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 31 毫秒
1.
The Epstein-Barr virus gH-gL complex includes a third glycoprotein, gp42, which is the product of the BZLF2 open reading frame (ORF). gp42 has been implicated as critical to infection of the B lymphocyte by virtue of its interaction with HLA class II on the B-cell surface. A neutralizing antibody that reacts with gp42 inhibits virus-cell fusion and blocks binding of gp42 to HLA class II; antibody to HLA class II can inhibit infection, and B cells that lack HLA class II can only be infected if HLA class II expression is restored. To confirm whether gp42 is an essential component of the virion, we derived a recombinant virus with a selectable marker inserted into the BZLF2 ORF to interrupt expression of the protein. A complex of gH and gL was expressed by the recombinant virus in the absence of gp42. Recombinant virus egressed from the cell normally and could bind to receptor-positive cells. It had, however, lost the ability to infect or transform B lymphocytes. Treatment with polyethylene glycol restored the infectivity of recombinant virus, confirming that gp42 is essential for penetration of the B-cell membrane.Entry of enveloped viruses into mammalian cells requires that the virion envelope fuse with the cell membrane after attachment to the cell surface. Herpesviruses require the functions of multiple protein species to mediate this event, and in keeping with the common origin and diverse habitats of these viruses, some of the proteins involved in penetration appear to be conserved throughout the family and some appear to be restricted to individual members or more closely related members with similar tropism. The two glycoproteins gH and gL fall into the first category of conserved proteins. Glycoprotein gH has been implicated as a major player in virus-cell fusion in many herpesviruses (8, 10, 11, 22, 28, 32, 34), and gL is an essential partner which is required for folding and transport of gH out of the endoplasmic reticulum (6, 19, 21, 27, 28, 35, 38, 45). The gH and gL homologs of Epstein-Barr virus (EBV) are gp85, the product of the BXLF2 open reading frame (ORF) (13, 31), and gp25, the product of the BKRF2 ORF (45), and these homologs appear to behave much as their counterparts in other herpesviruses do (45). However, a third glycoprotein, gp42, associates with the EBV gH-gL complex and falls into the second category of proteins, those with a more restricted distribution.Glycoprotein gp42 is the product of the BZLF2 ORF (26), and although there may be a functionally similar protein in cytomegalovirus (18, 24), it is not predicted to have a homolog in other human herpesviruses. It does, however, have a homolog in ORF51 of equine herpes virus 2 (43). Both EBV and equine herpes virus 2 infect B lymphocytes (1), and several lines of evidence suggest that, at least in the case of EBV, gp42 is critical to the infection of this cell type. A monoclonal antibody (MAb) called F-2-1 that reacts with gp42 has no affect on EBV attachment to its receptor, complement receptor type 2 (CR2) (CD21), but inhibits fusion of the virus with the B-cell membrane and neutralizes infection (29). Glycoprotein gp42 interacts with the β1 domain of the HLA class II protein HLA-DR (39), and MAb F-2-1 interferes with this interaction (25). Like F-2-1, a MAb to HLA-DR or a soluble form of gp42 can block B-cell transformation, and B-cell lines which lack expression of HLA class II are not susceptible to superinfection with EBV unless expression of HLA class II is restored (25). Collectively these observations suggest that gp42, probably by virtue of its interaction with HLA class II, is essential to infection of the B lymphocyte. To answer directly the question of whether gp42 is an indispensable glycoprotein, we derived a virus that could be definitively shown to lack expression of the molecule and examined its ability to infect normal resting B lymphocytes. We report here that virus with expression of gp42 blocked can exit cells normally and can bind to receptor-positive target cells. However, it is unable to penetrate into cells and initiate infection.  相似文献   

2.
The human immunodeficiency virus type 1 (HIV-1) vpu gene encodes a type I anchored integral membrane phosphoprotein with two independent functions. First, it regulates virus release from a post-endoplasmic reticulum (ER) compartment by an ion channel activity mediated by its transmembrane anchor. Second, it induces the selective down regulation of host cell receptor proteins (CD4 and major histocompatibility complex class I molecules) in a process involving its phosphorylated cytoplasmic tail. In the present work, we show that the Vpu-induced proteolysis of nascent CD4 can be completely blocked by peptide aldehydes that act as competitive inhibitors of proteasome function and also by lactacystin, which blocks proteasome activity by covalently binding to the catalytic β subunits of proteasomes. The sensitivity of Vpu-induced CD4 degradation to proteasome inhibitors paralleled the inhibition of proteasome degradation of a model ubiquitinated substrate. Characterization of CD4-associated oligosaccharides indicated that CD4 rescued from Vpu-induced degradation by proteasome inhibitors is exported from the ER to the Golgi complex. This finding suggests that retranslocation of CD4 from the ER to the cytosol may be coupled to its proteasomal degradation. CD4 degradation mediated by Vpu does not require the ER chaperone calnexin and is dependent on an intact ubiquitin-conjugating system. This was demonstrated by inhibition of CD4 degradation (i) in cells expressing a thermally inactivated form of the ubiquitin-activating enzyme E1 or (ii) following expression of a mutant form of ubiquitin (Lys48 mutated to Arg48) known to compromise ubiquitin targeting by interfering with the formation of polyubiquitin complexes. CD4 degradation was also prevented by altering the four Lys residues in its cytosolic domain to Arg, suggesting a role for ubiquitination of one or more of these residues in the process of degradation. The results clearly demonstrate a role for the cytosolic ubiquitin-proteasome pathway in the process of Vpu-induced CD4 degradation. In contrast to other viral proteins (human cytomegalovirus US2 and US11), however, whose translocation of host ER molecules into the cytosol occurs in the presence of proteasome inhibitors, Vpu-targeted CD4 remains in the ER in a transport-competent form when proteasome activity is blocked.

The human immunodeficiency virus type 1 (HIV-1)-specific accessory protein Vpu performs two distinct functions in the viral life cycle (11, 12, 29, 34, 46, 47, 5052; reviewed in references 31 and 55): enhancement of virus particle release from the cell surface, and the selective induction of proteolysis of newly synthesized membrane proteins. Known targets for Vpu include the primary virus receptor CD4 (63, 64) and major histocompatibility complex (MHC) class I molecules (28). Vpu is an oligomeric class I integral membrane phosphoprotein (35, 48, 49) with a structurally and functionally defined domain architecture: an N-terminal transmembrane anchor and C-terminal cytoplasmic tail (20, 34, 45, 47, 50, 65). Vpu-induced degradation of endoplasmic reticulum (ER) membrane proteins involves the phosphorylated cytoplasmic tail of the protein (50), whereas the virion release function is mediated by a cation-selective ion channel activity associated with the membrane anchor (19, 31, 45, 47).CD4 is a 55-kDa class I integral membrane glycoprotein that serves as the primary coreceptor for HIV entry into cells. CD4 consists of a large lumenal domain, a transmembrane peptide, and a 38-residue cytoplasmic tail. It is expressed on the surface of a subset of T lymphocytes that recognize MHC class II-associated peptides, and it plays a pivotal role in the development and maintenance of the immune system (reviewed in reference 30). Down regulation of CD4 in HIV-1-infected cells is mediated through several independent mechanisms (reviewed in references 5 and 55): intracellular complex formation of CD4 with the HIV envelope protein gp160 (8, 14), endocytosis of cell surface CD4 induced by the HIV-1 nef gene product (1, 2), and ER degradation induced by the HIV-1 vpu gene product (63, 64).Vpu-induced degradation of CD4 is an example of ER-associated protein degradation (ERAD). ERAD is a common outcome when proteins in the secretory pathway are unable to acquire their native structure (4). Although it was thought that ERAD occurs exclusively inside membrane vesicles of the ER or other related secretory compartments, this has gained little direct experimental support. Indeed, there are several recent reports that ERAD may actually represent export of the target protein to the cytosol, where it is degraded by cytosolic proteases. It was found that in yeast, a secreted protein, prepro-α-factor (pαF), is exported from microsomes and degraded in the cytosol in a proteasome-dependent manner (36). This process was dependent on the presence of calnexin, an ER-resident molecular chaperone that interacts with N-linked oligosaccharides containing terminal glucose residues (3). In mammalian cells, two human cytomegalovirus (HCMV) proteins, US2 and US11, were found to cause the retranslocation of MHC class I molecules from the ER to the cytosol, where they are destroyed by proteasomes (61, 62). In the case of US2, class I molecules were found to associate with a protein (Sec61) present in the channel normally used to translocate newly synthesized proteins into the ER (termed the translocon), leading to the suggestion that the ERAD substrates are delivered to the cytosol by retrograde transport through the Sec61-containing pore (61). Fujita et al. (24) reported that, similar to these findings, the proteasome-specific inhibitor lactacystin (LC) partially blocked CD4 degradation in transfected HeLa cells coexpressing CD4, Vpu, and HIV-1 Env glycoproteins. In the present study, we show that Vpu-induced CD4 degradation can be completely blocked by proteasome inhibitors, does not require the ER chaperone calnexin, but requires the function of the cytosolic polyubiquitination machinery which apparently targets potential ubiquitination sites within the CD4 cytoplasmic tail. Our findings point to differences between the mechanism of Vpu-mediated CD4 degradation and ERAD processes induced by the HCMV proteins US2 and US11 (61, 62).  相似文献   

3.
Human immunodeficiency virus type 1 (HIV-1) uses a variety of chemokine receptors as coreceptors for virus entry, and the ability of the virus to be neutralized by antibody may depend on which coreceptors are used. In particular, laboratory-adapted variants of the virus that use CXCR4 as a coreceptor are highly sensitive to neutralization by sera from HIV-1-infected individuals, whereas primary isolates that use CCR5 instead of, or in addition to, CXCR4 are neutralized poorly. To determine whether this dichotomy in neutralization sensitivity could be explained by differential coreceptor usage, virus neutralization by serum samples from HIV-1-infected individuals was assessed in MT-2 cells, which express CXCR4 but not CCR5, and in mitogen-stimulated human peripheral blood mononuclear cells (PBMC), where multiple coreceptors including CXCR4 and CCR5 are available for use. Our results showed that three of four primary isolates with a syncytium-inducing (SI) phenotype and that use CXCR4 and CCR5 were neutralized poorly in both MT-2 cells and PBMC. The fourth isolate, designated 89.6, was more sensitive to neutralization in MT-2 cells than in PBMC. We showed that the neutralization of 89.6 in PBMC was not improved when CCR5 was blocked by having RANTES, MIP-1α, and MIP-1β in the culture medium, indicating that CCR5 usage was not responsible for the decreased sensitivity to neutralization in PBMC. Consistent with this finding, a laboratory-adapted strain of virus (IIIB) was significantly more sensitive to neutralization in CCR5-deficient PBMC (homozygous Δ32-CCR5 allele) than were two of two SI primary isolates tested. The results indicate that the ability of HIV-1 to be neutralized by sera from infected individuals depends on factors other than coreceptor usage.Human immunodeficiency virus type 1 (HIV-1), the etiologic agent of AIDS, utilizes the HLA class II receptor, CD4, as its primary receptor to gain entry into cells (17, 30). Entry is initiated by a high-affinity interaction between CD4 and the surface gp120 of the virus (32). Subsequent to this interaction, conformational changes that permit fusion of the viral membrane with cellular membranes occur within the viral transmembrane gp41 (9, 58, 59). In addition to CD4, one or more recently described viral coreceptors are needed for fusion to take place. These coreceptors belong to a family of seven-transmembrane G-protein-coupled proteins and include the CXC chemokine receptor CXCR4 (3, 4, 24, 44), the CC chemokine receptors CCR5 (1, 12, 13, 18, 21, 23, 45) and, less commonly, CCR3 and CCR2b (12, 21), and two related orphan receptors termed BONZO/STRL33 and BOB (19, 34). Coreceptor usage by HIV-1 can be blocked by naturally occurring ligands, including SDF-1 for CXCR4 (4, 44), RANTES, MIP-1α, and MIP-1β in the case of CCR5 (13, 45), and eotaxin for CCR3 (12).The selective cellular tropisms of different strains of HIV-1 may be determined in part by coreceptor usage. For example, all culturable HIV-1 variants replicate initially in mitogen-stimulated human peripheral blood mononuclear cells (PBMC), but only a minor fraction are able to infect established CD4+ T-cell lines (43). This differential tropism is explained by the expression of CXCR4 together with CCR5 and other CC chemokine coreceptors on PBMC and the lack of expression of CCR5 on most T-cell lines (5, 10, 19, 35, 39, 50, 53). Indeed, low-passage field strains (i.e., primary isolates) of HIV-1 that fail to replicate in T-cell lines use CCR5 as their major coreceptor and are unable to use CXCR4 (1, 12, 18, 21, 23, 28). Because these isolates rarely produce syncytia in PBMC and fail to infect MT-2 cells, they are often classified as having a non-syncytium-inducing (NSI) phenotype. Primary isolates with a syncytium-inducing (SI) phenotype are able to use CXCR4 alone or, more usually, in addition to CCR5 (16, 20, 51). HIV-1 variants that have been passaged multiple times in CD4+ T-cell lines, and therefore considered to be laboratory adapted, exhibit a pattern of coreceptor usage that resembles that of SI primary isolates. Most studies have shown that the laboratory-adapted strain IIIB uses CXCR4 alone (3, 13, 20, 24, 51) and that MN and SF-2 use CXCR4 primarily and CCR5 to a lesser degree (11, 13). Sequences within the V3 loop of gp120 have been shown to be important, either directly or indirectly, for the interaction of HIV-1 with both CXCR4 (52) and CCR5 (12, 14, 54, 60). This region of gp120 contains multiple determinants of cellular tropism (43) and is a major target for neutralizing antibodies to laboratory-adapted HIV-1 but not to primary isolates (29, 46, 57).It has been known for some time that the ability of sera from HIV-1-infected individuals to neutralize laboratory-adapted strains of HIV-1 does not predict their ability to neutralize primary isolates in vitro (7). In general, the former viruses are highly sensitive to neutralization whereas the latter viruses are neutralized poorly by antibodies induced in response to HIV-1 infection (7, 43). Importantly, neutralizing antibodies generated by candidate HIV-1 subunit vaccines have been highly specific for laboratory-adapted viruses (26, 37, 38). In principle, the dichotomy in neutralization sensitivity between these two categories of virus could be related to coreceptor usage. To test this, we investigated whether the use of CXCR4 in the absence of CCR5 would render SI primary isolates highly sensitive to neutralization in vitro by sera from HIV-1-infected individuals. Two similar studies using human monoclonal antibodies and soluble CD4 have been reported (31a, 55).  相似文献   

4.
Four new monoclonal antibodies (MAbs) that inhibit human T-cell lymphotropic virus type 1 (HTLV-1)-induced syncytium formation were produced by immunizing BALB/c mice with HTLV-1-infected MT2 cells. Immunoprecipitation studies and binding assays of transfected mouse cells showed that these MAbs recognize class II major histocompatibility complex (MHC) molecules. Previously produced anti-class II MHC antibodies also blocked HTLV-1-induced cell fusion. Coimmunoprecipitation and competitive MAb binding studies indicated that class II MHC molecules and HTLV-1 envelope glycoproteins are not associated in infected cells. Anti-MHC antibodies had no effect on human immunodeficiency virus type 1 (HIV-1) syncytium formation by cells coinfected with HIV-1 and HTLV-1, ruling out a generalized disruption of cell membrane function by the antibodies. High expression of MHC molecules suggested that steric effects of bound anti-MHC antibodies might explain their inhibition of HTLV-1 fusion. An anti-class I MHC antibody and a polyclonal antibody consisting of several nonblocking MAbs against other molecules bound to MT2 cells at levels similar to those of class II MHC antibodies, and they also blocked HTLV-1 syncytium formation. Dose-response experiments showed that inhibition of HTLV-1 syncytium formation correlated with levels of antibody bound to the surface of infected cells. The results show that HTLV-1 syncytium formation can be blocked by protein crowding or steric effects caused by large numbers of immunoglobulin molecules bound to the surface of infected cells and have implications for the structure of the cellular HTLV-1 receptor(s).Human T-cell lymphotropic virus type 1 (HTLV-1) is a type C retrovirus and the etiologic agent of adult T-cell leukemia (43, 56, 59) and HTLV-1-associated myelopathy or tropical spastic paraparesis (15, 17, 49, 61). Although HTLV-1 shows tropism primarily for T cells, it can infect a variety of cell types including cells from some nonhuman species (6, 9, 27, 46, 48, 60, 62). Infection by free HTLV-1 tends to be highly inefficient, and the virus appears to be transmitted primarily by the cell-to-cell route (37). The HTLV-1 envelope glycoprotein is synthesized as a 61-kDa precursor which is cleaved into surface (gp46) and transmembrane (gp21) proteins (40, 57). gp46 is thought to serve as the virus attachment protein, as does gp120 for human immunodeficiency virus (HIV) (40, 57). Although previous reports have identified host cell molecules which might potentially mediate virus binding (9, 14), the cellular receptor for HTLV-1 has not been definitively identified. A recent study in which affinity chromatography was carried out with a gp46 peptide has provided evidence that the heat shock protein HSC70 binds directly to gp46 and may serve as a virus receptor (47).gp21 contains an N-terminal hydrophobic fusion domain and likely serves as a fusion protein similar to HIV gp41 (12, 61). Like many other retroviruses, HTLV-1 can induce syncytium formation between infected cells and certain uninfected cell types (28, 39). However, there are no data to indicate that virus transmission or virus persistence in vivo depends on syncytium formation. It is thought that cell-cell fusion involves the same receptors and occurs in a manner similar to virus-cell fusion. For this reason, HTLV-1 syncytium assays have been used to screen for cell surface molecules that may serve as virus receptors (13, 14, 25, 29). Monoclonal antibodies (MAbs) against a number of membrane proteins including members of the tetraspanner family (30, 31) have been found to block syncytium formation. My colleagues and I recently reported that expression of the cell adhesion molecule vascular cell adhesion molecule 1 (VCAM-1) on uninfected cells can confer sensitivity to HTLV-1-mediated syncytium formation (25). In this previous study, we were not able to block HTLV-1 cell fusion with MAbs against the major VCAM-1 counterreceptor VLA-4 (25). Others have reported that MAbs to other adhesion molecules including intercellular adhesion molecule 3 (ICAM-3) also block HTLV-1 syncytium formation (29). We have demonstrated that adhesion molecules also facilitate HIV type 1 (HIV-1) infection and syncytium formation (16, 24). Thus, adhesion molecules may be important accessory molecules for retroviruses generally.Earlier studies on accessory molecules involved in HTLV-1 biology have been extended by immunizing mice with HTLV-1-infected cells and screening for MAbs that block VCAM-1-supported HTLV-1 syncytium formation. Four new MAbs that completely block HTLV-1-mediated cell fusion have been generated. The MAbs were all determined to be specific for class II major histocompatibility complex (MHC) molecules. These MAbs had no effect on syncytium formation induced by HIV-1. Studies on the mechanism by which the MAbs mediate this effect have revealed a novel mode of antibody blockade of virus-induced cell fusion: protein crowding at the infected cell surface resulting in steric blockade of critical receptor-ligand interactions.  相似文献   

5.
Most individuals infected with human immunodeficiency virus type 1 (HIV-1) initially harbor macrophage-tropic, non-syncytium-inducing (M-tropic, NSI) viruses that may evolve into T-cell-tropic, syncytium-inducing viruses (T-tropic, SI) after several years. The reasons for the more efficient transmission of M-tropic, NSI viruses and the slow evolution of T-tropic, SI viruses remain unclear, although they may be linked to expression of appropriate chemokine coreceptors for virus entry. We have examined plasma viral RNA levels and the extent of CD4+ T-cell depletion in SCID mice reconstituted with human peripheral blood leukocytes following infection with M-tropic, dual-tropic, or T-tropic HIV-1 isolates. The cell tropism was found to determine the course of viremia, with M-tropic viruses producing sustained high viral RNA levels and sparing some CD4+ T cells, dual-tropic viruses producing a transient and lower viral RNA spike and extremely rapid depletion of CD4+ T cells, and T-tropic viruses causing similarly lower viral RNA levels and rapid-intermediate rates of CD4+ T-cell depletion. A single amino acid change in the V3 region of gp120 was sufficient to cause one isolate to switch from M-tropic to dual-tropic and acquire the ability to rapidly deplete all CD4+ T cells.The envelope gene of human immunodeficiency virus type 1 (HIV-1) determines the cell tropism of the virus (11, 32, 47, 62), the use of chemokine receptors as cofactors for viral entry (4, 17), and the ability of the virus to induce syncytia in infected cells (55, 60). Cell tropism is closely linked to but probably not exclusively determined by the ability of different HIV-1 envelopes to bind CD4 and the CC or the CXC chemokine receptors and initiate viral fusion with the target cell. Macrophage-tropic (M-tropic) viruses infect primary cultures of macrophages and CD4+ T cells and use CCR5 as the preferred coreceptor (2, 5, 15, 23, 26, 31). T-cell-tropic (T-tropic) viruses can infect primary cultures of CD4+ T cells and established T-cell lines, but not primary macrophages. T-tropic viruses use CXCR4 as a coreceptor for viral entry (27). Dual-tropic viruses have both of these properties and can use either CCR5 or CXCR4 (and infrequently other chemokine receptors [25]) for viral entry (24, 37, 57). M-tropic viruses are most frequently transmitted during primary infection of humans and persist throughout the duration of the infection (63). Many, but not all, infected individuals show an evolution of virus cell tropism from M-tropic to dual-tropic and finally to T-tropic with increasing time after infection (21, 38, 57). Increases in replicative capacity of viruses from patients with long-term infection have also been noted (22), and the switch to the syncytium-inducing (SI) phenotype in T-tropic or dual-tropic isolates is associated with more rapid disease progression (10, 20, 60). Primary infection with dual-tropic or T-tropic HIV, although infrequent, often leads to rapid disease progression (16, 51). The viral and host factors that determine the higher transmission rate of M-tropic HIV-1 and the slow evolution of dual- or T-tropic variants remain to be elucidated (4).These observations suggest that infection with T-tropic, SI virus isolates in animal model systems with SCID mice grafted with human lymphoid cells or tissue should lead to a rapid course of disease (1, 8, 4446). While some studies in SCID mice grafted with fetal thymus and liver are in agreement with this concept (33, 34), our previous studies with the human peripheral blood leukocyte-SCID (hu-PBL-SCID) mouse model have shown that infection with M-tropic isolates (e.g., SF162) causes more rapid CD4+ T-cell depletion than infection with T-tropic, SI isolates (e.g., SF33), despite similar proviral copy numbers, and that this property mapped to envelope (28, 41, 43). However, the dual-tropic 89.6 isolate (19) caused extremely rapid CD4+ T-cell depletion in infected hu-PBL-SCID mice that was associated with an early and transient increase in HIV-1 plasma viral RNA (29). The relationship between cell tropism of the virus isolate and the pattern of disease in hu-PBL-SCID mice is thus uncertain. We have extended these studies by determining the kinetics of HIV-1 RNA levels in serial plasma samples of hu-PBL-SCID mice infected with primary patient isolates or laboratory stocks that differ in cell tropism and SI properties. The results showed significant differences in the kinetics of HIV-1 replication and CD4+ T-cell depletion that are determined by the cell tropism of the virus isolate.  相似文献   

6.
7.
The high-affinity in vivo interaction between soluble HIV-1 envelope glycoprotein (Env) immunogens and primate CD4 results in conformational changes that alter the immunogenicity of the gp120 subunit. Because the conserved binding site on gp120 that directly interacts with CD4 is a major vaccine target, we sought to better understand the impact of in vivo Env-CD4 interactions during vaccination. Rhesus macaques were immunized with soluble wild-type (WT) Env trimers, and two trimer immunogens rendered CD4 binding defective through distinct mechanisms. In one variant, we introduced a mutation that directly disrupts CD4 binding (368D/R). In the second variant, we introduced three mutations (423I/M, 425N/K, and 431G/E) that disrupt CD4 binding indirectly by altering a gp120 subdomain known as the bridging sheet, which is required for locking Env into a stable interaction with CD4. Following immunization, Env-specific binding antibody titers and frequencies of Env-specific memory B cells were comparable between the groups. However, the quality of neutralizing antibody responses induced by the variants was distinctly different. Antibodies against the coreceptor binding site were elicited by WT trimers but not the CD4 binding-defective trimers, while antibodies against the CD4 binding site were elicited by the WT and the 423I/M, 425N/K, and 431G/E trimers but not the 368D/R trimers. Furthermore, the CD4 binding-defective trimer variants stimulated less potent neutralizing antibody activity against neutralization-sensitive viruses than WT trimers. Overall, our studies do not reveal any potential negative effects imparted by the in vivo interaction between WT Env and primate CD4 on the generation of functional T cells and antibodies in response to soluble Env vaccination.The HIV-1 Envs mediate the entry of the virus into target cells and are the only virally encoded proteins exposed on the surface of the virus. HIV-1 Env is the sole target for neutralizing antibodies (Abs) and therefore is an important component of a vaccine designed to elicit protective antibody responses (4, 20). The viral spike is a trimer comprised of three heterodimers of the exterior envelope glycoprotein, gp120, noncovalently attached to the transmembrane protein, gp41. The gp120 subunit binds the primary receptor, CD4 (7), to form or to expose the gp120 coreceptor binding elements, which interact with the viral coreceptor, primarily CCR5 (1, 9, 12, 45). The highly conserved coreceptor binding site (CoRbs) overlaps the gp120 bridging sheet and also contains both proximal and distal elements of V3 (18, 32, 43, 45).In attempts to mimic the native trimeric structure of Env present on the virus, various forms of soluble Env trimers were designed (reviewed in reference 14). One design consists of cleavage-defective trimers derived from the primary R5 isolate YU2 that possess a heterologous trimerization motif derived from T4 bacteriophage fibritin (F; YU2 gp140-F) (3, 21, 34, 40, 50, 51). We recently demonstrated that the immunization of monkeys, but not rabbits, with gp140-F trimers resulted in the generation of Abs directed against the CoRbs of gp120 capable of cross-neutralizing HIV-2 (15). CoRbs-directed Abs (also referred to as CD4-induced, or CD4i, Abs) also were elicited in rabbits transgenic for human CD4 (15). Taken together, these data strongly suggest that Env interacts with high-affinity primate CD4 in vivo, resulting in the formation, or exposure, of a highly immunogenic gp120 determinant that overlaps the CoRbs. Early in infection, the frequency of HIV-1-infected individuals with significant antibody responses against the CoRbs is high (8, 33), and CoRbs-directed antibody responses are elicited abundantly in humans inoculated with Env-based immunogens (15). Collectively, these data suggest that the recognition of the HIV-1 CoRbs by naïve B cells is greatly increased when Env is presented in complex with high-affinity primate CD4, leading to a productive Ab response against this epitope (41). With rare exceptions, the majority of CoRbs-directed monoclonal antibodies (MAbs) do not neutralize HIV-1 primary viruses in vitro, bringing into question the utility of this region as a relevant neutralization target (23, 31, 47, 49). Strategies aimed to diminish vaccine-elicited B-cell responses to the CoRbs, and shift responses toward more accessible neutralization targets, represent one approach to improve the design of Env-based vaccine candidates. The selective manipulation of Env immunogens to decrease their CD4 binding capacity may reduce the elicitation of CoRbs-directed Abs and circumvent potential occlusion effects of the conserved CD4 binding site caused by CD4 itself.In addition to the potential effects of in vivo Env-CD4 interactions on the Ab repertoire elicited by Env-based immunogens, interactions between Env and CD4 also may have consequences on CD4+ T-cell responses. CD4 is an important costimulatory molecule expressed on several subsets of T cells and antigen-presenting cells, and interactions with Env were shown to alter the function of CD4-expressing T cells in a number of in vitro systems (13, 37, 44). The elimination of the Env-CD4 interaction in the context of vaccination may be beneficial to improve the elicitation of helper T-cell responses and effective neutralizing Ab responses. In vivo evaluation in subjects possessing high-affinity CD4 (i.e., rhesus macaques or humans) of CD4 binding-competent and CD4 binding-deficient Env immunogens so far have not been described.To address these questions, we designed Env trimer variants rendered CD4 binding defective through two distinct mechanisms. In the first variant, the interaction between CD4 and HIV-1 Env was directly disrupted by the introduction of a mutation (368D/R) in the CD4 binding loop of the gp120 outer domain (29). This alteration abolishes the initial binding of CD4 and most CD4 binding site (CD4bs)-directed MAbs (42) to variant forms of gp120 and would be expected to do the same in the soluble stable timer context. The aim of the second variant was to decrease the CD4 binding affinity while preserving the antigenicity of the CD4bs (48). This variant was generated in the soluble gp140-F trimers by the introduction of three point mutations, 423I/M, 425N/K, and 431G/E, in the β20 strand region of gp120. These mutations were suggested to favor a helix rather than the β20/21 antiparallel strands visible in the gp120 structure (23, 31, 47, 49). In the monomeric context, mutations in the β20 strand region of gp120 abolish binding by CoRbs-directed Abs, presumably because the bridging sheet cannot form (48). The introduction of the 423I/M, 425N/K, and 431G/E mutations in the trimer context therefore should disrupt the normally high-affinity gp120-CD4 interaction, while recognition by CD4bs Abs would not be affected. Indeed, a recent study provides a mechanistic basis for the impact of these mutations on CD4 binding (52). This study revealed that CD4 interacts with gp120 by a two-step binding mechanism in which the first step involves a direct, but low-affinity, CD4 interaction with the gp120 outer domain, while the second step requires a conformational change in gp120 to fully stabilize the high-affinity gp120-CD4 interaction.Here, we exploit this two-step model to generate novel CD4 binding-defective soluble trimers that, unlike the 368D/R trimers, possess a CD4bs surface that retains recognition by well-described CD4bs Abs. By immunizing rhesus macaques with the wild-type (WT) and CD4 binding-defective trimer variants, we demonstrate that similar levels of Env-specific Ab and T-cell responses were elicited in the three groups, suggesting that in vivo interactions between CD4 binding-competent (WT) Env and CD4 do not measurably affect T-cell responses against Env in this immunization regimen. However, the quality of the Ab response was markedly different between the groups. As hypothesized, CoRbs-directed Abs were elicited only in animals inoculated with WT trimers and not in those inoculated with 368D/R or 423I/M, 425N/K, and 431G/E trimers (hereafter referred to as 368 and 423/425/431 trimers, respectively). Importantly, we show that the 423/425/431 trimers retain the capacity to elicit binding and neutralizing CD4bs-directed Abs. In conclusion, the results generated in this study suggest that CD4 engagement by the WT soluble Env trimers did not impair the overall magnitude of the elicited Env-specific antibody or T-cell responses. Furthermore, our data provide new insights into the characteristics of Env that impact immunogenicity. The data also provide a potential path forward for the design of Env immunogens that have the capacity to elicit neutralizing Abs against the conserved gp120 CD4 binding surface while eliminating both the elicitation of nonneutralizing CoRbs-directed Abs and the potential occlusion of the CD4 binding surface of gp120 by the engagement of the primary virus receptor, CD4.  相似文献   

8.
9.
10.
We have investigated the cellular uptake of Gag p24 shortly after exposure of cells to human immunodeficiency virus (HIV) particles. In the absence of envelope glycoprotein on virions or of viral receptors or coreceptors at the cell surface, p24 was incorporated in intracellular vesicles but not detected in the cytosolic subcellular fraction. When appropriate envelope-receptor interactions could occur, the nonspecific vesicular uptake was still intense and cytosolic p24 represented 10 to 40% of total intracellular p24. The measurement of cytosolic p24 early after exposure to HIV type 1 is a reliable assay for investigating virus entry and early events leading to authentic cell infection.The entry of human immunodeficiency virus type 1 (HIV-1) into target cells follows receptor-mediated attachment of viral particles to the cell surface. The cell surface receptor for HIV-1 is the CD4 molecule (7, 15), which promotes attachment of the particle to the cell surface. Fusion between the viral and plasma membranes leading to virus entry into the cytoplasm also requires interaction with a coreceptor. Various chemokine receptors ensure this function. The CXCR4 receptor is used by lymphotropic virus strains (10), whereas the entry of macrophage-tropic and of most primary isolates is processed through interaction with the CCR5 receptor (8, 9). Interactions with CD4 and with a coreceptor expose highly hydrophobic epitopes at the N terminus of the gp41 transmembrane component of envelope, leading to subsequent fusion between viral and cell membranes (6, 17, 34, 35).Several observations have suggested that the fusion process takes place at the cell surface: (i) HIV infection is pH independent, whereas infection by most viruses entering through the endocytic pathway is inhibited by weak bases and ionophore agents (20, 32); (ii) HIV fusion images have been observed at the cell surface (11); (iii) endocytosis of CD4 is not required for entry (18, 20, 25, 28, 32); and (iv) mutant CXCR5 receptors which are not endocytosed in response to ligand binding still function as HIV coreceptors (2). However, other considerations led to the assumption that although HIV entry is clearly pH independent, it may not necessarily be endocytosis independent: (i) images of HIV particles internalized in endocytic vesicles and undergoing fusion with endosomal membranes have been observed (11, 27), (ii) pH-independent entry via endosomal vesicles has been reported for poliovirus (29), (iii) binding and cross-linking by multivalent virus particles may induce endocytic behavior of cell surface receptors different from that induced by their natural ligands, and (iv) endocytosis of CD4 and that of coreceptors have not been simultaneously examined after HIV exposure. Moreover, since studies of virus entry have been performed with cells where the endocytic pathway is active, it is difficult to determine whether particular fusion events at the cell surface or in endosomal vesicles give rise to productive infection.With the aim of examining the role of endosomal HIV particle uptake, infection was synchronized by exposing cells to the virus at 4°C, cells were warmed at 37°C, and p24 was measured in the vesicular and cytosolic fractions of cell extracts. p24 was detected in intracellular vesicles regardless of whether exposure to virus particles could give rise to authentic infection or not. On the other hand, the detection of p24 in cytosolic fractions was strictly associated with authentic infectious events. However, it represented a minor fraction of intracellular p24. Thus, although vesicular uptake is quantitatively the main route of virus particle internalization, it is essentially a dead end with respect to cell infection.  相似文献   

11.
12.
Human immunodeficiency virus infection is characterized by a progressive decline in the number of peripheral blood CD4+ T lymphocytes, which finally leads to AIDS. This T-cell decline correlates with the degree of in vitro-induced lymphocyte apoptosis. However, such a correlation has not yet been described in feline AIDS, caused by feline immunodeficiency virus (FIV) infection. We therefore investigated the intensity of in vitro-induced apoptosis in peripheral blood lymphocytes from cats experimentally infected with a Swiss isolate of FIV for 1 year and for 6 years and from a number of long-term FIV-infected cats which were coinfected with feline leukemia virus. Purified peripheral blood lymphocytes were either cultured overnight under nonstimulating conditions or stimulated with phytohemagglutinin and interleukin-2 for 60 h. Under stimulating conditions, the isolates from the infected cats showed significantly higher relative counts of apoptotic cells than did those from noninfected controls (1-year-infected cats, P = 0.01; 6-year-infected cats, P = 0.006). The frequency of in vitro-induced apoptosis was inversely correlated with the CD4+ cell count (P = 0.002), bright CD8+ cell count (P = 0.009), and CD4/CD8 ratio (P = 0.01) and directly correlated with the percentage of bright major histocompatibility complex class II-positive peripheral blood lymphocytes (P = 0.004). However, we found no correlation between in vitro-induced apoptosis and the viral load in serum samples. Coinfection with feline leukemia virus enhanced the degree of in vitro-induced apoptosis compared with that in FIV monoinfected cats. We concluded that the degree of in vitro-induced apoptosis was closely related to FIV-mediated T-cell depletion and lymphocyte activation and could be used as an additional marker for disease progression in FIV infection.Feline immunodeficiency virus (FIV) infection is a naturally occurring infection, and disease progression in infected cats is associated with a decline in the number of CD4+ cells (2, 6, 22, 23, 36), a loss of bright CD8+ cells in the advanced stage of the disease (22), an increased number of activated T cells (39, 41), and a changed cytokine production, i.e., decreased production of interleukin-2 (IL-2) and concomitantly increased production of tumor necrosis factor alpha (TNF-α) (25, 26). The increased production of TNF-α has been reported to induce apoptosis in chronically FIV-infected cells (38). Apoptosis, a controlled mode of cell death (34), plays an important role in the regulation of immune responses (5, 14). As described for FIV (23), the hallmark of human immunodeficiency virus (HIV)-induced disease is the loss of T-helper cells (31, 43). Theoretically, cell loss can be caused by decreased production of cells, increased destruction, or a combination of the two mechanisms. Findings of an early infection of thymocytes followed by pathologic changes in the thymus support the model of decreased T-helper cell production triggered by HIV (13) and FIV (52). The destruction model is supported by findings of an increased number of peripheral blood T cells undergoing apoptosis upon HIV (20, 32) and FIV (11, 21, 33) infection. However, increased CD4+-T-cell turnover may not be the main cause of the observed T-helper cell decline in HIV-1 infection, as reviewed by others (44, 51). In addition, the degree of HIV-induced apoptosis correlates with the T-helper cell decline and disease progression (19, 40). However, such a relationship has not yet been described for FIV. It has been reported that cross-linking of CD4 molecules by HIV gp160 triggers apoptosis in noninfected CD4+ T cells (1). Investigation of this aspect in the feline system is especially interesting since FIV does not use the feline homologue of CD4 (50).The aim of the present study was to compare the degree of in vitro-induced lymphocyte apoptosis in FIV-infected cats with normal and decreased T-helper cell counts. We used two different culture conditions to trigger apoptosis in vitro: cells were either cultured overnight under nonstimulating conditions and in the absence of growth factors or cultured for 60 h in the presence of phytohemagglutinin, IL-2, and fetal calf serum. We additionally examined cats which were coinfected with the feline leukemia virus (FeLV). This coinfection is known to accelerate the progression toward feline AIDS (23) by an unknown mechanism (8).  相似文献   

13.
14.
A decoding algorithm is tested that mechanistically models the progressive alignments that arise as the mRNA moves past the rRNA tail during translation elongation. Each of these alignments provides an opportunity for hybridization between the single-stranded, -terminal nucleotides of the 16S rRNA and the spatially accessible window of mRNA sequence, from which a free energy value can be calculated. Using this algorithm we show that a periodic, energetic pattern of frequency 1/3 is revealed. This periodic signal exists in the majority of coding regions of eubacterial genes, but not in the non-coding regions encoding the 16S and 23S rRNAs. Signal analysis reveals that the population of coding regions of each bacterial species has a mean phase that is correlated in a statistically significant way with species () content. These results suggest that the periodic signal could function as a synchronization signal for the maintenance of reading frame and that codon usage provides a mechanism for manipulation of signal phase.[1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32]  相似文献   

15.
Peptides based on the second heptad repeat (HR2) of viral class I fusion proteins are effective inhibitors of virus entry. One such fusion inhibitor has been approved for treatment of human immunodeficiency virus-1 (T20, enfuvirtide). Resistance to T20 usually maps to the peptide binding site in HR1. To better understand fusion inhibitor potency and resistance, we combined virological, computational, and biophysical experiments with comprehensive mutational analyses and tested resistance to T20 and second and third generation inhibitors (T1249 and T2635). We found that most amino acid substitutions caused resistance to the first generation peptide T20. Only charged amino acids caused resistance to T1249, and none caused resistance to T2635. Depending on the drug, we can distinguish four mechanisms of drug resistance: reduced contact, steric obstruction, electrostatic repulsion, and electrostatic attraction. Implications for the design of novel antiviral peptide inhibitors are discussed.The HIV-1 envelope glycoprotein complex (Env),3 a class I viral fusion protein, is responsible for viral attachment to CD4+ target T cells and subsequent fusion of viral and cellular membranes resulting in release of the viral core in the cell. Other examples of viruses using class I fusion proteins are Coronaviridae (severe acute respiratory syndrome virus), Paramyxoviridae (Newcastle disease virus, human respiratory syncytial virus, Nipah virus, Hendra virus), and Orthomyxoviridae (influenza virus), some of which cause fatal diseases in humans (13). The entry process of these viruses is an attractive target for therapeutic intervention.The functional trimeric Env spike on HIV-1 virions consists of three gp120 and three gp41 molecules that are the products of cleavage of the precursor gp160 by cellular proteases such as furin (4, 5). The gp120 surface subunits are responsible for binding to the cellular receptors, whereas the gp41 subunits anchor the complex in the viral membrane and mediate the fusion of viral and cellular membranes. Env undergoes several conformational changes that culminate in membrane fusion. The gp120 subunit binds the CD4 receptor, resulting in creation and/or exposure of the binding site for a coreceptor, usually CCR5 or CXCR4 (6, 7). Two α-helical leucine zipper-like motifs, heptad repeat 1 (HR1) and heptad repeat 2 (HR2), located in the extracellular part of gp41, play a major role in the following conformational changes. Binding of the receptors to gp120 induces formation of the pre-hairpin intermediate of gp41 in which HR1 is exposed and the N-terminal fusion peptide is inserted into the target cell membrane (1, 812). Subsequently, three HR1 and three HR2 domains assemble into a highly stable six-helix bundle structure that juxtaposes the viral and cellular membranes for the membrane merger. Other viruses with class I viral fusion proteins use similar HR1-HR2-mediated membrane fusion for target cell entry.Peptides based on the HR domains of class I viral fusion proteins have proven to be efficient inhibitors of virus entry for a broad range of viruses (1317). The HIV-1 fusion inhibitor T20 (enfuvirtide (Fuzeon)) has been approved for clinical use. T20 mimics HR2 and can bind to HR1, thereby preventing the formation of the six-helix bundle (Fig. 1) (1821). T1249 is a second-generation fusion inhibitor with improved antiviral potency compared with the first-generation peptide T20 (2225). Recently, a series of more potent third-generation fusion inhibitors were designed (26, 27). These include T2635, which has an improved helical structure that increases stability and activity against both wild type (WT) HIV-1 and fusion inhibitor resistant variants.Open in a separate windowFIGURE 1.Schematic of the gp41 ectodomain. HR1 and HR2 are represented as cylinders, and position 38 in HR1 is indicated. Residues Gln-142, Asn-145, Glu-146, and Leu-149, which interact with residue 38, are underlined in the HR2 sequence. HR2-based peptide fusion inhibitors are shown underneath. Mutations introduced in T1249mut and T2635mut are bold and underlined. Numbering is based on the sequence of HXB2 gp41.Both the in vitro and in vivo selection of resistance has been described for T20 (2833) and T1249 (23, 3436). Resistance is often caused by mutations in the HR1 binding site of the fusion inhibitor. In particular, substitutions at positions 36 (G36D/M/S), 38 (V38A/W/M/E), and 43 (N43D/K) of gp41 can cause resistance. Strikingly, substitutions at position 38 can cause resistance to both T20 and T1249, but distinct amino acid substitutions are required. At position 38 only charged amino acids (V38E/R/K) cause resistance to T1249 (35). Surprisingly, none of the known T20 and T1249 resistance mutations at position 38 affect the susceptibility to the third generation inhibitor T2635.We hypothesized that the use of HIV-1 as a model system could provide a more detailed understanding of resistance to fusion inhibitors. We, therefore, analyzed the effect of all 20 amino acids at resistance hotspot 38 on Env function, viral fitness, biochemical properties of gp41, and resistance to the fusion inhibitors. From the results we can propose four resistance mechanisms that differ in the way the drug-target interaction is affected at the molecular level. Furthermore, we can deduce general principles on the mechanisms of resistance against fusion inhibitors and the requirements for effective antiviral drugs.  相似文献   

16.
DC-SIGN is a C-type lectin receptor of dendritic cells and is involved in the early stages of numerous infectious diseases. DC-SIGN is organized into a tetramer enabling multivalent interaction with pathogens. Once formed, the DC-SIGN-pathogen complex can be internalized into compartments of increasing acidity. We have studied the pH dependence of the oligomerization state and conformation of the entire extracellular domain and neck region. We present evidence for equilibrium between the monomeric and tetrameric states of the extracellular domain, which exhibits a marked dependence with respect to both pH and ionic strength. Using solution x-ray scattering we have obtained a molecular envelope of the extracellular domain in which a model has been built. Our results highlight the central role of the neck domain in the pH-sensitive control of the oligomerization state, in the extended conformation of the protein, and in carbohydrate recognition domain organization and presentation. This work opens new insight into the molecular mechanism of ligand release and points to new avenues to block the first step of this important infection pathway.Dendritic cells (DCs),2 which are found in peripheral tissues and act as sentinels against invading pathogens are considered to be the most efficient professional antigen presenting cells identified so far (1). A major subset of these DCs, dermal DCs, is characterized by the membrane expression of the DC-SIGN (dendritic cell-specific ICAM3 grabbing non-integrin) receptor (CD209). DC-SIGN is a calcium-dependent (C-type) lectin able to recognize highly glycosylated proteins. It is implicated in the early stages of many viral infections (2) and is spatially distributed in well defined membranous microdomains with an average diameter of 200 nm that act as docking platforms for pathogens and endogenous antigen attachment (3, 4). DC-SIGN binds to viral pathogens through their exposed glycoproteins such as HIV-1 envelope protein (gp120) (5), GP1 of Ebola (6), E1 and E2 of HCV (7, 8), and Dengue virus E glycoprotein (9). Apart from the viral world, DC-SIGN has also been implicated in infection processes involving fungi (10, 11), bacteria, such as Neisseria meningitidis (12) and Leishmania pifanoi (13), and parasites, such as Schistosoma mansoni (14). In addition, DC-SIGN mediates DC contacts with endothelial cells, naive T lymphocytes, and neutrophils by interacting with the endogenous adhesion molecules ICAM2 (15), ICAM3 (16), and the CD11b/CD18 integrin (17), respectively.DC-SIGN is a type II membrane protein comprising three main domains: a cytoplasmic region, a transmembrane segment, and an extracellular domain (ECD). The ECD can be divided into two structurally and functionally distinct regions: a neck region involved in the tetramerization of the receptor and a calcium-dependent carbohydrate-recognition domain (CRD), which is at the heart of the molecular recognition processes mediated by DC-SIGN.The cytoplasmic region contains recycling and internalization motifs important for targeting receptors, together with their associated ligands, to subcellular compartments as shown for DEC 205 and DC-SIGN itself (18, 19). Indeed, in some conditions, combined deletion of the triacidic cluster (EEE), the dileucine (LL), and the tyrosine-based (YXXL) internalization motifs induces the loss of DC capacity to enhance T-cell HIV-1 infection (19). The ability of DC-SIGN to promote T-cell infection by HIV-1 involves different pathways depending on the kinetics of the infection process. HIV transfers from DCs to T cells have been described to occur in two phases: a short-term phase (up to 24 h after viral exposure) and a long-term phase (from 24 to 72 h) (20, 21). The long-term phase is proposed to involve membrane fusion and subsequent DC infection and does not need to concern us further in this study. In contrast, short-term phase transfer involves virion internalization into DCs through vesicles. A pH dependence of HIV transfer has been suggested, based on the abrogation of the trans-enhancement of T-cell infection upon intracellular pH neutralization by concanamycin A (19). Indeed, internalization of HIV particles into low pH non-lysosomal compartments has been described in DC-SIGN-expressing cells (19). At the molecular level, a pH dependence of DC-SIGN binding to gp120 has been reported (22), as well as ligand release from DC-SIGN in pH conditions mimicking the acidic luminal environment of endosomes/lysosomes (23). However, no molecular explanation has yet been proposed for this phenomenon that might play a key role in the initiation mechanisms of DC-SIGN-mediated infectious processes. An effect of pH on the CRD binding properties has been reported (22) but no detailed molecular study of the pH dependence of the oligomerization properties of the neck domain, essential for the integrity of ECD, has yet been reported.The neck region comprises seven complete and one incomplete 23-amino acid long repeat. These repeats exhibit a distinct pattern of hydrophobic residues, positioned at regular intervals along an α-helical stretch that could favor coiled-coil interaction between helices from the four chains within a tetramer (24, 25). Comparison of circular dichroism spectra from the entire ECD and the CRD fragment led to a similar suggestion (26). The neck domain, which is responsible for tetramer formation and stability of the whole ECD, is thereby at the onset of the avidity mechanism that endows the DC-SIGN receptor with a very high affinity toward its target as compared with the weak affinity of isolated CRD. Indeed, the dissociation constant of the complex with the HIV gp120 envelope protein is in the nanomolar range with the whole ECD, whereas it is in the millimolar range when using monomeric CRD (22). Moreover, DC-SIGN neck region variants, resulting from alternative splicing and genomic polymorphism, exhibit altered sugar binding (27). DC-SIGN neck polymorphism has recently been correlated with differential susceptibility to HIV-1 and other pathogens as a result of modified DC-SIGN multimerization on the cell surface (27). Beyond the avidity mechanism already mentioned, the association of the four chains within the neck largely constraints the CRD spatial arrangement that must bear on the ligand interaction and recognition properties. However, despite the importance of the DC-SIGN tetrameric state in molecular recognitions, structural information is scarce and mostly restricted to the CRD moiety. High-resolution structures of the CRD in complex with various carbohydrate ligands are available (2831). The partial structure of a repeat has been obtained using a truncated dimeric form of the related homolog, DC-SIGNR (29, 32). However, structural information on the tetramer is still lacking although it is a prerequisite to a precise understanding of CRD presentation, pathogen recognition, and to rationalize emerging strategies for the design of DC-SIGN multivalent inhibitors (3336).Our study of the ECD, and more particularly of the neck domain, addresses two issues. First of all, we investigate the role of the neck domain in the structural organization of DC-SIGN ECD. We analyze its stability and dynamic as a function of pH conditions encountered in the various cell compartments. Our studies involve various constructs (CRD, ECD, and neck region) using several techniques: analytical ultracentrifugation, size exclusion chromatography (SEC), and circular dichroism (CD). We show that physiologically encountered pH variations significantly affect the protein oligomerization and structure. Slightly acidic pH destabilizes the tetramer as well as the CRD structure with possible consequences on the strength of ligand binding, which is strongly dependent on the multiplicity of interactions. In parallel, a molecular envelope of DC-SIGN ECD derived from small angle x-ray scattering (SAXS) measurements in solution highlights the extended organization of DC-SIGN and suggests a tighter arrangement of the four CRDs than hereto proposed.  相似文献   

17.
18.
19.
We have investigated whether the identity of the coreceptor (CCR5, CXCR4, or both) used by primary human immunodeficiency virus type 1 (HIV-1) isolates to enter CD4+ cells influences the sensitivity of these isolates to neutralization by monoclonal antibodies and CD4-based agents. Coreceptor usage was not an important determinant of neutralization titer for primary isolates in peripheral blood mononuclear cells. We also studied whether dualtropic primary isolates (able to use both CCR5 and CXCR4) were differentially sensitive to neutralization by the same antibodies when entering U87MG-CD4 cells stably expressing either CCR5 or CXCR4. Again, we found that the coreceptor used by a virus did not greatly affect its neutralization sensitivity. Similar results were obtained for CCR5- or CXCR4-expressing HOS cell lines engineered to express green fluorescent protein as a reporter of HIV-1 entry. Neutralizing antibodies are therefore unlikely to be the major selection pressure which drives the phenotypic evolution (change in coreceptor usage) of HIV-1 that can occur in vivo. In addition, the increase in neutralization sensitivity found when primary isolates adapt to growth in transformed cell lines in vitro has little to do with alterations in coreceptor usage.Human immunodeficiency virus type 1 (HIV-1) enters CD4+ T cells via an interaction with CD4 and coreceptor molecules, the most important of which yet identified are the chemokine receptors CXCR4 and CCR5 (4, 12, 23, 26, 28, 32). CXCR4 is used by T-cell line-tropic (T-tropic) primary isolates or T-cell line-adapted (TCLA) lab strains, whereas CCR5 is used by primary isolates of the macrophage-tropic (M-tropic) phenotype (4, 12, 23, 26, 28, 32). Most T-tropic isolates and some TCLA strains are actually dualtropic in that they can use both CXCR4 and CCR5 (and often other coreceptors such as CCR3, Bonzo/STRL33, and BOB/gpr15), at least in coreceptor-transfected cells (18, 24, 30, 54, 89). The M-tropic and T-tropic/dualtropic nomenclature has often been used interchangeably with the terms “non-syncytium-inducing” (NSI) and “syncytium-inducing” (SI), although it is semantically imprecise to do so.M-tropic viruses are those most commonly transmitted sexually (3, 33, 87, 106) and from mother to infant (2, 72, 81). If T-tropic strains are transmitted, or when they emerge, this is associated with a more rapid course of disease in both adults (17, 37, 46, 51, 52, 76, 78, 82, 92, 101) and children (6, 45, 84, 90). However, T-tropic viruses emerge in only about 40% of infected people, usually only several years after infection (76, 78). A well-documented, albeit anecdotal, study found that when a T-tropic strain was transmitted by direct transfer of blood, its replication was rapidly suppressed: the T-tropic virus was eliminated from the body, and M-tropic strains predominated (20). These results suggest that there is a counterselection pressure against the emergence of T-tropic strains during the early stages of HIV-1 infection in most people. But what is this pressure?Since the M-tropic and T-tropic phenotypes are properties mediated by the envelope glycoproteins whose function is to associate with CD4 and the coreceptors, a selection pressure differentially exerted on M- and T-tropic viruses could, in principle, act at the level of virus entry. In other words, neutralizing antibodies to the envelope glycoproteins, or the chemokine ligands of the coreceptors, could theoretically interfere more potently with the interactions of T-tropic strains with CXCR4 than with M-tropic viruses and CCR5. A differential effect of this nature could suppress the emergence of T-tropic viruses. Consistent with this possibility, neutralizing antibodies are capable of preventing the CD4-dependent association of gp120 with CCR5 (42, 94, 103), and chemokines can also prevent the coreceptor interactions of HIV-1 (8, 13, 23, 28, 70).Here, we explore whether the efficiency of HIV-1 neutralization is affected by coreceptor usage. Although earlier studies have not found T-tropic strains to be inherently more neutralization sensitive than M-tropic ones (20, 40, 44), previously available reagents and techniques may not have been adequate to fully address this question. One major problem is that even single residue changes can drastically affect both antibody binding to neutralization epitopes and the HIV-1 phenotype (25, 55, 62, 67, 83, 91), and so studies using relatively unrelated viruses and a fixed antibody (polyclonal or monoclonal) preparation have two variables to contend with: the viral phenotype (coreceptor use) and the antigenic structure of the virus and hence the efficiency of the antibody-virion interaction.We have used a new experimental strategy to explore whether coreceptor usage affects neutralization sensitivity in the absence of other confounding variables: the use of dualtropic viruses able to enter CD4+ cells via either CCR5 or CXCR4. By using a constant HIV-1 isolate or clone and the same monoclonal antibodies (MAbs) or CD4-based reagents as neutralizing agents, we can ensure that the only variable under study in the neutralization reaction is the nature of the coreceptor used for entry. Our major conclusion is that there is no strong association between coreceptor usage and neutralization sensitivity for primary HIV-1 isolates. Independent studies have reached the same conclusion (53a, 59). The emergence of T-tropic (SI) viruses in vivo may be unlikely to be due to escape from antibody-mediated selection pressure.  相似文献   

20.
A Boolean network is a model used to study the interactions between different genes in genetic regulatory networks. In this paper, we present several algorithms using gene ordering and feedback vertex sets to identify singleton attractors and small attractors in Boolean networks. We analyze the average case time complexities of some of the proposed algorithms. For instance, it is shown that the outdegree-based ordering algorithm for finding singleton attractors works in time for , which is much faster than the naive time algorithm, where is the number of genes and is the maximum indegree. We performed extensive computational experiments on these algorithms, which resulted in good agreement with theoretical results. In contrast, we give a simple and complete proof for showing that finding an attractor with the shortest period is NP-hard.[1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32]  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