HSV 2 gG

Herpes Simplex Virus-2 gG
Shipped with Ice Packs
In Stock

Description

Definition and Biological Significance

Herpes Simplex Virus Type 2 (HSV-2) glycoprotein G (gG) is a viral envelope protein critical for immune evasion and serodiagnosis. It exists in two forms:

  • Secreted amino-terminal portion (sgG-2): Shed into the extracellular space.

  • Mature glycosylated carboxy-terminal portion (mgG-2): Cell- and virion-associated, heavily O-glycosylated, with a molecular mass of ~120 kDa .

gG-2 is highly divergent from HSV-1 gG, making it a type-specific marker for serological discrimination . Its role in modulating host immune responses, particularly chemokine binding to inhibit neutrophil chemotaxis, underscores its importance in viral pathogenesis .

Role in Immune Response and Serodiagnosis

gG-2 is a dominant antibody target, with type-specific epitopes enabling precise serological differentiation:

  • sgG-2: Detected early in infection but associated with cross-reactivity in some assays .

  • mgG-2: Higher specificity in ELISA due to reduced non-specific binding .

Key Findings:

  • Seroconversion Detection: mgG-2-based ELISA shows delayed seroconversion compared to Western blot in longitudinal studies .

  • Vaccine Potential: Immunization with mgG-2 + CpG adjuvant induces protective immunity in murine models, though neutralization activity remains absent .

Clinical Applications and Diagnostic Challenges

Diagnostic MethodPerformanceLimitationsSource
sgG-2 ELISASensitivity: 94–97%; Specificity: 89–91%False positives in low-prevalence cohorts
mgG-2 ELISASensitivity: 97–99%; Specificity: 97%Higher cost due to native purification
Rapid Lateral Flow TestSensitivity: 94%; Specificity: 99%Requires validation in diverse populations

Genetic Variability Impact:
East African HSV-2 isolates exhibit polymorphisms at positions gG-2 291 (Gly vs. Glu), 338 (Lys vs. Arg), 372, and 542 (immunodominant region), potentially reducing assay sensitivity in these populations .

Genetic Diversity and Global Strain Variability

HSV-2 gG exhibits significant genetic divergence, particularly in East Africa:

  • Amino Acid Signatures:

    • Position 291: Glycine (African) vs. Glutamic acid (Global).

    • Position 338: Lysine (African) vs. Arginine (Global).

    • Position 542: Polymorphisms affect epitope recognition .

This diversity necessitates region-specific diagnostic assays to ensure accuracy .

Research Advancements and Future Directions

Study FocusKey OutcomeImplicationSource
Vaccine DevelopmentmgG-2 + CpG elicits protective immunityPotential human trials for HSV-2 prophylaxis
Assay OptimizationmgG-2 outperforms sgG-2 in specificityImproved diagnostic accuracy in low-prevalence settings
Genomic SurveillanceEast African strains require tailored reagentsEnhanced global diagnostic reliability

Product Specs

Introduction
Herpes simplex virus (HSV) enters host cells through a multi-step process involving interactions between viral glycoproteins and cell surface receptors. The virus's outer envelope, studded with these glycoproteins, binds to specific receptors on the cell surface. This binding initiates membrane fusion, creating a pore that allows the virus to enter the host cell. HSV entry mirrors the entry mechanisms of other viruses. Initially, complementary receptors on the virus and cell surfaces draw the two membranes close. Subsequently, the membranes begin to merge, forming a hemifusion state. Finally, a stable pore forms, enabling the viral envelope contents to enter the host cell.
Description
The HSV-2 gG protein is a synthetically produced protein containing the immunodominant regions of the HSV-2 gG protein.
Purity
The protein purity exceeds 99%, as determined by Analytical HPLC and Mass Spectrometry.
Physical Appearance
The product is a clear solution that has been sterilized by filtration.
Formulation
The HSV-2 gG solution is formulated in 25mM Tris-HCl buffer with a pH of 8.0.
Stability
For optimal stability, HSV-2 gG protein should be stored below -18°C. While the protein remains stable at 4°C for up to 1 week, repeated freeze-thaw cycles should be avoided.
Purification Method
HSV-2 gG protein was purified by Prep HPLC.
Specificity
Immunoreactive with sera of HSV-infected individuals.

Q&A

What is the processing pathway of HSV-2 glycoprotein G?

HSV-2 glycoprotein G undergoes a unique processing pathway unlike other HSV proteins. The gG-2 precursor protein is cotranslationally glycosylated to generate a high-mannose intermediate. This intermediate is subsequently cleaved during processing into two distinct portions: a secreted amino-terminal portion and a cell- and virion-associated carboxy-terminal portion. The carboxy-terminal high-mannose intermediate undergoes further processing through extensive O-glycosylation to produce the mature gG-2 (mgG-2), which has an apparent molecular mass of approximately 120 kDa. The secreted portion is released into infected cell medium, while the mature gG-2 becomes incorporated into the viral envelope .

How does gG-2 differ from other HSV envelope glycoproteins?

gG-2 exhibits several unique characteristics that distinguish it from other HSV envelope glycoproteins. Unlike glycoproteins B and D, which elicit cross-reactive B- and T-cell responses between HSV-1 and HSV-2, gG-2 induces strictly type-specific immune responses. This type-specificity makes gG-2 an ideal antigen for discriminating between HSV-1 and HSV-2 infections in serological tests. Additionally, gG-2 undergoes a distinctive cleavage process during maturation, resulting in both secreted and membrane-associated forms, a property not observed with other HSV glycoproteins .

What are the functional domains of mature gG-2?

The mature form of gG-2 (mgG-2) contains several functional domains that contribute to its biological activities. The amino-terminal signal sequence (first 22 residues) directs the protein into the secretory pathway. Following this region is the portion that becomes the secreted amino-terminal fragment. The carboxy-terminal portion contains heavily O-glycosylated regions and a transmembrane domain that anchors the protein in the viral envelope. Amino acid sequencing has confirmed that the N-terminal residues of the secreted portion begin at position 23 (sequence: GSGVPGPI), confirming that the first 22 amino acids constitute the signal sequence. The immunodominant region recognized in diagnostic applications appears to reside between residues 321-580, as this fragment retains the antigenicity of the native protein .

How can researchers detect and characterize gG-2 expression in clinical isolates?

Detecting and characterizing gG-2 expression in clinical isolates requires a multi-technique approach. Initially, researchers should screen isolates using anti-gG-2 monoclonal antibodies through techniques such as immunofluorescence or ELISA. For detailed characterization, a combination of immunoblotting and radioimmunoprecipitation can identify both the secreted amino-terminal and cell-associated carboxy-terminal portions of gG-2.

In immunoblotting analyses, researchers should look for the carboxy-terminal high-mannose intermediate (approximately 77 kDa) and the fully glycosylated mature gG-2 (approximately 120 kDa) in infected cell lysates. The secreted portion can be detected in virus-infected cell medium. For confirming protein identity, amino acid sequencing of the N-terminal residues of the secreted portion should match the expected sequence (GSGVPGPI for HSV-2 strain HG52). For comprehensive genetic characterization, PCR amplification and sequencing of the complete gG-2 gene will identify any mutations, particularly frameshift mutations within runs of guanine or cytosine nucleotides .

What expression systems are most effective for producing recombinant gG-2 for research applications?

The baculovirus expression system has proven highly effective for producing recombinant gG-2 fragments with native-like antigenicity. Using the Bac-to-Bac baculovirus expression system, researchers have successfully expressed fragments such as gG321-580His (incorporating residues 321-580 with a histidine tag). This system allows for proper post-translational modifications including glycosylation patterns that more closely resemble those of native viral proteins compared to bacterial expression systems.

For optimal expression, researchers should clone the target gG-2 fragment into a baculovirus transfer vector containing a strong promoter such as the polyhedrin promoter, add a purification tag (histidine tags work well), and then generate recombinant baculovirus according to standard protocols. Infection of insect cells (Sf9 or High Five) at an appropriate multiplicity of infection (MOI), followed by harvest at 72-96 hours post-infection typically yields good protein expression. Purification can be accomplished using affinity chromatography appropriate for the chosen tag (e.g., nickel affinity chromatography for His-tagged proteins) .

How can the immunogenicity of gG-2 be enhanced for vaccine development?

Enhancing the immunogenicity of gG-2 for vaccine development requires strategic adjuvant selection and optimization of delivery methods. Research has demonstrated that combining mature gG-2 (mgG-2) with CpG adjuvant significantly improves protective efficacy. CpG oligodeoxynucleotides act as TLR9 agonists that promote Th1-type immune responses, which appear particularly important for protection against HSV-2.

The optimal immunization protocol based on mouse studies involves at least three immunizations with mgG-2 plus CpG, spaced 2-3 weeks apart. This regimen elicits robust gamma interferon (IFN-γ) responses by splenic CD4+ T cells upon antigen restimulation, which correlates with protection. Additionally, the immunization induces antibodies capable of macrophage-mediated antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent complement-mediated cytolysis (ADCML), rather than neutralizing antibodies.

For advanced vaccine formulations, researchers should consider combining mgG-2 with other HSV-2 antigens that might induce complementary immune responses, and explore novel delivery systems such as nanoparticles or viral vectors to further enhance immunogenicity and ensure appropriate antigen processing and presentation .

What are the experimental approaches to study the role of gG-2 in viral pathogenesis?

Studying the role of gG-2 in viral pathogenesis requires multiple experimental approaches. One powerful approach involves comparing naturally occurring gG-2-negative clinical isolates with gG-2-positive strains in animal models of infection. Alternatively, researchers can generate recombinant viruses with specific mutations in the gG-2 gene using bacterial artificial chromosome (BAC) technology or CRISPR/Cas9-mediated genome editing.

The mouse intravaginal challenge model provides a relevant system for evaluating how gG-2 affects pathogenesis. Key parameters to measure include: (1) viral replication in the vaginal mucosa through periodic vaginal swabbing and viral titration, (2) disease progression using standardized scoring systems, (3) neuroinvasion by assessing viral loads in dorsal root ganglia and spinal cord tissues, and (4) mortality rates.

For mechanistic studies, researchers should analyze local and systemic immune responses, including cytokine/chemokine profiles in vaginal washes, antigen-specific T cell responses in draining lymph nodes, and antibody responses in serum. Cell culture systems can complement in vivo studies by examining how gG-2 affects viral cell-to-cell spread versus release of extracellular virions, as gG-2-negative mutants have shown impaired capacity to produce extracellular infectious particles while maintaining cell-to-cell spread .

How can researchers develop and validate a type-specific ELISA for HSV-2 using gG-2?

Developing a type-specific ELISA for HSV-2 using gG-2 requires careful antigen selection and assay optimization. Researchers should:

  • Choose an appropriate gG-2 antigen: Either purified native mgG-2 from infected cells (using methods like H. pomatia lectin affinity purification) or recombinant gG-2 fragments (such as gG321-580His expressed in baculovirus systems) can serve as diagnostic antigens.

  • Optimize coating conditions: Use carbonate buffer (pH 9.6) for native mgG-2 (approximately 100 μg/ml, diluted 1:6,000) or phosphate-buffered saline for recombinant fragments, and determine optimal coating concentration through checkerboard titration.

  • Block non-specific binding sites with appropriate blocking buffer (typically 1-5% BSA or non-fat milk).

  • Select appropriate detection system: For human samples, use enzyme-conjugated anti-human IgG (such as peroxidase-conjugated or alkaline phosphatase-conjugated goat anti-human IgG) at optimized dilutions (typically 1:3,000 to 1:3,500).

  • Establish cutoff values: Define cutoffs based on mean absorbance values of confirmed HSV-1 and HSV-2-negative sera, plus a fixed OD value (typically 0.2 OD units).

  • Validate the assay: Test against a panel of well-characterized serum samples, including HSV-1-positive/HSV-2-negative sera to confirm type-specificity, and compare results with established reference methods such as Western blot or commercial HSV-2 type-specific assays.

What are the limitations of gG-2-based serological testing in populations with gG-2-negative HSV-2 infections?

While gG-2-based serological testing is highly specific for differentiating HSV-2 from HSV-1 infections, it has important limitations when applied to populations with gG-2-negative HSV-2 infections. Patients infected with HSV-2 strains lacking functional gG-2 expression may not develop antibodies against gG-2, resulting in false-negative serological results. This represents a fundamental limitation of gG-2-based assays rather than a technical issue with assay design.

Research has shown that some patients carrying gG-2-negative HSV-2 isolates lack detectable antibodies against gG-2 in type-specific ELISA tests. Interestingly, other patients with gG-2-negative isolates do show antibodies against gG-2, suggesting they may harbor multiple HSV-2 strains or were previously infected with a gG-2-positive strain before acquiring a gG-2-negative variant.

The table below illustrates this phenomenon with endpoint titers against gG-2 and gG-1 antigens from patients with gG-2-negative HSV-2 isolates:

Patient no.Endpoint titer to gG-2 antigenEndpoint titer to gG-1 antigen
24344001,600
512400100
453800
147100
4444200200

Note: "–" indicates endpoint titer of <100.

To mitigate this limitation, researchers studying populations where gG-2-negative variants might be present should consider complementary approaches such as PCR detection of HSV-2 DNA from lesions or nucleic acid amplification tests for definitive diagnosis .

How do researchers optimize the specificity and sensitivity balance in gG-2-based diagnostic assays?

Optimizing the balance between specificity and sensitivity in gG-2-based diagnostic assays requires systematic evaluation of multiple parameters:

  • Antigen selection: Using immunodominant fragments of gG-2 rather than the complete protein can sometimes improve specificity without compromising sensitivity. Research has shown that the fragment comprising residues 321-580 contains sufficient immunodominant epitopes while reducing nonspecific interactions.

  • Signal-to-noise ratio enhancement: Researchers should optimize blocking conditions to minimize background while preserving specific signals. Different blocking agents (BSA, non-fat milk, commercial blocking buffers) should be compared for optimal performance.

  • Serum dilution optimization: Testing multiple serum dilutions can help identify the optimal dilution that maximizes discrimination between positive and negative samples. Typically, serum dilutions between 1:100 and 1:400 provide good performance.

  • Cutoff determination strategy: Multiple approaches exist for establishing cutoffs, including:

    • Statistical approaches (mean plus multiple standard deviations of negative controls)

    • ROC curve analysis to mathematically determine the optimal cutoff for balancing sensitivity and specificity

    • Adding a fixed OD value (e.g., 0.2) to the mean of negative controls

  • Equivocal zone implementation: Introducing an equivocal zone around the cutoff value helps manage borderline results that could be either false positives or false negatives.

  • Cross-reactivity elimination: Pre-absorption of sera with HSV-1 antigens can sometimes improve HSV-2 specificity by removing potentially cross-reactive antibodies.

How does the immune response to gG-2 differ from responses to other HSV-2 glycoproteins?

The immune response to gG-2 differs significantly from responses to other HSV-2 glycoproteins in several key aspects:

Type-specificity: The most distinctive feature of gG-2-directed immune responses is their strict type-specificity. While most HSV glycoproteins (such as gB and gD) elicit antibodies and T-cell responses that cross-react between HSV-1 and HSV-2 due to high sequence homology, gG-2 induces antibodies that are exclusively specific to HSV-2 with no cross-reactivity to HSV-1. This type-specificity forms the basis for using gG-2 in discriminating serodiagnosis.

Antibody functionality: Antibodies against gG-2 appear to function differently from those against other glycoproteins. While antibodies to glycoproteins like gB and gD often exhibit neutralizing activity (directly blocking viral entry), anti-gG-2 antibodies generally lack neutralizing capability. Instead, they mediate protection through other mechanisms such as antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent complement-mediated lysis (ADCML).

T-cell response profile: The T-cell response to gG-2 also shows distinctive features. Studies with mice immunized with mgG-2 plus CpG adjuvant have demonstrated that protection correlates with a robust gamma interferon (IFN-γ) response by CD4+ T cells upon antigen restimulation both in vitro and in vaginal secretions following challenge. This Th1-polarized response appears particularly important for protection against HSV-2 infection.

These unique immunological properties make gG-2 particularly valuable for type-specific diagnosis and as a potential vaccine component that could complement the immune responses induced by other HSV-2 glycoproteins .

What methodologies are used to evaluate T-cell responses to gG-2 in vaccine studies?

Evaluating T-cell responses to gG-2 in vaccine studies requires comprehensive methodological approaches that assess both quantity and functionality of responding T cells:

  • Ex vivo antigen restimulation assays: Splenocytes or lymph node cells from immunized animals are cultured with purified gG-2 antigen, and the response is measured through:

    • ELISPOT assays to enumerate cells secreting specific cytokines (particularly IFN-γ)

    • Intracellular cytokine staining followed by flow cytometry to identify specific T-cell subsets (CD4+ vs. CD8+) producing cytokines

    • Multiplex cytokine assays on culture supernatants to assess the cytokine profile breadth

  • In vivo T-cell functionality: Following immunization and challenge:

    • Cytokine measurements in vaginal washes (especially IFN-γ) 1 day post-infection provide evidence of functional T-cell responses at the infection site

    • Adoptive transfer of T cells from immunized to naïve animals can establish the protective capacity of gG-2-specific T cells

    • In vivo T-cell depletion (using anti-CD4 or anti-CD8 antibodies) can determine which T-cell subset is essential for protection

  • T-cell epitope mapping: Overlapping peptide libraries spanning the gG-2 sequence can identify specific epitopes recognized by T cells through:

    • T-cell proliferation assays

    • Cytokine ELISPOT with individual peptides

    • MHC tetramer staining for identified epitopes

  • Memory T-cell assessment: Long-term studies should evaluate the development of memory T cells by:

    • Phenotypic characterization of T cells (CD44, CD62L, CD127 expression)

    • Functional recall responses at extended timepoints post-immunization

    • Protection against delayed challenge

Research has shown that mice immunized with mgG-2 plus CpG develop gG-2-specific CD4+ T cells that produce IFN-γ upon restimulation, and these responses correlate with protection against vaginal HSV-2 challenge .

How can researchers distinguish between antibody responses to the secreted versus mature portions of gG-2?

Distinguishing between antibody responses to the secreted versus mature portions of gG-2 requires strategic experimental design and specific reagents:

  • Recombinant protein expression: Express the secreted amino-terminal portion and the mature carboxy-terminal portion of gG-2 as separate recombinant proteins. These can be produced in baculovirus expression systems with appropriate tags for purification.

  • Domain-specific ELISA: Develop separate ELISA assays using:

    • The purified secreted portion as coating antigen

    • The purified mature portion as coating antigen

    • Test patient sera against both antigens in parallel to determine reactivity patterns

  • Competitive inhibition assays: Pre-incubate sera with one portion (e.g., secreted) before testing reactivity against the other (e.g., mature) to identify portion-specific antibodies versus those that recognize shared epitopes.

  • Western blot analysis: Use immunoblotting with both portions separated by SDS-PAGE to visualize antibody binding patterns. The secreted portion (visible in virus-infected cell medium) and the mature gG-2 (in cell lysates) can be distinguished by their different molecular masses.

  • Absorption studies: Systematically deplete sera of antibodies specific to one portion by pre-absorption with the recombinant protein, then test the depleted sera for remaining reactivity to the other portion.

What techniques are used to study the post-translational modifications of gG-2?

Studying the post-translational modifications of gG-2 requires sophisticated analytical approaches due to its complex processing pathway and extensive glycosylation:

  • Glycosylation analysis:

    • N-glycan analysis: Treatment with endoglycosidases (PNGase F, Endo H) followed by mobility shift analysis on SDS-PAGE can assess N-glycan content and type

    • O-glycan analysis: Chemical methods (β-elimination) or specific O-glycosidases in combination with mass spectrometry can characterize O-glycan structures

    • Lectin binding assays: Different lectins (such as H. pomatia lectin used for purification of gG-2) bind specific glycan structures, allowing profiling of glycosylation patterns

  • Pulse-chase experiments:

    • Metabolic labeling with radioactive amino acids or sugars combined with chase periods of varying duration

    • Immunoprecipitation at different time points to track the conversion of the precursor to intermediate and mature forms

    • Analysis of secreted versus cell-associated fractions to monitor protein trafficking

  • Mass spectrometry applications:

    • Identification of exact cleavage sites between the secreted and mature portions

    • Mapping of glycosylation sites through glycopeptide analysis

    • Structural characterization of attached glycans

  • Site-directed mutagenesis:

    • Mutation of potential glycosylation sites to evaluate their importance for protein folding, cleavage, and function

    • Creation of truncation mutants to determine domains required for proper processing

These techniques have revealed that gG-2 undergoes cotranslational N-glycosylation generating a high-mannose intermediate, followed by proteolytic cleavage and extensive O-glycosylation of the carboxy-terminal portion. The heavily O-glycosylated mature gG-2 has an apparent molecular mass of approximately 120 kDa, significantly larger than predicted from the amino acid sequence alone, indicating the substantial contribution of glycosylation to the protein's structure .

How do natural mutations in the gG-2 gene affect protein expression and function?

Natural mutations in the gG-2 gene have significant effects on protein expression and function, providing valuable insights into structure-function relationships:

  • Frameshift mutations:
    Clinical HSV-2 isolates with frameshift mutations in the gG-2 gene have been identified, typically involving insertion or deletion of guanine or cytosine nucleotides within homopolymeric stretches (runs of five or more G or C). These mutations result in premature termination codons and can affect:

    • Complete absence of both secreted and mature gG-2 protein expression (as observed in four clinical isolates)

    • Partial expression where only the secreted portion is produced normally while the mature portion is truncated (as observed in one clinical isolate with a frameshift mutation upstream of but adjacent to the transmembrane region)

  • Functional consequences:

    • gG-2-negative HSV-2 isolates remain viable and can cause clinical lesions in immunocompetent patients, confirming that gG-2 is non-essential for viral replication and pathogenesis in vivo

    • In cell culture, mgG-2-negative HSV-2 mutants show altered viral spread patterns, primarily spreading cell-to-cell with impaired capacity to produce extracellular infectious particles

    • The rarity of gG-2-negative clinical isolates (only five identified in a large screening study) suggests that intact gG-2 provides some selective advantage during natural infection

  • Host immune responses:

    • Patients infected with gG-2-negative strains may lack antibodies against gG-2, potentially resulting in false-negative type-specific serological tests

    • Some patients with gG-2-negative isolates still possess gG-2 antibodies, suggesting either co-infection with multiple strains or sequential infection with different variants

These natural mutants serve as valuable tools for understanding the contributions of specific domains to gG-2 function and the role of this glycoprotein in the HSV-2 lifecycle .

What is the role of gG-2 in HSV-2 virion attachment, entry, and cell-to-cell spread?

The role of gG-2 in HSV-2 virion attachment, entry, and cell-to-cell spread remains incompletely understood but research has provided some important insights:

  • Virion attachment and entry:
    Unlike essential glycoproteins (gB, gD, gH/gL) that directly mediate virion attachment and fusion with host cell membranes, gG-2 appears to play a more accessory role in these processes. Experimental evidence suggests:

    • gG-2 is not absolutely required for virion attachment or entry, as gG-2-negative isolates remain infectious

    • Antibodies against gG-2 typically lack neutralizing activity, further indicating that gG-2 is not critically involved in the entry process

    • The mature form of gG-2 is incorporated into the viral envelope, where it could potentially modulate interactions with host cell receptors or other viral glycoproteins

  • Cell-to-cell spread:
    More significant roles for gG-2 have been suggested in cell-to-cell spread mechanisms:

    • An mgG-2-negative HSV-2 mutant was shown to spread primarily from cell to cell rather than through the production of extracellular infectious particles

    • This suggests that gG-2 may be particularly important for the efficient release of mature virions from infected cells

    • The secreted portion of gG-2 might influence the extracellular environment in ways that facilitate virion release or stability

  • Potential immunomodulatory functions:
    Some research suggests gG-2 may have immunomodulatory roles that indirectly affect viral spread:

    • The secreted portion could potentially interfere with host immune responses

    • The mature portion might shield other viral proteins from immune recognition

Understanding these functions is complicated by the processing of gG-2 into two distinct portions with potentially different roles. Further research using precisely engineered mutants that selectively affect either the secreted or mature portions would help clarify their specific contributions to viral entry and spread .

How does the antigenic structure of gG-2 compare between different HSV-2 strains and isolates?

The antigenic structure of gG-2 shows both conservation and variation between different HSV-2 strains and isolates, with important implications for diagnostics and vaccine development:

  • Epitope conservation:

    • The type-specific epitopes of gG-2 used in serodiagnosis appear to be highly conserved among most HSV-2 isolates, explaining the effectiveness of gG-2 as a diagnostic antigen

    • Both human sera and monoclonal antibodies directed against gG-2 typically recognize a broad range of clinical HSV-2 isolates

    • The immunodominant region comprising residues 321-580 contains particularly well-conserved epitopes, as evidenced by the high sensitivity and specificity of diagnostic tests using this fragment

  • Structural variations:

    • While complete absence of gG-2 expression is rare (as in the gG-2-negative isolates), more subtle variations in gG-2 structure may exist

    • The isolate VI-4444 shows a slight reduction in the apparent molecular mass of mature gG-2 (115 kDa versus the typical 120 kDa), suggesting possible differences in glycosylation patterns or other post-translational modifications

    • Polymorphisms in the gG-2 gene that don't abolish expression might still alter specific epitopes, potentially affecting recognition by some monoclonal antibodies

  • Implications for research:

    • Researchers developing diagnostic tests or vaccines based on gG-2 should consider testing multiple clinical isolates to ensure broad recognition

    • Monoclonal antibody panels targeting different epitopes of gG-2 can help characterize antigenic variations among isolates

    • Next-generation sequencing of the gG-2 gene from diverse clinical isolates would provide a more comprehensive view of natural variation in this protein

  • Functional consequences:

    • Whether antigenic variations in gG-2 correlate with differences in pathogenicity or immune evasion remains an important question for future research

    • The conservation of type-specific epitopes suggests selective pressure to maintain certain structural features of gG-2, despite it being non-essential for replication

While dramatic variations like complete absence of gG-2 are rare, more subtle antigenic differences may exist among HSV-2 strains and could influence host-pathogen interactions in ways that remain to be fully characterized .

Product Science Overview

Structure and Function of gG-2

Glycoprotein G-2 (gG-2) is a type-specific glycoprotein found on the envelope of HSV-2. It is encoded by the US4 gene and is involved in the virus’s ability to infect host cells. The primary function of gG-2 is to facilitate the attachment and entry of the virus into the host cell by interacting with cell surface receptors. This interaction is critical for the initiation of infection and the subsequent replication of the virus.

Role in Immune Response

gG-2 is also a major target for the host’s immune response. The immune system recognizes gG-2 as a foreign antigen and produces specific antibodies against it. These antibodies can neutralize the virus and prevent it from infecting new cells. The presence of gG-2-specific antibodies in the blood is a marker of HSV-2 infection and is used in serological tests to diagnose the infection .

Diagnostic Applications

The detection of antibodies against gG-2 is a cornerstone of serological testing for HSV-2. Commercially available assays, such as the HerpeSelect HSV-2 ELISA, utilize gG-2 to detect the presence of HSV-2-specific antibodies in the blood . These tests have significantly improved the accuracy of HSV-2 diagnosis, especially in distinguishing between HSV-1 and HSV-2 infections . The specificity of gG-2-based tests is high, making them reliable tools for clinical diagnosis.

Clinical Significance

The accurate diagnosis of HSV-2 infection is essential for managing the disease and preventing its transmission. HSV-2 is a chronic infection that can cause recurrent genital lesions and has been associated with an increased risk of acquiring and transmitting HIV. Therefore, identifying individuals with HSV-2 infection through gG-2-based serological tests is crucial for public health interventions and for guiding clinical management.

Research and Future Directions

Ongoing research is focused on understanding the detailed mechanisms of gG-2’s interaction with the host immune system and its role in viral pathogenesis. Advances in this area could lead to the development of new therapeutic strategies and vaccines targeting gG-2. Additionally, improving the sensitivity and specificity of gG-2-based diagnostic tests remains a priority to enhance the accuracy of HSV-2 detection in various populations .

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.