Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Hepatology ; 65(5): 1462-1477, 2017 05.
Article in English | MEDLINE | ID: mdl-28090674

ABSTRACT

Hepatitis C virus (HCV) infection is a common risk factor for the development of liver cancer. The molecular mechanisms underlying this effect are only partially understood. Here, we show that the HCV protein, nonstructural protein (NS) 5B, directly binds to the tumor suppressor, NORE1A (RASSF5), and promotes its proteosomal degradation. In addition, we show that NORE1A colocalizes to sites of HCV viral replication and suppresses the replication process. Thus, NORE1A has antiviral activity, which is specifically antagonized by NS5B. Moreover, the suppression of NORE1A protein levels correlated almost perfectly with elevation of Ras activity in primary human samples. Therefore, NORE1A inactivation by NS5B may be essential for maximal HCV replication and may make a major contribution to HCV-induced liver cancer by shifting Ras signaling away from prosenescent/proapoptotic signaling pathways. CONCLUSION: HCV uses NS5B to specifically suppress NORE1A, facilitating viral replication and elevated Ras signaling. (Hepatology 2017;65:1462-1477).


Subject(s)
Hepacivirus/physiology , Monomeric GTP-Binding Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Carcinoma, Hepatocellular/virology , Down-Regulation , HEK293 Cells , Humans , Liver/metabolism , Liver/virology , Liver Neoplasms/virology , Proteasome Endopeptidase Complex/metabolism
2.
J Virol ; 88(20): 11825-33, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25100850

ABSTRACT

To combat emerging coronaviruses, developing safe and efficient platforms to evaluate viral protease activities and the efficacy of protease inhibitors is a high priority. Here, we exploit a biosafety level 2 (BSL-2) chimeric Sindbis virus system to evaluate protease activities and the efficacy of inhibitors directed against the papain-like protease (PLpro) of severe acute respiratory syndrome coronavirus (SARS-CoV), a biosafety level 3 (BSL-3) pathogen. We engineered Sindbis virus to coexpress PLpro and a substrate, murine interferon-stimulated gene 15 (ISG15), and found that PLpro mediates removal of ISG15 (deISGylation) from cellular proteins. Mutation of the catalytic cysteine residue of PLpro or addition of a PLpro inhibitor blocked deISGylation in virus-infected cells. Thus, deISGylation is a marker of PLpro activity. Infection of alpha/beta interferon receptor knockout (IFNAR(-/-)) mice with these chimeric viruses revealed that PLpro deISGylation activity removed ISG15-mediated protection during viral infection. Importantly, administration of a PLpro inhibitor protected these mice from lethal infection, demonstrating the efficacy of a coronavirus protease inhibitor in a mouse model. However, this PLpro inhibitor was not sufficient to protect the mice from lethal infection with SARS-CoV MA15, suggesting that further optimization of the delivery and stability of PLpro inhibitors is needed. We extended the chimeric-virus platform to evaluate the papain-like protease/deISGylating activity of Middle East respiratory syndrome coronavirus (MERS-CoV) to provide a small-animal model to evaluate PLpro inhibitors of this recently emerged pathogen. This platform has the potential to be universally adaptable to other viral and cellular enzymes that have deISGylating activities. Importance: Evaluating viral protease inhibitors in a small-animal model is a critical step in the path toward antiviral drug development. We modified a biosafety level 2 chimeric virus system to facilitate evaluation of inhibitors directed against highly pathogenic coronaviruses. We used this system to demonstrate the in vivo efficacy of an inhibitor of the papain-like protease of severe acute respiratory syndrome coronavirus. Furthermore, we demonstrate that the chimeric-virus system can be adapted to study the proteases of emerging human pathogens, such as Middle East respiratory syndrome coronavirus. This system provides an important tool to rapidly assess the efficacy of protease inhibitors targeting existing and emerging human pathogens, as well as other enzymes capable of removing ISG15 from cellular proteins.


Subject(s)
Coronavirus/physiology , Disease Models, Animal , Papain/metabolism , Peptide Hydrolases/metabolism , Animals , Chlorocebus aethiops , Coronavirus/enzymology , Cricetinae , Mice , Vero Cells
3.
Eur J Med Chem ; 69: 931-41, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24161679

ABSTRACT

Hepatitis C virus (HCV) NS5B polymerase is an important and attractive target for the development of anti-HCV drugs. Here we report on the design, synthesis and evaluation of twenty-four novel allosteric inhibitors bearing the 4-thiazolidinone scaffold as inhibitors of HCV NS5B polymerase. Eleven compounds tested were found to inhibit HCV NS5B with IC50 values ranging between 19.8 and 64.9 µM. Compound 24 was the most active of this series with an IC50 of 5.6 µM. A number of these derivatives further exhibited strong inhibition against HCV 1b and 2a genotypes in cell based antiviral assays. Molecular docking analysis predicted that the thiazolidinone derivatives bind to the NS5B thumb pocket-II (TP-II). Our results suggest that further optimization of the thiazolidinone scaffold may be possible to yield new derivatives with improved enzyme- and cell-based activity.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepacivirus/enzymology , Thiazolidines/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/classification , Cells, Cultured , Dose-Response Relationship, Drug , Genotype , Hepacivirus/genetics , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Thiazolidines/chemical synthesis , Thiazolidines/chemistry , Viral Nonstructural Proteins/metabolism
4.
Chem Biol Drug Des ; 81(5): 607-14, 2013 May.
Article in English | MEDLINE | ID: mdl-23311976

ABSTRACT

Coumarins and coumestans represent an important family of compounds with diverse pharmacological properties. We recently identified coumestans as novel inhibitors of hepatitis C virus NS5B polymerase and predicted their binding in thumb pocket-1 (TP-1) of NS5B. As the coumarins are structurally related to coumestans by virtue of their common A- and B-rings, we postulated them to also exhibit similar binding interaction with NS5B and inhibit its polymerase function. We therefore investigated 24 coumarin and neoflavone derivatives as candidate NS5B inhibitors and identified 14 compounds inhibiting NS5B polymerase activity with IC50 values between 17 and 63 µm. Of these, the newly synthesized 6,8-diallyl-5,7-dihydroxycoumarin (8a) was produced in three steps in high chemical yield from floroglucinol and found to be the most potent of this series, exhibiting activity similar to the reference coumestan LQB-34. The binding site of 8a was mapped to TP-1 of NS5B by counter screening against P495L NS5B mutant, employed as a screen for TP-1 site binders. NS5B-TP-1-8a interaction map provided insight into 8a binding and offered clues for future SAR optimization.


Subject(s)
Antiviral Agents , Coumarins , Enzyme Inhibitors , Flavones , Hepacivirus/enzymology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Anticoagulants/chemical synthesis , Anticoagulants/chemistry , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Binding Sites , Coumarins/chemical synthesis , Coumarins/chemistry , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Flavones/chemical synthesis , Flavones/chemistry , Viral Nonstructural Proteins/chemistry
5.
Eur J Med Chem ; 58: 258-64, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23127989

ABSTRACT

Hepatitis C virus (HCV) NS5B polymerase is a key target for the development of anti-HCV drugs. Here we report on the identification of novel allosteric inhibitors of HCV NS5B through a combination of structure-based virtual screening and in vitro NS5B inhibition assays. One hundred and sixty thousand compounds from the Otava database were virtually screened against the thiazolone inhibitor binding site on NS5B (thumb pocket-2, TP-2), resulting in a sequential down-sizing of the library by 2.7 orders of magnitude to yield 59 NS5B non-nucleoside inhibitor (NNI) candidates. In vitro evaluation of the NS5B inhibitory activity of the 59 selected compounds resulted in a 14% hit rate, yielding 8 novel structural scaffolds. Of these, compound 1 bearing a 4-hydrazinoquinazoline scaffold was the most active (IC(50) = 16.0 µM). The binding site of all 8 NNIs was mapped to TP-2 of NS5B as inferred by a decrease in their inhibition potency against the M423T NS5B mutant, employed as a screen for TP-2 site binders. At 100 µM concentration, none of the eight compounds exhibited any cytotoxicity, and all except compound 8 exhibited between 40 and 60% inhibition of intracellular NS5B polymerase activity in BHK-NS5B-FRLuc reporter cells. These inhibitor scaffolds will form the basis for future optimization and development of more potent NS5B inhibitors.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Hydrazines/pharmacology , Quinazolinones/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Hydrazines/chemistry , Models, Molecular , Molecular Structure , Quinazolinones/chemistry , Structure-Activity Relationship , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
6.
PLoS One ; 7(2): e30802, 2012.
Article in English | MEDLINE | ID: mdl-22312431

ABSTRACT

Viruses have evolved elaborate mechanisms to evade or inactivate the complex system of sensors and signaling molecules that make up the host innate immune response. Here we show that human coronavirus (HCoV) NL63 and severe acute respiratory syndrome (SARS) CoV papain-like proteases (PLP) antagonize innate immune signaling mediated by STING (stimulator of interferon genes, also known as MITA/ERIS/MYPS). STING resides in the endoplasmic reticulum and upon activation, forms dimers which assemble with MAVS, TBK-1 and IKKε, leading to IRF-3 activation and subsequent induction of interferon (IFN). We found that expression of the membrane anchored PLP domain from human HCoV-NL63 (PLP2-TM) or SARS-CoV (PLpro-TM) inhibits STING-mediated activation of IRF-3 nuclear translocation and induction of IRF-3 dependent promoters. Both catalytically active and inactive forms of CoV PLPs co-immunoprecipitated with STING, and viral replicase proteins co-localize with STING in HCoV-NL63-infected cells. Ectopic expression of catalytically active PLP2-TM blocks STING dimer formation and negatively regulates assembly of STING-MAVS-TBK1/IKKε complexes required for activation of IRF-3. STING dimerization was also substantially reduced in cells infected with SARS-CoV. Furthermore, the level of ubiquitinated forms of STING, RIG-I, TBK1 and IRF-3 are reduced in cells expressing wild type or catalytic mutants of PLP2-TM, likely contributing to disruption of signaling required for IFN induction. These results describe a new mechanism used by CoVs in which CoV PLPs negatively regulate antiviral defenses by disrupting the STING-mediated IFN induction.


Subject(s)
Coronavirus NL63, Human/enzymology , Cysteine Endopeptidases/metabolism , Immunity, Innate , Membrane Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/enzymology , Signal Transduction/immunology , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biocatalysis , Cell Membrane/enzymology , Chlorocebus aethiops , Coronavirus 3C Proteases , Coronavirus NL63, Human/physiology , Cysteine Endopeptidases/chemistry , HEK293 Cells , Humans , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/metabolism , Interferons/metabolism , Membrane Proteins/chemistry , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Severe acute respiratory syndrome-related coronavirus/physiology , Ubiquitination/immunology , Vero Cells , Viral Proteins/chemistry
7.
Eur J Med Chem ; 49: 191-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22280819

ABSTRACT

Structure-based studies led to the identification of a constrained derivative of S-trityl-l-cysteine (STLC) scaffold as a candidate inhibitor of hepatitis C virus (HCV) NS5B polymerase. A panel of STLC derivatives were synthesized and investigated for their activity against HCV NS5B. Three STLC derivatives, 9, F-3070, and F-3065, were identified as modest HCV NS5B inhibitors with IC(50) values between 22.3 and 39.7 µM. F-3070 and F-3065 displayed potent inhibition of intracellular NS5B activity in the BHK-NS5B-FRLuc reporter and also inhibited HCV RNA replication in the Huh7/Rep-Feo1b reporter system. Binding mode investigations suggested that the STLC scaffold can be used to develop new NS5B inhibitors by further chemical modification at one of the trityl phenyl group.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cysteine/analogs & derivatives , Hepacivirus/enzymology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Cell Line , Cysteine/chemistry , Cysteine/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hepacivirus/drug effects , Hepatitis C/drug therapy , Hepatitis C/enzymology , Humans , Models, Molecular , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism
8.
J Med Chem ; 53(13): 4968-79, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20527968

ABSTRACT

The design, synthesis, X-ray crystal structure, molecular modeling, and biological evaluation of a series of new generation SARS-CoV PLpro inhibitors are described. A new lead compound 3 (6577871) was identified via high-throughput screening of a diverse chemical library. Subsequently, we carried out lead optimization and structure-activity studies to provide a series of improved inhibitors that show potent PLpro inhibition and antiviral activity against SARS-CoV infected Vero E6 cells. Interestingly, the (S)-Me inhibitor 15 h (enzyme IC(50) = 0.56 microM; antiviral EC(50) = 9.1 microM) and the corresponding (R)-Me 15 g (IC(50) = 0.32 microM; antiviral EC(50) = 9.1 microM) are the most potent compounds in this series, with nearly equivalent enzymatic inhibition and antiviral activity. A protein-ligand X-ray structure of 15 g-bound SARS-CoV PLpro and a corresponding model of 15 h docked to PLpro provide intriguing molecular insight into the ligand-binding site interactions.


Subject(s)
Antiviral Agents/chemistry , Enzyme Inhibitors/chemistry , Piperidines/chemistry , Severe Acute Respiratory Syndrome/drug therapy , Severe acute respiratory syndrome-related coronavirus/enzymology , Viral Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Chlorocebus aethiops , Coronavirus 3C Proteases , Crystallography, X-Ray , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Piperidines/chemical synthesis , Piperidines/metabolism , Piperidines/pharmacology , Severe Acute Respiratory Syndrome/virology , Structure-Activity Relationship , Vero Cells , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication/drug effects
9.
J Med Chem ; 52(16): 5228-40, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19645480

ABSTRACT

We describe here the design, synthesis, molecular modeling, and biological evaluation of a series of small molecule, nonpeptide inhibitors of SARS-CoV PLpro. Our initial lead compound was identified via high-throughput screening of a diverse chemical library. We subsequently carried out structure-activity relationship studies and optimized the lead structure to potent inhibitors that have shown antiviral activity against SARS-CoV infected Vero E6 cells. Upon the basis of the X-ray crystal structure of inhibitor 24-bound to SARS-CoV PLpro, a drug design template was created. Our structure-based modification led to the design of a more potent inhibitor, 2 (enzyme IC(50) = 0.46 microM; antiviral EC(50) = 6 microM). Interestingly, its methylamine derivative, 49, displayed good enzyme inhibitory potency (IC(50) = 1.3 microM) and the most potent SARS antiviral activity (EC(50) = 5.2 microM) in the series. We have carried out computational docking studies and generated a predictive 3D-QSAR model for SARS-CoV PLpro inhibitors.


Subject(s)
Antiviral Agents/chemical synthesis , Benzamides/chemical synthesis , Naphthalenes/chemical synthesis , Protease Inhibitors/chemical synthesis , Severe acute respiratory syndrome-related coronavirus/drug effects , Viral Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Benzamides/chemistry , Benzamides/pharmacology , Chlorocebus aethiops , Coronavirus 3C Proteases , Crystallography, X-Ray , Cysteine Endopeptidases , Drug Design , Models, Molecular , Naphthalenes/chemistry , Naphthalenes/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Quantitative Structure-Activity Relationship , Severe acute respiratory syndrome-related coronavirus/enzymology , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...