Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Virol ; 95(11)2021 05 10.
Article in English | MEDLINE | ID: mdl-33731460

ABSTRACT

In many enveloped virus families, including HIV and HSV, a crucial, yet unexploited, step in the viral life cycle is releasing particles from the infected cell membranes. This release process is mediated by host ESCRT complex proteins, which are recruited by viral structural proteins and provides the mechanical means for membrane scission and subsequent viral budding. The prazole drug, tenatoprazole, was previously shown to bind to ESCRT complex member Tsg101 and to quantitatively block the release of infectious HIV-1 from cells in culture. In this report we show that tenatoprazole and a related prazole drug, ilaprazole, effectively block infectious Herpes Simplex Virus (HSV)-1/2 release from Vero cells in culture. By electron microscopy, we found that both prazole drugs block the transit of HSV particles through the cell nuclear membrane resulting in their accumulation in the nucleus. Ilaprazole also quantitatively blocks the release of HIV-1 from 293T cells with an EC50 of 0.8-1.2 µM, which is much more potent than tenatoprazole. Our results indicate that prazole-based compounds may represent a class of drugs with potential to be broad-spectrum antiviral agents against multiple enveloped viruses, by interrupting cellular Tsg101 interaction with maturing virus, thus blocking the budding process that releases particles from the cell.ImportanceThese results provide the basis for the development of drugs that target enveloped virus budding that can be used ultimately to control multiple virus infections in humans.

2.
Nat Commun ; 8(1): 1391, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29123089

ABSTRACT

HIV-1 replication requires Tsg101, a component of cellular endosomal sorting complex required for transport (ESCRT) machinery. Tsg101 possesses an ubiquitin (Ub) E2 variant (UEV) domain with a pocket that can bind PT/SAP motifs and another pocket that can bind Ub. The PTAP motif in the viral structural precursor polyprotein, Gag, allows the recruitment of Tsg101 and other ESCRTs to virus assembly sites where they mediate budding. It is not known how or even whether the UEV Ub binding function contributes to virus production. Here, we report that disruption of UEV Ub binding by commonly used drugs arrests assembly at an early step distinct from the late stage involving PTAP binding disruption. NMR reveals that the drugs form a covalent adduct near the Ub-binding pocket leading to the disruption of Ub, but not PTAP binding. We conclude that the Ub-binding pocket has a chaperone function involved in bud initiation.


Subject(s)
DNA-Binding Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , HIV-1/metabolism , Transcription Factors/metabolism , Virus Assembly/physiology , Virus Release/physiology , gag Gene Products, Human Immunodeficiency Virus/metabolism , 2-Pyridinylmethylsulfinylbenzimidazoles/pharmacology , Anti-HIV Agents/pharmacology , Binding Sites , Cell Line, Tumor , DNA-Binding Proteins/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Esomeprazole/pharmacology , HEK293 Cells , HeLa Cells , Humans , Molecular Chaperones/metabolism , Protein Binding , Protein Domains , Transcription Factors/genetics , Ubiquitin/metabolism , Virus Assembly/drug effects , Virus Assembly/genetics , Virus Release/drug effects , Virus Release/genetics
3.
Adv Virol ; 2012: 532723, 2012.
Article in English | MEDLINE | ID: mdl-22666250

ABSTRACT

Pathogenic strains of viruses that infect humans are encapsulated in membranes derived from the host cell in which they infect. After replication, these viruses are released by a budding process that requires cell/viral membrane scission. As such, this represents a natural target for innate immunity mechanisms to interdict enveloped virus spread and recent advances in this field will be the subject of this paper.

4.
J Virol ; 85(14): 7153-61, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21543490

ABSTRACT

Budding of retroviruses from cell membranes requires ubiquitination of Gag and recruitment of cellular proteins involved in endosome sorting, including endosome sorting complex required for transport III (ESCRT-III) protein complex and vacuolar protein sorting 4 (VPS4) and its ATPase. In response to infection, a cellular mechanism has evolved that blocks virus replication early and late in the budding process through expression of interferon-stimulated gene 15 (ISG15), a dimer homologue of ubiquitin. Interferon treatment of DF-1 cells blocks avian sarcoma/leukosis virus release, demonstrating that this mechanism is functional under physiological conditions. The late block to release is caused in part by a loss in interaction between VPS4 and its coactivator protein LIP5, which is required to promote the formation of the ESCRT III-VPS4 double-hexamer complex to activate its ATPase. ISG15 is conjugated to two different LIP5-ESCRT-III-binding charged multivesicular body proteins, CHMP2A and CHMP5. Upon ISGylation of each, interaction with LIP5 is no longer detected. Two other ESCRT-III proteins, CHMP4B and CHMP6, are also conjugated to ISG15. ISGylation of CHMP2A, CHMP4B, and CHMP6 weakens their binding directly to VPS4, thereby facilitating the release of this protein from the membrane into the cytosol. The remaining budding complex fails to release particles from the cell membrane. Introducing a mutant of ISG15 into cells that cannot be conjugated to proteins prevents the ISG15-dependent mechanism from blocking virus release. CHMP5 is the primary switch to initiate the antiviral mechanism, because removal of CHMP5 from cells prevents ISGylation of CHMP2A and CHMP6.


Subject(s)
Cytokines/genetics , Interferons/physiology , Retroviridae/physiology , Ubiquitins/genetics , Blotting, Western , Cell Line , Electrophoresis, Polyacrylamide Gel , Humans , Mutagenesis, Site-Directed , RNA, Small Interfering , Virus Replication
5.
J Virol ; 84(9): 4725-36, 2010 May.
Article in English | MEDLINE | ID: mdl-20164219

ABSTRACT

The release of retroviruses from cells requires ubiquitination of Gag and recruitment of cellular proteins involved in endosome sorting, including the ESCRT-III proteins and the Vps4 ATPase. In response to infection, cells have evolved an interferon-induced mechanism to block virus replication through expression of the interferon-stimulated gene 15 (ISG15), a dimer homologue of ubiquitin, which interferes with ubiquitin pathways in cells. Previously, it has been reported that ISG15 expression inhibited the E3 ubiquitin ligase, Nedd4, and prevented association of the ESCRT-I protein Tsg101 with human immunodeficiency virus type 1 (HIV-1) Gag. The budding of avian sarcoma leukosis virus and HIV-1 Gag virus-like particles containing L-domain mutations can be rescued by fusion to ESCRT proteins, which cause entry into the budding pathway beyond these early steps. The release of these fusions from cells was susceptible to inhibition by ISG15, indicating that there was a block late in the budding process. We now demonstrate that the Vps4 protein does not associate with the avian sarcoma leukosis virus or the HIV-1 budding complexes when ISG15 is expressed. This is caused by a loss in interaction between Vps4 with its coactivator protein LIP5 needed to promote the formation of the ESCRT-III-Vps4 double-hexamer complex required for membrane scission and virus release. The inability of LIP5 to interact with Vps4 is the probable result of ISG15 conjugation to the ESCRT-III protein, CHMP5, which regulates the availability of LIP5. Thus, there appear to be multiple levels of ISG15-induced inhibition acting at different stages of the virus release process.


Subject(s)
Avian Sarcoma Viruses/immunology , Avian Sarcoma Viruses/physiology , Cytokines/immunology , HIV-1/immunology , HIV-1/physiology , Interferons/immunology , Ubiquitins/immunology , Virus Release , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/metabolism , Cell Line , Endosomal Sorting Complexes Required for Transport/metabolism , Fibroblasts/virology , Humans , Vacuolar Proton-Translocating ATPases
6.
Adv Virol ; 2009: 6239691-6239699, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19865606

ABSTRACT

Retroviruses have evolved a mechanism for the release of particles from the cell membrane that appropriates cellular protein complexes, referred to as ESCRT-I, -II, -III, normally involved in the biogenesis of multivesicular bodies. Three different classes of late assembly (L) domains encoded in Gag, with core sequences of PPXY, PTAP, and YPXL, recruit different components of the ESCRT machinery to form a budding complex for virus release. Here, we highlight recent progress in identifying the role of different ESCRT complexes in facilitating budding, ubiquitination, and membrane targeting of avian sarcoma and leukosis virus (ASLV) and human immunodeficiency virus, type 1 (HIV-1). These findings show that retroviruses adopt parallel budding pathways by recruiting different host factors from common cellular machinery for particle release.

7.
J Mol Biol ; 385(2): 568-79, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19014951

ABSTRACT

A tetramer model for human immunodeficiency virus type 1 (HIV-1) integrase (IN) with DNA representing long terminal repeat (LTR) termini was previously assembled to predict the IN residues that interact with the LTR termini; these predictions were experimentally verified for nine amino acid residues [Chen, A., Weber, I. T., Harrison, R. W. & Leis, J. (2006). Identification of amino acids in HIV-1 and avian sarcoma virus integrase subsites required for specific recognition of the long terminal repeat ends. J. Biol. Chem., 281, 4173-4182]. In a similar strategy, the unique amino acids found in avian sarcoma virus IN, rather than HIV-1 or Mason-Pfizer monkey virus IN, were substituted into the structurally related positions of HIV-1 IN. Substitutions of six additional residues (Q44, L68, E69, D229, S230, and D253) showed changes in the 3' processing specificity of the enzyme, verifying their predicted interaction with the LTR DNA. The newly identified residues extend interactions along a 16-bp length of the LTR termini and are consistent with known LTR DNA/HIV-1 IN cross-links. The tetramer model for HIV-1 IN with LTR termini was modified to include two IN binding domains for lens-epithelium-derived growth factor/p75. The target DNA was predicted to bind in a surface trench perpendicular to the plane of the LTR DNA binding sites of HIV-1 IN and extending alongside lens-epithelium-derived growth factor. This hypothesis is supported by the in vitro activity phenotype of HIV-1 IN mutant, with a K219S substitution showing loss in strand transfer activity while maintaining 3' processing on an HIV-1 substrate. Mutations at seven other residues reported in the literature have the same phenotype, and all eight residues align along the length of the putative target DNA binding trench.


Subject(s)
DNA/metabolism , HIV Integrase/genetics , HIV Integrase/metabolism , HIV-1/physiology , Amino Acid Substitution/genetics , Binding Sites , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Multimerization , Protein Structure, Quaternary
8.
J Biol Chem ; 283(44): 29822-30, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18723511

ABSTRACT

Members of the Nedd4 family of E3 ubiquitin ligases bind the L domain in avian sarcoma virus (ASV) Gag and facilitate viral particle release. Translational fusion of ASV Gag with an L domain deletion (Deltap2b) to proteins that comprise ESCRT-I, -II, and -III (the endocytic sorting complexes required for transport) rescued both Gag ubiquitination and particle release from cells. The ESCRT-I factors Vps37C or Tsg101 were more effective in rescue of Gag/Deltap2b budding than the ESCRT-II factor Eap20 or the ESCRT-III component CHMP6. Thus ESCRT components can substitute for Nedd4 family members in ASV Gag release. Unlike wild type, ASV Gag/Deltap2b -ESCRT chimeras failed to co-immunoprecipitate with co-expressed hemagglutinin-tagged Nedd4, indicating that Nedd4 was not stably associated with these Gag fusions. Release of the Gag-ESCRT-I or -II fusions was inhibited by a dominant negative mutant of Vps4 ATPase similar to wild type ASV Gag. In contrast to ASV Gag, HIV-1 Gag containing an L domain inactivating mutation (P7L) was efficiently rescued by fusion to a component of ESCRT-III (Chmp6) but not ESCRT-II (Eap20). Depletion of the endogenous pool of Eap20 (ESCRT-II) had little effect on HIV-1 Gag release but blocked ASV Gag release. In contrast, depletion of the endogenous pool of Vps37C (ESCRT-I) had little effect on ASV but blocked HIV-1 Gag release. Furthermore, an N-terminal fragment of Chmp6 inhibited both HIV-1 and ASV Gag release in a dominant negative manner. Taken together, these results indicate that ASV and HIV-1 Gag utilize different combinations of ESCRT proteins to facilitate the budding process, although they share some common elements.


Subject(s)
Avian Sarcoma Viruses/metabolism , HIV-1/metabolism , Vesicular Transport Proteins/metabolism , Binding Sites , Biological Transport , Cell Line , Endocytosis , Gene Products, gag/metabolism , Humans , Models, Biological , Mutation , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Ubiquitin/chemistry , Vesicular Transport Proteins/chemistry
9.
Virology ; 377(1): 30-8, 2008 Jul 20.
Article in English | MEDLINE | ID: mdl-18555885

ABSTRACT

The Late (L) domain of the avian sarcoma virus (ASV) Gag protein binds Nedd4 ubiquitin ligase E3 family members and is the determinant of efficient virus release in avian and mammalian cells. We previously demonstrated that Nedd4 and Tsg101 constitutively interact raising the possibility that Nedd4 links ASV Gag to the ESCRT machinery. We now demonstrate that covalently linking Tsg101 to ASV Gag lacking the Nedd4 binding site (Deltap2b-Tsg101) ablates the requirement for Nedd4, but the rescue of budding occurs by use of a different budding mechanism than that used by wild type ASV Gag. The evidence that Tsg101 and Nedd4 direct release by different pathways is: (i) Release of the virus-like particles (VLPs) assembled from Gag in DF-1, an avian cell line, was resistant to dominant-negative interference by a Tsg101 mutant previously shown to inhibit release of both HIV and Mo-MLV. (ii) Release of VLPs from DF-1 cells was resistant to siRNA-mediated depletion of the endogenous pool of Tsg101 in these cells. (iii) VLPs assembled from wild-type ASV Gag exhibited highly efficient release from endosome-like membrane domains enriched in the tetraspanin protein CD63 or a fluorescent analogue of the phospholipid phosphatidylethanolamine. However, the VLPs assembled from the L domain mutant Deltap2b or a chimeric Deltap2b-Tsg101 Gag lacked these domain markers even though the chimeric Gag was released efficiently compared to the Deltap2b mutant. These results suggest that Tsg101 and Nedd4 facilitate Gag release through functionally exchangeable but independent routes and that Tsg101 can replace Nedd4 function in facilitating budding but not directing through the same membranes.


Subject(s)
Avian Sarcoma Viruses/genetics , Avian Sarcoma Viruses/physiology , DNA-Binding Proteins/physiology , Gene Products, gag/genetics , Gene Products, gag/metabolism , Transcription Factors/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Avian Sarcoma Viruses/ultrastructure , Birds , COS Cells , Cell Line , Cell Membrane/ultrastructure , Cell Membrane/virology , Chlorocebus aethiops , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Endosomal Sorting Complexes Required for Transport , Genes, gag , Microscopy, Electron, Transmission , Nedd4 Ubiquitin Protein Ligases , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transfection
10.
J Biol Chem ; 281(7): 4173-82, 2006 Feb 17.
Article in English | MEDLINE | ID: mdl-16298997

ABSTRACT

A tetramer model for HIV-1 integrase (IN) with DNA representing 20 bp of the U3 and U5 long terminal repeats (LTR) termini was assembled using structural and biochemical data and molecular dynamics simulations. It predicted amino acid residues on the enzyme surface that can interact with the LTR termini. A separate structural alignment of HIV-1, simian sarcoma virus (SIV), and avian sarcoma virus (ASV) INs predicted which of these residues were unique. To determine whether these residues were responsible for specific recognition of the LTR termini, the amino acids from ASV IN were substituted into the structurally equivalent positions of HIV-1 IN, and the ability of the chimeras to 3 ' process U5 HIV-1 or ASV duplex oligos was determined. This analysis demonstrated that there are multiple amino acid contacts with the LTRs and that substitution of ASV IN amino acids at many of the analogous positions in HIV-1 IN conferred partial ability to cleave ASV substrates with a concomitant loss in the ability to cleave the homologous HIV-1 substrate. HIV-1 IN residues that changed specificity include Val(72), Ser(153), Lys(160)-Ile(161), Gly(163)-Val(165), and His(171)-Leu(172). Because a chimera that combines several of these substitutions showed a specificity of cleavage of the U5 ASV substrate closer to wild type ASV IN compared with chimeras with individual amino acid substitutions, it appears that the sum of the IN interactions with the LTRs determines the specificity. Finally, residues Ser(153) and Val(72) in HIV-1 IN are among those that change in enzymes that develop resistance to naphthyridine carboxamide- and diketo acid-related inhibitors in cells. Thus, amino acid residues involved in recognition of the LTRs are among these positions that change in development of drug resistance.


Subject(s)
Avian Sarcoma Viruses/enzymology , HIV Integrase/chemistry , HIV Long Terminal Repeat , Integrases/chemistry , Terminal Repeat Sequences , Amino Acid Sequence , Amino Acid Substitution , Integrase Inhibitors/pharmacology , Models, Molecular , Molecular Sequence Data
11.
Traffic ; 6(10): 880-94, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16138902

ABSTRACT

The functionally exchangeable L domains of HIV-1 and Rous sarcoma virus (RSV) Gag bind Tsg101 and Nedd4, respectively. Tsg101 and Nedd4 function in endocytic trafficking, and studies show that expression of Tsg101 or Nedd4 fragments interfere with release of HIV-1 or RSV Gag, respectively, as virus-like particles (VLPs). To determine whether functional exchangeability reflects use of the same trafficking pathway, we tested the effect on RSV Gag release of co-expression with mutated forms of Vps4, Nedd4 and Tsg101. A dominant-negative mutant of Vps4A, an AAA ATPase required for utilization of endosomal sorting proteins that was shown previously to interfere with HIV-1 budding, also inhibited RSV Gag release, indicating that RSV uses the endocytic trafficking machinery, as does HIV. Nedd4 and Tsg101 interacted in the presence or absence of Gag and, through its binding of Nedd4, RSV Gag interacted with Tsg101. Deletion of the N-terminal region of Tsg101 or the HECT domain of Nedd4 did not prevent interaction; however, three-dimensional spatial imaging suggested that the interaction of RSV Gag with full-length Tsg101 and N-terminally truncated Tsg101 was not the same. Co-expression of RSV Gag with the Tsg101 C-terminal fragment interfered with VLP release minimally; however, a significant fraction of the released VLPs was tethered to each other. The results suggest that, while Tsg101 is not required for RSV VLP release, alterations in the protein interfere with VLP budding/fission events. We conclude that RSV and HIV-1 Gag direct particle release through independent ESCRT-mediated pathways that are linked through Tsg101-Nedd4 interaction.


Subject(s)
Avian Sarcoma Viruses/metabolism , DNA-Binding Proteins/metabolism , Gene Products, gag/metabolism , HIV-1/metabolism , Protein Transport , Transcription Factors/metabolism , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Endosomal Sorting Complexes Required for Transport , Gene Products, gag/genetics , Hemagglutinins/metabolism , Humans , Nedd4 Ubiquitin Protein Ligases , Peptide Fragments/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vacuolar Proton-Translocating ATPases , Vesicular Transport Proteins
12.
Retrovirology ; 2: 58, 2005 Sep 27.
Article in English | MEDLINE | ID: mdl-16188035

ABSTRACT

A series of amino acid substitutions (M239F, M239G, P240F, V241G) were placed in the p10-CA protease cleavage site (VVAM*PVVI) to change the rate of cleavage of the junction. The effects of these substitutions on p10-CA cleavage by RSV PR were confirmed by measuring the kinetics of cleavage of model peptide substrates containing the wild type and mutant p10-CA sites. The effects of these substitutions on processing of the Gag polyprotein were determined by labeling Gag transfected COS-1 cells with 35S-Met and -Cys, and immunoprecipitation of Gag and its cleavage products from the media and lysate fractions. All substitutions except M239F caused decreases in detectable Gag processing and subsequent release from cells. Several of the mutants also caused defects in production of the three CA proteins. The p10-CA mutations were subcloned into an RSV proviral vector (RCAN) and introduced into a chick embryo fibroblast cell line (DF-1). All of the mutations except M239F blocked RSV replication. In addition, the effects of the M239F and M239G substitutions on the morphology of released virus particles were examined by electron microscopy. While the M239F particles appeared similar to wild type particles, M239G particles contained cores that were large and misshapen. These results suggest that mutations affecting cleavage at the p10-CA protease cleavage site block RSV replication and can have a negative impact on virus particle morphology.


Subject(s)
Aspartic Acid Endopeptidases/physiology , Avian Sarcoma Viruses/physiology , Capsid Proteins/chemistry , Gene Products, gag/metabolism , Animals , COS Cells , Capsid Proteins/physiology , Chlorocebus aethiops , Mutation , Structure-Activity Relationship , Virion/physiology , Virus Assembly , Virus Replication
13.
J Virol ; 78(24): 13943-53, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15564502

ABSTRACT

Rous sarcoma virus (RSV) budding requires an interaction of the L domain within the p2b region of Gag with cellular Nedd4-family E3 ubiquitin protein ligases. Members of our laboratories previously demonstrated that overexpression of a fragment of the chicken Nedd4-like protein (LDI-1 WW) inhibits Gag release in a dominant-negative manner (A. Kikonyogo, F. Bouamr, M. L. Vana, Y. Xiang, A. Aiyar, C. Carter, and J. Leis, Proc. Natl. Acad. Sci. USA 98:11199-11204, 2001). We have now identified the complete 3' end of LDI-1 and determined that it has a C-terminal ubiquitin ligase HECT domain, similar to other Nedd4 family members. While overexpression of the full-length LDI-1 clone (LDI-1 FL) had little effect on Gag budding, an LDI-1 FL mutant with a substitution in the HECT domain catalytic site blocked Gag release, similar to LDI-1 WW. The coexpression of Gag and hemagglutinin-tagged ubiquitin (HA-Ub) resulted in the detection of mono- and polyubiquitinated forms of Gag in cells and mostly monoubiquitinated Gag in virus-like particles (VLPs). When the Nedd4-binding site (L domain) was deleted, ubiquitinated Gag was not detected. Interestingly, the release of Gag with ubiquitin covalently linked to the C terminus (Gag-Ub) was still blocked by LDI-1 WW. To understand the mechanism of this inhibition, we examined cells expressing Gag and LDI-1 WW by electron microscopy. In the presence of LDI-1 WW, VLPs were found in electron-dense inclusion bodies in the cytoplasm of transfected cells. In contrast, when cells that coexpressed Gag-Ub and LDI-1 WW were examined, inclusion bodies were detected but did not contain VLPs. These results indicate that the ubiquitination of Gag is dependent upon Nedd4 binding to the L domain and suggest that Nedd4 has additional functions during RSV release besides the ubiquitination of Gag.


Subject(s)
Avian Sarcoma Viruses/growth & development , Gene Expression Regulation, Viral , Gene Products, gag/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Avian Sarcoma Viruses/genetics , Avian Sarcoma Viruses/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Endosomal Sorting Complexes Required for Transport , Gene Products, gag/chemistry , Humans , Mice , Microscopy, Confocal , Molecular Sequence Data , Mutation , Nedd4 Ubiquitin Protein Ligases , Rabbits , Ubiquitin-Protein Ligases/genetics , Virion/metabolism
14.
BMC Biol ; 2: 8, 2004 May 20.
Article in English | MEDLINE | ID: mdl-15153244

ABSTRACT

BACKGROUND: The 5' end of the Rous sarcoma virus (RSV) RNA around the primer-binding site forms a series of RNA secondary stem/loop structures (U5-IR stem, TpsiC interaction region, U5-leader stem) that are required for efficient initiation of reverse transcription. The U5-IR stem and loop also encode the U5 integrase (IN) recognition sequence at the level of DNA such that this region has overlapping biological functions in reverse transcription and integration. RESULTS: We have investigated the ability of RSV to tolerate mutations in and around the U5 IR stem and loop. Through the use of viral libraries with blocks of random sequence, we have screened for functional mutants in vivo, growing the virus libraries in turkey embryo fibroblasts. The library representing the U5-IR stem rapidly selects for clones that maintain the structure of the stem, and is subsequently overtaken by wild type sequence. In contrast, in the library representing the U5-IR loop, wild type sequence is found after five rounds of infection but it does not dominate the virus pool, indicating that the mutant sequences identified are able to replicate at or near wild type levels. CONCLUSION: These results indicate that the region of the RNA genome in U5 adjacent to the PBS tolerates much sequence variation even though it is required for multiple biological functions in replication. The in vivo selection method utilized in this study was capable of detecting complex patterns of selection as well as identifying biologically relevant viral mutants.


Subject(s)
Avian Sarcoma Viruses/genetics , Mutation , RNA, Small Nuclear/genetics , RNA, Viral/genetics , Avian Sarcoma Viruses/chemistry , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/virology , Gene Library , Genome , Nucleic Acid Conformation , RNA, Small Nuclear/chemistry , RNA, Viral/chemistry , Reverse Transcription , Transfection , Turkey
15.
J Virol ; 76(15): 7571-7, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12097570

ABSTRACT

Reverse transcription in avian sarcoma virus (ASV) initiates from the 3' end of a tRNA(Trp) primer, which anneals near the 5' end of the RNA genome. The region around the primer-binding site (PBS) forms an elaborate stem structure composed of the U5-inverted repeat (U5-IR) stem, the U5-leader stem, and the association of the tRNA primer with the PBS. There is evidence for an additional interaction between the viral U5 RNA and the T psi C loop of the tRNA(Trp) (U5-T psi C). We now demonstrate that this U5-T psi C interaction is necessary for efficient replication of ASV in culture. By randomizing specific biologically relevant regions of the viral RNA, thereby producing a library of mutant viruses, we are able to select, through multiple rounds of infection, those sequences imparting survival fitness to the virus. Randomizing the U5-T psi C interaction region of the viral RNA results in selection of largely wild-type sequences after five rounds of infection. Also recovered are mutant viruses that maintain their ability to base pair with the T psi C loop of the tRNA(Trp). To prove this interaction is specific to the tRNA primer, we constructed a second library, in which we altered the PBS to anneal to tRNA(Pro), while simultaneously randomizing the viral RNA U5-T psi C region. After five rounds of infection, the consensus sequence 5'-GPuPuCPy-3' emerged, which is complementary to the 5'-GGTTC-3' sequence found in the T psi C loop of tRNA(Pro). These observations confirm the importance of the U5-T psi C interaction in vivo.


Subject(s)
Avian Sarcoma Viruses/genetics , RNA, Transfer, Trp/metabolism , RNA, Viral/metabolism , Virus Replication , Animals , Avian Sarcoma Viruses/physiology , Base Sequence , Binding Sites/genetics , Cell Line , Gene Library , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Transfer, Trp/chemistry , RNA, Transfer, Trp/genetics , RNA, Viral/genetics
16.
J Biol Chem ; 277(21): 18357-64, 2002 May 24.
Article in English | MEDLINE | ID: mdl-11897790

ABSTRACT

Successful integration of viral genome into a host chromosome depends on interaction between viral integrase and its recognition sequences. We have used a reconstituted concerted human immunodeficiency virus, type 1 (HIV-1), integration system to analyze the role of integrase (IN) recognition sequences in formation of the IN-viral DNA complex capable of concerted integration. HIV-1 integrase was presented with substrates that contained all 4 bases at 8 mismatched positions that define the inverted repeat relationship between U3 and U5 long terminal repeats (LTR) termini and at positions 17-19, which are conserved in the termini. Evidence presented indicates that positions 17-20 of the IN recognition sequences are needed for a concerted DNA integration mechanism. All 4 bases were found at each randomized position in sequenced concerted DNA integrants, although in some instances there were preferences for specific bases. These results indicate that integrase tolerates a significant amount of plasticity as to what constitutes an IN recognition sequence. By having several positions randomized, the concerted integrants were examined for statistically significant relationships between selections of bases at different positions. The results of this analysis show not only relationships between different positions within the same LTR end but also between different positions belonging to opposite DNA termini.


Subject(s)
HIV Integrase/metabolism , Base Sequence , DNA Primers , HIV Long Terminal Repeat , Substrate Specificity
17.
J Mol Biol ; 316(3): 667-78, 2002 Feb 22.
Article in English | MEDLINE | ID: mdl-11866525

ABSTRACT

During assembly and morphogenesis of Rous sarcoma virus (RSV), proteolytic processing of the structural precursor (Pr76Gag) protein generates three capsid (CA) protein variants, CA476, CA479, and CA488. The proteins share identical N-terminal domains (NTDs), but are truncated at residues corresponding to gag codons 476, 479, and 488 in their CA C-terminal domains (CTDs). To characterize oligomeric forms of the RSV CA variants, we examined 2D crystals of the capsid proteins, assembled on lipid monolayers. Using electron microscopy and image analysis approaches, the CA proteins were observed to organize in hexagonal (p6) arrangements, where rings of membrane-proximal NTD hexamers were spaced at 95 A intervals. Differences between the oligomeric structures of the CA variants were most evident in membrane-distal regions, where apparent CTDs interconnect hexamer rings. In this region, CA488 connections were observed readily, while CA476 and CA479 contacts were resolved poorly, suggesting that in vivo processing of CA488 to the shorter forms may permit virions to adopt a dissembly-competent conformation. In addition to crystalline arrays, the CA479 and CA488 proteins formed small spherical particles with diameters of 165-175 A. The spheres appear to be arranged from hexamer or hexamer plus pentamer ring subunits that are related to the 2D crystal forms. Our results implicate RSV CA hexamer rings as basic elements in the assembly of RSV virus cores.


Subject(s)
Avian Sarcoma Viruses/metabolism , Avian Sarcoma Viruses/ultrastructure , Capsid/metabolism , Capsid/ultrastructure , Membrane Lipids/metabolism , Mutation/genetics , Amino Acid Sequence , Avian Sarcoma Viruses/genetics , Capsid/chemistry , Capsid/genetics , Crystallization , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary
18.
J Biol Chem ; 277(13): 10938-48, 2002 Mar 29.
Article in English | MEDLINE | ID: mdl-11788585

ABSTRACT

We have reconstituted concerted human immunodeficiency virus type 1 (HIV-1) integration with specially designed mini-donor DNA, a supercoiled plasmid acceptor, purified bacterial-derived HIV-1 integrase (IN), and host HMG-I(Y) protein (Hindmarsh, P., Ridky, T., Reeves, R., Andrake, M., Skalka, A. M., and Leis, J. (1999) J. Virol. 73, 2994-3003). Integration in this system is dependent upon the mini donor DNA having IN recognition sequences at both ends and the reaction products have all of the features associated with integration of viral DNA in vivo. Using this system, we explored the relationship between the HIV-1 U3 and U5 IN recognition sequences by analyzing substrates that contain either two U3 or two U5 terminal sequences. Both substrates caused severe defects to integration but with different effects on the mechanism indicating that the U3 and the U5 sequences are both required for concerted DNA integration. We have also used the reconstituted system to compare the mechanism of integration catalyzed by HIV-1 to that of avian sarcoma virus by analyzing the effect of defined mutations introduced into U3 or U5 ends of the respective wild type DNA substrates. Despite sequence differences between avian sarcoma virus and HIV-1 IN and their recognition sequences, the consequences of analogous base pair substitutions at the same relative positions of the respective IN recognition sequences were very similar. This highlights the common mechanism of integration shared by these two different viruses.


Subject(s)
HIV Integrase/metabolism , HIV-1/physiology , Mutation , Virus Integration , Base Pairing , Base Sequence , DNA Primers , HIV Integrase/chemistry , HIV Integrase/genetics , Plasmids
SELECTION OF CITATIONS
SEARCH DETAIL
...