Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta ; 1848(2): 408-16, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25450808

ABSTRACT

The reovirus p10 fusion-associated small transmembrane (FAST) proteins are the smallest known membrane fusion proteins, and evolved specifically to mediate cell-cell, rather than virus-cell, membrane fusion. The 36-40-residue ectodomains of avian reovirus (ARV) and Nelson Bay reovirus (NBV) p10 contain an essential intramolecular disulfide bond required for both cell-cell fusion and lipid mixing between liposomes. To more clearly define the functional, biochemical and biophysical features of this novel fusion peptide, synthetic peptides representing the p10 ectodomains of ARV and NBV were analyzed by solution-state NMR spectroscopy, circular dichroism spectroscopy, fluorescence spectroscopy-based hydrophobicity analysis, and liposome binding and fusion assays. Results indicate that disulfide bond formation promotes exposure of hydrophobic residues, as indicated by bis-ANS binding and time-dependent peptide aggregation under aqueous conditions, implying the disulfide bond creates a small, geometrically constrained, cystine noose. Noose formation is required for peptide partitioning into liposome membranes and liposome lipid mixing, and electron microscopy revealed that liposome-liposome fusion occurs in the absence of liposome tubulation. In addition, p10 fusion peptide activity, but not membrane partitioning, is dependent on membrane cholesterol.


Subject(s)
Cholesterol/chemistry , Cystine/chemistry , Liposomes/chemistry , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Hydrophobic and Hydrophilic Interactions , Membrane Fusion , Molecular Sequence Data , Orthoreovirus/chemistry , Orthoreovirus, Avian/chemistry , Protein Structure, Secondary , Viral Fusion Proteins/chemical synthesis
2.
Biochim Biophys Acta ; 1848(2): 721-30, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25475644

ABSTRACT

Severe acute respiratory syndrome-associated coronavirus (SARS-CoV) poses a serious public health hazard. The S2 subunit of the S glycoprotein of SARS-CoV carries out fusion between the virus and the host cells. However, the exact mechanism of the cell fusion process is not well understood. Current model suggests that a conformational transition, upon receptor recognition, of the two heptad core regions of S2 may expose the hydrophobic fusogenic peptide or fusion peptide for membrane insertion. Three regions of the S2 subunit have been proposed to be involved in cell-cell fusion. The N-terminal fusion peptide (FP, residues 770-788), an internal fusion peptide (IFP, residues 873-888) and the pre-transmembrane region (PTM, residues 1185-1202) demonstrated interactions with model lipid membranes and potentially involved in the fusion process. Here, we have determined atomic resolution structures of these three peptides in DPC detergent micelles by solution NMR. FP assumes α-helical conformation with significant distortion at the central Gly residues; enabling a close packing among sidechains of aromatic residues including W, Y and F. The 3-D structure of PMT is characterized by a helix-loop-helix with extensive aromatic interactions within the helices. IFP adopts a rather straight α-helical conformation defined by packing among sidechains of aromatic and aliphatic residues. Paramagnetic spin labeled NMR has demonstrated surface localization of PMT whereas FP and IFP inserted into the micelles. Collectively, data presented in this study will aid in understanding fusion mechanism of SARS-CoV.


Subject(s)
Membrane Fusion/physiology , Severe acute respiratory syndrome-related coronavirus/chemistry , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Detergents/chemistry , Humans , Micelles , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Thermodynamics , Viral Fusion Proteins/chemical synthesis
3.
J Pept Sci ; 20(6): 398-405, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24652714

ABSTRACT

Members of the Chordopoxvirinae subfamily possess an unusual 11 protein entry-fusion complex (EFC) that is highly conserved and present in all species. The mode of action of this EFC is unknown, and the interactions of the constituent proteins are uncharacterised. Here, we present the chemical synthesis of membrane domain truncated linear constructs of two EFC proteins in orf virus, ORFV036 and 049. By using Boc solid phase peptide synthesis and native chemical ligation methods, these truncated proteins have been readily prepared in milligram quantities. These robust synthetic protocols allow ready access to these polypeptides to facilitate biological studies.


Subject(s)
Orf virus/chemistry , Viral Fusion Proteins/chemical synthesis , Molecular Structure , Peptides/chemical synthesis , Peptides/chemistry , Viral Fusion Proteins/chemistry
4.
Intervirology ; 53(6): 381-9, 2010.
Article in English | MEDLINE | ID: mdl-20606459

ABSTRACT

OBJECTIVES: The deciphering of intracellular signaling pathways that are activated by the interaction between viral fusion peptides and cellular membranes are important for the understanding of both viral replication strategies and host defense mechanisms. METHODS: Fusion peptides of several enveloped viruses belonging to different virus families were prepared by standard 9-fluorenylmethoxycarbonyl polyamine solid-phase synthesis and used to stimulate U937 cells in vitro to analyze the phosphorylation patterns of the signaling pathways (PKC, Src, Akt, and MAPK pathways). Immunoprecipitation and Western blotting were carried out by using phosphospecific antibodies. All samples were also assayed for the presence of IL-10 and IFN-beta by ELISA and activation of nuclear factors (AP-1 and NF-kappaB). RESULTS: We have demonstrated that hydrophobic domains of fusion proteins are able to induce several transduction pathways that lead to cytokine (IFN-beta and IL-10) production, an event that appears to be dependent on early activation of AP-1 and NF-kappaB. CONCLUSIONS: The results obtained on the signaling activity of fusion peptides from different viruses enabled us to shed some light on the complex mechanism of viral entry and more precisely we focused on the exact signaling event induced by hydrophobic domains characteristic of fusion peptides interacting with the cell membrane.


Subject(s)
Host-Pathogen Interactions , Interferon-beta/biosynthesis , Interleukin-10/biosynthesis , Signal Transduction , Transcriptional Activation , Viral Fusion Proteins/immunology , Blotting, Western , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Immunoprecipitation , Monocytes/immunology , Viral Fusion Proteins/chemical synthesis
5.
Chembiochem ; 9(5): 758-67, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18311743

ABSTRACT

The molecular mechanism of entry of herpes viruses requires a multicomponent fusion system. Virus entry and cell-cell fusion of Herpes simplex virus (HSV) requires four glycoproteins: gD, gB and gH/gL. The role of gB remained elusive until recently, when the crystal structure of HSV-1 gB became available. Glycoprotein B homologues represent the most highly conserved group of herpes virus glycoproteins; however, despite the high degree of sequence and structural conservation, differences in post-translational processing are observed for different members of this virus family. Whereas gB of HSV is not proteolytically processed after oligomerization, most other gB homologues are cleaved by a cellular protease into subunits that remain linked through disulfide bonds. Proteolytic cleavage is common for activation of many other viral fusion proteins, so it remains difficult to envisage a common role for different herpes virus gB structures in the fusion mechanism. We selected bovine herpes virus type 1 (BoHV-1) and herpes simplex virus type 1 (HSV-1) as representative viruses expressing cleaved and uncleaved gBs, and have screened their amino acid sequences for regions of highly interfacial hydrophobicity. Synthetic peptides corresponding to such regions were tested for their ability to induce the fusion of large unilamellar vesicles and to inhibit herpes virus infection. These results underline that several regions of the gB protein are involved in the mechanism of membrane interaction.


Subject(s)
Peptide Fragments/chemistry , Viral Envelope Proteins/chemistry , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Cattle , Cell Line , Chlorocebus aethiops , Herpesvirus 1, Bovine/chemistry , Herpesvirus 1, Bovine/drug effects , Herpesvirus 1, Human/drug effects , Hydrophobic and Hydrophilic Interactions , Membrane Fusion , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Vero Cells , Viral Fusion Proteins/chemical synthesis , Viral Fusion Proteins/pharmacology
6.
J Biol Chem ; 281(17): 11965-71, 2006 Apr 28.
Article in English | MEDLINE | ID: mdl-16507566

ABSTRACT

The envelope glycoprotein, termed the spike protein, of severe acute respiratory syndrome coronavirus (SARS-CoV) is known to mediate viral entry. Similar to other class 1 viral fusion proteins, the heptad repeat regions of SARS-CoV spike are thought to undergo conformational changes from a prefusion form to a subsequent post-fusion form that enables fusion of the viral and host membranes. Recently, the structure of a post-fusion form of SARS-CoV spike, which consists of isolated domains of heptad repeats 1 and 2 (HR1 and HR2), has been determined by x-ray crystallography. To date there is no structural information for the prefusion conformations of SARS-CoV HR1 and HR2. In this work we present the NMR structure of the HR2 domain (residues 1141-1193) from SARS-CoV (termed S2-HR2) in the presence of the co-solvent trifluoroethanol. We find that in the absence of HR1, S2-HR2 forms a coiled coil symmetric trimer with a complex molecular mass of 18 kDa. The S2-HR2 structure, which is the first example of the prefusion form of coronavirus envelope, supports the current model of viral membrane fusion and gives insight into the design of structure-based antagonists of SARS.


Subject(s)
Membrane Glycoproteins/chemical synthesis , Models, Molecular , Repetitive Sequences, Amino Acid , Severe acute respiratory syndrome-related coronavirus/chemistry , Viral Envelope Proteins/chemistry , Viral Fusion Proteins/chemical synthesis , Crystallography, X-Ray , Hemagglutinins, Viral , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Protein Conformation , Repetitive Sequences, Amino Acid/genetics , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/metabolism , Solubility , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
7.
Biochemistry ; 43(44): 14064-71, 2004 Nov 09.
Article in English | MEDLINE | ID: mdl-15518555

ABSTRACT

Severe acute respiratory syndrome coronavirus (SARS-CoV) is a newly emergent virus responsible for a worldwide epidemic in 2003. The coronavirus spike proteins belong to class I fusion proteins, and are characterized by the existence of two heptad repeat (HR) regions, HR1 and HR2. The HR1 region in coronaviruses is predicted to be considerably longer than that in other type I virus fusion proteins. Therefore the exact binding sequence to HR2 from the HR1 is not clear. In this study, we defined the region of HR1 that binds to HR2 by a series of biochemical and biophysical measures. Subsequently the defined HR1 (902-952) and HR2 (1145-1184) chains, which are different from previously defined binding regions, were linked together by a flexible linker to form a single-chain construct, 2-Helix. This protein was expressed in Escherichia coli and forms a typical six-helix coiled coil bundle. Highly conserved HR regions between mouse hepatitis virus (MHV) and SARS-CoV spike proteins suggest a similar three-dimensional structure for the two fusion cores. Here, we constructed a homology model for SARS coronavirus fusion core based on our biochemical analysis and determined the MHV fusion core structure. We also propose an important target site for fusion inhibitor design and several strategies, which have been successfully used in fusion inhibitor design for human immunodeficiency virus (HIV), for the treatment of SARS infection.


Subject(s)
Membrane Glycoproteins/chemical synthesis , Models, Molecular , Protein Engineering , Recombinant Fusion Proteins/chemical synthesis , Repetitive Sequences, Amino Acid , Severe acute respiratory syndrome-related coronavirus/chemistry , Severe acute respiratory syndrome-related coronavirus/genetics , Viral Envelope Proteins/chemical synthesis , Viral Fusion Proteins/chemical synthesis , Amino Acid Sequence , Binding Sites/genetics , Computer Simulation , Genetic Vectors/chemical synthesis , Membrane Glycoproteins/genetics , Molecular Sequence Data , Murine hepatitis virus/chemistry , Murine hepatitis virus/genetics , Murine hepatitis virus/pathogenicity , Peptide Fragments/biosynthesis , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Binding/genetics , Protein Engineering/methods , Protein Structure, Secondary/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repetitive Sequences, Amino Acid/genetics , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Solubility , Spike Glycoprotein, Coronavirus , Structural Homology, Protein , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
8.
Biochemistry ; 43(41): 13097-105, 2004 Oct 19.
Article in English | MEDLINE | ID: mdl-15476403

ABSTRACT

Human immunodeficiency virus type 1 integrase catalyzes the integration of proviral DNA into the infected cell genome, so it is an important potential target for antiviral drug design. In an attempt to search for peptides that specifically interact with integrase (IN) and inhibit its function, we used an in vitro selection procedure, the phage display technique. A phage display library of random heptapeptides was used to screen for potential peptide ligands of HIV-1 IN. Several phage clones were identified that specifically bound IN. Two of the selected peptides (FHNHGKQ and HLEHLLF) exhibited a high affinity for IN and were chemically synthesized. High affinity was confirmed by a displacement assay which showed that these two synthetic peptides were able to compete with the phages expressing the corresponding peptide. These agents were assayed on the in vitro IN activities. While none of them inhibited the 3'-processing reaction, the FHNHGKQ peptide was found to be an inhibitor of the strand transfer reaction. Despite its high affinity for IN, the HLEHLLF peptide selected and assayed under the same conditions was unable to inhibit this reaction. We showed that the FHNHGKQ peptide inhibits specifically the strand transfer activity by competing with the target DNA for binding to IN. These IN-binding agents could be used as a base for developing new anti-integrase compounds as well as for structural studies of the still unknown three-dimensional structure of the entire integrase molecule.


Subject(s)
Bacteriophage M13 , HIV Integrase Inhibitors/chemistry , HIV Integrase/chemistry , Oligopeptides/chemistry , Peptide Library , Transcription, Genetic , Virus Integration , Binding, Competitive , Capsid Proteins , Catalysis , Catalytic Domain , DNA-Binding Proteins/chemical synthesis , DNA-Binding Proteins/metabolism , Dimerization , HIV Integrase/metabolism , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/metabolism , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Protein Binding , Substrate Specificity , Viral Fusion Proteins/chemical synthesis , Viral Fusion Proteins/metabolism
9.
Biochemistry ; 42(12): 3527-35, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12653557

ABSTRACT

In the HIV-1 gp41 and other viral fusion proteins, the minimal oligomerization state is believed to be trimeric with three N-terminal fusion peptides inserting into the membrane in close proximity. Previous studies have demonstrated that the fusion peptide by itself serves as a useful model fusion system, at least to the hemifusion stage in which the viral and target cell lipids are mixed. In the present study, HIV-1 fusion peptides were chemically synthesized and cross-linked at their C-termini to form dimers or trimers. C-terminal trimerization is their likely topology in the fusogenic form of the intact gp41 protein. The fusogenicity of the peptides was then measured in an intervesicle lipid mixing assay, and the assay results were compared to those of the monomer. For monomer, dimer, and trimer at peptide strand/lipid mol ratios between 0.0050 and 0.010, the final extent of lipid mixing for the dimer and trimer was 2-3 times greater than for the monomer. These data suggest that the higher local concentration of peptide strands in the cross-linked peptides enhances fusogenicity and that oligomerization of the fusion peptide in gp41 may enhance the rate of viral/target cell membrane fusion. For gp41, this effect is in addition to the role of the trimeric coiled-coil structure in bringing about apposition of viral and target cell membranes. NMR measurements on the membrane-associated dimeric fusion peptide were consistent with an extended structure at Phe-8, which is the same as has been observed for the membrane-bound monomer in the same lipid composition.


Subject(s)
HIV Envelope Protein gp41/chemistry , HIV-1 , Amino Acid Sequence , Cross-Linking Reagents , Dimerization , HIV Envelope Protein gp41/physiology , Humans , In Vitro Techniques , Lipids , Membrane Fusion , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Quaternary , Viral Fusion Proteins/chemical synthesis , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/physiology
10.
Biochemistry ; 41(35): 10866-76, 2002 Sep 03.
Article in English | MEDLINE | ID: mdl-12196026

ABSTRACT

The fusion peptide of the HIV fusion protein gp41 is required for viral fusion and entry into a host cell, but it is unclear whether this 23-residue peptide can fuse model membranes. We address this question for model membrane vesicles in the presence and absence of aggregating concentrations of poly(ethylene glycol) (PEG). PEG had no effect on the physical properties of peptide bound to membranes or free in solution. We tested for fusion of both highly curved and uncurved PC/PE/SM/CH (35:30:15:20 mol %) vesicles and highly curved PC/PE/CH (1:1:1) vesicles treated with peptide in the presence and absence of PEG. Fusion was never observed in the absence of PEG, although high peptide concentrations led to aggregation and rupture, especially in unstable PC/PE/CH (1:1:1) vesicles. When 5 wt % PEG was present to aggregate vesicles, peptide enhanced the rate of lipid mixing between curved PC/PE/SM/CH vesicles in proportion to the peptide concentration, with this effect leveling off at peptide/lipid (P/L) ratios approximately 1:200. Peptide produced an even larger effect on the rate of contents mixing but inhibited contents mixing at P/L ratios >1:200. No fusion enhancement was seen with uncurved vesicles. The rate of fusion was also enhanced by the presence of hexadecane, and peptide-induced rate enhancement was not observed in the presence of hexadecane. We conclude that gp41 fusion peptide does not induce vesicle fusion at subrupturing concentrations but can enhance fusion between highly curved vesicles induced to fuse by PEG. The different effects of peptide on the rates of lipid mixing and fusion pore formation suggest that, while gp41 fusion peptide does affect hemifusion, it mainly affects pore formation.


Subject(s)
HIV Envelope Protein gp41/chemistry , Lipid Bilayers/chemistry , Membrane Fusion , Peptide Fragments/chemistry , Polyethylene Glycols/chemistry , Viral Fusion Proteins/chemistry , Alkanes/chemistry , Amino Acid Sequence , Animals , Cattle , Cholesterol/chemistry , HIV Envelope Protein gp41/pharmacology , Humans , Kinetics , Membrane Fusion/drug effects , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Sphingomyelins/chemistry , Viral Fusion Proteins/chemical synthesis , Viral Fusion Proteins/pharmacology
11.
Exp Cell Res ; 269(1): 117-29, 2001 Sep 10.
Article in English | MEDLINE | ID: mdl-11525645

ABSTRACT

Inefficient nuclear incorporation of foreign DNA remains a critical roadblock in the development of effective nonviral gene delivery systems. DNA delivered by traditional protocols remains within endosomal/lysosomal vesicles, or is rapidly degraded in the cytoplasm. Verotoxin I (VT), an AB(5) subunit toxin produced by enterohaemorrhagic Escherichia coli, binds to the cell surface glycolipid, globotriaosylceramide (Gb(3)) and is internalized into preendosomes. VT is then retrograde transported to the Golgi, endoplasmic reticulum (ER), and nucleus of highly VT-sensitive cells. We have utilized this nuclear targeting of VT to design a unique delivery system which transports exogenous DNA via vesicular traffic to the nucleus. The nontoxic VT binding subunit (VTB) was fused to the lambda Cro DNA-binding repressor, generating a 14-kDa VTB-Cro chimera. VTB-Cro binds specifically via the Cro domain to a 25-bp DNA fragment containing the consensus Cro operator. VTB-Cro demonstrates simultaneous specific binding to Gb(3). Treatment of Vero cells with fluorescent-labeled Cro operator DNA in the presence of VTB-Cro, results in DNA internalization to the Golgi, ER, and nucleus, whereas fluorescent DNA alone is incorporated poorly and randomly within the cytoplasm. VTB-Cro mediated nuclear DNA transport is prevented by brefeldin A, consistent with Golgi/ER intracellular routing. Pretreatment with filipin had no effect, indicating that caveoli are not involved. This novel VTB-Cro shuttle protein may find practical applications in the fields of intracellular targeting, gene delivery, and gene therapy.


Subject(s)
Cell Nucleus/drug effects , DNA-Binding Proteins , DNA/metabolism , Genetic Vectors/metabolism , Immunotoxins/metabolism , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Shiga Toxin 1/metabolism , Trihexosylceramides/metabolism , Animals , Antiviral Agents/pharmacology , Binding Sites/drug effects , Binding Sites/physiology , Brefeldin A/pharmacology , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Chlorocebus aethiops , DNA/drug effects , Endocytosis/drug effects , Endocytosis/physiology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Filipin/pharmacology , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Protein Synthesis Inhibitors/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Repressor Proteins/genetics , Transport Vesicles/drug effects , Transport Vesicles/metabolism , Vero Cells/cytology , Vero Cells/drug effects , Vero Cells/metabolism , Viral Fusion Proteins/chemical synthesis , Viral Fusion Proteins/metabolism , Viral Proteins , Viral Regulatory and Accessory Proteins
12.
J Mol Biol ; 296(5): 1353-65, 2000 Mar 10.
Article in English | MEDLINE | ID: mdl-10698638

ABSTRACT

Viral fusion proteins contain a highly hydrophobic segment, named the fusion peptide, which is thought to be responsible for the merging of the cellular and viral membranes. Paramyxoviruses are believed to contain a single fusion peptide at the N terminus of the F1 protein. However, here we identified an additional internal segment in the Sendai virus F1 protein (amino acids 214-226) highly homologous to the fusion peptides of HIV-1 and RSV. A synthetic peptide, which includes this region, was found to induce membrane fusion of large unilamellar vesicles, at concentrations where the known N-terminal fusion peptide is not effective. A scrambled peptide as well as several peptides from other regions of the F1 protein, which strongly bind to membranes, are not fusogenic. The functional and structural characterization of this active segment suggest that the F1 protein has an additional internal fusion peptide that could participate in the actual fusion event. The presence of homologous regions in other members of the same family suggests that the concerted action of two fusion peptides, one N-terminal and the other internal, is a general feature of paramyxoviruses.


Subject(s)
Membrane Fusion , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Respirovirus/chemistry , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Amino Acid Sequence , Circular Dichroism , Dimerization , Endopeptidase K/metabolism , Lipid Metabolism , Liposomes/chemistry , Liposomes/metabolism , Liposomes/ultrastructure , Microscopy, Electron , Models, Biological , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Protein Binding , Protein Structure, Secondary , Sequence Homology, Amino Acid , Spectrometry, Fluorescence , Structure-Activity Relationship , Thermodynamics , Vacuoles/chemistry , Vacuoles/metabolism , Vacuoles/ultrastructure , Viral Fusion Proteins/chemical synthesis
13.
Virology ; 223(1): 103-12, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8806544

ABSTRACT

It has been suggested that a conserved heptad repeat region in paramyxovirus fusion (F) proteins is essential for viral fusion activity (Buckland et al., 1992; Sergel et al., 1994; Reitter et al., 1995) We have studied synthetic peptides containing the heptad repeat regions derived from the F proteins of human parainfluenza virus type 2 (Pl2) and type 3 (Pl3) for their function as potential inhibitors of virus-induced cell fusion as well as their effects on spread of viral infection. Two peptides containing sequences of heptad repeat B, adjacent to the transmembrane domain of the F protein, were synthesized for both Pl2 and Pl3 F proteins. We observed that the longer peptides [34 amino acids (a.a.) for Pl2F or 35 a.a. for Pl3F] which extend from heptad repeat B to the transmembrane domain showed complete inhibition of cell fusion induced by the respective virus as well as by the vaccinia-expressed F and HN proteins. The 50% effective concentration to inhibit virus-induced cell fusion was 2.1 microM for Pl2 and 1.2 microM for Pl3. Moreover, the inhibitory effects of each peptide on virus-induced cell fusion were found to be virus type-specific. These peptides were found to also inhibit viral entry and to prevent plaque formation when mixed with the virus inoculum. Furthermore, the peptides caused a reduction in virus yield when assayed 48 hr after low m.o.i. infection and in the size of viral plaques when added to the overlay. Shorter peptides (21 a.a. for Pl2F or 24 a.a. for Pl3F) which correspond to the partial sequence of heptad repeat B for Pl2F and the entire heptad repeat B for Pl3F showed partial inhibition of Pl2- or Pl3-induced cell fusion. These results indicate that peptides containing the heptad repeat B sequence have the potential to inhibit virus-induced cell fusion, virus entry, and spread of virus infection.


Subject(s)
Respirovirus/physiology , Viral Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , Bacteriophage T7 , Cell Line , Chlorocebus aethiops , HN Protein/genetics , HN Protein/metabolism , HeLa Cells , Humans , Membrane Fusion , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Respirovirus/drug effects , Vaccinia virus/genetics , Vero Cells , Viral Fusion Proteins/chemical synthesis , Viral Fusion Proteins/genetics , Viral Fusion Proteins/physiology
14.
EMBO J ; 14(22): 5524-31, 1995 Nov 15.
Article in English | MEDLINE | ID: mdl-8521809

ABSTRACT

A series of peptides derived from three domains within the fusion protein of Sendai virus was synthesized and examined for their potential to inhibit the fusion of the virus with human red blood cells. These domains include the 'fusion peptide' and two heptad repeats, one adjacent to the fusion peptide (SV-163) and the other to the transmembrane domain (SV-473). Of all the peptides tested, only SV-473 was highly inhibitive. Using fluorescently-labelled peptides, the mechanism through which the SV-473 peptide inhibits the haemolytic activity of the virus was investigated. The results suggest that interactions of the active peptide with virion elements and lipid membranes are involved. Since it has recently been found that synthetic peptides corresponding to putative coiled-coil domains of the human immunodeficiency virus (HIV) type 1 transmembrane protein gp41 are potent inhibitors of HIV, we discuss the general property of virus-derived coiled-coil peptides as inhibitors of viral infection.


Subject(s)
Conserved Sequence , Membrane Fusion , Parainfluenza Virus 1, Human/drug effects , Peptides/pharmacology , Viral Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Chick Embryo , Erythrocyte Membrane/virology , Humans , Membrane Fusion/drug effects , Molecular Sequence Data , Parainfluenza Virus 1, Human/metabolism , Parainfluenza Virus 1, Human/pathogenicity , Peptides/chemical synthesis , Repetitive Sequences, Nucleic Acid , Viral Fusion Proteins/chemical synthesis
15.
Bioconjug Chem ; 6(1): 43-53, 1995.
Article in English | MEDLINE | ID: mdl-7711103

ABSTRACT

We have developed a strategy for the synthesis of novel oligodeoxynucleotide (ODN)-peptide conjugates on a scale suitable for the investigation of their potential as antisense inhibitors of gene expression. These conjugates have the 3'-terminus of the antisense oligodeoxynucleotide linked covalently to the N-terminus of a peptide. This strategy allows the preparation of conjugates containing a peptide segment designed to facilitate intracellular delivery of the antisense oligodeoxynucleotide as well as providing protection against 3'-exonuclease digestion. To illustrate the synthetic approach we describe the preparation of a series of conjugates comprising antisense oligonucleotides to human immunodeficiency virus type 1 (HIV) linked to fusion peptides derived from the HIV transmembrane glycoprotein gp41. The conjugates were prepared by the total synthesis method, in which the peptide is assembled first by the N-(fluorenylmethoxycarbonyl) (Fmoc) solid-phase methodology. This is followed by derivatization of the amino terminus by reaction with an alpha,omega-hydroxycarboxylic acid derivative which converts the terminus to a protected aliphatic hydroxy group on which standard solid phase DNA synthesis by the phosphoramidite method is performed. The purified conjugates were characterized extensively by several analytical techniques including ion spray mass spectrometry. Thermal denaturation studies showed that the interaction of the ODN-peptide conjugate with its complementary strand was similar to that of unmodified oligonucleotides. Preparation by the total synthesis method gave the purified conjugate with overall yields in the range of 6-14%.


Subject(s)
HIV Envelope Protein gp41/chemistry , Oligonucleotides, Antisense/chemistry , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Base Sequence , Humans , Mass Spectrometry , Molecular Sequence Data , Nucleic Acid Denaturation , Oligonucleotides, Antisense/chemical synthesis , Protein Denaturation , Temperature , Viral Fusion Proteins/chemical synthesis
16.
Biochemistry ; 30(46): 11173-80, 1991 Nov 19.
Article in English | MEDLINE | ID: mdl-1932037

ABSTRACT

In the infectious entry pathway of influenza virus, the low pH of the endosomal compartment induces an irreversible conformational change in influenza virus hemagglutinin, leading to fusion of viral and endosomal membranes. In the current report, we characterized the low-pH-induced activation of hemagglutinin of influenza strain X31 by studying its interaction with a lipid monolayer. The surface activities of virions, of isolated hemagglutinins and its proteolytic fragments, and of a synthetic peptide mimicking the amino terminus of subunit 2 of hemagglutinin are compared. The data indicate that the surface activity of both virions and isolated hemagglutinin develop as a result of the low-pH-induced conformational change in hemagglutinin. The surface activity of isolated hemagglutinin is mainly caused by penetration into the lipid monolayer of protein domains other than the amino terminus of subunit 2 of hemagglutinin; domains in subunit 1 may be involved. The surface activity of virions appears to be a secondary effect of the conformational change and is explained by assuming a net transfer of viral lipids to the lipid monolayer.


Subject(s)
Hemagglutinins, Viral/isolation & purification , Influenza A virus/chemistry , Membrane Fusion , Membrane Lipids/chemistry , Viral Fusion Proteins/chemistry , Virion/chemistry , Amino Acid Sequence , Hemagglutinin Glycoproteins, Influenza Virus , Hemagglutinins, Viral/chemistry , Hydrogen-Ion Concentration , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Surface Properties , Viral Fusion Proteins/chemical synthesis
17.
J Gen Virol ; 72 ( Pt 6): 1293-9, 1991 Jun.
Article in English | MEDLINE | ID: mdl-1710646

ABSTRACT

The immunogenicity of chimeric peptides produced by collinear synthesis to contain both T and B cell epitopes from the fusion protein and the haemagglutinin of measles virus was studied in mice. The T cell epitope used was from the fusion protein (residues 288 to 302), which has been shown to be promiscuous in its binding to mouse major histocompatibility complex molecules. This epitope was coupled by (i) a glycine-glycine spacer to a B cell epitope from the fusion protein (residues 404 to 414) and (ii) either its amino or carboxy terminus to a neutralizing antibody epitope from the haemagglutinin (residues 188 to 199). The results obtained show that such chimeric peptides can indeed function as complete immunogens in a range of mouse strains of different H-2 haplotype, and can induce the production of antibodies which bind to the fusion protein and to measles virus. Furthermore, it was shown that the orientation of the T cell epitope with respect to the B cell epitope had a significant effect upon the immunogenicity and antigenic specificity of the chimera. This work gives further support to the concept of rationally designed synthetic peptide vaccines.


Subject(s)
Antibody Formation , B-Lymphocytes/immunology , Epitopes/analysis , Hemagglutinins, Viral/immunology , Measles virus/immunology , T-Lymphocytes/immunology , Viral Fusion Proteins/immunology , Amino Acid Sequence , Animals , Chimera , Hemagglutinins, Viral/chemical synthesis , Hemagglutinins, Viral/genetics , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Mice, Inbred CBA , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/immunology , Viral Fusion Proteins/chemical synthesis , Viral Fusion Proteins/genetics
18.
Biochim Biophys Acta ; 1065(1): 49-53, 1991 May 31.
Article in English | MEDLINE | ID: mdl-1646029

ABSTRACT

31P nuclear magnetic resonance spectroscopy (31P-NMR) was used to study phospholipid organization in hydrated preparations of N-methyl dioleoylphosphatidylethanolamine and a 'fusion peptide' with the sequence: FAGV-VLAGAALGVAAAAQI, which corresponds to the amino terminus of the F1 subunit of the membrane fusion protein of measles virus. These amino acids are believed to mediate syncytia formation, host-cell penetration and hemolysis by infectious virus. The presence of the peptide at 0.5 mole percent significantly facilitated the formation of isotropic 31P resonances. The effects at 1 mole percent peptide were substantially enhanced over the effects observed at 0.5 mole percent, leading to a decrease in the onset temperature of the formation of the isotropic 31P-NMR resonances by about 30 degrees C. The formation of such isotropic 31P-NMR resonances has been previously associated with an increased rate of fusion of large unilamellar vesicles composed of N-methyl dioleoylphosphatidylethanolamine. Enhanced fusion of octadecyl rhodamine-labelled Sendai virus with N-methyl dioleoylphosphatidylethanolamine large unilamellar vesicles was observed when the 'fusion peptide' was incorporated into the large unilamellar vesicles.


Subject(s)
Liposomes , Measles virus/physiology , Membrane Fusion/drug effects , Parainfluenza Virus 1, Human/physiology , Phosphatidylethanolamines/chemistry , Viral Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , Chick Embryo , Magnetic Resonance Spectroscopy/methods , Molecular Sequence Data , Parainfluenza Virus 1, Human/drug effects , Thermodynamics , Viral Fusion Proteins/chemical synthesis
19.
Arch Virol ; 117(1-2): 59-71, 1991.
Article in English | MEDLINE | ID: mdl-1706591

ABSTRACT

We have previously located a major neutralization site of the fusion protein of respiratory syncytial virus (RSV) in the polypeptide region extending from amino acids Ile221 to Glu232. In this report, 8 peptides corresponding to the six major hydrophilic regions of the F1 subunit were selected to analyse their immunogenic and protective capacities as well as their ability to block the high neutralization activities of 4 monoclonal antibodies (MAbs). Only 5 of the 8 peptides tested induced specific antibodies while all induced an in vitro interleukin-2 response of splenocytes from immunized mice. Peptide 3 (Ile221-Phe237) was able to elicit neutralizing antibodies, confirming our previous hypothesis concerning the location of a neutralization site. However, immunization with the latter did not induce significant reduction of virus in lungs of BALB/c mice upon challenge, probably due to an inadequate level of circulating neutralizing antibodies. Interestingly, peptides 2 (Asn216-Glu232), 3 (Ile221-Phe237), and 5 (Ser275-Ile288) blocked in vitro neutralization by four different F1 specific MAbs. A hypothesis is proposed to explain these results.


Subject(s)
Antigens, Viral/immunology , B-Lymphocytes/immunology , HN Protein , Respiratory Syncytial Viruses , Respirovirus Infections/prevention & control , T-Lymphocytes/immunology , Viral Proteins , Viral Vaccines/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Antibody Specificity , Antigens, Viral/chemical synthesis , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Interleukin-2/biosynthesis , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neutralization Tests , Respiratory Syncytial Viruses/immunology , Spleen/cytology , Spleen/immunology , Vaccines, Synthetic/immunology , Viral Envelope Proteins , Viral Fusion Proteins/chemical synthesis , Viral Fusion Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL