Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
1.
FEBS J ; 288(2): 427-433, 2021 01.
Article in English | MEDLINE | ID: mdl-32506843

ABSTRACT

Integrase strand transfer inhibitors (INSTIs) are important components of drug formulations that are used to treat people living with HIV, and second-generation INSTIs dolutegravir and bictegravir impart high barriers to the development of drug resistance. Reported 10 years ago, X-ray crystal structures of prototype foamy virus (PFV) intasome complexes explained how INSTIs bind integrase to inhibit strand transfer activity and provided initial glimpses into mechanisms of drug resistance. However, comparatively low sequence identity between PFV and HIV-1 integrases limited the depth of information that could be gleaned from the surrogate model system. Recent high-resolution structures of HIV-1 intasomes as well as intasomes from a closely related strain of simian immunodeficiency virus (SIV), which were determined using single-particle cryogenic electron microscopy, have overcome this limitation. The new structures reveal the binding modes of several advanced INSTI compounds to the HIV/SIV integrase active site and critically inform the structural basis of drug resistance. These findings will help guide the continued development of this important class of antiretroviral therapeutics.


Subject(s)
HIV Integrase Inhibitors/chemistry , HIV Integrase/chemistry , HIV-1/drug effects , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , Oxazines/chemistry , Piperazines/chemistry , Pyridones/chemistry , Simian Immunodeficiency Virus/drug effects , Amides , Animals , Catalytic Domain , Cryoelectron Microscopy , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , HIV Infections/drug therapy , HIV Infections/virology , HIV Integrase/genetics , HIV Integrase/metabolism , HIV Integrase Inhibitors/pharmacology , HIV-1/chemistry , HIV-1/enzymology , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Oxazines/pharmacology , Piperazines/pharmacology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Pyridones/pharmacology , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/enzymology , Spumavirus/chemistry , Spumavirus/drug effects , Spumavirus/enzymology
2.
J Virol ; 94(23)2020 11 09.
Article in English | MEDLINE | ID: mdl-32907979

ABSTRACT

Pandemic human immunodeficiency virus type 1 (HIV-1) is the result of the zoonotic transmission of simian immunodeficiency virus (SIV) from the chimpanzee subspecies Pan troglodytestroglodytes (SIVcpzPtt). The related subspecies Pan troglodytesschweinfurthii is the host of a similar virus, SIVcpzPts, which did not spread to humans. We tested these viruses with small-molecule capsid inhibitors (PF57, PF74, and GS-CA1) that interact with a binding groove in the capsid that is also used by CPSF6. While HIV-1 was sensitive to capsid inhibitors in cell lines, human macrophages, and peripheral blood mononuclear cells (PBMCs), SIVcpzPtt was resistant in rhesus FRhL-2 cells and human PBMCs but was sensitive to PF74 in human HOS and HeLa cells. SIVcpzPts was insensitive to PF74 in FRhL-2 cells, HeLa cells, PBMCs, and macrophages but was inhibited by PF74 in HOS cells. A truncated version of CPSF6 (CPSF6-358) inhibited SIVcpzPtt and HIV-1, while in contrast, SIVcpzPts was resistant to CPSF6-358. Homology modeling of HIV-1, SIVcpzPtt, and SIVcpzPts capsids and binding energy estimates suggest that these three viruses bind similarly to the host proteins cyclophilin A (CYPA) and CPSF6 as well as the capsid inhibitor PF74. Cyclosporine treatment, mutation of the CYPA-binding loop in the capsid, or CYPA knockout eliminated the resistance of SIVcpzPts to PF74 in HeLa cells. These experiments revealed that the antiviral capacity of PF74 is controlled by CYPA in a virus- and cell type-specific manner. Our data indicate that SIVcpz viruses can use infection pathways that escape the antiviral activity of PF74. We further suggest that the antiviral activity of PF74 capsid inhibitors depends on cellular cofactors.IMPORTANCE HIV-1 originated from SIVcpzPtt but not from the related virus SIVcpzPts, and thus, it is important to describe molecular infection by SIVcpzPts in human cells to understand the zoonosis of SIVs. Pharmacological HIV-1 capsid inhibitors (e.g., PF74) bind a capsid groove that is also a binding site for the cellular protein CPSF6. SIVcpzPts was resistant to PF74 in HeLa cells but sensitive in HOS cells, thus indicating cell line-specific resistance. Both SIVcpz viruses showed resistance to PF74 in human PBMCs. Modulating the presence of cyclophilin A or its binding to capsid in HeLa cells overcame SIVcpzPts resistance to PF74. These results indicate that early cytoplasmic infection events of SIVcpzPts may differ between cell types and affect, in an unknown manner, the antiviral activity of capsid inhibitors. Thus, capsid inhibitors depend on the activity or interaction of currently uncharacterized cellular factors.


Subject(s)
Anti-HIV Agents/pharmacology , Capsid Proteins/chemistry , Capsid Proteins/drug effects , Capsid Proteins/metabolism , Capsid/drug effects , Simian Immunodeficiency Virus/drug effects , mRNA Cleavage and Polyadenylation Factors/chemistry , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Binding Sites , Capsid Proteins/genetics , Cell Line , Cyclophilin A/genetics , Cyclophilin A/metabolism , Gene Knockout Techniques , HEK293 Cells , HIV-1 , HeLa Cells , Humans , Indazoles/pharmacology , Indoles/pharmacology , Leukocytes, Mononuclear/virology , Macrophages/virology , Models, Molecular , Pan troglodytes/virology , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Protein Conformation , Protein Interaction Domains and Motifs , Pyridines/pharmacology , Sequence Alignment , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/genetics , Zoonoses , mRNA Cleavage and Polyadenylation Factors/genetics
3.
J Struct Biol ; 209(1): 107426, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31733279

ABSTRACT

We describe a semiautomated approach to segment Env spikes from the membrane envelope of Simian Immunodeficiency Virus visualized by cryoelectron tomography of frozen-hydrated specimens. Multivariate data analysis is applied to a large set of overlapping subvolumes extracted semiautomatically from the viral envelope and does not utilize a template of the target structure. The major manual step used in the method involves determination of six points that define an ellipsoid approximating the virion shape. The approach is robust to departures of the actual virion from this starting ellipsoid. A point cage of sufficient density is generated to ensure that any spike-like protein is identified multiple times. Subsequently translational alignment of class averages to a cylindrical reference on a curved surface separates subvolumes with spikes from those without. Spike containing subvolumes identified multiple times are removed by proximity analysis. Slightly different procedures segment spikes in the equatorial and the polar regions. Once all spikes are segmented, further alignment of class averages using separately the polar and spin angles produces recognizable spike images. Our approach localized 96% of the equatorial spikes and 85% of all spikes identified manually; it identifies a significant number of additional spikes missed by manual selection. Two types of spike shapes were segmented, one with near 3-fold symmetry resembling the conventional spike, the other had a T-shape resembling the spike structure obtained when antibodies such as PG9 bind to HIV Env. The approach should be applicable to segmentation of any protein spikes extending from a cellular or virion envelope.


Subject(s)
Cryoelectron Microscopy/methods , Image Processing, Computer-Assisted/methods , Viral Envelope/chemistry , env Gene Products, Human Immunodeficiency Virus/chemistry , Algorithms , Electron Microscope Tomography/methods , HIV-1/chemistry , Simian Immunodeficiency Virus/chemistry , Viral Envelope/classification , Viral Proteins/chemistry , Virion/chemistry
4.
Virology ; 519: 180-189, 2018 06.
Article in English | MEDLINE | ID: mdl-29729526

ABSTRACT

The twin-cysteine motif (TCM) in the V2 loop region of gp120, identified in our previous report on the simian immunodeficiency virus mac239 (SIVmac239), is a conserved evolutionary element in all primate lentiviruses except for HIV-1 which has lost the TCM during cross-species transmission. In this study, we have further explored the TCM in other SIV and HIV-2 strains. Our data shows that strains from different evolutionary lineages have different phenotypes when the twin-cysteines are removed. In the SIVsm/HIV-2 lineage, removal of the twin-cysteines decreases envelope trimer stability, but in the SIVagm lineage, a blockage of gp160 processing is observed. Molecular modeling has confirmed that the twin-cysteines do form a disulfide bond in the gp120 subunit, which interacts with the V1 loop to stabilize the envelope trimer. Therefore, we hypothesize that if the TCM is added back to HIV-1, it will enhance envelope stability for vaccine immunogen design.


Subject(s)
Amino Acid Motifs , Cysteine/chemistry , HIV Envelope Protein gp120/chemistry , HIV-1/chemistry , HIV-2/chemistry , Simian Immunodeficiency Virus/chemistry , Viral Envelope Proteins/chemistry , AIDS Vaccines , Amino Acid Sequence , Animals , Cell Line , Cysteine/genetics , Drug Design , HEK293 Cells , HIV Envelope Protein gp120/genetics , HIV-1/genetics , HIV-2/genetics , Humans , Models, Molecular , Protein Conformation , Protein Multimerization , Protein Stability , Simian Immunodeficiency Virus/genetics , Viral Envelope Proteins/genetics
5.
J Virol ; 91(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27795437

ABSTRACT

Human immunodeficiency virus type 2 (HIV-2) has already spread to different regions worldwide, and currently about 1 to 2 million people have been infected, calling for new antiviral agents that are effective on both HIV-1 and HIV-2 isolates. T20 (enfuvirtide), a 36-mer peptide derived from the C-terminal heptad repeat region (CHR) of gp41, is the only clinically approved HIV-1 fusion inhibitor, but it easily induces drug resistance and is not active on HIV-2. In this study, we first demonstrated that the M-T hook structure was also vital to enhancing the binding stability and inhibitory activity of diverse CHR-based peptide inhibitors. We then designed a novel short peptide (23-mer), termed 2P23, by introducing the M-T hook structure, HIV-2 sequences, and salt bridge-forming residues. Promisingly, 2P23 was a highly stable helical peptide with high binding to the surrogate targets derived from HIV-1, HIV-2, and simian immunodeficiency virus (SIV). Consistent with this, 2P23 exhibited potent activity in inhibiting diverse subtypes of HIV-1 isolates, T20-resistant HIV-1 mutants, and a panel of primary HIV-2 isolates, HIV-2 mutants, and SIV isolates. Therefore, we conclude that 2P23 has high potential to be further developed for clinical use, and it is also an ideal tool for exploring the mechanisms of HIV-1/2- and SIV-mediated membrane fusion. IMPORTANCE: The peptide drug T20 is the only approved HIV-1 fusion inhibitor, but it is not active on HIV-2 isolates, which have currently infected 1 to 2 million people and continue to spread worldwide. Recent studies have demonstrated that the M-T hook structure can greatly enhance the binding and antiviral activities of gp41 CHR-derived inhibitors, especially for short peptides that are otherwise inactive. By combining the hook structure, HIV-2 sequence, and salt bridge-based strategies, the short peptide 2P23 has been successfully designed. 2P23 exhibits prominent advantages over many other peptide fusion inhibitors, including its potent and broad activity on HIV-1, HIV-2, and even SIV isolates, its stability as a helical, oligomeric peptide, and its high binding to diverse targets. The small size of 2P23 would benefit its synthesis and significantly reduce production cost. Therefore, 2P23 is an ideal candidate for further development, and it also provides a novel tool for studying HIV-1/2- and SIV-mediated cell fusion.


Subject(s)
HIV Envelope Protein gp41/antagonists & inhibitors , HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , HIV-2/drug effects , Peptides/pharmacology , Simian Immunodeficiency Virus/drug effects , Binding Sites , Drug Design , Drug Resistance, Viral/drug effects , Enfuvirtide , HIV Envelope Protein gp41/metabolism , HIV Envelope Protein gp41/pharmacology , HIV Fusion Inhibitors/chemical synthesis , HIV-1/chemistry , HIV-1/metabolism , HIV-2/chemistry , HIV-2/metabolism , Humans , Peptide Fragments/pharmacology , Peptides/chemical synthesis , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/metabolism , Structure-Activity Relationship , Virus Internalization/drug effects
6.
Retrovirology ; 13(1): 89, 2016 Dec 29.
Article in English | MEDLINE | ID: mdl-28034301

ABSTRACT

BACKGROUND: The nucleocapsid (NC) domain of HIV-1 Gag is responsible for specific recognition and packaging of genomic RNA (gRNA) into new viral particles. This occurs through specific interactions between the Gag NC domain and the Psi packaging signal in gRNA. In addition to this critical function, NC proteins are also nucleic acid (NA) chaperone proteins that facilitate NA rearrangements during reverse transcription. Although the interaction with Psi and chaperone activity of HIV-1 NC have been well characterized in vitro, little is known about simian immunodeficiency virus (SIV) NC. Non-human primates are frequently used as a platform to study retroviral infection in vivo; thus, it is important to understand underlying mechanistic differences between HIV-1 and SIV NC. RESULTS: Here, we characterize SIV NC chaperone activity for the first time. Only modest differences are observed in the ability of SIV NC to facilitate reactions that mimic the minus-strand annealing and transfer steps of reverse transcription relative to HIV-1 NC, with the latter displaying slightly higher strand transfer and annealing rates. Quantitative single molecule DNA stretching studies and dynamic light scattering experiments reveal that these differences are due to significantly increased DNA compaction energy and higher aggregation capability of HIV-1 NC relative to the SIV protein. Using salt-titration binding assays, we find that both proteins are strikingly similar in their ability to specifically interact with HIV-1 Psi RNA. In contrast, they do not demonstrate specific binding to an RNA derived from the putative SIV packaging signal. CONCLUSIONS: Based on these studies, we conclude that (1) HIV-1 NC is a slightly more efficient NA chaperone protein than SIV NC, (2) mechanistic differences between the NA interactions of highly similar retroviral NC proteins are revealed by quantitative single molecule DNA stretching, and (3) SIV NC demonstrates cross-species recognition of the HIV-1 Psi RNA packaging signal.


Subject(s)
Genome, Viral , HIV-1/chemistry , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/metabolism , RNA, Viral/genetics , Simian Immunodeficiency Virus/chemistry , HIV-1/genetics , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/physiology , Nucleic Acid Conformation , Nucleocapsid Proteins/genetics , Protein Binding , Reverse Transcription , Simian Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/genetics
7.
J Immunol ; 197(10): 3999-4013, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27733554

ABSTRACT

HIV sequence diversity and the propensity of eliciting immunodominant responses targeting variable regions of the HIV proteome are hurdles in the development of an effective AIDS vaccine. An HIV-derived conserved element (CE) p24gag plasmid DNA (pDNA) vaccine is able to redirect immunodominant responses to otherwise subdominant and often more vulnerable viral targets. By homology to the HIV immunogen, seven CE were identified in SIV p27Gag Analysis of 31 rhesus macaques vaccinated with full-length SIV gag pDNA showed inefficient induction (58% response rate) of cellular responses targeting these CE. In contrast, all 14 macaques immunized with SIV p27CE pDNA developed robust T cell responses recognizing CE. Vaccination with p27CE pDNA was also critical for the efficient induction and increased the frequency of Ag-specific T cells with cytotoxic potential (granzyme B+ CD107a+) targeting subdominant CE epitopes, compared with the responses elicited by the p57gag pDNA vaccine. Following p27CE pDNA priming, two booster regimens, gag pDNA or codelivery of p27CE+gag pDNA, significantly increased the levels of CE-specific T cells. However, the CE+gag pDNA booster vaccination elicited significantly broader CE epitope recognition, and thus, a more profound alteration of the immunodominance hierarchy. Vaccination with HIV molecules showed that CE+gag pDNA booster regimen further expanded the breadth of HIV CE responses. Hence, SIV/HIV vaccine regimens comprising CE pDNA prime and CE+gag pDNA booster vaccination significantly increased cytotoxic T cell responses to subdominant highly conserved Gag epitopes and maximized response breadth.


Subject(s)
Cytotoxicity, Immunologic , Epitopes/immunology , Gene Products, gag/immunology , HIV Infections/prevention & control , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Vaccines, DNA/immunology , AIDS Vaccines/immunology , Animals , Cytokines/immunology , HIV/immunology , HIV/physiology , HIV Infections/immunology , HIV Infections/virology , Immunization Schedule , Immunization, Secondary/methods , Macaca mulatta , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , Vaccines, DNA/administration & dosage
8.
J Virol ; 90(24): 10993-11006, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27630234

ABSTRACT

Strong antibody (Ab) responses against V1V2 epitopes of the human immunodeficiency virus type 1 (HIV-1) gp120 envelope (Env) correlated with reduced infection rates in studies of HIV, simian-human immunodeficiency virus (SHIV), and simian immunodeficiency virus (SIV). In order to focus the Ab response on V1V2, we used six V1V2 sequences and nine scaffold proteins to construct immunogens which were tested using various immunization regimens for their ability to induce cross-reactive and biologically active V2 Abs in rabbits. A prime/boost immunization strategy was employed using gp120 DNA and various V1V2-scaffold proteins. The rabbit polyclonal Ab responses (i) were successfully focused on the V1V2 region, with weak or only transient responses to other Env epitopes, (ii) displayed broad cross-reactive binding activity with gp120s and the V1V2 regions of diverse strains from clades B, C, and E, (iii) included V2 Abs with specificities similar to those found in HIV-infected individuals, and (iv) remained detectable ≥1 year after the last boosting dose. Importantly, sera from rabbits receiving V1V2-scaffold immunogens displayed Ab-dependent cellular phagocytosis whereas sera from rabbits receiving only gp120 did not. The results represent the first fully successful example of reverse vaccinology in the HIV vaccine field with rationally designed epitope scaffold immunogens inducing Abs that recapitulate the epitope specificity and biologic activity of the human monoclonal Abs from which the immunogens were designed. Moreover, this is the first immunogenicity study using epitope-targeting, rationally designed vaccine constructs that induced an Fc-mediated activity associated with protection from infection with HIV, SIV, and SHIV. IMPORTANCE: Novel immunogens were designed to focus the antibody response of rabbits on the V1V2 epitopes of HIV-1 gp120 since such antibodies were associated with reduced infection rates of HIV, SIV, and SHIV. The vaccine-induced antibodies were broadly cross-reactive with the V1V2 regions of HIV subtypes B, C and E and, importantly, facilitated Fc-mediated phagocytosis, an activity not induced upon immunization of rabbits with gp120. This is the first immunogenicity study of vaccine constructs that focuses the antibody response on V1V2 and induces V2-specific antibodies with the ability to mediate phagocytosis, an activity that has been associated with protection from infection with HIV, SIV, and SHIV.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/biosynthesis , HIV Envelope Protein gp120/immunology , HIV Infections/prevention & control , Immunization, Secondary , Immunogenicity, Vaccine , Simian Acquired Immunodeficiency Syndrome/prevention & control , AIDS Vaccines/administration & dosage , AIDS Vaccines/biosynthesis , AIDS Vaccines/genetics , Amino Acid Sequence , Animals , Cross Reactions , Drug Design , Epitopes/chemistry , Epitopes/immunology , Female , Gene Expression , HIV Envelope Protein gp120/biosynthesis , HIV Envelope Protein gp120/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/chemistry , HIV-1/genetics , HIV-1/immunology , Humans , Models, Molecular , Peptide Mapping , Phagocytosis/drug effects , Protein Structure, Secondary , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology
9.
J Virol ; 89(13): 6887-94, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25903330

ABSTRACT

UNLABELLED: Recent evidence suggests that even in treated infections, human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) replication may continue in lymph nodes (LN), serving as a potential virus reservoir. Here we investigated the effects of lentivirus infection on natural killer (NK) cell frequencies, phenotypes, and functions in naive and acutely or chronically SIVmac239-infected rhesus macaques. Compared to that in naive animals, we observed a 3-fold-greater frequency of cytotoxic CD16(+) CD56(-) NK cells in LN of chronically infected macaques. However, NK cells did not appear to be trafficking to LN, as homing markers CD62L and CCR7 did not increase on circulating NK cells during infection. LN NK cells demonstrated enhanced cytotoxicity in acute infection, with 2-fold increases in perforin expression and 3-fold increases in CD107a expression following mitogen stimulation. Lysis of K562 cells by LN NK cells from acutely infected animals was greater than lysis by preinfection samples from the same animals. LN NK cells from chronically infected animals lysed K562 cells more efficiently than LN NK cells from uninfected animals, but importantly, surrogate markers of cytotoxicity in infected macaques were disproportionately greater than ex vivo killing. Furthermore, Tim-3, an indicator of activation and/or exhaustion, was upregulated 3-fold on LN NK cells in chronically infected animals. Collectively, these data suggest that LN NK cells are skewed toward a cytotoxic phenotype during SIV infection but may become dysfunctional and exhausted in chronic disease. IMPORTANCE: The accumulation of CD16(+) CD56(-) NK cells in the SIV-infected lymph node without changes in NK homing to the LN could suggest that these cells are differentiating in situ. Surprisingly, this increase in frequency of the cytotoxic subset of NK cells is not accompanied by an increase of similar magnitude in the cytolytic function of LN lymphocytes. This functional modulation, together with the higher Tim-3 expression observed on LN NK cells isolated from chronically infected animals than on those from naive macaques, is indicative of an exhausted phenotype. This exhaustion could contribute to the robust replication of HIV and SIV in the LN during acute and chronic stages of infection, allowing the survival of infected cells and maintenance of a viral reservoir.


Subject(s)
Killer Cells, Natural/immunology , Lymph Nodes/immunology , Lymph Nodes/virology , Receptors, IgG/analysis , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Animals , CD56 Antigen/analysis , Lymphocyte Subsets/immunology , Macaca mulatta , Simian Immunodeficiency Virus/chemistry
10.
Virology ; 476: 19-25, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25499532

ABSTRACT

Vpr and Vpx are primate lentivirus proteins that manipulate the cellular CRL4 ubiquitin ligase complex. While Vpr is common to all primate lentiviruses, Vpx is only encoded by HIV-2 and a limited range of SIVs. Although Vpr and Vpx share a high degree of homology they are known to induce markedly different effects in host cell biology through the recruitment of different substrates to CRL4. Here we explore the interaction of HIV-1 Vpr and SIVmac Vpx with the CRL4 substrate receptor DCAF1. Through mutational analysis of DCAF1 we demonstrate that although Vpr and Vpx share a highly similar DCAF1-binding motif, they interact with a different set of residues in DCAF1. In addition, we show that Vpx recruits SAMHD1 through a protein-protein interface that includes interactions of SAMHD1 with both Vpx and DCAF1, as was first suggested in crystallography data by (Schwefel, D., Groom, H.C.T., Boucherit, V.C., Christodoulou, E., Walker, P.A., Stoye, J.P., Bishop, K.N., Taylor, I.A., 2014. Structural basis of lentiviral subversion of a cellular protein degradation pathway., Nature, 505, 234-238).


Subject(s)
Carrier Proteins/metabolism , HIV-1/metabolism , Simian Immunodeficiency Virus/metabolism , Viral Regulatory and Accessory Proteins/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Motifs , Amino Acid Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , HIV-1/chemistry , HIV-1/genetics , Humans , Molecular Sequence Data , Protein Binding , Protein Serine-Threonine Kinases , Protein Structure, Tertiary , Sequence Alignment , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/genetics , Ubiquitin-Protein Ligases , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/genetics , vpr Gene Products, Human Immunodeficiency Virus/chemistry , vpr Gene Products, Human Immunodeficiency Virus/genetics
11.
J Virol ; 88(18): 10289-302, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24991000

ABSTRACT

UNLABELLED: Following retrovirus entry, the viral capsid (CA) disassembles into its component capsid proteins. The rate of this uncoating process, which is regulated by CA-CA interactions and by the association of the capsid with host cell factors like cyclophilin A (CypA), can influence the efficiency of reverse transcription. Inspection of the CA sequences of lentiviruses reveals that several species of simian immunodeficiency viruses (SIVs) have lost the glycine-proline motif in the helix 4-5 loop important for CypA binding; instead, the helix 4-5 loop in these SIVs exhibits an increase in the number of glutamine residues. In this study, we investigated the role of these glutamine residues in SIVmac239 replication. Changes in these residues, particularly glutamine 89 and glutamine 92, resulted in a decreased efficiency of core condensation, decreased stability of the capsids in infected cells, and blocks to reverse transcription. In some cases, coexpression of two different CA mutants produced chimeric virions that exhibited higher infectivity than either parental mutant virus. For this complementation of infectivity, glutamine 89 was apparently required on one of the complementing pair of mutants and glutamine 92 on the other. Modeling suggests that glutamines 89 and 92 are located on the distal face of hexameric capsid spokes and thus are well positioned to contribute to interhexamer interactions. Requirements to evade host restriction factors like TRIMCyp may drive some SIV lineages to evolve means other than CypA binding to stabilize the capsid. One solution used by several SIV strains consists of glutamine-based bonding. IMPORTANCE: The retroviral capsid is an assembly of individual capsid proteins that surrounds the viral RNA. After a retrovirus enters a cell, the capsid must disassemble, or uncoat, at a proper rate. The interactions among capsid proteins contribute to this rate of uncoating. We found that some simian immunodeficiency viruses use arrays of glutamine residues, which can form hydrogen bonds efficiently, to keep their capsids stable. This strategy may allow these viruses to forego the use of capsid-stabilizing factors from the host cell, some of which have antiviral activity.


Subject(s)
Capsid Proteins/chemistry , Capsid Proteins/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Capsid , Capsid Proteins/genetics , Cyclophilin A/metabolism , Glutamine/chemistry , Glutamine/metabolism , HIV Infections/virology , HIV-1/chemistry , HIV-1/genetics , HIV-1/metabolism , Humans , Macaca , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Sequence Alignment , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/genetics
12.
Virology ; 452-453: 202-11, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24606697

ABSTRACT

We have previously designed a method to construct viable recombinant Yellow Fever (YF) 17D viruses expressing heterologous polypeptides including part of the Simian Immunodeficiency Virus (SIV) Gag protein. However, the expressed region, encompassing amino acid residues from 45 to 269, was genetically unstable. In this study, we improved the genetic stability of this recombinant YF 17D virus by introducing mutations in the IRES element localized at the 5' end of the SIV gag gene. The new stable recombinant virus elicited adaptive immune responses similar to those induced by the original recombinant virus. It is, therefore, possible to increase recombinant stability by removing functional motifs from the insert that may have deleterious effects on recombinant YF viral fitness.


Subject(s)
AIDS Vaccines/genetics , Gene Products, gag/genetics , HIV Infections/virology , Simian Immunodeficiency Virus/genetics , Yellow fever virus/genetics , AIDS Vaccines/chemistry , AIDS Vaccines/immunology , Amino Acid Sequence , Animals , Base Sequence , Cytokines/immunology , Female , Gene Products, gag/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , HIV Infections/immunology , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nucleic Acid Conformation , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/immunology , Yellow fever virus/immunology
13.
J Virol ; 88(6): 3320-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24390335

ABSTRACT

UNLABELLED: The human immunodeficiency virus type 1 (HIV-1)-encoded virion infectivity factor (Vif) is required to inactivate the host restriction factor APOBEC3 by engaging Cullin 5 (Cul5)-RING ubiquitin ligase (CRL5). Core binding factor beta (CBF-ß) is a novel regulator of Vif-CRL5 function; as yet, its mechanism of regulation remains unclear. In the present study, we demonstrate that CBF-ß promotion of Vif-CRL5 assembly is independent of its influence on Vif stability and is also a conserved feature of primate lentiviral Vif proteins. Furthermore, CBF-ß is critical for the formation of the Vif-ElonginB/ElonginC-Cul5 core E3 ubiquitin ligase complex in vitro. CBF-ß from diverse vertebrate species supported HIV-1 Vif function, indicating the conserved nature of Vif-CBF-ß interfaces. Considering the importance of the interaction between Vif and CBF-ß in viral CRL5 function, disrupting this interaction represents an attractive pharmacological intervention against HIV-1. IMPORTANCE: HIV-1 encodes virion infectivity factor (Vif) to inactivate its host's antiviral APOBEC3 proteins. Vif triggers APOBEC3 degradation by forming Vif-Cullin 5 (Cul5)-RING ubiquitin ligase (CRL5). Core binding factor beta (CBF-ß) is a novel regulator of Vif-CRL5 function whose mechanism of regulation remains poorly defined. In the present study, we demonstrate that the promotion of Vif-CRL5 assembly by CBF-ß can be separated from its influence on Vif stability. The promotion of Vif-CRL5 assembly, but not the influence on Vif stability, is conserved among primate lentiviral Vif proteins: we found that CBF-ß from diverse vertebrate species supported HIV-1 Vif function. Considering the importance of the interaction between Vif and CBF-ß in viral CRL5 function and HIV-1 replication, disrupting this interaction is an attractive strategy against HIV-1.


Subject(s)
Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Cullin Proteins/metabolism , Evolution, Molecular , HIV Infections/metabolism , HIV-1/metabolism , Simian Immunodeficiency Virus/metabolism , Ubiquitin-Protein Ligases/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Core Binding Factor beta Subunit/chemistry , Cullin Proteins/genetics , Elongin , Gene Products, vif/chemistry , Gene Products, vif/genetics , Gene Products, vif/metabolism , HIV Infections/enzymology , HIV Infections/genetics , HIV Infections/virology , HIV-1/chemistry , HIV-1/genetics , Humans , Molecular Sequence Data , Protein Binding , Sequence Alignment , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics , vif Gene Products, Human Immunodeficiency Virus/chemistry , vif Gene Products, Human Immunodeficiency Virus/genetics
14.
Virology ; 449: 140-9, 2014 Jan 20.
Article in English | MEDLINE | ID: mdl-24418547

ABSTRACT

Previous studies have shown that apolipoprotein B mRNA editing, enzyme catalytic, polypeptide G (APOBEC3G; hA3G) and F (APOBEC3F; hA3F) proteins interact with a nonlinear binding site located at the N-terminal region of the HIV-1 Vif protein. We have analyzed the role of 12 positively charged amino acids of the N-terminal region of the SIV Vif. Simian-human immunodeficiency viruses (SHIV) were constructed that expressed each of these amino acid substitutions. These viruses were examined for replication in the presence of rhesus macaque APOBEC3 proteins (rhA3A-rhA3H), incorporation of the different A3 proteins into virions, and replication in rhesus macaque PBMC. Similar to other studies, we found that K27 was essential for rhA3G activity and rhA3F but was not important for restriction of SHIVΔvif by rhA3A, rhA3D or rhA3H. Our results identified the arginine at position 14 of the SIV Vif as a critical residue for virus restriction by rhA3D, rhA3G and rhA3H.


Subject(s)
Cytidine Deaminase/metabolism , Gene Products, vif/chemistry , Gene Products, vif/metabolism , Simian Acquired Immunodeficiency Syndrome/enzymology , Simian Immunodeficiency Virus/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Cytidine Deaminase/antagonists & inhibitors , Cytidine Deaminase/genetics , Gene Products, vif/genetics , HIV Infections/enzymology , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , HIV-1/metabolism , Humans , Macaca mulatta , Molecular Sequence Data , Protein Binding , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/genetics
15.
Nature ; 505(7482): 234-8, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24336198

ABSTRACT

Lentiviruses contain accessory genes that have evolved to counteract the effects of host cellular defence proteins that inhibit productive infection. One such restriction factor, SAMHD1, inhibits human immunodeficiency virus (HIV)-1 infection of myeloid-lineage cells as well as resting CD4(+) T cells by reducing the cellular deoxynucleoside 5'-triphosphate (dNTP) concentration to a level at which the viral reverse transcriptase cannot function. In other lentiviruses, including HIV-2 and related simian immunodeficiency viruses (SIVs), SAMHD1 restriction is overcome by the action of viral accessory protein x (Vpx) or the related viral protein r (Vpr) that target and recruit SAMHD1 for proteasomal degradation. The molecular mechanism by which these viral proteins are able to usurp the host cell's ubiquitination machinery to destroy the cell's protection against these viruses has not been defined. Here we present the crystal structure of a ternary complex of Vpx with the human E3 ligase substrate adaptor DCAF1 and the carboxy-terminal region of human SAMHD1. Vpx is made up of a three-helical bundle stabilized by a zinc finger motif, and wraps tightly around the disc-shaped DCAF1 molecule to present a new molecular surface. This adapted surface is then able to recruit SAMHD1 via its C terminus, making it a competent substrate for the E3 ligase to mark for proteasomal degradation. The structure reported here provides a molecular description of how a lentiviral accessory protein is able to subvert the cell's normal protein degradation pathway to inactivate the cellular viral defence system.


Subject(s)
Carrier Proteins/metabolism , HIV/chemistry , HIV/physiology , Monomeric GTP-Binding Proteins/metabolism , Proteolysis , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Cercocebus atys/virology , Crystallography, X-Ray , Host-Pathogen Interactions , Humans , Models, Molecular , Molecular Sequence Data , Monomeric GTP-Binding Proteins/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases , SAM Domain and HD Domain-Containing Protein 1 , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/physiology , Ubiquitin-Protein Ligases , Ubiquitination , vpr Gene Products, Human Immunodeficiency Virus/chemistry , vpr Gene Products, Human Immunodeficiency Virus/metabolism
16.
Nature ; 505(7484): 502-8, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24352234

ABSTRACT

A major challenge for the development of a highly effective AIDS vaccine is the identification of mechanisms of protective immunity. To address this question, we used a nonhuman primate challenge model with simian immunodeficiency virus (SIV). We show that antibodies to the SIV envelope are necessary and sufficient to prevent infection. Moreover, sequencing of viruses from breakthrough infections revealed selective pressure against neutralization-sensitive viruses; we identified a two-amino-acid signature that alters antigenicity and confers neutralization resistance. A similar signature confers resistance of human immunodeficiency virus (HIV)-1 to neutralization by monoclonal antibodies against variable regions 1 and 2 (V1V2), suggesting that SIV and HIV share a fundamental mechanism of immune escape from vaccine-elicited or naturally elicited antibodies. These analyses provide insight into the limited efficacy seen in HIV vaccine trials.


Subject(s)
AIDS Vaccines/immunology , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/immunology , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , Disease Susceptibility/immunology , Female , Founder Effect , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/chemistry , Humans , Immune Evasion/immunology , Macaca mulatta , Male , Molecular Sequence Data , Phylogeny , Risk , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , env Gene Products, Human Immunodeficiency Virus/immunology
17.
J Virol ; 87(23): 13048-52, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24027336

ABSTRACT

Disruption of the conserved motif GYxxØ in the simian immunodeficiency virus (SIV) SIVmac239 envelope (Env) cytoplasmic tail resulted in a virus (ΔGY) that exhibited a high plasma peak but uniquely failed to acutely deplete mucosal CD4(+) T cells. Here, we show that ΔGY containing a flanking S727P mutation that was acquired in ΔGY-infected macaques reacquired the ability to rapidly deplete CD4(+) T cells in lamina propria. This suggests that the GYxxØ motif and S727P each contribute to SIV's targeting to mucosal tissues.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gene Products, env/genetics , Gene Products, env/metabolism , Mucous Membrane/immunology , Mutation, Missense , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Amino Acid Motifs , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , Gene Products, env/chemistry , Macaca , Male , Mucous Membrane/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/metabolism
18.
Cell Host Microbe ; 14(1): 85-92, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23870316

ABSTRACT

HIV-1 resulted from cross-species transmission of SIVcpz, a simian immunodeficiency virus that naturally infects chimpanzees. SIVcpz, in turn, is a recombinant between two SIV lineages from Old World monkeys. Lentiviral interspecies transmissions are partly driven by the evolution and capacity of viral accessory genes, such as vpx, vpr, and vif, to antagonize host antiviral factors, such as SAMHD1 and the APOBEC3 proteins. We show that vpx, which in other lentiviruses antagonizes SAMHD1, was deleted during the creation of SIVcpz. This genomic deletion resulted in the reconstruction of the overlapping vif gene by "overprinting," creating a unique vif that overlaps in its 3' end with the vpr gene and can antagonize hominid APOBEC3s. Moreover, passage of SIVs through chimpanzees facilitated the subsequent adaptation of HIV-1 to humans. Thus, HIV-1 originated through a series of gene loss and adaptation events that generated its chimpanzee precursor and lowered the species barrier to human infection.


Subject(s)
Evolution, Molecular , Gene Deletion , HIV Infections/virology , HIV-1/genetics , Hominidae/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Amino Acid Sequence , Animals , Gene Products, vif/chemistry , Gene Products, vif/genetics , Gene Products, vif/metabolism , Gene Products, vpr/chemistry , Gene Products, vpr/genetics , Gene Products, vpr/metabolism , HIV-1/chemistry , HIV-1/classification , HIV-1/metabolism , Haplorhini , Humans , Molecular Sequence Data , Pan troglodytes , Phylogeny , Sequence Alignment , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/classification , Simian Immunodeficiency Virus/metabolism
19.
Biophys Chem ; 171: 54-62, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23176826

ABSTRACT

In this study we used an engineered six-helix bundle construct corresponding to the fusogenic core of the SIV gp41 protein as a model system to investigate the folding of a trimeric protein, which acquires a compact structure upon association of largely unstructured monomeric peptides. Thirteen mutants were generated in order to gain information about the thermodynamic and kinetic roles of topologically conserved tertiary interactions to folding and stability. The effect of the mutations was assessed by circular dichroism spectroscopy from urea-induced equilibrium unfolding experiments and in time-resolved mode to follow the kinetics of refolding and unfolding. While individual experiments can be interpreted in terms of a simple monomer-trimer refolding/unfolding reaction mechanism, comparison of equilibrium and kinetic data reveals that some variants clearly deviate from this two-state behavior and that most proteins cannot be classified as two-state folders without some reservations. Nevertheless, following "quasi-φ-value" and "quasi-ß(T)-value" analyses, we propose that the highest-energy barrier along the folding pathway is passed in the trimeric state, after the C-terminal half of each monomer chain is "fixed" in anti-parallel orientation to the surface of the central, still nascent N-terminal coiled-coil.


Subject(s)
Membrane Glycoproteins/chemistry , Protein Folding , Retroviridae Proteins/chemistry , Simian Immunodeficiency Virus/chemistry , Kinetics , Membrane Glycoproteins/genetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Retroviridae Proteins/genetics , Simian Immunodeficiency Virus/genetics , Thermodynamics
20.
J Phys Chem B ; 116(46): 13713-21, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23094791

ABSTRACT

Fusion peptides of type I fusion glycoproteins are structural elements of several enveloped viruses which enable the fusion between host and virus membranes. It is generally suggested that these peptides can promote the early fusion steps by inducing membrane curvature and that they adopt a tilted helical conformation in membranes. Although this property has been the subject of several experimental and in silico studies, an extensive sampling of the membrane peptide interaction has not yet been done. In this study, we performed coarse-grained molecular dynamic simulations in which the lipid bilayer self-assembles around the peptide. The simulations indicate that the SIV fusion peptide can adopt two different orientations in a DPPC bilayer, a major population which adopts a tilted interfacial orientation and a minor population which is perpendicular to the bilayer. The simulations also indicate that for the SIV mutant that does not induce fusion in vitro the tilt is abolished.


Subject(s)
Molecular Dynamics Simulation , Peptides/chemistry , Simian Immunodeficiency Virus/chemistry , Viral Fusion Proteins/chemistry , Lipid Bilayers/chemistry , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...