Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
AIDS Res Hum Retroviruses ; 35(10): 941-947, 2019 10.
Article in English | MEDLINE | ID: mdl-31280582

ABSTRACT

Fusion inhibitors are antiretroviral (ARV) drugs that prevent HIV-1 entry into host cells. Enfuvirtide (ENF) is the only ARV drug marketed in this class and, like other HIV drugs, it has been associated with the emergence and selection of therapeutic-resistant HIV-1 strains. The aims of this work were to develop a computational tool capable of identifying and classifying mutations associated with resistance to Enfuvirtide and to evaluate the prevalence of these mutations among the HIV-1 sequences deposited in public databases. The HIVfird (HIV-1 fusion inhibitor resistance detector) was developed using the PHP programming language, using 30 DNA bases obtained from the HIV-1 HXB2 gp41 protein as a reference. To assess the level of resistance in HIV-1 populations, sequences were retrieved from the Los Alamos National Laboratory (LANL) database. The HIVfird is hosted at www.hivfird.ics.ufba.br, fully functional and available for public use. Twenty-five amino acid substitutions and 15 combinations were found to be associated with some level of resistance to ENF. These mutations are located at positions 36-45 of gp41, with 36, 38, 43, and 44 having the greatest diversity and frequency of variations associated with drug resistance. Resistance mutations were found in 3.16% and 4.67% of the circulating HIV-1 isolates in the world and Brazil, respectively. Subtype B showed a significantly higher ENF resistance rate (4.9%) compared to other genetic forms, while subtype C presented the lowest rate (0.9%). We present here HIVfird, an online tool that might assist in the therapeutic management of HIV-1 patients with multiple drug failure and in population-based analysis of drug resistance.


Subject(s)
DNA Mutational Analysis/methods , DNA, Viral/genetics , Drug Resistance, Viral , Enfuvirtide/pharmacokinetics , HIV Envelope Protein gp41/genetics , HIV Fusion Inhibitors/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Mutation , Software , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Databases, Genetic , Drug Resistance, Viral/genetics , Enfuvirtide/therapeutic use , Global Health , HIV Envelope Protein gp41/physiology , HIV Fusion Inhibitors/therapeutic use , HIV Infections/virology , HIV-1/classification , HIV-1/genetics , HIV-1/isolation & purification , Humans , Internet , Mutation, Missense , Sequence Alignment , Sequence Homology, Nucleic Acid
2.
J Virol ; 93(11)2019 06 01.
Article in English | MEDLINE | ID: mdl-30894471

ABSTRACT

Binding of the gp120 surface subunit of the envelope glycoprotein (Env) of HIV-1 to CD4 and chemokine receptors on target cells triggers refolding of the gp41 transmembrane subunit into a six-helix bundle (6HB) that promotes fusion between virus and host cell membranes. To elucidate details of Env entry and potential differences between viruses that use CXCR4 (X4) or CCR5 (R5) coreceptors, we generated viruses that are resistant to peptide fusion inhibitors corresponding to the first heptad repeat region (HR1) of gp41 that target fusion-intermediate conformations of Env. Previously we reported that an R5 virus selected two resistance pathways, each defined by an early gp41 resistance mutation in either HR1 or the second heptad repeat (HR2), to escape inhibition by an HR1 peptide, but preferentially selected the HR1 pathway to escape inhibition by a trimer-stabilized HR1 peptide. Here, we report that an X4 virus selected the same HR1 and HR2 resistance pathways as the R5 virus to escape inhibition by the HR1 peptide. However, in contrast to the R5 virus, the X4 virus selected a unique mutation in HR2 to escape inhibition by the trimer-stabilized peptide. Significantly, both of these X4 and R5 viruses acquired gp41 resistance mutations that improved the thermostability of the six-helix bundle, but they selected different gp120 adaptive mutations. These findings show that these X4 and R5 viruses use a similar resistance mechanism to escape from HR1 peptide inhibition but different gp120-gp41 interactions to regulate Env conformational changes.IMPORTANCE HIV-1 fuses with cells when the gp41 subunit of Env refolds into a 6HB after binding to cellular receptors. Peptides corresponding to HR1 or HR2 interrupt gp41 refolding and inhibit HIV infection. Previously, we found that a CCR5 coreceptor-tropic HIV-1 acquired a key HR1 or HR2 resistance mutation to escape HR1 peptide inhibitors but only the key HR1 mutation to escape a trimer-stabilized HR1 peptide inhibitor. Here, we report that a CXCR4 coreceptor-tropic HIV-1 selected the same key HR1 or HR2 mutations to escape inhibition by the HR1 peptide but different combinations of HR1 and HR2 mutations to escape the trimer-stabilized HR1 peptide. All gp41 mutations enhance 6HB stability to outcompete inhibitors, but gp120 adaptive mutations differed between these R5 and X4 viruses, providing new insights into gp120-gp41 functional interactions affecting Env refolding during HIV entry.


Subject(s)
HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp41/metabolism , HIV-1/genetics , Anti-Retroviral Agents/pharmacology , Drug Resistance, Viral/drug effects , Glycoproteins/genetics , HEK293 Cells , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp41/physiology , HIV Fusion Inhibitors/pharmacology , HIV Infections/genetics , HIV Seropositivity , HIV-1/physiology , Humans , Mutation , Protein Conformation , Receptors, CCR5/genetics , Receptors, CXCR4/genetics
3.
J Virol ; 90(23): 10587-10599, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27654295

ABSTRACT

The trimeric HIV-1 envelope glycoprotein spike (Env) mediates viral entry into cells by using a spring-loaded mechanism that allows for the controlled insertion of the Env fusion peptide into the target membrane, followed by membrane fusion. Env is the focus of vaccine research aimed at inducing protective immunity by antibodies as well as efforts to develop drugs that inhibit the viral entry process. The molecular factors contributing to Env stability and decay need to be understood better in order to optimally design vaccines and therapeutics. We generated viruses with resistance to VIR165, a peptidic inhibitor that binds the fusion peptide of the gp41 subunit and prevents its insertion into the target membrane. Interestingly, a number of escape viruses acquired substitutions in the C1 domain of the gp120 subunit (A60E, E64K, and H66R) that rendered these viruses dependent on the inhibitor. These viruses could infect target cells only when VIR165 was present after CD4 binding. Furthermore, the VIR165-dependent viruses were resistant to soluble CD4-induced Env destabilization and decay. These data suggest that VIR165-dependent Env proteins are kinetically trapped in the unliganded state and require the drug to negotiate CD4-induced conformational changes. These studies provide mechanistic insight into the action of the gp41 fusion peptide and its inhibitors and provide new ways to stabilize Env trimer vaccines. IMPORTANCE: Because of the rapid development of HIV-1 drug resistance, new drug targets need to be explored continuously. The fusion peptide of the envelope glycoprotein can be targeted by anchor inhibitors. Here we describe virus escape from the anchor inhibitor VIR165. Interestingly, some escape viruses became dependent on the inhibitor for cell entry. We show that the identified escape mutations stabilize the ground state of the envelope glycoprotein and should thus be useful in the design of stabilized envelope-based HIV vaccines.


Subject(s)
HIV Envelope Protein gp120/physiology , HIV Envelope Protein gp41/physiology , HIV-1/physiology , Virus Internalization , Amino Acid Sequence , Amino Acid Substitution , Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacology , Benzylamines , CD4 Antigens/metabolism , CD4 Antigens/pharmacology , Cyclams , Genes, env , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/genetics , HIV-1/drug effects , HIV-1/genetics , Heterocyclic Compounds/pharmacology , Humans , Models, Biological , Models, Molecular , Mutation , Protein Conformation , Protein Stability , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Virus Internalization/drug effects
4.
Biochem Biophys Res Commun ; 473(4): 1185-1190, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27079239

ABSTRACT

Enveloped virus, such as HIV-1, employs membrane fusion mechanism to invade into host cell. HIV-1 gp41 ectodomain uses six-helix bundle configuration to accomplish this process. Using molecular dynamic simulations, we confirmed the stability of this six-helix bundle by showing high occupancy of hydrogen bonds and hydrophobic interactions. Key residues and interactions important for the bundle integration were characterized by force-induced unfolding simulations of six-helix bundle, exhibiting the collapse order of these groups of interactions. Moreover, our results in some way concerted with a previous theory that the formation of coiled-coil choose a route which involved cooperative interactions between the N-terminal and C-terminal helix.


Subject(s)
HIV Envelope Protein gp41/physiology , HIV Envelope Protein gp41/ultrastructure , Membrane Fusion/physiology , Models, Biological , Molecular Dynamics Simulation , Computer Simulation , HIV Envelope Protein gp41/chemistry , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Mechanotransduction, Cellular/physiology , Membrane Fluidity/physiology , Micromanipulation/methods , Protein Conformation , Protein Folding , Protein Subunits , Stress, Mechanical , Structure-Activity Relationship
5.
Virol Sin ; 31(3): 207-18, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27117672

ABSTRACT

A transmission bottleneck occurs during each human immunodeficiency virus (HIV) transmission event, which allows only a few viruses to establish new infection. However, the genetic characteristics of the transmitted viruses that are preferentially selected have not been fully elucidated. Here, we analyzed amino acids changes in the envelope protein during simian immunodeficiency virus (SIV)/HIV deep transmission history and current HIV evolution within the last 15-20 years. Our results confirmed that the V1V2 region of gp120 protein, particularly V1, was preferentially selected. A shorter V1 region was preferred during transmission history, while during epidemic, HIV may evolve to an expanded V1 region gradually and thus escape immune recognition. We then constructed different HIV-1 V1 mutants using different HIV-1 subtypes to elucidate the role of the V1 region in envelope function. We found that the V1 region, although highly variable, was indispensable for virus entry and infection, probably because V1 deletion mutants exhibited impaired processing of gp160 into mature gp120 and gp41. Additionally, the V1 region affected Env incorporation. These results indicated that the V1 region played a critical role in HIV transmission and infection.


Subject(s)
HIV Envelope Protein gp120/physiology , HIV Infections/transmission , HIV-1/physiology , Membrane Glycoproteins/physiology , Peptide Fragments/physiology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/physiology , Viral Envelope Proteins/physiology , Animals , HEK293 Cells , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/metabolism , HIV Envelope Protein gp41/physiology , HIV Infections/virology , HIV-1/genetics , HIV-2/genetics , HIV-2/physiology , Humans , Membrane Fusion/physiology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
6.
Retrovirology ; 12: 81, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26399966

ABSTRACT

BACKGROUND: The structure of HIV-1 envelope glycoprotein (Env) is flexible and heterogeneous on whole virions. Although functional Env complexes are thought to require trimerization of cleaved gp41/gp120 heterodimers, variable processing can result in the potential incorporation of non-functional uncleaved proteins (gp160), non-trimeric arrangements of gp41/gp120 heterodimers, and gp120 depleted gp41 stumps. The potential distribution of functional and non-functional Env forms across replication-competent viral populations may have important implications for neutralizing and non-neutralizing antibody functions. This study applied an immuno-bead viral capture assay (VCA) to interrogate the potential distribution (heterologous vs homologous) of functional and non-functional forms of virion associated Env. RESULTS: The VCA revealed a significant association between depletion of infectious virions and virion Env incorporation, but not between infectivity and p24-gag. Three distinct subpopulations of virions were identified within pools of genetically homogenous viral particles. Critically, a significant subpopulation of infectious virions were exclusively captured by neutralizing antibodies (nAbs) indicative of a homologous distribution of functional trimeric Env forms. A second infectious subpopulation bound both neutralizing and non-neutralizing antibodies (nnAbs) representative of a heterologous distribution of Env forms, while a third non-infectious subpopulation was predominantly bound by nnAbs recognizing gp41 stumps. CONCLUSIONS: The observation that a distinct and significant subpopulation of infectious virions is exclusively captured by neutralizing antibodies has important implications for understanding antibody binding and neutralization, as well as other antibody effector functions.


Subject(s)
HIV Envelope Protein gp120/physiology , HIV Envelope Protein gp160/physiology , HIV-1/physiology , Virion/isolation & purification , Virion/physiology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp160/chemistry , HIV Envelope Protein gp160/immunology , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/immunology , HIV Envelope Protein gp41/physiology , HIV-1/immunology , Humans , Immunoassay , Protein Binding , Virion/immunology
7.
Proc Natl Acad Sci U S A ; 112(24): 7575-80, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26034275

ABSTRACT

Lentiviruses such as HIV-1 encode envelope glycoproteins (Env) with long cytoplasmic tails (CTs) that include motifs mediating interactions with host-cell-trafficking factors. We demonstrated recently that Rab11-family interacting protein 1C (FIP1C) is required for CT-dependent incorporation of Env into HIV-1 particles. Here, we used viruses bearing targeted substitutions within CT to map the FIP1C-dependent incorporation of Env. We identified YW795 as a critical motif mediating cell-type-dependent Env incorporation. Disruption of YW795 reproduced the cell-type-dependent particle incorporation of Env that had previously been observed with large truncations of CT. A revertant virus bearing a single amino acid change near the C terminus of CT restored wild-type levels of Env incorporation, Gag-Env colocalization on the plasma membrane, and viral replication. These findings highlight the importance of YW795 in the cell-type-dependent incorporation of Env and support a model of HIV assembly in which FIP1C/RCP mediates Env trafficking to the particle assembly site.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , HIV Envelope Protein gp41/physiology , HIV-1/physiology , Membrane Proteins/physiology , Amino Acid Motifs , Amino Acid Substitution , Cell Line , Cell Membrane/virology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/genetics , HIV-1/genetics , HeLa Cells , Humans , Mutagenesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/virology , Tyrosine/chemistry , Virion/physiology , Virus Assembly , Virus Replication , gag Gene Products, Human Immunodeficiency Virus/metabolism
8.
ACS Chem Biol ; 10(5): 1247-57, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25646644

ABSTRACT

The conformational rearrangement of N- and C-heptad repeats (NHR, CHR) of the HIV-1 glycoprotein-41 (gp41) ectodomain into a trimer of hairpins triggers virus-cell fusion by bringing together membrane-spanning N- and C-terminal domains. Peptides derived from the NHR and CHR inhibit fusion by targeting a prehairpin intermediate state of gp41. Typically, peptides derived from the CHR are low nanomolar inhibitors, whereas peptides derived from the NHR are low micromolar inhibitors. Here, we describe the inhibitory activity of swapped-domain gp41 mimics of the form CHR-loop-NHR, which were designed to form reverse hairpin trimers exposing NHR grooves. We observed low nanomolar inhibition of HIV fusion in constructs that possessed the following properties: an extended NHR C-terminus, an exposed conserved hydrophobic pocket on the NHR, high helical content, and trimer stability. Low nanomolar activity was independent of CHR length. CD studies in membrane mimetic dodecylphosphocholine micelles suggested that bioactivity could be related to the ability of the inhibitors to interact with a membrane-associated prehairpin intermediate. The swapped-domain design resolves the problem of unstable and weakly active NHR peptides and suggests a different mechanism of action from that of CHR peptides in inhibition of HIV-1 fusion.


Subject(s)
HIV Envelope Protein gp41/physiology , HIV Infections/prevention & control , Amino Acid Sequence , Biopolymers/chemistry , Cell Line , Circular Dichroism , HIV Envelope Protein gp41/chemistry , Humans , Molecular Sequence Data , Protein Structure, Secondary
9.
PLoS One ; 7(9): e44874, 2012.
Article in English | MEDLINE | ID: mdl-22970321

ABSTRACT

During the process of HIV-1 fusion with the target cell, the N-terminal heptad repeat (NHR) of gp41 interacts with the C-terminal heptad repeat (CHR) to form fusogenic six-helix bundle (6-HB) core. We previously identified a crucial residue for 6-HB formation and virus entry--Lys63 (K63) in the C-terminal region of NHR (aa 54-70), which forms a hydrophobic cavity. It can form an important salt bridge with Asp121 (D121) in gp41 CHR. Here, we found another important conserved residue for virus fusion and entry, Arg46 (R46), in the N-terminal region of NHR (aa 35-53), which forms a hydrogen bond with a polar residue, Asn43 (N43), in NHR, as a part of the hydrogen-bond network. R46 can also form a salt bridge with a negatively charged residue, Glu137 (E137), in gp41 CHR. Substitution of R46 with the hydrophobic residue Ala (R46A) or the negatively charged residue Glu (R46E) resulted in disruption of the hydrogen bond network, breakage of the salt bridge and reduction of 6-HB's stability, leading to impairment of viral fusion and decreased inhibition of N36, an NHR peptide. Similarly, CHR peptide C34 with substitution of E137 for Ala (E137A) or Arg (E137R) also exhibited reduced inhibitory activity against HIV-1 infection and HIV-1-mediated cell-to-cell fusion. These results suggest that the positively charged residue R46 and its hydrogen bond network, together with the salt bridge between R46 and E137, are important for viral fusion and entry and may therefore serve as a target for designing novel HIV fusion/entry inhibitors.


Subject(s)
Arginine/metabolism , HIV Envelope Protein gp41/physiology , Membrane Fusion , Amino Acid Sequence , Arginine/chemistry , Circular Dichroism , HIV Envelope Protein gp41/chemistry , Humans , Hydrogen Peroxide , Molecular Sequence Data , Repetitive Sequences, Amino Acid , Sequence Homology, Amino Acid
10.
Biochemistry ; 50(32): 6966-72, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21736372

ABSTRACT

HIV entry occurs by concerted conformational changes in the envelope protein complex on the surface of the virus. This complex is made up of a trimer of heterodimers of two subunits: surface subunit, gp120, and transmembrane subunit, gp41. Conformational changes in the envelope complex allow gp41 to mediate membrane fusion leading to exposure of two gp41 regions: N-heptad repeat (NHR) and C-heptad repeat (CHR). Peptides from the NHR or the CHR have been found to inhibit HIV entry. Herein we show that we can covalently inhibit HIV viral entry by permanently trapping the gp41 intermediate on the virus surface using a covalently reactive group on inhibitory peptides. This is evidence showing that vulnerable conformational intermediates exist transiently during HIV viral entry, and the details presented herein will facilitate development of envelope as a target for therapeutics and potential chemopreventive agents that could disable the virus before contact with the host cell.


Subject(s)
HIV Envelope Protein gp41/physiology , HIV-1/physiology , Membrane Fusion , Amino Acid Sequence , Cell Line , HIV Envelope Protein gp41/chemistry , Humans , Models, Molecular , Molecular Sequence Data
11.
J Biol Chem ; 286(31): 27156-66, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21659530

ABSTRACT

Although the HIV-1 Env gp120 and gp41 ectodomain have been extensively characterized in terms of structure and function, similar characterizations of the C-terminal tail (CTT) of HIV gp41 remain relatively limited and contradictory. The current study was designed to examine in detail CTT sequence conservation relative to gp120 and the gp41 ectodomain and to examine the conservation of predicted physicochemical and structural properties across a number of divergent HIV clades and groups. Results demonstrate that CTT sequences display intermediate levels of sequence evolution and diversity in comparison to the more diverse gp120 and the more conserved gp41 ectodomain. Despite the relatively high level of CTT sequence variation, the physicochemical properties of the lentivirus lytic peptide domains (LLPs) within the CTT are evidently highly conserved across clades/groups. Additionally, predictions using PEP-FOLD indicate a high level of structural similarity in the LLP regions that was confirmed by circular dichroism measurements of secondary structure of LLP peptides from clades B, C, and group O. Results demonstrate that LLP peptides adopt helical structure in the presence of SDS or trifluoroethanol but are predominantly unstructured in aqueous buffer. Thus, these data for the first time demonstrate strong conservations of characteristic CTT physicochemical and structural properties despite substantial sequence diversity, apparently indicating a delicate balance between evolutionary pressures and the conservation of CTT structure and associated functional roles in virus replication.


Subject(s)
Conserved Sequence , HIV Envelope Protein gp41/physiology , HIV-1/physiology , Virus Replication/physiology , Amino Acid Sequence , Circular Dichroism , HIV Envelope Protein gp41/chemistry , HIV-1/classification , Molecular Sequence Data , Phylogeny , Protein Structure, Secondary , Sequence Homology, Amino Acid
12.
J Exp Med ; 208(3): 439-54, 2011 Mar 14.
Article in English | MEDLINE | ID: mdl-21357743

ABSTRACT

Interference with virus entry is known to be the principle mechanism of HIV neutralization by antibodies, including 2F5 and 4E10, which bind to the membrane-proximal external region (MPER) of the gp41 envelope protein. However, to date, the precise molecular events underlying neutralization by MPER-specific antibodies remain incompletely understood. In this study, we investigated the capacity of these antibodies to irrevocably sterilize HIV virions. Long-term effects of antibodies on virions can differ, rendering neutralization either reversible or irreversible. MPER-specific antibodies irreversibly neutralize virions, and this capacity is associated with induction of gp120 shedding. Both processes have similar thermodynamic properties and slow kinetics requiring several hours. Antibodies directed to the CD4 binding site, V3 loop, and the MPER can induce gp120 shedding, and shedding activity is detected with high frequency in plasma from patients infected with divergent genetic HIV-1 subtypes. Importantly, as we show in this study, induction of gp120 shedding is closely associated with MPER antibody inhibition, constituting either a primary event leading to virion neutralization or representing an immediate consequence thereof, and thus needs to be factored into the mechanistic processes underlying their activity.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp41/immunology , HIV-1/immunology , Virus Shedding/immunology , Antibodies, Monoclonal/immunology , Antibodies, Viral/physiology , Blotting, Western , HIV Envelope Protein gp120/physiology , HIV Envelope Protein gp41/physiology , HIV Infections/immunology , HIV Infections/virology , Humans , Kinetics , Neutralization Tests , Virion/immunology , Virus Shedding/physiology
13.
Enferm Infecc Microbiol Clin ; 29 Suppl 5: 45-50, 2011 Dec.
Article in Spanish | MEDLINE | ID: mdl-22305669

ABSTRACT

Determination of HIV-1 tropism is mandatory before using CCR5 antagonists in clinical practice. One drug of this class, maraviroc, has been approved for the treatment of HIV infection. The phenotypic assay, TrofileTM, was clinically validated in the clinical development program of maraviroc and has been widely used to select candidates for maraviroc therapy. Phenotypic tests, however, have the disadvantage of being complex, are costly and time-consuming, and their accessibility is limited, which hampers their routine use in clinical diagnosis. Genotypic assays, based on sequencing the third hypervariable (V3 loop) of the viral gene env, interpreted according to various genotypic bioinformatic tools, such as geno2pheno and PSSM, are faster and cheaper than phenotypic assays, and are also more accessible. In retrospective analyses of the maraviroc pivotal trials, genotypic methods using either conventional ("bulk") or deep-sequencing technology predicted virologic response to maraviroc similarly to phenotypic assays and are now included within several European recommendations to guide the clinical use of CCR5 antagonists.


Subject(s)
Anti-HIV Agents/pharmacology , CCR5 Receptor Antagonists , HIV Infections/drug therapy , HIV-1/physiology , Viral Tropism , Virology/methods , Algorithms , Anti-HIV Agents/therapeutic use , Clinical Trials as Topic , Cyclohexanes/pharmacology , Cyclohexanes/therapeutic use , Europe , Genes, env , Genotype , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/physiology , HIV Envelope Protein gp41/physiology , HIV Infections/virology , HIV-1/genetics , Humans , Maraviroc , Peptide Fragments/genetics , Phenotype , Practice Guidelines as Topic , Receptors, CCR5/physiology , Receptors, CXCR4/physiology , Retrospective Studies , Triazoles/pharmacology , Triazoles/therapeutic use , Viral Tropism/drug effects , Viral Tropism/genetics , Virus Internalization/drug effects
14.
PLoS Pathog ; 6(11): e1001195, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-21124990

ABSTRACT

The human monoclonal antibody (mAb) HK20 neutralizes a broad spectrum of primary HIV-1 isolates by targeting the highly conserved heptad repeat 1 (HR1) of gp41, which is transiently exposed during HIV-1 entry. Here we present the crystal structure of the HK20 Fab in complex with a gp41 mimetic 5-Helix at 2.3 Å resolution. HK20 employs its heavy chain CDR H2 and H3 loops to bind into a conserved hydrophobic HR1 pocket that is occupied by HR2 residues in the gp41 post fusion conformation. Compared to the previously described HR1-specific mAb D5, HK20 approaches its epitope with a different angle which might favor epitope access and thus contribute to its higher neutralization breadth and potency. Comparison of the neutralization activities of HK20 IgG, Fab and scFv employing both single cycle and multiple cycle neutralization assays revealed much higher potencies for the smaller Fab and scFv over IgG, implying that the target site is difficult to access for complete antibodies. Nevertheless, two thirds of sera from HIV-1 infected individuals contain significant titers of HK20-inhibiting antibodies. The breadth of neutralization of primary isolates across all clades, the higher potencies for C-clade viruses and the targeting of a distinct site as compared to the fusion inhibitor T-20 demonstrate the potential of HK20 scFv as a therapeutic tool.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/physiology , HIV-1/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Crystallization , Crystallography, X-Ray , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/metabolism , Humans , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Mutation/genetics , Neutralization Tests , Protein Conformation , Surface Plasmon Resonance
15.
Biochemistry ; 49(24): 5057-65, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20481578

ABSTRACT

On the basis of mutagenesis, biochemical, and structural studies, heptad repeat 1 of HIV gp41 (HR1) has been shown to play numerous critical roles in HIV entry, including interacting with gp120 in prefusion states and interacting with gp41 heptad repeat 2 (HR2) in the fusion state. Moreover, HR1 is the site of therapeutic intervention by enfuviritide, a peptide analogue of HR2. In this study, the functional importance of each amino acid residue in gp41 HR1 has been systematically examined by alanine scanning mutagenesis, with subsequent characterization of the mutagenic effects on folding (as measured by incorporation into virions), association with gp120, and membrane fusion. The mutational effects on entry can be grouped into three classes: (1) wild type (defined as >40% of wild-type entry), (2) impaired (defined as 5-40% of wild-type entry), and (3) nonfunctional (defined as <5% of wild-type entry). Interestingly, the majority of HR1 mutations (77%) exhibit impaired or nonfunctional entry. Surprisingly, effects of mutations on folding, association, or fusion are not correlated to heptad position; however, folding defects are most often found in the N-terminal region of HR1. Moreover, disruption of the gp41-gp120 interaction is correlated to the C-terminal region of HR1, suggesting that this region interacts most closely with gp120. In summary, the sensitivity of gp41 HR1 to alanine substitutions suggests that even subtle changes in the local environment may severely affect envelope function, thereby strengthening the notion that HR1 is an attractive site for therapeutic intervention.


Subject(s)
Alanine/genetics , HIV Envelope Protein gp120/physiology , HIV Envelope Protein gp41/genetics , HIV-1/physiology , Amino Acid Sequence , Amino Acid Substitution , Cell Line , HIV Envelope Protein gp41/physiology , HIV-1/genetics , Humans , Membrane Fusion , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Folding , Repetitive Sequences, Amino Acid , Virus Internalization
16.
Subcell Biochem ; 51: 77-108, 2010.
Article in English | MEDLINE | ID: mdl-20213541

ABSTRACT

Up to now less than a handful of viral cholesterol-binding proteins have been characterized, in HIV, influenza virus and Semliki Forest virus. These are proteins with roles in virus entry or morphogenesis. In the case of the HIV fusion protein gp41 cholesterol binding is attributed to a cholesterol recognition consensus (CRAC) motif in a flexible domain of the ectodomain preceding the trans-membrane segment. This specific CRAC sequence mediates gp41 binding to a cholesterol affinity column. Mutations in this motif arrest virus fusion at the hemifusion stage and modify the ability of the isolated CRAC peptide to induce segregation of cholesterol in artificial membranes.Influenza A virus M2 protein co-purifies with cholesterol. Its proton translocation activity, responsible for virus uncoating, is not cholesterol-dependent, and the transmembrane channel appears too short for integral raft insertion. Cholesterol binding may be mediated by CRAC motifs in the flexible post-TM domain, which harbours three determinants of binding to membrane rafts. Mutation of the CRAC motif of the WSN strain attenuates virulence for mice. Its affinity to the raft-non-raft interface is predicted to target M2 protein to the periphery of lipid raft microdomains, the sites of virus assembly. Its influence on the morphology of budding virus implicates M2 as factor in virus fission at the raft boundary. Moreover, M2 is an essential factor in sorting the segmented genome into virus particles, indicating that M2 also has a role in priming the outgrowth of virus buds.SFV E1 protein is the first viral type-II fusion protein demonstrated to directly bind cholesterol when the fusion peptide loop locks into the target membrane. Cholesterol binding is modulated by another, proximal loop, which is also important during virus budding and as a host range determinant, as shown by mutational studies.


Subject(s)
Carrier Proteins/metabolism , Cholesterol/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/physiology , Lipid Bilayers/metabolism , Membrane Microdomains/physiology , Molecular Sequence Data , Pancreatic Elastase , Protein Binding , Viral Fusion Proteins/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/physiology , Virus Internalization
17.
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
18.
J Virol ; 84(1): 201-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19846514

ABSTRACT

The entry of human immunodeficiency virus type 1 (HIV-1) into a target cell entails a series of conformational changes in the gp41 transmembrane glycoprotein that mediates the fusion of the viral and target cell membranes. A trimer-of-hairpins structure formed by the association of two heptad repeat (HR) regions of the gp41 ectodomain has been implicated in a late step of the fusion pathway. Earlier native and intermediate states of the protein are postulated to mediate the antiviral activity of the fusion inhibitor enfuvirtide and of broadly neutralizing monoclonal antibodies (NAbs), but the details of these structures remain unknown. Here, we report the identification and crystal structure of a dimerization domain in the C-terminal ectodomain of gp41 (residues 630 to 683, or C54). Two C54 monomers associate to form an asymmetric, antiparallel coiled coil with two distinct C-terminal alpha-helical overhangs. This dimer structure is conferred largely by interactions within a central core that corresponds to the sequence of enfuvirtide. The mutagenic alteration of the dimer interface severely impairs the infectivity of Env-pseudotyped viruses. Moreover, the C54 structure binds tightly to both the 2F5 and 4E10 NAbs and likely represents a potential intermediate conformation of gp41. These results should enhance our understanding of the molecular basis of the gp41 fusogenic structural transitions and thereby guide rational, structure-based efforts to design new fusion inhibitors and vaccine candidates intended to induce broadly neutralizing antibodies.


Subject(s)
HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/physiology , HIV-1/pathogenicity , Virus Internalization , Antibodies, Monoclonal/immunology , Crystallography, X-Ray , Enfuvirtide , HIV Envelope Protein gp41/immunology , HIV Fusion Inhibitors/chemistry , Humans , Membrane Fusion , Mutagenesis , Peptide Fragments/chemistry , Protein Conformation , Protein Multimerization
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...