Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters

Publication year range
1.
Annu Rev Cell Dev Biol ; 30: 111-39, 2014.
Article in English | MEDLINE | ID: mdl-25000995

ABSTRACT

In biomembrane fusion pathways, membranes are destabilized through insertions of amphipathic protein segments, lipid reorganization via hemifusion, protein restructuring, and dimpling of the membranes. Four classes of membrane proteins are known in virus and cell fusion. Class I virus-cell fusion proteins (fusogens) are α-helix-rich prefusion trimers that form coiled-coil structures that insert hydrophobic fusion peptides or loops (FPs or FLs) into membranes and refold into postfusion trimers. Class II virus-cell fusogens are ß-sheet-rich prefusion homo- or heterodimers that insert FLs into membranes, ending in postfusion trimers. Class III virus-cell fusogens are trimers with both α-helices and ß-sheets that dissociate into monomers, insert FLs into membranes, and oligomerize into postfusion trimers. Class IV reoviral cell-cell fusogens are small proteins with FLs that oligomerize to fuse membranes. Class I cell-cell fusogens (Syncytins) were captured by mammals from retroviruses, and class II cell-cell fusogens (EFF-1/AFF-1) fuse membranes via homotypic zippering. Mechanisms and fusogens for most cell fusion events are unknown.


Subject(s)
Cell Fusion , Membrane Fusion , Viral Fusion Proteins/physiology , Animals , Gene Products, env/physiology , Hemagglutinin Glycoproteins, Influenza Virus/physiology , Humans , Membrane Glycoproteins/physiology , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Pregnancy Proteins/physiology , Protein Conformation , Structure-Activity Relationship , Viral Envelope Proteins/physiology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/classification , env Gene Products, Human Immunodeficiency Virus/physiology
2.
PLoS Biol ; 17(1): e3000114, 2019 01.
Article in English | MEDLINE | ID: mdl-30650070

ABSTRACT

During the entry process, the human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein (Env) trimer undergoes a sequence of conformational changes triggered by both CD4 and coreceptor engagement. Resolving the conformation of these transient entry intermediates has proven challenging. Here, we fine-mapped the antigenicity of entry intermediates induced by increasing CD4 engagement of cell surface-expressed Env. Escalating CD4 triggering led to the sequential adoption of different pre-fusion conformational states of the Env trimer, up to the pre-hairpin conformation, that we assessed for antibody epitope presentation. Maximal accessibility of the coreceptor binding site was detected below Env saturation by CD4. Exposure of the fusion peptide and heptad repeat 1 (HR1) required higher CD4 occupancy. Analyzing the diverse antigenic states of the Env trimer, we obtained key insights into the transitions in epitope accessibility of broadly neutralizing antibodies (bnAbs). Several bnAbs preferentially bound CD4-triggered Env, indicating a potential capacity to neutralize both pre- and post-CD4 engagement, which needs to be explored. Assessing binding and neutralization activity of bnAbs, we confirm antibody dissociation rates as a driver of incomplete neutralization. Collectively, our findings highlight a need to resolve Env conformations that are neutralization-relevant to provide guidance for immunogen development.


Subject(s)
CD4 Antigens/metabolism , Epitopes/immunology , env Gene Products, Human Immunodeficiency Virus/chemistry , Antibodies, Neutralizing/metabolism , Antibodies, Neutralizing/physiology , HIV Envelope Protein gp120/chemistry , HIV-1/immunology , Humans , Protein Binding , Protein Conformation , Protein Multimerization , env Gene Products, Human Immunodeficiency Virus/metabolism , env Gene Products, Human Immunodeficiency Virus/physiology
3.
J Virol ; 94(16)2020 07 30.
Article in English | MEDLINE | ID: mdl-32493821

ABSTRACT

Infection of human immunodeficiency virus type 1 (HIV-1) is subject to restriction by cellular factors. Serine incorporator 5 (SERINC5) and interferon-inducible transmembrane 3 (IFITM3) proteins represent two of these restriction factors, which inhibit HIV-1 entry into target cells. Both proteins impede fusion of the viral membrane with the cellular membrane and the formation of a viral fusion pore, and both are countered by the HIV-1 envelope glycoprotein (Env). Given the immense and lasting pressure which Env endures from host adaptive immune responses, it is important to understand whether and how HIV-1 Env is able to maintain the resistance to SERINC5 and IFITM3 throughout the course of infection. We have thus examined a panel of HIV-1 Env clones that were isolated at different stages of viral infection-transmission, acute, and chronic. While HIV-1 Env clones from the transmission stage are resistant to both SERINC5 and IFITM3, as infection progresses into the acute and chronic stages, the resistance to IFITM3 but not to SERINC5 is gradually lost. We further discovered a significant correlation between the resistance of HIV-1 Env to soluble CD4 inhibition and the resistance to SERINC5 but not to IFITM3. Interestingly, the miniprotein CD4 mimetic M48U1 sensitizes HIV-1 Env to the inhibition by SERINC5 but not IFITM3. Together, these data indicate that SERINC5 and IFITM3 exert differential inhibitory pressures on HIV-1 Env over different stages of HIV-1 infection and that HIV-1 Env uses varied strategies to resist these two restriction factors.IMPORTANCE HIV-1 Env protein is exposed to the inhibition not only by humoral response, but also by host restriction factors, including serine incorporator 5 (SERINC5) and interferon-inducible transmembrane 3 (IFITM3). This study investigates how HIV-1 envelope glycoprotein (Env) manages to overcome the pressures from all these different host inhibition mechanisms over the long course of viral infection. HIV-1 Env preserves the resistance to SERINC5 but becomes sensitive to IFITM3 when infection progresses into the chronic stage. Our study also supports the possibility of using CD4 mimetic compounds to sensitize HIV-1 Env to the inhibition by SERINC5 as a potential therapeutic strategy.


Subject(s)
HIV Infections/metabolism , HIV-1/metabolism , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism , Cell Line , Glycoproteins/metabolism , HEK293 Cells , HIV Infections/physiopathology , Host-Pathogen Interactions , Humans , Membrane Proteins/physiology , Protein Binding , RNA-Binding Proteins/physiology , Viral Envelope Proteins/metabolism , Virion/metabolism , Virus Internalization/drug effects , env Gene Products, Human Immunodeficiency Virus/metabolism , env Gene Products, Human Immunodeficiency Virus/physiology
4.
PLoS Pathog ; 15(7): e1007925, 2019 07.
Article in English | MEDLINE | ID: mdl-31260493

ABSTRACT

HIV-1 does not persistently infect macaques due in part to restriction by several macaque host factors. This has been partially circumvented by generating chimeric SIV/HIV-1 viruses (SHIVs) that encode SIV antagonist of known restriction factors. However, most SHIVs replicate poorly in macaques unless they are further adapted in culture and/or macaques (adapted SHIVs). Therefore, development of SHIVs encoding HIV-1 sequences derived directly from infected humans without adaptation (unadapted SHIVs) has been challenging. In contrast to the adapted SHIVs, the unadapted SHIVs have lower replication kinetics in macaque lymphocytes and are sensitive to type-1 interferon (IFN). The HIV-1 Envelope (Env) in the chimeric virus determines both the reduced replication and the IFN-sensitivity differences. There is limited information on macaque restriction factors that specifically limit replication of the more biologically relevant, unadapted SHIV variants. In order to identify the IFN-induced host factor(s) that could contribute to the inhibition of SHIVs in macaque lymphocytes, we measured IFN-induced gene expression in immortalized pig-tailed macaque (Ptm) lymphocytes using RNA-Seq. We found 147 genes that were significantly upregulated upon IFN treatment in Ptm lymphocytes and 31/147 were identified as genes that encode transmembrane helices and thus are likely present in membranes where interaction with viral Env is plausible. Within this group of upregulated genes with putative membrane-localized proteins, we identified several interferon-induced transmembrane protein (IFITM) genes, including several previously uncharacterized Ptm IFITM3-related genes. An evolutionary genomic analysis of these genes suggests the genes are IFITM3 duplications not found in humans that are both within the IFITM locus and also dispersed elsewhere in the Ptm genome. We observed that Ptm IFITMs are generally packaged at higher levels in unadapted SHIVs when compared to adapted SHIVs. CRISPR/Cas9-mediated knockout of Ptm IFITMs showed that depletion of IFITMs partially rescues the IFN sensitivity of unadapted SHIV. Moreover, we found that the depletion of IFITMs also increased replication of unadapted SHIV in the absence of IFN treatment, suggesting that Ptm IFITMs are likely important host factors that limit replication of unadapted SHIVs. In conclusion, this study shows that Ptm IFITMs selectively restrict replication of unadapted SHIVs. These findings suggest that restriction factors including IFITMs vary in their potency against different SHIV variants and may play a role in selecting for viruses that adapt to species-specific restriction factors.


Subject(s)
HIV-1/physiology , HIV-1/pathogenicity , Simian Immunodeficiency Virus/physiology , Simian Immunodeficiency Virus/pathogenicity , env Gene Products, Human Immunodeficiency Virus/physiology , Adaptation, Physiological , Animals , Genes, env , HIV-1/genetics , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Host Specificity , Humans , Interferon-alpha/metabolism , Macaca nemestrina/genetics , Macaca nemestrina/immunology , Macaca nemestrina/virology , Protein Processing, Post-Translational , Reassortant Viruses/genetics , Reassortant Viruses/pathogenicity , Reassortant Viruses/physiology , Simian Immunodeficiency Virus/genetics , Virus Replication
5.
Acta Virol ; 59(3): 209-20, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26435143

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) 92UG046 Env protein, obtained from a CD4-independent HIV-1 primary isolate (Zerhouni et al., 2004), has the ability to initiate an infection in HeLa cells expressing CD4 when carrying the full-length (FL) Env, but uses CD8 molecules for receptor-mediated entry when carrying a truncated Env (CT84). To determine whether a specific length or structure in the cytoplasmic tail (CT) is responsible for this alteration of tropism, we compared a series of Env constructs with different CT truncations and the presence or absence of an amphipathic alpha- helical sequence. We found that truncated constructs containing the alpha-helical LLP-2 structure in their CT domains conferred a switch from CD4 to CD8 tropism. The results support the conclusion that the structure of the CT domain can play an important role in determining receptor specificity.


Subject(s)
Cytoplasm/chemistry , HIV-1/physiology , Receptors, HIV/physiology , env Gene Products, Human Immunodeficiency Virus/chemistry , Amino Acid Sequence , Animals , Cells, Cultured , Humans , Mice , Molecular Sequence Data , Protein Structure, Tertiary , Tropism , env Gene Products, Human Immunodeficiency Virus/physiology
6.
Adv Exp Med Biol ; 790: 128-49, 2013.
Article in English | MEDLINE | ID: mdl-23884589

ABSTRACT

The retrovirus family contains several important human and animal pathogens, including the human immunodeficiency virus (HIV), the causative agent of acquired immunodeficiency syndrome (AIDS). Studies with retroviruses were instrumental to our present understanding of the cellular entry of enveloped viruses in general. For instance, studies with alpharetroviruses defined receptor engagement, as opposed to low pH, as a trigger for the envelope protein-driven membrane fusion. The insights into the retroviral entry process allowed the generation of a new class of antivirals, entry inhibitors, and these therapeutics are at present used for treatment of HIV/AIDS. In this chapter, we will summarize key concepts established for entry of avian sarcoma and leukosis virus (ASLV), a widely used model system for retroviral entry. We will then review how foamy virus and HIV, primate- and human retroviruses, enter target cells, and how the interaction of the viral and cellular factors involved in the cellular entry of these viruses impacts viral tropism, pathogenesis and approaches to therapy and vaccine development.


Subject(s)
Retroviridae/physiology , Virus Internalization , Animals , Avian Leukosis Virus/physiology , Avian Sarcoma Viruses/physiology , HIV/physiology , Humans , Hydrogen-Ion Concentration , Receptors, Virus/physiology , Viral Tropism , env Gene Products, Human Immunodeficiency Virus/physiology
7.
J Gen Virol ; 93(Pt 12): 2635-2645, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22956734

ABSTRACT

Understanding the properties of viruses preferentially establishing infection during perinatal transmission of human immunodeficiency virus type 1 (HIV-1) is critical for the development of effective measures to prevent transmission. A previous study demonstrated that the newly transmitted viruses (in infants) of chronically infected mother-infant pairs (MIPs) were fitter in terms of growth, which was imparted by their envelope (Env) glycoprotein V1-V5 regions, than those in the corresponding chronically infected mothers. In order to investigate whether the higher fitness of transmitted viruses was conferred by their higher entry efficiency directed by the V1-V5 regions during perinatal transmission, the fusogenicity of Env containing V1-V5 regions derived from transmitted and non-tranmsmitted viruses of five chronically infected MIPs and two acutely infected MIPs was analysed using two different cell-cell fusion assays. The results showed that, in one chronically infected MIP, a higher fusion efficiency was induced by the infant Env V1-V5 compared with that of the corresponding mother. Moreover, the V4-V5 regions played an important role in discriminating the transmitted and non-transmitted viruses in this pair. However, neither a consistent pattern nor significant differences in fusogenicity mediated by the V1-V5 regions between maternal and infant variants was observed in the other MIPs. This study suggests that there is no consistent and significant correlation between viral fitness selection and entry efficiency directed by the V1-V5 regions during perinatal transmission. Other factors such as the route and timing of transmission may also be involved.


Subject(s)
HIV Infections/transmission , HIV Infections/virology , HIV-1/physiology , HIV-1/pathogenicity , Infectious Disease Transmission, Vertical , Pregnancy Complications, Infectious/virology , env Gene Products, Human Immunodeficiency Virus/physiology , Adult , Amino Acid Sequence , Base Sequence , DNA, Viral/genetics , Female , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/physiology , HIV-1/genetics , Humans , Infant, Newborn , Male , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/physiology , Pregnancy , Sequence Homology, Amino Acid , Viral Fusion Proteins/genetics , Viral Fusion Proteins/physiology , Virus Internalization , Young Adult , env Gene Products, Human Immunodeficiency Virus/genetics
8.
Curr HIV/AIDS Rep ; 9(1): 52-63, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22237899

ABSTRACT

HIV-1 is completely dependent upon the Env protein to enter cells. The virus typically replicates in activated CD4+ T cells due to viral entry requirements for the CCR5 coreceptor and for high surface levels of the CD4 receptor. This is the case for the transmitted virus and for most of the virus sampled in the blood. Over the course of infection, the env gene can evolve to encode a protein with altered receptor and coreceptor usage allowing the virus to enter alternative host cells. In about 50% of HIV-1 infections, the viral population undergoes coreceptor switching, usually late in disease, allowing the virus to use CXCR4 to enter a different subset of CD4+ T cells. Neurocognitive disorders occur in about 10% of infections, also usually late in disease, but caused (ultimately) by viral replication in the brain either in CD4+ T cells or macrophage and/or microglia. Expanded host range is significantly intertwined with pathogenesis. Identification and characterization of such HIV-1 variants may be useful for early detection which would allow intervention to reduce viral pathogenesis in these alternative cell types.


Subject(s)
HIV Infections/transmission , HIV-1/chemistry , env Gene Products, Human Immunodeficiency Virus/physiology , Central Nervous System/virology , Humans , Macrophages/virology , Phenotype , T-Lymphocytes/virology
9.
J Virol ; 84(7): 3382-95, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20089658

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein (Env) on whole virions is heterogeneous, so molecular analysis of Env with monoclonal antibodies (MAbs) is challenging. Virus capture assays (VCAs) involving immobilized MAbs are typically used, but these assays suffer from immobilization artifacts and do not provide binding constants. Furthermore, we show here that certain HIV-1 neutralizing MAbs, including 2G12, 4E10, 2F5, Z13e1, and D5, will capture virion particles completely devoid of Env. We modified the VCA such that MAbs and virions are incubated in solution, and unbound MAbs are removed prior to the capture step. This modification nearly eliminated evidence of Env-independent binding by MAbs to virions and allowed determination of apparent affinity constants in solution. Three important qualitative observations were further revealed. First, neutralizing MAbs 2F5, 4E10, and Z13e1 against the membrane-proximal external region (MPER) of HIV-1 gp41 were found to capture virions efficiently only if a significant amount of uncleaved gp160 or synthetic MPER peptide was present. Second, we show how non-native forms of Env vary by Env genotype and that Env from HIV-1(JR-FL) is more homogeneously trimeric than that from HIV-1(JR-CSF). Third, we determined that Env containing all or parts of gp41, including uncleaved gp160, binds spontaneously to free virions. This exogenous Env is an indiscriminate molecular "bridge" between Env-specific Ab and virions and can affect VCA analyses, particularly using pseudotyped virions. Heterogeneity in Env from endogenous and exogenous sources might also subvert humoral immunity to HIV-1, so in-solution VCAs may help to dissect this heterogeneity for vaccine design purposes.


Subject(s)
Antibodies, Monoclonal/immunology , HIV-1/isolation & purification , AIDS Vaccines/immunology , Cell Line , Enzyme-Linked Immunosorbent Assay , HIV Envelope Protein gp41/physiology , HIV-1/immunology , Humans , Virion/immunology , Virion/isolation & purification , env Gene Products, Human Immunodeficiency Virus/physiology
10.
J Virol ; 83(22): 11715-25, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726524

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1)-mediated depletion of CD4+ lymphocytes in an infected individual is the hallmark of progression to AIDS. However, the mechanism for this depletion remains unclear. To identify mechanisms of HIV-1-mediated CD4 T-cell death, two similar viral isolates obtained from a rapid progressor patient with significantly different pathogenic phenotypes were studied. One isolate (R3A) demonstrates enhanced pathogenesis in both in vivo models and relevant ex vivo lymphoid organ model systems compared to another isolate, R3B. The pathogenic determinants were previously mapped to the V5-gp41 envelope region, correlating functionally with enhanced fusion activity and elevated CXCR4 binding affinity. To further elucidate specific differences between R3A and R3B within the V5-gp41 domains that enhance CD4 depletion, R3A-R3B chimeras to study the V5-gp41 region were developed. Our data demonstrate that six residues in the ectodomain of R3A provide the major determinant for both enhanced Env-cell fusion and pathogenicity. Furthermore, three amino acid differences in the heptad repeat 2 (HR-2) domain of R3A determined its fusion activity and significantly elevated its pathogenic activity. The chimeric viruses with enhanced fusion activity, but not elevated CXCR4 affinity, correlated with high pathogenicity in the thymus organ. We conclude that the functional domain of a highly pathogenic HIV-1 Env is determined by mutations in the HR-2 region that contribute to enhanced fusion and CD4 T-cell depletion.


Subject(s)
HIV-1/pathogenicity , env Gene Products, Human Immunodeficiency Virus/physiology , CD4-Positive T-Lymphocytes/virology , Cell Line , HIV Envelope Protein gp41/physiology , HIV Infections/virology , Mutagenesis, Site-Directed , Receptors, CXCR4/physiology , Virus Attachment , Virus Internalization
11.
J Virol ; 83(22): 11966-78, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19740980

ABSTRACT

Tetherin (CD317/BST-2), an interferon-induced membrane protein, restricts the release of nascent retroviral particles from infected cell surfaces. While human immunodeficiency virus type 1 (HIV-1) encodes the accessory gene vpu to overcome the action of tetherin, the lineage of primate lentiviruses that gave rise to HIV-2 does not. It has been previously reported that the HIV-2 envelope glycoprotein has a Vpu-like function in promoting virus release. Here we demonstrate that the HIV-2 Rod envelope glycoprotein (HIV-2 Rod Env) is a tetherin antagonist. Expression of HIV-2 Rod Env, but not that of HIV-1 or the closely related simian immunodeficiency virus (SIV) SIVmac1A11, counteracts tetherin-mediated restriction of Vpu-defective HIV-1 in a cell-type-specific manner. This correlates with the ability of the HIV-2 Rod Env to mediate cell surface downregulation of tetherin. Antagonism requires an endocytic motif conserved across HIV/SIV lineages in the gp41 cytoplasmic tail, but specificity for tetherin is governed by extracellular determinants in the mature Env protein. Coimmunoprecipitation studies suggest an interaction between HIV-2 Rod Env and tetherin, but unlike studies with Vpu, we found no evidence of tetherin degradation. In the presence of HIV-2 Rod Env, tetherin localization is restricted to the trans-Golgi network, suggesting Env-mediated effects on tetherin trafficking sequester it from virus assembly sites on the plasma membrane. Finally, we recapitulated these observations in HIV-2-infected CD4+ T-cell lines, demonstrating that tetherin antagonism and sequestration occur at physiological levels of Env expression during virus replication.


Subject(s)
Antigens, CD/physiology , HIV-2/physiology , Membrane Glycoproteins/physiology , env Gene Products, Human Immunodeficiency Virus/physiology , CD4-Positive T-Lymphocytes/physiology , Cell Line , Down-Regulation , GPI-Linked Proteins , Golgi Apparatus/physiology , HIV Infections/virology , HIV-1/physiology , HeLa Cells , Humans , Jurkat Cells
12.
J Virol ; 83(22): 11588-98, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726514

ABSTRACT

The membrane-spanning domain (MSD) of the envelope (Env) glycoprotein from human (HIV) and simian immunodeficiency viruses plays a key role in anchoring the Env complex into the viral membrane but also contributes to its biological function in fusion and virus entry. In HIV type 1 (HIV-1), it has been predicted to span 27 amino acids, from lysine residue 681 to arginine 707, and encompasses an internal arginine at residue 694. By examining a series of C-terminal-truncation mutants of the HIV-1 gp41 glycoprotein that substituted termination codons for amino acids 682 to 708, we show that this entire region is required for efficient viral infection of target cells. Truncation to the arginine at residue 694 resulted in an Env complex that was secreted from the cells. In contrast, a region from residues 681 to 698, which contains highly conserved hydrophobic residues and glycine motifs and extends 4 amino acids beyond 694R, can effectively anchor the protein in the membrane, allow efficient transport to the plasma membrane, and mediate wild-type levels of cell-cell fusion. However, these fusogenic truncated Env mutants are inefficiently incorporated into budding virions. Based on the analysis of these mutants, a "snorkeling" model, in which the flanking charged amino acid residues at 681 and 694 are buried in the lipid while their side chains interact with polar head groups, is proposed for the HIV-1 MSD.


Subject(s)
HIV-1/genetics , Virus Integration/genetics , Virus Internalization , env Gene Products, Human Immunodeficiency Virus/genetics , Animals , Binding Sites , COS Cells , Cell Line , Chlorocebus aethiops , Gene Expression Profiling , HIV Envelope Protein gp41/biosynthesis , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/physiology , HIV Infections/virology , HIV-1/pathogenicity , HIV-1/physiology , Humans , Virus Integration/physiology , env Gene Products, Human Immunodeficiency Virus/biosynthesis , env Gene Products, Human Immunodeficiency Virus/physiology
13.
Retrovirology ; 6: 75, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19671162

ABSTRACT

BACKGROUND: The HIV-1 Env glycoprotein mediates virus entry by catalyzing direct fusion between the virion membrane and the target cell plasma membrane. Env is composed of two subunits: gp120, which binds to CD4 and the coreceptor, and gp41, which is triggered upon coreceptor binding to promote the membrane fusion reaction. Env on the surface of infected cells is a trimer consisting of three gp120/gp41 homo-dimeric protomers. An emerging question concerns cooperative interactions between the protomers in the trimer, and possible implications for Env function. RESULTS: We extended studies on cooperative subunit interactions within the HIV-1 Env trimer, using analysis of functional complementation between coexpressed inactive variants harboring different functional deficiencies. In assays of Env-mediated cell fusion, complementation was observed between variants with a wide range of defects in both the gp120 and gp41 subunits. The former included gp120 subunits mutated in the CD4 binding site or incapable of coreceptor interaction due either to mismatched specificity or V3 loop mutation. Defective gp41 variants included point mutations at different residues within the fusion peptide or heptad repeat regions, as well as constructs with modifications or deletions of the membrane proximal tryptophan-rich region or the transmembrane domain. Complementation required the defective variants to be coexpressed in the same cell. The observed complementation activities were highly dependent on the assay system. The most robust activities were obtained with a vaccinia virus-based expression and reporter gene activation assay for cell fusion. In an alternative system involving Env expression from integrated provirus, complementation was detected in cell fusion assays, but not in virus particle entry assays. CONCLUSION: Our results indicate that Env function does not require every subunit in the trimer to be competent for all essential activities. Through cross-talk between subunits, the functional determinants on one defective protomer can cooperatively interact to trigger the functional determinants on an adjacent protomer(s) harboring a different defect, leading to fusion. Cooperative subunit interaction is a general feature of the Env trimer, based on complementation activities observed for a highly diverse range of functional defects.


Subject(s)
HIV-1/physiology , Protein Multimerization , Protein Subunits/physiology , Virus Internalization , env Gene Products, Human Immunodeficiency Virus/physiology , Cell Fusion , Genes, Reporter , Genetic Complementation Test , Genetic Vectors , HIV-1/genetics , Humans , Mutant Proteins/genetics , Mutant Proteins/physiology , Mutation, Missense , Protein Subunits/genetics , Sequence Deletion , Vaccinia virus/genetics , env Gene Products, Human Immunodeficiency Virus/genetics
14.
J Virol ; 82(23): 11758-66, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18815295

ABSTRACT

The envelope gene (env) of human immunodeficiency virus type 1 (HIV-1) undergoes rapid divergence from the transmitted sequence and increasing diversification during the prolonged course of chronic infection in humans. In about half of infected individuals or more, env evolution leads to expansion of the use of entry coreceptor from CCR5 alone to CCR5 and CXCR4. The stochastic nature of this coreceptor switch is not well explained by host selective forces that should be relatively constant between infected individuals. Moreover, differences in the incidence of coreceptor switching among different HIV-1 subtypes suggest that properties of the evolving virus population drive the switch. We evaluated the functional properties of sequential env clones from a patient with evidence of coreceptor switching at 5.67 years of infection. We found an abrupt decline in the ability of viruses to use CCR5 for entry at this time, manifested by a 1- to 2-log increase in susceptibility to CCR5 inhibitors and a reduced ability to infect cell lines with low CCR5 expression. There was an abnormally rapid 5.4% divergence in env sequences from 4.10 to 5.76 years of infection, with the V3 and V4/V5 regions showing the greatest divergence and evidence of positive selection. These observations suggest that a decline in the fitness of R5 virus populations may be one driving force that permits the emergence of R5X4 variants.


Subject(s)
Receptors, CCR5/physiology , Receptors, CXCR4/physiology , CCR5 Receptor Antagonists , Humans , Receptors, CCR5/analysis , Receptors, CCR5/chemistry , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/physiology
15.
J Virol ; 82(23): 11609-18, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18786994

ABSTRACT

Selection of a minor viral genotype during perinatal transmission of human Immunodeficiency virus type 1 (HIV-1) has been observed, but there is a lack of information on the correlation of the restrictive transmission with biological properties of the virus, such as replicative fitness. Recombinant viruses expressing the enhanced green fluorescent protein or the Discosoma sp. red fluorescent (DsRed2) protein carrying the V1 to V5 regions of env from seven mother-infant pairs (MIPs) infected by subtype C HIV-1 were constructed, and competition assays were carried out to compare the fitness between the transmitted and nontransmitted viruses. Flow cytometry was used to quantify the frequency of infected cells, and the replicative fitness was determined based on a calculation that takes into account replication of competing viruses in a single infection versus dual infections. Transmitted viruses from five MIPs with the mothers chronically infected showed a restrictive env genotype, and all the recombinant viruses carrying the infants' Env had higher replicative fitness than those carrying the Env from the mothers. This growth fitness is lineage specific and can be observed only within the same MIP. In contrast, in two MIPs where the mothers had undergone recent acute infection, the viral Env sequences were similar between the mothers and infants and showed no further restriction in quasispecies during perinatal transmission. The recombinant viruses carrying the Env from the infants' viruses also showed replication fitness similar to those carrying the mothers' Env proteins. Our results suggest that newly transmitted viruses from chronically infected mothers have been selected to have higher replicative fitness to favor transmission, and this advantage is conferred by the V1 to V5 region of Env of the transmitted viruses. This finding has important implications for vaccine design or development of strategies to prevent HIV-1 transmission.


Subject(s)
Acquired Immunodeficiency Syndrome/transmission , HIV-1/physiology , Infectious Disease Transmission, Vertical , Virus Replication , env Gene Products, Human Immunodeficiency Virus/physiology , Female , Humans , Infant , Infectious Disease Transmission, Vertical/prevention & control , Pregnancy , Recombination, Genetic
16.
Article in English | MEDLINE | ID: mdl-19842409

ABSTRACT

HIV-1 tat gene function and immunogenicity of HIV-1 Tat protein from 3 low (PS01, PS40, PS58) and 3 high (PS19, PS65, LP22) viral load infected, untreated and asymptomatic individuals from Thailand were compared. Levels of Tat-dependent chloramphenicol acetyltransferase (CAT) induced in HL3T1 cells with tat1 gene from HIV-1 isolates of high viral load group was significantly higher than those from low viral load group. HIV-1 subtype determination using env (C2-V4) gene demonstrated that 2/3 (PS01 and PS40) and 1/3 (PS58) from low viral load group were CRF01_AE and subtype B, while all 3 HIV-1 isolates from high viral load group were CRF01_AE. However, all 3 HIV-1 tat nucleotide sequences from low viral load group, which contained env CRF01_AE sequence, belonged to subtype B whereas all those from high viral load group contained CRF01_AE sequence. HIV Tat recombinant proteins from these groups were tested for immunogenicity in mice. All recombinant Tat proteins (except from PS58) were immunogenic in a dose-dependent manner, but with significantly differences of the immunogenicity levels between high and low viral load groups. These results indicated that HIV-1 subtype B tat gene activities might be associated with reduced disease progression of HIV-1 CRF01_AE infected individuals.


Subject(s)
Genes, tat/physiology , HIV Infections/virology , HIV-1/genetics , env Gene Products, Human Immunodeficiency Virus/physiology , Adult , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Disease Progression , Epitopes/genetics , Female , HIV-1/immunology , Humans , Male , Mice , Recombinant Proteins , Viral Load , Young Adult
17.
Retrovirology ; 5: 23, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18312646

ABSTRACT

BACKGROUND: Numerous studies have shown that viral quasi-species with genetically diverse envelope proteins (Env) replicate simultaneously in patients infected with the human immunodeficiency virus type 1 (HIV-1). Less information is available concerning the extent that envelope sequence diversity translates into a diversity of phenotypic properties, including infectivity and resistance to entry inhibitors. METHODS: To study these questions, we isolated genetically distinct contemporaneous clonal viral populations from the plasma of 5 HIV-1 infected individuals (n = 70), and evaluated the infectivity of recombinant viruses expressing Env proteins from the clonal viruses in several target cells. The sensitivity to entry inhibitors (enfuvirtide, TAK-799), soluble CD4 and monoclonal antibodies (2G12, 48d, 2F5) was also evaluated for a subset of the recombinant viruses (n = 20). RESULTS: Even when comparisons were restricted to viruses with similar tropism, the infectivity for a given target cell of viruses carrying different Env proteins from the same patient varied over an approximately 10-fold range, and differences in their relative ability to infect different target cells were also observed. Variable region haplotypes associated with high and low infectivity could be identified for one patient. In addition, clones carrying unique mutations in V3 often displayed low infectivity. No correlation was observed between viral infectivity and sensitivity to inhibition by any of the six entry inhibitors evaluated, indicating that these properties can be dissociated. Significant inter-patient differences, independent of infectivity, were observed for the sensitivity of Env proteins to several entry inhibitors and their ability to infect different target cells. CONCLUSION: These findings demonstrate the marked functional heterogeneity of HIV-1 Env proteins expressed by contemporaneous circulating viruses, and underscore the advantage of clonal analyses in characterizing the spectrum of functional properties of the genetically diverse viral populations present in a given patient.


Subject(s)
Genetic Variation , HIV Infections/virology , HIV-1/drug effects , HIV-1/physiology , env Gene Products, Human Immunodeficiency Virus/physiology , Adult , Antibodies, Monoclonal/immunology , Cell Line , HIV Antibodies/immunology , HIV Fusion Inhibitors/pharmacology , HIV-1/genetics , HIV-1/isolation & purification , Humans , Inhibitory Concentration 50 , Male , Phylogeny , Sequence Analysis, DNA , Sequence Homology, Amino Acid , env Gene Products, Human Immunodeficiency Virus/genetics
18.
J Clin Invest ; 123(9): 3848-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23979165

ABSTRACT

HIV-1 protease inhibitors (PIs) are among the most effective antiretroviral drugs. They are characterized by highly cooperative dose-response curves that are not explained by current pharmacodynamic theory. An unresolved problem affecting the clinical use of PIs is that patients who fail PI-containing regimens often have virus that lacks protease mutations, in apparent violation of fundamental evolutionary theory. Here, we show that these unresolved issues can be explained through analysis of the effects of PIs on distinct steps in the viral life cycle. We found that PIs do not affect virion release from infected cells but block entry, reverse transcription, and post-reverse transcription steps. The overall dose-response curves could be reconstructed by combining the curves for each step using the Bliss independence principle, showing that independent inhibition of multiple distinct steps in the life cycle generates the highly cooperative dose-response curves that make these drugs uniquely effective. Approximately half of the inhibitory potential of PIs is manifest at the entry step, likely reflecting interactions between the uncleaved Gag and the cytoplasmic tail (CT) of the Env protein. Sequence changes in the CT alone, which are ignored in current clinical tests for PI resistance, conferred PI resistance, providing an explanation for PI failure without resistance.


Subject(s)
Drug Resistance, Viral , HIV Protease Inhibitors/pharmacology , HIV Protease/metabolism , HIV-1/drug effects , Atazanavir Sulfate , CD4-Positive T-Lymphocytes/virology , Darunavir , Dose-Response Relationship, Drug , HEK293 Cells , HIV Protease/genetics , HIV-1/enzymology , HIV-1/physiology , Humans , Lopinavir/pharmacology , Mutation, Missense , Oligopeptides/pharmacology , Pyridines/pharmacology , Sulfonamides/pharmacology , Virus Internalization/drug effects , env Gene Products, Human Immunodeficiency Virus/physiology
19.
Cell Death Differ ; 17(5): 811-20, 2010 May.
Article in English | MEDLINE | ID: mdl-19876065

ABSTRACT

p53 binding protein-1 (53BP1) participates in checkpoint signaling during the DNA damage response (DDR) and during mitosis. In this study we report that 53BP1 aggregates in nuclear foci within syncytia elicited by the human immunodeficiency virus (HIV)-1 envelope. 53BP1 aggregation occurs as a consequence of nuclear fusion (karyogamy (KG)). It colocalizes partially with the promyelomonocytic leukemia protein (PML), and the ataxia telangiectasia mutated kinase (ATM), the two components of the DDR that mediate apoptosis induced by the HIV-1 envelope. ATM-dependent phosphorylation of 53BP1 on serines 25 and 1778 (53BP1S25P and 53BP1S1778P) occurs at these DNA damage foci. 53BP1S25P was also detected in syncytia present in the lymph nodes or frontal brain sections from HIV-1-infected carriers, as well as in peripheral blood mononucleated cells from HIV-1-infected individuals, correlating with viral load. Knockdown of 53BP1 caused HIV-1 envelope-induced syncytia to enter abnormal mitoses, leading to their selective destruction through mitochondrion-dependent and caspase-dependent pathways. In conclusion, depletion of 53BP1 triggers the demise of HIV-1-elicited syncytia through mitotic catastrophe.


Subject(s)
HIV-1/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Adult , Apoptosis/genetics , Apoptosis/physiology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Damage/genetics , DNA Damage/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Giant Cells/metabolism , HeLa Cells , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Male , Mitosis/genetics , Mitosis/physiology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1 , env Gene Products, Human Immunodeficiency Virus/metabolism , env Gene Products, Human Immunodeficiency Virus/physiology
20.
Autophagy ; 4(3): 273-5, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17993779

ABSTRACT

HIV-1 infection is characterized by a progressive CD4 T cell depletion. It is now accepted that apoptosis of uninfected bystander CD4 T lymphocytes plays a major role in AIDS development. Viral envelope glycoproteins (Env) are mainly involved in inducing this cell death process, but the mechanisms triggered by HIV-1 leading to immunodeficiency are still poorly understood. Recently, we have demonstrated that autophagy is a prerequisite for Env-mediated apoptosis in uninfected CD4 T cells, underlining its role in HIV-1 infection. However, occurrence of autophagy in HIV-1-infected cells has not yet been described. Several hypotheses are discussed, based on the comparison with data from other viral infections.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , HIV Infections/pathology , HIV-1/physiology , Animals , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , HIV Infections/metabolism , HIV Infections/virology , env Gene Products, Human Immunodeficiency Virus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL