Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 104(44): 17335-40, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17956988

ABSTRACT

Previous findings have suggested that class IIa histone deacetylases (HDACs) (HDAC4, -5, -7, and -9) are inactive on acetylated substrates, thus differing from class I and IIb enzymes. Here, we present evidence supporting this view and demonstrate that class IIa HDACs are very inefficient enzymes on standard substrates. We identified HDAC inhibitors unable to bind recombinant human HDAC4 while showing inhibition in a typical HDAC4 enzymatic assay, suggesting that the observed activity rather reflects the involvement of endogenous copurified class I HDACs. Moreover, an HDAC4 catalytic domain purified from bacteria was 1,000-fold less active than class I HDACs on standard substrates. A catalytic Tyr is conserved in all HDACs except for vertebrate class IIa enzymes where it is replaced by His. Given the high structural conservation of HDAC active sites, we predicted the class IIa His-Nepsilon2 to be too far away to functionally substitute the class I Tyr-OH in catalysis. Consistently, a Tyr-to-His mutation in class I HDACs severely reduced their activity. More importantly, a His-976-Tyr mutation in HDAC4 produced an enzyme with a catalytic efficiency 1,000-fold higher than WT, and this "gain of function phenotype" could be extended to HDAC5 and -7. We also identified trifluoroacetyl-lysine as a class IIa-specific substrate in vitro. Hence, vertebrate class IIa HDACs may have evolved to maintain low basal activities on acetyl-lysines and to efficiently process restricted sets of specific, still undiscovered natural substrates.


Subject(s)
Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Vertebrates , Amino Acid Sequence , Animals , Binding Sites , Catalysis , Enzyme Activation , HeLa Cells , Histidine/genetics , Histidine/metabolism , Histone Deacetylases/classification , Histone Deacetylases/genetics , Humans , Models, Molecular , Mutation/genetics , Protein Structure, Tertiary , Substrate Specificity , Urochordata , Vertebrates/genetics
2.
Nat Med ; 12(10): 1147-50, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16980968

ABSTRACT

Pharmacological interventions that increase myofiber size counter the functional decline of dystrophic muscles. We show that deacetylase inhibitors increase the size of myofibers in dystrophin-deficient (MDX) and alpha-sarcoglycan (alpha-SG)-deficient mice by inducing the expression of the myostatin antagonist follistatin in satellite cells. Deacetylase inhibitor treatment conferred on dystrophic muscles resistance to contraction-coupled degeneration and alleviated both morphological and functional consequences of the primary genetic defect. These results provide a rationale for using deacetylase inhibitors in the pharmacological therapy of muscular dystrophies.


Subject(s)
Enzyme Inhibitors/pharmacology , Muscles/enzymology , Muscles/pathology , Muscular Dystrophy, Animal/drug therapy , Animals , Dystrophin/genetics , Fibrosis/pathology , Follistatin/metabolism , Hydroxamic Acids/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscles/drug effects , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Phenylbutyrates/pharmacology , Sarcoglycans/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/enzymology , Valproic Acid/pharmacology
3.
J Virol ; 75(20): 9939-46, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11559826

ABSTRACT

The NS2-NS3 region of the hepatitis C virus polyprotein encodes a proteolytic activity that is required for processing of the NS2/3 junction. Membrane association of NS2 and the autocatalytic nature of the NS2/3 processing event have so far constituted hurdles to the detailed investigation of this reaction. We now report the first biochemical characterization of the self-processing activity of a purified NS2/3 precursor. Using multiple sequence alignments, we were able to define a minimal domain, devoid of membrane-anchoring sequences, which was still capable of performing the processing reaction. This truncated protein was efficiently expressed and processed in Escherichia coli. The processing reaction could be significantly suppressed by growth in minimal medium in the absence of added zinc ions, leading to the accumulation of an unprocessed precursor protein in inclusion bodies. This protein was purified to homogeneity, refolded, and shown to undergo processing at the authentic NS2/NS3 cleavage site with rates comparable to those observed using an in vitro-translated full-length NS2/3 precursor. Size-exclusion chromatography and a dependence of the processing rate on the concentration of truncated NS2/3 suggested a functional multimerization of the precursor protein. However, we were unable to observe trans cleavage activity between cleavage-site mutants and active-site mutants. Furthermore, the cleavage reaction of the wild-type protein was not inhibited by addition of a mutant that was unable to undergo self-processing. Site-directed mutagenesis data and the independence of the processing rate from the nature of the added metal ion argue in favor of NS2/3 being a cysteine protease having Cys993 and His952 as a catalytic dyad. We conclude that a purified protein can efficiently reproduce processing at the NS2/3 site in the absence of additional cofactors.


Subject(s)
Hepacivirus/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Culture Media , Dimerization , Escherichia coli/genetics , Hepacivirus/genetics , Inclusion Bodies/metabolism , Molecular Sequence Data , Protein Precursors/genetics , Protein Precursors/isolation & purification , Recombinant Proteins/biosynthesis , Sequence Alignment , Sequence Analysis, Protein , Viral Nonstructural Proteins/genetics , Zinc
4.
Curr Med Chem ; 8(8): 919-932, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11375758

ABSTRACT

Hepatitis C is a predominantly chronic viral infection, affecting 1-3% of the world population. The causative agent, the hepatitis C virus (HCV), has a positive strand-RNA genome that is utilized, in infected cells, as an mRNA to drive the synthesis of a large polyprotein precursor. This precursor subsequently undergoes proteolytic maturation to generate all of the functional, both structural and nonstructural proteins necessary for viral replication and assembly. The proteolytic activity that is responsible for the generation of the mature viral polymerase as well as for most of the cleavages occurring in the nonstructural region of the polyprotein is expressed by the virus itself and is contained in its nonstructural protein 3 (NS3). Here, the N-terminal 180 amino acids form a chymotrypsin-like serine protease domain. Full activation of this protease is achieved only after complexation with another viral protein, the cofactor protein NS4A. Together, NS3 and NS4A form the active, heterodimeric serine protease that presently is the target of medicinal chemistry efforts aiming at the development of inhibitors with potential antiviral activity. We here review the recent progress in our understanding of the structure and function of the enzyme and in the development of selective and potent NS3 protease inhibitors.


Subject(s)
Serine Proteinase Inhibitors/chemistry , Binding Sites , Forecasting , Hepacivirus/drug effects , Hepacivirus/enzymology , Humans , Models, Molecular , Molecular Conformation , Protein Conformation , Serine Proteinase Inhibitors/pharmacology , Viral Nonstructural Proteins/chemistry
5.
Biochemistry ; 40(3): 631-40, 2001 Jan 23.
Article in English | MEDLINE | ID: mdl-11170379

ABSTRACT

Maturational cleavage of the hepatitis C virus polyprotein involves the viral chymotrypsin-like serine protease NS3. The substrate binding site of this enzyme is unusually flat and featureless. We here show that NS3 has a highly asymmetric charge distribution that is characterized by strong positive potentials in the vicinity of its active site and in the S5/S6 region. Using electrostatic potential calculations, we identified determinants of this positive potential, and the role of six different residues was explored by site-directed mutagenesis. Mutation of residues in the vicinity of the active site led to changes in k(cat) values of a peptide substrate indicating that basic amino acids play a role in the stabilization of the transition state. Charge neutralization in the S5/S6 region increased the K(m) values of peptide substrates in a manner that depended on the presence of negatively charged residues in the P5 and P6 positions. K(i) values of hexapeptide acids spanning P6-P1 (product inhibitors) were affected by charge neutralization in both the active site region and the S5/S6 region. Pre-steady-state kinetic data showed that the electrostatic surface potential is used by this enzyme to enhance collision rates between peptidic ligands and the active site. Calculations of the interaction energies of protease-substrate or protease-inhibitor complexes showed that electrostatic interaction energies oppose the formation of a tightly bound complex due to an unfavorable change in the desolvation energy. We propose that desolvation costs are minimized by avoiding the formation of individual ion pair interactions through the use of clusters of positively charged residues in the generation of local electrostatic potentials.


Subject(s)
Catalytic Domain , Hepacivirus/enzymology , Serine Endopeptidases/chemistry , Viral Nonstructural Proteins/chemistry , Amino Acid Sequence , Arginine/genetics , Catalytic Domain/genetics , Enzyme Stability/genetics , Hepacivirus/genetics , Kinetics , Lysine/genetics , Macromolecular Substances , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptides/chemistry , Protein Binding/genetics , Serine/genetics , Serine Proteinase Inhibitors/chemistry , Static Electricity , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics
6.
Biochemistry ; 39(42): 12898-906, 2000 Oct 24.
Article in English | MEDLINE | ID: mdl-11041854

ABSTRACT

Infection by Hepatitis C Virus (HCV) leads to a slowly progressing disease that over two decades can lead to liver cirrhosis or liver cancer. Currently, one of the most promising approaches to anti-HCV therapy is the development of inhibitors of the NS3/4A protease, which is essential for maturation of the viral polyprotein. Several substrate-derived inhibitors of NS3/4A have been described, all taking advantage of binding to the S subsite of the enzyme. Inspection of the S' subsite of NS3/4A shows binding pockets which might be exploited for inhibitor binding, but due to the fact that ground-state binding to the S' subsite is not used by the substrate, this does not represent a suitable starting point. We have now optimized S'-binding in the context of noncleavable decapeptides spanning P6-P4'. Binding was sequentially increased by introduction of the previously optimized P-region [Ingallinella et al. (1998) Biochemistry 37, 8906-8914], change of the P4' residue, and combinatorial optimization of positions P2'-P3'. The overall process led to an increase in binding of more than 3 orders of magnitude, with the best decapeptide showing IC(50) < 200 pM. The binding mode of the decapeptides described in the present work shares features with the binding mode of the natural substrates, together with novel interactions within the S' subsite. Therefore, these peptides may represent an entry point for a novel class of NS3 inhibitors.


Subject(s)
Hepacivirus/enzymology , Serine Proteinase Inhibitors/chemical synthesis , Viral Nonstructural Proteins/antagonists & inhibitors , Amino Acids/genetics , Binding, Competitive/genetics , Combinatorial Chemistry Techniques , Drug Design , Hepacivirus/drug effects , Hydrolysis , Models, Chemical , Mutagenesis, Site-Directed , Oligopeptides/chemical synthesis , Oligopeptides/genetics , Oligopeptides/metabolism , Peptide Fragments/chemical synthesis , Peptide Fragments/genetics , Peptide Fragments/metabolism , Serine Proteinase Inhibitors/metabolism , Static Electricity , Structure-Activity Relationship , Substrate Specificity/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
7.
J Virol ; 74(22): 10563-70, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11044100

ABSTRACT

Identifying protease cleavage sites contributes to our understanding of their specificity and biochemical properties and can help in designing specific inhibitors. One route to this end is the generation and screening of random libraries of cleavage sites. Both synthetic and phage-displayed libraries have been extensively used in vitro. We describe a novel system based on recombinant Sindbis virus which can be used to identify cleavage sites in vivo, thus eliminating the need for a purified enzyme and overcoming the problem of choosing the correct in vitro conditions. As a model we used the serine protease of the hepatitis C virus (HCV). We engineered the gene coding for this enzyme and two specific cleavage sites in the Sindbis virus structural gene and constructed libraries of viral genomes with a random sequence at either of the cleavage sites. The system was designed so that only viral genomes coding for sequences cleaved by the protease would produce viable viruses. With this system we selected viruses containing sequences mirroring those of the natural HCV protease substrates which were cleaved with comparable efficiencies.


Subject(s)
Hepacivirus/genetics , Peptide Library , Serine Endopeptidases/metabolism , Sindbis Virus/genetics , Antibodies/immunology , Hepacivirus/enzymology , Immunoblotting , Protein Biosynthesis , RNA, Viral , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Analysis, DNA , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Sindbis Virus/metabolism , Substrate Specificity
8.
Semin Liver Dis ; 20(1): 69-83, 2000.
Article in English | MEDLINE | ID: mdl-10895433

ABSTRACT

Infection with the hepatitis C virus (HCV) is the major cause of non-A, non-B hepatitis worldwide. The viral genome, a positive-sense, single-stranded, 9.6-kb long RNA molecule, is translated into a single polyprotein of about 3,000 amino acids. The viral polyprotein is proteoytically processed to yield all the mature viral gene products. The genomic order of HCV has been determined to be C-->E1-->E2-->p7-->NS2-->NS3-->NS4A-->NS4B-->NS5A++ +-->NS5B. C, E1, and E2 are the virion structural proteins. Whereas the function of p7 is currently unknown, NS2 to NS5B are thought to be the nonstructural proteins. Generation of the mature nonstructural proteins relies on the activity of viral proteinases. Cleavage at the NS2-NS3 junction is accomplished by a metal-dependent autocatalytic proteinase encoded within NS2 and the N-terminus of NS3. The remaining downstream cleavages are effected by a serine proteinase contained also within the N-terminal region of NS3. NS3, in addition, contains an RNA helicase domain at its C-terminus. NS3 forms a heterodimeric complex with NS4A. The latter is a membrane protein that acts as a cofactor of the proteinase. Although no function has yet been attributed to NS4B, NS5A has been recently suggested to be involved in mediating the resistance of the HCV to the action of interferon. Finally, the NS5B protein has been shown to be the viral RNA-dependent RNA polymerase. This article reviews the current understanding of the structure and the function of the various HCV nonstructural proteins with particular emphasis on their potential as targets for the development of novel antiviral agents and vaccines.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/immunology , Viral Hepatitis Vaccines/pharmacology , Viral Nonstructural Proteins/immunology , Viral Proteins/immunology , Hepacivirus/drug effects , Humans , RNA, Viral/physiology , Sensitivity and Specificity , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/drug effects , Viral Proteins/chemistry , Viral Proteins/drug effects
9.
J Biol Chem ; 275(20): 15106-13, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10809747

ABSTRACT

A serine protease domain contained within the viral NS3 protein is a key player in the maturational processing of the hepatitis C virus polyprotein and a prime target for the development of antiviral drugs. In the present work, we describe a dansylated hexapeptide inhibitor of this enzyme. Active site occupancy by this compound could be monitored following fluorescence resonance energy transfer between the dansyl fluorophore and protein tryptophan residues and could be used to 1) unambiguously assess active site binding of NS3 protease inhibitors, 2) directly determine equilibrium and pre-steady-state parameters of enzyme-inhibitor complex formation, and 3) dissect, using site-directed mutagenesis, the contribution of single residues of NS3 to inhibitor binding in direct binding assays. The assay was also used to characterize the inhibition of the NS3 protease by its cleavage products. We show that enzyme-product inhibitor complex formation depends on the presence of an NS4A cofactor peptide. Equilibrium and pre-steady-state data support an ordered mechanism of ternary (enzyme-inhibitor-cofactor) complex formation, requiring cofactor complexation prior to inhibitor binding.


Subject(s)
Hepacivirus/enzymology , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemistry , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Binding Sites , Dansyl Compounds , Energy Transfer , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligopeptides/chemistry , Oligopeptides/metabolism , Oligopeptides/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serine Proteinase Inhibitors/metabolism , Serine Proteinase Inhibitors/pharmacology , Spectrometry, Fluorescence , Substrate Specificity , Tryptophan
10.
Biochemistry ; 39(7): 1849-61, 2000 Feb 22.
Article in English | MEDLINE | ID: mdl-10677236

ABSTRACT

The replication of the hepatitis C virus (HCV), an important human pathogen, crucially depends on the proteolytic maturation of a large viral polyprotein precursor. The viral nonstructural protein 3 (NS3) harbors a serine protease domain that plays a pivotal role in this process, being responsible for four out of the five cleavage events that occur in the nonstructural region of the HCV polyprotein. We here show that hexapeptide, tetrapeptide, and tripeptide alpha-ketoacids are potent, slow binding inhibitors of this enzyme. Their mechanism of inhibition involves the rapid formation of a noncovalent collision complex in a diffusion-limited, electrostatically driven association reaction followed by a slow isomerization step resulting in a very tight complex. pH dependence experiments point to the protonated catalytic His 57 as an important determinant for formation of the collision complex. K(i) values of the collision complexes vary between 3 nM and 18.5 microM and largely depend on contacts made by the peptide moiety of the inhibitors. Site-directed mutagenesis indicates that Lys 136 selectively participates in stabilization of the tight complex but not of the collision complex. A significant solvent isotope effect on the isomerization rate constant is suggestive of a chemical step being rate limiting for tight complex formation. The potency of these compounds is dominated by their slow dissociation rate constants, leading to complex half-lives of 11-48 h and overall K(i) values between 10 pM and 67 nM. The rate constants describing the formation and the dissociation of the tight complex are relatively independent of the peptide moiety and appear to predominantly reflect the intrinsic chemical reactivity of the ketoacid function.


Subject(s)
Hepacivirus/enzymology , Keto Acids/chemistry , Oligopeptides/chemistry , Serine Endopeptidases/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Alanine/analogs & derivatives , Alanine/chemistry , Aminobutyrates/chemistry , Binding Sites , Humans , Inhibitory Concentration 50 , Keto Acids/metabolism , Kinetics , Nuclear Magnetic Resonance, Biomolecular , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Serine Endopeptidases/metabolism , Spectrometry, Fluorescence , Structure-Activity Relationship , Viral Nonstructural Proteins/metabolism
12.
Protein Eng ; 12(11): 1005-11, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10585507

ABSTRACT

We have been interested for some time in establishing a strategy for deriving lead compounds from macromolecule ligands such as minibody variants. A minibody is a minimized antibody variable domain whose two loops are amenable to combinatorial mutagenesis. This approach can be especially useful when dealing with 'difficult' targets. One such target is the NS3 protease of hepatitis C virus (HCV), a human pathogen that is believed to infect about 100 million individuals worldwide and for which an effective therapy is not yet available. Based on known inhibitor specificity (residues P6-P1) of NS3 protease, we screened a number of minibodies from our collection and we were able to identify a competitive inhibitor of this enzyme. We thus validated an aspect of recognition by HCV NS3 protease, namely that an acid anchor is necessary for inhibitor activity. In addition, the characterization of the minibody inhibitor led to the synthesis of a constrained hexapeptide mimicking the bioactive loop of the parent macromolecule. The cyclic peptide is a lead compound prone to rapid optimization through solid phase combinatorial chemistry. We therefore confirmed that the potential of turning a protein ligand into a low molecular weight active compound for lead discovery is achievable and can complement more traditional drug discovery approaches.


Subject(s)
Enzyme Inhibitors/chemistry , Hepacivirus/enzymology , Immunoglobulin Variable Region/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Binding Sites , Binding, Competitive , Combinatorial Chemistry Techniques , Enzyme Inhibitors/immunology , Hepacivirus/immunology , Immunoglobulin Variable Region/pharmacology , Kinetics , Models, Molecular , Peptides, Cyclic/chemistry , Peptides, Cyclic/immunology , Peptides, Cyclic/pharmacology , Recombinant Proteins/chemistry , Viral Nonstructural Proteins/immunology
13.
Protein Sci ; 8(7): 1445-54, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10422832

ABSTRACT

Conformational changes occurring within the NS3 protease domain from the hepatitis C virus Bk strain (NS3(1-180)) under different physico-chemical conditions either in the absence or in the presence of its cofactor Pep4A were investigated by limited proteolysis experiments. Because the surface accessibility of the protein is affected by conformational changes, when comparative experiments were carried out on NS3(1-180) either at different glycerol concentrations or in the presence of Pep4A, differential peptide maps were obtained from which protein regions involved in the structural changes could be inferred. The surface topology of isolated NS3(1-180) in solution was essentially consistent with the crystal structure of the protein with the N-terminal segment showing a high conformational flexibility. At higher glycerol concentration, the protease assumed a more compact structure showing a decrease in the accessibility of the N-terminal segment that either was forced to interact with the protein or originate intermolecular interactions with neighboring molecules. Binding of the cofactor Pep4A caused the displacement of the N-terminal arm from the protein moiety, leading this segment to again adopt an open and flexible conformation, thus suggesting that the N-terminus of the protease contributes only marginally to the stability of the complex. The observed conformational changes might be directly correlated with the activation mechanism of the protease by either the cosolvent or the cofactor peptide because they lead to tighter packing of the substrate binding site.


Subject(s)
Hepacivirus/enzymology , Viral Nonstructural Proteins/chemistry , Amino Acid Sequence , Glycerol/chemistry , Hydrolysis , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Peptide Mapping , Protein Conformation
14.
J Mol Biol ; 289(2): 371-84, 1999 Jun 04.
Article in English | MEDLINE | ID: mdl-10366511

ABSTRACT

The solution structure of the hepatitis C virus (BK strain) NS3 protein N-terminal domain (186 residues) has been solved by NMR spectroscopy. The protein is a serine protease with a chymotrypsin-type fold, and is involved in the maturation of the viral polyprotein. Despite the knowledge that its activity is enhanced by the action of a viral protein cofactor, NS4A, the mechanism of activation is not yet clear. The analysis of the folding in solution and the differences from the crystallographic structures allow the formulation of a model in which, in addition to the NS4A cofactor, the substrate plays an important role in the activation of the catalytic mechanism. A unique structural feature is the presence of a zinc-binding site exposed on the surface, subject to a slow conformational exchange process.


Subject(s)
Hepacivirus/enzymology , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Catalysis , Computer Graphics , Conserved Sequence , Crystallography, X-Ray , Enzyme Activation , Escherichia coli , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Solutions , Thermodynamics
15.
J Mol Biol ; 289(2): 385-96, 1999 Jun 04.
Article in English | MEDLINE | ID: mdl-10366512

ABSTRACT

The interactions of peptide inhibitors, obtained by the optimization of N-terminal cleavage products of natural substrates, with the protease of human hepatitis C virus (HCV) are characterized by NMR and modelling studies. The S-binding region of the enzyme and the bound conformation of the ligands are experimentally determined. The NMR data are then used as the experimental basis for modelling studies of the structure of the complex. The S-binding region involves the loop connecting strands E2 and F2, and appears shallow and solvent-exposed. The ligand binds in an extended conformation, forming an antiparallel beta-sheet with strand E2 of the protein, with the P1 carboxylate group in the oxyanion hole.


Subject(s)
Hepacivirus/enzymology , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Binding Sites , Humans , Ligands , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Oligopeptides/chemistry , Oligopeptides/pharmacology , Protein Conformation , Protein Structure, Secondary , Serine Proteinase Inhibitors/pharmacology , Solutions , Viral Nonstructural Proteins/antagonists & inhibitors
16.
Biochemistry ; 38(17): 5620-32, 1999 Apr 27.
Article in English | MEDLINE | ID: mdl-10220351

ABSTRACT

The hepatitis C virus nonstructural 3 protein (NS3) possesses a serine protease activity in the N-terminal one-third, whereas RNA-stimulated NTPase and helicase activities reside in the C-terminal portion. The serine protease activity is required for proteolytic processing at the NS3-NS4A, NS4A-NS4B, NS4B-NS5A, and NS5A-NS5B polyprotein cleavage sites. NS3 forms a complex with NS4A, a 54-residue polypeptide that was shown to act as an essential cofactor of the NS3 protease. We have expressed in Escherichia coli the NS3-NS4A precursor; cleavage at the junction between NS3 and NS4A occurs during expression in the bacteria cells, resulting in the formation of a soluble noncovalent complex with a sub-nanomolar dissociation constant. We have assessed the minimal ionic strength and detergent and glycerol concentrations required for maximal proteolytic activity and stability of the purified NS3-NS4A complex. Using a peptide substrate derived from the NS5A-NS5B junction, the catalytic efficiency (kcat/Km) of NS3-NS4A-associated protease under optimized conditions was 55 000 s-1 M-1, very similar to that measured with a recombinant complex purified from eukaryotic cells. Dissociation of the NS3-NS4A complex was found to be fully reversible. No helicase activity was exhibited by the purified NS3-NS4A complex, but NS3 was fully active as a helicase upon dissociation of NS4A. On the other hand, both basal and poly(U)-induced NTPase activity and ssRNA binding activity associated with the NS3-NS4A complex were very similar to those exhibited by NS3 alone. Therefore, NS4A appears to uncouple the ATPase/ssRNA binding and RNA unwinding activities associated with NS3.


Subject(s)
DNA Helicases/metabolism , Hepacivirus/enzymology , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/metabolism , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Binding Sites , Enzyme Activation/genetics , Escherichia coli/genetics , Kinetics , Macromolecular Substances , Molecular Sequence Data , Protein Biosynthesis , RNA, Viral/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Serine Endopeptidases/genetics , Substrate Specificity , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/isolation & purification
17.
Biochemistry ; 38(16): 5206-15, 1999 Apr 20.
Article in English | MEDLINE | ID: mdl-10213628

ABSTRACT

The interaction of the hepatitis C virus (HCV) NS3 protease domain with its NS4A cofactor peptide (Pep4AK) was investigated at equilibrium and at pre-steady state under different physicochemical conditions. Equilibrium dissociation constants of the NS3-Pep4AK complex varied by several orders of magnitude depending on buffer additives. Glycerol, NaCl, detergents, and peptide substrates were found to stabilize this interaction. The extent of glycerol-induced stabilization varied in an HCV strain-dependent way with at least one determinant mapping to an NS3-NS4A interaction site. Conformational transitions affecting at least the first 18 amino acids of NS3 were the main energy barriers for both the association and the dissociation reactions of the complex. However, deletion of this N-terminal portion of the protease molecule only slightly influenced equilibrium dissociation constants determined under different physicochemical conditions. Limited proteolysis experiments coupled with mass spectrometric identification of cleavage fragments suggested a high degree of conformational flexibility affecting at least the first 21 residues of NS3. The accessibility of this region of the protease to limited chymotryptic digestion did not significantly change in any condition tested, whereas a significant reduction of chymotryptic cleavages within the NS3 core was detected under conditions of high NS3-Pep4AK complex affinity. We conclude the following: (1) The N-terminus of the NS3 protease that, according to the X-ray crystal structure, makes extensive contacts with the cofactor peptide is highly flexible in solution and contributes only marginally to the thermodynamic stability of the complex. (2) Affinity enhancement is accomplished by several factors through a general stabilization of the fold of the NS3 molecule.


Subject(s)
Hepacivirus/enzymology , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Chemical Phenomena , Chemistry, Physical , Detergents , Enzyme Stability , Glycerol/metabolism , Hepacivirus/metabolism , Humans , Hydrogen-Ion Concentration , Macromolecular Substances , Molecular Sequence Data , Osmolar Concentration , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Substrate Specificity , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
18.
Anal Biochem ; 266(2): 192-7, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9888975

ABSTRACT

A novel radiometric in vitro assay for discovery of inhibitors of hepatitis C viral protease activity, suitable for high-throughput screening, was developed. The NS3 protein of hepatitis C virus (HCV) contains a serine protease, whose function is to process the majority of the nonstructural proteins of the viral polyprotein. The viral NS4A protein is a cofactor of NS3 protease activity in the cleavage of NS3-NS4A, NS4A-NS4B, NS4B-NS5A, and NS5A-NS5B junctions. To establish an in vitro assay system we used NS3 proteases from different HCV strains, purified from Escherichia coli and a synthetic radiolabeled peptide substrate that mimics the NS4A-NS4B junction. Upon incubation with the enzyme the substrate was separated from the radiolabeled cleavage product by addition of an ion exchange resin. The assay was performed in a microtiter plate format and offered the potential for assaying numerous samples using a laboratory robot. Taking advantage of these features, we used the assay to optimize reaction conditions by simultaneously varying different buffer components. We showed that physicochemical conditions affect NS3 protease activity in a strain-specific way. Furthermore, the sensitivity of the assay makes it suitable for detection and detailed mechanistic characterization of inhibitors with low-nanomolar affinities for the HCV serine protease.


Subject(s)
Radiometry/methods , Viral Nonstructural Proteins/analysis , Buffers , Endopeptidases/metabolism , Kinetics , Peptides/pharmacology , Protease Inhibitors/pharmacology , Sensitivity and Specificity , Time Factors , Viral Nonstructural Proteins/antagonists & inhibitors
19.
J Viral Hepat ; 6 Suppl 1: 23-30, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10760031

ABSTRACT

The NS3 serine proteinase is regarded as one of the preferred targets for the development of therapeutic agents against hepatitis C virus (HCV). Possible mechanisms of NS3 inhibitors include: (i) interference with the activation of the enzyme by its NS4A cofactor; (ii) binding to the structural zinc site; and (iii) binding to the active site. These mechanisms have been explored in detail by structural analysis of the enzyme. (i) The NS4A cofactor binds to the amino-terminal beta-barrel domain of the NS3 proteinase bringing about several conformational changes that result in enzyme activation. The interaction between NS3 and NS4A involves a very large surface area and therefore it is not a likely target for the development of inhibitors. (ii) The NS3 proteinase contains a structural zinc binding site. Spectroscopic studies have shown that changes in the conformation of this metal-binding site correlate with changes in the specific activity of the enzyme, and the NS3 proteinase is inhibited by compounds capable of extracting zinc from its native coordination sphere. (iii) Based on the observation that the NS3 proteinase undergoes inhibition by its cleavage products, potent, active site-directed inhibitors have been generated. Kinetic studies, site-directed mutagenesis, and molecular modelling have been used to characterize the interactions between the NS3 proteinase and its product inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Binding Sites , Mutagenesis, Site-Directed , Structure-Activity Relationship , Viral Nonstructural Proteins/chemistry , Zinc/metabolism
20.
Biochemistry ; 37(33): 11459-68, 1998 Aug 18.
Article in English | MEDLINE | ID: mdl-9708981

ABSTRACT

Hepatitis C virus (HCV) infection is a major health problem that leads to cirrhosis and hepatocellular carcinoma in a substantial number of infected individuals, estimated to be 100-200 million worldwide. Unfortunately, immunotherapy or other effective treatments for HCV infection are not yet available, and interferon administration has limited efficacy. Different approaches to HCV therapy are being explored, and these include inhibition of the viral proteinase, helicase, and RNA-dependent RNA polymerase and development of a vaccine. Here we present the design of selective inhibitors with nanomolar potencies of HCV NS3 proteinase based on eglin c. These eglin c mutants were generated by reshaping the inhibitor active site-binding loop, and the results emphasize the role played by residues P5-P4' in enzyme recognition. In addition, alanine scanning experiments provide evidence that the N terminus of eglin c also contributes to NS3 binding. These eglin inhibitors offer a unique tool for accurately assessing the requirements for effective inhibition of the enzymatic activity of NS3 and at the same time can be considered lead compounds for the identification of other NS3 inhibitors in targeted design efforts.


Subject(s)
Hepacivirus/enzymology , Protein Engineering , Serine Proteinase Inhibitors/chemical synthesis , Serpins/chemical synthesis , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Humans , Kinetics , Leeches , Macromolecular Substances , Peptide Fragments/metabolism , Protein Binding , Protein Engineering/methods , Proteins , RNA Helicases , Serine Endopeptidases , Serine Proteinase Inhibitors/metabolism , Serine Proteinase Inhibitors/pharmacology , Serpins/metabolism , Serpins/pharmacology , Structure-Activity Relationship , Substrate Specificity , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...