CoV-2 Membrane Env.

Coronavirus 2019 Membrane Envelope, Recombinant
Shipped with Ice Packs
In Stock

Description

Membrane Protein (M)

  • Scaffolding: M dimers organize viral components via electrostatic interactions with the nucleocapsid (N) protein and RNA .

  • Membrane curvature: Oligomerization induces membrane bending, critical for spherical virion morphology .

  • Spike (S) protein regulation: Retains S proteins in the ERGIC, reducing premature cell-cell fusion .

Envelope Protein (E)

  • Viroporin activity: Promotes virion release and immune evasion .

  • Syncytia suppression: Coexpression with M reduces S-mediated cell fusion by 60% .

Host Membrane Interaction Mechanisms

The membrane-binding peptide (MBP) in the S2 subunit facilitates viral entry:

  • Cholesterol dependence: Binding affinity to cholesterol-enriched membranes is 5× higher than sphingomyelin membranes .

  • Disulfide bridge stability: A conserved Cys-Cys bond in MBP enhances mechanical stability during membrane attachment .

Table 2: Key Interactions and Mutational Impacts

Interaction/MutationFunctional ConsequenceSource
M dimerizationScaffolds viral assembly
M-N protein bindingMediates RNA packaging
MBP-Chol interactionEnables membrane fusion (ΔG = -8.2 kcal/mol)
Hinge region deletionAbolishes virion formation

Therapeutic Implications

  • M protein: Immunogenic epitopes on its intravirion domain make it a vaccine candidate .

  • E protein: Ion channel inhibitors (e.g., hexamethylene amiloride) reduce viral titers by 90% .

  • MBP targeting: Cholesterol depletion or MBP-disrupting mutations (e.g., R846A) inhibit infection .

Outstanding Research Questions

  1. How do M conformations transition during assembly?

  2. Can M’s electropositive surface be exploited for RNA-interference therapies?

  3. What is the stoichiometry of M-E-S complexes in native virions?

Product Specs

Introduction

In December 2019, a novel coronavirus, designated 2019-nCoV, emerged in Wuhan, China, causing viral pneumonia in humans. This virus was linked to a seafood market in the city.

Genetic analysis revealed that 2019-nCoV shares 87% sequence identity with the bat-derived SARS-CoV-2 identified in Zhoushan, eastern China, in 2018. Notably, both viruses possess similar receptor-binding domains (RBDs) despite some amino acid variations. This suggests that 2019-nCoV might utilize the human ACE2 receptor (angiotensin-converting enzyme 2) for cell entry, similar to SARS-CoV.

While bats are considered the likely reservoir of 2019-nCoV, researchers hypothesize that an intermediate animal host, potentially from the seafood market, played a role in its transmission to humans. Recombination analysis indicates that the virus's spike glycoprotein might be a product of recombination between a bat coronavirus and an unidentified coronavirus.

Description

This recombinant fusion protein is produced in E. coli and consists of the full-length Membrane (M) and Envelope (E) proteins of the Wuhan-Hu-1 strain of Coronavirus 2019. The protein has a molecular weight of 34.2 kDa and includes a C-terminal His tag for purification purposes.

Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation

This CoV-2 Membrane Envelope protein solution is supplied in a buffer solution of 1x PBS (phosphate-buffered saline).

Stability

The CoV-2 Membrane Envelope fusion protein is shipped to you frozen on ice packs to maintain its stability. Upon receiving the protein, it's crucial to store it at -20°C for long-term preservation.

Purity

The purity of this protein is greater than 90%, as determined by SDS-PAGE analysis.

Source

E.Coli

Q&A

What is the structural composition of the SARS-CoV-2 E protein?

The SARS-CoV-2 E protein is a 75-residue integral membrane protein composed of a long transmembrane helix (residues 8-43) and a short cytoplasmic helix (residues 53-60) connected by a complex linker that exhibits some internal mobility. High-resolution solid-state NMR spectroscopy has revealed that in lipid bilayers mimicking the endoplasmic reticulum-Golgi intermediate compartment (ERGIC) membrane, the transmembrane domain forms a five-helix bundle surrounding a narrow pore. The protein deviates from ideal α-helical geometry due to three phenylalanine residues that stack within and between helices .

How does the oligomeric structure of E protein form in membranes?

The E protein transmembrane domain assembles into a homopentameric structure in lipid membranes. This pentameric assembly creates a cation-selective channel critical for viral function. The structural integrity is maintained through specific interhelical interactions, particularly the stacking of phenylalanine residues and interdigitation of valine and leucine residues, which create a relatively dehydrated pore compared to viroporins from influenza viruses and HIV. This highly organized structure appears to be sensitive to membrane environment conditions, suggesting potential conformational flexibility .

What advanced techniques are most effective for resolving the high-resolution structure of E protein in native-like environments?

The most effective approach for determining E protein structure at high resolution in membrane-like environments combines solid-state nuclear magnetic resonance (ssNMR) spectroscopy with properly designed membrane mimetics. Researchers have successfully used ssNMR to achieve a 2.1-Å resolution structure of the E protein transmembrane domain. This methodology is superior to solution NMR for membrane proteins as it allows proteins to be studied in lipid bilayers rather than detergent micelles, providing a more physiologically relevant structural context. The technique requires isotopic labeling of the protein (typically with 13C and 15N), reconstitution into lipid bilayers mimicking the ERGIC membrane composition, and collection of multiple correlation spectra to establish distance constraints and orientation parameters .

What specific functions does the E protein serve in the SARS-CoV-2 life cycle?

The E protein plays multiple critical roles in the SARS-CoV-2 life cycle. It facilitates viral assembly by participating in the formation of the virion structure alongside other structural proteins (S, M, and N). The E protein traffics through the endoplasmic reticulum to the ERGIC, where virion assembly occurs. Its ion channel activity is instrumental in modifying the cellular environment to facilitate viral replication and assembly. Additionally, the E protein contributes to virion release by potentially altering membrane curvature and facilitating membrane scission during the budding process, similar to the function of influenza virus M2 protein .

How does the E protein's ion channel activity contribute to viral replication?

The E protein forms a cation-selective channel across cellular membranes, particularly in the ERGIC. This channel activity is believed to modify ionic conditions within cellular compartments to create an environment conducive to viral replication and assembly. By altering ion concentrations and pH in these compartments, the E protein may facilitate conformational changes in viral and host proteins necessary for efficient virion assembly. The ion channel function may also contribute to membrane remodeling required for virion budding. Notably, mutations that abolish channel activity have been associated with reduced virus pathogenicity, indicating the critical nature of this function for viral fitness .

What experimental approaches can effectively measure the ion channel conductance properties of E protein in different membrane compositions?

To measure E protein ion channel conductance, researchers commonly employ several complementary techniques:

  • Planar lipid bilayer electrophysiology: This method involves reconstituting purified E protein into planar lipid bilayers of defined composition, followed by measuring ionic currents under various voltage conditions. This approach allows precise control of lipid composition, protein concentration, and ionic conditions.

  • Patch-clamp electrophysiology with reconstituted vesicles: E protein can be reconstituted into liposomes and then measured using patch-clamp techniques to record single-channel conductance and ion selectivity.

  • Fluorescence-based ion flux assays: Liposomes containing E protein can be loaded with ion-sensitive fluorophores to monitor ion flux in response to various gradients and inhibitors.

For maximum physiological relevance, these measurements should be conducted using lipid compositions that mimic the ERGIC membrane, where the E protein naturally functions. These experiments should systematically vary membrane thickness, charge, and fluidity to determine how membrane properties influence channel function .

How does the E protein contribute to SARS-CoV-2 pathogenesis?

The E protein contributes significantly to SARS-CoV-2 pathogenesis through multiple mechanisms. It disrupts the airway epithelial barrier by down-regulating the expression of tight junctional proteins, compromising respiratory epithelial integrity. The E protein also activates Toll-like receptors (TLR) 2 and 4 and downstream c-Jun N-terminal kinase (JNK) signaling pathways, which leads to increased intracellular chloride concentration through up-regulation of phosphodiesterase 4D (PDE4D) expression in airway epithelial cells. This elevated chloride concentration promotes the phosphorylation of serum/glucocorticoid regulated kinase 1 (SGK1), contributing to heightened airway inflammation. These pathogenic effects highlight the E protein's central role in the inflammatory response characteristic of severe COVID-19 .

What is the relationship between E protein and host inflammatory responses?

The E protein plays a crucial role in triggering and modulating host inflammatory responses. It activates the host inflammasome, leading to the production of pro-inflammatory cytokines. The protein's ion channel activity appears to be directly linked to inflammation, as channel activity inhibition reduces inflammatory responses. The E protein triggers TLR2/4 signaling cascades that culminate in heightened inflammatory cytokine production. Furthermore, by increasing intracellular chloride concentrations through PDE4D upregulation, the E protein promotes SGK1 phosphorylation, which further intensifies inflammatory processes. Experimental evidence suggests that blocking SGK1 or PDE4 can alleviate the inflammatory response induced by the E protein, pointing to potential therapeutic approaches .

What methodologies can be used to evaluate the impact of E protein on epithelial barrier function?

To evaluate the E protein's impact on epithelial barrier function, researchers can employ several complementary approaches:

  • Transepithelial electrical resistance (TEER) measurements: This method quantifies epithelial monolayer integrity by measuring electrical resistance across cell layers expressing E protein compared to controls.

  • Fluorescein isothiocyanate (FITC)-dextran permeability assays: These assess barrier permeability by measuring the passage of fluorescently labeled dextran molecules of various molecular weights across epithelial monolayers.

  • Immunofluorescence microscopy: This visualizes the localization and expression levels of tight junction proteins (e.g., claudins, occludin, ZO-1) in the presence of E protein.

  • Western blotting: This quantifies changes in tight junction protein expression levels.

  • Real-time PCR: This measures mRNA expression changes in genes encoding junction proteins.

These methods should be applied to relevant respiratory epithelial cell models (primary human bronchial epithelial cells or air-liquid interface cultures) expressing wild-type or mutant E protein to comprehensively characterize barrier dysfunction mechanisms .

What compounds have shown inhibitory effects against SARS-CoV-2 E protein?

Several compounds have demonstrated inhibitory effects against the SARS-CoV-2 E protein, with amiloride derivatives showing particular promise. Hexamethylene amiloride (HMA) and 5-(N-ethyl-N-isopropyl)amiloride (EIPA) exhibit the strongest binding affinity and antiviral potency. Dimethylamiloride (DMA) shows moderate activity, while the parent compound amiloride demonstrates significantly weaker binding. The efficacy correlates with the presence of bulky hydrophobic groups in the 5' position of the amiloride pyrazine ring, suggesting that these moieties play essential roles in binding to the E protein. Additionally, amantadine has been reported to inhibit the E protein's channel activity, though with less potency than HMA .

What is the mechanism of inhibitor binding to the E protein?

Inhibitors such as hexamethylene amiloride (HMA) bind primarily to the N-terminal region (residues 6-18) of the SARS-CoV-2 E protein. NMR studies have revealed that the polar amino-terminal lumen of the channel serves as the principal binding site for these compounds. The binding mechanism likely involves interactions between the inhibitor and specific residues lining the channel pore, particularly involving Asn15. This is evidenced by the N15A mutation, which abolishes HMA binding while significantly altering the protein conformation near the binding site. The structure-activity relationship among amiloride derivatives indicates that hydrophobic moieties at the 5' position of the pyrazine ring enhance binding affinity, suggesting hydrophobic interactions with the channel interior .

How can structure-based drug design be optimized for developing novel E protein inhibitors?

Structure-based drug design for novel E protein inhibitors can be optimized through a multi-faceted approach:

  • Refined binding site targeting: Focus on the N-terminal region (residues 6-18) identified as the principal binding site for current inhibitors, with particular attention to interactions with Asn15 and adjacent residues.

  • Pharmacophore modeling: Develop comprehensive pharmacophore models based on the structure-activity relationships of amiloride derivatives, emphasizing the importance of bulky hydrophobic groups at the 5' position.

  • Molecular dynamics simulations: Employ extensive MD simulations to capture the conformational dynamics of the E protein channel and identify transient binding pockets or allosteric sites.

  • Fragment-based screening: Utilize NMR-based fragment screening to identify novel chemical scaffolds with binding capacity to the E protein.

  • Rational modification strategy: Design compounds that combine optimal channel blocking properties with favorable pharmacokinetic profiles, potentially through hybrid molecules that incorporate features of effective ion channel blockers.

  • Validation pipeline: Establish a systematic validation pipeline employing biophysical binding assays (ITC, SPR), functional assays (electrophysiology), and cellular antiviral assays to comprehensively evaluate candidate inhibitors .

How does the SARS-CoV-2 E protein compare structurally to E proteins from other coronaviruses?

The SARS-CoV-2 E protein shares significant structural similarity with E proteins from other coronaviruses, particularly SARS-CoV-1, with which it shares approximately 94% sequence identity. Both form pentameric cation channels with similar transmembrane topology. When comparing the E protein across human coronaviruses, there are conserved structural elements, particularly in the transmembrane domain, though specific amino acid differences exist that may influence pathogenicity. Unlike the viroporins of influenza viruses (M2) and HIV-1 (Vpu), the SARS-CoV-2 E protein forms a more compact and rigid helical bundle with a narrower, less hydrated pore. This is attributed to its higher percentage of hydrophobic residues compared to the more polar residues found in influenza M2 channels .

What functional differences exist between the E proteins of SARS-CoV-2 and other coronaviruses?

While functionally similar in many respects, the E proteins from different coronaviruses exhibit notable differences that may influence pathogenicity and virus-host interactions. The SARS-CoV-2 E protein appears to form a cation channel that is structurally more rigid than those of other viruses. Unlike the influenza M2 protein, which undergoes rigid-body fast uniaxial rotation at high temperatures, the SARS-CoV-2 E protein remains relatively immobilized in membranes, suggesting extensive interaction with lipids. These structural differences likely translate to functional variations in ion selectivity, gating properties, and interactions with host factors. Additionally, the specific role in inflammation and epithelial barrier disruption may vary among coronavirus E proteins, potentially contributing to differences in disease presentation and severity .

What experimental designs can effectively compare the channel activities of E proteins from different coronaviruses?

To effectively compare channel activities of E proteins from different coronaviruses, researchers should implement a comprehensive experimental design that includes:

  • Parallel protein expression and purification: Express and purify E proteins from multiple coronaviruses (SARS-CoV-2, SARS-CoV-1, MERS-CoV, etc.) using identical expression systems and purification protocols to minimize methodology-induced variations.

  • Standardized reconstitution protocols: Reconstitute all E protein variants into liposomes of identical lipid composition mimicking the ERGIC membrane environment, maintaining consistent protein-to-lipid ratios.

  • Comparative electrophysiology: Conduct planar lipid bilayer or patch-clamp electrophysiology measurements under identical conditions (voltage, ion concentrations, pH) to directly compare:

    • Ion selectivity profiles

    • Single-channel conductance

    • Voltage dependence

    • Open probability

    • Gating kinetics

  • Inhibitor sensitivity profiling: Test a panel of channel inhibitors (amiloride derivatives, amantadine, etc.) against each E protein variant to develop detailed structure-activity relationships.

  • Mutagenesis studies: Create corresponding mutations in conserved positions across different coronavirus E proteins to evaluate the impact on channel function.

  • Computational modeling: Complement experimental data with molecular dynamics simulations to visualize ion permeation pathways and identify structural determinants of functional differences .

What key residues in the E protein are essential for its function?

Several key residues in the SARS-CoV-2 E protein have been identified as critical for its function. Asn15 plays a crucial role in maintaining the protein conformation near the inhibitor binding site, as the N15A mutation abolishes HMA binding and increases the production of virus-like particles. The phenylalanine residues (particularly Phe20, Phe23, and Phe26) are essential for maintaining the structural integrity of the transmembrane domain through their stacking interactions within and between helices. Additionally, the valine and leucine residues contribute to the interdigitation that stabilizes the pentameric structure. In SARS-CoV studies, mutations at Val25 (V25F) resulted in escape mutants with compensatory mutations at positions 19, 27, 30, and 37, suggesting these residues are also critical for maintaining channel function .

How do mutations in the E protein affect viral pathogenicity?

Mutations in the E protein can significantly impact viral pathogenicity, primarily through alterations in ion channel activity and protein-protein interactions. Studies with SARS-CoV have demonstrated that mutations abolishing E protein's ion channel activity lead to attenuated viruses with reduced pathogenicity. The V25F mutation in SARS-CoV E protein resulted in viruses with diminished fitness, though escape mutants with compensatory mutations (L19A, L27S, T30I, L37R) emerged that restored channel activity. These findings suggest that precise structural maintenance of the channel is essential for viral virulence. Additionally, mutations affecting the E protein's interaction with host factors likely impact inflammatory responses and epithelial barrier disruption, further modulating pathogenicity. The bipartite nature of the E protein, with its N-terminal and C-terminal halves potentially interacting semi-independently with viral and host proteins, suggests that mutations in different regions may have distinct effects on viral pathogenesis .

What advanced methodologies can be employed to systematically analyze the effects of E protein mutations?

To systematically analyze the effects of E protein mutations, researchers can employ several advanced methodologies:

  • Deep mutational scanning: This comprehensive approach involves creating a library of all possible single amino acid substitutions across the E protein sequence, followed by functional selection and next-generation sequencing to map mutational effects on viral fitness.

  • Cryo-EM structural analysis of mutants: Apply high-resolution cryo-electron microscopy to resolve structural changes induced by specific mutations in the context of the full virion or virus-like particles.

  • Multi-parameter patch-clamp analysis: Conduct detailed electrophysiological characterization of mutant E proteins to assess changes in conductance, ion selectivity, voltage dependence, and inhibitor sensitivity.

  • Protein-protein interaction mapping: Use proximity labeling approaches (BioID, APEX) combined with mass spectrometry to identify how mutations alter E protein's interactome.

  • In vivo pathogenesis models: Develop reverse genetics systems to introduce specific E protein mutations into infectious SARS-CoV-2 clones and evaluate their effects in appropriate animal models.

  • Real-time conformational dynamics: Apply single-molecule FRET or EPR spectroscopy to mutant E proteins to capture dynamic conformational changes under various conditions.

  • Combinatorial mutation analysis: Create and analyze double and triple mutants to understand compensatory effects and epistatic interactions between residues.

This integrated approach would provide comprehensive insights into structure-function relationships of the E protein and identify potential vulnerabilities for therapeutic targeting .

How can E protein research inform the development of attenuated coronavirus vaccines?

E protein research can significantly inform attenuated coronavirus vaccine development through several strategic approaches. Since E protein deletion or channel-inactivating mutations result in attenuated viruses with reduced pathogenicity, these modifications represent prime candidates for creating live attenuated vaccine platforms. Researchers can identify specific mutations that maintain immunogenicity while reducing virulence by systematically engineering modifications in channel-forming residues or protein-protein interaction domains. The detailed structural understanding of E protein allows for rational design of mutations that specifically target pathogenic functions while preserving antigenic properties. Additionally, combining E protein modifications with mutations in other viral proteins could create optimally balanced attenuation. Success with this approach is supported by previous studies showing that coronaviruses with E protein deletions or modifications can serve as attenuated vaccine candidates, providing protection against challenge with virulent strains while eliminating the risk of severe disease .

What role does the E protein play in the emergence of SARS-CoV-2 variants with enhanced transmissibility?

While spike protein mutations receive the most attention in SARS-CoV-2 variant analysis, the E protein may also contribute to enhanced viral transmissibility through several mechanisms. The E protein's ion channel activity and role in virion assembly and release directly impact viral production efficiency. Subtle mutations in the E protein could optimize these functions, potentially leading to increased viral load and enhanced transmission. The protein's ability to disrupt epithelial barriers may influence viral shedding patterns, affecting transmission dynamics. Additionally, E protein variants might modulate the inflammatory response in ways that alter symptom presentation or duration of viral shedding. Comprehensive analysis of E protein sequences across variants of concern, combined with functional characterization of any identified mutations, would provide valuable insights into whether E protein changes contribute to the transmissibility differences observed among SARS-CoV-2 variants .

What experimental systems can be developed to study E protein interactions with host factors in near-native conditions?

To study E protein interactions with host factors under near-native conditions, several advanced experimental systems can be developed:

  • SARS-CoV-2 E protein interactome mapping: Employ proximity-based labeling approaches (BioID, TurboID, APEX) in relevant respiratory epithelial cell models to capture transient and stable E protein interactions in physiological membrane contexts.

  • Organoid-based expression systems: Utilize human airway or lung organoids expressing tagged E protein to study interactions in complex, differentiated tissue architectures that better recapitulate the native environment.

  • Split-protein complementation assays: Develop fluorescence or luminescence-based complementation systems optimized for membrane proteins to visualize E protein-host protein interactions in living cells.

  • Cryo-electron tomography: Apply cellular cryo-ET to visualize E protein distribution and interactions within cellular compartments at near-molecular resolution.

  • Nanobody-based detection systems: Develop specific nanobodies against the E protein to enable super-resolution microscopy of E protein localization and trafficking without disrupting function.

  • Reconstituted membrane systems: Create synthetic membrane systems incorporating both purified E protein and candidate host interaction partners in native-like lipid compositions to study direct interactions and functional consequences.

These methodologies would provide unprecedented insights into how the E protein engages with the host cellular machinery during infection, potentially revealing new therapeutic targets .

Product Science Overview

Introduction

The Coronavirus 2019 Membrane Envelope, Recombinant, refers to the recombinant form of the membrane (M) and envelope (E) proteins of the SARS-CoV-2 virus, which is responsible for the COVID-19 pandemic. These proteins play crucial roles in the virus’s structure, assembly, and pathogenesis.

Structure and Function

The membrane (M) protein is the most abundant structural protein in the coronavirus. It spans the viral envelope and is responsible for maintaining the shape of the virus. The M protein interacts with other structural proteins, such as the spike (S) and nucleocapsid (N) proteins, to facilitate the assembly and budding of new virions .

The envelope (E) protein is a small, integral membrane protein involved in several aspects of the virus’s life cycle, including assembly, budding, envelope formation, and pathogenesis . It functions as an ion channel and interacts with both viral and host cell proteins to modulate the virus’s replication and release .

Recombinant Forms

Recombinant forms of the M and E proteins are produced using genetic engineering techniques. These recombinant proteins are used in various research applications, including the development of vaccines and diagnostic assays. By expressing these proteins in a controlled environment, researchers can study their structure, function, and interactions in detail .

Role in Vaccine Development

The M and E proteins are highly conserved among coronaviruses, making them potential targets for vaccine development. DNA vaccines expressing the M and E proteins have shown promise in preclinical studies. These vaccines can elicit cellular immune responses, which are crucial for long-term immunity against the virus . Although these vaccines may not induce robust humoral immunity, they can provide partial protection and are valuable in optimizing vaccination strategies .

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.