Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 493
Filter
1.
Anal Chem ; 74(18): 4653-61, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12349967

ABSTRACT

The formation of a range of precursor ion charge states from a single concentrated and purified charge state, followed by activation of each charge state, is introduced as a means to obtain more protein structural information than is available from dissociation of a single charge state alone. This approach is illustrated using off-resonance collisional activation of the [M + 8H]8+ to [M + 6H]6+ precursor ions of the bacteriophage MS2 viral coat protein following concentration and purification of the [M + 8H]8+ charge state. This range of charge states was selected on the basis of an ion trap collisional activation study of the effects of precursor ion charge state on the dissociation of the [M + 12H]12+ to [M + 5H]5+ ions. Gas-phase ion/ion proton-transfer reactions and the ion parking technique were applied to purify and concentrate selected precursor ion charge states as well as to simplify the product ion spectra. The high-charge-state ions fragment preferentially at the N-terminal side of proline residues while the product ion spectra of the lowest charge states investigated are dominated by C-terminal aspartic acid cleavages. Maximum structural information is obtained by fragmentation of the intermediate-charge states.


Subject(s)
Capsid/chemistry , Amino Acid Sequence , Capsid/isolation & purification , Gases , Ions , Levivirus/chemistry , Mass Spectrometry , Molecular Sequence Data
2.
Article in Chinese | MEDLINE | ID: mdl-12198557

ABSTRACT

Rice ragged stunt oryzavirus (RRSV) is a member of the genus oryzavirus within the family Reoviridae. Its genome consists of ten segments of dsRNA. The functions of all products encoded by these viral genome segments, except one encoded by S9, have not yet been elucidated. In the present study, the ORF of S 8 of RRSV-Philippines isolate was sequenced and expressed in E. coli. The 67 kD product of S8 could be self-cleaved into two fragments with molecular weights of 43 kD and 26 kD. Western blotting indicated that both 67 kD and 43 kD products were major structural proteins of the virus. It was also found that the 67 kD protein could self aggregate into aggregates with higher sedimentation rate in sucrose gradients during centrifugation. Moreover, the self-aggregation process could be accelerated by the complex of S6 product and genome dsRNAs of RRSV. These results suggest that the S8 products, 67 kD or 43 kD, may be the structural components of the viral inner-capsid.


Subject(s)
Capsid/metabolism , Oryza/virology , Plant Viruses/metabolism , RNA Viruses/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western/methods , Capsid/genetics , Capsid/isolation & purification , Gene Expression , Hemiptera , Molecular Sequence Data , Nucleotides/analysis , Open Reading Frames , Plant Viruses/genetics , RNA Viruses/genetics , RNA, Viral , Sequence Analysis, Protein , Sequence Analysis, RNA
3.
Clin Diagn Lab Immunol ; 9(3): 639-48, 2002 May.
Article in English | MEDLINE | ID: mdl-11986273

ABSTRACT

We developed a quantitative enzyme immunoassay (EIA) for antibody to hepatitis E virus (HEV) by using truncated HEV capsid protein expressed in the baculovirus system to improve seroepidemiology, to contribute to hepatitis E diagnosis, and to enable vaccine evaluations. Five antigen lots were characterized; we used a reference antiserum to standardize antigen potency. We defined Walter Reed antibody units (WR U) with a reference antiserum by using the four-parameter logistic model, established other reference pools as assay standards, and determined the conversion factor: 1 WR U/ml = 0.125 World Health Organization unit (WHO U) per ml. The EIA performed consistently; median intra- and inter-test coefficients of variation were 9 and 12%, respectively. The accurate minimum detection limit with serum diluted 1:1,000 was 5.6 WR U/ml; the test could detect reliably a fourfold antibody change. In six people followed from health to onset of hepatitis E, the geometric mean antibody level rose from 7.1 WR U/ml to 1,924.6 WR U/ml. We used the presence of 56- and 180-kDa bands by Western blotting as a confirmatory test and to define true-negative and -positive serum specimens. A receiver-operating characteristics plot identified 30 WR U/ml as an optimum cut-point (sensitivity, 86%; specificity, 89%). The EIA detected antibody more sensitively than a commercially available test. The EIA was transferred to another laboratory, where four operators matched reference laboratory results for a panel of unknowns. Quantitation of antibody to HEV and confirmation of its specificity by Western blotting make HEV serology more meaningful.


Subject(s)
Hepatitis Antibodies/analysis , Hepatitis E/diagnosis , Immunoenzyme Techniques/methods , Capsid/immunology , Capsid/isolation & purification , Hepatitis Antigens/immunology , Hepatitis Antigens/isolation & purification , Hepatitis E/immunology , Hepatitis E/virology , Hepatitis E virus/immunology , Humans , Reagent Kits, Diagnostic , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification , Reproducibility of Results
4.
J Virol ; 76(11): 5667-77, 2002 Jun.
Article in English | MEDLINE | ID: mdl-11991995

ABSTRACT

Virions of human immunodeficiency virus type 1 (HIV-1) and other lentiviruses contain conical cores consisting of a protein shell composed of the viral capsid protein (CA) surrounding an internal viral ribonucleoprotein complex. Although genetic studies have implicated CA in both early and late stages of the virus replication cycle, the mechanism of core disassembly following penetration of target cells remains undefined. Using quantitative assays for analyzing HIV-1 core stability in vitro, we identified point mutations in CA that either reduce or increase the stability of the HIV-1 core without impairing conical core formation in virions. Alterations in core stability resulted in severely attenuated HIV-1 replication and impaired reverse transcription in target cells with only minimal effects on viral DNA synthesis in permeabilized virions in vitro. We conclude that formation of a viral core of optimal stability is a prerequisite for efficient HIV-1 infection and suggest that disassembly of the HIV-1 core is a regulated step in infection that may be an attractive target for pharmacologic intervention.


Subject(s)
HIV-1/physiology , Viral Core Proteins/physiology , Virus Replication/physiology , Capsid/genetics , Capsid/isolation & purification , Cell Line, Transformed , Cells, Cultured , HIV-1/genetics , HIV-1/metabolism , HeLa Cells , Humans , Kinetics , Mutagenesis, Site-Directed , T-Lymphocytes/cytology , Transcription, Genetic , Viral Core Proteins/genetics , Viral Core Proteins/isolation & purification
5.
J Virol ; 76(11): 5784-92, 2002 Jun.
Article in English | MEDLINE | ID: mdl-11992006

ABSTRACT

The emergence of viral escape mutants is usually a highly undesirable phenomenon. This phenomenon is frequently observed in antiviral drug applications for the treatment of viral infections and can undermine long-term therapeutic success. Here, we propose a strategy for evaluating a given antiviral approach in terms of its potential to provoke the appearance of resistant virus mutants. By use of Q beta RNA phage as a model system, the effect of an antiviral gene therapy, i.e., a virus-specific repressor protein expressed by a recombinant Escherichia coli host, was studied over the course of more than 100 generations. In 13 experiments carried out in parallel, 12 phage populations became resistant and 1 became extinct. Sequence analysis revealed that only two distinct phage mutants emerged in the 12 surviving phage populations. For both escape mutants, sequence variations located in the repressor binding site of the viral genomic RNA, which decrease affinity for the repressor protein, conferred resistance to translational repression. The results clearly suggest the feasibility of the proposed strategy for the evaluation of antiviral approaches in terms of their potential to allow resistant mutants to appear. In addition, the strategy proved to be a valuable tool for observing virus-specific molecular targets under the impact of antiviral drugs.


Subject(s)
Allolevivirus/genetics , Capsid/genetics , Evolution, Molecular , Allolevivirus/growth & development , Allolevivirus/physiology , Capsid/isolation & purification , Capsid/metabolism , Gene Expression , Genetic Therapy , Genetic Vectors , Mutagenesis , Plasmids , RNA, Viral/metabolism , Sequence Analysis, RNA
6.
Electrophoresis ; 23(6): 896-902, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11920874

ABSTRACT

In vivo, the icosahedral capsid of human rhinoviruses undergoes well-defined transitions during the infection pathway. Native virus, sedimenting at 150S, is converted to subviral particles with a sedimentation coefficient of 135S, which have lost the innermost capsid protein VP4. Upon release of the genomic RNA empty 80S capsids remain. Similar structural modifications are observed in vitro upon exposure to low pH and/or elevated temperature. Virions are stabilized against these transitions by various antiviral compounds, which bind to a hydrophobic pocket in the capsid protein VP1. Using capillary electrophoresis the kinetics of viral decay in the presence of such hydrophobic drugs was investigated. Assuming first-order kinetics, the increase of the time constant reflects the extent of stabilization. Exposure of the virions to 55 degrees C after presaturation with the antivirals increased the time constants (as compared to native virus) by a factor of 8-30, from a few minutes to several ten minutes. Denaturation of the stabilized capsid gave rise to heterogeneous material rather than to defined subviral particles. This was confirmed by electron microscopy and indicates that the structural modification of the virus follows a kinetically well-defined pathway which is disturbed by the drugs resulting in disorganized disruption of the virion.


Subject(s)
Antiviral Agents/pharmacology , Benzoates/pharmacology , Capsid/drug effects , Isoxazoles/pharmacology , Pyrroles/pharmacology , Rhinovirus/drug effects , Capsid/isolation & purification , Electrophoresis, Capillary/methods , HeLa Cells , Heating , Humans , Isoelectric Point , Kinetics , Rhinovirus/isolation & purification , Temperature , Time Factors
7.
Cell ; 108(2): 283-95, 2002 Jan 25.
Article in English | MEDLINE | ID: mdl-11832217

ABSTRACT

Cell entry by nonenveloped animal viruses requires membrane penetration without membrane fusion. The reovirus penetration agent is the outer-capsid protein, Mu1. The structure of Mu1, complexed with its "protector" protein, Sigma3, and the fit of this Mu1(3)Sigma3(3) heterohexameric complex into the cryoEM image of an intact virion, reveal molecular events essential for viral penetration. Autolytic cleavage divides Mu1 into myristoylated Mu1N and Mu1C. A long hydrophobic pocket can receive the myristoyl group. Dissociation of Mu1N, linked to a major conformational change of the entire Mu1 trimer, must precede myristoyl-group insertion into the cellular membrane. A myristoyl switch, coupling exposure of the fatty acid chain, autolytic cleavage of Mu1N, and long-range molecular rearrangement of Mu1C, thus appears to be part of the penetration mechanism.


Subject(s)
Capsid Proteins , Capsid/chemistry , Protein Structure, Tertiary , Reoviridae/physiology , Amino Acid Sequence , Animals , Binding Sites , Capsid/isolation & purification , Capsid/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Protein Subunits , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/isolation & purification , RNA-Binding Proteins/metabolism , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/isolation & purification , Viral Proteins/metabolism , Virion/chemistry , Virion/ultrastructure
8.
Proc Natl Acad Sci U S A ; 99(3): 1353-8, 2002 Feb 05.
Article in English | MEDLINE | ID: mdl-11805304

ABSTRACT

Pentamer formation by Vp1, the major capsid protein of simian virus 40, requires an interdigitation of structural elements from the Vp1 monomers [Liddington, R. C., Yan, Y., Moulai, J., Sahli, R., Benjamin, T. L. & Harrison, S. C. (1991) Nature (London) 354, 278-284]. Our analyses reveal that disulfide-linked Vp1 homooligomers are present in the simian virus 40-infected cytoplasm and that they are derived from a 41-kDa monomeric intermediate containing an intrachain disulfide bond(s). The 41-kDa species, emerging within 5 min of pulse labeling with [(35)S]methionine, is converted into a 45-kDa, disulfide-free Vp1 monomer and disulfide-bonded dimers through pentamers. The covalent oligomer formation is blocked in the presence of a sulfhydryl-modifying reagent. We propose that there are two stages in this Vp1 disulfide bonding. First, the newly synthesized Vp1 monomers acquire intrachain bonds as they fold and begin to interact. Next, these bonds are replaced with intermolecular bonds as the monomers assemble into pentamers. This sequential appearance of transitory disulfide bonds is consistent with a role for sulfhydryl-disulfide redox reactions in the coordinate folding of Vp1 chains into pentamers. The cytoplasmic Vp1 does not colocalize with marker proteins of the endoplasmic reticulum. This paper demonstrates in vivo disulfide formations and exchanges coupled to the folding and oligomerization of a mammalian protein in the cytoplasm, outside the secretory pathway. Such disulfide dynamics may be a general phenomenon for other cysteine-bearing mammalian proteins that fold in the cytoplasm.


Subject(s)
Capsid Proteins , Capsid/chemistry , Capsid/metabolism , Protein Folding , Simian virus 40/physiology , Animals , Capsid/isolation & purification , Cell Line , Chlorocebus aethiops , Cross-Linking Reagents , Cytoplasm/metabolism , Disulfides , Macromolecular Substances , Methionine/metabolism , Models, Molecular , Oxidation-Reduction , Protein Conformation , Succinimides , Sulfur Radioisotopes
9.
J Biotechnol ; 93(1): 1-14, 2002 Jan 31.
Article in English | MEDLINE | ID: mdl-11690690

ABSTRACT

Aqueous two-phase systems allow for the unequal distribution of proteins and other molecules in water-rich solutions containing phase separating polymers or surfactants. One approach to improve the partitioning properties of recombinant proteins is to produce the proteins as fused to certain peptide tags. However, the rational design of such tags has proven difficult since it involves a compromise between multivariate parameters such as partitioning properties, solvent accessibility and production/secretion efficiency. In this work, a novel approach for the identification of suitable peptide tag extensions has been investigated. Using the principles of selection, rather than design, peptide sequences contributing to an improved partitioning have been identified using phage display technology. A 40 million member phagemid library of random nona-peptides, displayed as fusion to the major coat protein pVIII of the filamentous phage M13, was employed in the selection of top-phase partitioning phage particles in a PEG/sodium phosphate system. After multiple cycles of selection by partitioning, peptides with high frequencies of both tyrosine and proline residues were found to be over represented in selected clones. The identified peptide sequences, or derivatives thereof, were subsequently individually analyzed for their partitioning behavior as displayed on phage, as free synthetic peptides and as genetically fused to a recombinant model target protein. The results showed that novel peptide sequences capable of enhancing top-phase partitioning without interfering with protein production and secretion indeed could be identified for the aqueous two-phase system investigated.


Subject(s)
Capsid Proteins , Peptide Library , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Amino Acid Sequence , Bacteriophage M13/chemistry , Base Sequence , Biotechnology , Capsid/chemistry , Capsid/genetics , Capsid/isolation & purification , DNA, Recombinant/genetics , Drug Design , Escherichia coli/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Protein Engineering , Recombinant Fusion Proteins/genetics , Solutions , Water
10.
Proc Natl Acad Sci U S A ; 98(25): 14631-6, 2001 Dec 04.
Article in English | MEDLINE | ID: mdl-11724937

ABSTRACT

For all retroviruses, the completion of the viral budding process correlates with the activation of the viral protease by an unknown mechanism, and, as the structural (Gag) polyproteins are cleaved by the viral protease, maturation of the immature virus-like particle into an infectious virion. Unlike most retroviruses, the Mason-Pfizer monkey virus Gag polyproteins assemble into immature capsids within the cytoplasm of the cell before the viral budding event. The results reported here describe a unique experimental system in which Mason-Pfizer monkey virus immature capsids are removed from the cell, and the protease is activated in vitro by the addition of a reducing agent. The cleavage of the protease from the precursor form is a primary event, which proceeds with a half time of 14 min, and is followed by authentic processing of the Gag polyproteins. Activity of the viral protease in vitro depends on pH, with an increase in catalytic rates at acidic and neutral pH. The initiation of protease activity within immature capsids in vitro demonstrates that viral protease activity is sensitive to oxidation-reduction conditions, and that the viral protease can be activated in the absence of viral budding.


Subject(s)
Capsid/metabolism , Endopeptidases/metabolism , Mason-Pfizer monkey virus/metabolism , Animals , COS Cells , Capsid/isolation & purification , Capsid/ultrastructure , Enzyme Activation , Gene Products, gag/metabolism , Hydrogen-Ion Concentration , Kinetics , Mason-Pfizer monkey virus/growth & development , Mason-Pfizer monkey virus/ultrastructure , Microscopy, Electron , Protein Precursors/metabolism , Protein Processing, Post-Translational
11.
Nucleic Acids Res ; 29(22): E113, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11713333

ABSTRACT

A method was developed to screen bacteria for synthesis of mutant proteins with altered assembly and solubility properties using bacteriophage MS2 coat protein as a model self-associating protein. Colonies expressing coat protein from a plasmid were covered with an agarose overlay under conditions that caused the lysis of some of the cells in each colony. The proteins thus liberated diffused through the overlay at rates depending on their molecular sizes. After transfer of the proteins to a nitrocellulose membrane, probing with coat protein-specific antiserum revealed spots whose sizes and intensities were related to the aggregation state of coat protein. The method was employed in the isolation of assembly defective mutants and to find soluble variants of an aggregation-prone coat protein mutant.


Subject(s)
Capsid Proteins , Capsid/genetics , Capsid/isolation & purification , RNA-Binding Proteins/genetics , RNA-Binding Proteins/isolation & purification , Amino Acid Substitution , Bacteriophage T7/genetics , Dimerization , Electrophoresis, Agar Gel/methods , Electrophoresis, Polyacrylamide Gel/methods , Escherichia coli/genetics , Escherichia coli/virology , Genetic Vectors/genetics , Models, Molecular , Molecular Structure , Mutation , Promoter Regions, Genetic/genetics , Protein Conformation , Solubility
12.
Biotechnol Bioeng ; 75(4): 451-5, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11668444

ABSTRACT

The release of protein and DNA from nonrecombinant E. coli JM101 and recombinant E. coli HMS174(DE3) expressing L1 (the major viral coat protein of human papillomavirus type 16) as an inclusion body was demonstrated at high cell density (OD(600) = 160). For the nonrecombinant strain, extraction efficiency decreased significantly as cell mass increased, with a high viscosity increase in the postextraction broth. A different dependence on cell concentration was observed for the recombinant strain, with total protein extraction efficiency exceeding 85% for both uninduced and induced cells. Almost complete release of the recombinant L1 protein was achieved at high cell concentration (OD(600) = 80 approximately 160) without the use of reducing agent. This greatly extends the concentration range for chemical extraction.


Subject(s)
Capsid/genetics , Capsid/isolation & purification , Escherichia coli/growth & development , Papillomaviridae/chemistry , Recombinant Proteins/isolation & purification , Biomass , Cell Count , Escherichia coli/virology , Papillomaviridae/genetics , Recombinant Proteins/genetics
13.
Protein Expr Purif ; 23(1): 75-83, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11570848

ABSTRACT

The efficiencies of different procedures for purification of the capsid protein (CA) of Mason-Pfizer monkey virus are compared. Plasmids encoding both wild-type CA and two C-terminally modified sequences of CA suitable for affinity chromatography purification were prepared. CA was expressed in Escherichia coli (i) as a wild-type protein, (ii) C-terminally extended with a six-histidine tag (CA 6His), and (iii) as a protein containing a C-terminal fusion to a viral protease cleavage site followed by a six-histidine tag (CA 6aa6His). Electron microscopy was used for comparison of the resulting proteins, as CA is a structural protein with no enzymatic activity. We have found that these C-terminal fusions dramatically influenced the properties and morphology of structures formed by CA protein in E. coli. The formation of amorphous aggregates of CA was abolished and CA 6His and CA 6aa6His proteins formed organized structures. CA and CA 6aa6His accumulated in bacteria in inclusion bodies as insoluble proteins, CA 6His was found in a soluble form. Both six-histidine-tagged proteins were purified using affinity chromatography under either native (CA 6His) or denaturing (CA 6aa6His) conditions. CA protein was purified under denaturing conditions using gel-filtration chromatography followed by refolding. All proteins were obtained at a purity >98%. Both aforementioned C-terminal extensions led to dramatic changes in behavior of the products and they also affected the tendency to form organized structures within E. coli. We show here that the widely used histidine anchor may significantly alter the properties of the protein of interest.


Subject(s)
Capsid/isolation & purification , Mason-Pfizer monkey virus/chemistry , Binding Sites , Capsid/genetics , Capsid/metabolism , Chromatography, Affinity/methods , Chromatography, Affinity/standards , Cloning, Molecular , Endopeptidases/genetics , Endopeptidases/metabolism , Histidine/pharmacology , Inclusion Bodies/drug effects , Inclusion Bodies/ultrastructure , Microscopy, Electron , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Solubility
14.
J Mol Biol ; 310(5): 1027-37, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11501993

ABSTRACT

In tailed icosahedral bacteriophages the connection between the 5-fold symmetric environment of the portal vertex in the capsid and the 6-fold symmetric phage tail is formed by a complex interface structure. The current study provides the detailed analysis of the assembly and structural organisation of such an interface within a phage having a long tail. The region of the interface assembled as part of the viral capsid (connector) was purified from DNA-filled capsids of the Bacillus subtilis bacteriophage SPP1. It is composed of oligomers of gp6, the SPP1 portal protein, of gp15, and of gp16. The SPP1 connector structure is formed by a mushroom-like portal protein whose cap faces the interior of the viral capsid in intact virions, an annular structure below the stem of the mushroom, and a second narrower annulus that is in direct contact with the helical tail extremity. The layered arrangement correlates to the stacking of gp6, gp15, and gp16 on top of the tail. The gp16 ring is exposed to the virion outside. During SPP1 morphogenesis, gp6 participates in the procapsid assembly reaction, an early step in the assembly pathway, while gp15 and gp16 bind to the capsid portal vertex after viral chromosome encapsidation. gp16 is processed during or after tail attachment to the connector region. The portal protein gp6 has 12-fold cyclical symmetry in the connector structure, whereas assembly-naïve gp6 exhibits 13-fold symmetry. We propose that it is the interaction of gp6 with other viral morphogenetic proteins that drives its assembly into the 12-mer state.


Subject(s)
Bacillus subtilis/virology , Bacteriophages/chemistry , Bacteriophages/ultrastructure , Capsid/metabolism , Microscopy, Immunoelectron , Viral Proteins/metabolism , Viral Tail Proteins/metabolism , Bacteriophages/isolation & purification , Bacteriophages/metabolism , Binding Sites , Capsid/chemistry , Capsid/isolation & purification , Capsid/ultrastructure , Models, Biological , Protein Binding , Protein Structure, Quaternary , Viral Proteins/chemistry , Viral Proteins/isolation & purification , Viral Proteins/ultrastructure , Viral Tail Proteins/chemistry , Viral Tail Proteins/isolation & purification , Viral Tail Proteins/ultrastructure , Virus Assembly
15.
Virology ; 285(2): 302-12, 2001 Jul 05.
Article in English | MEDLINE | ID: mdl-11437664

ABSTRACT

Adenoviruses of serotype Ad3 (subgenus B) use a still-unknown host cell receptor for viral attachment, whereas viruses from all other known subgenera use the coxsackie and adenovirus receptor (CAR). The receptor binding domain (head) of the Ad3 fiber protein has been expressed in Escherichia coli inclusion bodies. After denaturation and renaturation using a rapid dilution method, crystals of trimeric head were obtained. The 1.6 A resolution X-ray structure shows a strict conservation of the beta-sheet scaffold of the protein very similar to the head structures of the CAR-binding serotypes Ad2, Ad5, and Ad12. The conformation of the loops is different, with the exception of the AB loop, which forms the center of the interface in the Ad12-CAR complex structure. The structure explains why a mutation in Ad5 of one residue in the AB loop to glutamic acid, as in Ad3, abrogates binding to CAR. It is possible that the Ad3 receptor binding site is nevertheless situated similar to the CAR binding site, although it cannot be excluded that other regions of the relatively hydrophobic head surface may be used.


Subject(s)
Adenoviruses, Human/chemistry , Capsid Proteins , Capsid/chemistry , Adenoviruses, Human/classification , Adenoviruses, Human/metabolism , Amino Acid Sequence , Capsid/genetics , Capsid/isolation & purification , Capsid/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Crystallography, X-Ray , Gene Expression , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Renaturation , Protein Structure, Secondary , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Serotyping
16.
J Chromatogr A ; 921(1): 39-48, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11461012

ABSTRACT

For cancer gene therapy, a recombinant adenovirus serotype 5 named RPR/INGN201 has been constructed by susbtitution of the E1 region with human tumor suppressor gene p53. The protein components of RPR/INGN201 virions were separated by reversed-phase HPLC and were individually identified by electrospray time-of-flight mass spectrometry and N-terminal sequencing, both on intact proteins and on their proteolytic fragments after trypsin digestion. Twenty-five peptide components of the proteome (including fiber) with greater than 0.25-0.5% contribution to the protein content of the virus were identified and characterized. Fiber was confirmed to be partially glycosylated (both the non-glycosylated and the monoglycosylated states were identified), and two proteins were isolated and identified as phosphorylation derivatives, namely protein V (non-phosphorylated and monophosphorylated) and protein IIIa (mono- and diphosphorylated). This new analytical tool proved to be very useful not only for refining our current knowledge of the polypeptide repertoire of purified infectious virions but also for monitoring and very rapidly identifying structural modifications resulting from changes in the manufacturing process. It was also used successfully for the characterization of various adenoviral constructs.


Subject(s)
Adenoviridae/genetics , Capsid/chemistry , Genetic Therapy , Neoplasms/therapy , Peptides/analysis , Capsid/isolation & purification , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Electrophoresis, Polyacrylamide Gel , Humans , Spectrometry, Mass, Electrospray Ionization
17.
Acta Virol ; 45(1): 61-3, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11394580

ABSTRACT

The coat protein (CP) gene of Prunus necrotic ringspot virus (PNRSV) was cloned into pET 16b vector and expressed in Escherichia coli. CP-enriched fractions were prepared from whole cell lysate by differential centrifugation. The fraction sedimenting at 20,000 x g for 30 mins was used for preparation of a rabbit antiserum to CP. This antiserum had a titer of 1:2048 and reacted in a double-antibody sandwich ELISA (DAS-ELISA).


Subject(s)
Capsid/isolation & purification , Ilarvirus/genetics , Base Sequence , Capsid/genetics , Cloning, Molecular , Genes, Viral , Ilarvirus/isolation & purification , Molecular Sequence Data , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Trees/virology
18.
Virology ; 283(2): 353-7, 2001 May 10.
Article in English | MEDLINE | ID: mdl-11336560

ABSTRACT

The immunogenicity of capsomeres of human papillomavirus type 33 was evaluated in a dose-response analysis. Capsomeres were obtained free of capsids by expression of L1 carrying the single point mutation C427S. Neutralizing antibodies were detected using an in vitro pseudoinfection assay. Capsomeres induced type-specific, neutralizing antibodies in mice even in the absence of adjuvant. The neutralization titers of immune sera raised without adjuvant were 10- to 20-fold lower than those of antisera to virus-like particles, but virtually identical using Freund's adjuvant. These data indicate that capsomeres may substitute for virus-like particles in future vaccines when used with an adjuvant appropriate for human vaccination.


Subject(s)
Antibodies, Viral/biosynthesis , Capsid/immunology , Papillomaviridae/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Capsid/genetics , Capsid/isolation & purification , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Humans , Immunization , Mice , Mice, Inbred BALB C , Neutralization Tests , Papillomaviridae/metabolism , Virion/immunology , Virion/isolation & purification
19.
J Virol ; 75(6): 2879-90, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11222713

ABSTRACT

The capsid of Kaposi's sarcoma-associated herpesvirus (KSHV) was visualized at 24-A resolution by cryoelectron microscopy. Despite limited sequence similarity between corresponding capsid proteins, KSHV has the same T=16 triangulation number and much the same capsid architecture as herpes simplex virus (HSV) and cytomegalovirus (CMV). Its capsomers are hexamers and pentamers of the major capsid protein, forming a shell with a flat, close-packed, inner surface (the "floor") and chimney-like external protrusions. Overlying the floor at trigonal positions are (alpha beta(2)) heterotrimers called triplexes. The floor structure is well conserved over all three viruses, and the most variable capsid features reside on the outer surface, i.e., in the shapes of the protrusions and triplexes, in which KSHV resembles CMV and differs from HSV. Major capsid protein sequences from the three subfamilies have some similarity, which is closer between KSHV and CMV than between either virus and HSV. The triplex proteins are less highly conserved, but sequence analysis identifies relatively conserved tracts. In alphaherpesviruses, the alpha-subunit (VP19c in HSV) has a 100-residue N-terminal extension and an insertion near the C terminus. The small basic capsid protein sequences are highly divergent: whereas the HSV and CMV proteins bind only to hexons, difference mapping suggests that the KSHV protein, ORF65, binds around the tips of both hexons and pentons.


Subject(s)
Capsid Proteins , Capsid/ultrastructure , Cytomegalovirus/ultrastructure , Herpesvirus 1, Human/ultrastructure , Herpesvirus 8, Human/ultrastructure , Amino Acid Sequence , Capsid/genetics , Capsid/isolation & purification , Capsid/metabolism , Cell Line , Cryoelectron Microscopy , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Humans , Molecular Sequence Data , Phylogeny
SELECTION OF CITATIONS
SEARCH DETAIL
...