Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
EMBO Rep ; 10(10): 1132-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19713960

ABSTRACT

Several studies have shown that ribosomal proteins (RPs) are important mediators of p53 activation in response to nucleolar disruption; however, the pathways that control this signalling function of RPs are currently unknown. We have recently shown that RPs are targets for the ubiquitin-like molecule NEDD8, and that NEDDylation protects RPs from destabilization. Here, we identify NEDD8 as a crucial regulator of L11 RP signalling to p53. A decrease in L11 NEDDylation during nucleolar stress causes relocalization of L11 from the nucleolus to the nucleoplasm. This not only provides the signal for p53 activation, but also makes L11 susceptible to degradation. Mouse double minute 2 (MDM2) -mediated NEDDylation protects L11 from degradation and this is required for p53 stabilization during nucleolar stress. By controlling the correct localization and stability of L11, NEDD8 acts as a crucial, new regulator of nucleolar signalling to p53.


Subject(s)
Cell Nucleolus/metabolism , Ribosomal Proteins/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Ubiquitins/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Humans , Mice , NEDD8 Protein , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Ubiquitins/genetics
3.
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
4.
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
5.
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
6.
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
7.
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
SELECTION OF CITATIONS
SEARCH DETAIL