Recombinant Human coronavirus 229E Envelope small membrane protein (E)

Shipped with Ice Packs
In Stock

Description

Viral Assembly and Egress

  • Membrane Pore Formation: TMD forms ion channels, facilitating viral progeny release and modulating host membrane integrity .

  • Golgi Recruitment: CTD’s PDZ-binding motif interacts with host junction proteins (e.g., PALS1), redirecting them to the ER-Golgi intermediate compartment (ERGIC) for viral assembly .

Immune Modulation

  • Immune Evasion: Glycosylation at N-glycosylation sites (e.g., N265, N319) may shield epitopes, reducing neutralizing antibody binding .

  • Host Stress Response: Disruption of E protein function attenuates viral replication and pathogenesis .

Recombinant Production and Biochemical Properties

ParameterDetails
Expression SystemE. coli with N-terminal 10xHis-tag
Purity>95% (via SDS-PAGE and HPLC)
Storage-20°C (liquid), -80°C (lyophilized); avoid freeze-thaw cycles
ApplicationsStructural studies (cryo-EM, X-ray crystallography), antiviral drug testing

Key Findings:

  • Glycosylation Profiling: Native E protein exhibits N-linked glycans at specific sites (e.g., N265, N319), which are critical for receptor engagement and immune evasion .

  • Membrane Topology: Recombinant E protein retains the native Nt lum/Ct cyt topology in mammalian membranes, confirmed via protease protection assays .

Antiviral Drug Development

  • Target Validation: Inhibitors of E protein’s PDZ-binding motif or ion channel activity are under investigation for broad-spectrum antiviral therapies .

  • Vaccine Development: Recombinant E protein serves as a candidate antigen for subunit vaccines, though its role in immune protection remains under study .

Clinical Significance

  • Viral Fitness: Deletion of E protein in HCoV-229E severely impairs virion morphogenesis, suggesting its critical role in replication .

  • Therapeutic Targets: E protein’s ion channel activity and host protein interactions make it a candidate for antiviral intervention, particularly in combination therapies .

Comparative Analysis with Other Coronaviruses

FeatureHCoV-229E E ProteinSARS-CoV-2 E Protein
Sequence Identity94.7% with other human coronaviruses (e.g., HCoV-NL63) Lower homology to mild coronaviruses
PDZ-Binding MotifDLLV (residues 72–75)DLLV (conserved)
OligomerizationPentameric/dimeric states observed Homopentamer in micelles; dimer in lipid bilayers

Future Directions

  • Structural Dynamics: High-resolution studies of E protein’s conformational changes during viral assembly.

  • Therapeutic Strategies: Designing small-molecule inhibitors targeting E protein’s ion channel or PDZ-binding motif.

  • Cross-Species Transmission: Investigating E protein’s role in zoonotic spillover events.

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format currently in stock. However, if you have specific requirements for the format, please indicate them in your order. We will accommodate your request whenever possible.
Lead Time
Delivery time may vary depending on the purchasing method and location. Please contact your local distributor for specific delivery time information.
Note: Our proteins are shipped with standard blue ice packs by default. If you require dry ice shipping, please inform us in advance. Additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend centrifuging the vial briefly before opening to ensure the contents settle to the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers can use this as a reference.
Shelf Life
Shelf life is influenced by various factors, including storage conditions, buffer composition, temperature, and the inherent stability of the protein.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type will be determined during the manufacturing process.
The tag type will be determined during the production process. If you have a specific tag type preference, please inform us, and we will prioritize developing the specified tag.
Synonyms
E; sM; 5; Envelope small membrane protein; E protein; sM protein
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-77
Protein Length
full length protein
Species
Human coronavirus 229E (HCoV-229E)
Target Names
E
Target Protein Sequence
MFLKLVDDHALVVNVLLWCVVLIVILLVCITIIKLIKLCFTCHMFCNRTVYGPIKNVYHI YQSYMHIDPFPKRVIDF
Uniprot No.

Target Background

Function
Plays a central role in virus morphogenesis and assembly. Functions as a viroporin, self-assembling in host membranes to form pentameric protein-lipid pores that facilitate ion transport. Also implicated in the induction of apoptosis.
Gene References Into Functions
  1. These findings suggest that lipids play a functional role in E protein ion channel activity, forming a protein-lipid pore. This represents a novel concept for CoV E protein ion channel structure. PMID: 22832120
Database Links

KEGG: vg:918761

Protein Families
Alphacoronaviruses E protein family
Subcellular Location
Host Golgi apparatus membrane; Single-pass type III membrane protein.

Q&A

What is the primary structure of HCoV-229E envelope protein?

The HCoV-229E envelope protein is a small membrane protein consisting of 77 amino acids with the sequence: MFLKLVDDHALVVNVLLWCVVLIVILLVCITIIKLIKLCFTCHMFCNRTVYGPIKNVYHIYQSYMHIDPFPKRVIDF . It has a molecular weight of approximately 8.4-12 kDa and contains three distinct domains: a short hydrophilic N-terminal domain (NTD), a hydrophobic transmembrane domain (TMD) forming an amphipathic α-helix, and a hydrophilic C-terminal domain (CTD) that comprises most of the protein . The TMD enables oligomerization of E proteins to form ion-conductive pores across membranes, while the CTD contains a β-coil-β motif that functions as a Golgi-complex targeting signal .

How does the HCoV-229E E protein compare structurally to other coronavirus E proteins?

Coronavirus E proteins share a common architecture despite having variable sequence homology. Comparative analysis shows that HCoV-229E E protein shares structural similarities with other coronavirus E proteins, particularly within the transmembrane domain. Like other coronavirus E proteins, the HCoV-229E E protein forms pentameric α-helical bundles that are responsible for ion channel activity . Evidence from studies on SARS-CoV E protein indicates that these channels form pentameric left-handed parallel bundles with regular α-helices . The conserved structural features across coronavirus E proteins suggest common functional mechanisms, though specific amino acid variations may influence host interactions and pathogenicity.

What are the known functional roles of HCoV-229E E protein in the viral lifecycle?

The HCoV-229E E protein serves multiple critical functions:

  • Membrane permeabilization through ion channel activity

  • Virion assembly and morphogenesis

  • Involvement in viral tropism determination

  • Golgi complex localization via its C-terminal domain

Experimental evidence shows that absence or inactivation of E protein results in attenuated viruses, demonstrating its importance in viral pathogenesis . The E protein works in concert with other structural proteins (S, M, and N) to facilitate viral assembly and release .

What are the optimal conditions for recombinant expression of HCoV-229E E protein?

The most efficient expression system for producing functional recombinant HCoV-229E E protein is bacterial expression in E. coli with an N-terminal His-tag . For optimal expression:

  • Clone the full-length sequence (amino acids 1-77) into an appropriate expression vector with an N-terminal His-tag

  • Transform into an E. coli expression strain (BL21(DE3) or equivalent)

  • Induce expression with IPTG at lower temperatures (16-25°C) to reduce inclusion body formation

  • Include membrane-mimicking detergents (such as DPC) during purification to maintain protein solubility

The resulting protein can be stored as a lyophilized powder or in a Tris/PBS-based buffer with 6% trehalose at pH 8.0 . Repeated freeze-thaw cycles should be avoided, and addition of 5-50% glycerol is recommended for long-term storage at -20°C/-80°C .

What purification strategies yield the highest purity and functional activity for recombinant HCoV-229E E protein?

A multi-step purification strategy is required to obtain high-purity, functionally active HCoV-229E E protein:

  • Initial capture using immobilized metal affinity chromatography (IMAC) with Ni-NTA resin to leverage the His-tag

  • Secondary purification via hydrophobic interaction chromatography

  • Final polishing using size-exclusion chromatography to remove aggregates

This approach consistently yields protein with >90% purity as determined by SDS-PAGE . For functional studies, it's crucial to verify that the purified protein maintains its native conformation. This can be assessed through circular dichroism spectroscopy to confirm α-helical secondary structure content and ion channel activity assays to verify functional integrity.

How can researchers validate the correct folding and oligomerization of recombinant HCoV-229E E protein?

Validation of proper folding and oligomerization can be accomplished through multiple complementary approaches:

  • Biophysical characterization:

    • Circular dichroism (CD) spectroscopy to confirm α-helical content

    • Size-exclusion chromatography coupled with multi-angle light scattering (SEC-MALS) to determine oligomeric state

    • Analytical ultracentrifugation to verify pentameric assembly

  • Structural analysis:

    • Solution NMR in dodecylphosphatidylcholine micelles to assess secondary structure

    • Negative stain electron microscopy to visualize oligomeric assemblies

  • Functional validation:

    • Planar lipid bilayer electrophysiology to confirm ion channel activity

    • Fluorescence-based ion flux assays using reconstituted proteoliposomes

How can recombinant HCoV-229E E protein be used for ion channel inhibitor screening?

The ion channel activity of coronavirus E proteins makes them potential targets for antiviral therapeutics. A methodological approach for inhibitor screening includes:

  • Preparation of E protein channels:

    • Reconstitute purified E protein into lipid bilayers or liposomes

    • Confirm channel formation through electrophysiological measurements

  • Inhibitor screening protocol:

    • Primary screening using fluorescence-based ion flux assays with potential inhibitors

    • Secondary validation using electrophysiological recording of channel activity in the presence of hit compounds

    • Dose-response analysis to determine IC50 values

  • Validation with known inhibitors:

    • Use hexamethylene amiloride (HMA) as a positive control, which has been shown to inhibit coronavirus E protein ion channel activity

    • Compare with amiloride, which does not inhibit these channels, as a negative control

  • Correlation with antiviral activity:

    • Test promising inhibitors in viral replication assays to establish structure-activity relationships

    • Perform time-of-addition studies to confirm that inhibition occurs during viral assembly/release phases

What mutagenesis strategies are most effective for studying structure-function relationships in HCoV-229E E protein?

Systematic mutagenesis approaches can reveal critical functional residues in the E protein:

  • Domain-specific mutagenesis:

    • Target the transmembrane domain to identify residues critical for ion channel formation and oligomerization

    • Modify C-terminal domain residues to investigate Golgi-targeting and protein-protein interactions

    • Alter N-terminal residues to study membrane topology and insertion

  • Specific mutation types:

    • Alanine scanning mutagenesis of the transmembrane domain to identify pore-lining residues

    • Conservative vs. non-conservative substitutions at charged residues to assess ion selectivity

    • Cysteine substitutions combined with disulfide crosslinking to map protein-protein interfaces

  • Functional assessment of mutants:

    • Electrophysiological characterization to measure changes in ion conductance and selectivity

    • Subcellular localization studies to identify trafficking defects

    • Virus-like particle (VLP) assays to evaluate effects on assembly and release

How can cryo-EM be optimized for structural analysis of HCoV-229E E protein channels?

Cryo-electron microscopy (cryo-EM) offers advantages for structural studies of membrane proteins like the E protein. An optimized workflow includes:

  • Sample preparation:

    • Express and purify E protein to >95% homogeneity

    • Reconstitute in nanodiscs or amphipols to maintain native-like membrane environment

    • Optimize protein concentration (typically 2-5 mg/mL) and buffer conditions

  • Grid preparation:

    • Use Quantifoil R1.2/1.3 holey carbon grids with glow-discharge treatment (20 mA for 30s)

    • Apply 3 μL of sample and blot for 2.5 s at 4°C with 100% humidity before vitrification

    • Prepare multiple grids with varying protein concentrations and blotting times

  • Data collection parameters:

    • Collect on a 300-keV Titan Krios transmission electron microscope equipped with a direct detector

    • Use a defocus range between -1.5 and -2 μm and a magnification of 81,000x (pixel size ~1.08 Å)

    • Distribute a total dose of 50 e-/Å2 over 50 frames with exposure time of 2.5 s

  • Image processing considerations:

    • Perform motion correction and CTF estimation

    • Use focused refinement strategies to account for the small size of the pentameric channel

    • Consider symmetry-based approaches (C5 symmetry) to improve resolution

How does HCoV-229E E protein differ functionally from SARS-CoV-2 E protein?

While both proteins serve similar roles in their respective viruses, key functional differences exist:

FeatureHCoV-229E E proteinSARS-CoV-2 E protein
Length77 amino acids 75 amino acids
Channel activityForms pentameric ion channels Forms pentameric ion channels with higher cation selectivity
PathogenicityAssociated with mild respiratory disease Contributes to inflammatory pathology and cytokine storm
InhibitionSensitive to hexamethylene amiloride (HMA) Shows different inhibitor sensitivity profile
PDZ-binding motifLess well-characterizedContains a PDZ-binding motif that interacts with host proteins

These differences reflect the varied pathogenic potential of the viruses and highlight potential targets for selective therapeutic intervention.

What experimental approaches can detect interactions between HCoV-229E E protein and other viral proteins?

Several complementary methods can characterize protein-protein interactions:

  • Co-immunoprecipitation (Co-IP):

    • Express tagged versions of E protein and potential interaction partners

    • Perform pull-down assays followed by western blotting

    • Include appropriate controls to rule out non-specific binding

  • Proximity-based labeling:

    • Generate BioID or TurboID fusions with E protein

    • Express in relevant cell types during viral infection

    • Identify biotinylated proteins through streptavidin pull-down and mass spectrometry

  • Fluorescence resonance energy transfer (FRET):

    • Create fluorescent protein fusions with E protein and potential partners

    • Measure FRET efficiency in live cells to detect interactions

    • Perform acceptor photobleaching FRET for quantitative analysis

  • Bimolecular fluorescence complementation (BiFC):

    • Split fluorescent protein approach with fragments fused to E protein and potential partners

    • Monitor fluorescence reconstitution as indication of protein-protein proximity

    • Combine with subcellular markers to determine interaction localization

How do post-translational modifications affect HCoV-229E E protein function?

While less extensively characterized than other viral proteins, potential post-translational modifications (PTMs) of the E protein may regulate its function:

  • Palmitoylation:

    • Cysteine residues in the E protein may undergo palmitoylation

    • This modification can affect membrane association, protein stability, and channel function

    • Detection methods include metabolic labeling with palmitate analogs and mass spectrometry

  • Phosphorylation:

    • Serine, threonine, and tyrosine residues may be phosphorylated

    • This can regulate protein-protein interactions and subcellular localization

    • Site-directed mutagenesis of potential phosphorylation sites can reveal functional consequences

  • Ubiquitination:

    • May regulate E protein stability and turnover

    • Can be detected through immunoprecipitation followed by ubiquitin-specific western blotting

    • Proteasome inhibitors can be used to assess degradation pathways

What evidence supports targeting HCoV-229E E protein for antiviral development?

Several lines of evidence suggest the E protein is a viable therapeutic target:

  • E protein is essential for optimal viral replication, as demonstrated by attenuated viruses lacking functional E protein

  • The ion channel activity can be specifically inhibited by compounds like hexamethylene amiloride (HMA)

  • Targeting E protein may reduce inflammatory pathology associated with coronavirus infections

  • The E protein's high conservation within the alphacoronavirus genus suggests a low probability of escape mutations

Effective E protein inhibitors could potentially serve as broad-spectrum antivirals against multiple coronaviruses, given the structural similarities in E proteins across coronavirus species.

How can recombinant HCoV-229E E protein be utilized in virus-like particle (VLP) production for vaccine research?

VLPs represent a promising approach for coronavirus vaccine development, and recombinant E protein plays a critical role:

  • Optimal VLP composition:

    • Co-expression of S, M, E, and N proteins in mammalian cells produces coronavirus-like particles

    • E protein is essential for efficient VLP formation and release

    • The protein ratio should be optimized (typically lower E protein expression relative to other structural proteins)

  • Expression systems for VLP production:

    • Mammalian cell lines (HEK293T, Expi293) provide proper post-translational modifications

    • Baculovirus-insect cell systems offer higher yield for large-scale production

    • DNA or RNA transfection methods can be employed depending on the system

  • Purification and characterization of VLPs:

    • Differential centrifugation followed by density gradient ultracentrifugation

    • Negative stain electron microscopy to confirm VLP morphology

    • Western blotting to verify protein composition

    • Dynamic light scattering for size distribution analysis

  • Immunological assessment:

    • Evaluation of antibody responses against multiple viral proteins

    • Analysis of T cell responses to gauge cellular immunity

    • Challenge studies in appropriate animal models

What methodologies can assess the role of HCoV-229E E protein in modulating host immune responses?

Understanding E protein's immunomodulatory effects requires multifaceted approaches:

  • In vitro immune response models:

    • Transfect or transduce recombinant E protein into relevant immune cells (macrophages, dendritic cells)

    • Measure cytokine/chemokine production through ELISA, multiplex assays, or qRT-PCR

    • Assess inflammasome activation through caspase-1 activity and IL-1β processing

  • Signaling pathway analysis:

    • Employ phospho-specific antibodies to identify activated signaling cascades

    • Use pathway inhibitors to determine the mechanism of immune modulation

    • Perform gene expression profiling to identify transcriptional changes

  • Protein-protein interaction screening:

    • Yeast two-hybrid or mammalian two-hybrid screens to identify host factors

    • Affinity purification coupled with mass spectrometry (AP-MS)

    • Validate interactions through co-immunoprecipitation and functional assays

  • Comparative studies with mutant E proteins:

    • Generate E protein variants lacking specific domains or functions

    • Compare immunomodulatory effects to identify critical regions

    • Correlate with viral pathogenesis in cellular or animal models

How might artificial intelligence approaches accelerate inhibitor discovery for HCoV-229E E protein?

AI-driven methods offer several advantages for E protein drug discovery:

  • Structure-based virtual screening:

    • Use validated E protein structures as templates for molecular docking

    • Employ machine learning algorithms to score and rank potential inhibitors

    • Prioritize compounds with predicted binding to critical residues in the channel pore

  • Deep learning for compound optimization:

    • Train models on known ion channel inhibitors to predict activity

    • Generate novel chemical scaffolds with desired physicochemical properties

    • Optimize lead compounds for improved potency and selectivity

  • Molecular dynamics simulations:

    • Predict binding modes and conformational changes upon inhibitor binding

    • Calculate binding free energies to prioritize compounds for experimental testing

    • Identify allosteric binding sites beyond the channel pore

  • Integration with experimental data:

    • Implement active learning approaches that incorporate experimental feedback

    • Develop quantitative structure-activity relationship (QSAR) models

    • Combine with high-throughput screening data to improve predictive power

What approaches can detect structural dynamics of HCoV-229E E protein in membrane environments?

Understanding the dynamic behavior of E protein requires specialized techniques:

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS):

    • Measures accessibility of backbone amide hydrogens to solvent

    • Can detect conformational changes upon oligomerization or ligand binding

    • Compatible with detergent-solubilized or liposome-reconstituted protein

  • Single-molecule FRET (smFRET):

    • Label E protein with donor-acceptor fluorophore pairs at strategic positions

    • Monitor distance changes between domains during gating or assembly

    • Can detect rare or transient conformational states

  • Solid-state NMR spectroscopy:

    • Provides atomic-level information in lipid bilayer environments

    • Can determine orientation of transmembrane helices

    • Detects local structural changes upon inhibitor binding

  • Molecular dynamics simulations:

    • Model E protein pentamer in explicit lipid bilayers

    • Simulate ion permeation and gating mechanisms

    • Predict effects of mutations on channel stability and function

How can multi-coronavirus studies of E proteins inform pandemic preparedness?

Comparative studies across coronavirus E proteins can provide insights for future pandemic responses:

  • Evolutionary analysis:

    • Sequence conservation analysis to identify invariant residues as therapeutic targets

    • Positive selection analysis to detect rapidly evolving regions

    • Ancestral sequence reconstruction to understand evolutionary trajectories

  • Structure-function comparisons:

    • Determine structures of E proteins from diverse coronaviruses

    • Compare ion selectivity and conductance properties

    • Identify conserved vs. virus-specific interactions with host proteins

  • Broad-spectrum inhibitor development:

    • Screen compounds against multiple coronavirus E proteins

    • Identify pharmacophores that target conserved structural features

    • Develop combination approaches targeting multiple viral proteins

  • Predictive modeling for emerging coronaviruses:

    • Develop algorithms to predict E protein functions from sequence

    • Create models to assess pandemic potential based on E protein features

    • Establish rapid response protocols for characterizing novel E proteins

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 2025 TheBiotek. All Rights Reserved.