Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Elife ; 102021 09 03.
Article in English | MEDLINE | ID: mdl-34477106

ABSTRACT

Class A G-protein-coupled receptors (GPCRs) normally function as monomers, although evidence from heterologous expression systems suggests that they may sometimes form homodimers and/or heterodimers. This study aims to evaluate possible functional interplay of endogenous µ- and δ-opioid receptors (MORs and DORs) in mouse neurons. Detecting GPCR dimers in native tissues, however, has been challenging. Previously, MORs and DORs co-expressed in transfected cells have been reported to form heterodimers, and their possible co-localization in neurons has been studied in knock-in mice expressing genetically engineered receptors fused to fluorescent proteins. Here, we find that single cholinergic neurons in the mouse striatum endogenously express both MORs and DORs. The receptors on neurons from live brain slices were fluorescently labeled with a ligand-directed labeling reagent, NAI-A594. The selective activation of MORs and DORs, with DAMGO (µ-agonist) and deltorphin (δ-agonist) inhibited spontaneous firing in all cells examined. In the continued presence of agonist, the firing rate returned to baseline as the result of receptor desensitization with the application of deltorphin but was less observed with the application of DAMGO. In addition, agonist-induced internalization of DORs but not MORs was detected. When MORs and DORs were activated simultaneously with [Met5]-enkephalin, desensitization of MORs was facilitated but internalization was not increased. Together, these results indicate that while MORs and DORs are expressed in single striatal cholinergic interneurons, the two receptors function independently.


Subject(s)
Cholinergic Neurons/metabolism , Corpus Striatum/metabolism , Interneurons/physiology , Receptors, Opioid, delta/genetics , Receptors, Opioid, mu/genetics , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism
2.
Elife ; 82019 10 07.
Article in English | MEDLINE | ID: mdl-31589142

ABSTRACT

Identifying neurons that have functional opioid receptors is fundamental for the understanding of the cellular, synaptic and systems actions of opioids. Current techniques are limited to post hoc analyses of fixed tissues. Here we developed a fluorescent probe, naltrexamine-acylimidazole (NAI), to label opioid receptors based on a chemical approach termed 'traceless affinity labeling'. In this approach, a high affinity antagonist naltrexamine is used as the guide molecule for a transferring reaction of acylimidazole at the receptor. This reaction generates a fluorescent dye covalently linked to the receptor while naltrexamine is liberated and leaves the binding site. The labeling induced by this reagent allowed visualization of opioid-sensitive neurons in rat and mouse brains without loss of function of the fluorescently labeled receptors. The ability to locate endogenous receptors in living tissues will aid considerably in establishing the distribution and physiological role of opioid receptors in the CNS of wild type animals.


Subject(s)
Brain Chemistry , Neurons/chemistry , Receptors, Opioid/analysis , Staining and Labeling/methods , Animals , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Fluorometry/methods , Mice, Inbred C57BL , Rats, Sprague-Dawley
3.
J Cell Biol ; 217(2): 541-553, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29288153

ABSTRACT

Mammalian cells replicate their chromosomes via a temporal replication program. The ASAR6 and ASAR15 genes were identified as loci that when disrupted result in delayed replication and condensation of entire human chromosomes. ASAR6 and ASAR15 are monoallelically expressed long noncoding RNAs that remain associated with the chromosome from which they are transcribed. The chromosome-wide effects of ASAR6 map to the antisense strand of an L1 retrotransposon within ASAR6 RNA, deletion or inversion of which delayed replication of human chromosome 6. Furthermore, ectopic integration of ASAR6 or ASAR15 transgenes into mouse chromosomes resulted in delayed replication and condensation, an increase in H3K27me3, coating of the mouse chromosome with ASAR RNA, and a loss of mouse Cot-1 RNA expression in cis. Targeting the antisense strand of the L1 within ectopically expressed ASAR6 RNA restored normal replication timing. Our results provide direct evidence that L1 antisense RNA plays a functional role in chromosome-wide replication timing of mammalian chromosomes.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 6/genetics , DNA Replication Timing/genetics , RNA, Antisense/genetics , RNA, Long Noncoding/genetics , Animals , Cells, Cultured , Humans , Long Interspersed Nucleotide Elements/genetics , Mice
4.
AIDS Res Hum Retroviruses ; 31(10): 992-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26114311

ABSTRACT

The CCR5 coreceptor amino terminus and extracellular (ECL) loops 1 and 2 have been implicated in HIV-1 infections, with species differences in these regions inhibiting zoonoses. Interactions of gp120 with CD4 and CCR5 reduce constraints on metastable envelope subunit gp41, enabling gp41 conformational changes needed for infection. We previously selected HIV-1JRCSF variants that efficiently use CCR5(Δ18) with a deleted amino terminus or CCR5(HHMH) with ECL2 from an NIH/Swiss mouse. Unexpectedly, the adaptive gp120 mutations were nearly identical, suggesting that they function by weakening gp120's grip on gp41 and/or by increasing interactions with ECL1. To analyze this and further wean HIV-1 from human CCR5, we selected variants using CCR5(HMMH) with murine ECL1 and 2 sequences. HIV-1JRCSF mutations adaptive for CCR5(Δ18) and CCR5(HHMH) were generally maladaptive for CCR5(HMMH), whereas the converse was true for CCR5(HMMH) adaptations. The HIV-1JRCSF variant adapted to CCR5(HMMH) also weakly used intact NIH/Swiss mouse CCR5. Our results strongly suggest that HIV-1JRCSF makes functionally critical contacts with human ECL1 and that adaptation to murine ECL1 requires multiple mutations in the crown of gp120's V3 loop.


Subject(s)
HIV Envelope Protein gp120/metabolism , HIV-1/physiology , Receptors, CCR5/metabolism , Receptors, HIV/metabolism , Virus Attachment , Adaptation, Biological , Animals , HIV Envelope Protein gp120/genetics , HIV-1/genetics , Humans , Mice , Mutation , Protein Binding , Protein Interaction Mapping
5.
J Virol ; 89(7): 3988-4001, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25631074

ABSTRACT

UNLABELLED: We have examined the interactions of wild-type (WT) and matrix protein-deleted (ΔMA) HIV-1 precursor Gag (PrGag) proteins in virus-producing cells using a biotin ligase-tagging approach. To do so, WT and ΔMA PrGag proteins were tagged with the Escherichia coli promiscuous biotin ligase (BirA*), expressed in cells, and examined. Localization patterns of PrGag proteins and biotinylated proteins overlapped, consistent with observations that BirA*-tagged proteins biotinylate neighbor proteins that are in close proximity. Results indicate that BirA*-tagged PrGag proteins biotinylated themselves as well as WT PrGag proteins in trans. Previous data have shown that the HIV-1 Envelope (Env) protein requires an interaction with MA for assembly into virions. Unexpectedly, ΔMA proteins biotinylated Env, whereas WT BirA*-tagged proteins did not, suggesting that the presence of MA made Env inaccessible to biotinylation. We also identified over 50 cellular proteins that were biotinylated by BirA*-tagged PrGag proteins. These included membrane proteins, cytoskeleton-associated proteins, nuclear transport factors, lipid metabolism regulators, translation factors, and RNA-processing proteins. The identification of these biotinylated proteins offers new insights into HIV-1 Gag protein trafficking and activities and provides new potential targets for antiviral interference. IMPORTANCE: We have employed a novel strategy to analyze the interactions of the HIV-1 structural Gag proteins, which involved tagging wild-type and mutant Gag proteins with a biotin ligase. Expression of the tagged proteins in cells allowed us to analyze proteins that came in close proximity to the Gag proteins as they were synthesized, transported, assembled, and released from cells. The tagged proteins biotinylated proteins encoded by the HIV-1 pol gene and neighbor Gag proteins, but, surprisingly, only the mutant Gag protein biotinylated the HIV-1 Envelope protein. We also identified over 50 cellular proteins that were biotinylated, including membrane and cytoskeletal proteins and proteins involved in lipid metabolism, nuclear import, translation, and RNA processing. Our results offer new insights into HIV-1 Gag protein trafficking and activities and provide new potential targets for antiviral interference.


Subject(s)
HIV-1/physiology , Host-Pathogen Interactions , Protein Interaction Mapping , gag Gene Products, Human Immunodeficiency Virus/metabolism , Biotin/analysis , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Genes, Reporter , Repressor Proteins/genetics , Repressor Proteins/metabolism , Staining and Labeling/methods
6.
J Virol ; 88(8): 4304-18, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24478426

ABSTRACT

UNLABELLED: HIV-1 membranes contain gp120-gp41 trimers. Binding of gp120 to CD4 and a coreceptor (CCR5 or CXCR4) reduces the constraint on metastable gp41, enabling a series of conformational changes that cause membrane fusion. An analytic difficulty occurs because these steps occur slowly and asynchronously within cohorts of adsorbed virions. We previously isolated HIV-1JRCSF variants that efficiently use CCR5 mutants severely damaged in the tyrosine-sulfated amino terminus or extracellular loop 2. Surprisingly, both independent adaptations included gp120 mutations S298N, F313L, and N403S, supporting other evidence that they function by weakening gp120's grip on gp41 rather than by altering gp120 binding to specific CCR5 sites. Although several natural HIV-1 isolates reportedly use CCR5(Δ18) (CCR5 with a deletion of 18 N-terminal amino acids, including the tyrosine-sulfated region) when the soluble tyrosine-sulfated peptide is present, we show that HIV-1JRCSF with the adaptive mutations [HIV-1JRCSF(Ad)] functions approximately 100 times more efficiently and that coreceptor activation is reversible, enabling synchronous efficient entry control under physiological conditions. This system revealed that three-stranded gp41 folding intermediates susceptible to the inhibitor enfuvirtide form slowly and asynchronously on cell surface virions but resolve rapidly, with virions generally forming only one target. Adsorbed virions asynchronously and transiently become competent for entry at 37°C but are inactivated if the CCR5 peptide is absent during their window of opportunity. This competency is conferred by endocytosis, which results in inactivation if the peptide is absent. For both wild-type and adapted HIV-1 isolates, early gp41 refolding steps obligatorily occur on cell surfaces, whereas the final step(s) is endosomal. This system powerfully dissects HIV-1 entry and inhibitor mechanisms. IMPORTANCE: We present a powerful means to reversibly and efficiently activate or terminate HIV-1 entry by adding or removing a tyrosine-sulfated CCR5 peptide from the culture medium. This system uses stable cell clones and a variant of HIV-1JRCSF with three adaptive mutations. It enabled us to show that CCR5 coreceptor activation is rapidly reversible and to dissect aspects of entry that had previously been relatively intractable. Our analyses elucidate enfuvirtide (T-20) function and suggest that HIV-1 virions form only one nonredundant membrane fusion complex on cell surfaces. Additionally, we obtained novel and conclusive evidence that HIV-1 entry occurs in an assembly line manner, with some steps obligatorily occurring on cell surfaces and with final membrane fusion occurring in endosomes. Our results were confirmed for wild-type HIV-1. Thus, our paper provides major methodological and mechanistic insights about HIV-1 infection.


Subject(s)
Endocytosis , HIV Envelope Protein gp41/pharmacology , HIV Fusion Inhibitors/pharmacology , HIV Infections/virology , HIV-1/physiology , Peptide Fragments/pharmacology , Peptides/pharmacology , Virus Internalization/drug effects , Amino Acid Sequence , CCR5 Receptor Antagonists , Cell Line , Endocytosis/drug effects , Enfuvirtide , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/physiopathology , HIV-1/drug effects , HIV-1/genetics , Humans , Molecular Sequence Data , Peptides/chemical synthesis , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Sequence Alignment , Tyrosine/analogs & derivatives , Tyrosine/chemistry
7.
Proc Natl Acad Sci U S A ; 109(20): 7829-34, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22547820

ABSTRACT

Despite structural knowledge of broadly neutralizing monoclonal antibodies (NMAbs) complexed to HIV-1 gp120 and gp41 envelope glycoproteins, virus inactivation mechanisms have been difficult to prove, in part because neutralization assays are complex and were previously not understood. Concordant with recent evidence that HIV-1 titers are determined by a race between entry of cell-attached virions and competing inactivation processes, we show that NMAb 2G12, which binds to gp120 N-glycans with α (1, 2)-linked mannose termini and inhibits replication after passive transfer into patients, neutralizes by slowing entry of adsorbed virions. Accordingly, apparent neutralization is attenuated when a kinetically competing virus inactivation pathway is blocked. Moreover, removing 2G12 from media causes its dissociation from virions coupled to accelerated entry and restored infectivity, demonstrating the reversibility of neutralization. A difference between 2G12 dissociation and infectivity recovery rates implies that the inhibited complexes at virus-cell junctions contain several 2G12's that must dissociate before entry commences. Quantitative microscopy of 2G12 binding and dissociation from single virions and studies using a split CCR5 coreceptor suggest that 2G12 competitively inhibits interactions between gp120's V3 loop and the tyrosine sulfate-containing CCR5 amino terminus, thereby reducing assembly of complexes that catalyze entry. These results reveal a unique reversible kinetic mechanism for neutralization by an antibody that binds near a critical V3 region in the glycan shield of gp120.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/immunology , HIV Envelope Protein gp120/metabolism , HIV-1/immunology , Models, Molecular , Virus Internalization , Antibodies, Monoclonal/immunology , Broadly Neutralizing Antibodies , Crystallography , HIV Antibodies , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/immunology , HeLa Cells , Humans , Kinetics , Microscopy, Fluorescence , Protein Binding
8.
J Virol ; 84(6): 3106-10, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20042508

ABSTRACT

By using immunofluorescence microscopy to observe and analyze freshly made HIV-1 virions adsorbed onto cells, we found that they are inherently highly infectious, rather than predominantly defective as previously suggested. Surprisingly, polycations enhance titers 20- to 30-fold by stabilizing adsorption and preventing a previously undescribed process of rapid dissociation, strongly implying that infectivity assays for many viruses are limited not only by inefficient virus diffusion onto cells but also by a postattachment race between entry and dissociation. This kinetic competition underlies inhibitory effects of CCR5 antagonists and explains why adaptive HIV-1 mutations overcome many cell entry limitations by accelerating entry.


Subject(s)
Cells, Cultured/virology , HIV-1/metabolism , HIV-1/pathogenicity , Virion/pathogenicity , Adsorption , CCR5 Receptor Antagonists , HIV-1/genetics , HeLa Cells , Humans , Virion/metabolism , Virus Inactivation , Virus Internalization
9.
J Virol ; 83(16): 8289-92, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19474095

ABSTRACT

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.


Subject(s)
Cell Line/virology , HIV Infections/immunology , HIV-1/immunology , Leukemia Virus, Murine , Neutralization Tests , Cell Line/immunology , HIV Infections/virology , HIV-1/chemistry , Humans , Specimen Handling
10.
J Mol Biol ; 374(1): 64-79, 2007 Nov 16.
Article in English | MEDLINE | ID: mdl-17920626

ABSTRACT

Binding of the human immunodeficiency virus (HIV-1) envelope glycoprotein gp120 to the CCR5 co-receptor reduces constraints on the metastable transmembrane subunit gp41, thereby enabling gp41 refolding, fusion of viral and cellular membranes, and infection. We previously isolated adapted HIV-1(JRCSF) variants that more efficiently use mutant CCR5s, including CCR5(Delta18) lacking the important tyrosine sulfate-containing amino terminus. Effects of mutant CCR5 concentrations on HIV-1 infectivities were highly cooperative, implying that several may be required. However, because wild-type CCR5 efficiently mediates infections at trace concentrations that were difficult to measure accurately, analyses of its cooperativity were not feasible. New HIV-1(JRCSF) variants efficiently use CCR5(HHMH), a chimera containing murine extracellular loop 2. The adapted virus induces large syncytia in cells containing either wild-type or mutant CCR5s and has multiple gp120 mutations that occurred independently in CCR5(Delta18)-adapted virus. Accordingly, these variants interchangeably use CCR5(HHMH) or CCR5(Delta18). Additional analyses strongly support a novel energetic model for allosteric proteins, implying that the adaptive mutations reduce quaternary constraints holding gp41, thus lowering the activation energy barrier for membrane fusion without affecting bonds to specific CCR5 sites. In accordance with this mechanism, highly adapted HIV-1s require only one associated CCR5(HHMH), whereas poorly adapted viruses require several. However, because they are allosteric ensembles, complexes with additional co-receptors fuse more rapidly and efficiently than minimal ones. Similarly, wild-type HIV-1(JRCSF) is highly adapted to wild-type CCR5 and minimally requires one. The adaptive mutations cause resistances to diverse entry inhibitors and cluster appropriately in the gp120 trimer interface overlying gp41. We conclude that membrane fusion complexes are allosteric machines with an ensemble of compositions, and that HIV-1 adapts to entry limitations by gp120 mutations that reduce its allosteric hold on gp41. These results provide an important foundation for understanding the mechanisms that control membrane fusion and HIV-1's facile adaptability.


Subject(s)
Allosteric Regulation , HIV Envelope Protein gp120/physiology , HIV-1/physiology , Membrane Fusion , Receptors, CCR5/physiology , Adaptation, Biological , Animals , COS Cells , Chlorocebus aethiops , Flow Cytometry , HIV Envelope Protein gp120/chemistry , HeLa Cells , Humans , Models, Molecular , Mutation , Receptors, CCR5/chemistry , Sulfates/chemistry , Virus Replication
11.
Retrovirology ; 4: 55, 2007 Aug 08.
Article in English | MEDLINE | ID: mdl-17686153

ABSTRACT

BACKGROUND: HIV-1 envelope glycoprotein (Env) induces membrane fusion as a result of sequential binding to CD4 and chemokine receptors (CCR5 or CXCR4). The critical determinants of CCR5 coreceptor function are the N-terminal domain (Nt) and the second extracellular loop. However, mutations in gp120 adapt HIV-1 to grow on cells expressing the N-terminally truncated CCR5(Delta 18) (Platt et al., J. Virol. 2005, 79: 4357-68). RESULTS: We have functionally characterized the adapted Env (designated Env(NYP)) using a quantitative cell-cell fusion assay. The rate of fusion with target cells expressing wild-type CCR5 and the resistance to fusion inhibitors was virtually identical for wild-type Env and Env(NYP), implying that the coreceptor affinity had not increased as a result of adaptation. In contrast, Env(NYP)-induced fusion with cells expressing CCR5(Delta 18) occurred at a slower rate and was extremely sensitive to the CCR5 binding inhibitor, Sch-C. Resistance to Sch-C drastically increased after pre-incubation of Env(NYP)- and CCR5(Delta 18)-expressing cells at a temperature that was not permissive to fusion. This indicates that ternary Env(NYP)-CD4-CCR5(Delta 18) complexes accumulate at sub-threshold temperature and that low-affinity interactions with the truncated coreceptor are sufficient for triggering conformational changes in the gp41 of Env(NYP) but not in wild-type Env. We also demonstrated that the ability of CCR5(Delta 18) to support fusion and infection mediated by wild-type Env can be partially reconstituted in the presence of a synthetic sulfated peptide corresponding to the CCR5 Nt. Pre-incubation of wild-type Env- and CCR5(Delta 18)-expressing cells with the sulfated peptide at sub-threshold temperature markedly increased the efficiency of fusion. CONCLUSION: We propose that, upon binding the Nt region of CCR5, wild-type Env acquires the ability to productively engage the extracellular loop(s) of CCR5 - an event that triggers gp41 refolding and membrane merger. The adaptive mutations in Env(NYP) enable it to more readily release its hold on gp41, even when it interacts weakly with a severely damaged coreceptor in the absence of the sulfopeptide.


Subject(s)
HIV Envelope Protein gp120/metabolism , HIV-1/physiology , Membrane Fusion , Receptors, CCR5/metabolism , Virus Internalization , Amino Acid Sequence , Anti-HIV Agents/pharmacology , CCR5 Receptor Antagonists , Cyclic N-Oxides/pharmacology , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Fusion Inhibitors/pharmacology , HIV-1/genetics , HeLa Cells , Humans , Kinetics , Membrane Fusion/genetics , Mutation , Oximes , Piperidines/pharmacology , Protein Binding , Protein Conformation , Protein Folding , Protein Structure, Tertiary , Pyridines/pharmacology , Receptors, CCR5/genetics , Sequence Deletion , Temperature
12.
J Biol Chem ; 280(33): 29570-7, 2005 Aug 19.
Article in English | MEDLINE | ID: mdl-15975927

ABSTRACT

Molecular mimicry of chemokine ligands has been described for several pathogens. Toxoplasma gondii produces a protein, cyclophilin-18 (C-18), which binds to the human immunodeficiency virus (HIV) co-receptor CCR5 and inhibits fusion and infection of T cells and macrophages by R5 viruses but not by X4 viruses. We recently identified structural determinants of C-18 required for anti-HIV activity (Yarovinsky, F., Andersen, J. F., King, L. R., Caspar, P., Aliberti, J., Golding, H., and Sher, A. (2004) J. Biol. Chem. 279, 53635-53642). Here we have elucidated the fine specificity of CCR5 residues involved in binding and HIV inhibitory potential of C-18. To delineate the regions of CCR5 involved in C-18 binding, we analyzed C-18 inhibition of cells expressing CXCR4/CCR5 chimeric receptors and CCR5 with a truncated N terminus (Delta2-19). These experiments identified a critical role for the N terminus of CCR5 in C-18 binding and anti-HIV activity. Studies with a large panel of CCR5 N-terminal peptides, including Tyr-sulfated analogues, truncated peptides, and alanine-scanning mutants, suggested that each of the 12-17 amino acids in the N terminus of CCR5 are essential for C-18 binding and inhibitory activity. Tyr sulfation did not improve C-18 reactivity. This finding is of interest because the same CCR5 N-terminal region was shown previously to play a key role in binding of HIV-1 envelope glycoproteins. The elucidation of the functional C-18-binding mechanism may help in the rational design of novel antiviral agents against HIV.


Subject(s)
Cyclophilins/pharmacology , HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Protozoan Proteins/pharmacology , Receptors, CCR5/physiology , Toxoplasma/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cyclophilins/metabolism , Humans , Mice , Molecular Sequence Data , Receptors, CCR5/chemistry
13.
J Virol ; 79(7): 4347-56, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15767435

ABSTRACT

Replication of human immunodeficiency virus type 1 (HIV-1) in diverse conditions limiting for viral entry into cells frequently leads to adaptive mutations in the V3 loop of the gp120 envelope glycoprotein. This has suggested that the V3 loop limits the efficiencies of HIV-1 infections, possibly by directly affecting gp120-coreceptor affinities. In contrast, V3 loop mutations that enable HIV-1(JR-CSF) to use the low-affinity mutant coreceptor CCR5(Y14N) are shown here to have negligible effects on the virus-coreceptor affinity but to dramatically accelerate the irreversible conformational conversion of the envelope gp41 subunits from a three-stranded coil into a six-helix bundle. This slow step is blocked irreversibly by the inhibitor T-20. To further evaluate the role of entry rates in controlling infection efficiencies and viral adaptations, we developed methods to quantitatively measure viral entry kinetics. The virions were adsorbed by spinoculation at 4 degrees C onto HeLa-CD4/CCR5 cell clones that either had limiting or saturating concentrations of CCR5. After warming to 37 degrees C, the completion of entry was monitored over time by the resistance of infections to the competitive CCR5 inhibitor TAK-779. Our results suggest that the efficiency of entry of cell-attached infectious HIV-1 is principally controlled by three kinetic processes. The first is a lag phase that is caused in part by the concentration-dependent reversible association of virus with CD4 and CCR5 to form an equilibrium assemblage of complexes. Second, this assembly step lowers but does not eliminate a large activation energy barrier for a rate-limiting, CCR5-dependent conformational change in gp41 that is sensitive to blockage by T-20. The rate of infection therefore depends on the fraction of infectious virions that are sufficiently saturated with CCR5 to undergo this conformational change and on the magnitude of the activation energy barrier. Although only a small fraction of fully assembled viral complexes overcome this barrier per hour, the ensuing steps of entry are rapidly completed within 5 to 10 min. Thus, this barrier limits the overall flow rate at which the attached virions enter cells, but it has no effect on the lag time that precedes this entry flow. Third, a relatively rapid and kinetically dominant process of viral inactivation, which may partly involve endocytosis, competes with infectious viral entry. Our results suggest that the V3 loop of gp120 has a major effect on the rate-limiting coreceptor-dependent conformational change in gp41 and that adaptive viral mutations, including V3 loop mutations, function kinetically by accelerating this inherently slow step in the entry pathway.


Subject(s)
HIV Fusion Inhibitors/pharmacology , HIV-1/physiology , Virus Replication/drug effects , Amides/pharmacology , Anti-HIV Agents/pharmacology , CCR5 Receptor Antagonists , CD4 Antigens/physiology , Enfuvirtide , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/physiology , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/pharmacology , HIV Envelope Protein gp41/physiology , HIV-1/drug effects , HeLa Cells , Humans , Kinetics , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptide Fragments/physiology , Protein Conformation/drug effects , Quaternary Ammonium Compounds/pharmacology , Receptors, CCR5/genetics , Receptors, CCR5/physiology
14.
J Virol ; 79(7): 4357-68, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15767436

ABSTRACT

By selecting the R5 human immunodeficiency virus type 1 (HIV-1) strain JR-CSF for efficient use of a CCR5 coreceptor with a badly damaged amino terminus [i.e., CCR5(Y14N)], we previously isolated variants that weakly utilize CCR5(Delta18), a low-affinity mutant lacking the normal tyrosine sulfate-containing amino-terminal region of the coreceptor. These previously isolated HIV-1(JR-CSF) variants contained adaptive mutations situated exclusively in the V3 loop of their gp120 envelope glycoproteins. We now have weaned the virus from all dependency on the CCR5 amino terminus by performing additional selections with HeLa-CD4 cells that express only a low concentration of CCR5(Delta18). The adapted variants had additional mutations in their V3 loops, as well as one in the V2 stem (S193N) and four alternative mutations in the V4 loop that eliminated the same N-linked oligosaccharide from position N403. Assays using pseudotyped viruses suggested that these new gp120 mutations all made strong contributions to use of CCR5(Delta18) by accelerating a rate-limiting CCR5-dependent conformational change in gp41 rather than by increasing viral affinity for this damaged coreceptor. Consistent with this interpretation, loss of the V4 N-glycan at position N403 also enhanced HIV-1 use of a different low-affinity CCR5 coreceptor with a mutation in extracellular loop 2 (ECL2) [i.e., CCR5(G163R)], whereas the double mutant CCR5(Delta18,G163R) was inactive. We conclude that loss of the N-glycan at position N403 helps to convert the HIV-1 envelope into a hair-trigger form that no longer requires strong interactions with both the CCR5 amino terminus and ECL2 but efficiently uses either site alone. These results demonstrate a novel functional role for a gp120 N-linked oligosaccharide and a high degree of adaptability in coreceptor usage by HIV-1.


Subject(s)
HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/physiology , HIV-1/genetics , HIV-1/physiology , Receptors, CCR5/physiology , Adaptation, Biological , Amides/pharmacology , Amino Acid Substitution , Cell Line , Cloning, Molecular , DNA Mutational Analysis , Enfuvirtide , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/pharmacology , HIV Envelope Protein gp41/physiology , HIV Fusion Inhibitors/pharmacology , HeLa Cells , Humans , Models, Molecular , Mutation , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptide Fragments/physiology , Polysaccharides/chemistry , Polysaccharides/physiology , Quaternary Ammonium Compounds/pharmacology , Receptors, CCR5/chemistry , Receptors, CCR5/genetics , Virus Replication
15.
J Virol ; 76(21): 11133-8, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12368356

ABSTRACT

The viral infectivity factor (Vif) of human immunodeficiency virus type 1 (HIV-1) neutralizes an unidentified antiviral pathway that occurs only in nonpermissive (NP) cells. Using a yeast two-hybrid screen of a human lymphocyte cDNA library, we identified several potential Vif partners. One, the nuclear body protein Sp140, was found specifically in all NP cells (n = 12 cell lines tested; P < or = 0.001), and HIV-1 infection induced its partial dispersal from nuclear bodies into cytosolic colocalization with Vif. Our results implicate Sp140 in a response to HIV-1 that may be related to or coordinated with the pathway that inactivates HIV-1 lacking vif.


Subject(s)
Antigens, Nuclear/metabolism , Gene Products, vif/metabolism , HIV-1/metabolism , Transcription Factors/metabolism , Antigens, Nuclear/genetics , Binding Sites , Gene Products, vif/genetics , HL-60 Cells , HeLa Cells , Humans , Jurkat Cells , Lymphocytes/metabolism , Lymphocytes/virology , Mutagenesis , Transcription Factors/genetics , Tumor Cells, Cultured , U937 Cells , vif Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...