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.
重组蛋白NE2包含了戊型肝炎病毒(HEV)衣壳蛋白(pORF2)的aa394~606片段.在NE2上已鉴定出了2个HEV中和表位,并获得了3个识别中和表位的单克隆抗体(MAb)8C11、13D8和8H3.这3个MAb间的交叉阻断ELISA实验发现,8C11和13D8可以彼此完全阻断,8H3对8C11和13D8均不能阻断,而8C11非但不能阻断8H3,反而显著增强了8H3与抗原的结合.用生物传感器进行的抗体与抗原结合的动力学分析也证实了这一现象.这些结果提示,在NE2上8H3表位区域受到抗原上某些结构的掩盖,而8C11与NE2的结合引起了抗原空间结构的改变,导致了掩盖8H3表位的结构的去除和8H3表位的充分暴露.免疫捕获RT-PCR发现,8C11同样可以显著增强8H3对天然HEV病毒的捕获能力,提示这种结合诱导的衣壳蛋白空间构象改变在天然HEV病毒颗粒上同样存在.  相似文献   

2.
The henipaviruses, represented by Hendra (HeV) and Nipah (NiV) viruses are highly pathogenic zoonotic paramyxoviruses with uniquely broad host tropisms responsible for repeated outbreaks in Australia, Southeast Asia, India and Bangladesh. The high morbidity and mortality rates associated with infection and lack of licensed antiviral therapies make the henipaviruses a potential biological threat to humans and livestock. Henipavirus entry is initiated by the attachment of the G envelope glycoprotein to host cell membrane receptors. Previously, henipavirus-neutralizing human monoclonal antibodies (hmAb) have been isolated using the HeV-G glycoprotein and a human naïve antibody library. One cross-reactive and receptor-blocking hmAb (m102.4) was recently demonstrated to be an effective post-exposure therapy in two animal models of NiV and HeV infection, has been used in several people on a compassionate use basis, and is currently in development for use in humans. Here, we report the crystal structure of the complex of HeV-G with m102.3, an m102.4 derivative, and describe NiV and HeV escape mutants. This structure provides detailed insight into the mechanism of HeV and NiV neutralization by m102.4, and serves as a blueprint for further optimization of m102.4 as a therapeutic agent and for the development of entry inhibitors and vaccines.  相似文献   

3.
The membrane-proximal external region (MPER) of the human immunodeficiency virus (HIV) envelope glycoprotein (gp41) is critical for viral fusion and infectivity and is the target of three of the five known broadly neutralizing HIV type 1 (HIV-1) antibodies, 2F5, Z13, and 4E10. Here, we report the crystal structure of the Fab fragment of Z13e1, an affinity-enhanced variant of monoclonal antibody Z13, in complex with a 12-residue peptide corresponding to the core epitope (W670NWFDITN677) at 1.8-Å resolution. The bound peptide adopts an S-shaped conformation composed of two tandem, perpendicular helical turns. This conformation differs strikingly from the α-helical structure adopted by an overlapping MPER peptide bound to 4E10. Z13e1 binds to an elbow in the MPER at the membrane interface, making relatively few interactions with conserved aromatics (Trp672 and Phe673) that are critical for 4E10 recognition. The comparison of the Z13e1 and 4E10 epitope structures reveals a conformational switch such that neutralization can occur by the recognition of the different conformations and faces of the largely amphipathic MPER. The Z13e1 structure provides significant new insights into the dynamic nature of the MPER, which likely is critical for membrane fusion, and it has significant implications for mechanisms of HIV-1 neutralization by MPER antibodies and for the design of HIV-1 immunogens.The continued spread of human immunodeficiency virus (HIV) worldwide and, in particular, in sub-Saharan Africa, where an estimated 22 million people currently are living with HIV/AIDS, underscores the urgent need for a preventative vaccine. However, despite nearly 25 years of intense international research, a vaccine is not yet available. Passive immunization with broadly neutralizing antibodies can confer sterilizing protection against infection in animal models (4, 12, 39-41, 51, 64), providing encouragement for the development of an antibody-inducing component of an HIV type 1 (HIV-1) vaccine. Such a vaccine should elicit neutralizing antibodies with activity against the broadest range of primary circulating isolates. However, a lack of understanding of how to raise potent, cross-reactive antibodies by immunization, the so-called neutralizing antibody problem, is a major hurdle in this effort (6, 24, 72). Thus, an understanding of the structure and presentation of neutralizing epitopes on the virus and the antibodies that recognize them is vital for vaccine development.The targets of antibody neutralization are the surface envelope (Env) glycoprotein trimers (gp120/gp41) that mediate the fusion of the viral membrane with that of the host. The majority of antibodies elicited during natural infection or immunization show limited or no cross-reactivity against diverse isolates. However, a few rare, broadly neutralizing, monoclonal antibodies have been isolated from HIV-1-infected individuals and exhibit activity against a wide range of isolates by binding to functionally conserved epitopes exposed on native gp120/gp41 trimers. These epitopes include the CD4 binding site, recognized by antibody b12, and a relatively well-conserved cluster of N-linked glycans, located on the outer domain of gp120, that is recognized by antibody 2G12 (12, 13, 71, 76). V3-directed antibodies, which are common in natural infection, also are able to sporadically neutralize across clades, as exemplified by 447-52D and F425-B4e8 (7, 16, 49, 66). The identification of three broadly neutralizing antibodies, 2F5, Z13, and 4E10, that target the conserved tryptophan-rich membrane-proximal external region (MPER) of gp41 has implicated this region as a highly promising vaccine target and has, therefore, spurred interest in its structural characterization (15, 35, 45, 47, 48, 50, 80).The MPER plays a critical, but not fully understood, role in membrane fusion and is situated between the C-terminal heptad repeat (CHR) and the transmembrane domain (TM) of gp41 (Fig. (Fig.1).1). Following the binding of gp120 to the cell surface receptors CD4 and CXCR4/CCR5, the gp41 glycoprotein undergoes a series of conformational changes that trigger the membrane fusion activity. Notably, a relatively long-lived prehairpin intermediate of gp41 is formed, in which the coiled-coil of the N-terminal heptad repeats (NHR) extends so as to enable the fusion peptides to embed into the target membrane. In the postfusion or fusogenic state, the CHR and NHR reassemble into an antiparallel 6-helix bundle in a process that drives membrane fusion (18). The MPER contains several functionally conserved tryptophan residues that are critical for membrane fusion and viral entry, although the structural basis for their specific role has not been firmly established (22, 44, 58). Their mutation to alanine leads to the attenuation of viral infectivity, which is most pronounced for Trp666 and Trp672 (numbered according to the HXB2 isolate) (46, 58, 78). In addition, peptides based on the MPER can induce membrane leakage (68). Such membrane-disrupting properties of the MPER have been suggested to be functionally important in the expansion of the fusion pore created after receptor engagement (42, 44, 58, 68, 77).Open in a separate windowFIG. 1.Major features of gp41 include the fusion peptide (FP), NHR, CHR, TM, and cytoplasmic domain (CD). The MPER is located between the CHR and TM regions of gp41. The core epitopes of 2F5 (green), Z13e1 (yellow), and 4E10 (orange) are indicated. The epitope of Z13e1 is located between those of 2F5 and 4E10, but it overlaps more closely with 4E10.From initial explorations using solution nuclear magnetic resonance, the structure of a 19-residue MPER peptide (residues 665 to 683) was found to be helical in dodecylphosphocholine micelles, with the hydrophobic and hydrophilic residues distributed evenly around the helix axis (62). Another study found that an MPER peptide comprising residues 659 to 671 adopts a 310-helix in water (10). More recently, the structure of an MPER peptide (residues 662 to 683) in liposomes was elucidated by a combination of nuclear magnetic resonance and spin-label electron paramagnetic resonance (69), and it was found to adopt a kinked, amphipathic structure composed of two helices connected by a short hinge (Phe673 and Asn674). Crystal structures of Fab 2F5 in complex with a 7-mer (E662LDKWAS668) and 17-mer encompassing residues 654 to 670 previously had revealed a mostly extended conformation characterized by a central β-turn involving Asp664, Lys665, and Trp666 (47, 48). This motif is the key recognition determinant for 2F5 and becomes deeply buried in the antibody combining site, suggesting that it is exposed at some stage in viral entry (45, 47, 78). The crystal structure of Fab 4E10 in complex with peptide-spanning residues W670NWFDITNW678 revealed an amphipathic α-helical structure with a narrow hydrophilic face (15). The N terminus of the 4E10 epitope forms a 310-helix that transitions into a regular α-helix at residue Asp674 and continues to Lys683, which constitutes the end of the gp41 ectodomain (14). Thus, while the structure of the MPER within functional, membrane-embedded Env trimers is not known, the observation that unconstrained peptides are able to adopt more than one defined structure suggests an inherent degree of flexibility.Like 4E10, Z13 was identified from an HIV-1-infected individual, the former being isolated from an immortalized B-cell line and the latter from a bone marrow RNA phage display library (80). The epitope of MAb Z13 spans residues S668LWNWFDITN677, as determined by peptide mapping, scanning mutagenesis, and antibody competition studies (46, 80). This region lies between the 2F5 and 4E10 epitopes but overlaps more closely with 4E10 (Fig. (Fig.1).1). 4E10 and Z13 are both able to neutralize primary as well as laboratory-adapted isolates; nevertheless, Z13 is not as broadly neutralizing as 4E10, which has the greatest breadth of any HIV-1 antibody described to date (9). Z13e1 is an affinity-enhanced variant of Z13 and was evolved by randomizing the complementarity determining region (CDR) L3 loop sequence to identify tighter-binding mutants using phage display (46). Z13e1 displays higher affinity for both peptide and recombinant gp41 substrates, as well as increased neutralization potency, suggesting that the L3 mutations optimize binding to the linear MPER epitope. The neutralization breadth of Z13e1 is limited by the requirement for Asn671 and Asp674 in the MPER, which are approximately 71 and 58% conserved, respectively, among sequences in the Los Alamos HIV sequence database (80). Based on the clear relationship between Env trimer binding and neutralization, the neutralizing activity of Z13e1 derives from binding to a functional trimer (8, 20, 25, 43, 52, 55, 60, 73, 74). While Z13e1 and 4E10 have identical affinities for optimized linear peptides, Z13e1 is still about an order of magnitude less potent than 4E10 against a variety of primary isolates. Although the occlusion of the Z13e1 epitope on virion-associated trimers is thought to be the major limitation (46), the structural basis for the lower potency of Z13e1 relative to those of 2F5 and 4E10 is unclear.Whereas neutralization by 4E10 depends critically on Trp672 and Phe673, Z13e1 instead requires the flanking Asn671 and Asp674 residues (46). Based on a helical model of the MPER, it was predicted that Z13e1 binds the narrow hydrophilic face that displays Asn671, Asp674, and Asn677 that is opposite that recognized by 4E10. As Z13e1 and 4E10 bind to functional trimers, both epitopes must be exposed at some stage before membrane fusion (20). To examine how Z13e1 recognizes its MPER epitope, we determined the crystal structure of Fab Z13e1 in complex with a 12-residue peptide corresponding to the core epitope with C-terminal flanking lysines to aid peptide solubility (W670NWFDITN677KKKK). The crystal structure at 1.8-Å resolution uncovers a conformation of the MPER that is distinct from that visualized in complex with 4E10. Our findings show that Z13e1 and 4E10 recognize different conformers of the MPER and reveal a novel conformational switch that is relevant for HIV-1 neutralization and membrane fusion.  相似文献   

4.
Primary bovine embryonic kidney cell was successfully replaced by the green monkey cell line (Vero) for the isolation and seroneutralization of rinderpest virus.  相似文献   

5.
6.
在用噬菌体表面呈现系统获得人源抗甲型肝炎(甲肝)病毒中和性基因工程Fab抗体的基础上,对所获得的4株中和性Fab抗体轻重链可变区基因进行了序列分析、可溶性表达及生物学特性鉴定.4株Fab抗体重链可变区拥有99%同源的核苷酸序列和相同的CDR区氨基酸序列,属于VHⅢ基因家族.而轻链可变区核苷酸序列同源性为95%和相似的CDR区氨基酸序列,属于VL5基因家族.这些重组抗体都能与人甲肝恢复期血清及具有中和活性的鼠抗甲肝单克隆抗体产生竞争抑制反应,表明其针对甲肝病毒结构蛋白上的主要抗原决定簇.  相似文献   

7.
8.
为了探讨人源抗甲型肝炎(甲肝)病毒scFv—Fc融合抗体在酵母中的表达特性,将获得的人源抗甲肝病毒中和性单链可变区抗体(scFv抗体)基因克隆入含信号肽及人IgG1Fc抗体基因的酵母细胞表达载体中,获得了一株中和性人源抗甲肝病毒pPiscFv—FcHA16融合抗体的分泌表达,并对表达产物进行了纯化。同时对表达产物的生物学特性进行了一系列鉴定。表达的pPiscFv—FcHA16融合抗体为具有不同糖基化形式的同源二聚体,与相应的CHO细胞表达的IgG抗体相比,pPiscFv—FcHA16融合抗体仍保持很好的抗原结合活性,以及与中和性鼠抗甲肝病毒单克隆抗体的竞争抑制能力。同时也保持了对甲肝病毒的体外中和活性。这些结果表明,在酵母中表达的单链可变区(scFv)与IgG1Fc区的融合抗体具有很好的生物学活性,有希望用做体外诊断,用纯化相应的抗原,或者可能用于体内预防与治疗。  相似文献   

9.
10.
Prevention of the initial infection of mucosal dendritic cells (DC) and interruption of the subsequent transmission of HIV-1 from DC to T cells are likely to be important attributes of an effective human immunodeficiency virus type 1 (HIV-1) vaccine. While anti-HIV-1 neutralizing antibodies have been difficult to elicit by immunization, there are several human monoclonal antibodies (MAbs) that effectively neutralize virus infection of activated T cells. We investigated the ability of three well-characterized neutralizing MAbs (IgG1b12, 2F5, and 2G12) to block HIV-1 infection of human DC. DC were generated from CD14+ blood cells or obtained from cadaveric human skin. The MAbs prevented viral entry into purified DC and the ensuing productive infection in DC/T-cell cultures. When DC were first pulsed with HIV-1, MAbs blocked the subsequent transmission to unstimulated CD3+ T cells. Thus, neutralizing antibodies can block HIV-1 infection of DC and the cell-to-cell transmission of virus from infected DC to T cells. These data suggest that neutralizing antibodies could interrupt the initial events associated with mucosal transmission and regional spread of HIV-1.  相似文献   

11.
12.
Effective vaccination programs have dramatically reduced the number of measles-related deaths globally. Although all the available data suggest that measles eradication is biologically feasible, a structural and biochemical basis for the single serotype nature of measles virus (MV) remains to be provided. The hemagglutinin (H) protein, which binds to two discrete proteinaceous receptors, is the major neutralizing target. Monoclonal antibodies (MAbs) recognizing distinct epitopes on the H protein were characterized using recombinant MVs encoding the H gene from different MV genotypes. The effects of various mutations on neutralization by MAbs and virus fitness were also analyzed, identifying the location of five epitopes on the H protein structure. Our data in the present study demonstrated that the H protein of MV possesses at least two conserved effective neutralizing epitopes. One, which is a previously recognized epitope, is located near the receptor-binding site (RBS), and thus MAbs that recognize this epitope blocked the receptor binding of the H protein, whereas the other epitope is located at the position distant from the RBS. Thus, a MAb that recognizes this epitope did not inhibit the receptor binding of the H protein, rather interfered with the hemagglutinin-fusion (H-F) interaction. This epitope was suggested to play a key role for formation of a higher order of an H-F protein oligomeric structure. Our data also identified one nonconserved effective neutralizing epitope. The epitope has been masked by an N-linked sugar modification in some genotype MV strains. These data would contribute to our understanding of the antigenicity of MV and support the global elimination program of measles.  相似文献   

13.
14.
15.
16.
The TZM-bl cell line that is commonly used to assess neutralizing antibodies against human immunodeficiency virus type 1 (HIV-1) was recently reported to be contaminated with an ecotropic murine leukemia virus (MLV) (Y. Takeuchi, M. O. McClure, and M. Pizzato, J. Virol. 82:12585-12588, 2008), raising questions about the validity of results obtained with this cell line. Here we confirm this observation and show that HIV-1 neutralization assays performed with a variety of serologic reagents in a similar cell line that does not harbor MLV yield results that are equivalent to those obtained in TZM-bl cells. We conclude that MLV contamination has no measurable effect on HIV-1 neutralization when TZM-bl cells are used as targets for infection.It was recently reported that TZM-bl cells, which are commonly used to assess neutralizing antibodies (Abs) against human immunodeficiency virus type 1 (HIV-1), are contaminated with an ecotropic murine leukemia virus (MLV) (22). TZM-bl (also called JC.53bl-13) is a HeLa cell derivative that was engineered by amphotropic retroviral transduction to express CD4 and CCR5 (17) and was further engineered with an HIV-1-based vector to contain Tat-responsive reporter genes for firefly luciferase (Luc) and Escherichia coli β-galactosidase (24). These engineered features made TZM-bl cells highly susceptible to HIV-1 infection in a readily quantifiable assay for neutralizing Abs. Many published studies used this cell line for assessments of HIV-1 neutralization; these include several recent reports describing the magnitude, breadth, and epitope specificity of the neutralizing Ab response in infected individuals (14, 18-20), neutralization escape (25), and the neutralization phenotype of transmitted/founder viruses (10). TZM-bl cells are also gaining popularity for assessments of vaccine-elicited neutralizing Ab responses (13). The validity of these and other published results, together with a rationale for the continued use of TZM-bl cells in assessing neutralizing Abs against HIV-1, are very dependent on establishing to what extent, if any, MLV contamination affects the outcome of the assay.It was suggested that ecotropic MLV entered TZM-bl cells via the progenitor JC.53 cell line as an amphotropic MLV pseudotype (22). In this regard, JC.53 cells were constructed from HeLa cells in two stages by using ping-pong technology to amplify the pSFF vector derived from the replication-defective and highly truncated Friend spleen focus-forming virus (3). When used with this vector, this procedure has previously resulted in stable vector expression (17) without formation of replication-competent MLV recombinants (8, 11). A panel of HeLa-CD4 clones was made that express different amounts of CD4 and where the high-expression HI-J clone was used to make a derivative panel of clones (termed JC), including JC.53, that expressed diverse levels of CCR5 (9, 16, 17). In addition, the HeLa-CD4 clone HI-R that expressed low levels of CD4 was used to make another panel of CCR5-expressing clones (termed RC). To investigate this newly reported issue, cell extracts from these clonal panels and from TZM-bl cells were analyzed for MLV Gag antigens by Western immunoblotting. Representative data, as shown in Fig. Fig.1A,1A, confirm that JC.53 and TZM-bl cells express MLV Gag antigens, whereas the progenitor HI-J clone of HeLa-CD4 cells and many but not all of the other HeLa-CD4/CCR5 clones in the JC panel lack MLV antigens.Open in a separate windowFIG. 1.Characterization of HeLa clones for MLV Gag expression, HIV-1 susceptibility, and cell surface expression of HIV-1 fusion receptors. (A) MLV Gag antigen expression in HeLa cells and derivative clones expressing CD4 or CD4 and CCR5. Cell lysates were prepared from the cell clones and analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and Western blotting with Abs to MLV Gag antigens (upper blot). The lysates were also probed with anti-tubulin antibodies (lower blot). Lane 1, HeLa cells; lanes 2 and 3, HeLa CD4 clones HI-R and HI-J, respectively; lanes 4, 5, and 6, HeLa-CD4/CCR5 clones JC.10, JC.48, and JC.53, respectively; lane 7, TZM-bl cells; lane 8, psi-2 packaging cells positive for MLV Gag. (B) HIV-1 infectivity on the HeLa-CD4/CCR5 JC panel. Target cells were infected with HIV-1 isolate JRCSF that had been produced from clone JC.53 cells (black) or with JRCSF produced from transfected HEK293T cells (red). The target cells were also infected with the JR-FL isolate produced from peripheral blood mononuclear cells (PBMC; green). The HeLa-CD4/CCR5 target cells had a CCR5 expression range of 2 × 103 (clone JC.10) to 1.3 × 105 (clones JC.53 and TZM-bl) CCR5 molecules/cell. Each set of three data points at a given CCR5 expression level represents a single HeLa-CD4/CCR5 JC clone. None of the HIV-1 isolates was able to infect HeLa-CD4 cells lacking CCR5. The blue asterisks indicate clones that are negative for MLV Gag proteins. Clones JC.48 (used for subsequent infection and neutralization assays) and JC.53 (progenitor of TZM-bl cells) are specifically labeled. (C) Surface expression of CD4, CCR5, and CXCR4 on TZM-bl and JC.48 cells was assessed by flow cytometry using the same stocks of cells that were used in infection and neutralization assays in Fig. Fig.2.2. Surface staining was performed with phycoerythrin-conjugated mouse monoclonal Abs to CD4, CCR5 (CD195), and CXCR4 (CD184). Background staining was performed with isotype-matched control Abs. All Abs for flow cytometry were purchased from BD Biosciences Pharmingen (San Diego, CA). Results are shown as the mean fluorescence intensity (MFI) of positive cells. Most cells (>90%) stained positive in each case.Initial studies of HI-R cells and other clonal panels that were made using these methods also suggested a lack of MLV antigens (data not shown). We then determined the titers of replication-competent HIV-1JRCSF preparations using JC.53 and TZM-bl cells as well as other representative HeLa-CD4/CCR5 clones in the JC panel. The results are plotted in Fig. Fig.1B1B as a function of cellular CCR5 content. Clones having more than a low threshold level of ∼8,000 CCR5/cell were equally susceptible to infection regardless of whether they contained MLV antigens, clearly demonstrating that HIV-1JRCSF titers were not significantly affected by MLV. As expected, titers obtained with JC.53 and TZM-bl cells were also equivalent. In addition, these results demonstrate that HIV-1JRCSF preparations made in JC.53 cells and in cells lacking MLV antigens (i.e., HEK293T cells and human peripheral blood mononuclear cells) were unable to infect HeLa cells lacking CCR5. The results in Fig. Fig.1B1B were expected because previous studies demonstrated that ecotropic MLVs cannot infect human cells or even bind to the human CAT-1 receptor paralog (1, 6, 21, 23). Moreover, it has been shown that ecotropic host range MLVs do not interfere with superinfection by any retrovirus capable of infecting human cells, including gibbon ape leukemia virus, amphotropic MLV, baboon endogenous virus, and feline leukemia virus subgroup C (21). In view of the report by Takeuchi et al. (22), we were surprised to find that JC.53 and TZM-bl cells express very small amounts of ecotropic MLV Env glycoproteins, as indicated by immunofluorescence microscopy and by their resistance to complement-dependent killing by a cytotoxic antiserum specific for MLV envelope glycoproteins (6). Nevertheless, the cell clones that contained MLV Gag all released ecotropic host range virions that replicated in murine NIH 3T3 cells but not in human cells (data not shown).To determine whether MLV affects the measurement of neutralizing Abs in TZM-bl cells, parallel assays were performed in TZM-bl and JC.48 cells; these latter cells were determined to be MLV free by Western blot analysis (Fig. (Fig.1)1) and by an inability to transfer MLV infection to NIH 3T3 cells (data not shown). Because JC.48 cells express CCR5 at somewhat lower levels than JC.53 cells (∼2-fold lower; Fig. Fig.1B),1B), it may be expected that they would be less susceptible to HIV-1 infection than are TZM-bl cells. Differences in susceptibility to HIV-1 infection may require the use of adjusted virus doses to achieve equivalent assay performance when measuring neutralizing Abs. Indeed, levels of CD4 and CCR5 were approximately twofold lower on JC.48 cells than on TZM-bl cells, whereas levels of CXCR4 were approximately equal (Fig. (Fig.1C).1C). We therefore measured the susceptibility of both cell lines to infection by three molecularly cloned Env-pseudotyped viruses, each bearing an Env from a different CCR5-tropic HIV-1 subtype B virus (SF162.LS, Bal.26, and QH0692.42). Infection was quantified by Luc activity expressed as relative luminescence units (RLU). Because JC.48 cells do not contain a reporter gene, the Env-pseudotyped viruses were prepared by cotransfection with the NL4-3.Luc.R-E- reporter backbone plasmid (7). Identical Luc-containing, Env-pseudotyped virus stocks were used in both cell lines. As shown in Fig. Fig.2A,2A, the infectivity of each pseudotyped virus was somewhat diminished in JC.48 cells compared to the infectivity in TZM-bl cells. Nonetheless, the levels of infectivity in JC.48 cells remained acceptable for neutralization assays.Open in a separate windowFIG. 2.HIV-1 infectivity and neutralization in TZM-bl and JC.48.CD4.CCR5 cells. (A) TZM-bl and JC.48 cells were incubated with serial fourfold dilutions (11 dilutions total) of three HIV-1 Env-pseudotyped viruses in quadruplicate in 96-well culture plates. Luc activity was measured after 48 h of incubation and is expressed as RLU after subtraction of background luminescence from cell control wells. Squares, TZM-bl cells; triangles, JC.48 cells. (B) Neutralization assays were performed with three HIV-1 Env-pseudotyped viruses in either TZM-bl or JC.48 cells. Input virus doses were adjusted to yield equivalent infectivity in both cell lines. Black bars, TZM-bl; gray bars, JC.48. Top panel: sCD4, monoclonal Abs, and HIVIG (purified immunoglobulin G from pooled HIV-1-positive plasmas). Bottom panel: individual HIV-1-positive plasma samples. The same three stocks of virus were used in both experiments. All three Env-pseudotyped viruses were prepared with the NL4-3.Luc.R-E- reporter backbone plasmid.With this information in hand, neutralization assays were performed in JC.48 and TZM-bl cells using adjusted virus doses that yielded equivalent infectivity levels in both cell lines. These neutralization assays were performed in a 96-well format as described previously (12), where the 50% inhibitory dose (ID50) was reported as either the concentration or sample dilution at which RLU were reduced by 50% compared to RLU in virus control wells (cells plus virus without test sample) after subtraction of background RLU from cell control wells (cells only). A wide variety of serologic reagents was tested, including sCD4, a monoclonal Ab to the CD4 binding site of gp120 (immunoglobulin G1b12) (15); a monoclonal Ab that recognizes a glycan-specific epitope on gp120 (2G12) (5); two monoclonal Abs that recognize adjacent epitopes in the membrane proximal external region of gp41 (2F5 and 4E10) (2, 4); and serum samples from seven antiretroviral-naive HIV-1-infected individuals. As shown in Fig. Fig.2B,2B, results in the two cell lines were similar for all three viruses and all serologic reagents tested. Indeed, ID50 values in the two cell types agreed within twofold, which is within the normal range of variability of the assay. These results indicate that equivalent neutralization results were obtained in both cell lines.In summary, we found no evidence that ecotropic MLV contamination in TZM-bl cells has a measurable effect on HIV-1 neutralization when these cells are used as targets for infection. This outcome indicates that the presence of ecotropic MLV in TZM-bl cells does not alter the ability of Ab to neutralize HIV-1, nor does it interfere with the detection of neutralization by using HIV-1 Tat-regulated reporter gene expression in a single-cycle infection assay. However, we discourage the use of TZM-bl cells to generate HIV-1 stocks, because the latter would likely be contaminated with ecotropic MLV and contain pseudovirions with mixtures of HIV-1 and ecotropic MLV Env glycoproteins. For this reason, we have begun efforts to produce an uncontaminated, second-generation panel of HeLa-CD4/CCR5 cell clones that express diverse amounts of CCR5 and to isolate a TZM-bl variant lacking MLV antigens.  相似文献   

17.
Russian Journal of Bioorganic Chemistry - Humanization of antibodies for the development of novel therapeutic agents with low immunogenicity remains a topical problem in modern science. In the...  相似文献   

18.
The structure of a complex between human rhinovirus 2 (HRV2) and the Fab fragment of neutralizing monoclonal antibody (MAb) 3B10 has been determined to 25-Å resolution by cryoelectron microscopy and three-dimensional reconstruction techniques. The footprint of 3B10 on HRV2 is very similar to that of neutralizing MAb 8F5, which binds bivalently across the icosahedral twofold axis. However, the 3B10 Fab fragment (Fab-3B10) is bound in an orientation, inclined at approximately 45° to the surface of the virus capsid, which is compatible only with monovalent binding of the antibody. The canyon around the fivefold axis is not directly obstructed by the bound Fab. The X-ray structures of a closely related HRV (HRV1A) and a Fab fragment were fitted to the density maps of the HRV2–Fab-3B10 complex obtained by cryoelectron microscope techniques. The footprint of 3B10 on the viral surface is largely on VP2 but also covers the VP3 loop centered on residue 3064 and the VP1 loop centered on residue 1267. MAb 3B10 can interact directly with VP2 residue 2164, the site of an escape mutation on VP2, and with VP1 residues 1264 to 1267, the site of a deletion escape mutation. Deletion of these residues shortens the VP1 loop, moving it away from the MAb binding site. All structural and biochemical evidence indicates that MAb 3B10 binds to a conformation epitope on HRV2.  相似文献   

19.
20.
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