19 patients .

  • Seroconversion Timing: Most patients seroconvert to IgG against both S1 and nucleocapsid (N) proteins by week 2 post-symptom onset, with antibody levels showing significant correlation with the number of days after symptoms appeared .

  • Antibody Correlation: There is a strong correlation between IgG responses against S1 and N proteins, suggesting that both can be effective markers for assessing immune status .

  • Cross-Reactivity Properties: S1-based serological assays demonstrate high specificity for SARS-CoV-2, with no significant cross-reactivity observed with antibodies against other human coronaviruses, including MERS-CoV, hCoV-OC43, hCoV-NL63, hCoV-229E, and hCoV-HKU1 .

This understanding of antibody kinetics is crucial for researchers designing serological studies, evaluating vaccine responses, or developing therapeutic antibodies. The early appearance of both IgM and IgG antibodies against S1 suggests that this protein domain elicits a rapid immune response, which may have implications for both protection and pathogenesis in COVID-19 .

How should researchers design serological studies to distinguish between natural infection and vaccination-induced antibody responses to S1?

When designing serological studies to differentiate between natural infection and vaccination-induced immunity, researchers should implement a multi-faceted approach:

  • Multiplex Antigen Profiling: Natural infection typically induces antibodies against multiple viral antigens, while most vaccines primarily induce S-specific antibodies. Include both S1 and N protein assays, as presence of N-specific antibodies generally indicates prior infection, not vaccination (for most current vaccines) .

  • Epitope-Specific Analysis: Design assays that detect antibodies against specific epitopes within S1 that are differentially presented in natural infection versus vaccination. For example:

    • RBD-specific antibodies (present in both scenarios)

    • N-terminal domain (NTD) antibodies (may show different profiles)

    • Antibodies against specific regions of S1 outside the RBD

  • Avidity Testing: Include antibody avidity measurements, as vaccination often produces higher-avidity antibodies compared to natural infection, particularly following booster doses.

  • IgG Subclass Profiling: Analyze IgG1/IgG3/IgG4 subclass distribution, which may differ between infection and vaccination responses.

  • Serological Algorithm: Implement a decision tree incorporating results from multiple assays:

    • S1+/N+ suggests natural infection

    • S1+/N- suggests vaccination without infection

    • Quantitative S1:N ratio analysis can help with ambiguous cases

  • Longitudinal Sampling: When possible, collect samples at multiple timepoints to capture temporal dynamics that may differ between vaccine and infection responses .

This comprehensive approach enables more accurate differentiation between immune responses and minimizes misclassification in epidemiological studies.

What methodological approaches can be used to study the neurotoxic effects of SARS-CoV-2 S1 protein in cellular and animal models?

To effectively investigate the neurotoxic effects of SARS-CoV-2 S1 protein, researchers should consider the following methodological approaches for both in vitro and in vivo studies:

  • In Vitro Cellular Models:

    • Primary Neuronal Cultures: Isolated from hippocampus or cortex of rodents to assess direct neurotoxicity through viability assays (MTT, LDH release), calcium imaging, and electrophysiology

    • Brain Organoids: Human-derived 3D cultures to model complex neural circuits and cell-cell interactions

    • Blood-Brain Barrier Models: Transwell systems with brain endothelial cells to study S1 penetration mechanisms

    • Mixed Glial-Neuronal Cultures: To examine cell-specific responses and intercellular signaling

  • In Vivo Animal Models:

    • Direct Hippocampal Injection: As demonstrated in the provided research, intracerebral administration of S1 protein can be used to assess local effects on cognition and neuronal viability

    • Intracerebroventricular Delivery: For broader CNS exposure

    • Systemic Administration: To determine if peripherally administered S1 can cross the BBB and induce central effects

    • Transgenic Models: Expressing human ACE2 receptors for greater translational relevance

  • Analytical Techniques:

    • Behavioral Testing: Novel object recognition, novel location tests, elevated plus maze, and open field tests to assess cognitive function and anxiety-like behavior

    • Immunohistochemistry: Using markers such as NeuN for neurons, GFAP for astrocytes, and Iba-1 for microglia to assess cellular responses

    • Cell Quantification: Stereological methods to accurately quantify neuronal loss in specific brain regions

    • Molecular Analysis: RNA-seq, proteomics, and metabolomics to identify mechanistic pathways

    • Functional Neuroimaging: In vivo imaging techniques to assess brain activity and metabolism

  • Controls and Variables to Consider:

    • Dose-Response Relationships: Test multiple concentrations of S1 protein

    • Timing of Administration: Acute versus chronic exposure

    • Regional Specificity: Compare effects in different brain regions

    • Age and Sex Differences: Include both male and female animals of different ages

    • Appropriate Controls: Heat-inactivated S1 protein, other viral proteins, or unrelated proteins of similar size

By employing these complementary approaches, researchers can comprehensively characterize the neurotoxic potential of SARS-CoV-2 S1 protein and elucidate mechanisms underlying neurological symptoms observed in COVID-19 patients.

How can researchers accurately assess the functional impact of mutations at position 681 in the SARS-CoV-2 spike protein?

To accurately assess the functional impact of mutations at position 681 in the SARS-CoV-2 spike protein, researchers should implement a comprehensive experimental framework:

  • Biochemical Cleavage Assays:

    • In vitro furin cleavage assays with purified recombinant spike proteins (wild-type vs. mutants)

    • Quantitative Western blotting to measure the ratio of cleaved to uncleaved spike protein

    • Enzyme kinetics analysis to determine changes in Km and Vmax parameters for furin processing

  • Cell-Based Systems:

    • Pseudovirus entry assays comparing wild-type and mutant spike proteins

    • Cell-cell fusion assays to measure membrane fusion efficiency

    • Live-cell imaging to track spike protein trafficking and processing

    • Flow cytometry for quantitative assessment of surface expression

  • Structural Analysis:

    • Cryo-electron microscopy to determine conformational changes induced by mutations

    • Molecular dynamics simulations to predict structural alterations around the cleavage site

    • Hydrogen-deuterium exchange mass spectrometry to assess local structural flexibility

  • Contextual Analysis:

    • Study mutations in isolated form and in combination with other variant-specific mutations

    • Compare effects in different genetic backgrounds (original strain vs. variant backbones)

    • Assess impact in the context of different cell types relevant to SARS-CoV-2 tropism

  • Viral Replication Studies:

    • Reverse genetics systems to generate recombinant viruses with specific mutations

    • Growth curve analysis in relevant cell lines and primary human airway cultures

    • Competition assays between wild-type and mutant viruses

  • Control Considerations:

    • Include multiple reference strains (original Wuhan strain, Alpha, Delta variants)

    • Analyze dose-dependent effects of furin inhibitors

    • Test impact of different proteases (TMPRSS2, cathepsins) on spike processing in mutants

The research indicates that changes at position 681 must be analyzed in their specific variant context, as the P681R mutation produced different effects in the A.23.1 spike compared to when introduced into the original SARS-CoV-2 spike. This suggests that successful mutations work in concert with other genomic changes through natural selection processes .

What are the key differences in antibody epitopes between SARS-CoV-2 S1 and corresponding regions in other human coronaviruses?

The S1 subunit of SARS-CoV-2 contains unique epitopes that distinguish it from other human coronaviruses, which has significant implications for diagnostic test development and cross-protection studies:

  • Sequence Homology Analysis:
    The SARS-CoV-2 S1 subunit shares only limited sequence identity with other human coronaviruses:

    • 64% with SARS-CoV

    • 57% with MERS-CoV

    • 9-37% with endemic human coronaviruses (hCoV-OC43, hCoV-NL63, hCoV-229E, hCoV-HKU1)

    This limited homology explains the high specificity of S1-based serological assays.

  • Receptor Binding Domain (RBD) Differences:
    The RBD within S1 is particularly variable across coronavirus species, reflecting adaptation to different cellular receptors:

    • SARS-CoV-2 and SARS-CoV: Both bind ACE2 but have significant structural differences in the receptor-binding motif

    • MERS-CoV: Binds DPP4 (CD26) instead of ACE2

    • Common cold coronaviruses: Utilize various receptors including APN (229E) and sialic acids (OC43)

  • Immunological Cross-Reactivity:
    Experimental data demonstrates that SARS-CoV-2 S1-based ELISAs specifically detect antibodies from COVID-19 seropositive samples but not those from individuals with prior exposure to other human coronaviruses, including MERS-CoV, hCoV-OC43, hCoV-NL63, hCoV-229E, or hCoV-HKU1 .

  • Epitope Mapping Considerations:
    When designing epitope mapping studies, researchers should focus on:

    • Regions unique to SARS-CoV-2 (for specificity)

    • Conserved regions across betacoronaviruses (for potential cross-protective responses)

    • Post-translational modifications, particularly glycosylation patterns that may affect epitope recognition

This high epitope specificity makes S1-based serological assays particularly valuable for distinguishing SARS-CoV-2 infections from other coronavirus infections without cross-reactivity concerns .

How does SARS-CoV-2 S1 protein induce neuroinflammatory responses, and what signaling pathways are involved?

SARS-CoV-2 S1 protein induces complex neuroinflammatory responses through multiple cellular mechanisms and signaling pathways. Based on experimental evidence from mouse models, the following mechanisms have been identified:

  • Glial Cell Activation:

    • Astrocyte Activation: S1 protein administration significantly increases GFAP-immunoreactive astrocytes in the hippocampus, with fluorescence intensity elevating by 59-63% in both dorsal and ventral regions .

    • Microglial Activation: Dramatic increases in Iba-1 immunoreactivity occur following S1 protein exposure, with microglia adopting reactive morphology (more circular cell bodies and shorter branch lengths) .

  • Neuronal Death Mechanisms:

    • S1 protein injection reduces hippocampal neuronal density by approximately 35% in CA1 and DG regions of the dorsal hippocampus and by 20% in the ventral hippocampus .

    • This neuronal loss correlates with cognitive deficits and anxiety-like behaviors in experimental animals.

  • Potential Signaling Pathways:
    Several signaling cascades are likely involved in S1-induced neuroinflammation:

    • TLR4 Signaling: S proteins can activate toll-like receptors, particularly TLR4, triggering NFκB-mediated inflammatory responses

    • NLRP3 Inflammasome Activation: Leading to IL-1β and IL-18 production

    • Oxidative Stress Pathways: Including NADPH oxidase activation and ROS production

    • Microglial Polarization: Shifting toward pro-inflammatory M1 phenotype

  • Blood-Brain Barrier Effects:

    • S1 protein may compromise BBB integrity through interaction with endothelial cells

    • This disruption could facilitate peripheral immune cell infiltration, amplifying neuroinflammation

  • Cognitive Impact Correlation:
    The neuroinflammatory changes induced by S1 protein directly correlate with behavioral deficits in:

    • Novel object recognition

    • Novel location tests

    • Elevated plus maze performance

    • Open field test behavior

These findings align with clinical observations of neurological symptoms in COVID-19 patients and suggest that the S1 protein alone, even without active viral replication, can induce significant neuroinflammation and subsequent cognitive impairment .

What are the comparative kinetics of antibody responses to different domains within SARS-CoV-2 S1 protein?

Antibody responses to different domains within SARS-CoV-2 S1 protein display distinct kinetic profiles that have important implications for diagnostics, therapeutics, and vaccine development. Current research reveals several key patterns:

  • Domain-Specific Response Timing:

    • RBD-specific antibodies: Typically appear first, detectable within the first week post-symptom onset

    • N-terminal domain (NTD) antibodies: Generally develop slightly later but reach comparable titers

    • Other S1 domain antibodies: Follow variable kinetics depending on epitope accessibility and immunogenicity

  • Antibody Class Switching:

    • IgM antibodies against S1 can be detected as early as the first week after symptom onset

    • Most patients seroconvert to IgG by the second week post-symptom onset

    • IgG antibodies against both S1 and nucleocapsid (N) proteins show significant correlation with days post-symptom onset

  • Neutralizing vs. Binding Antibody Development:

    • Initial antibody responses may bind S1 but lack neutralizing capacity

    • High-quality neutralizing antibodies typically develop 2-3 weeks post-infection

    • The ratio of neutralizing to binding antibodies increases over time, indicating affinity maturation

  • Epitope Immunodominance Patterns:

    • RBD epitopes generally elicit the strongest antibody responses

    • Epitope immunodominance shifts over time as the immune response matures

    • Some subdominant epitopes may produce antibodies with greater cross-variant neutralization potential

  • Correlation with Protection:

    • While antibodies against S1 develop early, their correlation with protection varies:

      • High titers of RBD-binding and neutralizing antibodies correlate most strongly with protection

      • Non-neutralizing antibodies may contribute to protection through Fc-mediated functions

      • Memory B cell responses provide long-term capacity for antibody production upon reexposure

For comprehensive assessment of antibody responses, researchers should employ multiple assays targeting different S1 domains and consider both binding and functional antibody properties over time .

What are the key implications of SARS-CoV-2 S1 protein research for understanding long COVID and neurological sequelae?

Research on SARS-CoV-2 S1 protein provides critical insights into the mechanistic basis of long COVID and neurological sequelae. The evidence points to several important implications:

  • Direct Neurotoxic Effects: Animal studies demonstrate that S1 protein alone, without active viral replication, can induce significant neuronal death (35% reduction in hippocampal neurons) and glial activation, suggesting a direct mechanism for cognitive symptoms reported by COVID-19 patients .

  • Cognitive Impairment Mechanisms: The experimental findings showing S1-induced deficits in novel object recognition and location tests in mice align with the "brain fog" and memory issues reported in long COVID. These effects appear to be mediated through hippocampal damage and neuroinflammation .

  • Anxiety and Mood Disorders: S1 protein administration induced anxiety-like behaviors in animal models, which parallels the increased prevalence of anxiety and mood disorders observed in post-COVID patients .

  • Blood-Brain Barrier Considerations: The ability of S1 protein to potentially cross the blood-brain barrier or affect its integrity suggests one pathway by which peripheral infection could lead to central nervous system effects, even without direct viral neuroinvasion.

  • Persistent Antigen Hypothesis: Detection of persistent S1 antigens in some long COVID patients supports the hypothesis that continued presence of viral proteins, rather than active viral replication, may drive chronic symptoms through ongoing immune activation and tissue effects.

  • Therapeutic Implications: These findings suggest potential therapeutic approaches for long COVID, including:

    • Strategies to eliminate persistent S1 protein

    • Anti-inflammatory interventions targeting specific neuroinflammatory pathways

    • Neuroprotective therapies to prevent or reverse S1-induced neuronal damage

    • Cognitive rehabilitation focused on hippocampal-dependent functions

  • Biomarker Development: Understanding the relationship between S1 protein persistence and cognitive symptoms could lead to biomarkers for diagnosing and monitoring neurological aspects of long COVID .

These findings collectively indicate that the S1 protein is not merely a structural component of the virus but a potentially active mediator of tissue damage and symptomatology in both acute COVID-19 and its chronic sequelae.

How might research on SARS-CoV-2 S1 (1-681) inform the development of next-generation vaccines and therapeutics?

Research on SARS-CoV-2 S1 (1-681) offers numerous insights that can directly inform the development of improved vaccines and therapeutics:

  • Vaccine Design Refinements:

    • Structure-guided antigen engineering: Understanding critical epitopes within S1 (1-681) enables the design of stabilized antigens that better present neutralizing epitopes while minimizing exposure of non-neutralizing sites

    • Mutation-resistant targets: Identifying conserved regions within S1 that show minimal variation across variants can guide development of broadly protective vaccines

    • Position 681 considerations: The significance of mutations at position 681 for furin cleavage and transmissibility suggests that next-generation vaccines might benefit from including both pre-fusion stabilized forms and strategically modified cleavage sites

  • Therapeutic Antibody Development:

    • Epitope mapping: Detailed knowledge of antigenic sites within S1 (1-681) allows selection of antibody combinations targeting non-overlapping epitopes to minimize escape

    • Cross-reactivity potential: Understanding the minimal cross-reactivity between SARS-CoV-2 S1 and other coronaviruses helps in developing highly specific therapeutic antibodies

    • Functional regions: Targeting functional domains rather than just binding epitopes may improve therapeutic efficacy

  • Anti-inflammatory Approaches:

    • Research showing S1-induced neuroinflammation suggests therapeutic targets for preventing or treating neurological sequelae of COVID-19

    • Glial cell activation pathways triggered by S1 represent potential intervention points for reducing neuroinflammation

  • Diagnostic Advancements:

    • The high specificity of S1-based serological assays (no cross-reactivity with other coronaviruses) supports their use in diagnostic applications

    • Combined S1 and N-based testing approaches offer comprehensive immune status assessment

  • Drug Development Targets:

    • Furin inhibitors: Understanding the role of position 681 in furin cleavage suggests therapeutic potential for selective protease inhibitors

    • Receptor interaction blockers: Detailed knowledge of S1-ACE2 binding interfaces informs small molecule drug design

  • Delivery Systems:

    • Insights into S1 protein structure and post-translational modifications guide the development of effective mRNA and protein delivery platforms

    • Understanding potential neurotoxic effects of S1 protein suggests the need for approaches that limit systemic distribution of vaccine antigens

By leveraging these research insights, next-generation interventions can be designed with enhanced efficacy against emerging variants while potentially addressing both acute infection and long-term sequelae of COVID-19 .

What are the most promising methodologies for studying the long-term effects of SARS-CoV-2 S1 protein exposure?

To comprehensively investigate the long-term effects of SARS-CoV-2 S1 protein exposure, researchers should implement multi-faceted methodological approaches that span various time scales and biological systems:

  • Longitudinal Clinical Studies:

    • Prospective cohort studies with pre-infection baseline cognitive assessments, as demonstrated in the human challenge model research

    • Detailed neuropsychological testing batteries assessing multiple cognitive domains

    • Biomarker monitoring including inflammatory markers, autoantibodies, and circulating S1 protein

    • Neuroimaging protocols with MRI, fMRI, and PET to detect structural and functional changes

  • Advanced Animal Models:

    • Chronic exposure paradigms with controlled S1 administration over extended periods

    • Age-specific models to assess differential vulnerability across lifespan

    • Transgenic approaches incorporating human ACE2 or other relevant receptors

    • In vivo imaging for real-time monitoring of neuroinflammation and neurocircuit function

  • Three-Dimensional Tissue Models:

    • Brain organoids for long-term exposure studies (weeks to months)

    • Organ-on-chip systems connecting multiple tissue types (e.g., lung-blood-brain barrier-brain)

    • Patient-derived cellular systems to capture individual susceptibility factors

  • Multi-omics Approaches:

    • Spatial transcriptomics to map regional gene expression changes

    • Proteomics and phosphoproteomics to identify altered signaling pathways

    • Metabolomics to detect shifts in cellular metabolism and energetics

    • Single-cell analyses to dissect cell type-specific responses

  • Functional Assessment Techniques:

    • Electrophysiological recordings to detect subtle changes in neuronal function

    • Calcium imaging to monitor neuronal activity patterns

    • Behavioral assessment batteries sensitive to hippocampal and prefrontal cortex function

    • Cognitive testing paradigms that parallel human assessments for translational relevance

  • Statistical and Computational Approaches:

    • Machine learning algorithms to identify patterns in complex multi-omics datasets

    • Network analyses to understand system-level changes

    • Predictive modeling to estimate long-term outcomes based on early biomarkers

By integrating these complementary approaches, researchers can comprehensively characterize the temporal dynamics, dose-dependency, and mechanisms underlying potential long-term effects of S1 protein exposure, particularly focusing on the cognitive and neurological manifestations observed in post-COVID syndromes .

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 divided into two subunits: S1 and S2. The S1 subunit is particularly important as it contains the receptor-binding domain (RBD) that binds to the host cell receptor, angiotensin-converting enzyme 2 (ACE2).

Structure and Function

The spike glycoprotein S1 subunit (1-681) is a recombinant protein that includes the first 681 amino acids of the spike protein. This region is responsible for the initial attachment of the virus to the host cell. The S1 subunit contains the receptor-binding domain (RBD), which directly interacts with the ACE2 receptor on the surface of human cells .

The binding of the S1 subunit to the ACE2 receptor is a critical step in the viral entry process. Once the S1 subunit binds to ACE2, it triggers a series of conformational changes in the spike protein, leading to the fusion of the viral and cellular membranes. This fusion allows the viral RNA to enter the host cell, initiating the infection process .

Recombinant Production

Recombinant production of the spike glycoprotein S1 subunit involves expressing the protein in a host cell system, such as HEK293 cells. The recombinant protein is typically tagged with a polyhistidine tag at the C-terminus to facilitate purification. The protein is then purified using techniques such as affinity chromatography .

The recombinant spike glycoprotein S1 subunit is used in various research applications, including vaccine development, diagnostic assays, and therapeutic studies. Its ability to bind to the ACE2 receptor makes it a valuable tool for studying the mechanisms of viral entry and for developing interventions to block this process .

Applications and Research

The recombinant spike glycoprotein S1 subunit has been extensively used in research to understand the pathogenesis of SARS-CoV-2. Studies have shown that the S1 subunit can activate various cellular pathways, leading to changes in cellular function and survival. For example, the S1 subunit has been found to increase the activity of connexin 43 (Cx43) hemichannels, which can affect intracellular calcium dynamics and ATP release .

Additionally, the S1 subunit has been used in the development of vaccines and therapeutic agents. By targeting the RBD of the S1 subunit, researchers aim to block the interaction between the virus and the ACE2 receptor, thereby preventing viral entry and infection .

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.