Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 14422, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32879358

ABSTRACT

Alphaviruses are (re-)emerging arboviruses of public health concern. The nsP3 gene product is one of the key players during viral replication. NsP3 comprises three domains: a macro domain, a zinc-binding domain and a hypervariable region. The macro domain is essential at both early and late stages of the replication cycle through ADP-ribose (ADPr) binding and de-ADP-ribosylation of host proteins. However, both its specific role and the precise molecular mechanism of de-ADP-ribosylation across specific viral families remains to be elucidated. Here we investigate by X-ray crystallography the mechanism of ADPr reactivity in the active site of Getah virus macro domain, which displays a peculiar substitution of one of the conserved residues in the catalytic loop. ADPr adopts distinct poses including a covalent bond between the C''1 of the ADPr and a conserved Togaviridae-specific cysteine. These different poses observed for ADPr may represent snapshots of the de-ADP-ribosylation mechanism, highlighting residues to be further characterised.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Alphavirus/enzymology , Molecular Docking Simulation , Viral Nonstructural Proteins/chemistry , Viral Proteases/chemistry , ADP-Ribosylation , Adenosine Diphosphate Ribose/chemistry , Binding Sites , Protein Binding , Viral Nonstructural Proteins/metabolism , Viral Proteases/metabolism
2.
Cells ; 9(4)2020 04 15.
Article in English | MEDLINE | ID: mdl-32326388

ABSTRACT

Alphaviruses can infect a broad range of vertebrate hosts, including birds, horses, primates, and humans, in which infection can lead to rash, fever, encephalitis, and arthralgia or arthritis. They are most often transmitted by mosquitoes in which they establish persistent, asymptomatic infections. Currently, there are no vaccines or antiviral therapies for any alphavirus. Several Old World alphaviruses, including Semliki Forest virus, Ross River virus and chikungunya virus, activate or hyperactivate the phosphatidylinositol-3-kinase (PI3K)-AKT pathway in vertebrate cells, potentially influencing many cellular processes, including survival, proliferation, metabolism and autophagy. Inhibition of PI3K or AKT inhibits replication of several alphaviruses either in vitro or in vivo, indicating the importance for viral replication. In this review, we discuss what is known about the mechanism(s) of activation of the pathway during infection and describe those effects of PI3K-AKT activation which could be of advantage to the alphaviruses. Such knowledge may be useful for the identification and development of therapies.


Subject(s)
Alphavirus/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Alphavirus/metabolism , Animals , Autophagy , Enzyme Activation , Humans
3.
J Virol ; 91(18)2017 09 15.
Article in English | MEDLINE | ID: mdl-28701392

ABSTRACT

Alphaviruses are positive-strand RNA viruses expressing their replicase as a polyprotein, P1234, which is cleaved to four final products, nonstructural proteins nsP1 to nsP4. The replicase proteins together with viral RNA and host factors form membrane invaginations termed spherules, which act as the replication complexes producing progeny RNAs. We have previously shown that the wild-type alphavirus replicase requires a functional RNA template and active polymerase to generate spherule structures. However, we now find that specific partially processed forms of the replicase proteins alone can give rise to membrane invaginations in the absence of RNA or replication. The minimal requirement for spherule formation was the expression of properly cleaved nsP4, together with either uncleaved P123 or with the combination of nsP1 and uncleaved P23. These inactive spherules were morphologically less regular than replication-induced spherules. In the presence of template, nsP1 plus uncleaved P23 plus nsP4 could efficiently assemble active replication spherules producing both negative-sense and positive-sense RNA strands. P23 alone did not have membrane affinity, but could be recruited to membrane sites in the presence of nsP1 and nsP4. These results define the set of viral components required for alphavirus replication complex assembly and suggest the possibility that it could be reconstituted from separately expressed nonstructural proteins.IMPORTANCE All positive-strand RNA viruses extensively modify host cell membranes to serve as efficient platforms for viral RNA replication. Alphaviruses and several other groups induce protective membrane invaginations (spherules) as their genome factories. Most positive-strand viruses produce their replicase as a polyprotein precursor, which is further processed through precise and regulated cleavages. We show here that specific cleavage intermediates of the alphavirus replicase can give rise to spherule structures in the absence of viral RNA. In the presence of template RNA, the same intermediates yield active replication complexes. Thus, partially cleaved replicase proteins play key roles that connect replication complex assembly, membrane deformation, and the different stages of RNA synthesis.


Subject(s)
Alphavirus/enzymology , Host-Pathogen Interactions , Organelle Biogenesis , Protein Multimerization , RNA-Dependent RNA Polymerase/metabolism , Virus Replication , Protein Binding
4.
Virus Res ; 234: 44-57, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28104453

ABSTRACT

Alphaviruses are typically arthropod-borne, and many are important pathogens such as chikungunya virus. Alphaviruses encode four nonstructural proteins (nsP1-4), initially produced as a polyprotein P1234. nsP4 is the core RNA-dependent RNA polymerase but all four nsPs are required for RNA synthesis. The early replication complex (RC) formed by the polyprotein P123 and nsP4 synthesizes minus RNA strands, and the late RC composed of fully processed nsP1-nsP4 is responsible for the production of genomic and subgenomic plus strands. Different parts of nsP4 recognize the promoters for minus and plus strands but the binding also requires the other nsPs. The alphavirus polymerase has been purified and is capable of de novo RNA synthesis only in the presence of the other nsPs. The purified nsP4 also has terminal adenylyltransferase activity, which may generate the poly(A) tail at the 3' end of the genome. Membrane association of the nsPs is vital for replication, and alphaviruses induce membrane invaginations called spherules, which form a microenvironment for RNA synthesis by concentrating replication components and protecting double-stranded RNA intermediates. The RCs isolated as crude membrane preparations are active in RNA synthesis in vitro, but high-resolution structure of the RC has not been achieved, and thus the arrangement of viral and possible host components remains unknown. For some alphaviruses, Ras-GTPase-activating protein (Src-homology 3 (SH3) domain)-binding proteins (G3BPs) and amphiphysins have been shown to be essential for RNA replication and are present in the RCs. Host factors offer an additional target for antivirals, as only few alphavirus polymerase inhibitors have been described.


Subject(s)
Alphavirus/enzymology , Alphavirus/physiology , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/metabolism , Virus Replication , Cell Membrane/enzymology , Cell Membrane/virology , Promoter Regions, Genetic , Protein Binding , Viral Nonstructural Proteins/metabolism
5.
Biosci Rep ; 35(3)2015 Apr 22.
Article in English | MEDLINE | ID: mdl-26182358

ABSTRACT

Alphavirus nsP2 proteins are multifunctional and essential for viral replication. The protease role of nsP2 is critical for virus replication as only the virus protease activity is used for processing of the viral non-structural polypeptide. Chikungunya virus is an emerging disease problem that is becoming a world-wide health issue. We have generated purified recombinant chikungunya virus nsP2 proteins, both full length and a truncated protease domain from the C-terminus of the nsP2 protein. Enzyme characterization shows that the protease domain alone has different properties compared with the full length nsP2 protease. We also show chikungunya nsP2 protease possesses different substrate specificity to the canonical alphavirus nsP2 polyprotein cleavage specificity. Moreover, the chikungunya nsP2 also appears to differ from other alphavirus nsP2 in its distinctive ability to recognize small peptide substrates.


Subject(s)
Chikungunya virus/enzymology , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Alphavirus/enzymology , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/isolation & purification , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Stability , Kinetics , Peptides/metabolism , Protein Domains , Protein Engineering/methods , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Substrate Specificity , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/isolation & purification , Viral Proteins/metabolism
6.
Biol Direct ; 10: 16, 2015 Apr 11.
Article in English | MEDLINE | ID: mdl-25886938

ABSTRACT

BACKGROUND: Members of the alphavirus supergroup include human pathogens such as chikungunya virus, hepatitis E virus and rubella virus. They encode a capping enzyme with methyltransferase-guanylyltransferase (MTase-GTase) activity, which is an attractive drug target owing to its unique mechanism. However, its experimental study has proven very difficult. RESULTS: We examined over 50 genera of viruses by sequence analyses. Earlier studies showed that the MTase-GTase contains a "Core" region conserved in sequence. We show that it is followed by a long extension, which we termed "Iceberg" region, whose secondary structure, but not sequence, is strikingly conserved throughout the alphavirus supergroup. Sequence analyses strongly suggest that the minimal capping domain corresponds to the Core and Iceberg regions combined, which is supported by earlier experimental data. The Iceberg region contains all known membrane association sites that contribute to the assembly of viral replication factories. We predict that it may also contain an overlooked, widely conserved membrane-binding amphipathic helix. Unexpectedly, we detected a sequence homolog of the alphavirus MTase-GTase in taxa related to nodaviruses and to chronic bee paralysis virus. The presence of a capping enzyme in nodaviruses is biologically consistent, since they have capped genomes but replicate in the cytoplasm, where no cellular capping enzyme is present. The putative MTase-GTase domain of nodaviruses also contains membrane-binding sites that may drive the assembly of viral replication factories, revealing an unsuspected parallel with the alphavirus supergroup. CONCLUSIONS: Our work will guide the functional analysis of the alphaviral MTase-GTase and the production of domains for structure determination. The identification of a homologous domain in a simple model system, nodaviruses, which replicate in numerous eukaryotic cell systems (yeast, flies, worms, mammals, and plants), can further help crack the function and structure of the enzyme.


Subject(s)
Alphavirus/genetics , Methyltransferases/genetics , Nucleotidyltransferases/genetics , Viral Proteins/genetics , Alphavirus/enzymology , Antiviral Agents/chemistry , Chikungunya virus/enzymology , Chikungunya virus/genetics , Computational Biology , Gene Deletion , Genes, Viral , Hepatitis E virus/enzymology , Hepatitis E virus/genetics , Methyltransferases/chemistry , Mutation , Nucleotidyltransferases/chemistry , Phylogeny , Protein Structure, Tertiary , Rubella virus/enzymology , Rubella virus/genetics , Sequence Analysis, DNA , Viral Proteins/chemistry , Virus Replication
7.
J Virol ; 88(21): 12242-53, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25100849

ABSTRACT

The alphavirus capsid protein (CP) is a serine protease that possesses cis-proteolytic activity essential for its release from the nascent structural polyprotein. The released CP further participates in viral genome encapsidation and nucleocapsid core formation, followed by its attachment to glycoproteins and virus budding. Thus, protease activity of the alphavirus capsid is a potential antialphaviral target to arrest capsid release, maturation, and structural polyprotein processing. However, the discovery of capsid protease inhibitors has been hampered due to the lack of a suitable screening assay and of the crystal structure in its active form. Here, we report the development of a trans-proteolytic activity assay for Aura virus capsid protease (AVCP) based on fluorescence resonance energy transfer (FRET) for screening protease inhibitors. Kinetic parameters using fluorogenic peptide substrates were estimated, and the K(m) value was found to be 2.63 ± 0.62 µM while the k(cat)/K(m) value was 4.97 × 10(4) M(-1) min(-1). Also, the crystal structure of the trans-active form of AVCP has been determined to 1.81-Å resolution. Structural comparisons of the active form with the crystal structures of available substrate-bound mutant and inactive blocked forms of the capsid protease identify conformational changes in the active site, the oxyanion hole, and the substrate specificity pocket residues, which could be critical for rational drug design. IMPORTANCE The alphavirus capsid protease is an attractive antiviral therapeutic target. In this study, we have described the formerly unappreciated trans-proteolytic activity of the enzyme and for the first time have developed a FRET-based protease assay for screening capsid protease inhibitors. Our structural studies unveil the structural features of the trans-active protease, which has been previously proposed to exist in the natively unfolded form (M. Morillas, H. Eberl, F. H. Allain, R. Glockshuber, and E. Kuennemann, J. Mol. Biol. 376:721-735, 2008, doi:http://dx.doi.org/10.1016/j.jmb.2007.11.055). The different enzymatic forms have been structurally compared to reveal conformational variations in the active and substrate binding sites. The flexible active-site residue Ser218, the disordered C-terminal residues after His261, and the presence of a water molecule in the oxyanion hole of AVCPΔ2 (AVCP with a deletion of the last two residues at the C terminus) reveal the effect of the C-terminal Trp267 deletion on enzyme structure. New structural data reported in this study along with the fluorogenic assay will be useful in substrate specificity characterization, high-throughput protease inhibitor screening, and structure-based development of antiviral drugs.


Subject(s)
Alphavirus/enzymology , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Protease Inhibitors/isolation & purification , Crystallography, X-Ray , Drug Evaluation, Preclinical , Fluorescence Resonance Energy Transfer , Kinetics , Models, Molecular , Protein Conformation
8.
PLoS One ; 7(12): e51288, 2012.
Article in English | MEDLINE | ID: mdl-23251484

ABSTRACT

The nucleocapsid core interaction with endodomains of glycoproteins plays a critical role in the alphavirus life cycle that is essential to virus budding. Recent cryo-electron microscopy (cryo-EM) studies provide structural insights into key interactions between capsid protein (CP) and trans-membrane glycoproteins E1 and E2. CP possesses a chymotrypsin-like fold with a hydrophobic pocket at the surface responsible for interaction with glycoproteins. In the present study, crystal structures of the protease domain of CP from Aura virus and its complex with dioxane were determined at 1.81 and 1.98 Å resolution respectively. Due to the absence of crystal structures, homology models of E1 and E2 from Aura virus were generated. The crystal structure of CP and structural models of E1 and E2 were fitted into the cryo-EM density map of Venezuelan equine encephalitis virus (VEEV) for detailed analysis of CP-glycoprotein interactions. Structural analysis revealed that the E2 endodomain consists of a helix-loop-helix motif where the loop region fits into the hydrophobic pocket of CP. Our studies suggest that Cys397, Cys418 and Tyr401 residues of E2 are involved in stabilizing the structure of E2 endodomain. Density map fitting analysis revealed that Pro405, a conserved E2 residue is present in the loop region of the E2 endodomain helix-loop-helix structure and makes intermolecular hydrophobic contacts with the capsid. In the Aura virus capsid protease (AVCP)-dioxane complex structure, dioxane occupies the hydrophobic pocket on CP and structurally mimics the hydrophobic pyrollidine ring of Pro405 in the loop region of E2.


Subject(s)
Alphavirus/enzymology , Dioxanes/chemistry , Glycoproteins/chemistry , Peptide Hydrolases/chemistry , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid
9.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 11): 1394-8, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22102240

ABSTRACT

The C-terminal protease domain of capsid protein from Aura virus expressed in a bacterial expression system has been purified to homogeneity and crystallized. Crystals suitable for X-ray diffraction analysis were obtained by the vapour-diffusion method using 0.1 M bis-tris and polyethylene glycol monomethyl ether 2000. Crystals of the C-terminal protease domain of capsid protein in complex with dioxane were also produced and crystal data were obtained. Both crystals belonged to space group C2, with unit-cell parameters a = 79.6, b = 35.2, c = 49.5 Å. High-resolution data sets were collected to a resolution of 1.81 Å for the native protein and 1.98 Å for the complex. Preliminary crystallographic studies suggested the presence of a single molecule in the crystallographic asymmetric unit, with a solvent content of 38.5%.


Subject(s)
Alphavirus/enzymology , Capsid/enzymology , Dioxanes/chemistry , Peptide Hydrolases/chemistry , Crystallization , Crystallography, X-Ray , Dioxanes/metabolism
10.
J Mol Graph Model ; 29(1): 46-53, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20483643

ABSTRACT

The alphavirus nsP2 protease is essential for correct processing of the alphavirus nonstructural polyprotein (nsP1234) and replication of the viral genome. We have combined molecular dynamics simulations with our structural studies to reveal features of the nsP2 protease catalytic site and S1'-S4 subsites that regulate the specificity of the protease. The catalytic mechanism of the nsP2 protease appears similar to the papain-like cysteine proteases, with the conserved catalytic dyad forming a thiolate-imidazolium ion pair in the nsP2-activated state. Substrate binding likely stabilizes this ion pair. Analysis of bimolecular complexes of Venezuelan equine encephalitis virus (VEEV) nsP2 protease with each of the nsP1234 cleavage sites identified protease residues His(510), Ser(511), His(546) and Lys(706) as critical for cleavage site recognition. Homology modelling and molecular dynamics simulations of diverse alphaviruses and their cognate cleavage site sequences revealed general features of substrate recognition that operate across alphavirus strains as well as strain specific covariance between binding site and cleavage site residues. For instance, compensatory changes occurred in the P3 and S3 subsite residues to maintain energetically favourable complementary binding surfaces. These results help explain how alphavirus nsP2 proteases recognize different cleavage sites within the nonstructural polyprotein and discriminate between closely related cleavage targets.


Subject(s)
Alphavirus/enzymology , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Biocatalysis , Models, Molecular , Molecular Sequence Data , Structure-Activity Relationship , Substrate Specificity
11.
Antiviral Res ; 87(2): 111-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19616028

ABSTRACT

The alphaviruses were amongst the first arboviruses to be isolated, characterized and assigned a taxonomic status. They are globally very widespread, infecting a large variety of terrestrial animals, insects and even fish, and circulate both in the sylvatic and urban/peri-urban environment, causing considerable human morbidity and mortality. Nevertheless, despite their obvious importance as pathogens, there are currently no effective antiviral drugs with which to treat humans or animals infected by any of these viruses. The EU-supported project-VIZIER (Comparative Structural Genomics of Viral Enzymes Involved in Replication, FP6 PROJECT: 2004-511960) was instigated with an ultimate view of contributing to the development of antiviral therapies for RNA viruses, including the alphaviruses [Coutard, B., Gorbalenya, A.E., Snijder, E.J., Leontovich, A.M., Poupon, A., De Lamballerie, X., Charrel, R., Gould, E.A., Gunther, S., Norder, H., Klempa, B., Bourhy, H., Rohayemj, J., L'hermite, E., Nordlund, P., Stuart, D.I., Owens, R.J., Grimes, J.M., Tuckerm, P.A., Bolognesi, M., Mattevi, A., Coll, M., Jones, T.A., Aqvist, J., Unger, T., Hilgenfeld, R., Bricogne, G., Neyts, J., La Colla, P., Puerstinger, G., Gonzalez, J.P., Leroy, E., Cambillau, C., Romette, J.L., Canard, B., 2008. The VIZIER project: preparedness against pathogenic RNA viruses. Antiviral Res. 78, 37-46]. This review highlights some of the major features of alphaviruses that have been investigated during recent years. After describing their classification, epidemiology and evolutionary history and the expanding geographic distribution of Chikungunya virus, we review progress in understanding the structure and function of alphavirus replicative enzymes achieved under the VIZIER programme and the development of new disease control strategies.


Subject(s)
Alphavirus Infections/epidemiology , Alphavirus Infections/virology , Alphavirus/classification , Alphavirus/pathogenicity , Biomedical Research/trends , Alphavirus/drug effects , Alphavirus/enzymology , Animals , Biomedical Research/organization & administration , Chikungunya virus/pathogenicity , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/virology , Enzymes/chemistry , Enzymes/genetics , Enzymes/metabolism , European Union , Humans , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
12.
Vaccine ; 24(24): 5110-8, 2006 Jun 12.
Article in English | MEDLINE | ID: mdl-16725231

ABSTRACT

The immunogenicity and efficacy of nucleic acid vaccines can be greatly enhanced when antigen production is under the control of an alphaviral replicase enzyme. However, replicase-mediated mRNA overproduction does not necessarily result in enhanced antigen level. Instead, the strong adaptive immune response of alphavirus replicon-based vectors is due to their production of double-stranded RNA (dsRNA) intermediates, which trigger innate immunity. Because viral infections are known to trigger innate immune responses that lead to the rapid production of Type I Interferons (IFNs), namely IFN-alpha and IFN-beta, we investigated the role of Type I IFNs in the enhanced immunogenicity of replicase-based DNA vaccines. In vitro, cells transfected with replicase-based plasmids produce significantly more Type I IFNs than cells transfected with a conventional DNA plasmid. In vivo, replicase-based DNA vaccines yield stronger humoral responses in the absence of Type I IFN signaling but the lack of this signaling pathway in IFN-alphabeta receptor-/- (knockout) mice abolishes T cell mediated efficacy against tumors of both conventional and alphavirus replicase-based DNA vaccines. Moreover, the co-delivery of an IFNalpha-encoding plasmid significantly improved the efficacy of a weakly immunogenic conventional plasmid. These results suggest a central role for Type I IFNs in the mechanism of replicase-based DNA vaccines and indicate that vaccines can be enhanced by enabling their capacity to triggering innate anti-viral defense pathways.


Subject(s)
Alphavirus/enzymology , Interferon Type I/physiology , Melanoma, Experimental/prevention & control , Membrane Glycoproteins/genetics , Oxidoreductases/genetics , Replicon , Vaccines, DNA/immunology , Animals , Immunization , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Oxidoreductases/immunology , Plasmids
14.
J Biol Chem ; 275(23): 17281-7, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10748213

ABSTRACT

Both genomic and subgenomic RNAs of the Alphavirus have m(7)G(5')ppp(5')N (cap0 structure) at their 5' end. Previously it has been shown that Alphavirus-specific nonstructural protein Nsp1 has guanine-7N-methyltransferase and guanylyltransferase activities needed in the synthesis of the cap structure. During normal cap synthesis the 5' gamma-phosphate of the nascent viral RNA chain is removed by a specific RNA 5'-triphosphatase before condensation with GMP, delivered by the guanylyltransferase. Using a novel RNA triphosphatase assay, we show here that nonstructural protein Nsp2 (799 amino acids) of Semliki Forest virus specifically cleaves the gamma,beta-triphosphate bond at the 5' end of RNA. The same activity was demonstrated for Nsp2 of Sindbis virus, as well as for the amino-terminal fragment of Semliki Forest virus Nsp2-N (residues 1-470). The carboxyl-terminal part of Semliki Forest virus Nsp2-C (residues 471-799) had no RNA triphosphatase activity. Replacement of Lys-192 by Asn in the nucleotide-binding site completely abolished RNA triphosphatase and nucleoside triphosphatase activities of Semliki Forest virus Nsp2 and Nsp2-N. Here we provide biochemical characterization of the newly found function of Nsp2 and discuss the unique properties of the entire Alphavirus-capping apparatus.


Subject(s)
Acid Anhydride Hydrolases/metabolism , Alphavirus/enzymology , Cysteine Endopeptidases/metabolism , GTP Phosphohydrolases/metabolism , RNA Caps/metabolism , Semliki forest virus/enzymology , Amino Acid Sequence , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Guanosine Triphosphate/metabolism , Kinetics , Molecular Sequence Data , Recombinant Proteins/metabolism , Substrate Specificity
15.
J Virol ; 71(12): 9713-21, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9371637

ABSTRACT

Intracellular applications of ribozymes have been limited partly by the availability of suitable high-expression systems. For RNA effectors, consideration of an RNA virus vector system for delivery and expression is reasonable. We show that alphavirus replicons can be highly efficient nonintegrating ribozyme-expressing vectors. Using a hammerhead ribozyme targeted to a highly conserved sequence in the U5 region of the human immunodeficiency virus type 1 (HIV-1) long terminal repeat, we demonstrate that a full-length 8.3-kb Semliki Forest virus ribozyme (SFVRz) chimeric RNA maintains catalytic activity. SFVRz is packaged into viral particles, and these particles transduce mammalian cells efficiently. SFVRz-transduced BHK cells were found to produce large amounts of genomic and subgenomic forms of ribozyme-containing RNAs that are functional in cleaving a U5-tagged mRNA. The RNase protection assay shows that HIV-1 U5-chloramphenicol acetyltransferase mRNA expressed intracellularly from an RNA polymerase II promoter is quantitatively eliminated in SFVRz-transduced BHK cells.


Subject(s)
HIV-1/metabolism , RNA, Catalytic/biosynthesis , Replicon , Semliki forest virus/enzymology , Alphavirus/enzymology , Alphavirus/genetics , Animals , CD8 Antigens/biosynthesis , Catalysis , Cell Line , Cricetinae , Gene Expression , Genetic Vectors , Genome, Viral , Humans , RNA, Catalytic/genetics , Subcellular Fractions
16.
Proc Natl Acad Sci U S A ; 92(2): 507-11, 1995 Jan 17.
Article in English | MEDLINE | ID: mdl-7831320

ABSTRACT

After the start of transcription, the 5' ends of eukaryotic mRNA molecules are modified by the addition of a guanylyl residue to form a cap structure, G(5')ppp(5')N. The guanylyltransferase (GTP:mRNA guanylyltransferase, EC 2.7.7.50) reaction responsible for cap formation usually proceeds via a covalent enzyme-GMP intermediate. We have studied the alphavirus-specific guanylyltransferase by incubating lysates from Semliki Forest and Sindbis virus-infected cells with [alpha-32P]GTP, using vaccinia virus and mock-infected cells as controls. One additional 32P-labeled protein was detected in alphavirus-infected cells but only in the presence of S-adenosylmethionine. This protein was identified as the nonstructural protein nsP1. The properties of the covalent enzyme-guanylate complex were studied with Semliki Forest virus nsP1 expressed in recombinant baculovirus-infected cells. S-Adenosylmethionine and divalent cations were required for the complex formation. The reaction was specific for guanylate nucleotides (GTP, dGTP) and was inhibited by pyrophosphate. nsP1 could be labeled with S-adenosyl[methyl-3H]methionine but only under conditions in which the nsP1-guanylate complex was formed. 7-Methyl-GMP was released from the nsP1-guanylate complex by treatment with acid or acidic hydroxylamine. Similar treatment of vaccinia virus capping enzyme released GMP. These findings suggest that in the capping of alphavirus mRNAs the guanine is methylated before linkage to the mRNA molecule.


Subject(s)
Alphavirus/enzymology , Methyltransferases/metabolism , RNA Cap Analogs/metabolism , RNA Caps/biosynthesis , RNA, Viral/metabolism , Cations, Divalent , Cells, Cultured , Diphosphates , Guanosine Triphosphate/metabolism , Methylation , Methyltransferases/chemistry , RNA Cap Analogs/isolation & purification , Recombinant Proteins/metabolism , S-Adenosylmethionine/metabolism , Substrate Specificity
17.
FEBS Lett ; 288(1-2): 201-5, 1991 Aug 19.
Article in English | MEDLINE | ID: mdl-1652473

ABSTRACT

A computer-assisted comparative analysis of the amino acid sequences of (putative) thiol proteases encoded by the genomes of several diverse groups of positive-stranded RNA viruses and distantly related to the family of cellular papain-like proteases is presented. A high level of similarity was detected between the leader protease of foot-and-mouth-disease virus and the protease of murine hepatitis coronavirus which cleaves the N-terminal p28 protein from the polyprotein. Statistically significant alignment of a portion of the rubella virus polyprotein with cellular papain-like proteases was obtained, leading to tentative identification of the papain-like protease as the enzyme mediating processing of the non-structural proteins of this virus. Specific grouping between the sequences of the proteases of alpha-viruses, and poty- and bymoviruses was revealed. It was noted that papain-like proteases of positive-stranded RNA viruses are much more variable both in their sequences and in genomic locations than chymotrypsin-related proteases found in the same virus class. A novel conserved domain of unknown function has also been identified which flanks the papain-like proteases of alpha-, rubi- and coronaviruses.


Subject(s)
Cysteine Endopeptidases/chemistry , Papain/chemistry , RNA Viruses/enzymology , Alphavirus/enzymology , Amino Acid Sequence , Aphthovirus/enzymology , Coronaviridae/enzymology , Molecular Sequence Data , Plant Viruses/enzymology , Protein Sorting Signals/chemistry , Rubella virus/enzymology , Sequence Alignment
18.
J Cell Biol ; 107(6 Pt 1): 2075-86, 1988 Dec.
Article in English | MEDLINE | ID: mdl-2904446

ABSTRACT

Using morphological and cell biological techniques, we have shown that the RNA replicase of Semliki Forest and Sindbis virus (two closely related alphaviruses) is located in complex ribonucleoprotein structures associated with the cytoplasmic surface of modified secondary lysosomes and endosomes. These nucleoprotein complexes often form a bridge between the membrane of the endocytic vacuole and the rough endoplasmic reticulum where the synthesis of the structural proteins of these viruses occurs. The results suggest that these cytopathic vacuoles constitute sites not only for viral RNA synthesis, but also for translation of structural proteins, and for the assembly of nucleocapsids.


Subject(s)
Alphavirus/growth & development , Endosomes/enzymology , Lysosomes/enzymology , RNA Nucleotidyltransferases/metabolism , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/metabolism , Virus Replication , Alphavirus/enzymology , Animals , Cell Compartmentation , Cell Line , Endocytosis , Endosomes/ultrastructure , Fluorescent Antibody Technique , Immunohistochemistry , Lysosomes/ultrastructure , Microscopy, Electron , Precipitin Tests , Ribonucleoproteins/metabolism , Vacuoles/enzymology , Vacuoles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...