Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 19675, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33184473

ABSTRACT

Nora virus, a virus of Drosophila, encapsidates one of the largest single-stranded RNA virus genomes known. Its taxonomic affinity is uncertain as it has a picornavirus-like cassette of enzymes for virus replication, but the capsid structure was at the time for genome publication unknown. By solving the structure of the virus, and through sequence comparison, we clear up this taxonomic ambiguity in the invertebrate RNA virosphere. Despite the lack of detectable similarity in the amino acid sequences, the 2.7 Å resolution cryoEM map showed Nora virus to have T = 1 symmetry with the characteristic capsid protein ß-barrels found in all the viruses in the Picornavirales order. Strikingly, α-helical bundles formed from the extended C-termini of capsid protein VP4B and VP4C protrude from the capsid surface. They are similar to signalling molecule folds and implicated in virus entry. Unlike other viruses of Picornavirales, no intra-pentamer stabilizing annulus was seen, instead the intra-pentamer stability comes from the interaction of VP4C and VP4B N-termini. Finally, intertwining of the N-termini of two-fold symmetry-related VP4A capsid proteins and RNA, provides inter-pentamer stability. Based on its distinct structural elements and the genetic distance to other picorna-like viruses we propose that Nora virus, and a small group of related viruses, should have its own family within the order Picornavirales.


Subject(s)
Capsid/ultrastructure , Picornaviridae/ultrastructure , Receptors, Virus/metabolism , Binding Sites , Biological Evolution , Capsid/metabolism , Capsid/physiology , Cryoelectron Microscopy , Models, Molecular , Phylogeny , Picornaviridae/classification , Picornaviridae/physiology , Protein Stability , RNA, Viral/metabolism
3.
J Virol ; 87(20): 10918-35, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23926354

ABSTRACT

Boid inclusion body disease (BIBD) is a progressive, usually fatal disease of constrictor snakes, characterized by cytoplasmic inclusion bodies (IB) in a wide range of cell types. To identify the causative agent of the disease, we established cell cultures from BIBD-positive and -negative boa constrictors. The IB phenotype was maintained in cultured cells of affected animals, and supernatants from these cultures caused the phenotype in cultures originating from BIBD-negative snakes. Viruses were purified from the supernatants by ultracentrifugation and subsequently identified as arenaviruses. Purified virus also induced the IB phenotype in naive cells, which fulfilled Koch's postulates in vitro. One isolate, tentatively designated University of Helsinki virus (UHV), was studied in depth. Sequencing confirmed that UHV is a novel arenavirus species that is distinct from other known arenaviruses including those recently identified in snakes with BIBD. The morphology of UHV was established by cryoelectron tomography and subtomographic averaging, revealing the trimeric arenavirus spike structure at 3.2-nm resolution. Immunofluorescence, immunohistochemistry, and immunoblotting with a polyclonal rabbit antiserum against UHV and reverse transcription-PCR (RT-PCR) revealed the presence of genetically diverse arenaviruses in a large cohort of snakes with BIBD, confirming the causative role of arenaviruses. Some snakes were also found to carry arenavirus antibodies. Furthermore, mammalian cells (Vero E6) were productively infected with UHV, demonstrating the potential of arenaviruses to cross species barriers. In conclusion, we propose the newly identified lineage of arenaviruses associated with BIBD as a novel taxonomic entity, boid inclusion body disease-associated arenaviruses (BIBDAV), in the family Arenaviridae.


Subject(s)
Arenaviridae Infections/veterinary , Arenavirus/classification , Arenavirus/isolation & purification , Snakes/virology , Animals , Arenaviridae Infections/virology , Arenavirus/genetics , Arenavirus/ultrastructure , Cells, Cultured , Cluster Analysis , Cryoelectron Microscopy , Electron Microscope Tomography , Inclusion Bodies , Molecular Sequence Data , Phylogeny , RNA, Viral/genetics , Sequence Analysis, DNA , Ultracentrifugation , Virion/ultrastructure
4.
Biomaterials ; 34(33): 8491-503, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23932247

ABSTRACT

We have developed a highly efficient method for the radiolabeling of phytantriol (PHYT)/oleic acid (OA)-based hexosomes based on the surface chelation of technetium-99m ((99m)Tc) to preformed hexosomes using the polyamine 1, 12-diamino-3, 6, 9-triazododecane (SpmTrien) as chelating agent. We also report on the unsuccessful labeling of cubosomes using the well-known chelating agent hexamethylpropyleneamine oxime (HMPAO). The (99m)Tc-labeled SpmTrien-hexosomes ((99m)Tc-SpmTrien-hexosomes) were synthesized with good radiolabeling (84%) and high radiochemical purity (>90%). The effect of radiolabeling on the internal nanostructure and the overall size of these aqueous dispersions was investigated by using synchrotron small angle X-ray scattering (SAXS), dynamic light scattering (DLS), and transmission electron cryo microscopy (cryo-TEM). Further, we show the utility of (99m)Tc-SpmTrien-hexosomes for the in vivo imaging of healthy mice using single photon emission computed tomography (SPECT) in combination with computed tomography (CT), i.e. SPECT/CT. SPECT/CT experiments of subcutaneously administered (99m)Tc-SpmTrien-hexosomes to the flank of mice showed a high stability in vivo allowing imaging of the distribution of the radiolabeled hexosomes for up to 24 h. These injected (99m)Tc-SpmTrien-hexosomes formed a deposit within the subcutaneous adipose tissue, displaying a high biodistribution of ≈ 343% injected dose/g tissue (%ID/g), with negligible uptake in other organs and tissues. The developed (99m)Tc labeling method for PHYT/OA-based hexosomes could further serve as a useful tool for investigating and imaging the in vivo performance of cubosomal and hexosomal drug nanocarriers.


Subject(s)
Technetium , Tomography, Emission-Computed, Single-Photon/methods , Animals , Chelating Agents/chemistry , Mice
5.
Proc Natl Acad Sci U S A ; 110(26): 10604-9, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23733949

ABSTRACT

It has been proposed that viruses can be divided into a small number of structure-based viral lineages. One of these lineages is exemplified by bacterial virus Hong Kong 97 (HK97), which represents the head-tailed dsDNA bacteriophages. Seemingly similar viruses also infect archaea. Here we demonstrate using genomic analysis, electron cryomicroscopy, and image reconstruction that the major coat protein fold of newly isolated archaeal Haloarcula sinaiiensis tailed virus 1 has the canonical coat protein fold of HK97. Although it has been anticipated previously, this is physical evidence that bacterial and archaeal head-tailed viruses share a common architectural principle. The HK97-like fold has previously been recognized also in herpesviruses, and this study expands the HK97-like lineage to viruses from all three domains of life. This is only the second established lineage to include archaeal, bacterial, and eukaryotic viruses. Thus, our findings support the hypothesis that the last common universal ancestor of cellular organisms was infected by a number of different viruses.


Subject(s)
Archaeal Viruses/chemistry , Archaeal Viruses/ultrastructure , Capsid Proteins/chemistry , Haloarcula/virology , Archaeal Viruses/genetics , Capsid/chemistry , Capsid/ultrastructure , Capsid Proteins/genetics , Computer Simulation , Cryoelectron Microscopy , Genome, Viral , Imaging, Three-Dimensional , Models, Molecular , Molecular Sequence Data , Protein Folding
6.
J Bacteriol ; 195(8): 1727-34, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23396908

ABSTRACT

Chlorosomes are large light-harvesting complexes found in three phyla of anoxygenic photosynthetic bacteria. Chlorosomes are primarily composed of self-assembling pigment aggregates. In addition to the main pigment, bacteriochlorophyll c, d, or e, chlorosomes also contain variable amounts of carotenoids. Here, we use X-ray scattering and electron cryomicroscopy, complemented with absorption spectroscopy and pigment analysis, to compare the morphologies, structures, and pigment compositions of chlorosomes from Chloroflexus aurantiacus grown under two different light conditions and Chlorobaculum tepidum. High-purity chlorosomes from C. aurantiacus contain about 20% more carotenoid per bacteriochlorophyll c molecule when grown under low light than when grown under high light. This accentuates the light-harvesting function of carotenoids, in addition to their photoprotective role. The low-light chlorosomes are thicker due to the overall greater content of pigments and contain domains of lamellar aggregates. Experiments where carotenoids were selectively extracted from intact chlorosomes using hexane proved that they are located in the interlamellar space, as observed previously for species belonging to the phylum Chlorobi. A fraction of the carotenoids are localized in the baseplate, where they are bound differently and cannot be removed by hexane. In C. tepidum, carotenoids cannot be extracted by hexane even from the chlorosome interior. The chemical structure of the pigments in C. tepidum may lead to π-π interactions between carotenoids and bacteriochlorophylls, preventing carotenoid extraction. The results provide information about the nature of interactions between bacteriochlorophylls and carotenoids in the protein-free environment of the chlorosome interior.


Subject(s)
Carotenoids/chemistry , Chloroflexus/metabolism , Light , Phycobiliproteins/chemistry , Phycobiliproteins/physiology , Bacterial Chromatophores , Carotenoids/metabolism , Chloroflexus/cytology , Molecular Structure , Organelles/physiology , Pigments, Biological , X-Ray Diffraction
7.
J Virol ; 87(7): 3943-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23365426

ABSTRACT

Coxsackievirus A9 (CVA9) is an important pathogen of the Picornaviridae family. It utilizes cellular receptors from the integrin αv family for binding to its host cells prior to entry and genome release. Among the integrins tested, it has the highest affinity for αvß6, which recognizes the arginine-glycine-aspartic acid (RGD) loop present on the C terminus of viral capsid protein, VP1. As the atomic model of CVA9 lacks the RGD loop, we used surface plasmon resonance, electron cryo-microscopy, and image reconstruction to characterize the capsid-integrin interactions and the conformational changes on genome release. We show that the integrin binds to the capsid with nanomolar affinity and that the binding of integrin to the virion does not induce uncoating, thereby implying that further steps are required for release of the genome. Electron cryo-tomography and single-particle image reconstruction revealed variation in the number and conformation of the integrins bound to the capsid, with the integrin footprint mapping close to the predicted site for the exposed RGD loop on VP1. Comparison of empty and RNA-filled capsid reconstructions showed that the capsid undergoes conformational changes when the genome is released, so that the RNA-capsid interactions in the N termini of VP1 and VP4 are lost, VP4 is removed, and the capsid becomes more porous, as has been reported for poliovirus 1, human rhinovirus 2, enterovirus 71, and coxsackievirus A7. These results are important for understanding the structural basis of integrin binding to CVA9 and the molecular events leading to CVA9 cell entry and uncoating.


Subject(s)
Antigens, Neoplasm/metabolism , Capsid Proteins/metabolism , Enterovirus B, Human/physiology , Integrins/metabolism , Models, Molecular , Virus Attachment , Virus Uncoating/physiology , Antigens, Neoplasm/chemistry , Cryoelectron Microscopy , Enterovirus B, Human/genetics , Enterovirus B, Human/metabolism , Integrins/chemistry , Protein Binding , Protein Conformation , Surface Plasmon Resonance , Virus Uncoating/genetics
8.
J Virol ; 87(6): 3248-60, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23283946

ABSTRACT

Extremophilic archaea, both hyperthermophiles and halophiles, dominate in habitats where rather harsh conditions are encountered. Like all other organisms, archaeal cells are susceptible to viral infections, and to date, about 100 archaeal viruses have been described. Among them, there are extraordinary virion morphologies as well as the common head-tailed viruses. Although approximately half of the isolated archaeal viruses belong to the latter group, no three-dimensional virion structures of these head-tailed viruses are available. Thus, rigorous comparisons with bacteriophages are not yet warranted. In the present study, we determined the genome sequences of two of such viruses of halophiles and solved their capsid structures by cryo-electron microscopy and three-dimensional image reconstruction. We show that these viruses are inactivated, yet remain intact, at low salinity and that their infectivity is regained when high salinity is restored. This enabled us to determine their three-dimensional capsid structures at low salinity to a ∼10-Šresolution. The genetic and structural data showed that both viruses belong to the same T-number class, but one of them has enlarged its capsid to accommodate a larger genome than typically associated with a T=7 capsid by inserting an additional protein into the capsid lattice.


Subject(s)
Archaea/virology , Archaeal Viruses/genetics , Archaeal Viruses/ultrastructure , DNA, Viral/chemistry , DNA, Viral/genetics , Genome, Viral , Virion/ultrastructure , Archaeal Viruses/isolation & purification , Archaeal Viruses/physiology , Capsid/ultrastructure , Cryoelectron Microscopy , Imaging, Three-Dimensional , Microbial Viability/drug effects , Molecular Sequence Data , Sequence Analysis, DNA , Sodium Chloride/metabolism
9.
J Virol ; 86(15): 7858-66, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22593166

ABSTRACT

African horsesickness (AHS) is a devastating disease of horses. The disease is caused by the double-stranded RNA-containing African horsesickness virus (AHSV). Using electron cryomicroscopy and three-dimensional image reconstruction, we determined the architecture of an AHSV serotype 4 (AHSV-4) reference strain. The structure revealed triple-layered AHS virions enclosing the segmented genome and transcriptase complex. The innermost protein layer contains 120 copies of VP3, with the viral polymerase, capping enzyme, and helicase attached to the inner surface of the VP3 layer on the 5-fold axis, surrounded by double-stranded RNA. VP7 trimers form a second, T=13 layer on top of VP3. Comparative analyses of the structures of bluetongue virus and AHSV-4 confirmed that VP5 trimers form globular domains and VP2 trimers form triskelions, on the virion surface. We also identified an AHSV-7 strain with a truncated VP2 protein (AHSV-7 tVP2) which outgrows AHSV-4 in culture. Comparison of AHSV-7 tVP2 to bluetongue virus and AHSV-4 allowed mapping of two domains in AHSV-4 VP2, and one in bluetongue virus VP2, that are important in infection. We also revealed a protein plugging the 5-fold vertices in AHSV-4. These results shed light on virus-host interactions in an economically important orbivirus to help the informed design of new vaccines.


Subject(s)
African Horse Sickness Virus/ultrastructure , Models, Molecular , Virion/ultrastructure , African Horse Sickness/metabolism , African Horse Sickness Virus/metabolism , Animals , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Chlorocebus aethiops , Horses/virology , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , RNA, Viral/chemistry , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Structure-Activity Relationship , Vero Cells , Viral Core Proteins/chemistry , Viral Core Proteins/metabolism , Virion/metabolism
10.
J Virol ; 86(13): 7207-15, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22514349

ABSTRACT

Coxsackievirus A7 (CAV7) is a rarely detected and poorly characterized serotype of the Enterovirus species Human enterovirus A (HEV-A) within the Picornaviridae family. The CAV7-USSR strain has caused polio-like epidemics and was originally thought to represent the fourth poliovirus type, but later evidence linked this strain to the CAV7-Parker prototype. Another isolate, CAV7-275/58, was also serologically similar to Parker but was noninfectious in a mouse model. Sequencing of the genomic region encoding the capsid proteins of the USSR and 275/58 strains and subsequent comparison with the corresponding amino acid sequences of the Parker strain revealed that the Parker and USSR strains are nearly identical, while the 275/58 strain is more distant. Using electron cryomicroscopy and three-dimensional image reconstruction, the structures of the CAV7-USSR virion and empty capsid were resolved to 8.2-Å and 6.1-Å resolutions, respectively. This is one of the first detailed structural analyses of the HEV-A species. Using homology modeling, reconstruction segmentation, and flexible fitting, we constructed a pseudoatomic T = 1 (pseudo T = 3) model incorporating the three major capsid proteins (VP1 to VP3), addressed the conformational changes of the capsid and its constituent viral proteins occurring during RNA release, and mapped the capsid proteins' variable regions to the structure. During uncoating, VP4 and RNA are released analogously to poliovirus 1, the interfaces of VP2 and VP3 are rearranged, and VP1 rotates. Variable regions in the capsid proteins were predicted to map mainly to the surface of VP1 and are thus likely to affect the tropism and pathogenicity of CAV7.


Subject(s)
Capsid Proteins/genetics , Capsid Proteins/ultrastructure , Enterovirus/genetics , Enterovirus/ultrastructure , Virus Internalization , Cryoelectron Microscopy , Enterovirus/physiology , Humans , Image Processing, Computer-Assisted , Models, Biological , Molecular Sequence Data , Protein Conformation , Sequence Analysis, DNA
11.
J Struct Biol ; 178(3): 215-24, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22508104

ABSTRACT

Bacteriophage P4 is dependent on structural proteins supplied by a helper phage, P2, to assemble infectious virions. Bacteriophage P2 normally forms an icosahedral capsid with T=7 symmetry from the gpN capsid protein, the gpO scaffolding protein and the gpQ portal protein. In the presence of P4, however, the same structural proteins are assembled into a smaller capsid with T=4 symmetry. This size determination is effected by the P4-encoded protein Sid, which forms an external scaffold around the small P4 procapsids. Size responsiveness (sir) mutants in gpN fail to assemble small capsids even in the presence of Sid. We have produced large and small procapsids by co-expression of gpN with gpO and Sid, respectively, and applied cryo-electron microscopy and three-dimensional reconstruction methods to visualize these procapsids. gpN has an HK97-like fold and interacts with Sid in an exposed loop where the sir mutations are clustered. The T=7 lattice of P2 has dextro handedness, unlike the laevo lattices of other phages with this fold observed so far.


Subject(s)
Bacteriophage P2/chemistry , Bacteriophage P2/ultrastructure , Capsid/chemistry , Capsid/diagnostic imaging , Myoviridae/chemistry , Myoviridae/ultrastructure , Bacteriophage P2/genetics , Cryoelectron Microscopy , Models, Biological , Mutation , Myoviridae/genetics , Protein Structure, Secondary , Ultrasonography
12.
J Virol ; 86(9): 5376-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22379079

ABSTRACT

Enveloped double-stranded RNA (dsRNA) bacterial virus Pseudomonas phage ϕ6 has been developed into an advanced assembly system where purified virion proteins and genome segments self-assemble into infectious viral particles, inferring the assembly pathway. The most intriguing step is the membrane assembly occurring inside the bacterial cell. Here, we demonstrate that the middle virion shell, made of protein 8, associates with the expanded viral core particle and the virus-specific membrane vesicle.


Subject(s)
Bacteriophage phi 6/physiology , Capsid Proteins/metabolism , Viral Envelope Proteins/metabolism , Lipid Metabolism , Protein Binding , Virus Assembly
13.
J Virol Methods ; 181(1): 6-11, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22265819

ABSTRACT

Recombinant expression of the norovirus capsid protein VP1 leads to self-assembly of non-infectious virus-like particles (VLPs), which are recognized as promising vaccine candidates against norovirus infections. To overcome the scalability issues connected to the ultracentrifugation-based purification strategies used in previous studies, an anion exchange-based purification method for norovirus VLPs was developed in this study. The method consists of precipitation by polyethylene glycol (PEG) and a single anion exchange chromatography step for purifying baculovirus-expressed GII.4 norovirus VLPs, which can be performed within one day. High product purity was obtained using chromatography. The purified material also contained fully assembled monodispersed VLPs, which were recognized by human sera containing polyclonal antibodies against norovirus GII.4.


Subject(s)
Chromatography, Ion Exchange/methods , Norovirus/genetics , Virology/methods , Virosomes/genetics , Virosomes/isolation & purification , Baculoviridae , Capsid Proteins/genetics , Capsid Proteins/metabolism , Fractional Precipitation , Genetic Vectors , Polyethylene Glycols/chemistry
14.
J Virol Methods ; 179(1): 1-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21600929

ABSTRACT

Noroviruses are an important cause of epidemic acute gastroenteritis in humans. In this study the production and characterization of GII.4 norovirus virus-like particles (VLPs) in insect cells is reported. Furthermore, the expression of corresponding norovirus polyhistidine-tagged P domain protein in Escherichia coli is described. The protruding P domain of the norovirus capsid is known to contain determinants for antibody and receptor binding. Therefore, P domain proteins were studied as an alternative diagnostic tool for evaluating norovirus infection. Analyses by dynamic light scattering and cryo-electron microscopy revealed the presence of intact VLPs with an average diameter of about 40 nm. Immunostaining and ELISA assays using norovirus-specific human sera revealed that VLPs and the P domain are recognized by norovirus-specific antibodies and by their putative receptor. The VLPs and P domain protein are potentially useful in the development of diagnostic and vaccination tools for noroviruses.


Subject(s)
Norovirus/genetics , Norovirus/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Virosomes/immunology , Virosomes/isolation & purification , Animals , Antibodies, Viral/blood , Caliciviridae Infections/diagnosis , Caliciviridae Infections/prevention & control , Cell Line , Escherichia coli/genetics , Gene Expression , Humans , Immunoassay , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Spodoptera , Viral Vaccines/immunology , Virosomes/genetics , Virosomes/metabolism
15.
J Control Release ; 147(1): 136-43, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20624434

ABSTRACT

Liposomes embedded with gold nanoparticles show light-triggered contents release. We investigated the mechanism of the light-induced changes and functionality of the light-induced release in the cells. The real time small angle X-ray scattering (SAXS) analysis revealed time-dependent phase transitions in distearoylphosphatidylcholine (DSPC)/dipalmitoylphosphatidylcholine (DPPC) liposomes upon heating. Similar changes were observed when gold nanoparticle-embedded liposomes were exposed to the UV light: gold nanoparticles absorb light energy and transfer it to heat, thereby causing lipid phase transition from gel phase to rippled phase, and further to fluid phase. Without UV light exposure the gold nanoparticles did not affect the liposomal bilayer periodicity. The light-triggered release of hydrophilic fluorescent probe (calcein) from the gold nanoparticle-loaded liposomes was demonstrated with fluorescence-activated cell sorting after liposome internalization into the ARPE-19 cells. The liposome formulations did not decrease the cell viability in vitro. In conclusion, the light-triggered release from the liposomes is functional in the cells, and the release is triggered by thermal phase changes in the lipid bilayers.


Subject(s)
Drug Delivery Systems/methods , Gold/chemistry , Light , Metal Nanoparticles/chemistry , Photochemical Processes , 1,2-Dipalmitoylphosphatidylcholine/adverse effects , 1,2-Dipalmitoylphosphatidylcholine/chemistry , 1,2-Dipalmitoylphosphatidylcholine/radiation effects , Animals , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Dogs , Gold/adverse effects , Gold/radiation effects , Humans , Hydrophobic and Hydrophilic Interactions , Kidney Tubules/cytology , Kidney Tubules/drug effects , Kidney Tubules/radiation effects , Lipid Bilayers/adverse effects , Lipid Bilayers/chemistry , Lipid Bilayers/radiation effects , Liposomes , Metal Nanoparticles/adverse effects , Metal Nanoparticles/radiation effects , Microscopy, Confocal , Phase Transition , Phosphatidylcholines/adverse effects , Phosphatidylcholines/chemistry , Phosphatidylcholines/radiation effects , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/radiation effects , Scattering, Small Angle , Solubility , X-Ray Diffraction
16.
J Virol ; 84(17): 8509-19, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20554778

ABSTRACT

Human parechovirus (HPEV) infections are very common in early childhood and can be severe in neonates. It has been shown that integrins are important for cellular infectivity of HPEV1 through experiments using peptide blocking assays and function-blocking antibodies to alpha(V) integrins. The interaction of HPEV1 with alpha(V) integrins is presumably mediated by a C-terminal RGD motif in the capsid protein VP1. We characterized the binding of integrins alpha(V)beta(3) and alpha(V)beta(6) to HPEV1 by biochemical and structural studies. We showed that although HPEV1 bound efficiently to immobilized integrins, alpha(V)beta(6) bound more efficiently than alpha(V)beta(3) to immobilized HPEV1. Moreover, soluble alpha(V)beta(6), but not alpha(V)beta(3), blocked HPEV1 cellular infectivity, indicating that it is a high-affinity receptor for HPEV1. We also showed that HPEV1 binding to integrins in vitro could be partially blocked by RGD peptides. Using electron cryo-microscopy and image reconstruction, we showed that HPEV1 has the typical T=1 (pseudo T=3) organization of a picornavirus. Complexes of HPEV1 and integrins indicated that both integrin footprints reside between the 5-fold and 3-fold symmetry axes. This result does not match the RGD position predicted from the coxsackievirus A9 X-ray structure but is consistent with the predicted location of this motif in the shorter C terminus found in HPEV1. This first structural characterization of a parechovirus indicates that the differences in receptor binding are due to the amino acid differences in the integrins rather than to significantly different viral footprints.


Subject(s)
Antigens, Neoplasm/metabolism , Integrin alphaVbeta3/metabolism , Integrins/metabolism , Parechovirus/metabolism , Picornaviridae Infections/metabolism , Amino Acid Sequence , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Cell Line , Humans , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/genetics , Integrins/chemistry , Integrins/genetics , Molecular Conformation , Molecular Sequence Data , Parechovirus/chemistry , Parechovirus/genetics , Picornaviridae Infections/virology , Protein Binding , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
17.
J Virol ; 84(10): 4889-97, 2010 May.
Article in English | MEDLINE | ID: mdl-20219926

ABSTRACT

Hantaviruses (family Bunyaviridae) are rodent-borne emerging viruses that cause a serious, worldwide threat to human health. Hantavirus diseases include hemorrhagic fever with renal syndrome and hantavirus cardiopulmonary syndrome. Virions are enveloped and contain a tripartite single-stranded negative-sense RNA genome. Two types of glycoproteins, G(N) and G(C), are embedded in the viral membrane and form protrusions, or "spikes." The membrane encloses a ribonucleoprotein core, which consists of the RNA segments, the nucleocapsid protein, and the RNA-dependent RNA polymerase. Detailed information on hantavirus virion structure and glycoprotein spike composition is scarce. Here, we have studied the structures of Tula hantavirus virions using electron cryomicroscopy and tomography. Three-dimensional density maps show how the hantavirus surface glycoproteins, membrane, and ribonucleoprotein are organized. The structure of the G(N)-G(C) spike complex was solved to 3.6-nm resolution by averaging tomographic subvolumes. Each spike complex is a square-shaped assembly with 4-fold symmetry. Spike complexes formed ordered patches on the viral membrane by means of specific lateral interactions. These interactions may be sufficient for creating membrane curvature during virus budding. In conclusion, the structure and assembly principles of Tula hantavirus exemplify a unique assembly paradigm for enveloped viruses.


Subject(s)
Orthohantavirus/physiology , Virion/ultrastructure , Virus Assembly , Animals , Chlorocebus aethiops , Cryoelectron Microscopy , Electron Microscope Tomography , Imaging, Three-Dimensional , Vero Cells
18.
J Bacteriol ; 191(21): 6701-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19717605

ABSTRACT

The green filamentous bacterium Chloroflexus aurantiacus employs chlorosomes as photosynthetic antennae. Chlorosomes contain bacteriochlorophyll aggregates and are attached to the inner side of a plasma membrane via a protein baseplate. The structure of chlorosomes from C. aurantiacus was investigated by using a combination of cryo-electron microscopy and X-ray diffraction and compared with that of Chlorobi species. Cryo-electron tomography revealed thin chlorosomes for which a distinct crystalline baseplate lattice was visualized in high-resolution projections. The baseplate is present only on one side of the chlorosome, and the lattice dimensions suggest that a dimer of the CsmA protein is the building block. The bacteriochlorophyll aggregates inside the chlorosome are arranged in lamellae, but the spacing is much greater than that in Chlorobi species. A comparison of chlorosomes from different species suggested that the lamellar spacing is proportional to the chain length of the esterifying alcohols. C. aurantiacus chlorosomes accumulate larger quantities of carotenoids under high-light conditions, presumably to provide photoprotection. The wider lamellae allow accommodation of the additional carotenoids and lead to increased disorder within the lamellae.


Subject(s)
Chloroflexus/metabolism , Light-Harvesting Protein Complexes/physiology , Organelles/physiology , Bacterial Chromatophores , Bacteriochlorophylls/physiology , Cell Membrane , Intracellular Membranes , Organelles/ultrastructure , X-Ray Diffraction
19.
Curr Biol ; 19(2): 95-107, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19150238

ABSTRACT

BACKGROUND: Generation of membrane curvature is critical for the formation of plasma membrane protrusions and invaginations and for shaping intracellular organelles. Among the central regulators of membrane dynamics are the BAR superfamily domains, which deform membranes into tubular structures. In contrast to the relatively well characterized BAR and F-BAR domains that promote the formation of plasma membrane invaginations, I-BAR domains induce plasma membrane protrusions through a poorly understood mechanism. RESULTS: We show that I-BAR domains induce strong PI(4,5)P(2) clustering upon membrane binding, bend the membrane through electrostatic interactions, and remain dynamically associated with the inner leaflet of membrane tubules. Thus, I-BAR domains induce the formation of dynamic membrane protrusions to the opposite direction than do BAR and F-BAR domains. Strikingly, comparison of different I-BAR domains revealed that they deform PI(4,5)P(2)-rich membranes through distinct mechanisms. IRSp53 and IRTKS I-BARs bind membranes mainly through electrostatic interactions, whereas MIM and ABBA I-BARs additionally insert an amphipathic helix into the membrane bilayer, resulting in larger tubule diameter in vitro and more efficient filopodia formation in vivo. Furthermore, FRAP analysis revealed that whereas the mammalian I-BAR domains display dynamic association with filopodia, the C. elegans I-BAR domain forms relatively stable structures inside the plasma membrane protrusions. CONCLUSIONS: These data define I-BAR domain as a functional member of the BAR domain superfamily and unravel the mechanisms by which I-BAR domains deform membranes to induce filopodia in cells. Furthermore, our work reveals unexpected divergence in the mechanisms by which evolutionarily distinct groups of I-BAR domains interact with PI(4,5)P(2)-rich membranes.


Subject(s)
Cell Membrane , Membrane Proteins , Protein Conformation , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Cell Line , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Fluorescence Recovery After Photobleaching , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Models, Molecular , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Structure, Tertiary , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Static Electricity
20.
J Mol Biol ; 383(3): 529-38, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-18793644

ABSTRACT

The polymerase complexes of double-stranded RNA (dsRNA) viruses are multifunctional RNA processing machineries that carry out viral genome packaging, replication, and transcription. The polymerase complex forms the innermost virion shell and is structurally related in dsRNA viruses infecting a diversity of host organisms. In this study, we analyzed the properties and functions of the minor polymerase complex protein P7 of dsRNA bacteriophage phi6 using terminally truncated P7 polypeptides and an in vitro self-assembly system established for the phi6 polymerase complex. The N-terminally truncated P7 failed to dimerize, whereas C-terminally truncated P7 polypeptides formed functional dimers that were incorporated into the polymerase complex. Nevertheless, the polymerase complex assembly kinetics and stability were altered by the incorporation of the C-terminally truncated P7. Using the in vitro assembly system for phi6 nucleocapsids and subsequent infectivity assays, we confirmed that full-length P7 is necessary for the formation of infectious viral particles. Contrary to previous results, we found that P7 must be incorporated into polymerase complexes during shell assembly.


Subject(s)
Bacteriophage phi 6/physiology , Capsid Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , Virus Assembly/physiology , Amino Acid Sequence , Animals , Capsid Proteins/chemistry , Capsid Proteins/genetics , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...