Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Comput Biol Chem ; 89: 107400, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33068917

ABSTRACT

A series of alkylated benzimidazole derivatives was synthesized and screened for their anti-HIV, anti-YFV, and broad-spectrum antiviral properties. The physicochemical parameters and drug-like properties of the compounds were assessed first, and then docking studies and MD simulations on HIV-RT allosteric sites were conducted to find the possible mode of their action. DFT analysis was also performed to confirm the nature of the hydrogen bonding interaction of active compounds. The in silico studies indicated that the molecules behaved like possible NNRTIs. The nature - polar or non-polar and position of the substituent present at fifth, sixth, and N-1 positions of the benzimidazole moiety played an important role in determining the antiviral properties of the compounds. Among the various compounds, 2-(5,6-dibromo-2-chloro-1H-benzimidazol-1-yl)ethan-1-ol (3a) showed anti-HIV activity with an appreciably low IC50 value as 0.386 × 10-5µM. Similarly, compound 2b, 3-(2-chloro-5-nitro-1H-benzimidazol-1-yl) propan-1-ol, showed excellent inhibitory property against the yellow fever virus (YFV) with EC50 value as 0.7824 × 10-2µM.


Subject(s)
Benzimidazoles/pharmacology , HIV/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Yellow fever virus/drug effects , Animals , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacokinetics , Catalytic Domain , Chlorocebus aethiops , Density Functional Theory , HIV/enzymology , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/metabolism , Microbial Sensitivity Tests , Models, Chemical , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/pharmacokinetics , Structure-Activity Relationship , Vero Cells , Yellow fever virus/enzymology
2.
Biochim Biophys Acta Gen Subj ; 1864(4): 129521, 2020 04.
Article in English | MEDLINE | ID: mdl-31931019

ABSTRACT

BACKGROUND: The Yellow Fever virus (YFV) is transmitted by mosquitos and causes an infection with symptoms including fever, headaches and nausea. In 20-50% of the cases, the disease may evolve to a visceral stage, reaching high mortality rates. YFV NS2B-NS3 protease has been identified as an important drug target. METHODS: Herein, we describe the crystal structure of the NS2B-NS3 protease from the 2017 YFV Brazilian circulating strain using X-ray crystallography. Furthermore, we used a combination of biochemical and biophysical assays to characterize the enzyme and investigate the impact of the polymorphisms observed in different YFV circulating strains. RESULTS: Surprisingly, the crystal structure of YFV protease seems to adopt the closed conformation without the presence of a binding partner. Although D88E and K121R mutants exhibited a lower affinity for the substrate, both revealed to be more processive, resulting in a similar catalytic efficiency in relation to the WT protease. Still, both mutants showed an accentuated decrease in stability when compared with the WT. CONCLUSIONS: The crystal structure of YFV NS2B-NS3 in closed conformation might be an important tool for the development of new drugs, as well as understanding the activation mechanism of viral proteases. Biochemical analyses indicate that the NS2B-NS3 protease of the circulating strain of YFV is more stable than previous strains. GENERAL SIGNIFICANCE: The YFV NS2B-NS3 protease is the first flaviviral structure described in its closed conformation when in a free form, implying that external factors might induce the activation of the enzyme.


Subject(s)
Polymorphism, Single Nucleotide/genetics , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Yellow fever virus/enzymology , Brazil , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Helicases/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/metabolism
3.
Immunity ; 43(1): 1-2, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26200004

ABSTRACT

The innate immune sensor RIG-I recognizes viral RNA while avoiding unwanted activation by self RNA. In this issue of Immunity, Schuberth-Wagner et al. (2015) show that a histidine residue in the RNA binding pocket of RIG-I sterically excludes the cap1 structure of self RNA, thereby preventing downstream activation.


Subject(s)
DEAD-box RNA Helicases/genetics , Immune Tolerance/genetics , RNA Processing, Post-Transcriptional/genetics , RNA/genetics , Yellow fever virus/enzymology , Animals , Humans
4.
Immunity ; 43(1): 41-51, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26187414

ABSTRACT

The cytosolic helicase retinoic acid-inducible gene-I (RIG-I) initiates immune responses to most RNA viruses by detecting viral 5'-triphosphorylated RNA (pppRNA). Although endogenous mRNA is also 5'-triphosphorylated, backbone modifications and the 5'-ppp-linked methylguanosine ((m7)G) cap prevent immunorecognition. Here we show that the methylation status of endogenous capped mRNA at the 5'-terminal nucleotide (N1) was crucial to prevent RIG-I activation. Moreover, we identified a single conserved amino acid (H830) in the RIG-I RNA binding pocket as the mediator of steric exclusion of N1-2'O-methylated RNA. H830A alteration (RIG-I(H830A)) restored binding of N1-2'O-methylated pppRNA. Consequently, endogenous mRNA activated the RIG-I(H830A) mutant but not wild-type RIG-I. Similarly, knockdown of the endogenous N1-2'O-methyltransferase led to considerable RIG-I stimulation in the absence of exogenous stimuli. Studies involving yellow-fever-virus-encoded 2'O-methyltransferase and RIG-I(H830A) revealed that viruses exploit this mechanism to escape RIG-I. Our data reveal a new role for cap N1-2'O-methylation in RIG-I tolerance of self-RNA.


Subject(s)
DEAD-box RNA Helicases/genetics , Immune Tolerance/genetics , RNA Processing, Post-Transcriptional/genetics , RNA/genetics , Yellow fever virus/enzymology , Amino Acid Sequence , Animals , Cells, Cultured , DEAD Box Protein 58 , Enzyme Activation/genetics , Enzyme Activation/immunology , Histidine/genetics , Humans , Methylation , Methyltransferases/genetics , Mice , Protein Structure, Tertiary , RNA/chemistry , RNA/immunology , RNA, Viral/immunology , Receptors, Immunologic , Yellow fever virus/genetics
5.
Biochem Biophys Res Commun ; 407(4): 640-4, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21419753

ABSTRACT

Here we report the hydrolytic behavior of recombinant YFV NS2B/NS3 protease against FRET substrates mimicking the prime and non-prime region of the natural polyprotein cleavage sites. While the P2-P'1 motif is the main factor associated with the catalytic efficiency of Dengue (DV) and West Nile Virus (WNV) protease, we show that the k(cat)/K(m) of YFV NS2B/NS3 varied by more than two orders of magnitude, despite the presence of the same motif in all natural substrates. The catalytic significance of this homogeneity - a unique feature among worldwide prominent flavivirus - was kinetically analyzed using FRET peptides containing all possible combinations of two and three basic amino acids in tandem, and Arg and Lys residues produced distinct effects on k(cat)/K(m). The parallel of our data with those obtained in vivo by Chambers et al. (1991) restrains the idea that these sites co-evolved with the NS2B/NS3 protease to promote highly efficient hydrolysis and supports the notion that secondary substrate interaction distant from cleavage sites are the main factor associated with the different hydrolytic rates on YFV NS2B-NS3pro natural substrates.


Subject(s)
Viral Nonstructural Proteins/chemistry , Yellow fever virus/enzymology , Amino Acid Motifs , Hydrogen-Ion Concentration , Hydrolysis , Peptides/chemistry , RNA Helicases/chemistry , RNA Helicases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Substrate Specificity , Viral Nonstructural Proteins/genetics
6.
Virus Res ; 141(1): 101-4, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19185594

ABSTRACT

Serine/Threonine phosphorylation of the nonstructural protein 5 (NS5) is a conserved feature of flaviviruses, but the identity and function(s) of the responsible kinase(s) remain unknown. Serine 56 in the methyltransferase domain of NS5 can be phosphorylated intracellularly, is conserved in all flaviviruses, and is a critical residue in the catalytic mechanism. A negative charge at this residue inactivates the 2'-0 methyltransferase activity necessary to form a 5' cap structure of the viral RNA. Here we show pharmacologic inhibition of Casein Kinase 1 (CK1) suppresses yellow fever virus (YFV) production. We also demonstrate the alpha isoform of Casein Kinase 1 (CK1alpha), a kinase previously identified as phosphorylating Hepatitis C Virus NS5A protein, also phosphorylates serine 56 of YFV methyltransferase. Overall these results suggest CK1 activity can influence flaviviral replication.


Subject(s)
Casein Kinase I/metabolism , Flavivirus Infections/enzymology , Methyltransferases/metabolism , Viral Proteins/metabolism , Yellow fever virus/enzymology , Casein Kinase I/chemistry , Casein Kinase I/genetics , Cell Line , Flavivirus/chemistry , Flavivirus/enzymology , Flavivirus/physiology , Humans , Methyltransferases/chemistry , Methyltransferases/genetics , Phosphorylation , Substrate Specificity , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication , Yellow fever virus/chemistry , Yellow fever virus/physiology
7.
J Gen Virol ; 88(Pt 8): 2223-2227, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17622626

ABSTRACT

A recombinant form of yellow fever virus (YFV) NS3 protease, linked via a nonapeptide to the minimal NS2B co-factor sequence (CF40-gly-NS3pro190), was expressed in Escherichia coli and shown to be catalytically active. It efficiently cleaved the fluorogenic tetrapeptide substrate Bz-norleucine-lysine-arginine-arginine-AMC, which was previously optimized for dengue virus NS2B/3 protease. A series of small peptidic inhibitors based on this substrate sequence readily inhibited its enzymic activity. To understand the structure-activity relationship of the inhibitors, they were docked into a homology model of the YFV NS2B/NS3 protease structure. The results revealed that the P1 and P2 positions are most important for inhibitor binding, whilst the P3 and P4 positions have much less effect. These findings indicate that the characteristics of YFV protease are very similar to those reported for dengue and West Nile virus proteases, and suggest that pan-flavivirus NS3 protease drugs may be developed for flaviviral diseases.


Subject(s)
Enzyme Inhibitors/pharmacology , Oligopeptides/metabolism , Viral Nonstructural Proteins/drug effects , Viral Nonstructural Proteins/metabolism , Yellow fever virus/enzymology , Amino Acid Sequence , Antiviral Agents/pharmacology , Binding Sites/physiology , Kinetics , Molecular Sequence Data , Oligopeptides/chemistry , RNA Helicases/chemistry , RNA Helicases/drug effects , RNA Helicases/genetics , RNA Helicases/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Sequence Alignment , Serine Endopeptidases/chemistry , Serine Endopeptidases/drug effects , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Substrate Specificity , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
8.
J Virol ; 79(16): 10268-77, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16051820

ABSTRACT

Yellow fever virus (YFV), a member of the Flavivirus genus, has a plus-sense RNA genome encoding a single polyprotein. Viral protein NS3 includes a protease and a helicase that are essential to virus replication and to RNA capping. The 1.8-A crystal structure of the helicase region of the YFV NS3 protein includes residues 187 to 623. Two familiar helicase domains bind nucleotide in a triphosphate pocket without base recognition, providing a site for nonspecific hydrolysis of nucleoside triphosphates and RNA triphosphate. The third, C-terminal domain has a unique structure and is proposed to function in RNA and protein recognition. The organization of the three domains indicates that cleavage of the viral polyprotein NS3-NS4A junction occurs in trans.


Subject(s)
Flavivirus/enzymology , Viral Nonstructural Proteins/chemistry , West Nile virus/enzymology , Yellow fever virus/enzymology , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Binding Sites , Catalysis , Molecular Sequence Data , Protein Structure, Tertiary , RNA/metabolism , RNA Helicases/chemistry , RNA Helicases/physiology , Serine Endopeptidases/chemistry , Serine Endopeptidases/physiology , Viral Nonstructural Proteins/physiology
9.
J Gen Virol ; 86(Pt 5): 1403-1413, 2005 May.
Article in English | MEDLINE | ID: mdl-15831952

ABSTRACT

A series of 46 charged-to-alanine mutations in the yellow fever virus NS2B-NS3 protease, previously characterized in cell-free and transient cellular expression systems, was tested for their effects on virus recovery. Four distinct plaque phenotypes were observed in cell culture: parental plaque-size (13 mutants), reduced plaque-size (17 mutants), small plaque-size (8 mutants) and no plaque-formation (8 mutants). No mutants displayed any temperature sensitivity based on recovery of virus after RNA transfection at 32 versus 37 degrees C. Most small plaque-mutants were defective in growth efficiency compared with parental virus. However not all small plaque-mutants had defective 2B/3 cleavage, with some showing selective defects at other non-structural protein cleavage sites. Revertant viruses were recovered for six mutations that caused reduced plaque sizes. Same-site and second-site mutations occurred in NS2B, and one second-site mutation occurred in the NS3 protease domain. Some reversion mutations ameliorated defects in cleavage activity and plaque size caused by the original mutation. These data indicate that certain mutations that reduce NS2B-NS3 protease cleavage activity cause growth restriction of yellow fever virus in cell culture. However, for at least two mutations, processing defects other than impaired cleavage activity at the 2B/3 site may account for the mutant phenotype. The existence of reversion mutations primarily in NS2B rather than NS3, suggests that the protease domain is less tolerant of structural perturbation compared with the NS2B protein.


Subject(s)
Amino Acid Substitution , RNA Helicases/genetics , RNA Helicases/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Yellow fever virus/enzymology , DNA Mutational Analysis , Humans , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Tertiary , RNA Helicases/chemistry , Serine Endopeptidases/chemistry , Viral Nonstructural Proteins/chemistry , Viral Plaque Assay , Yellow fever virus/genetics , Yellow fever virus/growth & development
10.
J Virol ; 79(3): 1943-7, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15650220

ABSTRACT

It is not yet clear to what extent depletion of intracellular GTP pools contributes to the antiviral activity of ribavirin. Therefore, the antiviral activities of (i) ribavirin, (ii) its 5-ethynyl analogue, 5-ethynyl-1-beta-D-ribofuranosylimidazole-4-carboxamide (EICAR), and (iii) mycophenolic acid (MPA) (a compound that inhibits only cellular IMP dehydrogenase activity) were studied on the replication of flaviviruses and paramyxoviruses. In addition, the effects of these three compounds on intracellular GTP pools were assessed. A linear correlation was observed over a broad concentration range between the antiviral activities of ribavirin, EICAR, and MPA and the effects of these compounds on GTP pool depletion. When the 50% effective concentrations (EC50s) for the antiviral activities of ribavirin, EICAR, and MPA were plotted against the respective EC50 values for GTP pool depletion, a linear correlation was calculated. These data provide compelling evidence that the predominant mechanism of action of ribavirin in vitro against flavi- and paramyxoviruses is based on inhibition of cellular IMP dehydrogenase activity.


Subject(s)
Antiviral Agents/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Parainfluenza Virus 3, Human/drug effects , Ribavirin/pharmacology , Yellow fever virus/drug effects , Animals , Antiviral Agents/chemistry , Chlorocebus aethiops , Guanosine Triphosphate/metabolism , Microbial Sensitivity Tests/methods , Mycophenolic Acid/chemistry , Mycophenolic Acid/pharmacology , Parainfluenza Virus 3, Human/enzymology , Ribavirin/chemistry , Ribonucleosides/chemistry , Ribonucleosides/pharmacology , Vero Cells , Virus Replication/drug effects , Yellow fever virus/enzymology
11.
J Virol ; 78(2): 1032-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14694136

ABSTRACT

Three consecutive plaque purifications of four chimeric yellow fever virus-dengue virus (ChimeriVax-DEN) vaccine candidates against dengue virus types 1 to 4 were performed. The genome of each candidate was sequenced by the consensus approach after plaque purification and additional passages in cell culture. Our data suggest that the nucleotide sequence error rate for SP6 RNA polymerase used in the in vitro transcription step to initiate virus replication was as high as 1.34 x 10(-4) per copied nucleotide and that the error rate of the yellow fever virus RNA polymerase employed by the chimeras for genome replication in infected cells was as low as 1.9 x 10(-7) to 2.3 x 10(-7). Clustering of beneficial mutations that accumulated after multiple virus passages suggests that the N-terminal part of the prM protein, a specific site in the middle of the E protein, and the NS4B protein may be essential for nucleocapsid-envelope interaction during flavivirus assembly.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Dengue/prevention & control , Dengue Virus/genetics , Sequence Analysis, DNA , Serial Passage , Transcription, Genetic , Vaccines, Synthetic , Viral Plaque Assay , Viral Vaccines , Virus Assembly , Virus Replication , Yellow fever virus/enzymology , Yellow fever virus/genetics
12.
Virology ; 275(2): 335-47, 2000 Sep 30.
Article in English | MEDLINE | ID: mdl-10998334

ABSTRACT

Charged-to-alanine substitutions and deletions within the yellow fever virus NS2B-NS3(181) protease were analyzed for effects on protease function. During cell-free translation of NS2B-3(181) polyproteins, mutations at three charge clusters markedly impaired cis cleavage activity: a single N-terminal cluster in the conserved domain of NS2B (residues ELKK(52-55)) and two in NS3 (ED(21-22), and residue H(47)). These mutations inhibited other protease-dependent cleavages of a transiently expressed nonstructural polyprotein, although differential effects occurred. NS2B and NS3(181) proteins harboring these mutations were impaired in their ability to associate for trans cleavage activity. N-terminal deletions in NS3 also implicated residues ED(21-22) in the association with NS2B. Deletions within NS2B revealed that the conserved domain alone provided minimal cofactor activity, with optimal function requiring both flanking hydrophobic regions. NS2B-3(181)- and NS3(181)-green fluorescent protein fusion proteins were used to determine the intracellular distribution of the protease complex. The former localized in membrane-based vesicular structures, whereas the latter localized poorly. The data suggest that NS2B-NS3 complex formation requires charge interactions involving the N-terminus of the conserved domain of NS2B and 22 N-terminal residues of NS3. A role for the putative transmembrane regions of NS2B in targeting of NS3 to intracellular membranes is also suggested.


Subject(s)
Endopeptidases/metabolism , Viral Nonstructural Proteins/metabolism , Yellow fever virus/metabolism , Amino Acid Sequence , Amino Acid Substitution , Electrophoresis, Polyacrylamide Gel , Endopeptidases/genetics , Fluorescent Antibody Technique , Green Fluorescent Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , RNA Helicases , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Serine Endopeptidases , Viral Nonstructural Proteins/genetics , Yellow fever virus/enzymology
13.
J Virol ; 67(2): 989-96, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8380474

ABSTRACT

The nonstructural protein NS3 of the prototypic flavivirus, yellow fever virus, was investigated for possession of an NTPase activity. The entire NS3 protein coding sequence and an amino-terminal truncated version thereof were engineered into Escherichia coli expression plasmids. Bacteria harboring these plasmids produced the expected polypeptides, which upon cell disruption were found in an insoluble aggregated material considerably enriched for the NS3-related polypeptides. Solubilization and renaturation of these materials, followed by examination of their ability to hydrolyze ATP, revealed an ATPase activity present in both the full-length and amino-terminal truncated NS3 preparations but not in a similarly prepared fraction from E. coli cells engineered to express an unrelated polypeptide. The amino-terminal truncated NS3 polypeptide was further enriched to greater than 95% purity by ion-exchange and affinity chromatography. Throughout the purification scheme, the ATPase activity cochromatographed with the recombinant NS3 polypeptide. The enzymatic activity of the purified material was shown to be a general NTPase and was dramatically stimulated by the presence of particular single-stranded polyribonucleotides. These results are discussed in view of similar activities identified for proteins of other positive-strand RNA viruses.


Subject(s)
Phosphoric Monoester Hydrolases/metabolism , Polynucleotides/pharmacology , Viral Nonstructural Proteins/metabolism , Yellow fever virus/enzymology , Adenosine Triphosphatases/drug effects , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Base Sequence , Cloning, Molecular , Escherichia coli/genetics , Molecular Sequence Data , Nucleoside-Triphosphatase , Phosphoric Monoester Hydrolases/drug effects , Phosphoric Monoester Hydrolases/genetics , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Viral Nonstructural Proteins/drug effects , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/isolation & purification
14.
J Virol ; 65(9): 4749-58, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1651406

ABSTRACT

The proteins of flaviviruses are translated as a single long polyprotein which is co- and posttranslationally processed by both cellular and viral proteinases. We have studied the processing of flavivirus polyproteins in vitro by a viral proteinase located within protein NS3 that cleaves at least three sites within the nonstructural region of the polyprotein, acting primarily autocatalytically. Recombinant polyproteins in which part of the polyprotein is derived from yellow fever virus and part from dengue virus were used. We found that polyproteins containing the yellow fever virus cleavage sites were processed efficiently by the yellow fever virus enzyme, by the dengue virus enzyme, and by various chimeric enzymes. In contrast, dengue virus cleavage sites were cleaved inefficiently by the dengue virus enzyme and not at all by the yellow fever virus enzyme. Studies with chimeric proteinases and with site-directed mutants provided evidence for a direct interaction between the cleavage sites and the proposed substrate-binding pocket of the enzyme. We also found that the efficiency and order of processing could be altered by site-directed mutagenesis of the proposed substrate-binding pocket.


Subject(s)
Endopeptidases/metabolism , Flavivirus/enzymology , Proteins/metabolism , Amino Acid Sequence , Binding Sites , Capsid/metabolism , Cloning, Molecular , DNA Mutational Analysis , Dengue Virus/enzymology , Molecular Sequence Data , Recombinant Fusion Proteins/metabolism , Species Specificity , Structure-Activity Relationship , Substrate Specificity , Viral Core Proteins/metabolism , Viral Nonstructural Proteins , Yellow fever virus/enzymology
15.
Proc Natl Acad Sci U S A ; 87(22): 8898-902, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2147282

ABSTRACT

Sequence homology and molecular modeling studies have suggested that the N-terminal one-third of the flavirvirus nonstructural protein NS3 functions as a trypsin-like serine protease. To examine the putative proteolytic activity of NS3, segments of the yellow fever virus genome were subcloned into plasmid transcription/translation vectors and cell-free translation products were characterized. The results suggest that a protease activity encoded within NS2B and the N-terminal one-third of yellow fever virus NS3 is capable of cis-acting site-specific proteolysis at the NS2B-NS3 cleavage site and dilution-insensitive cleavage of the NS2A-NS2B site. Site-directed mutagenesis of the His-53, Asp-77, and Ser-138 residues of NS3 that compose the proposed catalytic triad implicates this domain as a serine protease. Infectious virus was not recovered from mammalian cells transfected with RNAs transcribed from full-length yellow fever virus cDNA templates containing mutations at Ser-138 (which abolish or dramatically reduce protease activity in vitro), suggesting that the protease is required for viral replication.


Subject(s)
Capsid/metabolism , Serine Endopeptidases/genetics , Viral Core Proteins/metabolism , Yellow fever virus/enzymology , Capsid/genetics , Cloning, Molecular , DNA Mutational Analysis , Genes, Viral , Protein Biosynthesis , Protein Precursors/metabolism , Viral Core Proteins/genetics , Viral Nonstructural Proteins , Viral Proteins/metabolism , Viral Structural Proteins/genetics , Yellow fever virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...