CoV-2 Spike (318-542)

Coronavirus 2019 Spike Receptor Binding Domain (318-542 a.a.) Recombinant
Shipped with Ice Packs
In Stock

Description

ACE2 Binding Mechanism

The RBD forms critical interactions with ACE2 through:

  • Residue Lys417: Forms ionic bonds with ACE2 Asp30

  • Residues Gly446/Tyr449: Stabilize hydrophobic interactions

  • Flexible Loop (438–506): Facilitates conformational changes for receptor engagement

Binding kinetics demonstrate high affinity:

ParameterValueMethod
K<sub>D</sub>3.60 nMBiacore SPR
K<sub>on</sub>1.12 × 10<sup>5</sup> M<sup>-1</sup>s<sup>-1</sup>BLI
K<sub>off</sub>4.03 × 10<sup>-4</sup> s<sup>-1</sup>BLI

Neutralizing Antibody Epitopes

Four distinct epitope groups target this region:

  1. Group I: Overlaps with CR3022 epitope (residues 369–386), non-ACE2 competing

  2. Group II: Blocks ACE2 binding via steric hindrance (e.g., MD62, NT<sub>50</sub> = 0.4 μg/mL)

  3. Group III/IV: Binds distal RBD regions, synergizes with Group II antibodies

Cross-Reactivity

  • 75% sequence identity with SARS-CoV-1 RBD

  • Minimal cross-neutralization with MERS-CoV (<5% NT<sub>50</sub>)

Endothelial Dysregulation

The RBD induces endothelial-mesenchymal transition (EndMT) via:

  • TLR4-ROS-TGF-β2 axis: Upregulates MRTF-A/B nuclear translocation

  • ACE2 downregulation: Stimulates TGF-β1 secretion

Neurological Effects

  • Activates microglia via TLR4, releasing IL-1β and CXCL8

  • Disrupts blood-brain barrier integrity at concentrations ≥100 nM

Therapeutic and Diagnostic Applications

AntibodyEpitope GroupNT<sub>50</sub> (μg/mL)ACE2 Blockade
MD65II0.4Complete
MD29I13.0None
MD47IV8.2Partial

Stability and Formulation

  • Lyophilized Stability: >24 months at -80°C (carrier-free)

  • Reconstitution: 500 μg/mL in PBS yields >95% monomeric form

  • Aggregation Threshold: <5% at concentrations ≤1 mg/mL

Product Specs

Introduction

First identified in Wuhan, China, in December 2019, the 2019 novel coronavirus (2019-nCoV) is a human coronavirus responsible for causing viral pneumonia. Genetic analysis reveals that 2019-nCoV shares 87% of its identity with the bat-derived severe acute respiratory syndrome coronavirus (SARS-CoV-2) discovered in Zhoushan, eastern China, in 2018. Despite some differences, the receptor-binding domain (RBD) structure of 2019-nCoV is similar to that of 2018 SARS-CoV, suggesting that 2019-nCoV may also bind to the human angiotensin-converting enzyme 2 (ACE2) receptor. While bats are considered the likely natural reservoir of 2019-nCoV, an intermediate animal host, potentially from the seafood market in Wuhan, is suspected. Research suggests that 2019-nCoV may be a recombinant virus, with its spike glycoprotein exhibiting elements from both bat coronaviruses and an unknown coronavirus.

Description

This product consists of the recombinant Coronavirus 2019 Spike Receptor Binding Domain (amino acids 318-542), with a molecular weight of 25.7 kDa. Expressed in E. coli, this protein features a C-terminal 6xHis tag and undergoes purification via a proprietary chromatographic method.

Physical Appearance

Sterile Filtered clear solution.

Formulation

The CoV-2 Spike protein solution is formulated in PBS (phosphate-buffered saline) with 25mM K2CO3 (potassium carbonate).

Stability

For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. To further enhance long-term stability, the addition of a carrier protein (0.1% HSA or BSA) is advisable. Minimize freeze-thaw cycles to maintain product integrity.

Purity

The purity of the protein is determined to be greater than 95% based on 10% SDS-PAGE analysis with Coomassie blue staining.

Applications

This product is suitable for various applications, including ELISA (enzyme-linked immunosorbent assay), Western blot analysis, and lateral flow immunoassays.

Source

Escherichia Coli.

Amino Acid Sequence

HMRVQPTESI VRFPNITNLC PFGEVFNATR FASVYAWNRK RISNCVADYS VLYNSASFS TFKCYGVSPT KLNDLCFTNV YADSFVIRGD EVRQIAPGQT GKIADYNYKL PDDFTGCVI AWNSNNLDSKV GGNYNYLYRL FRKSNLKPFE RDISTEIYQA GSTPCNGVEG FNCYFPLQSY GFQPTNGVGY QPYRVVVLSF ELLHAPATVC GPKKSTNLVK NKCVNFNLE 

Q&A

What is the structural significance of the SARS-CoV-2 spike protein region 318-542?

The region 318-542 of the SARS-CoV-2 spike protein contains critical elements of the receptor-binding domain (RBD), which is essential for viral attachment to the human ACE2 receptor. This region undergoes significant conformational changes during the viral infection process. Specifically, the spike proteins must bind to ACE2 receptors and then dramatically change shape, folding in on themselves to enable the virus to fuse its membrane with host cell membranes . Structural biology studies using cryo-electron microscopy (cryo-EM) have revealed that mutations within this region can stabilize the spike protein, potentially increasing infectivity by ensuring more functional spikes are available to bind to ACE2 receptors . Researchers investigating this region should consider both its static structure and dynamic conformational changes when designing experiments.

What methods are available for expressing and purifying the 318-542 region for laboratory studies?

For laboratory studies requiring the isolated 318-542 region, several expression systems can be employed. Bacterial expression systems (particularly E. coli) can be used for high-yield production, though proper folding may be a challenge due to the complexity of this region. Mammalian expression systems (HEK293 or CHO cells) often provide better folding and post-translational modifications but at lower yields. For purification, researchers typically use a combination of affinity chromatography (utilizing His-tags or other fusion tags), followed by size exclusion chromatography to ensure homogeneity. When designing constructs, researchers should consider including stabilizing mutations that maintain the prefusion conformation, as demonstrated in recent studies where simulation-driven approaches identified tryptophan substitutions that impart kinetic and thermodynamic stabilization . Quality control should include functional binding assays to ACE2 and structural verification via circular dichroism or, ideally, cryo-EM to confirm proper folding.

How do mutations within the 318-542 region affect antibody neutralization and escape?

Mutations within the 318-542 region can significantly impact antibody neutralization efficacy due to this region's role in the receptor-binding domain. Research using phage display libraries constructed from COVID-19 patients has identified multiple neutralizing monoclonal antibodies that specifically target distinct epitopes on the viral spike RBD . These antibodies exhibit varying mechanisms of action - some directly abrogate RBD binding to human ACE2 receptors, while others may interfere with conformational changes necessary for membrane fusion . To study neutralization escape, researchers should employ competitive binding assays with panels of monoclonal antibodies targeting different epitopes within the region. Deep mutational scanning approaches can systematically identify escape mutations by creating libraries of single amino acid substitutions and selecting for variants that maintain ACE2 binding but escape neutralization. Additionally, structural studies combining cryo-EM and X-ray crystallography can precisely map antibody binding sites and inform rational immunogen design strategies that focus immune responses on conserved epitopes within this region.

What experimental approaches can be used to study the pathological effects of the spike protein region 318-542?

The pathological effects of the spike protein region 318-542 can be studied through multiple experimental approaches. In vitro fibrinogen clotting assays have demonstrated that spike protein induces anomalous clotting in both purified fluorescent fibrinogen and in platelet-poor plasma (PPP) from healthy individuals . These clots demonstrate amyloid-like properties when stained with Thioflavin T . Scanning electron microscopy and fluorescence microscopy can reveal large, dense anomalous and amyloid masses in whole blood and PPP samples exposed to spike protein . Mass spectrometry techniques have confirmed that spike protein causes structural changes to fibrin(ogen), complement 3, and prothrombin, making these proteins less resistant to trypsinization . For researchers investigating this specific region (318-542), comparing its effects to those of the full spike or other regions can help determine whether the observed pathologies are region-specific. Flow analysis methods can be employed to assess how microclots formed in the presence of this region may impair blood flow . Additionally, proteomics approaches can identify broader interactions with human plasma proteins to understand the full scope of potential pathological effects.

How can computational approaches enhance the design of stabilized versions of the 318-542 region for vaccine development?

Computational approaches have proven invaluable for designing stabilized versions of spike protein regions for vaccine development. Molecular simulations provide mechanistic characterization of the spike trimer's opening, informing the strategic placement of stabilizing mutations . Specifically, simulation-driven approaches have successfully identified tryptophan substitutions that impart kinetic and thermodynamic stabilization to maintain the prefusion conformation . These computational predictions should be validated through experimental characterization via cryo-EM to confirm the molecular basis of stabilization . Researchers can employ molecular dynamics simulations, structure-based energy calculations, and machine learning approaches trained on existing structural data to predict stabilizing mutations. Integration of experimental feedback into iterative computational design cycles has proven particularly effective, leading to engineered immunogens with increased protein expression, superior thermostability, and preserved immunogenicity . For the 318-542 region specifically, computational approaches should focus on maintaining proper folding of the RBD while enhancing stability, as structural integrity is critical for eliciting neutralizing antibodies that recognize the native conformation.

What are the optimal detection methods for quantifying the 318-542 region in biological samples, and how can detection sensitivity be improved?

Detecting and quantifying the SARS-CoV-2 spike protein region 318-542 in biological samples requires highly sensitive methods. Ultrasensitive immunoassays like the SPEAR SARS-CoV-2 Spike Protein assay can detect sub-femtomolar concentrations of the S1 spike protein subunit from as little as 1 μL of diluted sample . For researchers developing detection methods, key considerations include antibody selection (monoclonal antibodies targeting conserved epitopes within the 318-542 region), sample preparation techniques to overcome interference from anti-spike antibodies in patient samples, and signal amplification strategies. Proprietary antibody-reducing sample treatments can allow detection of total spike protein, even in the presence of anti-spike antibodies, enabling accurate and consistent measurements . Additional approaches include digital ELISA platforms, which offer exceptional sensitivity through single-molecule detection, and mass spectrometry-based methods that can provide both quantification and structural characterization. For improved sensitivity, researchers should consider multiplexed detection systems targeting multiple epitopes within the 318-542 region and implement rigorous validation using both recombinant standards and clinical samples with known viral loads.

How does the furin cleavage site proximal to the 318-542 region influence spike protein function and viral pathogenicity?

While the furin cleavage site itself is not within the 318-542 region, its proximity and functional relationship make it relevant to researchers studying this region. Mutations near the furin cleavage site, such as P681R, add a basic amino acid that enhances the fusion activity of the SARS-CoV-2 spike protein, likely due to increased cleavage by cellular furin protease . This type of mutation has been observed in variants of concern, including a similar change (P681H) in the B.1.1.7 lineage . To study the interplay between the 318-542 region and the furin cleavage site, researchers can employ cell-cell fusion assays to quantify fusion efficiency with various mutations. Pseudovirus entry assays using constructs with mutations in both regions can assess their combined effects on viral entry. Biochemical approaches including in vitro protease susceptibility assays can determine how the conformation of the 318-542 region influences accessibility of the furin site to proteases. Structural studies combining hydrogen-deuterium exchange mass spectrometry with cryo-EM can map conformational changes that propagate between these regions. Understanding this functional relationship is critical for comprehending how distant mutations may have synergistic effects on viral fitness and pathogenicity.

What controls and validation steps are necessary when studying interactions between the 318-542 region and human proteins?

When studying interactions between the SARS-CoV-2 spike protein region 318-542 and human proteins, rigorous controls and validation steps are essential. Primary controls should include: (1) A negative control using a structurally similar but functionally distinct protein domain to confirm specificity; (2) Denatured 318-542 protein to distinguish between conformation-dependent and -independent interactions; (3) Competitive inhibition controls with soluble ACE2 or known neutralizing antibodies; and (4) Dose-response assessments to establish interaction kinetics. Validation should employ orthogonal techniques - if initial screening uses biolayer interferometry (BLI) with K​D values ranging from 0.4-5.8 nM as observed with various antibodies , confirmation should follow with surface plasmon resonance, microscale thermophoresis, or isothermal titration calorimetry. For cellular interactions, both binding (flow cytometry, immunofluorescence) and functional assays (cell fusion, pseudovirus entry) should be performed. Mass spectrometry can verify direct protein-protein interactions and identify binding interfaces . Researchers should also validate that observed interactions occur under physiologically relevant conditions by varying pH, ionic strength, and temperature to mimic different cellular compartments where viral-host protein interactions might occur.

What evidence exists for the persistence of the 318-542 fragment in Long COVID/PASC patients, and how might it contribute to pathology?

Evidence regarding the persistence of SARS-CoV-2 spike protein fragments, including the 318-542 region, in Long COVID/PASC patients is emerging as an area of significant research interest. Ultrasensitive detection methods capable of measuring sub-femtomolar concentrations of spike protein are now being employed to investigate this phenomenon . Studies have documented significant hypercoagulation in patients suffering from Long COVID/PASC, similar to coagulation abnormalities seen in acute infection . The spike protein has been shown to interact with plasma proteins, causing fibrin(ogen), prothrombin, and other coagulation factors to become resistant to normal breakdown processes, potentially contributing to the formation of microclots . These microclots may impair blood flow and could explain some persistent symptoms . For researchers investigating this hypothesis, longitudinal sampling with controls for antibody interference is crucial, as anti-spike antibodies can mask detection of spike protein fragments . Comparison of fragment levels between patients with different symptom profiles may help establish clinically relevant thresholds and determine if specific regions like 318-542 correlate with particular manifestations of Long COVID. Mechanistic studies in appropriate cell and animal models are needed to confirm whether the observed correlations represent causal relationships.

How can the 318-542 region be optimized for next-generation vaccine design to enhance protection against emerging variants?

Optimizing the 318-542 region for next-generation vaccine design requires strategies that address both increased immunogenicity and broader protection against emerging variants. Structure-based vaccine design approaches should focus on stabilizing this region in its prefusion conformation, as studies have shown that the original spike protein could dissociate, reducing its ability to induce strong neutralizing antibody responses . Simulation-driven design approaches have successfully identified specific substitutions, such as tryptophan mutations, that provide kinetic and thermodynamic stabilization to spike protein constructs . These engineered immunogens have demonstrated increased protein expression, superior thermostability, and preserved immunogenicity against sarbecoviruses . For broader protection, researchers should consider multivalent display approaches that present the 318-542 region from multiple variants simultaneously. Computational epitope mapping can identify conserved regions within 318-542 that are targeted by broadly neutralizing antibodies, allowing for focused immune responses on these sites. Iterative design cycles incorporating structural characterization via cryo-EM should verify that modifications maintain the desired conformation . Prime-boost strategies with heterologous 318-542 constructs may expand the breadth of neutralizing antibody responses. Finally, adjuvant optimization specific to this protein region could enhance both the magnitude and quality of immune responses, particularly focusing on durability and mucosal immunity.

How can researchers distinguish between specific effects of the 318-542 region on coagulation versus general inflammatory responses?

Distinguishing between the specific effects of the SARS-CoV-2 spike protein region 318-542 on coagulation versus general inflammatory responses requires carefully controlled experimental designs. Researchers should employ purified systems with defined components to isolate direct effects on coagulation factors. Studies have demonstrated that spike protein directly induces anomalous clotting in purified fluorescent fibrinogen, suggesting a specific mechanism independent of inflammatory mediators . Mass spectrometry analysis has confirmed that spike protein causes structural changes to fibrinogen, complement 3, and prothrombin, making these proteins resistant to trypsinization . To differentiate these direct effects from inflammation-mediated coagulation, researchers should conduct parallel experiments with and without immune cells or inflammatory mediators. Time-course studies can help establish the sequence of events - whether coagulation abnormalities precede inflammatory responses or vice versa. Dose-response experiments with isolated 318-542 region versus other spike domains can determine region-specific effects. Mechanistic dissection using specific inhibitors of coagulation pathways versus inflammatory signaling cascades can further delineate these processes. Additionally, comparative studies between the effects of 318-542 and known inflammatory triggers (like LPS) or direct coagulation activators (like thrombin) can provide benchmarks for characterizing the observed phenomena. Multiparameter analysis combining coagulation and inflammatory readouts with principal component analysis can help identify distinct versus overlapping pathways.

Product Science Overview

Introduction

The Coronavirus 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to extensive research into its structure and mechanisms of infection. One of the critical components of SARS-CoV-2 is the spike (S) protein, which facilitates the virus’s entry into host cells. The spike protein contains a specific region known as the Receptor Binding Domain (RBD), which is crucial for binding to the host cell receptor, angiotensin-converting enzyme 2 (ACE2).

Structure and Function

The RBD of the SARS-CoV-2 spike protein spans amino acids 318 to 542. This domain is responsible for the initial interaction with the ACE2 receptor on the surface of host cells. The binding of the RBD to ACE2 is a key step in the viral entry process, allowing the virus to fuse with the host cell membrane and release its genetic material into the cell .

Recombinant RBD

Recombinant RBD refers to the RBD produced through recombinant DNA technology. This involves inserting the gene encoding the RBD into an expression system, such as bacteria, yeast, or mammalian cells, to produce the protein in large quantities. Recombinant RBD is used in various applications, including vaccine development, diagnostic assays, and therapeutic research.

Applications in Vaccine Development

The RBD is a prime target for vaccine development due to its critical role in viral entry. Vaccines that elicit an immune response against the RBD can potentially block the virus from binding to ACE2, thereby preventing infection. Several COVID-19 vaccines, including mRNA vaccines, have been designed to induce antibodies against the RBD .

Diagnostic and Therapeutic Uses

Recombinant RBD is also used in diagnostic assays to detect antibodies against SARS-CoV-2 in patient samples. These assays help determine whether an individual has been exposed to the virus and has developed an immune response. Additionally, recombinant RBD is being explored as a therapeutic agent. For example, it can be used to develop monoclonal antibodies that specifically target the RBD, neutralizing the virus and preventing it from infecting cells .

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.