Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(2): e0263853, 2022.
Article in English | MEDLINE | ID: mdl-35213606

ABSTRACT

The Semliki Forest Virus (SFV) is an RNA virus with a positive-strand that belongs to the Togaviridae family's Alphavirus genus. An epidemic was observed among French troops stationed in the Central African Republic, most likely caused by the SFV virus. The two transmembrane proteins El and E2 and the peripheral protein E3 make up the viral spike protein. The virus binds to the host cell and is internalized via endocytosis; endosome acidification causes the E1/E2 heterodimer to dissociate and the E1 subunits to trimerize. Lupenone was evaluated against the E1 spike protein of SFV in this study based on state-of-the-art cheminformatics approaches, including molecular docking, molecular dynamics simulation, and binding free energy calculation. The molecular docking study envisaged major interactions of Lupenone with binding cavity residues involved non-bonded van der Waal's and Pi-alkyl interactions. Molecular dynamic simulation of a time scale 200 ns corroborated interaction pattern with molecular docking studies between Lupenone and E1 spike protein. Nevertheless, Lupenone intearcation with the E1 spike protein conforming into a stable complex substantiated by free energy landscape (FEL), PCA analysis. Free energy decomposition of the binding cavity resdiues of E1 spike protein also ensured the efficient non-bonded van der Waal's interaction contributing most energy to interact with the Lupenone. Therefore, Lupenone interacted strongly at the active site conforming into higher structural stability throughout the dynamic evolution of the complex. Thus, this study perhaps comprehend the efficiency of Lupenone as lead molecule against SFV E1 spike protein for future therapeutic purpose.


Subject(s)
Molecular Docking Simulation , Semliki forest virus/chemistry , Triterpenes/chemistry , Viral Fusion Proteins/chemistry
2.
Biochim Biophys Acta ; 1858(11): 2671-2680, 2016 11.
Article in English | MEDLINE | ID: mdl-27425030

ABSTRACT

Semliki Forest virus (SFV) is a well-characterized alphavirus that infects cells via endocytosis and an acid-triggered fusion step using class II fusion proteins. Membrane fusion is mediated by the viral spike protein, a heterotrimer of two transmembrane subunits, E1 and E2, and a peripheral protein, E3. Sequence analysis of the E1 ectodomain of a number of alphaviruses demonstrated the presence of a highly conserved hydrophobic domain on the E1 ectodomain. This sequence was proposed to be the fusion peptide of SFV and is believed to be the domain of E1 that interacts with the target membrane and triggers fusion. Here, we investigate the structure and the interaction with lipid membrane models of 76YQCKVYTGVYPFMWGGAYCFC96 sequence from SFV, named SFV21, using optical method (ellipsometry) and vibrational spectroscopiy approaches (Polarization Modulation infra-Red Reflection Absorption Spectroscopy, PMIRRAS, and polarized ATR-FTIR). We demonstrate a structural flexibility of SFV21 sequence whether the lateral pressure and the lipid environment. In a lipid environment that mimics eukaryotic cell membranes, a conformational transition from an α-helix to a ß-sheet is induced in the presence of lipid by increasing the peptide to lipid ratio, which leads to important perturbations in the membrane organisation.


Subject(s)
Cholesterol/chemistry , Dimyristoylphosphatidylcholine/chemistry , Membrane Lipids/chemistry , Semliki forest virus/chemistry , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Membrane Fusion , Peptides/chemistry , Phosphatidylethanolamines/chemistry , Phosphatidylglycerols/chemistry , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Multimerization , Unilamellar Liposomes/chemistry
3.
J Virol ; 89(15): 7536-49, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25972559

ABSTRACT

UNLABELLED: Semliki Forest virus (SFV) provides a well-characterized model system to study the pathogenesis of virus encephalitis. Several studies have used virus derived from the molecular clone SFV4. SFV4 virus does not have the same phenotype as the closely related L10 or the prototype virus from which its molecular clone was derived. In mice, L10 generates a high-titer plasma viremia, is efficiently neuroinvasive, and produces a fatal panencephalitis, whereas low-dose SFV4 produces a low-titer viremia, rarely enters the brain, and generally is avirulent. To determine the genetic differences responsible, the consensus sequence of L10 was determined and compared to that of SFV4. Of the 12 nucleotide differences, six were nonsynonymous; these were engineered into a new molecular clone, termed SFV6. The derived virus, SFV6, generated a high-titer viremia and was efficiently neuroinvasive and virulent. The phenotypic difference mapped to a single amino acid residue at position 162 in the E2 envelope glycoprotein (lysine in SFV4, glutamic acid in SFV6). Analysis of the L10 virus showed it contained different plaque phenotypes which differed in virulence. A lysine at E2 247 conferred a small-plaque avirulent phenotype and glutamic acid a large-plaque virulent phenotype. Viruses with a positively charged lysine at E2 162 or 247 were more reliant on glycosaminoglycans (GAGs) to enter cells and were selected for by passage in BHK-21 cells. Interestingly, viruses with the greatest reliance on binding to GAGs replicated to higher titers in the brain and more efficiently crossed an in vitro blood-brain barrier (BBB). IMPORTANCE: Virus encephalitis is a major disease, and alphaviruses, as highlighted by the recent epidemic of chikungunya virus (CHIKV), are medically important pathogens. In addition, alphaviruses provide well-studied experimental systems with extensive literature, many tools, and easy genetic modification. In this study, we elucidate the genetic basis for the difference in phenotype between SFV4 and the virus stocks from which it was derived and correct this by engineering a new molecular clone. We then use this clone in one comprehensive study to demonstrate that positively charged amino acid residues on the surface of the E2 glycoprotein, mediated by binding to GAGs, determine selective advantage and plaque size in BHK-21 cells, level of viremia in mice, ability to cross an artificial BBB, efficiency of replication in the brain, and virulence. Together with studies on Sindbis virus (SINV), this study provides an important advance in understanding alphavirus, and probably other virus, encephalitis.


Subject(s)
Alphavirus Infections/virology , Blood-Brain Barrier/virology , Encephalitis/virology , Semliki forest virus/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Brain/virology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Semliki forest virus/chemistry , Semliki forest virus/genetics , Semliki forest virus/pathogenicity , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viremia/virology , Virulence
4.
J Proteome Res ; 13(3): 1702-12, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24467287

ABSTRACT

Cross-species viral transmission subjects parent and progeny alphaviruses to differential post-translational processing of viral envelope glycoproteins. Alphavirus biogenesis has been extensively studied, and the Semliki Forest virus E1 and E2 glycoproteins have been shown to exhibit differing degrees of processing of N-linked glycans. However the composition of these glycans, including that arising from different host cells, has not been determined. Here we determined the chemical composition of the glycans from the prototypic alphavirus, Semliki Forest virus, propagated in both arthropod and rodent cell lines, by using ion-mobility mass spectrometry and collision-induced dissociation analysis. We observe that both the membrane-proximal E1 fusion glycoprotein and the protruding E2 attachment glycoprotein display heterogeneous glycosylation that contains N-linked glycans exhibiting both limited and extensive processing. However, E1 contained predominantly highly processed glycans dependent on the host cell, with rodent and mosquito-derived E1 exhibiting complex-type and paucimannose-type glycosylation, respectively. In contrast, the protruding E2 attachment glycoprotein primarily contained conserved under-processed oligomannose-type structures when produced in both rodent and mosquito cell lines. It is likely that glycan processing of E2 is structurally restricted by steric-hindrance imposed by local viral protein structure. This contrasts E1, which presents glycans characteristic of the host cell and is accessible to enzymes. We integrated our findings with previous cryo-electron microscopy and crystallographic analyses to produce a detailed model of the glycosylated mature virion surface. Taken together, these data reveal the degree to which virally encoded protein structure and cellular processing enzymes shape the virion glycome during interspecies transmission of Semliki Forest virus.


Subject(s)
Membrane Glycoproteins/chemistry , Polysaccharides/analysis , Protein Processing, Post-Translational , Semliki forest virus/chemistry , Viral Envelope Proteins/chemistry , Virion/chemistry , Aedes , Animals , Carbohydrate Sequence , Cell Line , Cricetinae , Glycomics , Glycosylation , Host Specificity , Mass Spectrometry/methods , Membrane Glycoproteins/metabolism , Models, Molecular , Molecular Sequence Data , Polysaccharides/chemistry , Semliki forest virus/metabolism , Viral Envelope Proteins/metabolism , Virion/metabolism
5.
J Virol ; 86(7): 3588-94, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22258261

ABSTRACT

The enveloped alphaviruses infect cells via a low-pH-triggered membrane fusion reaction mediated by the viral transmembrane protein E1. During fusion, E1 inserts into the target membrane and refolds to a hairpin-like postfusion conformation in which domain III (DIII) and the juxtamembrane stem pack against a central core trimer. Although zinc has previously been shown to cause a striking block in alphavirus fusion with liposome target membranes, the mechanism of zinc's effect on the E1 fusion protein is not understood. Here we developed a cell culture system to study zinc inhibition of fusion and infection of the alphavirus Semliki Forest virus (SFV). Inclusion of 2 mM ZnCl(2) in the pH 5.75 fusion buffer caused a decrease of ∼5 logs in SFV fusion at the plasma membrane. Fusion was also inhibited by nickel, a chemically related transition metal. Selection for SFV zinc resistance identified a key histidine residue, H333 on E1 DIII, while other conserved E1 histidine residues were not involved. An H333N mutation conferred resistance to both zinc and nickel, with properties in keeping with the known pH-dependent chelation of these metals by histidine. Biochemical studies demonstrated that zinc strongly inhibits formation of the postfusion E1 trimer in wild-type SFV but not in an H333 mutant. Together our results suggest that zinc acts by blocking the fold-back of DIII via its interaction with H333.


Subject(s)
Alphavirus Infections/virology , Down-Regulation/drug effects , Semliki forest virus/drug effects , Semliki forest virus/physiology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Virus Internalization/drug effects , Zinc/pharmacology , Animals , Cell Line , Cricetinae , Humans , Protein Structure, Tertiary , Semliki forest virus/chemistry , Semliki forest virus/genetics , Viral Fusion Proteins/genetics
6.
J Virol ; 86(1): 553-65, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22031949

ABSTRACT

Semliki Forest virus (SFV) is a member of the Alphavirus genus, which produces its replicase proteins in the form of a nonstructural (ns) polyprotein precursor P1234. The maturation of the replicase occurs in a temporally controlled manner by protease activity of nsP2. The template preference and enzymatic capabilities of the alphaviral replication complex have a very important connection with its composition, which is irreversibly altered by proteolysis. The final cleavage of the 2/3 site in the ns polyprotein apparently leads to significant rearrangements within the replication complex and thus denotes the "point of no return" for viral replication progression. Numerous studies have devised rules for when and how ns protease acts, but how the alphaviral 2/3 site is recognized remained largely unexplained. In contrast to the other two cleavage sites within the ns polyprotein, the 2/3 site evidently lacks primary sequence elements in the vicinity of the scissile bond sufficient for specific protease recognition. In this study, we sought to investigate the molecular details of the regulation of the 2/3 site processing in the SFV ns polyprotein. We present evidence that correct macromolecular assembly, presumably strengthened by exosite interactions rather than the functionality of the individual nsP2 protease, is the driving force for specific substrate targeting. We conclude that structural elements within the macrodomain of nsP3 are used for precise positioning of a substrate recognition sequence at the catalytic center of the protease and that this process is coordinated by the exact N-terminal end of nsP2, thus representing a unique regulation mechanism used by alphaviruses.


Subject(s)
Polyproteins/metabolism , RNA-Dependent RNA Polymerase/metabolism , Semliki forest virus/enzymology , Viral Proteins/metabolism , Alphavirus Infections/virology , Amino Acid Sequence , Animals , Cell Line , Cricetinae , Molecular Sequence Data , Polyproteins/chemistry , Polyproteins/genetics , Protein Processing, Post-Translational , Proteolysis , RNA-Dependent RNA Polymerase/genetics , Semliki forest virus/chemistry , Semliki forest virus/genetics , Semliki forest virus/physiology , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication
7.
J Virol ; 84(22): 11624-33, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20826687

ABSTRACT

Semliki Forest virus (SFV) is an enveloped alphavirus that infects cells by a low-pH-triggered membrane fusion reaction mediated by the viral E1 protein. E1 inserts into target membranes and refolds to a hairpin-like homotrimer containing a central core trimer and an outer layer composed of domain III and the juxtamembrane stem region. The key residues involved in mediating E1 trimerization are not well understood. We recently showed that aspartate 188 in the interface of the core trimer plays a critical role. Substitution with lysine (D188K) blocks formation of the core trimer and E1 trimerization and strongly inhibits virus fusion and infection. Here, we have isolated and characterized revertants that rescued the fusion and growth defects of D188K. These revertants included pseudorevertants containing acidic or polar neutral residues at E1 position 188 and a second-site revertant containing an E1 K176T mutation. Computational analysis using multiconformation continuum electrostatics revealed an important interaction bridging D188 of one chain with K176 of the adjacent chain in the core trimer. E1 K176 is completely conserved among the alphaviruses, and mutations of K176 to threonine (K176T) or isoleucine (K176I) produced similar fusion phenotypes as D188 mutants. Together, our data support a model in which a ring of three salt bridges formed by D188 and K176 stabilize the core trimer, a key intermediate of the alphavirus fusion protein.


Subject(s)
Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mutation, Missense , Protein Multimerization , Semliki forest virus/physiology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Virus Internalization , Amino Acid Sequence , Amino Acid Substitution , Animals , Cricetinae , Membrane Fusion , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Semliki forest virus/chemistry , Semliki forest virus/genetics , Semliki forest virus/metabolism , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
8.
J Virol ; 83(16): 7996-8003, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19474104

ABSTRACT

Although enveloped virus assembly in the host cell is a crucial step in the virus life cycle, it remains poorly understood. One issue is how viruses include lipids in their membranes during budding from infected host cells. To analyze this issue, we took advantage of the fact that baby hamster kidney cells can be infected by two different viruses, namely, vesicular stomatitis virus and Semliki Forest virus, from the Rhabdoviridae and Togaviridae families, respectively. We purified the host plasma membrane and the two different viruses after exit from the host cells and analyzed the lipid compositions of the membranes by quantitative shotgun mass spectrometry. We observed that the lipid compositions of these otherwise structurally different viruses are virtually indistinguishable, and only slight differences were detected between the viral lipid composition and that of the plasma membrane. Taken together, the facts that the lipid compositions of the two viruses are so similar and that they strongly resemble the composition of the plasma membrane suggest that these viruses exert little selection in including lipids in their envelopes.


Subject(s)
Alphavirus Infections/virology , Cell Membrane/chemistry , Membrane Lipids/chemistry , Rhabdoviridae Infections/virology , Semliki forest virus/chemistry , Vesiculovirus/chemistry , Virus Assembly , Alphavirus Infections/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cell Membrane/virology , Cricetinae , Host-Pathogen Interactions , Mass Spectrometry , Membrane Lipids/genetics , Membrane Lipids/metabolism , Rhabdoviridae Infections/metabolism , Semliki forest virus/physiology , Vesiculovirus/physiology
9.
J Gen Virol ; 89(Pt 6): 1457-1466, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18474562

ABSTRACT

Semliki Forest virus (SFV) non-structural protein 1 (nsP1) is a major component of the virus replicase complex. It has previously been studied in cells infected with virus or using transient or stable expression systems. To extend these studies, tetracycline-inducible stable cell lines expressing SFV nsP1 or its palmitoylation-negative mutant (nsP16D) were constructed. The levels of protein expression and the subcellular localization of nsP1 in induced cells were similar to those in virus-infected cells. The nsP1 expressed by stable, inducible cell lines or by SFV-infected HEK293 T-REx cells was a stable protein with a half-life of approximately 5 h. In contrast to SFV infection, induction of nsP1 expression had no detectable effect on cellular transcription, translation or viability. Induction of expression of nsP1 or nsP16D interfered with multiplication of SFV, typically resulting in a 5-10-fold reduction in virus yields. This reduction was not due to a decrease in the number of infected cells, indicating that nsP1 expression does not block virus entry or initiation of replication. Expression of nsP1 interfered with virus genomic RNA synthesis and delayed accumulation of viral subgenomic RNA translation products. Expression of nsP1 with a mutation in the palmitoylation site reduced synthesis of genomic and subgenomic RNAs and their products of translation, and this effect did not resolve with time. These results are in agreement with data published previously, suggesting a role for nsP1 in genomic RNA synthesis.


Subject(s)
Alphavirus Infections/virology , Semliki forest virus/chemistry , Semliki forest virus/physiology , Viral Nonstructural Proteins/physiology , Animals , Cricetinae , Down-Regulation , Humans , Intracellular Space/metabolism , RNA-Dependent RNA Polymerase/chemistry , Viral Proteins/chemistry , Virus Replication
10.
J Virol ; 81(20): 11218-25, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17686870

ABSTRACT

Cells expressing the low pH-triggered class II viral fusion protein E1 of Semliki Forest virus (SFV) were fused to target cells. Fusion was monitored by electrical capacitance and aqueous dye measurements. Electrical voltage-clamp measurements showed that SFV E1-induced cell-cell fusion occurred quickly after acidification for a trans-negative potential across the target membrane (i.e., negative potential inside the target cell) but that a trans-positive potential eliminated all fusion. Use of an ionophore to control potentials for a large population of cells confirmed the dependence of fusion on voltage polarity. In contrast, fusion induced by the class I fusion proteins of human immunodeficiency virus, avian sarcoma leukosis virus, and influenza virus was independent of the voltage polarity across the target cell. Initial pore size and pore growth were also independent of voltage polarity for the class I proteins. An intermediate of SFV E1-induced fusion was created by transient acidification at low temperature. Membranes were hemifused at this intermediate state, and raising the temperature at neutral pH allowed full fusion to occur. Capacitance measurements showed that maintaining a trans-positive potential definitely blocked fusion at steps following the creation of the hemifusion intermediate and may have inhibited fusion at prior steps. It is proposed that the trans-negative voltage across the endosomal membrane facilitates fusion after low-pH-induced conformational changes of SFV E1 have occurred.


Subject(s)
Cell Fusion , Membrane Potentials/physiology , Semliki forest virus/physiology , Viral Envelope Proteins/physiology , Viral Fusion Proteins/physiology , Cell Line , Cell Membrane Permeability , Electric Capacitance , Electrophysiology , Endosomes , Humans , Hydrogen-Ion Concentration , Intracellular Membranes/physiology , Permeability , Semliki forest virus/chemistry , Temperature
11.
Biopolymers ; 85(5-6): 422-37, 2007.
Article in English | MEDLINE | ID: mdl-17211889

ABSTRACT

Abeta fibrils, which are central to the pathology of Alzheimer's disease, form a cross-beta-structure that contains likely parallel beta-sheets with a salt bridge between residues Asp23 and Lys28. Recent studies suggest that soluble oligomers of amyloid peptides have neurotoxic effects in cell cultures, raising the interest in studying the structures of these intermediate forms. Here, we present three models of possible soluble Abeta forms based on the sequences similarities, assumed to support local structural similarities, of the Abeta peptide with fragments of three proteins (adhesin, Semliki Forest virus capsid protein, and transthyretin). These three models share a similar structure in the C-terminal region composed of two beta-strands connected by a loop, which contain the Asp23-Lys28 salt bridge. This segment is also structurally well conserved in Abeta fibril forms. Differences between the three monomeric models occur in the N-terminal region and in the C-terminal tail. These three models might sample some of the most stable conformers of the soluble Abeta peptide within oligomeric assemblies, which were modeled here in the form of dimers, trimers, tetramers, and hexamers. The consistency of these models is discussed with respect to available experimental and theoretical data.


Subject(s)
Amyloid beta-Peptides/chemistry , Peptide Fragments/chemistry , Adhesins, Escherichia coli/chemistry , Amino Acid Sequence , Capsid Proteins/chemistry , Dimerization , Humans , Models, Molecular , Molecular Sequence Data , Prealbumin/chemistry , Protein Conformation , Protein Folding , Semliki forest virus/chemistry , Semliki forest virus/enzymology , Sequence Alignment
12.
J Neuroimmunol ; 184(1-2): 149-55, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17257687

ABSTRACT

Anti-SFV antibodies generated during SFV infection may affect CNS function due to cross-reactivity with a peptide of oligodendrocyte myelin glycoprotein. To explore this possibility, total IgG from SFV immunized or normal control rabbits was unilaterally microinfused into the subthalamic region of normal rat brain. Behavior of the IgG-infused rats was determined using a bioassay, measuring rotational locomotion following systemic injection of apomorphine. Anti-SFV IgG-infused rats demonstrated a significantly increased (p<0.005) ipsilateral turning response compared to control rats, persisting for at least a month. Results suggest that brain cross-reactive antibodies in anti-SFV IgG may affect brain function.


Subject(s)
Antibodies/pharmacology , Apomorphine/pharmacology , Behavior, Animal/drug effects , Dopamine Agonists/pharmacology , Semliki forest virus/chemistry , Viral Proteins/immunology , Animals , Brain/drug effects , Enzyme-Linked Immunosorbent Assay/methods , Male , Motor Activity/drug effects , Rats , Rats, Inbred F344 , Rotarod Performance Test , Time Factors
13.
J Biol Chem ; 282(9): 6752-62, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17192272

ABSTRACT

Semliki Forest virus is among the prototypes for Class II virus fusion and targets the endosomal membrane. Fusion protein E1 and its envelope companion E2 are both anchored in the viral membrane and form an external shell with protruding spikes. In acid environments, mimicking the early endosomal milieu, surface epitopes in the virus rearrange along with exposure of the fusion loop. To visualize this transformation into a fusogenic stage, we determined the structure of the virus at gradually lower pH values. The results show that while the fusion loop is available for external interaction and the shell and stalk domains of the spike begin to deteriorate, the E1 and E2 remain in close contact in the spike head. This unexpected observation points to E1 and E2 cooperation beyond the fusion loop exposure stage and implies a more prominent role for E2 in guiding membrane close encounter than has been earlier anticipated.


Subject(s)
Cryoelectron Microscopy , Semliki forest virus/chemistry , Semliki forest virus/pathogenicity , Viral Envelope Proteins/chemistry , Viral Fusion Proteins/chemistry , Hydrogen-Ion Concentration , Membrane Fusion , Molecular Structure , Phase Transition , Protein Conformation , Viral Envelope Proteins/physiology , Viral Fusion Proteins/physiology
14.
J Virol ; 80(19): 9599-607, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16973563

ABSTRACT

The E1 envelope protein of the alphavirus Semliki Forest virus (SFV) is a class II fusion protein that mediates low pH-triggered membrane fusion during virus infection. Like other class I and class II fusion proteins, during fusion E1 inserts into the target membrane and rearranges to form a trimeric hairpin structure. The postfusion structures of the alphavirus and flavivirus fusion proteins suggest that the "stem" region connecting the fusion protein domain III to the transmembrane domain interacts along the trimer core during the low pH-induced conformational change. However, the location of the E1 stem in the SFV particle and its rearrangement and functional importance during fusion are not known. We developed site-directed polyclonal antibodies to the N- or C-terminal regions of the SFV E1 stem and used them to study the stem during fusion. The E1 stem was hidden on neutral pH virus but became accessible after low pH-triggered dissociation of the E2/E1 heterodimer. The stem packed onto the trimer core in the postfusion conformation and became inaccessible to antibody binding. Generation of the E1 homotrimer on fusion-incompetent membranes identified an intermediate conformation in which domain III had folded back but stem packing was incomplete. Our data suggest that E1 hairpin formation occurs by the sequential packing of domain III and the stem onto the trimer core and indicate a tight correlation between stem packing and membrane merger.


Subject(s)
Antibodies, Viral/immunology , Semliki forest virus/metabolism , Viral Fusion Proteins/immunology , Viral Fusion Proteins/metabolism , Animals , Cell Line , Cricetinae , Dimerization , Hydrogen-Ion Concentration , Protein Conformation , Semliki forest virus/chemistry , Semliki forest virus/genetics , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics
15.
J Virol ; 80(17): 8493-502, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16912299

ABSTRACT

Macro domains constitute a protein module family found associated with specific histones and proteins involved in chromatin metabolism. In addition, a small number of animal RNA viruses, such as corona- and toroviruses, alphaviruses, and hepatitis E virus, encode macro domains for which, however, structural and functional information is extremely limited. Here, we characterized the macro domains from hepatitis E virus, Semliki Forest virus, and severe acute respiratory syndrome coronavirus (SARS-CoV). The crystal structure of the SARS-CoV macro domain was determined at 1.8-Angstroms resolution in complex with ADP-ribose. Information derived from structural, mutational, and sequence analyses suggests a close phylogenetic and, most probably, functional relationship between viral and cellular macro domain homologs. The data revealed that viral macro domains have relatively poor ADP-ribose 1"-phosphohydrolase activities (which were previously proposed to be their biologically relevant function) but bind efficiently free and poly(ADP-ribose) polymerase 1-bound poly(ADP-ribose) in vitro. Collectively, these results suggest to further evaluate the role of viral macro domains in host response to viral infection.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Hepatitis E virus/chemistry , Histones/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Semliki forest virus/chemistry , Severe acute respiratory syndrome-related coronavirus/chemistry , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/chemistry , Amino Acid Sequence , Crystallography , Hepatitis E virus/metabolism , Histones/chemistry , Models, Molecular , Molecular Sequence Data , Poly(ADP-ribose) Polymerases/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Semliki forest virus/metabolism , Structure-Activity Relationship
16.
Structure ; 14(1): 75-86, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16407067

ABSTRACT

Semliki Forest virus (SFV) is enveloped by a lipid bilayer enclosed within a glycoprotein cage made by glycoproteins E1 and E2. E1 is responsible for inducing membrane fusion, triggered by exposure to the acidic environment of the endosomes. Acidic pH induces E1/E2 dissociation, allowing E1 to interact with the target membrane, and, at the same time, to rearrange into E1 homotrimers that drive the membrane fusion reaction. We previously reported a preliminary Calpha trace of the monomeric E1 glycoprotein ectodomain and its organization on the virus particle. We also reported the 3.3 A structure of the trimeric, fusogenic conformation of E1. Here, we report the crystal structure of monomeric E1 refined to 3 A resolution and describe the amino acids involved in contacts in the virion. These results identify the major determinants for the E1/E2 icosahedral shell formation and open the way to rational mutagenesis approaches to shed light on SFV assembly.


Subject(s)
Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Semliki forest virus/chemistry , Semliki forest virus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Glycoproteins , Histidine/genetics , Lipids/physiology , Membrane Fusion/physiology , Membrane Fusion Proteins/chemistry , Membrane Fusion Proteins/genetics , Membrane Fusion Proteins/metabolism , Membrane Glycoproteins/genetics , Molecular Sequence Data , Mutation , Protein Binding , Protein Structure, Tertiary , Semliki forest virus/genetics , Viral Envelope Proteins/genetics
17.
J Gen Virol ; 85(Pt 6): 1695-1701, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15166454

ABSTRACT

Studies using the alphavirus Semliki Forest virus have indicated that the viral E1 fusion protein forms two types of pore: fusion pores and ion-permeable pores. The formation of ion-permeable pores has not been generally accepted, partly because it was not evident how the protein might form these different pores. Here it is proposed that the choice of the target membrane determines whether a fusion pore or ion-permeable pores are formed. The fusion protein is activated in the endosome and for steric reasons only a fraction of the activated molecules can interact with the endosomal membrane. This target membrane reaction forms the fusion pore. It is proposed that the rest of the activated molecules interact with the membrane in which the protein is anchored and that this self-membrane reaction leads to formation of ion-permeable pores, which can be detected in the target membrane after fusion of the viral membrane into the target membrane.


Subject(s)
Membrane Fusion , Semliki forest virus/physiology , Viral Fusion Proteins/chemistry , Hydrogen-Ion Concentration , Semliki forest virus/chemistry , Viral Fusion Proteins/physiology
18.
J Mol Biol ; 338(1): 159-67, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-15050831

ABSTRACT

The capsid protein of Semliki Forest virus constitutes the N-terminal part of a large viral polyprotein. It consists of an unstructured basic segment (residues 1-118) and a 149 residue serine protease module (SFVP, residues 119-267) comprised of two beta-barrel domains. Previous in vivo and in vitro translation experiments have demonstrated that SFVP folds co-translationally during synthesis of the viral polyprotein and rapidly cleaves itself off the nascent chain. To test whether fast co-translation folding of SFVP is an intrinsic property of the polypeptide chain or whether folding is accelerated by cellular components, we investigated spontaneous folding of recombinant SFVP in vitro. The results show that the majority of unfolded SFVP molecules fold faster than any previously studied two-domain protein (tau=50 ms), and that folding of the N-terminal domain precedes structure formation of the C-terminal domain. This shows that co-translational folding of SFVP does not require additional cellular components and suggests that rapid folding is the result of molecular evolution towards efficient virus biogenesis.


Subject(s)
Capsid Proteins/chemistry , Capsid Proteins/metabolism , Capsid/chemistry , Peptide Hydrolases/metabolism , Protein Folding , Protein Processing, Post-Translational , Semliki forest virus/chemistry , Capsid Proteins/genetics , Humans , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
19.
J Virol ; 78(7): 3514-23, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15016874

ABSTRACT

The fusion proteins of the alphaviruses and flaviviruses have a similar native structure and convert to a highly stable homotrimer conformation during the fusion of the viral and target membranes. The properties of the alpha- and flavivirus fusion proteins distinguish them from the class I viral fusion proteins, such as influenza virus hemagglutinin, and establish them as the first members of the class II fusion proteins. Understanding how this new class carries out membrane fusion will require analysis of the structural basis for both the interaction of the protein subunits within the homotrimer and their interaction with the viral and target membranes. To this end we report a purification method for the E1 ectodomain homotrimer from the alphavirus Semliki Forest virus. The purified protein is trimeric, detergent soluble, retains the characteristic stability of the starting homotrimer, and is free of lipid and other contaminants. In contrast to the postfusion structures that have been determined for the class I proteins, the E1 homotrimer contains the fusion peptide region responsible for interaction with target membranes. This E1 trimer preparation is an excellent candidate for structural studies of the class II viral fusion proteins, and we report conditions that generate three-dimensional crystals suitable for analysis by X-ray diffraction. Determination of the structure will provide our first high-resolution views of both the low-pH-induced trimeric conformation and the target membrane-interacting region of the alphavirus fusion protein.


Subject(s)
Semliki forest virus/chemistry , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/isolation & purification , Animals , Cell Line , Centrifugation, Density Gradient , Cricetinae , Crystallization , Crystallography, X-Ray , Glycoproteins/chemistry , Glycoproteins/isolation & purification , Glycoproteins/metabolism , Hydrogen-Ion Concentration , Light , Liposomes , Protein Binding , Protein Structure, Quaternary , Scattering, Radiation , Solubility , Viral Fusion Proteins/metabolism
20.
Nature ; 427(6972): 320-5, 2004 Jan 22.
Article in English | MEDLINE | ID: mdl-14737160

ABSTRACT

Fusion of biological membranes is mediated by specific lipid-interacting proteins that induce the formation and expansion of an initial fusion pore. Here we report the crystal structure of the ectodomain of the Semliki Forest virus fusion glycoprotein E1 in its low-pH-induced trimeric form. E1 adopts a folded-back conformation that, in the final post-fusion form of the full-length protein, would bring the fusion peptide loop and the transmembrane anchor to the same end of a stable protein rod. The observed conformation of the fusion peptide loop is compatible with interactions only with the outer leaflet of the lipid bilayer. Crystal contacts between fusion peptide loops of adjacent E1 trimers, together with electron microscopy observations, suggest that in an early step of membrane fusion, an intermediate assembly of five trimers creates two opposing nipple-like deformations in the viral and target membranes, leading to formation of the fusion pore.


Subject(s)
Semliki forest virus/chemistry , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Amino Acid Sequence , Cell Membrane/chemistry , Cell Membrane/metabolism , Crystallography, X-Ray , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Semliki forest virus/ultrastructure , Viral Fusion Proteins/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...