首页 | 本学科首页   官方微博 | 高级检索  
   检索      


Herpes Simplex Virus Glycoprotein B Associates with Target Membranes via Its Fusion Loops
Authors:Brian P Hannah  Tina M Cairns  Florent C Bender  J Charles Whitbeck  Huan Lou  Roselyn J Eisenberg  Gary H Cohen
Institution:Department of Microbiology, School of Dental Medicine,1. Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 191042.
Abstract:Herpes simplex virus (HSV) glycoproteins gB, gD, and gH/gL are necessary and sufficient for virus entry into cells. Structural features of gB are similar to those of vesicular stomatitis virus G and baculovirus gp64, and together they define the new class III group of fusion proteins. Previously, we used mutagenesis to show that three hydrophobic residues (W174, Y179, and A261) within the putative gB fusion loops are integral to gB function. Here we expanded our analysis, using site-directed mutagenesis of each residue in both gB fusion loops. Mutation of most of the nonpolar or hydrophobic amino acids (W174, F175, G176, Y179, and A261) had severe effects on gB function in cell-cell fusion and null virus complementation assays. Of the six charged amino acids, mutation of H263 or R264 also negatively affected gB function. To further analyze the mutants, we cloned the ectodomains of the W174R, Y179S, H263A, and R264A mutants into a baculovirus expression system and compared them with the wild-type (WT) form, gB730t. As shown previously, gB730t blocks virus entry into cells, suggesting that gB730t competes with virion gB for a cell receptor. All four mutant proteins retained this function, implying that fusion loop activity is separate from gB-receptor binding. However, unlike WT gB730t, the mutant proteins displayed reduced binding to cells and were either impaired or unable to bind naked, cholesterol-enriched liposomes, suggesting that it may be gB-lipid binding that is disrupted by the mutations. Furthermore, monoclonal antibodies with epitopes proximal to the fusion loops abrogated gB-liposome binding. Taken together, our data suggest that gB associates with lipid membranes via a fusion domain of key hydrophobic and hydrophilic residues and that this domain associates with lipid membranes during fusion.Herpes simplex virus (HSV) entry into cells requires four viral envelope glycoproteins (gB, gD, and the heterodimer gH/gL) as well as a cell surface gD receptor (reviewed in references 31, 42, 43, and 49). When gD binds its receptor, it undergoes conformational changes that are essential to activate the fusion machinery, gB and gH/gL. In addition to being essential for virus entry, both gH/gL and gB play important roles in primary fusion events that occur during egress of the capsid from the nuclei of infected cells (22). gB and gH/gL constitute the core fusion machinery of all members of the Herpesviridae.The mechanisms by which gB and gH/gL function individually and in concert during fusion are topics of intense investigations. Peptides based on predicted heptad repeats in gH block virus entry and have the ability to bind and disrupt model membranes (24, 26, 27). In addition, gH/gL can achieve hemifusion of adjacent cells in the absence of other herpesvirus proteins (50). These studies imply that gH/gL has fusogenic properties. Previously, we showed that both virion gB and soluble wild-type (WT) gB (gB730t), but not gD or gH/gL, bind to cells and associate with lipid rafts (10). Like gH/gL, several synthetic gB peptides induced the fusion of large unilamellar vesicles and inhibited herpesvirus infection (23, 24). Thus, it appears that both gB and gH/gL may be fusion proteins, a theory strengthened by data showing that either gB or gH/gL is sufficient for membrane fusion during nuclear egress (22). Additionally, gB730t blocks virus entry into cells deficient in heparan sulfate proteoglycans (HSPGs), suggesting that it competes with virion gB for an obligate cell surface receptor (9). A recent study suggested that paired immunoglobulin-like type 2 receptor alpha (PILRα) may serve this role for at least some cell types (47).The crystal structure of gB is now known for both HSV type 1 (HSV-1) (32) and Epstein-Barr virus (EBV) (6). Interestingly, gB is structurally related to two other viral fusion proteins, the vesicular stomatitis virus (VSV) G protein (45) and the baculovirus gp64 protein (34). VSV G, gB, and most recently, baculovirus gp64 were placed into a newly formed group of fusion proteins, the class III proteins. Class III fusion proteins have similar individual domain structures and contain a central three-stranded coiled coil reminiscent of the class I proteins. Whereas class I proteins have an N-terminal fusion peptide, class III proteins have internal bipartite fusion loops within domain I (shown in Fig. Fig.1A1A for gB) which are similar to the single fusion loop of class II fusion proteins. However, the class II fusion loop is composed entirely of hydrophobic amino acids, whereas the fusion loops of gB have both hydrophobic and charged residues (32, 34, 45). Unlike G or gp64, which are the sole fusion proteins for their respective viruses, gB requires gH/gL to function in fusion and entry.Open in a separate windowFIG. 1.HSV gB hydrophobic ridge is surrounded by charged residues on the surface of the molecule. A ribbon diagram of the HSV protomer (A) and molecular surface representation of the trimer (B) are shown. In each, one protomer is colored by secondary structure succession, using blue (domain I), green (domain II), yellow (domain III), orange (domain IV), and red (domain V). The box in panel A shows the primary amino acid sequences of the fusion loops. The box in panel B shows the base of the gB trimer, rotated 90°. For the boxes in both panels A and B, highlighted hydrophobic residues are colored in blue and charged residues are shown in red. All structural figures were generated, in part, using PyMOL Molecular Graphics System software.In our previous study, we used site-directed mutagenesis to show that three hydrophobic amino acids within the gB loops (W174, Y179, and A261) are essential for gB function (29). Similar studies of VSV G, gp64, and EBV gB support the notion that hydrophobic amino acids of both fusion loops are critical for fusion (34, 44, 51) and together constitute a fusion domain. Recently, bimolecular complementation was used to show that gB and gH/gL interact with each other concomitantly with fusion and that this interaction is triggered by binding of gD to its cellular receptor (3, 4). Thus, gB may function cooperatively with gH/gL, yet each may have some fusogenic potential on its own.The goal of the experiments reported here was twofold. First, we wanted to complete our mutagenic analysis of all of the residues in the two putative fusion loops of HSV gB. Our data show that the two fusion loops constitute a structural “subdomain” wherein key hydrophobic amino acids form a ridge that is supported on both sides by charged residues. We hypothesize that two charged residues on one side of the ridge enhance the ability of the hydrophobic residues to interact with target membranes and to function in fusion.Our second goal was to assess the effects of mutations in the fusion loops on the function of gB in cell binding, blocking of entry, and insertion into lipid membranes. Therefore, we constructed recombinant baculoviruses, with each carrying the gene for a truncated version (residues 31 to 730) of one of four mutant forms of gB (W174R, Y179S, H263A, and R264A). We found that the mutant proteins were able to efficiently block virus entry, suggesting that the fusion loops do not participate in protein-receptor binding. However, all four mutant proteins were impaired in cell binding compared to WT gB730t. Whereas WT gB730t associated with liposomes in a flotation assay, soluble truncated forms of HSV gD and gH/gL did not, consistent with our previous finding that gB730t associates with lipid rafts on cell surfaces (8). In contrast to WT gB730t, the gB mutant proteins were either impaired or unable to bind liposomes. Our data suggest that gB has an intrinsic ability to associate with a target membrane via its fusion domain.
Keywords:
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号