首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
We have examined mutations in the ectodomain of the human immunodeficiency virus type 1 transmembrane glycoprotein gp41 within a region immediately adjacent to the membrane-spanning domain for their effect on the outcome of the fusion cascade. Using the recently developed three-color assay (I. Muñoz-Barroso, S. Durell, K. Sakaguchi, E. Appella, and R. Blumenthal, J. Cell Biol. 140:315–323, 1998), we have assessed the ability of the mutant gp41s to transfer lipid and small solutes from susceptible target cells to the gp120-gp41-expressing cells. The results were compared with the syncytium-inducing capabilities of these gp41 mutants. Two mutant proteins were incapable of mediating both dye transfer and syncytium formation. Two mutant proteins mediated dye transfer but were less effective at inducing syncytium formation than was wild-type gp41. The most interesting mutant proteins were those that were not capable of inducing syncytium formation but still mediated dye transfer, indicating that the fusion cascade was blocked beyond the stage of small fusion pore formation. Fusion mediated by the mutant gp41s was inhibited by the peptides DP178 and C34.

The human immunodeficiency virus type 1 (HIV-1) gp120-gp41 fusion machine consists of an assembly of viral envelope glycoprotein oligomers which forms a molecular scaffold responsible for bringing the viral membrane close to the target cell membrane and creating the architecture that enables lipid bilayers to merge (7). The fusion reaction undergoes multiple steps before the final event occurs which allows delivery of the nucleocapsid into the cell. In the case of influenza virus hemagglutinin (HA), we have dissected these steps kinetically and analyzed the molecular features of the kinetic intermediates (1). In order to examine the modus operandi of the fusion machine, mutations in various domains of viral envelope glycoproteins have been examined for their effect on the outcome of the fusion cascade. For instance, replacement of the membrane-spanning domain of influenza virus HA with a glycosylphosphatidylinositol anchor results in a very stable hemifusion intermediate (6). Moreover, single-site mutations in the fusion peptide of HA significantly affect fusion pore dilation (9). Recently, cytoplasmic tail acylation mutants of influenza virus HA were identified which induce transfer of lipids and small aqueous molecules but do not induce syncytium formation (4a).High-resolution crystallographic determinations (4, 10, 11) of gp41 fragments from HIV-1 have revealed a bent-in-half, antiparallel, heterotrimeric coiled-coil structure. This is made up of a triple-stranded coiled coil of α-helices from the leucine zipper-like 4-3 repeat domain in gp41 close to the N-terminal fusion peptide termed HR1 (8) flanked by α-helices from the domain in gp41 close to the C-terminal membrane anchor termed HR2 (8). Comparison with the crystal structure of the influenza virus HA2 subunits in a low-pH-induced conformation (2) reveals common structural motifs which provide growing support for the “spring-loaded” type of mechanistic models (3). In this scenario, activation of the fusion protein results in release of the fusion peptide and extension of the central coiled-coil structure. The new positioning of the fusion peptides at the tip of the stalk provides for easy contact with the target cell membrane. A small group of proximal fusion proteins which are simultaneously inserted into both the viral and target membranes would constitute a potential fusion site. A concerted collapse of this protein complex, actuated by the bending in half of the stalks at a central hinge region, would presumably position the C-terminal transmembrane anchors and N-terminal fusion peptides on top of each other in the center, bring the two membranes into contact, and thus allow formation of the fusion pore (7). In this study, we examined the effects on the various stages of the fusion reaction of mutations in the region between HR2 and the transmembrane (TM) anchor (Fig. (Fig.1)1) described in detail by Salzwedel et al. (8a). Open in a separate windowFIG. 1Amino acid sequence and mutations in gp41. FP is the predicted fusion peptide region, and HR1 and HR2 (8) represent, respectively, the N-terminal and C-terminal α-helices of the triple-stranded coiled coil (4, 11). Mutations in the region between HR2 and the TM anchor include deletions of amino acids 665 to 682 and 678 to 682, insertion of a FLAG sequence (YKDDDD), insertion of a DAF sequence (PNKGSGTTS), scrambling of the underlined sequence to SC7 (INNWNFT), and replacement of the five tryptophans with alanines [W(1-5)A]. Peptide C34 represents HR2 amino acids 628 to 663, and peptide DP178 represents amino acids 638 to 673.Mutagenesis of HIV-1 env, construction of plasmids, cell surface expression, CD4 binding, and cell fusion were performed as previously described (8a). The simian virus 40-based env expression plasmids (1 μg of DNA) were transfected into COS-1 cells in 35-mm-diameter plates by using DEAE-dextran (1 mg/ml). At 14 h posttransfection, the cells were replated, and starting at 36 to 48 h posttransfection, they were incubated with 20 μM CMAC (7-amino-4-chloromethylcoumarin) in Dulbecco modified Eagle medium overnight at 37°C. All constructs expressed similar amounts of envelope glycoprotein on the cell surface (8a). The transfected cells were then washed and incubated in fresh medium for 2 h at 37°C before addition of HeLa-CD4 cells which were labeled in the membrane with octadecyl indocarbocyanine (DiI) and in the cytosol with calcein as previously described (7). The method used to detect cell-cell fusion was a three-color assay (7) based on the redistribution of fluorescent probes between effector and target cells upon fusion. The application of three different probes was used to monitor lipid versus cytosolic mixing in the same cell population. Fluorescently labeled gp120-gp41-expressing cells and CD4+ cells were cocultured at a 1:10 ratio for 2 h at 37°C in uncoated microwells (MatTek Corp., Ashland, Mass.). Bright-field and fluorescent images were acquired with an Olympus IX70 microscope coupled to a charge-coupled device camera (Princeton Instruments, Trenton, N.J.) with a 40× UplanApo oil immersion objective. Fluorescein isothiocyanate (exciter, BP470-490; beam splitter, DM505; emitter, BA515-550), rhodamine (exciter, BP530-550; beam splitter, DM570; emitter, BA590), and 4′,6-diamidino-2-phenylindole (DAPI) (exciter, D360/40; beam splitter, 400DCLP; emitter, D450/60) optical filter cubes were carefully chosen to avoid spillover when observing the fluorescence of the three dyes. For each sample, three or four different fields were collected, and data were analyzed by overlaying the images using Metamorph software (Universal Imaging Corporation, West Chester, Pa.). The percentage of lipid mixing and cytoplasmic mixing was calculated as 100 times the number of COS-1 cells stained with DiI and calcein divided by the total number of COS-1–HeLa-CD4 conjugates. Although not all COS-1 cells express env since the transfection efficiency is not 100%, env-expressing COS-1 cells are more likely to adhere to HeLa-CD4 cells.Figure Figure22 shows a montage of video images taken 2 h following incubation of COS-1 cells expressing wild-type (WT), W(1-5)A, and +DAF env with HeLa-CD4 cells at 37°C. As described in detail in the legend to Fig. Fig.2,2, we clearly observed COS-1 cells attached to HeLa-CD4 cells, which showed continuity of all three dyes (CMAC, calcein, and DiI). We know that for +DAF and W(1-5)A env-expressing COS-1 cells, these images do not represent syncytia since even small heterokaryons will show up in the MAGI cell assay (6a), which is based on the transfer of HIV-1 Tat coexpressed with env in COS-1 cells to HeLa-CD4 cells as a result of cell fusion. This transfer induces the expression of a β-galactosidase reporter gene engineered in HeLa-CD4 (MAGI) cells under the control of the viral long terminal repeat promoter (8a). Because the β-galactosidase has been modified to contain a nuclear targeting signal, the nuclei of the resulting heterokaryons stain dark blue with 5-bromo-4-chloro-3-indolyl-β-d-galactopyranoside (X-Gal) in situ. The MAGI assay is extremely sensitive and clearly identifies syncytia as small as two nuclei. Such nuclei were common for the Δ678-682 mutant, which produced syncytia with an average size of ∼5 nuclei (Fig. (Fig.3).3). The assay can detect even a few of these fusion events per 100,000 cells. In the MAGI assay, we did not observe any blue nuclei with the W(1-5)A and +DAF constructs, an experiment repeated several times. The three-color assay therefore reveals a distinct phenotype exhibited by the +DAF and W(1-5)A mutant envelope glycoproteins, which form small fusion pores allowing movement of lipids and small molecules (<1,000 Da) but not of large molecules (HIV-1 Tat is about 14 kDa [4b]). Open in a separate windowFIG. 2Three-color assay for WT and mutant HIV-1 gp41s. Simian virus 40-based env expression plasmids (1 μg) containing WT (A to D), W(1-5)A (E to H), and +DAF (I to L) env genes were transfected into COS-1 cells in 35-mm plates using DEAE-dextran (1 μg/ml). At 14 h posttransfection, the cells were replated, and starting at 36 to 48 h posttransfection they were incubated with 20 μM CMAC in Dulbecco modified Eagle medium overnight at 37°C. All constructs expressed similar amounts of envelope glycoprotein on the cell surface (8a). The transfected cells were then washed and incubated in fresh medium for 2 h at 37°C before addition of HeLa-CD4 cells which were labeled in the membrane with DiI and in the cytosol with calcein as previously described (7). The COS-1 cells, labeled with CMAC, were cocultured 1:10 at 37°C for 2 h with HeLa-CD4 cells labeled with DiI and calcein, and images were examined by bright-field microscopy (A, E, and I) and fluorescence microscopy for CMAC staining (B, F, and J), for DiI staining (C, G, and K), and for calcein staining (D, H, and L). CMAC is a fluorescent chloromethyl derivative that freely diffuses through the membranes of live cells. Once inside the cell, this mildly thiol-reactive probe undergoes what is believed to be a glutathione S-transferase-mediated reaction to produce membrane-impermeant fluorescent dye adducts with glutathione, as well as with other intracellular components. Staining of COS-1 cells with CMAC gives rise to bright fluorescence due to reaction with proteins in the perinuclear, endoplasmic reticulum, and Golgi regions, which are immobile, as well as to weaker fluorescence due to the fluorescent glutathione adduct (molecular mass, ∼600 Da) in the cytosol, which is able to diffuse through small fusion pores. The COS-1 cells identified by CMAC staining (B, F, and J) are large and often appear multinuclear, although we do not know whether the round granular structures seen by bright-field microscopy of the COS-1 cells are nuclei or large granules. Panels A to D show one large cell triple stained with CMAC, DiI, and calcein (indicated by a star). DiI is internalized after 2 h at 37°C and appears punctate with nuclear sparing due to its localization in membranes of intracellular organelles. Calcein (465 Da) is evenly distributed throughout the cell (D). One large, granular COS-1 cell (A, left) is only stained with CMAC (B); its lack of staining with DiI (C) and calcein (D) indicates that it has not fused with HeLa-CD4 cells. In panel F, a large structure is seen which seems in continuity with CMAC. However, since the bottom left part of this structure is not in continuity with DiI (G) and calcein (H), it represent two cells. The top right cell (indicated by a star) is in continuity with CMAC, DiI, and calcein. Since COS-1 cells expressing W(1-5)A env do not produce blue nuclei when incubated with MAGI cells (see Fig. Fig.3),3), which requires transfer of the 14-kDa HIV-1 Tat protein (see text), we conclude that this COS-1–HeLa-CD4 conjugate represents a phenotype in which small fusion pores form, allowing movement of lipids and small molecules (<1,000 Da) but not of large molecules. The same phenotype is seen with COS-1 cells expressing DAF env: the COS-1–HeLa-CD4 conjugate indicated by a star in panels J, K, and L is in continuity with CMAC, DiI, and calcein but does not allow transfer of HIV-1 Tat (see Fig. Fig.33).Open in a separate windowFIG. 3Fusogenic activity of WT and mutant HIV-1 gp41s. The three-color assay was performed as described in the legend to Fig. Fig.2.2. Since multiple rounds of fusion may interfere with quantitation in the case of WT and mutant env genes which produce a large number of blue nuclei after 24 h at 37°C (grey bars), incubations were done for 2 h at 37°C. Black bars represent 100 times the number of COS-1 cells stained with DiI and calcein over the total number of COS-1–HeLa-CD4 conjugates measured in the three-color assay. The data are representative of five separate experiments. In each experiment, a total of 30 to 50 COS-1–HeLa-CD4 conjugates were counted. The number of nuclei per syncytium (grey bars) was obtained from the MAGI assay (8a) and represents the ability of HIV-1 Tat to transfer from COS-1 cells to HeLa-CD4 cells.We tallied data from many cell pairs similar to those shown in Fig. Fig.22 and plotted the average percentage of COS-1 cells stained with DiI and calcein. Figure Figure33 shows the data for the WT and a number of mutants described by Salzwedel et al. (8a). The data fall into three groups, in which the envelope glycoproteins mediate (i) both dye and HIV-1 Tat redistribution (WT, Δ678-682, and SC7), (ii) neither dye nor HIV-1 Tat redistribution (Δ665-682 and +FLAG), or (iii) dye but not HIV-1 Tat redistribution [W(1-5)A and +DAF]. The latter represents a nonexpanding fusion pore phenotype.Dye redistribution induced by WT and mutant gp41s was inhibited by the peptide inhibitors DP178 and C34 (Fig. (Fig.4).4). The latter peptide is from the HR2 sequence (residues 628 to 663) which forms the flanking peptide of the heterotrimeric coiled coil in the crystal structure. DP178 is frameshifted 10 amino acids toward the C terminus (residues 638 to 673). The inhibition data indicate that dye redistribution mediated by WT and mutant gp41 molecules is specific for the gp120-gp41-induced fusion reaction and not due to nonspecific transfer. Interestingly, W(1-5)A and SC7 exhibited greater sensitivity than the WT to DP178 inhibition. In the case of C34, inhibition was about the same for the WT and the two mutants. We observed no inhibition by DP178 or C34 of HIV-2 env-mediated fusion at up to 100 nM peptide (data not shown). Open in a separate windowFIG. 4Inhibition of cell-cell fusion by DP178 and C34 peptides. Cell fusion was calculated as a percentage of the control by using the three-color assay method shown in Fig. Fig.22 and and33 and described in the text for the WT, W(1-5)A, and SC7.Although the crystal structure of the gp41 core (4, 11) is based on the HR1-HR2 coiled coil, it is possible that in intact gp41 the bundle is extended to include amino acids downstream from HR2 and upstream from HR1. Extension of the coiled coil might lead to tilting of the TM anchor, which is presumably important for producing sufficient lipid curvature to form a fusion junction (1). Removal of amino acids 665 to 682 may leave no possibility to form this extended coiled coil. Similarly, insertion of the FLAG sequence, which contains four aspartic acid residues, would presumably insert charged residues into a hydrophobic domain, which could also prevent extension of the coiled coil. The other mutations presumably allow extended coiled-coil formation but reduce its efficiency because of weaker interactions between the amino acids in the extended region. The coiled-coil structure might be so frail in mutant gp41s W(1-5)A and +DAF that it is not present for a sufficient amount of time to create the fusion pore dilation necessary to allow transfer of HIV-1 Tat. Since the Δ678-682 and SC7 proteins are, to a limited extent, capable of inducing syncytium formation and dye transfer, we surmise that they possess intermediate extended coiled-coil-forming propensities.Based on the structural information about the gp41 core (4, 10, 11), it has been proposed that the binding site for the peptide inhibitors is in the HR1 bundle. The C34 and DP178 peptides presumably bind in the same way as the corresponding amino acid sequence regions of the three HR2 helices in the crystal structures. At this position, the peptides would sterically block the regular binding of the HR2 helices to the inner core of HR1 helices and thus prevent formation of the bent-in-half, antiparallel, heterotrimeric coiled-coil structure presumably required to bring the viral and target cell membranes into contact for fusion. Since C34 corresponds to HR2 with no amino acids in the extended region, we do not expect any enhanced inhibitory effect on fusion mediated by the mutant gp41s. Figure Figure4b4b shows that this is the case. Since DP178 does contain 10 amino acids downstream from HR2 whose interaction with amino acids upstream from HR1 is weaker in the mutants, we expect greater sensitivity to DP178 inhibition in the mutant proteins. This does seem to be the case, as shown in Fig. Fig.44a.The recent high-resolution X-ray crystallographic determination of the structure of the gp41 core from HIV-1 provides well-defined landmarks in the terrain the viral envelope glycoproteins navigate following CD4 and coreceptor-induced conformational changes (5). The structures include neither fusion peptides and TM anchors nor regions between those domains and HR1 and HR2, respectively, which are crucial for fusion activity. Therefore, mutagenesis of those undetermined domains combined with sensitive assays for the activity of the modified proteins will lead to refinement of our thinking about the HIV-1 gp120-gp41 fusion machine.  相似文献   

2.
The entry of human immunodeficiency virus type 1 (HIV-1) into a target cell entails a series of conformational changes in the gp41 transmembrane glycoprotein that mediates the fusion of the viral and target cell membranes. A trimer-of-hairpins structure formed by the association of two heptad repeat (HR) regions of the gp41 ectodomain has been implicated in a late step of the fusion pathway. Earlier native and intermediate states of the protein are postulated to mediate the antiviral activity of the fusion inhibitor enfuvirtide and of broadly neutralizing monoclonal antibodies (NAbs), but the details of these structures remain unknown. Here, we report the identification and crystal structure of a dimerization domain in the C-terminal ectodomain of gp41 (residues 630 to 683, or C54). Two C54 monomers associate to form an asymmetric, antiparallel coiled coil with two distinct C-terminal α-helical overhangs. This dimer structure is conferred largely by interactions within a central core that corresponds to the sequence of enfuvirtide. The mutagenic alteration of the dimer interface severely impairs the infectivity of Env-pseudotyped viruses. Moreover, the C54 structure binds tightly to both the 2F5 and 4E10 NAbs and likely represents a potential intermediate conformation of gp41. These results should enhance our understanding of the molecular basis of the gp41 fusogenic structural transitions and thereby guide rational, structure-based efforts to design new fusion inhibitors and vaccine candidates intended to induce broadly neutralizing antibodies.The entry of human immunodeficiency virus type 1 (HIV-1) into its target cell to establish an infection requires the fusion of viral and cellular membranes, a process that is mediated by the viral envelope glycoprotein (Env) through interactions with receptors on the target cell membrane (CD4 and a coreceptor, such as CCR-5 or CXCR-4) (14). HIV-1 Env is synthesized as the glycoprotein precursor gp160, which oligomerizes in the endoplasmic reticulum and subsequently is cleaved by the cellular furin endoprotease to create a metastable state that is primed for the induction of membrane fusion activity (19). The resulting Env complex is a trimeric structure comprising three gp120 surface glycoproteins, each associated noncovalently with one of three subunits of the gp41 transmembrane glycoprotein (24, 27, 47, 48). This native (prefusion) Env spike protrudes from the virus surface and is the target for neutralizing antibodies (NAbs) (reviewed in reference 3). It is generally accepted that HIV-1 membrane fusion is promoted by a series of receptor binding-triggered conformational changes in the Env complex, culminating in the formation of an energetically stable trimer of α-helical hairpins in gp41 (10, 14).The core structure of the trimer-of-hairpins is an antiparallel six-helix bundle: a central, three-stranded coiled coil formed by the first heptad repeat (HRN) region of gp41 is sheathed by three α-helices derived from the second HR (HRC) (5, 27, 42, 44). HRN is immediately C terminal to the fusion peptide, while HRC is adjacent to the transmembrane helix anchored in the viral membrane. The interaction of gp120 with CD4 and a chemokine receptor is thought to alter intersubunit interactions in the native Env complex, leading to gp41 reorganization into a postulated prehairpin intermediate (reviewed in references 10 and 14). At this point, the N-terminal HRN coiled-coil trimer is formed, relocating the fusion peptides to allow them to insert into the cellular membrane. The HRC region then is thought to jackknife so as to pack against the inner coiled-coil core and form the postfusion trimer-of-hairpin structure that brings the attached target cell and viral membranes together. Evidence for the existence of these different gp41 conformational states in the fusion pathway is indirect, being inferred from the antiviral activity of peptides derived from the two HR regions of gp41 (20, 45). These peptide inhibitors likely act in a dominant-negative manner by binding to the prehairpin intermediate, preventing the formation of the trimer-of-hairpins (6, 13, 27, 31). This intermediate is relatively stable, with a half-life of many minutes, as detected by the capacity of such peptides to inhibit fusion once prefusion gp41 has undergone a conformational transition (21, 31). Although mounting evidence indicates that the prefusogenic and intermediate states are important targets for drug- and vaccine-elicited NAbs (reviewed in references 3 and 10), little is known about their structures and how they modulate gp41 fusogenicity or serve as targets for inhibition.The C-terminal part of the gp41 ectodomain consists of HRC (or C34) and the membrane-proximal external region (MPER) (Fig. (Fig.1).1). The C34 peptide is intrinsically disordered in isolation and forms an outer-layer α-helix only in the six-helix bundle (27, 29). Structural studies of the trimeric coiled-coil state of the MPER and of its bent helix conformation after binding to lipid membranes have begun to provide clues regarding the function of this unusual and important NAb-associated segment (25, 41). The MPER is the established target for two very rare but broadly reactive NAbs, 2F5 and 4E10/z13, which are elicited during natural human infection (50). These neutralizing epitopes seem to be poorly exposed on the surface of both HIV-1-infected cells and virions (reviewed in reference 3). Their exposure is enhanced or triggered by receptor binding but diminishes on the formation of the trimer-of-hairpins, suggesting that both of the NAbs target a more extended intermediate conformation rather than the native gp41 structure (8, 12). Despite extensive efforts, how structural aspects of the MPER explain its antigenicity and immunogenicity remains unclear. Here, we report the identification of the C-terminal dimerization domain of gp41 and present the 1.65-Å crystal structure of this domain. We characterize the role of this antiparallel two-stranded coiled-coil structure in NAb reactivity and viral function. Our study provides a potential structure for the fusion-intermediate state of gp41 and for the future design of new HIV-1 immunogens that may elicit broad and potent NAbs.Open in a separate windowFIG. 1.Structural and functional domains of HIV-1 gp41. (Upper) Schematic view of gp41 showing the location of the fusion peptide (FP), the two HR regions, the MPER, the transmembrane segment (TM), and the cytoplasmic region (CP). HRC and MPER are depicted in blue and green, respectively. (Lower) Sequences of the C56, C54, C54N656L, and C39 peptides employed in the study. The Asn-656→Leu mutation in C54N656L is shown in red. The sequences of T-20 and core epitopes recognized by the human 2F5 and 4E10 MAbs are indicated.  相似文献   

3.
The membrane-anchored proteins of enveloped viruses form labile spikes on the virion surface, primed to undergo large-scale conformational changes culminating in virus-cell membrane fusion and viral entry. The prefusion form of these envelope glycoproteins thus represents an important molecular target for antiviral intervention. A critical roadblock to this endeavor has been our inability to produce the prefusion envelope glycoprotein trimer for biochemical and structural analysis. Through our studies of the GPC envelope glycoprotein of the hemorrhagic fever arenaviruses, we have shown that GPC is unique among class I viral fusion proteins in that the mature complex retains a stable signal peptide (SSP) in addition to the conventional receptor-binding and transmembrane fusion subunits. In this report we show that the recombinant GPC precursor can be produced as a discrete native-like trimer and that its proteolytic cleavage generates the mature glycoprotein. Proteoliposomes containing the cleaved GPC mediate pH-dependent membrane fusion, a characteristic feature of arenavirus entry. This reaction is inhibited by arenavirus-specific monoclonal antibodies and small-molecule fusion inhibitors. The in vitro reconstitution of GPC-mediated membrane-fusion activity offers unprecedented opportunities for biochemical and structural studies of arenavirus entry and its inhibition. To our knowledge, this report is the first to demonstrate functional reconstitution of membrane fusion by a viral envelope glycoprotein.  相似文献   

4.
5.
6.
The membrane-spanning domain (MSD) of the envelope (Env) glycoprotein from human (HIV) and simian immunodeficiency viruses plays a key role in anchoring the Env complex into the viral membrane but also contributes to its biological function in fusion and virus entry. In HIV type 1 (HIV-1), it has been predicted to span 27 amino acids, from lysine residue 681 to arginine 707, and encompasses an internal arginine at residue 694. By examining a series of C-terminal-truncation mutants of the HIV-1 gp41 glycoprotein that substituted termination codons for amino acids 682 to 708, we show that this entire region is required for efficient viral infection of target cells. Truncation to the arginine at residue 694 resulted in an Env complex that was secreted from the cells. In contrast, a region from residues 681 to 698, which contains highly conserved hydrophobic residues and glycine motifs and extends 4 amino acids beyond 694R, can effectively anchor the protein in the membrane, allow efficient transport to the plasma membrane, and mediate wild-type levels of cell-cell fusion. However, these fusogenic truncated Env mutants are inefficiently incorporated into budding virions. Based on the analysis of these mutants, a “snorkeling” model, in which the flanking charged amino acid residues at 681 and 694 are buried in the lipid while their side chains interact with polar head groups, is proposed for the HIV-1 MSD.Human immunodeficiency virus type 1 (HIV-1) infection is initiated by fusion of the viral membrane with that of the target cell and is mediated by the viral envelope glycoprotein (Env). HIV-1 Env, a type 1 membrane-spanning glycoprotein, is a trimeric complex composed of three noncovalently linked heterodimers of gp120, the receptor-binding surface (SU) component, and gp41, the membrane-spanning, transmembrane (TM) component (12, 26, 44, 45). The gp120 and gp41 glycoproteins are synthesized as a precursor gp160 glycoprotein, which is encoded by the env gene. The gp160 precursor is cotranslationally glycosylated and, following transport to the trans-Golgi network, is cleaved into the mature products by a member of the furin family of endoproteases (45). Mature Env proteins are transported to the plasma membrane, where they are rapidly endocytosed or incorporated into virions (5, 33, 43). Recent evidence suggests that endocytosis and intracellular trafficking of Env is required for its interaction with Gag precursors and for efficient assembly into virions (20).HIV-1 Env molecules function as quasistable “spring-loaded” fusion machines. Recent studies have suggested that several regions of gp120 are reoriented following CD4 binding so that a planar “bridging sheet,” which forms the binding site for the coreceptor (CCR5 or CXCR4), can form (6, 7). Coreceptor binding is necessary for additional conformational changes in gp41 and for complete fusion (3). The gp41 monomer has three subdomains, an ectodomain, a membrane-spanning domain (MSD), and a cytoplasmic domain (39). The ectodomain of gp41, which mediates membrane fusion, is composed of a fusion peptide, two heptad repeats, and a tryptophan-rich membrane-proximal external region. Following the binding of gp120 to the CD4 receptor and the CCR5/CXCR4 coreceptor, conformational changes are induced in Env that result in the exposure of the gp41 fusion peptide (32). This peptide inserts into the target cell membrane, allowing gp41 to form a bridge between the viral and cellular membranes. Interaction of the heptad repeats to form a six-helix bundle then brings the target and viral membranes together, allowing membrane fusion to occur (24).While heptad repeat regions 1 and 2 in the N-terminal ectodomain play key roles in Env-mediated fusion by bringing the viral and cell membranes into close proximity, an important function of gp41 is to anchor the glycoprotein complex within the host-derived viral membrane (18). The precise boundaries of the HIV-1 MSD have not been clearly defined; however, the MSD is one of the most conserved regions in the gp41 sequence. Based on the initial functional studies of HIV-1, the MSD of Env was defined as a stretch of 25 predominantly hydrophobic amino acids that span residues K681 to R705 in the NL4-3 sequence (14, 16, 18). These residues were suggested to cross the viral membrane in the form of an alpha helix, the length of which is approximately equal to the theoretical depth of a membrane bilayer. A major caveat of this model is that it places a basic amino acid residue (R694) into the hydrophobic center of the lipid bilayer. While some transmembrane proteins do contain charged amino acid residues in their MSDs, it is normally considered to be energetically unfavorable without some mechanism to neutralize the charge (8, 13). Point mutation studies have yielded varying results, but in general, substitution of K681 is detrimental to fusion and infectivity while mutation of R694 or R705 has only a limited effect on these activities (16, 29). On the other hand, accumulating data argue for a different intramembrane structure of the HIV-1 MSD. Serial small deletions (3 amino acid residues) in the region between R694 and R705 showed normal cell-cell fusion, although larger deletions were detrimental (29), suggesting that, with respect to the biological functions of the Env glycoprotein, the length of this region is more important than its amino acid conservation.Previous C-terminal-truncation studies of simian immunodeficiency virus (SIV) Env (19, 41) suggested that the entire 27-amino-acid region is not required for the biological function of the protein. In the case of SIV, only the 15 apolar amino acids flanked by K689 and R705 (equivalent to K681 and R694 in HIV) and 6 additional amino acids (for a total of 23 amino acids) were required for near-wild-type (WT) fusion (19, 41). Two subsequent residues were required (total, 25 amino acids) for virus-cell entry and infectivity, while a length of 21 amino acid residues was sufficient for SIV Env to be incorporated into viral particles. These results led to a basic amino acid “snorkeling” model for the SIV MSD (41). In this model, the lysine and arginine (NL4-3 equivalents of K681 and R694) are buried in the lipid bilayer, while their long side chains are proposed to extend outward to the membrane surface and present the positively charged amino groups to the negatively charged head groups of the lipid bilayers. Applied to HIV-1 MSD, this model predicts a hydrophobic intramembrane core of only 12 amino acid residues (compared to 15 amino acid residues in the SIV MSD) between K681 and R694. The hydrophobic region C-terminal to K681 is not sufficient to effectively anchor the protein, since mutation of R694 to a stop codon yielded a nonfunctional protein that appeared to be retained in the endoplasmic reticulum (11). This contrasts with truncation experiments with the vesicular stomatitis virus (VSV) G glycoprotein, which have shown that a region of 12 hydrophobic amino acids flanked by basic residues is sufficient to anchor the protein in the membrane (1).In order to understand if the “snorkeling” model is applicable to the HIV-1 MSD, we constructed a series of nonsense mutants with HIV-1 gp41 truncated in single-amino-acid steps at the C terminus from residue R707 to residue R694. For each mutant Env, we determined the membrane stability, fusogenicity, and ability to mediate infectivity. The results of these studies suggest that the 12-residue “core” (36) plus three subsequent hydrophobic amino acids is the minimal anchor domain for HIV-1 Env, as well as the minimal sequence to mediate cell-cell fusion. In contrast to SIV Env, HIV-1 Env requires the entire 25-amino-acid region from K681 to R707 to mediate near-WT incorporation and infectivity.  相似文献   

7.
8.
Mutations were introduced into the ectodomain of the human immunodeficiency virus type 1 (HIV-1) transmembrane envelope glycoprotein, gp41, within a region immediately adjacent to the membrane-spanning domain. This region, which is predicted to form an α-helix, contains highly conserved hydrophobic residues and is unusually rich in tryptophan residues. In addition, this domain overlaps the epitope of a neutralizing monoclonal antibody, 2F5, as well as the sequence corresponding to a peptide, DP-178, shown to potently neutralize virus. Site-directed mutagenesis was used to create deletions, substitutions, and insertions centered around a stretch of 17 hydrophobic and uncharged amino acids (residues 666 to 682 of the HXB2 strain of HIV-1) in order to determine the role of this region in the maturation and function of the envelope glycoprotein. Deletion of the entire stretch of 17 amino acids abrogated the ability of the envelope glycoprotein to mediate both cell-cell fusion and virus entry without affecting the normal maturation, transport, or CD4-binding ability of the protein. This phenotype was also demonstrated by substituting alanine residues for three of the five tryptophan residues within this sequence. Smaller deletions, as well as multiple amino acid substitutions, were also found to inhibit but not block cell-cell fusion. These results demonstrate the crucial role of a tryptophan-rich motif in gp41 during a post-CD4-binding step of glycoprotein-mediated fusion. The basis for the invariant nature of the tryptophans, however, appears to be at the level of glycoprotein incorporation into virions. Even the substitution of phenylalanine for a single tryptophan residue was sufficient to reduce Env incorporation and drop the efficiency of virus entry approximately 10-fold, despite the fact that the same mutation had no significant effect on syncytium formation.  相似文献   

9.
10.
The envelope glycoprotein (Env) of human immunodeficiency virus type 1 (HIV-1) has been shown to redirect the site of virus assembly in polarized epithelial cells. To test whether localization of the glycoprotein exclusively to the endoplasmic reticulum (ER) could redirect virus assembly to that organelle in nonpolarized cells, an ER -retrieval signal was engineered into an epitope-tagged variant of Env. The epitope tag, attached to the C terminus of Env, did not affect the normal maturation and transport of the glycoprotein or the incorporation of Env into virions. The epitope-tagged Env was also capable of mediating syncytium formation and virus entry with a similar efficiency to that of wild-type Env. When the epitope was modified to contain a consensus K(X)KXX ER retrieval signal, however, the glycoprotein was no longer proteolytically processed into its surface and transmembrane subunits and Env could not be detected at the cell surface by biotinylation. Endoglycosidase H analysis revealed that the modified Env was not transported to the Golgi apparatus. Immunofluorescent staining patterns were also consistent with the exclusion of Env from the Golgi. As expected, cells expressing the modified Env failed to form syncytia with CD4+ permissive cells. Despite this tight localization of Env to the ER, when the modified Env was expressed in the context of virus, virions continued to be produced efficiently from the plasma membrane of transfected cells. However, these virions contained no detectable glycoprotein and were noninfectious. Electron microscopy revealed virus budding from the plasma membrane of these cells, but no virus was seen assembling at the ER membrane and no assembled virions were found within the cell. These results suggest that the accumulation of Env in an intracellular compartment is not sufficient to redirect the assembly of HIV Gag in nonpolarized cells.  相似文献   

11.
The gp41 envelope protein of human immunodeficiency virus type 1 (HIV-1) contains an alpha-helical core structure responsible for mediating membrane fusion during viral entry. Recent studies suggest that a conserved hydrophobic cavity in the coiled coil of this core plays a distinctive structural role in maintaining the fusogenic conformation of the gp41 molecule. Here we investigated the importance of this cavity in determining the structure and biological activity of the gp41 core by using the N34(L6)C28 model. The high-resolution crystal structures of N34(L6)C28 of two HIV-1 gp41 fusion-defective mutants reveal that each mutant sequence is accommodated in the six-helix bundle structure by forming the cavity with different sets of atoms. Remarkably, the mutant N34(L6)C28 cores are highly effective inhibitors of HIV-1 infection, with 5- to 16-fold greater activity than the wild-type molecule. The enhanced inhibitory activity by fusion-defective mutations correlates with local structural perturbations close to the cavity that destabilize the six-helix bundle. Taken together, these results indicate that the conserved hydrophobic coiled-coil cavity in the gp41 core is critical for HIV-1 entry and its inhibition and provides a potential antiviral drug target.  相似文献   

12.
Incorporation of the human immunodeficiency virus type 1 (HIV-1) envelope glycoproteins into assembling particles is crucial for virion infectivity. Genetic and biochemical data indicate that the matrix (MA) domain of Gag and the cytoplasmic tail of the transmembrane glycoprotein gp41 play an important role in coordinating Env incorporation; however, the molecular mechanism and possible role of host factors in this process remain to be defined. Recent studies suggested that Env incorporation is mediated by interactions between matrix and tail-interacting protein of 47 kDa (TIP47; also known as perilipin-3 and mannose-6-phosphate receptor-binding protein 1), a member of the perilipin, adipophilin, TIP47 (PAT) family of proteins implicated in protein sorting and lipid droplet biogenesis. We have confirmed by nuclear magnetic resonance spectroscopy titration experiments and surface plasmon resonance that MA binds TIP47. We also reevaluated the role of TIP47 in HIV-1 Env incorporation in HeLa cells and in the Jurkat T-cell line. In HeLa cells, TIP47 overexpression or RNA interference (RNAi)-mediated depletion had no significant effect on HIV-1 Env incorporation, virus release, or particle infectivity. Similarly, depletion of TIP47 in Jurkat cells did not impair HIV-1 Env incorporation, virus release, infectivity, or replication. Our results thus do not support a role for TIP47 in HIV-1 Env incorporation or virion infectivity.  相似文献   

13.
To evaluate antibody specificities induced by simian immunodeficiency virus (SIV) versus human immunodeficiency virus type 1 (HIV-1) envelope antigens in nonhuman primate (NHP), we profiled binding antibody responses to linear epitopes in NHP studies with HIV-1 or SIV immunogens. We found that, overall, HIV-1 Env IgG responses were dominated by V3, with the notable exception of the responses to the vaccine strain A244 Env that were dominated by V2, whereas the anti-SIVmac239 Env responses were dominated by V2 regardless of the vaccine regimen.  相似文献   

14.
The interaction of the human immunodeficiency virus type 1 (HIV-1) Pr55Gag molecule with the plasma membrane of an infected cell is an essential step of the viral life cycle. Myristic acid and positively charged residues within the N-terminal portion of MA constitute the membrane-binding domain of Pr55Gag. A separate assembly domain, termed the interaction (I) domain, is located nearer the C-terminal end of the molecule. The I domain is required for production of dense retroviral particles, but has not previously been described to influence the efficiency of membrane binding or the subcellular distribution of Gag. This study used a series of Gag-green fluorescent protein fusion constructs to define a region outside of MA which determines efficient plasma membrane interaction. This function was mapped to the nucleocapsid (NC) region of Gag. The minimal region in a series of C-terminally truncated Gag proteins conferring plasma membrane fluorescence was identified as the N-terminal 14 amino acids of NC. This same region was sufficient to create a density shift in released retrovirus-like particles from 1.13 to 1.17 g/ml. The functional assembly domain previously termed the I domain is thus required for the efficient plasma membrane binding of Gag, in addition to its role in determining the density of released particles. We propose a model in which the I domain facilitates the interaction of the N-terminal membrane-binding domain of Pr55Gag with the plasma membrane.  相似文献   

15.
An effective AIDS vaccine must control highly diverse circulating strains of human immunodeficiency virus type 1 (HIV-1). Among HIV-1 gene products, the envelope (Env) protein contains variable as well as conserved regions. In this report, an informatic approach to the design of T-cell vaccines directed to HIV-1 Env M group global sequences was tested. Synthetic Env antigens were designed to express mosaics that maximize the inclusion of common potential T-cell epitope (PTE) 9-mers and minimize the inclusion of rare epitopes likely to elicit strain-specific responses. DNA vaccines were evaluated using intracellular cytokine staining in inbred mice with a standardized panel of highly conserved 15-mer PTE peptides. One-, two-, and three-mosaic sets that increased theoretical epitope coverage were developed. The breadth and magnitude of T-cell immunity stimulated by these vaccines were compared to those for natural strain Envs; additional comparisons were performed on mutant Envs, including gp160 or gp145 with or without V regions and gp41 deletions. Among them, the two- or three-mosaic Env sets elicited the optimal CD4 and CD8 responses. These responses were most evident in CD8 T cells; the three-mosaic set elicited responses to an average of eight peptide pools, compared to two pools for a set of three natural Envs. Synthetic mosaic HIV-1 antigens can therefore induce T-cell responses with expanded breadth and may facilitate the development of effective T-cell-based HIV-1 vaccines.The development of AIDS vaccines has been advanced recently by demonstrations of increased survival and decreased viral load following vaccination with T-cell vaccines in nonhuman primate models (12, 19, 23, 26, 31, 37). Although such vaccine studies have implied that T cells may contribute to the control of viremia in the highly lethal simian immunodeficiency virus SIVmac251 challenge model, the applicability of these results in human studies remains uncertain. The major concern regarding the efficacy of human immunodeficiency virus (HIV) vaccines in humans is the extraordinary genetic diversity of the virus. The sequence similarity of HIV type 1 (HIV-1) envelope from diverse isolates within a clade can diverge by as much as 15%, and divergence between alternative clades may approach 30% (10). In addition, the diversity of the viral Gag gene product can approach similar levels, particularly in p17 and p15, which are much more diverse than p24 (6), although Gag does not have the extreme localized diversity seen in the highly variable regions of Env (6, 10). While the approach to viral diversity has been addressed in existing vaccines through the use of envelopes derived from representative viruses in the major clades, increasing knowledge about the genetic diversity of naturally occurring isolates has enabled alternative approaches that enhance population coverage of vaccine-elicited T-cell responses.Approaches under consideration include the use of central gene sequences based on ancestral, consensus, or center-of-the-tree genetic analyses (5, 10, 18, 31, 36). Such prototypes are derived by selection of the most common amino acids at each residue (10, 16, 17, 21, 25, 36), identifying the most recent common ancestor of diverging viruses in a vaccine target population (5, 10, 18, 36), or modeling the sequence at the center of the phylogenetic tree (29), respectively. Peptides based on any of these three centralized protein strategies enhanced the detection of T-cell responses in natural infection relative to the use of peptides based on natural strains; however, all three strategies behaved equivalently (7).The use of a single M group consensus/ancestral Env sequence has been shown to elicit T-cell responses with greater breadth of cross-reactivity than single natural strains in animal models (31, 36). Such central sequences do not exist in nature, and even phylogenetic ancestral reconstructions are just an approximate model of an ancestral state of the virus (8). Thus, central sequence strategies have provided evidence that various informatically derived gene products can elicit immune responses to T-cell epitopes found in diverse circulating strains, leading to the possibility of using computational strategies to design polyvalent vaccines which optimize T-cell coverage (6, 24). In this study, we have evaluated for the first time the ability of nonnatural mosaic Env immunogens (6) to elicit T-cell responses of increased cross-reactivity against epitopes represented in naturally circulating viruses in animals.Mosaic HIV-1 envelope genes were derived using an informatic approach, whereby in silico-generated recombinants of natural variants from the Los Alamos database M group Env alignment were created, scored, and selected in combination to optimize the coverage of 9-mers in the global database for a given vaccine cocktail size. While mosaic proteins are artificial constructs that do not occur in nature, they align well to natural proteins, and any short span found in mosaics will tend to be found repeatedly among natural strains (although some of the hypervariable loop regions of Env are so extremely variable that they are not repeated among circulating strains, and this necessitates bridging these regions with segments found in a single strain). In silico recombination breakpoints are constrained to create fusion points found in natural sequences. It is possible to provide increased breadth of coverage with a single mosaic, providing the maximum possible single-antigen diversity coverage for stretches of nine amino acids. Alternatively, multiple mosaics can increase the breadth of representation but have the drawback of requiring the synthesis of additional vectors for clinical use. Mosaics also preserve a natural Env-like sequence to retain normal antigen processing. Here, we have compared single-, double-, or triple-mosaic envelope antigen sets to naturally circulating strains or other derivatives for their ability to elicit immune responses of increased breadth. The data suggest that mosaic HIV-1 envelope sequences provide an approach that may be useful in the development of HIV vaccines that respond to T-cell epitopes represented in naturally circulating strains.  相似文献   

16.
In vivo passage of a poorly replicating, nonpathogenic simian-human immunodeficiency virus (SHIV-HXBc2) generated an efficiently replicating virus, KU-1, that caused rapid CD4+ T-lymphocyte depletion and AIDS-like illness in monkeys (S. V. Joag, Z. Li, L. Foresman, E. B. Stephens, L.-J. Zhao, I. Adany, D. M. Pinson, H. M. McClure, and O. Narayan, J. Virol. 70:3189–3197, 1996). The env gene of the KU-1 virus was used to create a molecularly cloned virus, SHIV-HXBc2P 3.2, that differed from a nonpathogenic SHIV-HXBc2 virus in only 12 envelope glycoprotein residues. SHIV-HXBc2P 3.2 replicated efficiently and caused rapid and persistent CD4+ T-lymphocyte depletion in inoculated rhesus macaques. Compared with the envelope glycoproteins of the parental SHIV-HXBc2, the SHIV-HXBc2P 3.2 envelope glycoproteins supported more efficient infection of rhesus monkey peripheral blood mononuclear cells. Both the parental SHIV-HXBc2 and the pathogenic SHIV-HXBc2P 3.2 used CXCR4 but none of the other seven transmembrane segment receptors tested as a second receptor. Compared with the parental virus, viruses with the SHIV-HXBc2P 3.2 envelope glycoproteins were more resistant to neutralization by soluble CD4 and antibodies. Thus, changes in the envelope glycoproteins account for the ability of the passaged virus to deplete CD4+ T lymphocytes rapidly and specify increased replicative capacity and resistance to neutralization.  相似文献   

17.
Inactivation of progeny virions with chimeric virion-associated proteins represents a novel therapeutic approach against human immunodeficiency virus (HIV) replication. The HIV type 1 (HIV-1) Vpr gene product, which is packaged into virions, is an attractive candidate for such a strategy. In this study, we developed Vpr-based fusion proteins that could be specifically targeted into mature HIV-1 virions to affect their structural organization and/or functional integrity. Two Vpr fusion proteins were constructed by fusing to the first 88 amino acids of HIV-1 Vpr the chloramphenicol acetyltransferase enzyme (VprCAT) or the last 18 C-terminal amino acids of the HIV-1 Vpu protein (VprIE). These Vpr fusion proteins were initially designed to quantify their efficiency of incorporation into HIV-1 virions when produced in cis from the provirus. Subsequently, CD4+ Jurkat T-cell lines constitutively expressing the VprCAT or the VprIE fusion protein were generated with retroviral vectors. Expression of the VprCAT or the VprIE fusion protein in CD4+ Jurkat T cells did not interfere with cellular viability or growth but conferred substantial resistance to HIV replication. The resistance to HIV replication was more pronounced in Jurkat-VprIE cells than in Jurkat-VprCAT cells. Moreover, the antiviral effect mediated by VprIE was dependent on an intact p6gag domain, indicating that the impairment of HIV-1 replication required the specific incorporation of Vpr fusion protein into virions. Gene expression, assembly, or release was not affected upon expression of these Vpr fusion proteins. Indeed, the VprIE and VprCAT fusion proteins were shown to affect the infectivity of progeny virus, since HIV virions containing the VprCAT or the VprIE fusion proteins were, respectively, 2 to 3 times and 10 to 30 times less infectious than the wild-type virus. Overall, this study demonstrated the successful transfer of resistance to HIV replication in tissue cultures by use of Vpr-based antiviral genes.  相似文献   

18.
Previous studies have shown that in addition to its function in specific RNA encapsidation, the human immunodeficiency virus type 1 (HIV-1) nucleocapsid (NC) is required for efficient virus particle assembly. However, the mechanism by which NC facilitates the assembly process is not clearly established. Formally, NC could act by constraining the Pr55gag polyprotein into an assembly-competent conformation or by masking residues which block the assembly process. Alternatively, the capacity of NC to bind RNA or make interprotein contacts might affect particle assembly. To examine its role in the assembly process, we replaced the NC domain in Pr55gag with polypeptide domains of known function, and the chimeric proteins were analyzed for their abilities to direct the release of virus-like particles. Our results indicate that NC does not mask inhibitory domains and does not act passively, by simply providing a stable folded monomeric structure. However, replacement of NC by polypeptides which form interprotein contacts permitted efficient virus particle assembly and release, even when RNA was not detected in the particles. These results suggest that formation of interprotein contacts by NC is essential to the normal HIV-1 assembly process.Human immunodeficiency virus type 1 (HIV-1) encodes three major genes, gag, pol, and env, which are commonly found in all mammalian retroviruses. It also encodes accessory genes whose protein products are important for regulation of its life cycle (6, 30, 35). However, of all the genes encoded by HIV-1, only the protein product of the gag gene has been found to be necessary and sufficient for the assembly of virus-like particles (11, 13, 17, 22, 32, 33). The HIV-1 Gag protein initially is expressed as a 55-kDa polyprotein precursor (Pr55gag), but during or shortly after particle release, Pr55gag ordinarily is cleaved by the viral protease (PR). The products of the protease action are the four major viral proteins matrix (MA), capsid (CA), nucleocapsid (NC), and p6, and the two spacer polypeptides p2 and p1, which represent sequences between CA and NC and between NC and p6, respectively (15, 19, 23, 30).The HIV-1 nucleocapsid proteins have two Cys-X2-Cys-X4-His-X4-Cys (Cys-His) motifs, reminiscent of the zinc finger motifs found in many DNA binding proteins, and NC has been shown to facilitate the specific encapsidation of HIV-1 genomic RNAs. In addition to its encapsidation function, NC influences virus particle assembly (7, 10, 17, 21, 40). In particular, Gag proteins lacking the NC domain fail to assemble virus particles efficiently. Nevertheless, some chimeric Gag proteins which carry foreign sequences in place of NC have been shown to assemble and release virus particles at wild-type (wt) levels (2, 37, 40). Thus, it appears that in some circumstances, the role that NC plays in virus particle assembly can be replaced. To date, it is not clear how NC affects particle assembly, although several possibilities might be envisioned. One possibility is that deletion of NC unmasks inhibitory sequences in p2 or the C terminus of CA. Alternatively, NC may simply provide a stable monomeric folded structure which locks CA or other Gag domains into an assembly-competent conformation. Another possibility is that NC facilitates assembly by forming essential protein-protein contacts between neighbor Prgag molecules, as suggested in cross-linking studies (21). Finally, the assembly role of NC may stem from its RNA binding capabilities, a hypothesis supported by studies of Campbell and Vogt (5), which have shown that RNA facilitates the in vitro assembly of retroviral Gag proteins into higher-order structures.To distinguish among possible mechanisms by which NC facilitates HIV-1 assembly, we replaced NC with polypeptides having known structural characteristics and examined particle assembly directed by these chimeric proteins. Using this approach, we have found that NC does not play a passive role in HIV-1 assembly as either a mask to assembly inhibitor domains or a nonspecific, stably folded structure. Rather, sequences known to form strong interprotein contacts were observed to enhance assembly, suggesting a similar role for the NC domain itself. With several assembly-competent chimeric proteins, we detected no particle-associated RNAs. These results suggest that while RNA may be essential to virus assembly in the context of the wt Pr55gag protein, it is dispensable for formation of virus-like particles from chimeric proteins.  相似文献   

19.
nef alleles derived from a large number of individuals infected with human immunodeficiency virus type 1 (HIV-1) were analyzed to investigate the frequency of disrupted nef genes and to elucidate whether specific amino acid substitutions in Nef are associated with different stages of disease. We confirm that deletions or gross abnormalities in nef are rarely present. However, a comparison of Nef consensus sequences derived from 41 long-term nonprogressors and from 50 individuals with progressive HIV-1 infection revealed that specific variations are associated with different stages of infection. Five amino acid variations in Nef (T15, N51, H102, L170, and E182) were more frequently observed among nonprogressors, while nine features (an additional N-terminal PxxP motif, A15, R39, T51, T157, C163, N169, Q170, and M182) were more frequently found in progressors. Strong correlations between the frequency of these variations in Nef and both the CD4(+)-cell count and the viral load were observed. Moreover, analysis of sequential samples obtained from two progressors revealed that several variations in Nef, which were more commonly observed in patients with low CD4(+)-T-cell counts, were detected only during or after progression to immunodeficiency. Our results indicate that sequence variations in Nef are associated with different stages of HIV-1 infection and suggest a link between nef gene function and the immune status of the infected individual.  相似文献   

20.
Receptors (FcγRs) for the constant region of immunoglobulin G (IgG) are an important link between humoral immunity and cellular immunity. To help define the role of FcγRs in determining the fate of human immunodeficiency virus type 1 (HIV-1) immune complexes, cDNAs for the four major human Fcγ receptors (FcγRI, FcγRIIa, FcγRIIb, and FcγRIIIa) were stably expressed by lentiviral transduction in a cell line (TZM-bl) commonly used for standardized assessments of HIV-1 neutralization. Individual cell lines, each expressing a different FcγR, bound human IgG, as evidence that the physical properties of the receptors were preserved. In assays with a HIV-1 multisubtype panel, the neutralizing activities of two monoclonal antibodies (2F5 and 4E10) that target the membrane-proximal external region (MPER) of gp41 were potentiated by FcγRI and, to a lesser extent, by FcγRIIb. Moreover, the neutralizing activity of an HIV-1-positive plasma sample known to contain gp41 MPER-specific antibodies was potentiated by FcγRI. The neutralizing activities of monoclonal antibodies b12 and 2G12 and other HIV-1-positive plasma samples were rarely affected by any of the four FcγRs. Effects with gp41 MPER-specific antibodies were moderately stronger for IgG1 than for IgG3 and were ineffective for Fab. We conclude that FcγRI and FcγRIIb facilitate antibody-mediated neutralization of HIV-1 by a mechanism that is dependent on the Fc region, IgG subclass, and epitope specificity of antibody. The FcγR effects seen here suggests that the MPER of gp41 could have greater value for vaccines than previously recognized.Fc receptors (FcRs) are differentially expressed on a variety of cells of hematopoietic lineage, where they bind the constant region of antibody (Ab) and provide a link between humoral and cellular immunity. Humans possess two classes of FcRs for the constant region of IgG (FcγRs) that, when cross-linked, are distinguished by their ability to either activate or inhibit cell signaling (69, 77, 79). The activating receptors FcγRI (CD64), FcγRIIa (CD32), and FcγRIII (CD16) signal through an immunoreceptor tyrosine-based activation motif (ITAM), whereas FcγRIIb (CD32) contains an inhibitory motif (ITIM) that counters ITAM signals and B-cell receptor signals. It has been suggested that a balance between activating and inhibitory FcγRs coexpressed on the same cells plays an important role in regulating adaptive immunity (23, 68). Moreover, the inhibitory FcγRIIb, being the sole FcγR on B cells, appears to play an important role in regulating self-tolerance (23, 68). The biologic role of FcγRs may be further influenced by differences in their affinity for immunoglobulin G (IgG); thus, FcγRI is a high-affinity receptor that binds monomeric IgG (mIgG) and IgG immune complexes (IC), whereas FcγRIIa, FcγRIIb, and FcγRIIIa are medium- to low-affinity receptors that preferentially bind IgG IC (10, 49, 78). FcγRs also exhibit differences in their relative affinity for the four IgG subclasses (10), which has been suggested to influence the balance between activating and inhibitory FcγRs (67).In addition to their participation in acquired immunity, FcγRs can mediate several innate immune functions, including phagocytosis of opsonized pathogens, Ab-dependent cell cytotoxicity (ADCC), antigen uptake by professional antigen-presenting cells, and the production of inflammatory cytokines and chemokines (26, 35, 41, 48, 69). In some cases, interaction of Ab-coated viruses with FcγRs may be exploited by viruses as a means to facilitate entry into FcγR-expressing cells (2, 33, 47, 84). Several groups have reported FcγR-mediated Ab-dependent enhancement (ADE) of HIV-1 infection in vitro (47, 51, 58, 63, 94, 96), whereas other reports have implicated FcγRs in efficient inhibition of the virus in vitro (19, 21, 29, 44-46, 62, 98) and possibly as having beneficial effects against HIV-1 in vivo (5, 27, 28, 42). These conflicting results are further complicated by the fact that HIV-1-susceptible cells, such as monocytes and macrophages, can coexpress more than one FcγR (66, 77, 79).HIV-1 entry requires sequential interactions between the viral surface glycoprotein, gp120, and its cellular receptor (CD4) and coreceptor (usually CCR5 or CXCR4), followed by membrane fusion that is mediated by the viral transmembrane glycoprotein gp41 (17, 106). Abs neutralize the virus by binding either gp120 or gp41 and blocking entry into cells. Several human monoclonal Abs that neutralize a broad spectrum of HIV-1 variants have attracted considerable interest for vaccine design. Epitopes for these monoclonal Abs include the receptor binding domain of gp120 in the case of b12 (71, 86), a glycan-specific epitope on gp120 in the case of 2G12 (13, 85, 86), and two adjacent epitopes in the membrane-proximal external region (MPER) of g41 in the cases of 2F5 and 4E10 (3, 11, 38, 93). At least three of these monoclonal Abs have been shown to interact with FcRs and to mediate ADCC (42, 43).A highly standardized and validated assay for neutralizing Abs against HIV-1 that quantifies reductions in luciferase (Luc) reporter gene expression after a single round of virus infection in TZM-bl cells has been developed (60, 104). TZM-bl (also called JC53BL-13) is a CXCR4-positive HeLa cell line that was engineered to express CD4 and CCR5 and to contain integrated reporter genes for firefly Luc and Escherichia coli β-galactosidase under the control of the HIV-1 Tat-regulated promoter in the long terminal repeat terminal repeat sequence (74, 103). TZM-bl cells are permissive to infection by a wide variety of HIV-1, simian immunodeficiency virus, and human-simian immunodeficiency virus strains, including molecularly cloned Env-pseudotyped viruses. Here we report the creation and characterization of four new TZM-bl cell lines, each expressing one of the major human FcγRs. These new cell lines were used to gain a better understanding of the individual roles that FcγRs play in determining the fate of HIV-1 IC. Two FcγRs that potentiated the neutralizing activity of gp41 MPER-specific Abs were identified.  相似文献   

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