Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Biomolecules ; 11(12)2021 12 20.
Article in English | MEDLINE | ID: mdl-34944553

ABSTRACT

Integrases of different retroviruses assemble as functional complexes with varying multimers of the protein. Retroviral integrases require a divalent metal cation to perform one-step transesterification catalysis. Tetrameric prototype foamy virus (PFV) intasomes assembled from purified integrase and viral DNA oligonucleotides were characterized for their activity in the presence of different cations. While most retroviral integrases are inactive in calcium, PFV intasomes appear to be uniquely capable of catalysis in calcium. The PFV intasomes also contrast with other retroviral integrases by displaying an inverse correlation of activity with increasing manganese beginning at relatively low concentrations. The intasomes were found to be significantly more active in the presence of chloride co-ions compared to acetate. While HIV-1 integrase appears to commit to a target DNA within 20 s, PFV intasomes do not commit to target DNA during their reaction lifetime. Together, these data highlight the unique biochemical activities of PFV integrase compared to other retroviral integrases.


Subject(s)
DNA/metabolism , Integrases/chemistry , Integrases/metabolism , Spumavirus/enzymology , Acetates/metabolism , Binding Sites , Chlorides/metabolism , Esterification , Manganese/metabolism , Oligonucleotides , Spumavirus/chemistry , Viral Proteins/chemistry , Viral Proteins/metabolism
2.
Viruses ; 13(8)2021 07 29.
Article in English | MEDLINE | ID: mdl-34452360

ABSTRACT

In most cases, proteolytic processing of the retroviral Pol portion of the Gag-Pol polyprotein precursor produces protease (PR), reverse transcriptase (RT), and integrase (IN). However, foamy viruses (FVs) express Pol separately from Gag and, when Pol is processed, only the IN domain is released. Here, we report a 2.9 Å resolution crystal structure of the mature PR-RT from prototype FV (PFV) that can carry out both proteolytic processing and reverse transcription but is in a configuration not competent for proteolytic or polymerase activity. PFV PR-RT is monomeric and the architecture of PFV PR is similar to one of the subunits of HIV-1 PR, which is a dimer. There is a C-terminal extension of PFV PR (101-145) that consists of two helices which are adjacent to the base of the RT palm subdomain, and anchors PR to RT. The polymerase domain of PFV RT consists of fingers, palm, thumb, and connection subdomains whose spatial arrangements are similar to the p51 subunit of HIV-1 RT. The RNase H and polymerase domains of PFV RT are connected by flexible linkers. Significant spatial and conformational (sub)domain rearrangements are therefore required for nucleic acid binding. The structure of PFV PR-RT provides insights into the conformational maturation of retroviral Pol polyproteins.


Subject(s)
Peptide Hydrolases/chemistry , Polyproteins/chemistry , RNA-Directed DNA Polymerase/chemistry , Spumavirus/chemistry , Crystallization , Peptide Hydrolases/metabolism , Polyproteins/metabolism , RNA-Directed DNA Polymerase/metabolism , Reverse Transcription
3.
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
4.
Viruses ; 11(7)2019 07 02.
Article in English | MEDLINE | ID: mdl-31269675

ABSTRACT

Reverse transcription describes the process of the transformation of single-stranded RNA into double-stranded DNA via an RNA/DNA duplex intermediate, and is catalyzed by the viral enzyme reverse transcriptase (RT). This event is a pivotal step in the life cycle of all retroviruses. In contrast to orthoretroviruses, the domain structure of the mature RT of foamy viruses is different, i.e., it harbors the protease (PR) domain at its N-terminus, thus being a PR-RT. This structural feature has consequences on PR activation, since the enzyme is monomeric in solution and retroviral PRs are only active as dimers. This review focuses on the structural and functional aspects of simian and prototype foamy virus reverse transcription and reverse transcriptase, as well as special features of reverse transcription that deviate from orthoretroviral processes, e.g., PR activation.


Subject(s)
Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , RNA-Directed DNA Polymerase/chemistry , RNA-Directed DNA Polymerase/metabolism , Spumavirus/enzymology , Viral Proteins/chemistry , Viral Proteins/metabolism , Animals , Humans , Peptide Hydrolases/genetics , RNA-Directed DNA Polymerase/genetics , Retroviridae Infections/virology , Spumavirus/chemistry , Spumavirus/genetics , Viral Proteins/genetics
5.
PLoS One ; 14(3): e0212764, 2019.
Article in English | MEDLINE | ID: mdl-30865665

ABSTRACT

Eukaryotic DNA binding proteins must access genomic DNA that is packaged into chromatin in vivo. During a productive infection, retroviral integrases (IN) must similarly interact with chromatin to integrate the viral cDNA genome. Here we examine the role of nucleosome DNA unwrapping in the retroviral integrase search for a target site. These studies utilized PFV intasomes that are comprised of a tetramer of PFV IN with two oligomers mimicking the viral cDNA ends. Modified recombinant human histones were used to generate nucleosomes with increased unwrapping rates at different DNA regions. These modifications included the acetylmimetic H3(K56Q) and the chemically engineered H4(K77ac, K79ac). While transcription factors and DNA damage sensors may search nucleosome bound DNA during transient unwrapping, PFV intasome mediated integration appears to be unaffected by increased nucleosome unwrapping. These studies suggest PFV intasomes do not utilize nucleosome unwrapping to search nucleosome targets.


Subject(s)
DNA, Viral/metabolism , Genome, Viral , Nucleosomes/metabolism , Spumavirus/metabolism , Virus Integration/physiology , Cell-Free System/chemistry , Cell-Free System/metabolism , DNA, Viral/chemistry , Histones/chemistry , Histones/metabolism , Humans , Nucleosomes/chemistry , Spumavirus/chemistry
6.
J Vis Exp ; (133)2018 Mar 19.
Article in English | MEDLINE | ID: mdl-29608167

ABSTRACT

A defining feature and necessary step of the retrovirus life cycle is the integration of the viral genome into the host genome. All retroviruses encode an integrase (IN) enzyme that catalyzes the covalent joining of viral to host DNA, which is known as strand transfer. Integration may be modeled in vitro with recombinant retroviral IN and DNA oligomers mimicking the ends of the viral genome. In order to more closely recapitulate the integration reaction that occurs in vivo, integration complexes are assembled from recombinant IN and synthetic oligomers by dialysis in a reduced salt concentration buffer. The integration complex, called an intasome, may be purified by size exclusion chromatography. In the case of prototype foamy virus (PFV), the intasome is a tetramer of IN and two DNA oligomers and is readily separated from monomeric IN and free oligomer DNA. The integration efficiency of PFV intasomes may be assayed under a variety of experimental conditions to better understand the dynamics and mechanics of retroviral integration.


Subject(s)
DNA, Viral/genetics , Integrases/isolation & purification , Spumavirus/chemistry
7.
Virology ; 514: 50-56, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29128756

ABSTRACT

Integrase (IN) is an essential enzyme in retroviral life cycle. It mediates viral cDNA integration into host cellular DNA. Feline foamy virus (FFV) is a member of the Spumavirus subfamily of Retroviridae. Recently, its life cycle has been proposed to be different from other retroviruses. Despite this important finding, FFV IN is not understood clearly. Here, we constructed point mutations in FFV IN C-terminal domain (CTD) to obtain a clear understanding of its integration mechanism. Mutation of the amino acid residues in FFV IN CTD interacting with target DNA reduced both IN enzymatic activities in vitro and viral productions in infected cells. Especially, the mutants, R307 and K340, made viral DNA integration less efficient and allowed accumulation of more unintegrated viral DNA, thereby suppressing viral replication. Therefore, we suggest that the CTD residues interacting with the target DNA play a significant role in viral DNA integration and replication.


Subject(s)
Cat Diseases/virology , DNA, Viral/genetics , Integrases/chemistry , Integrases/metabolism , Retroviridae Infections/veterinary , Spumavirus/enzymology , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Integration , Amino Acid Motifs , Animals , Cats , Cell Line , DNA, Viral/metabolism , Integrases/genetics , Retroviridae Infections/virology , Spumavirus/chemistry , Spumavirus/genetics , Spumavirus/physiology , Viral Proteins/genetics , Virus Replication
8.
BMC Struct Biol ; 17(1): 3, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28372592

ABSTRACT

BACKGROUND: The Spumaretrovirinae (foamy viruses) and the Orthoretrovirinae (e.g. HIV) share many similarities both in genome structure and the sequences of the core viral encoded proteins, such as the aspartyl protease and reverse transcriptase. Similarity in the gag region of the genome is less obvious at the sequence level but has been illuminated by the recent solution of the foamy virus capsid (CA) structure. This revealed a clear structural similarity to the orthoretrovirus capsids but with marked differences that left uncertainty in the relationship between the two domains that comprise the structure. METHODS: We have applied protein structure comparison methods in order to try and resolve this ambiguous relationship. These included both the DALI method and the SAP method, with rigorous statistical tests applied to the results of both methods. For this, we employed collections of artificial fold 'decoys' (generated from the pair of native structures being compared) to provide a customised background distribution for each comparison, thus allowing significance levels to be estimated. RESULTS: We have shown that the relationship of the two domains conforms to a simple linear correspondence rather than a domain transposition. These similarities suggest that the origin of both viral capsids was a common ancestor with a double domain structure. In addition, we show that there is also a significant structural similarity between the amino and carboxy domains in both the foamy and ortho viruses. CONCLUSIONS: These results indicate that, as well as the duplication of the double domain capsid, there may have been an even more ancient gene-duplication that preceded the double domain structure. In addition, our structure comparison methodology demonstrates a general approach to problems where the components have a high intrinsic level of similarity.


Subject(s)
Capsid/chemistry , Evolution, Molecular , Gene Duplication , Retroviridae/chemistry , Spumavirus/chemistry , Amino Acid Sequence , Capsid/metabolism , Genome, Viral , Protein Conformation , Protein Domains , Retroviridae/physiology , Sequence Homology , Spumavirus/physiology , Virus Assembly
9.
Virol J ; 13: 117, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27363487

ABSTRACT

BACKGROUND: Bovine foamy virus (BFV) encodes the transactivator BTas, which enhances viral gene transcription by binding to the long terminal repeat promoter and the internal promoter. In this study, we investigated the different replication capacities of two similar BFV full-length DNA clones, pBS-BFV-Y and pBS-BFV-B. RESULTS: Here, functional analysis of several chimeric clones revealed a major role for the C-terminal region of the viral genome in causing this difference. Furthermore, BTas-B, which is located in this C-terminal region, exhibited a 20-fold higher transactivation activity than BTas-Y. Sequence alignment showed that these two sequences differ only at amino acid 108, with BTas-B containing N108 and BTas-Y containing D108 at this position. Results of mutagenesis studies demonstrated that residue N108 is important for BTas binding to viral promoters. In addition, the N108D mutation in pBS-BFV-B reduced the viral replication capacity by about 1.5-fold. CONCLUSIONS: Our results suggest that residue N108 is important for BTas binding to BFV promoters and has a major role in BFV replication. These findings not only advances our understanding of the transactivation mechanism of BTas, but they also highlight the importance of certain sequence polymorphisms in modulating the replication capacity of isolated BFV clones.


Subject(s)
Cattle Diseases/virology , Gene Expression Regulation, Viral , Promoter Regions, Genetic , Retroviridae Infections/veterinary , Spumavirus/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Motifs , Animals , Cattle , Retroviridae Infections/virology , Spumavirus/chemistry , Spumavirus/genetics , Trans-Activators/genetics , Viral Proteins/genetics
10.
PLoS Pathog ; 12(7): e1005721, 2016 07.
Article in English | MEDLINE | ID: mdl-27399201

ABSTRACT

Foamy viruses (FV) belong to the genus Spumavirus, which forms a distinct lineage in the Retroviridae family. Although the infection in natural hosts and zoonotic transmission to humans is asymptomatic, FVs can replicate well in human cells making it an attractive gene therapy vector candidate. Here we present cryo-electron microscopy and (cryo-)electron tomography ultrastructural data on purified prototype FV (PFV) and PFV infected cells. Mature PFV particles have a distinct morphology with a capsid of constant dimension as well as a less ordered shell of density between the capsid and the membrane likely formed by the Gag N-terminal domain and the cytoplasmic part of the Env leader peptide gp18LP. The viral membrane contains trimeric Env glycoproteins partly arranged in interlocked hexagonal assemblies. In situ 3D reconstruction by subtomogram averaging of wild type Env and of a Env gp48TM- gp80SU cleavage site mutant showed a similar spike architecture as well as stabilization of the hexagonal lattice by clear connections between lower densities of neighboring trimers. Cryo-EM was employed to obtain a 9 Å resolution map of the glycoprotein in its pre-fusion state, which revealed extensive trimer interactions by the receptor binding subunit gp80SU at the top of the spike and three central helices derived from the fusion protein subunit gp48TM. The lower part of Env, presumably composed of interlaced parts of gp48TM, gp80SU and gp18LP anchors the spike at the membrane. We propose that the gp48TM density continues into three central transmembrane helices, which interact with three outer transmembrane helices derived from gp18LP. Our ultrastructural data and 9 Å resolution glycoprotein structure provide important new insights into the molecular architecture of PFV and its distinct evolutionary relationship with other members of the Retroviridae.


Subject(s)
Gene Products, env/ultrastructure , Glycoproteins/ultrastructure , Spumavirus/ultrastructure , Blotting, Western , Cell Line , Cryoelectron Microscopy , Humans , Image Processing, Computer-Assisted , Protein Conformation , Spumavirus/chemistry , Transfection
11.
Acta Virol ; 60(2): 181-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27265468

ABSTRACT

The successful foamy viruses (FVs) infection includes at least two essential events, attachment to the cell surface and fusion of the viral envelope with the cell membrane. For the FVs, membrane fusion between virus and cell is mediated by envelope glycoprotein (Env) transmembrane (TM) subunit gp47. Compared with other retroviruses, FV TM subunit shares a similar but not identical structural characteristic. This paper focuses on in sillico analyses of all 15 available FV TM subunits gp47 based on their amino acid sequences. The hydrophobicity analysis revealed that the 15 FVs gp47 had two prominent hydrophobic regions, the N-terminal fusion peptide (FP) and the C-terminal region, which included a membrane-spanning domain (MSD) and a membrane proximal ectodomain region (MPER). In most FVs gp47, two heptad repeats, the coiled coils characterized by repetition of 7-amino acid-motif, were found to be correspondently located downstream of FP (named "N-HR") and the upstream of MPER (named "C-HR"). Furthermore, the solvent accessibility and secondary structure were predicted for all FVs gp47. These observations suggested that FVs gp47 possessed several fusion domains, which were necessary in the process of lipid membrane fusion between FVs and the target cells.


Subject(s)
Cell Membrane/virology , Retroviridae Infections/veterinary , Retroviridae Infections/virology , Spumavirus/genetics , Viral Envelope Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Cats/virology , Cattle , Haplorhini/virology , Humans , Molecular Sequence Data , Primates/virology , Protein Structure, Tertiary , Spumavirus/chemistry , Viral Envelope Proteins/genetics
12.
Nature ; 530(7590): 358-61, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26887496

ABSTRACT

Retroviral integrase catalyses the integration of viral DNA into host target DNA, which is an essential step in the life cycle of all retroviruses. Previous structural characterization of integrase-viral DNA complexes, or intasomes, from the spumavirus prototype foamy virus revealed a functional integrase tetramer, and it is generally believed that intasomes derived from other retroviral genera use tetrameric integrase. However, the intasomes of orthoretroviruses, which include all known pathogenic species, have not been characterized structurally. Here, using single-particle cryo-electron microscopy and X-ray crystallography, we determine an unexpected octameric integrase architecture for the intasome of the betaretrovirus mouse mammary tumour virus. The structure is composed of two core integrase dimers, which interact with the viral DNA ends and structurally mimic the integrase tetramer of prototype foamy virus, and two flanking integrase dimers that engage the core structure via their integrase carboxy-terminal domains. Contrary to the belief that tetrameric integrase components are sufficient to catalyse integration, the flanking integrase dimers were necessary for mouse mammary tumour virus integrase activity. The integrase octamer solves a conundrum for betaretroviruses as well as alpharetroviruses by providing critical carboxy-terminal domains to the intasome core that cannot be provided in cis because of evolutionarily restrictive catalytic core domain-carboxy-terminal domain linker regions. The octameric architecture of the intasome of mouse mammary tumour virus provides new insight into the structural basis of retroviral DNA integration.


Subject(s)
Cryoelectron Microscopy , DNA, Viral/metabolism , DNA, Viral/ultrastructure , Integrases/chemistry , Integrases/ultrastructure , Mammary Tumor Virus, Mouse/enzymology , Protein Multimerization , Catalytic Domain , Crystallography, X-Ray , DNA, Viral/chemistry , Integrases/metabolism , Mammary Tumor Virus, Mouse/chemistry , Mammary Tumor Virus, Mouse/genetics , Mammary Tumor Virus, Mouse/ultrastructure , Models, Molecular , Protein Structure, Quaternary , Spumavirus/chemistry , Spumavirus/enzymology , Virus Integration
13.
Nature ; 523(7560): 366-9, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26061770

ABSTRACT

Retroviral integration is catalysed by a tetramer of integrase (IN) assembled on viral DNA ends in a stable complex, known as the intasome. How the intasome interfaces with chromosomal DNA, which exists in the form of nucleosomal arrays, is currently unknown. Here we show that the prototype foamy virus (PFV) intasome is proficient at stable capture of nucleosomes as targets for integration. Single-particle cryo-electron microscopy reveals a multivalent intasome-nucleosome interface involving both gyres of nucleosomal DNA and one H2A-H2B heterodimer. While the histone octamer remains intact, the DNA is lifted from the surface of the H2A-H2B heterodimer to allow integration at strongly preferred superhelix location ±3.5 positions. Amino acid substitutions disrupting these contacts impinge on the ability of the intasome to engage nucleosomes in vitro and redistribute viral integration sites on the genomic scale. Our findings elucidate the molecular basis for nucleosome capture by the viral DNA recombination machinery and the underlying nucleosome plasticity that allows integration.


Subject(s)
Nucleosomes/chemistry , Nucleosomes/virology , Spumavirus/metabolism , Virus Integration , Amino Acid Substitution , Binding Sites/genetics , Cryoelectron Microscopy , DNA/genetics , DNA/metabolism , DNA/ultrastructure , Genome/genetics , Histones/chemistry , Histones/metabolism , Histones/ultrastructure , Integrases/metabolism , Models, Molecular , Nucleosomes/genetics , Nucleosomes/ultrastructure , Protein Multimerization , Recombination, Genetic , Spumavirus/chemistry , Spumavirus/genetics , Spumavirus/ultrastructure
14.
Curr Top Med Chem ; 15(1): 43-9, 2015.
Article in English | MEDLINE | ID: mdl-25579571

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) integrase (IN) is an essential enzyme in the viral replication cycle as it catalyzes the insertion of the reverse transcribed viral DNA into host chromosome. The structure of prototype foamy virus (PFV) IN has structural and functional homology with HIV-1 IN (no full-length structure available). In this study, we have used PFV IN-DNA complex as a surrogate model for HIV-1 IN-DNA complex to investigate the binding modes of N-methyl pyrimidones (NMPs) by QM-polarized ligand docking (QPLD), binding free energy calculations and molecular dynamics simulations. The O,O,O donor atom triad of NMPs show metal chelation with divalent Mg(2+) ions in the active site of PFV IN, in perfect agreement with the proposed mechanism of IN strand transfer inhibitors (INSTIs). The results also show that the benzyl group of compounds fit into a pocket to displace the 3'-terminal adenosine of viral DNA from the IN active site making it unavailable for the nucleophile to attack the target DNA in the strand transfer (ST) reaction. The halobenzyl moiety show hydrophobic interactions with conserved PFV IN Tyr212 and Pro214 residues, corresponding to HIV-1 IN Tyr143 and Pro145, respectively. Molecular dynamics (MD) simulations gave important insights into the structural and chemical basis involved in ST inhibition. Based on MD results, hydrogen bond with Tyr212, coordinate bonds with Mg(2+) ions, and hydrophobic interactions play an important role in the stabilization of compounds. Our results provide additional insight into the possible mechanism of action and binding mode of NMPs, and might have implications for rational design of specific HIV-1 INSTIs with improved affinity and selectivity.


Subject(s)
Antiviral Agents/chemistry , Chelating Agents/chemistry , DNA, Viral/antagonists & inhibitors , Integrases/chemistry , Pyrimidinones/chemistry , Spumavirus/chemistry , Viral Proteins/antagonists & inhibitors , Amino Acid Motifs , Antiviral Agents/chemical synthesis , Catalytic Domain , Chelating Agents/chemical synthesis , DNA, Viral/chemistry , HIV-1/chemistry , HIV-1/enzymology , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Kinetics , Magnesium/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Binding , Pyrimidinones/chemical synthesis , Sequence Homology, Amino Acid , Spumavirus/enzymology , Structural Homology, Protein , Thermodynamics , Viral Proteins/chemistry
15.
Bing Du Xue Bao ; 30(4): 346-52, 2014 Jul.
Article in Chinese | MEDLINE | ID: mdl-25272585

ABSTRACT

Bel1, a transactivator of prototype foamy virus (PFV), plays pivotal roles in the replication of PFV. Previous studies have shown that Bel1 bears a nuclear localization signal (NLS), but its amino acid sequence remains unclear and the corresponding importins have not been identified. In this report, we inserted various fragments of Bel1 into an EGFP-GST fusion protein and investigated their subcellular localization by fluorescence microscopy. We found that the 215PRQKRPR221 fragment could direct nuclear localization, which accords with the consensus sequence K(K/R)X(K/R) of monopartite NLS. Point mutation experiments revealed that K218, R219, and R221 are essential for the nuclear localization of Bel1. The results of the GST-pulldown showed that the Bel1 fragment with residues 215-223, which bears the NLS, interacts with KPNA1, KPNA6, and KPNA7. This result suggests that KPNA1, KPNA6, and KPNA7 maybe involved in Bel1 nuclear translocation.


Subject(s)
Cell Nucleus/virology , Nuclear Localization Signals/genetics , Retroviridae Infections/metabolism , Retroviridae Proteins/metabolism , Spumavirus/genetics , Trans-Activators/metabolism , alpha Karyopherins/metabolism , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Humans , Nuclear Localization Signals/metabolism , Protein Binding , Protein Transport , Retroviridae Infections/genetics , Retroviridae Infections/virology , Retroviridae Proteins/chemistry , Retroviridae Proteins/genetics , Spumavirus/chemistry , Spumavirus/physiology , Trans-Activators/chemistry , Trans-Activators/genetics , alpha Karyopherins/genetics
16.
Virology ; 449: 215-23, 2014 Jan 20.
Article in English | MEDLINE | ID: mdl-24418555

ABSTRACT

Prototype foamy virus encodes a transactivator called Bel1 that enhances viral gene transcription and is essential for PFV replication. Nuclear localization of Bel1 has been reported to rely on two proximal basic motifs R(199)H(200) and R(221)R(222)R(223) that likely function together as a bipartite nuclear localization signal. In this study, we report that mutating R(221)R(222)R(223), but not R(199)H(200), relocates Bel1 from the nucleus to the cytoplasm, suggesting an essential role for R(221)R(222)R(223) in the nuclear localization of Bel1. Although not affecting the nuclear localization of Bel1, mutating R(199)H(200) disables Bel1 from transactivating PFV promoters. Results of EMSA reveal that the R(199)H(200) residues are vital for the binding of Bel1 to viral promoter DNA. Moreover, mutating R(199)H(200) in Bel1 impairs PFV replication to a much greater extent than mutating R(221)R(222)R(223). Collectively, our findings suggest that R(199)H(200) directly participate in Bel1 binding to viral promoter DNA and are indispensible for Bel1 transactivation activity.


Subject(s)
Cell Nucleus/virology , Gene Expression Regulation, Viral , Promoter Regions, Genetic , Retroviridae Infections/virology , Retroviridae Proteins/chemistry , Retroviridae Proteins/metabolism , Spumavirus/metabolism , Terminal Repeat Sequences , Trans-Activators/chemistry , Trans-Activators/metabolism , Amino Acid Motifs , Amino Acid Sequence , Humans , Molecular Sequence Data , Protein Binding , Protein Transport , Retroviridae Proteins/genetics , Spumavirus/chemistry , Spumavirus/genetics , Trans-Activators/genetics , Transcriptional Activation
17.
J Mol Biol ; 425(12): 2133-46, 2013 Jun 26.
Article in English | MEDLINE | ID: mdl-23542006

ABSTRACT

Although there are structures of the different domains of human immunodeficiency virus type 1 (HIV-1) integrase (IN), there is no structure of the entire protein. The recently determined crystal structures of the prototype foamy virus (PFV) IN tetramer, in complexes with viral DNA, led to the generation of models of full-length HIV-1 IN. These models were generated, in part, by superimposing the structures of the domains of HIV-1 IN onto the structure of full-length PFV IN. We developed a model for HIV-1 IN-based solely on its sequence alignment with PFV IN-that differs in several ways from the previous models. Specifically, in our model, the junction between the catalytic core domain and C-terminal domain adopts a helix-loop-helix motif that is similar to the corresponding segment of PFV IN and differs from the crystal structures of these two HIV-1 IN domains. The alignment of residues in the C-terminal domain also differs from the previous models. Our model can be used to explain the phenotype of previously published HIV-1 IN mutants. We made additional mutants, and the behavior of these new mutants provides additional support for the model.


Subject(s)
HIV Integrase/chemistry , HIV Integrase/metabolism , Mutagenesis, Site-Directed , Mutation, Missense , Amino Acid Sequence , HIV Integrase/genetics , HIV-1/chemistry , HIV-1/genetics , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Conformation , Sequence Alignment , Spumavirus/chemistry , Spumavirus/genetics
18.
PLoS One ; 8(1): e54929, 2013.
Article in English | MEDLINE | ID: mdl-23365687

ABSTRACT

HIV-1 integrase (IN) is an important target in the development of drugs against the AIDS virus. Drug design based on the structure of IN was markedly hampered due to the lack of three-dimensional structure information of HIV-1 IN-viral DNA complex. The prototype foamy virus (PFV) IN has a highly functional and structural homology with HIV-1 IN. Recently, the X-ray crystal complex structure of PFV IN with its cognate viral DNA has been obtained. In this study, both Gaussian network model (GNM) and anisotropy network model (ANM) have been applied to comparatively investigate the motion modes of PFV DNA-free and DNA-bound IN. The results show that the motion mode of PFV IN has only a slight change after binding with DNA. The motion of this enzyme is in favor of association with DNA, and the binding ability is determined by its intrinsic structural topology. Molecular docking experiments were performed to gain the binding modes of a series of diketo acid (DKA) inhibitors with PFV IN obtained from ANM, from which the dependability of PFV IN-DNA used in the drug screen for strand transfer (ST) inhibitors was confirmed. It is also found that the functional groups of keto-enol, bis-diketo, tetrazole and azido play a key role in aiding the recognition of viral DNA, and thus finally increase the inhibition capability for the corresponding DKA inhibitor. Our study provides some theoretical information and helps to design anti-AIDS drug based on the structure of IN.


Subject(s)
DNA, Viral/chemistry , HIV Integrase Inhibitors/chemistry , HIV Integrase/chemistry , HIV-1/chemistry , Keto Acids/chemistry , Molecular Docking Simulation , Spumavirus/chemistry , Amino Acid Sequence , Binding Sites , Drug Design , Humans , Isoenzymes/chemistry , Molecular Sequence Data , Protein Binding , Protein Conformation , Sequence Alignment , Structural Homology, Protein , Structure-Activity Relationship , Substrate Specificity
19.
Cell Microbiol ; 15(2): 227-36, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23051660

ABSTRACT

Foamy viruses (FVs), a unique type of retroviruses, are characterized by several unusual features in their replication strategy. FVs, common to all non-human primates and several other species, display an extremely broad tropism in vitro. Basically, all mammalian cells and species examined, but also cells of amphibian or bird origin, are permissive to FV glycoprotein (Env)-mediated capsid release into the cytoplasm. The nature of the broadly expressed, and potentially evolutionary conserved, FV entry receptor molecule(s) is poorly characterized. Although recent data indicate that proteoglycans serve as an important factor for FV Env-mediated target cell attachment, additional uncharacterized molecules appear to be essential for the pH-dependent fusion of viral and cellular lipid membranes after endocytic uptake of virions. Furthermore, FVs show a very special assembly strategy. Unlike other retroviruses, the FV capsid precursor protein (Gag) undergoes only very limited proteolytic processing during assembly. This results in an immature morphology of capsids found in released FV virions. In addition, the FV Gag protein appears to lack a functional membrane-targeting signal. As a consequence, FVs utilize a specific interaction between capsid and cognate viral glycoprotein for initiation of thebudding process. Genetic fusion of heterologous targeting domains for plasma but not endosomal membranes to FV Gag enables glycoprotein-independent particle egress. However, this is at the expense of normal capsid morphogenesis and infectivity. The low-level Gag precursor processing and the requirement for a reversible, artificial Gag membrane association for effective pseudotyping of FV capsids by heterologous glycoproteins strongly suggest that FVs require a transient interaction of capsids with cellular membranes for viral replication. Under natural condition, this appears to be achieved by the lack of a membrane-targeting function of the FV Gag protein and the accomplishment of capsid membrane attachment through an unusual specific interaction with the cognate glycoprotein.


Subject(s)
Capsid/chemistry , Gene Products, gag/genetics , Spumavirus/chemistry , Virion/chemistry , Virus Assembly/physiology , Animals , Capsid/metabolism , Capsid/ultrastructure , Cell Membrane/chemistry , Cell Membrane/virology , Endocytosis , Gene Products, gag/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Membrane Fusion , Spumavirus/metabolism , Spumavirus/ultrastructure , Virion/metabolism , Virion/ultrastructure , Virus Internalization , Virus Replication
20.
Acta Virol ; 56(4): 283-91, 2012.
Article in English | MEDLINE | ID: mdl-23237084

ABSTRACT

One of the most fascinating findings in retrovirology is the construction of viral vectors based on foamy viruses (FVs) for gene therapy. The envelope glycoprotein (Env), one of the structural proteins of FV, is an important antigen in the immunoassays, as it is highly specific. To compare the characteristics of all 15 available FV Envs, the phylogenesis, hydrophobicity, modifications, and conserved motifs were analyzed based on the Env sequences. Meanwhile, the secondary structures of transmembrane (TM) domains of FV Envs were predicted. The results of phylogenetic analyses based on Envs indicated that the foamy viruses from different hosts could form three groups. The hydrophobicity analysis revealed that FV Envs had two prominent hydrophobic regions, which was similar to other retroviruses. Though the glycosylation, ubiquitination, and the secondary structures of TM domains of FV Envs were in line with other retroviruses, the roles were distinctly different. Interestingly, the analyses of conserved motifs suggested that FV Envs possessed several specific functional motifs.


Subject(s)
Retroviridae Infections/veterinary , Retroviridae Infections/virology , Spumavirus/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Phylogeny , Protein Conformation , Sequence Alignment , Spumavirus/chemistry , Spumavirus/classification , Viral Envelope Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...