Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 292(27): 11165-11177, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28483922

ABSTRACT

Human cells detect RNA viruses through a set of helicases called RIG-I-like receptors (RLRs) that initiate the interferon response via a mitochondrial signaling complex. Many RNA viruses also encode helicases, which are sometimes covalently linked to proteases that cleave signaling proteins. One unresolved question is how RLRs interact with each other and with viral proteins in cells. This study examined the interactions among the hepatitis C virus (HCV) helicase and RLR helicases in live cells with quantitative microspectroscopic imaging (Q-MSI), a technique that determines FRET efficiency and subcellular donor and acceptor concentrations. HEK293T cells were transfected with various vector combinations to express cyan fluorescent protein (CFP) or YFP fused to either biologically active HCV helicase or one RLR (i.e. RIG-I, MDA5, or LGP2), expressed in the presence or absence of polyinosinic-polycytidylic acid (poly(I:C)), which elicits RLR accumulation at mitochondria. Q-MSI confirmed previously reported RLR interactions and revealed an interaction between HCV helicase and LGP2. Mitochondria in CFP-RIG-I:YFP-RIG-I cells, CFP-MDA5:YFP-MDA5 cells, and CFP-MDA5:YFP-LGP2 cells had higher FRET efficiencies in the presence of poly(I:C), indicating that RNA causes these proteins to accumulate at mitochondria in higher-order complexes than those formed in the absence of poly(I:C). However, mitochondria in CFP-LGP2:YFP-LGP2 cells had lower FRET signal in the presence of poly(I:C), suggesting that LGP2 oligomers disperse so that LGP2 can bind MDA5. Data support a new model where an LGP2-MDA5 oligomer shuttles NS3 to the mitochondria to block antiviral signaling.


Subject(s)
Hepacivirus/enzymology , Interferon-Induced Helicase, IFIH1/metabolism , Mitochondria/enzymology , Models, Biological , RNA Helicases/metabolism , Signal Transduction , Viral Nonstructural Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Hepacivirus/genetics , Humans , Interferon-Induced Helicase, IFIH1/genetics , Microscopy, Fluorescence/methods , Mitochondria/genetics , Poly I-C/pharmacology , RNA Helicases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Nonstructural Proteins/genetics
2.
ACS Chem Biol ; 9(10): 2393-403, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25126694

ABSTRACT

The hepatitis C virus (HCV) nonstructural protein 3 (NS3) is both a protease, which cleaves viral and host proteins, and a helicase that separates nucleic acid strands, using ATP hydrolysis to fuel the reaction. Many antiviral drugs, and compounds in clinical trials, target the NS3 protease, but few helicase inhibitors that function as antivirals have been reported. This study focuses on the analysis of the mechanism by which ebselen (2-phenyl-1,2-benzisoselenazol-3-one), a compound previously shown to be a HCV antiviral agent, inhibits the NS3 helicase. Ebselen inhibited the abilities of NS3 to unwind nucleic acids, to bind nucleic acids, and to hydrolyze ATP, and about 1 µM ebselen was sufficient to inhibit each of these activities by 50%. However, ebselen had no effect on the activity of the NS3 protease, even at 100 times higher ebselen concentrations. At concentrations below 10 µM, the ability of ebselen to inhibit HCV helicase was reversible, but prolonged incubation of HCV helicase with higher ebselen concentrations led to irreversible inhibition and the formation of covalent adducts between ebselen and all 14 cysteines present in HCV helicase. Ebselen analogues with sulfur replacing the selenium were just as potent HCV helicase inhibitors as ebselen, but the length of the linker between the phenyl and benzisoselenazol rings was critical. Modifications of the phenyl ring also affected compound potency over 30-fold, and ebselen was a far more potent helicase inhibitor than other, structurally unrelated, thiol-modifying agents. Ebselen analogues were also more effective antiviral agents, and they were less toxic to hepatocytes than ebselen. Although the above structure-activity relationship studies suggest that ebselen targets a specific site on NS3, we were unable to confirm binding to either the NS3 ATP binding site or nucleic acid binding cleft by examining the effects of ebselen on NS3 proteins lacking key cysteines.


Subject(s)
Antiviral Agents/pharmacology , Azoles/pharmacology , Hepatitis C/virology , Nucleic Acids/metabolism , Organoselenium Compounds/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects , Antioxidants/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Cell Proliferation , Electrophoretic Mobility Shift Assay , Hepacivirus/drug effects , Hepatitis C/drug therapy , Hepatitis C/metabolism , Humans , Hydrolysis , Isoindoles , Kinetics , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Models, Molecular , Protein Structure, Tertiary , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , Tumor Cells, Cultured
3.
J Biomol Screen ; 18(7): 761-81, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23536547

ABSTRACT

Helicases are ubiquitous motor proteins that separate and/or rearrange nucleic acid duplexes in reactions fueled by adenosine triphosphate (ATP) hydrolysis. Helicases encoded by bacteria, viruses, and human cells are widely studied targets for new antiviral, antibiotic, and anticancer drugs. This review summarizes the biochemistry of frequently targeted helicases. These proteins include viral enzymes from herpes simplex virus, papillomaviruses, polyomaviruses, coronaviruses, the hepatitis C virus, and various flaviviruses. Bacterial targets examined include DnaB-like and RecBCD-like helicases. The human DEAD-box protein DDX3 is the cellular antiviral target discussed, and cellular anticancer drug targets discussed are the human RecQ-like helicases and eIF4A. We also review assays used for helicase inhibitor discovery and the most promising and common helicase inhibitor chemotypes, such as nucleotide analogues, polyphenyls, metal ion chelators, flavones, polycyclic aromatic polymers, coumarins, and various DNA binding pharmacophores. Also discussed are common complications encountered while searching for potent helicase inhibitors and possible solutions for these problems.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , DNA Helicases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Viral Proteins/antagonists & inhibitors , Animals , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Bacterial Proteins/physiology , DNA Helicases/physiology , DNA Replication , High-Throughput Screening Assays , Humans , Protein Binding , Protein Biosynthesis , Viral Proteins/physiology
4.
Antiviral Res ; 96(2): 245-55, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22940425

ABSTRACT

The hepatitis C virus (HCV) multifunctional nonstructural protein 3 (NS3) is a protease that cleaves viral and host proteins and a helicase that separates DNA and RNA structures in reactions fueled by ATP hydrolysis. Li et al. (2012) recently synthesized a series of new NS3 helicase inhibitors from the benzothiazole dimer component of the fluorescent yellow dye primuline. This study further characterizes a subset of these primuline derivatives with respect to their specificity, mechanism of action, and effect on cells harboring HCV subgenomic replicons. All compounds inhibited DNA and RNA unwinding catalyzed by NS3 from different HCV genotypes, but only some inhibited the NS3 protease function, and few had any effect on HCV NS3 catalyzed ATP hydrolysis. A different subset contained potent inhibitors of RNA stimulated ATP hydrolysis catalyzed by the related NS3 protein from Dengue virus. In assays monitoring intrinsic protein fluorescence in the absence of nucleic acids, the compounds cooperatively bound NS3 with K(d)s that reflect their potency in assays. The fluorescent properties of the primuline derivatives both in vitro and in cells are also described. The primuline derivative that was the most active against subgenomic replicons in cells caused a 14-fold drop in HCV RNA levels (IC(50)=5±2µM). In cells, the most effective primuline derivative did not inhibit the cellular activity of NS3 protease but disrupted HCV replicase structures.


Subject(s)
Antiviral Agents/pharmacology , DNA Helicases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Serine Endopeptidases/metabolism , Thiazoles/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Cell Line , Fluorescence , Hepacivirus/drug effects , Humans , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Protein Binding , Thiazoles/chemistry
5.
Vaccine ; 24(22): 4716-26, 2006 May 29.
Article in English | MEDLINE | ID: mdl-16621195

ABSTRACT

Domain II of Mycobacterium tuberculosis (Mtb) heat shock protein 70 (HSP70), spanning amino acid residues 161-370, was covalently linked to a recently described synthetic dengue virus antigen to study the influence of the former on the immunogenicity of the latter. Using an enzyme-linked immunosorbent assay, dengue antigen-specific antibody titers elicited by the fusion protein in Balb/c mice were an order of magnitude higher than those elicited by either the synthetic dengue antigen alone or a physical mixture of the dengue antigen plus Mtb HSP70 domain II protein. Our data demonstrate that (i) Mtb HSP70 domain II is capable of potentiating B-cell response and (ii) it should be covalently linked to the target antigen to do so.


Subject(s)
Antibodies, Viral/blood , Bacterial Proteins/immunology , Dengue Virus/immunology , HSP70 Heat-Shock Proteins/immunology , Peptide Fragments/immunology , Recombinant Fusion Proteins/immunology , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/genetics , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...