Recombinant Human herpesvirus 6A Envelope glycoprotein H (gH)

Shipped with Ice Packs
In Stock

Description

Overview of Recombinant Human Herpesvirus 6A Envelope Glycoprotein H (gH)

Recombinant Human Herpesvirus 6A (HHV-6A) Envelope Glycoprotein H (gH) is a bioengineered protein produced through bacterial expression systems. It represents a critical component of the viral fusion machinery, essential for HHV-6A entry into host cells. This recombinant protein is widely used in virological research to study viral tropism, entry mechanisms, and immune interactions.

Role in Viral Entry

gH is part of a four-component complex (gH/gL/gQ1/gQ2) that binds to the cellular receptor CD46 . This interaction facilitates membrane fusion, mediated by glycoprotein B (gB) and the gH/gL complex .

Expression Systems

Recombinant gH is typically expressed in E. coli, leveraging bacterial protein synthesis machinery. Two formats are available:

  1. His-Tagged: Facilitates purification via nickel-affinity chromatography.

  2. Tag-Free: Used for studies requiring native-like protein interactions .

Purification and Quality Control

StepDetailsSource
PurificationNickel-affinity chromatography (His-tagged), size-exclusion chromatography
Purity AssessmentSDS-PAGE (>90% purity)
StorageLyophilized powder stored at -20°C/-80°C; avoid freeze-thaw cycles

Functional Studies

  • Binding Assays: Recombinant gH is used to study interactions with cellular receptors (e.g., CD46) and co-receptors (e.g., gp96) .

  • Epitope Mapping: Neutralizing antibodies targeting gH’s linear epitopes (e.g., residues near domain II) have been identified, though their role in viral neutralization remains under investigation .

Role in Pathogenesis

  • Immune Evasion: HHV-6A infection induces HLA-G expression, which may suppress NK cell activity and promote immune tolerance .

  • Latency and Reactivation: Integrated HHV-6A genomes in telomeres can reactivate, producing infectious virions. gH’s role in reactivation is an active area of study .

Comparative Analysis of Recombinant gH Products

Product IDStrainAA RangeTagBiological Activity (ELISA)Source
RFL16786HFUganda-110217–694HisConfirmed
Recombinant gH (GS)GS24–694His/Tag-freeConfirmed
Uganda-1102 gHUganda-110217–694HisFunctional binding

Key Research Insights

  1. Entry Mechanism: gH/gL/gQ1/gQ2 binds CD46, while gp96 facilitates viral attachment and fusion .

  2. Antigenic Targets: Neutralizing epitopes on gH are under investigation for vaccine development .

  3. Diagnostic Utility: Recombinant gH is used in serological assays to detect HHV-6A-specific antibodies .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Our proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a guideline.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
gH; LF2; U48; Envelope glycoprotein H; gH
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
17-694aa
Protein Length
Full Length of Mature Protein
Species
Human herpesvirus 6A (strain GS) (HHV-6 variant A) (Human B lymphotropic virus)
Target Names
gH
Target Protein Sequence
WRPLNISNSSHCRNGNFENPIVRPGFITFNFYTKNDTRIYQVPKCLLGSDITYHLFDAINTTESLTNYEKRVTRFYEPPMNDILRLSPVPSVKQFNLDRSIQPQVVYSLNMYPSQGIYYVRVVEVRQMQYDNVSCKLPNSLKELIFPVQVRCAKITRYVGEDIYTHFFTPDFMILYIQNPAGDLTMMYGNTTSINFKAPYKKSSFIFKQTLTDDLLLIVEKDVIDVQYRFISDATFVDETLNDVDEVEALLLKFNNLGIQTLLRGDCKKPNYAGIPQMMFLYGIVHFSYSTKNTGPMPVLRVLKTHENLLSIDSFVNRCVNVSEGTLQYPKMKEFLKYEPSDYSYITKNKSISVSTLLTYLATAYESNVTISKYKWTDIANTLQNIYEKHMFFTNLTFSDRETLFMLAEIANIIPTDERMQRHMQLLIGNLCNPVEIVSWARMLTADRAPNLENIYSPCASPVRRDVTNSFLKTVLTYASLDRYRSDMMEMLSVYRPPNMERVAAIQCLSPSEPAASLTLPNVTFVISPSYVIKGVSLTITTTIVATSIIITAIPLNSTCVSTNYKYAGQDLLVLRNISSQTCEFCQSVVMEYDDIDGPLQYIYIKNIDELKTLTDPNNNLLVPNTRTHYLLLAKNGSVFEMSEVGIDIDQVSIILVIIYILIAIIALFGLYRLIRLC
Uniprot No.

Target Background

Function

The heterodimeric glycoprotein H-glycoprotein L complex is crucial for the fusion of viral and host cell plasma membranes, enabling viral entry. Following initial receptor binding, membrane fusion is mediated by the fusion machinery comprising gB and the gH/gL heterodimer. It may also participate in the fusion between the virion envelope and the outer nuclear membrane during virion morphogenesis.

Gene References Into Functions
  1. This study demonstrates that gH, gL, gQ1, and gQ2 are all essential for the trafficking and proper folding of the gH/gL/gQ1/gQ2 complex, and consequently, for HHV-6 infection. PMID: 21849437
Database Links

KEGG: vg:1487928

Protein Families
Herpesviridae glycoprotein H family
Subcellular Location
Virion membrane; Single-pass type I membrane protein. Host cell membrane; Single-pass type I membrane protein. Host endosome membrane; Single-pass type I membrane protein.

Q&A

What is the basic structure of HHV-6A gH?

Recombinant Human herpesvirus 6A (strain Uganda-1102) envelope glycoprotein H is a mature protein spanning amino acids 17-694, with a UniProt ID of P68324 . The mature form of HHV-6 gH has a molecular mass of approximately 98-102 kDa after post-translational modifications . The protein contains 13 potential N-linked glycosylation sites, which contribute significantly to its structure and function . The amino acid sequence contains specific domains involved in protein-protein interactions, particularly regions that facilitate binding with glycoprotein L (gL) and other components of the entry complex .

What is the primary function of HHV-6A gH?

The heterodimer glycoprotein H-glycoprotein L (gH-gL) is required for the fusion of viral and plasma membranes leading to virus entry into the host cell . Following initial binding to host receptor, membrane fusion is mediated by the fusion machinery composed of glycoprotein B (gB) and the heterodimer gH/gL . Beyond viral entry, gH may also be involved in the fusion between the virion envelope and the outer nuclear membrane during virion morphogenesis, contributing to the viral lifecycle beyond the initial infection event . This dual role makes gH a particularly interesting target for understanding both viral entry and assembly mechanisms.

How does HHV-6A gH differ from other herpesvirus gH proteins?

HHV-6A gH forms part of a tetramer complex (gH/gL/gQ1/gQ2) that is characteristic of this virus family . While other herpesviruses also utilize gH-gL complexes for entry, the specific association with gQ1 and gQ2 proteins is unique to HHV-6A and HHV-6B and regulates their distinct receptor specificities . The HHV-6A gH shows structural similarities to other herpesvirus gH proteins but has evolved specific functional characteristics, particularly in terms of receptor recognition and cellular tropism targeting primarily T lymphocytes .

What expression systems are effective for producing recombinant HHV-6A gH?

Recombinant HHV-6A gH can be successfully produced using E. coli cell-free expression systems with either a His-tag or in tag-free formats . Alternatively, researchers have achieved expression using a transient in vivo vaccinia virus-T7 system that allows for proper post-translational modifications . For functional studies requiring proper folding and glycosylation, mammalian expression systems using COS-7 cells have been employed successfully . Each system offers different advantages: bacterial systems provide high yield but lack glycosylation, while mammalian systems offer proper processing but at lower yields.

What are the challenges in expressing functional HHV-6A gH?

A significant challenge in producing functional HHV-6A gH is achieving proper folding and glycosylation. When expressed alone, gH tends to aggregate and shows limited biological activity . Co-expression with gL is essential for proper processing, as both proteins form a stable complex that facilitates correct folding, trafficking through the endoplasmic reticulum and Golgi apparatus, and ultimately functional activity . Additionally, researchers must consider that in vitro transcription-translation systems yield products of 65K (gH) and 28K (gL), which can be processed by microsomes to 110K and 40K, respectively, mimicking the post-translational modifications observed in vivo .

How can the purity and quality of recombinant HHV-6A gH be assessed?

The purity of recombinant HHV-6A gH can be effectively determined using SDS-PAGE, with high-quality preparations typically showing >90% purity . Functional quality can be assessed through binding assays in a functional ELISA . For more rigorous analysis, researchers can employ immunoblotting with specific anti-gH antibodies such as mouse antiserum or monoclonal antibodies like 1D3 or 2E4 . When evaluating complex formation with gL, co-immunoprecipitation assays can confirm proper heterodimer assembly, which is crucial for functional studies .

How does HHV-6A gH interact with gL, and why is this interaction important?

HHV-6A gH forms a heterodimer with gL that is essential for proper protein processing and function . This interaction occurs in the endoplasmic reticulum and is a prerequisite for the proteins to migrate out of this compartment . The heterodimer formation is necessary for proper folding and trafficking through the cellular secretory pathway . Experimentally, this interaction can be demonstrated through co-immunoprecipitation assays where anti-gH antibodies can pull down both gH and gL when co-expressed . The biological significance of this interaction lies in its requirement for viral entry, as neither protein alone can mediate the membrane fusion needed for viral penetration into host cells .

What is the composition and function of the gH/gL/gQ1/gQ2 tetramer complex?

The HHV-6A envelope contains a distinctive tetramer complex composed of gH, gL, gQ1, and gQ2 . The gQ1/gQ2 complex forms a heterodimer that associates with the gH/gL heterodimer to create the complete tetramer . This tetramer is crucial for receptor recognition, particularly binding to the cellular receptor CD46 . The combination of gQ1 and gQ2 is critical for regulating the specificity of the tetramer's function for virus entry . Notably, the gH-gL complex alone is unable to bind to CD46, and the interaction with CD46 requires additional association with gQ-80K modified in HHV-6-infected cells .

How do the interactions between gH, gL, gQ1, and gQ2 differ between HHV-6A and HHV-6B?

While both HHV-6A and HHV-6B form gH/gL/gQ1/gQ2 tetramers, the specific combinations of gQ1 and gQ2 determine distinct receptor tropisms . HHV-6A tetramers preferentially bind to CD46, while HHV-6B tetramers recognize CD134 . Experimental evidence shows that switching gQ1 or gQ2 between HHV-6A and HHV-6B results in decreased cell fusion activity, highlighting the co-evolutionary specificity of these components . Furthermore, HHV-6B gQ2 cannot complement the function of HHV-6A gQ2 in HHV-6A propagation, further demonstrating the specific nature of these interactions . These differences explain the distinct cellular tropism and pathogenic properties of the two viral variants.

How does the HHV-6A gH complex recognize and bind to human CD46?

Human CD46 serves as the primary cellular receptor for HHV-6A, and this interaction is mediated through the gH/gL/gQ1/gQ2 tetramer complex . The binding occurs specifically between CD46 and the complete tetramer, as the gH-gL complex alone is insufficient for receptor recognition . This has been demonstrated through co-immunoprecipitation experiments where CD46(309his) associates with HHV-6A gH in the context of the complete viral glycoprotein complex . The interaction appears to require gQ components in their mature, properly glycosylated forms, specifically the 80-kDa U100 gene product that contains complex N-linked oligosaccharides .

What is the mechanism of membrane fusion mediated by HHV-6A gH?

Following receptor binding, membrane fusion is orchestrated by a complex machinery involving both the gH/gL/gQ1/gQ2 tetramer and glycoprotein B (gB) . The current model suggests that receptor binding triggers conformational changes in the tetramer, which then activates gB—the primary fusion protein . This process can be studied using cell-cell fusion assays that monitor the ability of viral glycoproteins to induce syncytia formation between cells expressing viral glycoproteins and those expressing the appropriate receptor . Specific regions of the gH protein contain fusion-related domains that undergo structural rearrangements during this process, exposing hydrophobic residues that interact with and destabilize the target cell membrane .

How can researchers experimentally investigate gH-mediated viral entry?

Researchers can employ several methodologies to study gH-mediated viral entry. Cell-cell fusion assays provide a direct measure of the fusion activity of viral glycoproteins and can be used to assess the contributions of individual components of the entry machinery . Co-immunoprecipitation studies allow for the investigation of protein-protein interactions, such as the association between gH and CD46 . Another approach involves expressing tagged versions of gH, such as those with the nine-amino acid epitope for monoclonal antibody LP14, which allows for tracking of the protein in various experimental settings . Additionally, recombinant protein binding assays using purified gH/gL or the complete tetramer can directly measure receptor binding kinetics and affinities .

What are the optimal conditions for storing and handling recombinant HHV-6A gH?

Recombinant HHV-6A gH is typically supplied as a lyophilized powder for optimal stability . Upon reconstitution, the protein should be stored at -80°C for long-term preservation or at -20°C for shorter periods. Working solutions should be maintained at 4°C and used within 24 hours to prevent degradation. The addition of stabilizing agents such as low concentrations of glycerol (10-15%) can improve stability . For functional assays, care should be taken to avoid repeated freeze-thaw cycles, which can lead to protein denaturation and loss of activity. Additionally, when working with the complete gH/gL/gQ1/gQ2 complex, consideration must be given to maintaining the integrity of all component interactions .

How can researchers effectively analyze gH interactions with other viral and cellular proteins?

Several methodological approaches are effective for studying gH interactions. Co-immunoprecipitation assays using specific antibodies against gH (such as 1D3 or 2E4), followed by Western blotting for interacting proteins, can identify protein complexes . For cellular localization studies, immunofluorescence microscopy has revealed that gH and gL localize to Golgi-like bodies in fibroblasts but distribute throughout the endoplasmic reticulum in T lymphocytes . Protein tagging strategies, such as His-tagging or epitope tagging, facilitate purification and detection in complex biological samples . Advanced techniques like surface plasmon resonance or biolayer interferometry can provide quantitative binding kinetics for gH interactions with receptors or other viral glycoproteins .

What cell culture systems are most appropriate for studying HHV-6A gH function?

The choice of cell culture system depends on the specific research questions being addressed. For viral propagation studies, T lymphocytes (such as JJhan cells) represent the natural target for HHV-6A and thus provide the most physiologically relevant system . For recombinant protein expression, COS-7 cells infected with recombinant vaccinia virus vTF7 have been successfully used to express functional gH and gL . When studying receptor interactions, cells expressing CD46 (such as human epithelial cells) are appropriate for binding assays . For cell-cell fusion assays, combinations of effector cells expressing viral glycoproteins and target cells expressing appropriate receptors can effectively measure fusion activity . Researchers should consider that the cellular localization of glycoproteins may differ between cell types, as observed with the differential localization of gH/gL in fibroblasts versus T lymphocytes .

How can structural biology approaches enhance our understanding of HHV-6A gH?

Advanced structural biology techniques offer significant potential for elucidating the molecular mechanisms of HHV-6A gH function. X-ray crystallography of the gH/gL complex or the complete tetramer would provide atomic-level details of protein-protein interfaces and conformational states . Cryo-electron microscopy (cryo-EM) is particularly suitable for studying the larger gH/gL/gQ1/gQ2 complex and could reveal how these components assemble and interact with receptors . Hydrogen-deuterium exchange mass spectrometry (HDX-MS) could identify flexible regions and conformational changes associated with receptor binding or fusion activation . Computational approaches such as molecular dynamics simulations based on homology models can predict structural transitions during the fusion process, generating testable hypotheses about functional domains .

What are the challenges and strategies for developing inhibitors targeting HHV-6A gH?

Developing inhibitors targeting HHV-6A gH presents several challenges and opportunities. The complex nature of the gH/gL/gQ1/gQ2 tetramer means that multiple interfaces could be targeted, including gH-gL, gH-gQ, or gH-receptor interactions . High-throughput screening assays based on protein-protein interaction disruption or cell fusion inhibition can identify lead compounds . Peptide inhibitors derived from interface regions between gH and its binding partners have shown promise in other herpesvirus systems and could be applied to HHV-6A . Structure-based drug design approaches would benefit significantly from solving the crystal structure of gH complexes . A key challenge is developing specificity for HHV-6A gH versus other herpesvirus gH proteins or host proteins . Combination approaches targeting multiple components of the entry machinery (gH, gL, gQ1, gQ2, and gB) might offer synergistic effects and reduce the emergence of resistance .

How might genetic variation in HHV-6A gH affect viral pathogenesis and immune evasion?

Genetic variation in HHV-6A gH could significantly impact viral fitness, tissue tropism, and immune recognition. Comparative sequence analysis of gH across clinical isolates could identify conserved regions essential for function versus variable regions potentially involved in immune evasion . Specific amino acid substitutions might alter binding affinity to CD46 or modify interactions with other components of the entry complex . Experimental approaches could include site-directed mutagenesis of key residues followed by functional assays for receptor binding, complex formation, and fusion activity . Structure-function correlations would be particularly valuable, especially if mapped onto three-dimensional models of the protein . Additionally, variations in glycosylation sites could affect protein folding, complex assembly, and recognition by neutralizing antibodies, as glycans often shield important epitopes from immune surveillance . Understanding these variations has implications for diagnostics, vaccine development, and antiviral strategies targeting conserved functional domains .

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.