Recombinant Human herpesvirus 2 Glycoprotein N

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format currently in stock. However, if you have specific format requirements, please indicate them when placing your order. We will prepare the product according to your needs.
Lead Time
Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
Note: All our proteins are shipped with standard blue ice packs by default. If you require dry ice shipping, please communicate with us in advance as 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 briefly centrifuging the vial prior to opening to ensure the contents are settled at 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 glycerol concentration is 50%. Customers may use this as a reference.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer ingredients, temperature, and the protein's intrinsic stability.
Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. The shelf life of 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 production. If you have a specific tag type preference, please inform us, and we will prioritize development with the specified tag.
Synonyms
gN; Envelope glycoprotein N
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
26-87
Protein Length
Full Length of Mature Protein
Species
Human herpesvirus 2 (strain HG52) (HHV-2) (Human herpes simplex virus 2)
Target Names
gN
Target Protein Sequence
EPPNAAGARGVIGDAQCRGDSAGVVSVPGVLVPFYLGMTSMGVCMIAHVYQICQRALAAG SA
Uniprot No.

Target Background

Function
Envelope glycoprotein crucial for proper maturation of gM and modulation of its membrane fusion activity. It also plays a critical role in virion morphogenesis.
Database Links

KEGG: vg:1487337

Protein Families
Herpesviridae glycoprotein N family
Subcellular Location
Virion membrane; Single-pass type I membrane protein. Host membrane; Single-pass type I membrane protein. Host Golgi apparatus, host trans-Golgi network.

Q&A

What expression systems are most suitable for producing recombinant HSV-2 glycoprotein N?

While specific data on glycoprotein N is limited, research on other HSV-2 glycoproteins suggests that eukaryotic expression systems are generally superior for recombinant herpesvirus glycoproteins. The baculovirus expression system using Spodoptera frugiperda (Sf9) cells has proven particularly effective for producing biologically active HSV-2 glycoproteins.

For example, research has demonstrated successful expression of HSV-2 glycoprotein D (gD2) in Sf9 cells, with yields reaching 192 mg/L under optimized conditions . This system preserves critical post-translational modifications, including glycosylation patterns essential for proper protein folding and immunogenicity.

Parameters for optimized expression in Sf9 cells include:

ParameterOptimized ValueImpact on Yield
Glucose concentration15 mMSupports high-density growth
Glutamine concentration0.4 g/LEnhances protein expression
Dissolved oxygen (DO)40%Maintains cell viability
Cell density9.6×10^6 cells/mLMaximizes volumetric yield

Alternative expression systems include mammalian cell lines (CHO, HEK293), which may better replicate human glycosylation patterns but typically yield lower protein quantities.

What purification strategies yield the highest purity and biological activity for recombinant HSV-2 glycoproteins?

For recombinant HSV-2 glycoproteins, multi-step purification protocols are necessary to maintain structural integrity and biological activity. Based on successful approaches for similar HSV-2 glycoproteins, an effective purification strategy should include:

  • Initial clarification via centrifugation (10,000×g for 30 minutes)

  • Affinity chromatography (typically using His-tag or immunoaffinity approaches)

  • Ion-exchange chromatography to remove contaminants

  • Size-exclusion chromatography for final polishing and buffer exchange

When glycoprotein D was expressed with a histidine tag in the baculovirus system, researchers achieved greater than 95% purity using nickel affinity chromatography followed by size-exclusion chromatography . Purification under non-denaturing conditions is critical for preserving conformational epitopes.

How can researchers assess the correct folding and glycosylation of recombinant HSV-2 glycoprotein N?

Proper folding and glycosylation are essential for biological activity of recombinant glycoproteins. Assessment approaches should include:

  • Structural analysis: Circular dichroism spectroscopy to evaluate secondary structure elements

  • Glycosylation profiling: Mass spectrometry to identify glycan structures and occupancy at predicted N-linked glycosylation sites

  • Functional assays: Binding studies with known interaction partners or antibodies that recognize conformational epitopes

  • Thermal shift assays: To evaluate protein stability and proper folding

Research on other HSV glycoproteins indicates that asparagine residues in the consensus sequence Asn-X-Ser/Thr serve as N-linked glycosylation sites. While glycosylation site positions are not always conserved among herpesvirus glycoprotein homologs, their presence is critical for proper function . Biophysical characterization should confirm that recombinant glycoprotein N contains similar post-translational modifications to the native viral protein.

What are the most effective methods for evaluating the immunogenicity of recombinant HSV-2 glycoprotein N?

Comprehensive immunogenicity assessment requires both in vitro and in vivo approaches:

In vitro methods:

  • Neutralization assays using serum from immunized animals to assess the capacity of antibodies to block viral infection

  • ELISA-based antibody titer determination

  • T-cell activation assays (e.g., ELISpot) to evaluate cellular immune responses

In vivo methods:

  • Animal immunization models (guinea pigs are preferred for HSV-2)

  • Challenge studies to assess protection against viral infection

  • Analysis of antibody isotype profiles (IgG, IgA) in serum and mucosal secretions

How can researchers effectively analyze the interaction between recombinant HSV-2 glycoprotein N and other viral proteins?

Understanding protein-protein interactions is crucial for elucidating glycoprotein N's role in viral biology. Methodological approaches include:

  • Co-immunoprecipitation: Using tagged recombinant glycoprotein N to pull down interaction partners

  • Surface plasmon resonance (SPR): For quantitative binding kinetics (kon, koff, KD)

  • Proximity ligation assays: To visualize protein interactions in situ

  • Yeast two-hybrid screening: For discovery of novel interaction partners

  • Cryo-electron microscopy: For structural analysis of glycoprotein complexes

Studies with other HSV-2 glycoproteins have demonstrated important interactions that mediate viral entry and immune evasion. For example, glycoprotein G has been shown to modify host signaling pathways by binding to and increasing the function of nerve growth factor (NGF) , suggesting that glycoproteins may have roles beyond structural components of the viral envelope.

What are the critical quality attributes for recombinant HSV-2 glycoprotein N in vaccine development?

For vaccine applications, recombinant glycoprotein quality must be rigorously assessed across multiple parameters:

Quality AttributeAnalytical MethodAcceptance Criteria
PuritySDS-PAGE, SEC-HPLC≥95% purity
IdentityMass spectrometry, Western blotMatches predicted sequence
PotencyNeutralization assayConsistent ED50 values
AggregationDLS, SEC<5% aggregates
EndotoxinLAL test<0.5 EU/mg protein
Host cell proteinsELISA<100 ppm
Glycosylation profileLC-MS/MSConsistent pattern batch-to-batch

How can epitope mapping of recombinant HSV-2 glycoprotein N improve diagnostic and therapeutic applications?

Detailed epitope mapping enables development of targeted diagnostics and therapeutics. A methodological approach includes:

  • Overlapping peptide arrays: Synthesize overlapping peptides spanning the entire glycoprotein N sequence

  • Alanine scanning mutagenesis: Systematically replace amino acids with alanine to identify critical residues

  • Hydrogen-deuterium exchange mass spectrometry: Map conformational epitopes

  • X-ray crystallography of antibody-antigen complexes: Determine precise epitope structure

Research on HSV-2 glycoprotein G demonstrated that specific peptide epitopes can distinguish between HSV-1 and HSV-2 antibodies. One study found that "Herpes simplex virus type 2-specific human seroreactivities were mainly seen against three peptides, peptides G2-64, G2-69, and G2-70, located in the C-terminal part of glycoprotein G" . The epitope of peptide G2-69 was mapped to the amino acid sequence RYAHPS, and it reacted with 93% of HSV-2 IgG-positive human serum samples while showing no reactivity with HSV-2 negative samples .

Similar epitope mapping of glycoprotein N could identify type-specific regions useful for diagnostic applications and neutralizing epitopes for therapeutic antibody development.

What strategies can enhance the efficacy of recombinant HSV-2 glycoprotein-based vaccines?

Based on lessons from previous HSV-2 glycoprotein vaccine trials, several strategies may improve efficacy:

  • Adjuvant optimization: Studies have shown that aluminum hydroxide combined with monophosphoryl lipid A (MPL) enhances immune responses to glycoprotein vaccines

  • Multi-valent formulations: Combining multiple glycoproteins may provide broader protection than single glycoprotein approaches

  • Prime-boost strategies: Sequential vaccination with DNA followed by protein, or viral vectors followed by protein

  • Mucosal delivery systems: To enhance secretory IgA production at sites of viral entry

  • Structure-based design: Engineering glycoproteins to expose neutralizing epitopes

Comparative studies between recombinant glycoprotein subunit vaccines and more complex vaccine platforms provide important insights. One study found that "CJ2-gD2 [a non-replicating dominant-negative HSV-2 virus vaccine] is 8 times more effective than the gD2-alum/MPL subunit vaccine in eliciting an anti-HSV-2 specific neutralizing antibody response and offers significantly superior protection against primary and recurrent HSV-2 genital infections" . This suggests that presentation of glycoproteins in a virus-like context may be advantageous compared to purified recombinant proteins alone.

How does the structural homology between HSV-1 and HSV-2 glycoproteins affect cross-protection strategies?

Cross-protection between HSV-1 and HSV-2 is an important consideration for vaccine development. Research approaches include:

  • Comparative structural analysis: Identify conserved and variable regions between HSV-1 and HSV-2 homologs

  • Cross-neutralization studies: Assess whether antibodies against one type neutralize the other

  • T-cell epitope mapping: Identify shared T-cell epitopes that might contribute to cross-protection

  • Challenge studies in animal models: Evaluate whether immunization against one type protects against the other

Research has shown complex relationships between prior HSV-1 immunity and HSV-2 protection. Clinical studies found that "prior HSV-1 infection did not appear to protect against HSV-2 infection" , despite the induction of HSV-2 binding and neutralizing antibodies in HSV-1-seropositive individuals.

The identification of type-specific and cross-reactive epitopes in glycoprotein N would inform the design of vaccines offering broader protection against both HSV types.

What are the optimal in vitro models for studying recombinant HSV-2 glycoprotein N function?

Selection of appropriate cellular models is critical for functional studies:

  • Cell lines for entry studies: Vero, HeLa, and human neuronal cell lines

  • Primary cells for physiological relevance: Human epithelial cells from genital mucosa, primary neurons

  • 3D tissue models: Reconstructed human vaginal or cervical epithelium

  • Co-culture systems: Epithelial cells with dendritic cells to study immune interactions

Functional assays should evaluate:

  • Binding to cellular receptors

  • Effects on viral entry

  • Interference with viral neutralization

  • Modulation of immune responses

Studies with HSV-2 glycoprotein G revealed its ability to modify host signaling pathways , suggesting that glycoproteins may have roles beyond structural components of the viral envelope. Similar functional studies with glycoprotein N would help elucidate its biological functions.

What animal models are most appropriate for evaluating recombinant HSV-2 glycoprotein N-based vaccines?

Animal model selection should be based on specific research questions:

Animal ModelAdvantagesLimitationsBest Applications
Guinea pigDevelops genital disease similar to humans; Establishes latency; Exhibits recurrencesLimited immunological reagentsVaccine efficacy; Disease pathogenesis
MouseWell-characterized immune system; Abundant reagents; Lower costLimited recurrent diseaseImmunogenicity studies; Mechanism investigations
Cotton ratPermissive for HSV replicationLimited disease recurrenceInitial screening of vaccine candidates
RabbitOcular model well-establishedLess relevant for genital diseaseOcular herpes studies

The guinea pig model has proven particularly valuable as it "develops genital disease similar to that seen in humans and, like humans, experiences spontaneous recurrences of genital herpetic lesions" . Important endpoints in animal studies include:

  • Protection against primary infection

  • Reduction in viral shedding

  • Prevention of latency establishment

  • Reduction in frequency and severity of recurrences

  • Viral DNA detection in dorsal root ganglia

Studies have shown that "no challenge wild-type HSV-2 viral DNA was detectable in dorsal root ganglia DNA isolated from CJ2-gD2-immunized guinea pigs on day 60 post-challenge" , establishing an important efficacy benchmark for HSV-2 vaccines.

How should researchers address discrepancies between in vitro neutralization titers and in vivo protection?

Neutralizing antibody titers often correlate poorly with actual protection, requiring sophisticated analysis approaches:

  • Multivariate analysis: Incorporate multiple immune parameters (antibody titers, T-cell responses, antibody functionality)

  • Systems serology: Comprehensive profiling of antibody features (isotype, glycosylation, Fc receptor binding, ADCC activity)

  • Machine learning algorithms: Identify non-obvious correlates of protection

  • Longitudinal sampling: Track immune parameters over time rather than at single timepoints

Clinical trials of HSV-2 glycoprotein D vaccines revealed this disconnect, finding "no differences in peak titer after vaccination for HSV-2 antibodies prior to infection between 'vaccinated infected' and 'vaccinated uninfected' persons" . This suggests that antibody quantity alone may not predict protection, and "more information about the cellular and mucosal immune responses to HSV-2 infection is needed before we can attempt to identify laboratory-based correlates of immune protection" .

What statistical approaches are most appropriate for analyzing protective efficacy in HSV-2 vaccine trials?

Robust statistical methods are essential for evaluating vaccine efficacy:

  • Survival analysis: Kaplan-Meier curves with log-rank tests to compare time to infection

  • Cox proportional hazards models: Adjust for covariates that might influence susceptibility

  • Mixed-effects models: For longitudinal analysis of recurrence rates

  • Bayesian approaches: Incorporate prior information and update probability estimates as data accumulates

Trials should be powered to detect differences in multiple endpoints:

  • Prevention of HSV-2 acquisition (seroconversion)

  • Reduction in clinical disease severity

  • Decrease in viral shedding frequency

  • Reduction in recurrence rate

Phase 3 trials of HSV-2 glycoprotein D vaccines highlight the importance of robust endpoints, as some vaccines showed promise in reducing clinical disease but failed to prevent HSV-2 acquisition . This suggests that vaccines that "do not affect acquisition of genital HSV-2 but do limit subsequent clinical disease manifestations could paradoxically increase the prevalence of subclinical infection" , potentially increasing transmission risk.

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.