Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Proc Natl Acad Sci U S A ; 116(21): 10504-10509, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31048506

ABSTRACT

To replicate in a new host, lentiviruses must adapt to exploit required host factors and evade species-specific antiviral proteins. Understanding how host protein variation drives lentivirus adaptation allowed us to expand the host range of HIV-1 to pigtail macaques. We have previously derived a viral swarm (in the blood of infected animals) that can cause AIDS in this new host. To further exploit this reagent, we generated infectious molecular clones (IMCs) from the viral swarm. We identified clones with high replicative capacity in pigtail peripheral blood mononuclear cells (PBMC) in vitro and used in vivo replication to select an individual IMC, named stHIV-A19 (for simian tropic HIV-1 clone A19), which recapitulated the phenotype obtained with the viral swarm. Adaptation of HIV-1 in macaques led to the acquisition of amino acid changes in viral proteins, such as capsid (CA), that are rarely seen in HIV-1-infected humans. Using stHIV-A19, we show that these CA changes confer a partial resistance to the host cell inhibitor Mx2 from pigtail macaques, but that complete resistance is associated with a fitness defect. Adaptation of HIV-1 to a new host will lead to a more accurate animal model and a better understanding of virus-host interactions.


Subject(s)
Adaptation, Biological , Disease Models, Animal , HIV Infections , HIV-1 , Animals , Capsid Proteins/genetics , Evolution, Molecular , Host Specificity , Macaca nemestrina , Virus Replication
2.
J Virol ; 94(1)2019 12 12.
Article in English | MEDLINE | ID: mdl-31619553

ABSTRACT

The matrix (MA) domain of HIV-1 Gag plays key roles in virus assembly by targeting the Gag precursor to the plasma membrane and directing the incorporation of the viral envelope (Env) glycoprotein into virions. The latter function appears to be in part dependent on trimerization of the MA domain of Gag during assembly, as disruption of the MA trimer interface impairs Env incorporation. Conversely, many MA mutations that impair Env incorporation can be rescued by compensatory mutations in the trimer interface. In this study, we sought to investigate further the biological significance of MA trimerization by isolating and characterizing compensatory mutations that rescue MA trimer interface mutants with severely impaired Env incorporation. By serially propagating MA trimerization-defective mutants in T cell lines, we identified a number of changes in MA, both within and distant from the trimer interface. The compensatory mutations located within or near the trimer interface restored Env incorporation and particle infectivity and permitted replication in culture. The structure of the MA lattice was interrogated by measuring the cleavage of the murine leukemia virus (MLV) transmembrane Env protein by the viral protease in MLV Env-pseudotyped HIV-1 particles bearing the MA mutations and by performing crystallographic studies of in vitro-assembled MA lattices. These results demonstrate that rescue is associated with structural alterations in MA organization and rescue of MA domain trimer formation. Our data highlight the significance of the trimer interface of the MA domain of Gag as a critical site of protein-protein interaction during HIV-1 assembly and establish the functional importance of trimeric MA for Env incorporation.IMPORTANCE The immature Gag lattice is a critical structural feature of assembling HIV-1 particles, which is primarily important for virion formation and release. While Gag forms a hexameric lattice, driven primarily by the capsid domain, the MA domain additionally trimerizes where three Gag hexamers meet. MA mutants that are defective for trimerization are deficient for Env incorporation and replication, suggesting a requirement for trimerization of the MA domain of Gag in Env incorporation. This study used a gain-of-function, forced viral evolution approach to rescue HIV-1 mutants that are defective for MA trimerization. Compensatory mutations that rescue virus replication do so by restoring Env incorporation and MA trimer formation. This study supports the importance of MA domain trimerization in HIV-1 replication and the potential of the trimer interface as a therapeutic target.


Subject(s)
HIV-1/genetics , Viral Matrix Proteins/chemistry , Virion/genetics , Virus Assembly , env Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/chemistry , Amino Acid Motifs , Amino Acid Substitution , Animals , Cell Line , Gene Expression , HIV-1/metabolism , HeLa Cells , Humans , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/metabolism , Mice , Models, Molecular , Mutation , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Multimerization , T-Lymphocytes/virology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Virion/metabolism , Virus Replication , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism
3.
J Virol ; 93(20)2019 10 15.
Article in English | MEDLINE | ID: mdl-31341054

ABSTRACT

The HIV/AIDS pandemic remains an important threat to human health. We have recently demonstrated that a novel microRNA (miR), miR-128, represses retrotransposon long interspaced element 1 (L1) by a dual mechanism, namely, by directly targeting the coding region of the L1 RNA and by repressing a required nuclear import factor (TNPO1). We have further determined that miR-128 represses the expression of all three TNPO proteins (transportins TNPO1, TNPO2, and TNPO3). Here, we establish that miR-128 also influences HIV-1 replication by repressing TNPO3, a factor that regulates HIV-1 nuclear import and viral; replication of TNPO3 is well established to regulate HIV-1 nuclear import and viral replication. Here, we report that type I interferon (IFN)-inducible miR-128 directly targets two sites in the TNPO3 mRNA, significantly downregulating TNPO3 mRNA and protein expression levels. Challenging miR-modulated Jurkat cells or primary CD4+ T-cells with wild-type (WT), replication-competent HIV-1 demonstrated that miR-128 reduces viral replication and delays spreading of infection. Manipulation of miR-128 levels in HIV-1 target cell lines and in primary CD4+ T-cells by overexpression or knockdown showed that reduction of TNPO3 levels by miR-128 significantly affects HIV-1 replication but not murine leukemia virus (MLV) infection and that miR-128 modulation of HIV-1 replication is reduced with TNPO3-independent HIV-1 virus, suggesting that miR-128-indued TNPO3 repression contributes to the inhibition of HIV-1 replication. Finally, we determine that anti-miR-128 partly neutralizes the IFN-mediated block of HIV-1. Thus, we have established a novel role of miR-128 in antiviral defense in human cells, namely inhibiting HIV-1 replication by altering the cellular milieu through targeting factors that include TNPO3.IMPORTANCE HIV-1 is the causative agent of AIDS. During HIV-1 infection, type I interferons (IFNs) are induced, and their effectors limit HIV-1 replication at multiple steps in its life cycle. However, the cellular targets of INFs are still largely unknown. In this study, we identified the interferon-inducible microRNA (miR) miR-128, a novel antiviral mediator that suppresses the expression of the host gene TNPO3, which is known to modulate HIV-1 replication. Notably, we observe that anti-miR-128 partly neutralizes the IFN-mediated block of HIV-1. Elucidation of the mechanisms through which miR-128 impairs HIV-1 replication may provide novel candidates for the development of therapeutic interventions.


Subject(s)
Gene Expression Regulation/drug effects , HIV Infections/genetics , HIV Infections/virology , HIV-1/physiology , Interferons/pharmacology , MicroRNAs/genetics , Virus Replication , beta Karyopherins/genetics , 3' Untranslated Regions , Cell Line , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Models, Biological , RNA Interference
4.
PLoS Pathog ; 13(9): e1006572, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28945790

ABSTRACT

Species-dependent variation in proteins that aid or limit virus replication determines the ability of lentiviruses to jump between host species. Identifying and overcoming these differences facilitates the development of animal models for HIV-1, including models based on chimeric SIVs that express HIV-1 envelope (Env) glycoproteins, (SHIVs) and simian-tropic HIV-1 (stHIV) strains. Here, we demonstrate that the inherently poor ability of most HIV-1 Env proteins to use macaque CD4 as a receptor is improved during adaptation by virus passage in macaques. We identify a single amino acid, A281, in HIV-1 Env that consistently changes during adaptation in macaques and affects the ability of HIV-1 Env to use macaque CD4. Importantly, mutations at A281 do not markedly affect HIV-1 Env neutralization properties. Our findings should facilitate the design of HIV-1 Env proteins for use in non-human primate models and thus expedite the development of clinically relevant reagents for testing interventions against HIV-1.


Subject(s)
HIV Envelope Protein gp120/chemistry , HIV Infections/virology , HIV-1/physiology , Viral Tropism/physiology , Adaptation, Physiological/physiology , Animals , CD4 Antigens/metabolism , Disease Models, Animal , Female , Flow Cytometry , HIV Envelope Protein gp120/genetics , HIV Infections/genetics , Humans , Immunoblotting , Macaca mulatta , Male , Polymerase Chain Reaction , Simian Immunodeficiency Virus
5.
Proc Natl Acad Sci U S A ; 111(7): 2728-33, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24449914

ABSTRACT

Transportin 3 (Tnpo3, Transportin-SR2) is implicated in nuclear import of splicing factors and HIV-1 replication. Herein, we show that the majority of cellular Tnpo3 binding partners contain arginine-serine (RS) repeat domains and present crystal structures of human Tnpo3 in its free as well as GTPase Ran- and alternative splicing factor/splicing factor 2 (ASF/SF2)-bound forms. The flexible ß-karyopherin fold of Tnpo3 embraces the RNA recognition motif and RS domains of the cargo. A constellation of charged residues on and around the arginine-rich helix of Tnpo3 HEAT repeat 15 engage the phosphorylated RS domain and are critical for the recognition and nuclear import of ASF/SF2. Mutations in the same region of Tnpo3 impair its interaction with the cleavage and polyadenylation specificity factor 6 (CPSF6) and its ability to support HIV-1 replication. Steric incompatibility of the RS domain and RanGTP engagement by Tnpo3 provides the mechanism for cargo release in the nucleus. Our results elucidate the structural bases for nuclear import of splicing factors and the Tnpo3-CPSF6 nexus in HIV-1 biology.


Subject(s)
Cell Nucleus/metabolism , HIV-1/physiology , Models, Molecular , Nuclear Proteins/metabolism , Protein Conformation , beta Karyopherins/chemistry , beta Karyopherins/metabolism , Active Transport, Cell Nucleus/physiology , Blotting, Western , Chromatography, Gel , Chromatography, Ion Exchange , Crystallography, X-Ray , HEK293 Cells , HIV-1/metabolism , Humans , Immunoprecipitation , Oligonucleotides/genetics , Protein Binding , Virus Replication/physiology , X-Ray Diffraction , mRNA Cleavage and Polyadenylation Factors/metabolism
6.
Nat Rev Immunol ; 6(11): 859-68, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17063186

ABSTRACT

Dendritic cells (DCs) are crucial for the generation and the regulation of adaptive immunity. Because DCs have a pivotal role in marshalling immune responses, HIV has evolved ways to exploit DCs, thereby facilitating viral dissemination and allowing evasion of antiviral immunity. Defining the mechanisms that underlie cell-cell transmission of HIV and understanding the role of DCs in this process should help us in the fight against HIV infection. This Review highlights the latest advances in our understanding of the interactions between DCs and HIV, focusing on the mechanisms of DC-mediated viral dissemination.


Subject(s)
Dendritic Cells/immunology , HIV Infections/immunology , HIV Infections/transmission , HIV/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/immunology , HIV Infections/virology , Humans , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , Virus Internalization
7.
Nucleic Acids Res ; 42(3): 2085-97, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24194608

ABSTRACT

Control over the simultaneous delivery of different functionalities and their synchronized intracellular activation can greatly benefit the fields of RNA and DNA biomedical nanotechnologies and allow for the production of nanoparticles and various switching devices with controllable functions. We present a system of multiple split functionalities embedded in the cognate pairs of RNA-DNA hybrids which are programmed to recognize each other, re-associate and form a DNA duplex while also releasing the split RNA fragments which upon association regain their original functions. Simultaneous activation of three different functionalities (RNAi, Förster resonance energy transfer and RNA aptamer) confirmed by multiple in vitro and cell culture experiments prove the concept. To automate the design process, a novel computational tool that differentiates between the thermodynamic stabilities of RNA-RNA, RNA-DNA and DNA-DNA duplexes was developed. Moreover, here we demonstrate that besides being easily produced by annealing synthetic RNAs and DNAs, the individual hybrids carrying longer RNAs can be produced by RNA polymerase II-dependent transcription of single-stranded DNA templates.


Subject(s)
DNA/chemistry , RNA/chemistry , Aptamers, Nucleotide/chemistry , Cell Line, Tumor , Fluorescence Resonance Energy Transfer , Humans , Models, Molecular , RNA Interference , RNA Polymerase II/metabolism , Thermodynamics , Transcription, Genetic
8.
J Virol ; 87(1): 648-58, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23097450

ABSTRACT

Retroviruses integrate into cellular DNA nonrandomly. Lentiviruses such as human immunodeficiency virus type 1 (HIV-1) favor the bodies of active genes and gene-enriched transcriptionally active regions of chromosomes. The interaction between lentiviral integrase and the cellular protein lens epithelium-derived growth factor (LEDGF)/p75 underlies the targeting of gene bodies, whereas recent research has highlighted roles for the HIV-1 capsid (CA) protein and cellular factors implicated in viral nuclear import, including transportin 3 (TNPO3) and nucleoporin 358 (NUP358), in the targeting of gene-dense regions of chromosomes. Here, we show that CA mutations, which include the substitution of Asp for Asn74 (N74D), significantly reduce the dependency of HIV-1 on LEDGF/p75 during infection and that this difference correlates with the efficiency of viral DNA integration. The distribution of integration sites mapped by Illumina sequencing confirms that the N74D mutation reduces integration into gene-rich regions of chromosomes and gene bodies and reveals previously unrecognized roles for NUP153 (another HIV-1 cofactor implicated in viral nuclear import) and LEDGF/p75 in the targeting of the viral preintegration complex to gene-dense regions of chromatin. A role for the CA protein in determining the dependency of HIV-1 on LEDGF/p75 during infection highlights a connection between the viral capsid and chromosomal DNA integration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Capsid Proteins/metabolism , DNA, Viral/metabolism , HIV-1/pathogenicity , Host-Pathogen Interactions , Nuclear Pore Complex Proteins/metabolism , Transcription Factors/metabolism , Virus Integration , Animals , Capsid Proteins/genetics , Cell Line , Humans , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense
9.
PLoS Pathog ; 8(8): e1002896, 2012.
Article in English | MEDLINE | ID: mdl-22956906

ABSTRACT

The HIV-1 genome enters cells inside a shell comprised of capsid (CA) protein. Variation in CA sequence alters HIV-1 infectivity and escape from host restriction factors. However, apart from the Cyclophilin A-binding loop, CA has no known interfaces with which to interact with cellular cofactors. Here we describe a novel protein-protein interface in the N-terminal domain of HIV-1 CA, determined by X-ray crystallography, which mediates both viral restriction and host cofactor dependence. The interface is highly conserved across lentiviruses and is accessible in the context of a hexameric lattice. Mutation of the interface prevents binding to and restriction by CPSF6-358, a truncated cytosolic form of the RNA processing factor, cleavage and polyadenylation specific factor 6 (CPSF6). Furthermore, mutations that prevent CPSF6 binding also relieve dependence on nuclear entry cofactors TNPO3 and RanBP2. These results suggest that the HIV-1 capsid mediates direct host cofactor interactions to facilitate viral infection.


Subject(s)
Capsid Proteins/metabolism , HIV Infections/virology , HIV-1/physiology , Virus Replication , mRNA Cleavage and Polyadenylation Factors/metabolism , Amino Acid Sequence , Antiviral Agents/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cell Line, Tumor , Conserved Sequence , Crystallography, X-Ray , HIV-1/genetics , Humans , Indoles/metabolism , Models, Molecular , Molecular Chaperones/metabolism , Molecular Sequence Data , Mutation , Nuclear Pore Complex Proteins/metabolism , Phenylalanine/analogs & derivatives , Phenylalanine/metabolism , Protein Binding , Sequence Alignment , Virus Internalization , beta Karyopherins/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics
10.
J Virol ; 86(7): 3851-60, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22301135

ABSTRACT

The antiviral factor CPSF6-358 restricts human immunodeficiency virus type 1 (HIV-1) infection through an interaction with capsid (CA), preventing virus nuclear entry and integration. HIV-1 acquires resistance to CPSF6-358 through an N74D mutation of CA that impairs binding of the antiviral factor. Here we examined the determinants within CPSF6-358 that are necessary for CA-specific interaction. Residues 314 to 322 include amino acids that are essential for CPSF6-358 restriction of HIV-1. Fusion of CPSF6 residues 301 to 358 to rhesus TRIM5α is also sufficient to restrict wild-type but not N74D HIV-1. Restriction is lost if CPSF6 residues in the amino acid 314 to 322 interaction motif are mutated. Examination of the CA targeting motif in CPSF6-358 did not reveal evidence of positive selection. Given the sensitivity of different primate lentiviruses to CPSF6-358 and apparent conservation of this interaction, our data suggest that CPSF6-358-mediated targeting of HIV-1 could provide a broadly effective antiviral strategy.


Subject(s)
Capsid/metabolism , HIV Infections/metabolism , HIV-1/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , Humans , Molecular Sequence Data , Primates , Protein Binding , Protein Structure, Tertiary
11.
J Virol ; 86(8): 4708-14, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22301145

ABSTRACT

The antiviral factor CPSF6-358 interferes with the nuclear entry of human immunodeficiency virus type 1 (HIV-1). HIV-1 acquires resistance to CPSF6-358 through the N74D mutation of the capsid (CA), which alters its nuclear entry pathway. Here we show that compared to wild-type (WT) HIV-1, N74D HIV-1 is more sensitive to cyclosporine, has increased sensitivity to nevirapine, and is impaired in macrophage infection prior to reverse transcription. These phenotypes suggest a difference in the N74D reverse transcription complex that manifests early after infection and prior to interaction with the nuclear pore. Overall, our data indicate that N74D HIV-1 replication in transformed cells requires cyclophilin A but is dependent on other interactions in macrophages.


Subject(s)
Capsid Proteins/genetics , Cyclophilin A/pharmacology , HIV-1/genetics , HIV-1/metabolism , Macrophages/drug effects , Macrophages/virology , Mutation , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Animals , Antiviral Restriction Factors , Aotidae , Capsid Proteins/metabolism , Carrier Proteins/pharmacology , Cell Division , Cell Line , HIV-1/drug effects , Humans , Microbial Sensitivity Tests , RNA-Directed DNA Polymerase/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
12.
J Virol ; 86(23): 12525-30, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22933296

ABSTRACT

It has been proposed that most drug-resistant mutants, resulting from a single-nucleotide change, exist at low frequency in human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus (SIV) populations in vivo prior to the initiation of antiretroviral therapy (ART). To test this hypothesis and to investigate the emergence of resistant mutants with drug selection, we developed a new ultrasensitive allele-specific PCR (UsASP) assay, which can detect drug resistance mutations at a frequency of ≥0.001% of the virus population. We applied this assay to plasma samples obtained from macaques infected with an SIV variant containing HIV-1 reverse transcriptase (RT) (RT-simian-human immunodeficiency [SHIV](mne)), before and after they were exposed to a short course of efavirenz (EFV) monotherapy. We detected RT inhibitor (RTI) resistance mutations K65R and M184I but not K103N in 2 of 2 RT-SHIV-infected macaques prior to EFV exposure. After three doses over 4 days of EFV monotherapy, 103N mutations (AAC and AAT) rapidly emerged and increased in the population to levels of ∼20%, indicating that they were present prior to EFV exposure. The rapid increase of 103N mutations from <0.001% to 20% of the viral population indicates that the replicating virus population size in RT-SHIV-infected macaques must be 10(6) or more infected cells per replication cycle.


Subject(s)
Benzoxazines/pharmacology , Drug Resistance, Viral/genetics , HIV Reverse Transcriptase/genetics , Macaca nemestrina , Polymerase Chain Reaction/methods , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/enzymology , Alkynes , Alleles , Animals , Benzoxazines/therapeutic use , Cyclopropanes , Mutation/drug effects , Mutation/genetics , Sequence Analysis, DNA , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/genetics
13.
J Virol ; 86(6): 3152-66, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22238316

ABSTRACT

Although xenotropic murine leukemia virus-related virus (XMRV) has been previously linked to prostate cancer and myalgic encephalomyelitis/chronic fatigue syndrome, recent data indicate that results interpreted as evidence of human XMRV infection reflect laboratory contamination rather than authentic in vivo infection. Nevertheless, XMRV is a retrovirus of undefined pathogenic potential that is able to replicate in human cells. Here we describe a comprehensive analysis of two male pigtailed macaques (Macaca nemestrina) experimentally infected with XMRV. Following intravenous inoculation with >10(10) RNA copy equivalents of XMRV, viral replication was limited and transient, peaking at ≤2,200 viral RNA (vRNA) copies/ml plasma and becoming undetectable by 4 weeks postinfection, though viral DNA (vDNA) in peripheral blood mononuclear cells remained detectable through 119 days of follow-up. Similarly, vRNA was not detectable in lymph nodes by in situ hybridization despite detectable vDNA. Sequencing of cell-associated vDNA revealed extensive G-to-A hypermutation, suggestive of APOBEC-mediated viral restriction. Consistent with limited viral replication, we found transient upregulation of type I interferon responses that returned to baseline by 2 weeks postinfection, no detectable cellular immune responses, and limited or no spread to prostate tissue. Antibody responses, including neutralizing antibodies, however, were detectable by 2 weeks postinfection and maintained throughout the study. Both animals were healthy for the duration of follow-up. These findings indicate that XMRV replication and spread were limited in pigtailed macaques, predominantly by APOBEC-mediated hypermutation. Given that human APOBEC proteins restrict XMRV infection in vitro, human XMRV infection, if it occurred, would be expected to be characterized by similarly limited viral replication and spread.


Subject(s)
Disease Models, Animal , Macaca nemestrina , Retroviridae Infections/virology , Virus Replication , Xenotropic murine leukemia virus-related virus/physiology , Animals , Antibodies, Viral/immunology , Humans , Male , Phylogeny , Retroviridae Infections/immunology , Xenotropic murine leukemia virus-related virus/classification , Xenotropic murine leukemia virus-related virus/genetics
14.
PLoS Pathog ; 7(12): e1002439, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22174692

ABSTRACT

Lentiviruses such as HIV-1 traverse nuclear pore complexes (NPC) and infect terminally differentiated non-dividing cells, but how they do this is unclear. The cytoplasmic NPC protein Nup358/RanBP2 was identified as an HIV-1 co-factor in previous studies. Here we report that HIV-1 capsid (CA) binds directly to the cyclophilin domain of Nup358/RanBP2. Fusion of the Nup358/RanBP2 cyclophilin (Cyp) domain to the tripartite motif of TRIM5 created a novel inhibitor of HIV-1 replication, consistent with an interaction in vivo. In contrast to CypA binding to HIV-1 CA, Nup358 binding is insensitive to inhibition with cyclosporine, allowing contributions from CypA and Nup358 to be distinguished. Inhibition of CypA reduced dependence on Nup358 and the nuclear basket protein Nup153, suggesting that CypA regulates the choice of the nuclear import machinery that is engaged by the virus. HIV-1 cyclophilin-binding mutants CA G89V and P90A favored integration in genomic regions with a higher density of transcription units and associated features than wild type virus. Integration preference of wild type virus in the presence of cyclosporine was similarly altered to regions of higher transcription density. In contrast, HIV-1 CA alterations in another patch on the capsid surface that render the virus less sensitive to Nup358 or TRN-SR2 depletion (CA N74D, N57A) resulted in integration in genomic regions sparse in transcription units. Both groups of CA mutants are impaired in replication in HeLa cells and human monocyte derived macrophages. Our findings link HIV-1 engagement of cyclophilins with both integration targeting and replication efficiency and provide insight into the conservation of viral cyclophilin recruitment.


Subject(s)
Capsid Proteins/metabolism , Cell Nucleus/virology , Cyclophilin A/metabolism , HIV Infections/metabolism , HIV-1/physiology , Virus Replication , Active Transport, Cell Nucleus/physiology , Blotting, Western , Cell Line , Cell Nucleus/metabolism , HeLa Cells , Humans , Macrophages/metabolism , Macrophages/virology , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Virus Replication/physiology
15.
Sci Rep ; 13(1): 10974, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37414787

ABSTRACT

The early events of HIV-1 infection involve the transport of the viral core into the nucleus. This event triggers the translocation of CPSF6 from paraspeckles into nuclear speckles forming puncta-like structures. Our investigations revealed that neither HIV-1 integration nor reverse transcription is required for the formation of puncta-like structures. Moreover, HIV-1 viruses without viral genome are competent for the induction of CPSF6 puncta-like structures. In agreement with the notion that HIV-1 induced CPSF6 puncta-like structures are biomolecular condensates, we showed that osmotic stress and 1,6-hexanediol induced the disassembly of CPSF6 condensates. Interestingly, replacing the osmotic stress by isotonic media re-assemble CPSF6 condensates in the cytoplasm of the cell. To test whether CPSF6 condensates were important for infection we utilized hypertonic stress, which prevents formation of CPSF6 condensates, during infection. Remarkably, preventing the formation of CPSF6 condensates inhibits the infection of wild type HIV-1 but not of HIV-1 viruses bearing the capsid changes N74D and A77V, which do not form CPSF6 condensates during infection1,2. We also investigated whether the functional partners of CPSF6 are recruited to the condensates upon infection. Our experiments revealed that CPSF5, but not CPSF7, co-localized with CPSF6 upon HIV-1 infection. We found condensates containing CPSF6/CPSF5 in human T cells and human primary macrophages upon HIV-1 infection. Additionally, we observed that the integration cofactor LEDGF/p75 changes distribution upon HIV-1 infection and surrounds the CPSF6/CPSF5 condensates. Overall, our work demonstrated that CPSF6 and CPSF5 are forming biomolecular condensates that are important for infection of wild type HIV-1 viruses.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Humans , Biomolecular Condensates , Capsid/metabolism , Capsid Proteins/metabolism , Cell Nucleus/metabolism , HIV Seropositivity/metabolism , HIV-1/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism , Virus Replication
16.
Nat Commun ; 14(1): 3782, 2023 06 24.
Article in English | MEDLINE | ID: mdl-37355754

ABSTRACT

The movement of viruses and other large macromolecular cargo through nuclear pore complexes (NPCs) is poorly understood. The human immunodeficiency virus type 1 (HIV-1) provides an attractive model to interrogate this process. HIV-1 capsid (CA), the chief structural component of the viral core, is a critical determinant in nuclear transport of the virus. HIV-1 interactions with NPCs are dependent on CA, which makes direct contact with nucleoporins (Nups). Here we identify Nup35, Nup153, and POM121 to coordinately support HIV-1 nuclear entry. For Nup35 and POM121, this dependence was dependent cyclophilin A (CypA) interaction with CA. Mutation of CA or removal of soluble host factors changed the interaction with the NPC. Nup35 and POM121 make direct interactions with HIV-1 CA via regions containing phenylalanine glycine motifs (FG-motifs). Collectively, these findings provide additional evidence that the HIV-1 CA core functions as a macromolecular nuclear transport receptor (NTR) that exploits soluble host factors to modulate NPC requirements during nuclear invasion.


Subject(s)
HIV-1 , Humans , Active Transport, Cell Nucleus/genetics , HIV-1/genetics , Capsid/metabolism , Cell Line , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Nuclear Pore/metabolism , Membrane Glycoproteins/metabolism
17.
J Virol ; 85(2): 1067-76, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21084490

ABSTRACT

The impact of antiretroviral therapy (ART) on the genetics of simian immunodeficiency virus (SIV) or human immunodeficiency virus (HIV) populations has been incompletely characterized. We analyzed SIV genetic variation before, during, and after ART in a macaque model. Six pigtail macaques were infected with an SIV/HIV chimeric virus, RT-SHIV(mne), in which SIV reverse transcriptase (RT) was replaced by HIV-1 RT. Three animals received a short course of efavirenz (EFV) monotherapy before combination ART was started. All macaques received 20 weeks of tenofovir, emtricitabine, and EFV. Plasma virus populations were analyzed by single-genome sequencing. Population diversity was measured by average pairwise difference, and changes in viral genetics were assessed by phylogenetic and panmixia analyses. After 20 weeks of ART, viral diversity was not different from pretherapy viral diversity despite more than 10,000-fold declines in viremia, indicating that, within this range, there is no relationship between diversity and plasma viremia. In two animals with consistent SIV RNA suppression to <15 copies/ml during ART, there was no evidence of viral evolution. In contrast, in the four macaques with viremias >15 copies/ml during therapy, there was divergence between pre- and during-ART virus populations. Drug resistance mutations emerged in two of these four animals, resulting in virologic failure in the animal with the highest level of pretherapy viremia. Taken together, these findings indicate that viral diversity does not decrease with suppressive ART, that ongoing replication occurs with viremias >15 copies/ml, and that in this macaque model of ART drug resistance likely emerges as a result of incomplete suppression and preexisting drug resistance mutations.


Subject(s)
Anti-Retroviral Agents/administration & dosage , Antiretroviral Therapy, Highly Active , Genetic Variation , HIV Reverse Transcriptase/genetics , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Animals , Cluster Analysis , Disease Models, Animal , Evolution, Molecular , Macaca , Phylogeny , Plasma/virology , RNA, Viral/genetics , Recombinant Proteins/genetics , Sequence Analysis, DNA , Simian Immunodeficiency Virus/classification , Simian Immunodeficiency Virus/isolation & purification , Viral Load
18.
Proc Natl Acad Sci U S A ; 106(11): 4425-9, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19255423

ABSTRACT

The lack of a primate model that utilizes HIV-1 as the challenge virus is an impediment to AIDS research; existing models generally employ simian viruses that are divergent from HIV-1, reducing their usefulness in preclinical investigations. Based on an understanding of species-specific variation in primate TRIM5 and APOBEC3 antiretroviral genes, we constructed simian-tropic (st)HIV-1 strains that differ from HIV-1 only in the vif gene. We demonstrate that such minimally modified stHIV-1 strains are capable of high levels of replication in vitro in pig-tailed macaque (Macaca nemestrina) lymphocytes. Importantly, infection of pig-tailed macaques with stHIV-1 results in acute viremia, approaching the levels observed in HIV-1-infected humans, and an ensuing persistent infection for several months. stHIV-1 replication was controlled thereafter, at least in part, by CD8+ T cells. We demonstrate the potential utility of this HIV-1-based animal model in a chemoprophylaxis experiment, by showing that a commonly used HIV-1 therapeutic regimen can provide apparently sterilizing protection from infection following a rigorous high-dose stHIV-1 challenge.


Subject(s)
Disease Models, Animal , HIV Infections/virology , HIV-1/genetics , Macaca , Animals , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Chemoprevention , Genetic Engineering , Viremia , Virus Replication
19.
iScience ; 25(1): 103593, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35005542

ABSTRACT

The HIV-1 capsid is the target for the antiviral drugs GS-CA1 and Lenacapavir (GS-6207). We investigated the mechanism by which GS-CA1 and GS-6207 inhibit HIV-1 infection. HIV-1 inhibition by GS-CA1 did not require CPSF6 in CD4+ T cells. Contrary to PF74 that accelerates uncoating of HIV-1, GS-CA1 and GS-6207 stabilized the core. GS-CA1, unlike PF74, allowed the core to enter the nucleus, which agrees with the fact that GS-CA1 inhibits infection after reverse transcription. Unlike PF74, GS-CA1 did not disaggregate preformed CPSF6 complexes in nuclear speckles, suggesting that PF74 and GS-CA1 have different mechanisms of action. GS-CA1 stabilized the HIV-1 core, possibly by inducing a conformational shift in the core; in agreement, HIV-1 cores bearing N74D regained their ability to bind CPSF6 in the presence of GS-CA1. We showed that GS-CA1 binds to the HIV-1 core, changes its conformation, stabilizes the core, and thereby prevents viral uncoating and infection.

20.
Nat Commun ; 13(1): 1474, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35304442

ABSTRACT

Retroviruses utilize the viral integrase (IN) protein to integrate a DNA copy of their genome into host chromosomal DNA. HIV-1 integration sites are highly biased towards actively transcribed genes, likely mediated by binding of the IN protein to specific host factors, particularly LEDGF, located at these gene regions. We here report a substantial redirection of integration site distribution induced by a single point mutation in HIV-1 IN. Viruses carrying the K258R IN mutation exhibit a high frequency of integrations into centromeric alpha satellite repeat sequences, as assessed by deep sequencing, a more than 10-fold increase over wild-type. Quantitative PCR and in situ immunofluorescence assays confirm this bias of the K258R mutant virus for integration into centromeric DNA. Immunoprecipitation studies identify host factors binding to IN that may account for the observed bias for integration into centromeres. Centromeric integration events are known to be enriched in the latent reservoir of infected memory T cells, as well as in elite controllers who limit viral replication without intervention. The K258R point mutation in HIV-1 IN is also present in databases of latent proviruses found in patients, and may reflect an unappreciated aspect of the establishment of viral latency.


Subject(s)
HIV Integrase , Point Mutation , Centromere/genetics , Centromere/metabolism , HIV Integrase/genetics , HIV Integrase/metabolism , Humans , Proviruses/genetics , Virus Integration/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL