Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Viruses ; 8(3): 80, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26985903

ABSTRACT

Clonal expansion of human T-lymphotropic virus type-1 (HTLV-1) infected cells in vivo is well documented. Unlike human immunodeficiency virus type 1 (HIV-1), HTLV-1 plasma RNA is sparse. The contribution of the "mitotic" spread of HTLV-1 compared with infectious spread of the virus to HTLV-1 viral burden in established infection is uncertain. Since extrachromosomal long terminal repeat (LTR) DNA circles are indicators of viral replication in HIV-1 carriers with undetectable plasma HIV RNA, we hypothesised that HTLV-1 LTR circles could indicate reverse transcriptase (RT) usage and infectious activity. 1LTR and 2LTR DNA circles were measured in HTLV-1 cell lines and peripheral blood mononuclear cells (PBMC) of asymptomatic carriers (ACs) and patients with HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) or adult T cell leukaemia/lymphoma (ATLL). 1LTR DNA circles were detected in 14/20 patients at a mean of 1.38/100 PBMC but did not differentiate disease status nor correlate with HTLV-1 DNA copies. 2LTR DNA circles were detected in 30/31 patients and at higher concentrations in patients with HTLV-1-associated diseases, independent of HTLV-1 DNA load. In an incident case the 2LTR DNA circle concentration increased 2.1 fold at the onset of HAM/TSP compared to baseline. Detectable and fluctuating levels of HTLV-1 DNA circles in patients indicate viral RT usage and virus replication. Our results indicate HTLV-1 viral replication capacity is maintained in chronic infection and may be associated with disease onset.


Subject(s)
Biomarkers/analysis , DNA, Circular/analysis , Human T-lymphotropic virus 1/physiology , Terminal Repeat Sequences , Virus Replication , Blood/virology , Carrier State/virology , Cells, Cultured , DNA, Circular/genetics , Female , HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Humans , Leukocytes, Mononuclear/virology , Male , RNA-Directed DNA Polymerase/metabolism
2.
Microbes Infect ; 15(6-7): 491-505, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23541980

ABSTRACT

Nonsense-mediated mRNA decay (NMD) is an essential and conserved cellular mRNA quality control mechanism. RNA signals to express viral genes from overlapping open reading frames potentially initiate NMD, nevertheless it is not clear whether viral RNAs are sensitive to NMD or if viruses have evolved mechanisms to evade NMD. Here we demonstrate that the genomic and full-length mRNAs of Human-T-cell Leukemia Virus type-I (HTLV-1), a retrovirus responsible for Adult T-cell Leukemia (ATL), are sensitive to NMD. They exhibit accelerated turnover in NMD-activated cells, while siRNA-mediated knockdown of NMD-master-regulator, UPF1, promotes enhanced stability of them. These effects on RNA stability were recapitulated by a reporter construct encoding the HTLV-1 translational frameshift signal of gag-pol. In agreement with the RNA stability, viral protein expression from the integrated provirus was inversely correlated with cellular NMD activity. We further demonstrated that the viral RNA-binding protein, Rex, approves the stability of viral RNA by inhibiting NMD. Significantly, Rex establishes a general block to NMD, as both NMD-responsive reporter transcripts and natural host-encoded NMD substrates were stabilized in the presence of Rex. Thus, we suggest that Rex not only stabilizes viral transcripts, but also perturbs cellular mRNA metabolism and host cell homeostasis via inhibition of NMD.


Subject(s)
Gene Products, rex/metabolism , Host-Pathogen Interactions , Human T-lymphotropic virus 1/physiology , Nonsense Mediated mRNA Decay , Virulence Factors/metabolism , Humans , RNA Stability , RNA, Viral/metabolism , Viral Proteins/biosynthesis
3.
J Immunol ; 187(1): 361-71, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21646298

ABSTRACT

Infection of human cells by human T cell leukemia virus type 1 (HTLV-1) is mediated by the viral envelope glycoproteins. The gp46 surface glycoprotein binds to cell surface receptors, including heparan sulfate proteoglycans, neuropilin 1, and glucose transporter 1, allowing the transmembrane glycoprotein to initiate fusion of the viral and cellular membranes. The envelope glycoproteins are recognized by neutralizing Abs and CTL following a protective immune response, and therefore, represent attractive components for a HTLV-1 vaccine. To begin to explore the immunological properties of potential envelope-based subunit vaccine candidates, we have used a soluble recombinant surface glycoprotein (gp46, SU) fused to the Fc region of human IgG (sRgp46-Fc) as an immunogen to vaccinate mice. The recombinant SU protein is highly immunogenic and induces high titer Ab responses, facilitating selection of hybridomas that secrete mAbs targeting SU. Many of these mAbs recognize envelope displayed on the surface of HTLV-1-infected cells and virions and several of the mAbs robustly antagonize envelope-mediated membrane fusion and neutralize pseudovirus infectivity. The most potently neutralizing mAbs recognize the N-terminal receptor-binding domain of SU, though there is considerable variation in neutralizing proficiency of the receptor-binding domain-targeted mAbs. By contrast, Abs targeting the C-terminal domain of SU tend to lack robust neutralizing activity. Importantly, we find that both neutralizing and poorly neutralizing Abs strongly stimulate neutrophil-mediated cytotoxic responses to HTLV-1-infected cells. Our data demonstrate that recombinant forms of SU possess immunological features that are of significant utility to subunit vaccine design.


Subject(s)
Antibodies, Neutralizing/toxicity , Deltaretrovirus Antibodies/toxicity , Gene Products, env/immunology , Human T-lymphotropic virus 1/immunology , Retroviridae Proteins, Oncogenic/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Virus Internalization , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/toxicity , Antibodies, Neutralizing/biosynthesis , Antibody-Dependent Cell Cytotoxicity/immunology , Deltaretrovirus Antibodies/biosynthesis , Gene Products, env/administration & dosage , Gene Products, env/genetics , HTLV-I Infections/immunology , HTLV-I Infections/prevention & control , HTLV-I Infections/virology , HeLa Cells , Human T-lymphotropic virus 1/pathogenicity , Humans , Jurkat Cells , Mice , Retroviridae Proteins, Oncogenic/administration & dosage , Retroviridae Proteins, Oncogenic/genetics , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use
4.
PLoS Pathog ; 7(2): e1001268, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-21304939

ABSTRACT

Refolding of viral class-1 membrane fusion proteins from a native state to a trimer-of-hairpins structure promotes entry of viruses into cells. Here we present the structure of the bovine leukaemia virus transmembrane glycoprotein (TM) and identify a group of asparagine residues at the membrane-distal end of the trimer-of-hairpins that is strikingly conserved among divergent viruses. These asparagines are not essential for surface display of pre-fusogenic envelope. Instead, substitution of these residues dramatically disrupts membrane fusion. Our data indicate that, through electrostatic interactions with a chloride ion, the asparagine residues promote assembly and profoundly stabilize the fusion-active structures that are required for viral envelope-mediated membrane fusion. Moreover, the BLV TM structure also reveals a charge-surrounded hydrophobic pocket on the central coiled coil and interactions with basic residues that cluster around this pocket are critical to membrane fusion and form a target for peptide inhibitors of envelope function. Charge-surrounded pockets and electrostatic interactions with small ions are common among class-1 fusion proteins, suggesting that small molecules that specifically target such motifs should prevent assembly of the trimer-of-hairpins and be of value as therapeutic inhibitors of viral entry.


Subject(s)
Ions/metabolism , Protein Folding , Retroviridae Proteins/chemistry , Retroviridae Proteins/physiology , Static Electricity , Amino Acid Sequence , Animals , Anti-Retroviral Agents/chemistry , Anti-Retroviral Agents/pharmacology , Catalytic Domain/drug effects , Cattle , Human T-lymphotropic virus 1/chemistry , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/metabolism , Humans , Hydrogen Bonding , Ions/chemistry , Leukemia Virus, Bovine/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology , Retroviridae/metabolism , Retroviridae/physiology , Retroviridae Proteins/metabolism , Surface Properties , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism
5.
Antimicrob Agents Chemother ; 53(2): 678-87, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19047654

ABSTRACT

The human T-cell lymphotropic virus type 1 (HTLV-1) is the cause of adult T-cell leukemia and inflammatory diseases including HTLV-1-associated myelopathy/tropical spastic paraparesis. HTLV-1 can be transmitted through sexual contact, mother-to-child transmission, and exposure to contaminated blood. Microbicides are agents that interfere with microbial infectivity at mucous membranes, and candidates are under development for use against sexually transmitted viruses such as human immunodeficiency virus type 1. We previously demonstrated that cell surface polyanionic heparan sulfate proteoglycans bind the HTLV-1 envelope glycoprotein surface subunit gp46, facilitating cell-cell and cell-free virus spread in vitro. We now show, using assays for Env-receptor binding inhibition, Env-induced cell-cell fusion, cell-cell virus spread, and pseudotype HTLV-1 infectivity, that the soluble polyanions PRO 2000 and dextran sulfate are potent inhibitors of HTLV-1 spread in vitro, with PRO 2000 being the more promising candidate. The results of these studies suggest that candidate topical microbicides may be of use in reducing HTLV-1 sexual transmission.


Subject(s)
Antiviral Agents/pharmacology , HTLV-I Infections/prevention & control , Human T-lymphotropic virus 1/drug effects , Naphthalenesulfonates/pharmacology , Polymers/pharmacology , Antiviral Agents/chemistry , Cell Fusion , Cell Survival/drug effects , Cells, Cultured , Gene Products, env/drug effects , HTLV-I Infections/transmission , HeLa Cells , Humans , Naphthalenesulfonates/chemistry , Peptides/pharmacology , Polymers/chemistry , Retroviridae Proteins, Oncogenic/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Virion/drug effects
6.
J Biol Chem ; 284(10): 6575-84, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19114713

ABSTRACT

A synthetic peptide based on the leash and alpha-helical region (LHR) of human T cell leukemia virus type 1 envelope is a potent inhibitor of viral entry into cells. The inhibitory peptide targets a triple-stranded coiled-coil motif of the fusion-active transmembrane glycoprotein and in a trans-dominant negative manner blocks resolution to the trimer-of-hairpins form. The LHR-mimetic is, therefore, functionally analogous to the C34/T20-type inhibitors of human immunodeficiency virus. Previous attempts to shorten the bioactive peptide produced peptides with severely attenuated activity. We now demonstrate that truncated peptides often suffer from poor solubility and impaired coiled coil binding properties, and we identify features that are critical to peptide function. In particular, the alpha-helical region of the LHR-mimetic is necessary but not sufficient for inhibitory activity. Moreover, two basic residues are crucial for coiled-coil binding and efficient inhibition of membrane fusion. By retaining these basic residues and a region of main chain peptide contacts with the coiled coil, a core LHR-mimetic was obtained that retains both the inhibitory properties and solubility profile of the parental peptide. Variants of the core peptide inhibit both membrane fusion and infection of cells by free viral particles, but unexpectedly, infection by virions was more susceptible to inhibition by low activity inhibitors than syncytium formation. The core inhibitor provides a valuable lead in the search for smaller more bio-available peptides and peptido-mimetics that possess anti-viral activity. Such molecules may be attractive candidates for therapeutic intervention in human T cell leukemia virus type 1 infections.


Subject(s)
Anti-Retroviral Agents/pharmacology , Biomimetic Materials/pharmacology , Human T-lymphotropic virus 1/metabolism , Peptides/chemistry , Peptides/pharmacology , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Amino Acid Motifs , Anti-Retroviral Agents/chemistry , Biomimetic Materials/chemistry , HTLV-I Infections/drug therapy , HTLV-I Infections/genetics , HTLV-I Infections/metabolism , HeLa Cells , Human T-lymphotropic virus 1/chemistry , Human T-lymphotropic virus 1/genetics , Humans , Peptides/genetics , Peptides/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
7.
Retrovirology ; 5: 70, 2008 Aug 04.
Article in English | MEDLINE | ID: mdl-18680566

ABSTRACT

BACKGROUND: Human T-cell leukaemia virus (HTLV-1) and bovine leukaemia virus (BLV) entry into cells is mediated by envelope glycoprotein catalyzed membrane fusion and is achieved by folding of the transmembrane glycoprotein (TM) from a rod-like pre-hairpin intermediate to a trimer-of-hairpins. For HTLV-1 and for several virus groups this process is sensitive to inhibition by peptides that mimic the C-terminal alpha-helical region of the trimer-of-hairpins. RESULTS: We now show that amino acids that are conserved between BLV and HTLV-1 TM tend to map to the hydrophobic groove of the central triple-stranded coiled coil and to the leash and C-terminal alpha-helical region (LHR) of the trimer-of-hairpins. Remarkably, despite this conservation, BLV envelope was profoundly resistant to inhibition by HTLV-1-derived LHR-mimetics. Conversely, a BLV LHR-mimetic peptide antagonized BLV envelope-mediated membrane fusion but failed to inhibit HTLV-1-induced fusion. Notably, conserved leucine residues are critical to the inhibitory activity of the BLV LHR-based peptides. Homology modeling indicated that hydrophobic residues in the BLV LHR likely make direct contact with a pocket at the membrane-proximal end of the core coiled-coil and disruption of these interactions severely impaired the activity of the BLV inhibitor. Finally, the structural predictions assisted the design of a more potent antagonist of BLV membrane fusion. CONCLUSION: A conserved region of the HTLV-1 and BLV coiled coil is a target for peptide inhibitors of envelope-mediated membrane fusion and HTLV-1 entry. Nevertheless, the LHR-based inhibitors are highly specific to the virus from which the peptide was derived. We provide a model structure for the BLV LHR and coiled coil, which will facilitate comparative analysis of leukaemia virus TM function and may provide information of value in the development of improved, therapeutically relevant, antagonists of HTLV-1 entry into cells.


Subject(s)
Antiviral Agents/pharmacology , Human T-lymphotropic virus 1/drug effects , Leukemia Virus, Bovine/drug effects , Peptides/pharmacology , Viral Envelope Proteins/chemistry , Virus Internalization/drug effects , Amino Acid Sequence , Amino Acid Substitution , Animals , Antiviral Agents/chemical synthesis , Conserved Sequence , HeLa Cells , Human T-lymphotropic virus 1/chemistry , Human T-lymphotropic virus 1/physiology , Humans , Leukemia Virus, Bovine/chemistry , Leukemia Virus, Bovine/physiology , Models, Molecular , Molecular Sequence Data , Peptides/chemical synthesis , Protein Structure, Tertiary , Sequence Alignment , Species Specificity , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
8.
J Virol ; 82(10): 4965-73, 2008 May.
Article in English | MEDLINE | ID: mdl-18305034

ABSTRACT

Viral fusion proteins mediate the entry of enveloped viral particles into cells by inducing fusion of the viral and target cell membranes. Activated fusion proteins undergo a cascade of conformational transitions and ultimately resolve into a compact trimer of hairpins or six-helix bundle structure, which pulls the interacting membranes together to promote lipid mixing. Significantly, synthetic peptides based on a C-terminal region of the trimer of hairpins are potent inhibitors of membrane fusion and viral entry, and such peptides are typically extensively alpha-helical. In contrast, an atypical peptide inhibitor of human T-cell leukemia virus (HTLV) includes alpha-helical and nonhelical leash segments. We demonstrate that both the C helix and C-terminal leash are critical to the inhibitory activities of these peptides. Amino acid side chains in the leash and C helix extend into deep hydrophobic pockets at the membrane-proximal end of the HTLV type 1 (HTLV-1) coiled coil, and these contacts are necessary for potent antagonism of membrane fusion. In addition, a single amino acid substitution within the inhibitory peptide improves peptide interaction with the core coiled coil and yields a peptide with enhanced potency. We suggest that the deep pockets on the coiled coil are ideal targets for small-molecule inhibitors of HTLV-1 entry into cells. Moreover, the extended nature of the HTLV-1-inhibitory peptide suggests that such peptides may be intrinsically amenable to modifications designed to improve inhibitory activity. Finally, we propose that leash-like mimetic peptides may be of value as entry inhibitors for other clinically important viral infections.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Human T-lymphotropic virus 1/growth & development , Peptides/chemistry , Peptides/pharmacology , Virus Internalization , Amino Acid Substitution , Cell Line , Giant Cells/virology , Humans , Models, Molecular , Peptides/chemical synthesis , Protein Structure, Quaternary , Protein Structure, Secondary , Sequence Deletion , Viral Fusion Proteins/antagonists & inhibitors
9.
J Biol Chem ; 282(50): 36724-35, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-17940280

ABSTRACT

The human T-cell leukemia virus transmembrane glycoprotein (TM) is a typical class 1 membrane fusion protein and a subunit of the viral envelope glycoprotein complex. Following activation, the TM undergoes conformational transitions from a native nonfusogenic state to a fusion-active pre-hairpin intermediate that subsequently resolves to a compact trimer-of-hairpins or six-helix bundle. Disruption of these structural transitions inhibits membrane fusion and viral entry and validates TM as an anti-viral and vaccine target. To investigate the immunological properties of fusion-active TM, we have generated a panel of monoclonal antibodies that recognize the coiled-coil domain of the pre-hairpin intermediate. Antibody reactivity is highly sensitive to the conformation of the coiled coil as binding is dramatically reduced or lost on denatured antigen. Moreover, a unique group of antibodies are 100-1000-fold more reactive with the coiled coil than the trimer-of-hairpins form of TM. The antibodies recognize virally expressed envelope, and significantly, some selectively bind to envelope only under conditions that promote membrane fusion. Most importantly, many of the antibodies potently block six-helix bundle formation in vitro. Nevertheless, viral envelope was remarkably resistant to neutralization by antibodies directed to the coiled coil. The data imply that the coiled coil of viral envelope is poorly exposed to antibody during membrane fusion. We suggest that resistance to neutralization by antibodies directed to fusion-associated structures is a common property of retroviral TM and perhaps of other viral class I fusion proteins. These observations have significant implications for vaccine design.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Human T-lymphotropic virus 1/immunology , Membrane Fusion/immunology , Viral Fusion Proteins/immunology , Virus Internalization , Animals , HeLa Cells , Human T-lymphotropic virus 1/genetics , Humans , Membrane Fusion/genetics , Mice , Protein Structure, Quaternary , Protein Structure, Secondary , Viral Fusion Proteins/genetics , Viral Vaccines/genetics , Viral Vaccines/immunology
10.
J Virol ; 81(11): 6019-31, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17376912

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) entry into cells is dependent upon the viral envelope glycoprotein-catalyzed fusion of the viral and cellular membranes. Following receptor activation of the envelope, the transmembrane glycoprotein (TM) is thought to undergo a series of fusogenic conformational transitions through a rod-like prehairpin intermediate to a compact trimer-of-hairpins structure. Importantly, synthetic peptides that interfere with the conformational changes of TM are potent inhibitors of membrane fusion and HTLV-1 entry, suggesting that TM is a valid target for antiviral therapy. To assess the utility of TM as a vaccine target and to explore further the function of TM in HTLV-1 pathogenesis, we have begun to examine the immunological properties of TM. Here we demonstrate that a recombinant trimer-of-hairpins form of the TM ectodomain is strongly immunogenic. Monoclonal antibodies raised against the TM immunogen specifically bind to trimeric forms of TM, including structures thought to be important for membrane fusion. Importantly, these antibodies recognize the envelope on virally infected cells but, surprisingly, fail to neutralize envelope-mediated membrane fusion or infection by pseudotyped viral particles. Our data imply that, even in the absence of overt membrane fusion, there are multiple forms of TM on virally infected cells and that some of these display fusion-associated structures. Finally, we demonstrate that many of the antibodies possess the ability to recruit complement to TM, suggesting that envelope-derived immunogens capable of eliciting a combination of neutralizing and complement-fixing antibodies would be of value as subunit vaccines for intervention in HTLV infections.


Subject(s)
Glycoproteins/chemistry , Glycoproteins/immunology , HTLV-I Antibodies/chemistry , Human T-lymphotropic virus 1/chemistry , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Amino Acid Motifs , Animals , Binding Sites, Antibody , Cell Line , Glycoproteins/metabolism , HTLV-I Antibodies/metabolism , HeLa Cells , Human T-lymphotropic virus 1/immunology , Human T-lymphotropic virus 1/pathogenicity , Humans , Membrane Fusion/immunology , Mice , Mice, Inbred BALB C , Neutralization Tests , Protein Conformation , Viral Envelope Proteins/metabolism
11.
J Gen Virol ; 88(Pt 2): 660-669, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17251585

ABSTRACT

Fusion of the viral and cellular membranes is a critical step in the infection of cells by the human T-cell leukemia virus type 1 (HTLV-1) and this process is catalysed by the viral envelope glycoproteins. During fusion, the transmembrane glycoprotein (TM) is thought to undergo a transition from a rod-like pre-hairpin conformation that is stabilized by a trimeric coiled coil to a more compact six-helix-bundle or trimer-of-hairpins structure. Importantly, synthetic peptides that interfere with the conformational changes of TM are potent inhibitors of membrane fusion and HTLV-1 entry, suggesting that the pre-hairpin motif is a valid target for antiviral therapy. Here, a stable, trimeric TM derivative that mimics the coiled-coil structure of fusion-active TM has been used to develop a plate-based assay to identify reagents that interfere with the formation of the six-helix bundle. The assay discriminates effectively between strong, weak and inactive peptide inhibitors of membrane fusion and has been used to identify a monoclonal antibody (mAb) that disrupts six-helix-bundle formation efficiently in vitro. The mAb is reactive with the C-helical region of TM, indicating that this region of TM is immunogenic. However, the mAb failed to neutralize HTLV-1 envelope-mediated membrane fusion, suggesting that, on native viral envelope, the epitope recognized by the mAb is obscured during fusion. This novel mAb will be of value in the immunological characterization of fusion-active structures of HTLV-1 TM. Moreover, the assay developed here will aid the search for therapeutic antibodies, peptides and small-molecule inhibitors targeting envelope and the HTLV-1 entry process.


Subject(s)
Antibodies, Monoclonal/pharmacology , Glycoproteins/metabolism , Human T-lymphotropic virus 1/metabolism , Membrane Fusion/drug effects , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Glycoproteins/chemistry , Glycoproteins/drug effects , HeLa Cells , Human T-lymphotropic virus 1/pathogenicity , Humans , Models, Molecular , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Protein Conformation , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/drug effects
12.
J Gen Virol ; 86(Pt 5): 1509-1513, 2005 May.
Article in English | MEDLINE | ID: mdl-15831964

ABSTRACT

Nuclear export of unspliced and incompletely spliced human immunodeficiency virus type 1 mRNA is mediated by the viral Rev protein. Rev binds to a structured RNA motif known as the Rev-response element (RRE), which is present in all Rev-dependent transcripts, and thereby promotes entry of the ribonucleoprotein complex into the nuclear-export pathway. Recent evidence indicates that a dimerization interface and a genetically separable "trimerization" interface are required for multimeric assembly of Rev on the RRE. In this report, the effect of mutations within the trimerization interface on Rev function was examined in mammalian cells. All trimerization-defective Rev molecules had profoundly compromised Rev function and a range of localization defects was observed. However, despite the potential for formation of heterodimers between functional and non-functional Rev proteins, trimerization-defective Rev mutants were unable to inhibit wild-type Rev function in a trans-dominant-negative manner.


Subject(s)
Gene Products, rev/genetics , Gene Products, rev/physiology , HIV-1/genetics , HIV-1/physiology , Active Transport, Cell Nucleus , Dimerization , Gene Products, rev/chemistry , HeLa Cells , Humans , Mutation , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , RNA, Messenger/metabolism , RNA, Messenger/physiology , RNA, Viral/metabolism , RNA, Viral/physiology , rev Gene Products, Human Immunodeficiency Virus
13.
J Virol ; 77(18): 9922-30, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12941902

ABSTRACT

The major receptors required for attachment and entry of the human T-cell leukemia virus type 1 (HTLV-1) remain to be identified. Here we demonstrate that a functional, soluble form of the HTLV-1 surface envelope glycoprotein, gp46, fused to an immunoglobulin Fc region (gp46-Fc) binds to heparan sulfate proteoglycans (HSPGs) on mammalian cells. Substantial binding of gp46-Fc to HeLa and Chinese hamster ovary (CHO) K1 cells that express HSPGs was detected, whereas binding to the sister CHO lines 2244, which expresses no HSPGs, and 2241, which expresses no glycosaminoglycans (GAGs), was much reduced. Enzymatic removal of HSPGs from HeLa and CHO K1 cells also reduced gp46-Fc binding. Dextran sulfate inhibited gp46-Fc binding to HSPG-expressing cells in a dose-dependent manner, whereas chondroitin sulfate was less effective. By contrast, dextran sulfate inhibited gp46-Fc binding to GAG-negative cells such as CHO 2244, CHO 2241, and Jurkat T cells weakly or not at all. Dextran sulfate inhibited HTLV-1 envelope glycoprotein (Env)-pseudotyped virus infection of permissive, HSPG-expressing target cells and blocked syncytium formation between HTLV-1 Env-expressing cells and HSPG-expressing permissive target cells. Finally, HSPG-expressing cells were more permissive for HTLV-1 Env-pseudotyped virus infection than HSPG-negative cells. Thus, similar to other pathogenic viruses, HTLV-1 may have evolved to use HSPGs as cellular attachment receptors to facilitate its propagation.


Subject(s)
Gene Products, env/metabolism , Heparan Sulfate Proteoglycans/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Animals , CHO Cells , Cricetinae , Dextran Sulfate/pharmacology , HeLa Cells , Human T-lymphotropic virus 1/physiology , Humans , Immunoglobulin Fc Fragments/metabolism , Membrane Fusion , Transcription, Genetic
14.
J Virol ; 77(5): 3281-90, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12584351

ABSTRACT

Retrovirus entry into cells is mediated by the viral envelope glycoproteins which, through a cascade of conformational changes, orchestrate fusion of the viral and cellular membranes. In the absence of membrane fusion, viral entry into the host cell cannot occur. For human T-cell leukemia virus type 1 (HTLV-1), synthetic peptides that mimic a carboxy-terminal region of the transmembrane glycoprotein (TM) ectodomain are potent inhibitors of membrane fusion and virus entry. Here, we demonstrate that this class of inhibitor targets a fusion-active structure of HTLV-1 envelope. In particular, the peptides bind specifically to a core coiled-coil domain of envelope, and peptide variants that fail to bind the coiled-coil lack inhibitory activity. Our data indicate that the inhibitory peptides likely function by disrupting the formation of a trimer-of-hairpins structure that is required for membrane fusion. Importantly, we also show that peptides exhibiting dramatically increased potency can be readily obtained. We suggest that peptides or peptide mimetics targeting the fusion-active structures of envelope may be of therapeutic value in the treatment of HTLV-1 infections.


Subject(s)
Antiviral Agents/pharmacology , Human T-lymphotropic virus 1/pathogenicity , Membrane Fusion/drug effects , Peptides/pharmacology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/drug effects , Amino Acid Sequence , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Cell Line , Giant Cells/drug effects , HeLa Cells , Human T-lymphotropic virus 1/drug effects , Humans , Models, Molecular , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/chemistry , Peptides/metabolism , Viral Envelope Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...