Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(9): e0272916, 2022.
Article in English | MEDLINE | ID: mdl-36103462

ABSTRACT

We recently published a preliminary assessment of the activity of a poly (ADP-ribose) polymerase (PARP) inhibitor, stenoparib, also known as 2X-121, which inhibits viral replication by affecting pathways of the host. Here we show that stenoparib effectively inhibits a SARS-CoV-2 wild type (BavPat1/2020) strain and four additional variant strains; alpha (B.1.1.7), beta (B.1.351), delta (B.1.617.2) and gamma (P.1) in vitro, with 50% effective concentration (EC50) estimates of 4.1 µM, 8.5 µM, 24.1 µM, 8.2 µM and 13.6 µM, respectively. A separate experiment focusing on a combination of 10 µM stenoparib and 0.5 µM remdesivir, an antiviral drug, resulted in over 80% inhibition of the alpha variant, which is substantially greater than the effect achieved with either drug alone, suggesting at least additive effects from combining the different mechanisms of activity of stenoparib and remdesivir.


Subject(s)
COVID-19 Drug Treatment , Poly(ADP-ribose) Polymerases , Adenosine Diphosphate , Humans , Poly(ADP-ribose) Polymerases/metabolism , Ribose , SARS-CoV-2
2.
Biochem Biophys Res Commun ; 578: 91-96, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34547629

ABSTRACT

The SARS-CoV-2 variant is rapidly spreading across the world and causes to resurge infections. We previously reported that CT-P59 presented its in vivo potency against Beta variants, despite its reduced activity in cell experiments. Yet, it remains uncertain to exert the antiviral effect of CT-P59 on Gamma, Delta and its associated variants (L452R). To tackle this question, we carried out cell tests and animal studies. CT-P59 showed neutralization against Gamma, Delta, Epsilon, and Kappa variants in cells, with reduced susceptibility. The mouse challenge experiments with Gamma and Delta variants substantiated in vivo potency of CT-P59 showing symptom remission and virus abrogation in the respiratory tract. Collectively, cell and animal studies showed that CT-P59 is effective against Gamma and Delta variants infection, hinting that CT-P59 has therapeutic potential for patients infected with Gamma, Delta and its associated variants.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Neutralizing/pharmacology , COVID-19 Drug Treatment , Disease Models, Animal , Immunoglobulin G/pharmacology , SARS-CoV-2/drug effects , Animals , Antiviral Agents/pharmacology , Body Weight/drug effects , COVID-19/virology , Female , Humans , Mice, Transgenic , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Survival Analysis
3.
Biochem Biophys Res Commun ; 566: 135-140, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34119826

ABSTRACT

The global circulation of newly emerging variants of SARS-CoV-2 is a new threat to public health due to their increased transmissibility and immune evasion. Moreover, currently available vaccines and therapeutic antibodies were shown to be less effective against new variants, in particular, the South African (SA) variant, termed 501Y.V2 or B.1.351. To assess the efficacy of the CT-P59 monoclonal antibody against the SA variant, we sought to perform as in vitro binding and neutralization assays, and in vivo animal studies. CT-P59 neutralized B.1.1.7 variant to a similar extent as to wild type virus. CT-P59 showed reduced binding affinity against a RBD (receptor binding domain) triple mutant containing mutations defining B.1.351 (K417N/E484K/N501Y) also showed reduced potency against the SA variant in live virus and pseudovirus neutralization assay systems. However, in vivo ferret challenge studies demonstrated that a therapeutic dosage of CT-P59 was able to decrease B.1.351 viral load in the upper and lower respiratory tracts, comparable to that observed for the wild type virus. Overall, although CT-P59 showed reduced in vitro neutralizing activity against the SA variant, sufficient antiviral effect in B.1.351-infected animals was confirmed with a clinical dosage of CT-P59, suggesting that CT-P59 has therapeutic potential for COVID-19 patients infected with SA variant.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , COVID-19/therapy , COVID-19/virology , Immunoglobulin G/therapeutic use , SARS-CoV-2 , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , Disease Models, Animal , Female , Ferrets , Humans , Immunoglobulin G/immunology , In Vitro Techniques , Neutralization Tests , Pandemics , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , South Africa , Viral Load/immunology
4.
Int J Food Microbiol ; 344: 109089, 2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33662900

ABSTRACT

Contamination of bivalve molluscs with viruses is well recognized as a food safety risk. A microbiological criterion for norovirus (NoV) and hepatitis A virus (HAV) in shellfish, however, does not exist in the European Union currently. The aim of this study was to evaluate the contamination levels of these viruses for fluctuation over a long period (2013-2017) in oyster (n = 266) and mussel samples (n = 490) using a method based on ISO/TS 15216-1: 2013. Samples were taken at different points in the food chain, either directly post-harvest, at Dutch dispatch centers or in retail stores, from September until March of each year. Altogether, 53.1% of the mussel and 31.6% of the oyster samples tested positive for NoV RNA. Simultaneous presence of NoV GI and GII RNA was observed in 31.6% of mussel and 10.2% of oyster samples. Contamination levels in NoV positive mussel samples collected post-harvest from B-areas were significantly higher than in those collected post-harvest from A-areas, or at dispatch centers or retail stores. Levels in oysters from dispatch were significantly lower than those collected in retail stores. Ready for sale mussels and oysters contained 2.04 and 1.76 mean log10 transformed NoV genome copies/gram (gc/g), respectively. GII levels were at a constant level in ready for sale mussels throughout all sampling periods in the study. This seemed to be true for oysters as well. HAV RNA was detected in only one of the tested mussel samples (n = 392) (typed HAV 1A) and in none of the tested oyster samples (n = 228). Critical evaluation of NoV and HAV levels in shellfish can be of help for risk assessment and risk management actions.


Subject(s)
Caliciviridae Infections/epidemiology , Hepatitis A virus/isolation & purification , Hepatitis A/epidemiology , Norovirus/isolation & purification , Ostreidae/virology , Animals , Caliciviridae Infections/veterinary , Food Chain , Food Contamination/analysis , Food Safety , Hepatitis A/veterinary , Hepatitis A virus/genetics , Humans , Netherlands/epidemiology , Norovirus/genetics , Shellfish/virology
5.
Emerg Microbes Infect ; 8(1): 1076-1085, 2019.
Article in English | MEDLINE | ID: mdl-31339457

ABSTRACT

Enterovirus A71 (EV-A71) is one of the main causative agents of hand-foot-and-mouth disease and is occasionally associated with severe neurological complications. EV-A71 pathophysiology is poorly understood due to the lack of small animal models that robustly support viral replication in relevant organs/tissues. Here, we show that adult severe combined immune-deficient (SCID) mice can serve as an EV-A71 infection model to study neurotropic determinants and viral tropism. Mice inoculated intraperitoneally with an EV-A71 clinical isolate had an initial infection of the lung compartment, followed by neuroinvasion and infection of (motor)neurons, resulting in slowly progressing paralysis of the limbs. We identified a substitution (V135I) in the capsid protein VP2 as a key requirement for neurotropism. This substitution was also present in a mouse-adapted variant, obtained by passaging the clinical isolate in the brain of one-day-old mice, and induced exclusive neuropathology and rapid paralysis, confirming its role in neurotropism. Finally, we showed that this residue enhances the capacity of EV-A71 to use mouse PSGL1 for viral entry. Our data reveal that EV-A71 initially disseminates to the lung and identify viral and host determinants that define the neurotropic character of EV-A71, pointing to a hitherto understudied role of PSGL1 in EV-A71 tropism and neuropathology.


Subject(s)
Capsid Proteins/genetics , Enterovirus A, Human/physiology , Enterovirus Infections/metabolism , Membrane Glycoproteins/metabolism , Neurons/virology , Animals , Capsid Proteins/metabolism , Enterovirus A, Human/genetics , Enterovirus A, Human/pathogenicity , Enterovirus Infections/genetics , Enterovirus Infections/virology , Host-Pathogen Interactions , Humans , Membrane Glycoproteins/genetics , Mice , Mice, SCID , Mutation, Missense , Neurons/metabolism , Viral Tropism , Virulence , Virus Internalization
6.
Lancet Infect Dis ; 19(7): 778-786, 2019 07.
Article in English | MEDLINE | ID: mdl-31133519

ABSTRACT

BACKGROUND: Salmonella spp are a major cause of food-borne outbreaks in Europe. We investigated a large multi-country outbreak of Salmonella enterica serotype Enteritidis in the EU and European Economic Area (EEA). METHODS: A confirmed case was defined as a laboratory-confirmed infection with the outbreak strains of S Enteritidis based on whole-genome sequencing (WGS), occurring between May 1, 2015, and Oct 31, 2018. A probable case was defined as laboratory-confirmed infection with S Enteritidis with the multiple-locus variable-number tandem repeat analysis outbreak profile. Multi-country epidemiological, trace-back, trace-forward, and environmental investigations were done. We did a case-control study including confirmed and probable cases and controls randomly sampled from the population registry (frequency matched by age, sex, and postal code). Odds ratios (ORs) for exposure rates between cases and controls were calculated with unmatched univariable and multivariable logistic regression. FINDINGS: 18 EU and EEA countries reported 838 confirmed and 371 probable cases. 509 (42%) cases were reported in 2016, after which the number of cases steadily increased. The case-control study results showed that cases more often ate in food establishments than did controls (OR 3·4 [95% CI 1·6-7·3]), but no specific food item was identified. Recipe-based food trace-back investigations among cases who ate in food establishments identified eggs from Poland as the vehicle of infection in October, 2016. Phylogenetic analysis identified two strains of S Enteritidis in human cases that were subsequently identified in salmonella-positive eggs and primary production premises in Poland, confirming the source of the outbreak. After control measures were implemented, the number of cases decreased, but increased again in March, 2017, and the increase continued into 2018. INTERPRETATION: This outbreak highlights the public health value of multi-country sharing of epidemiological, trace-back, and microbiological data. The re-emergence of cases suggests that outbreak strains have continued to enter the food chain, although changes in strain population dynamics and fewer cases indicate that control measures had some effect. Routine use of WGS in salmonella surveillance and outbreak response promises to identify and stop outbreaks in the future. FUNDING: European Centre for Disease Prevention and Control; Directorate General for Health and Food Safety, European Commission; and National Public Health and Food Safety Institutes of the authors' countries (see Acknowledgments for full list).


Subject(s)
Disease Outbreaks , Eggs/microbiology , Epidemiologic Studies , Salmonella Food Poisoning/diagnosis , Salmonella enteritidis/isolation & purification , Serogroup , Whole Genome Sequencing , Case-Control Studies , Europe/epidemiology , Female , Humans , Male , Poland , Salmonella Food Poisoning/epidemiology , Salmonella Food Poisoning/microbiology
7.
Int J Food Microbiol ; 296: 58-64, 2019 May 02.
Article in English | MEDLINE | ID: mdl-30849707

ABSTRACT

The aim of the present study was to assess pork liver and meat products present on the Dutch market for the presence of hepatitis E virus (HEV) RNA. HEV RNA was detected in 27.3% of 521 products sampled from Dutch retail stores in 2016. 12.7% of livers were positive for HEV RNA (n = 79), 70.7% of liverwurst (n = 99), 68.9% of liver pate (n = 90), but in none of the pork chops (n = 98), fresh sausages (n = 103) or wild boar meat (n = 52). The highest level of HEV RNA contamination was observed in a liver (reaching up to 1 × 106 copies/g), followed by ready to eat liverwurst and liver pate (up to 3 × 104 copies/g and 7 × 104 copies/g respectively). Sequence analyses revealed mainly genotype 3c, but also some 3a, 3e and 3f strains. One strain derived from a liver sample was 100% (493 nt) identical with one isolated from a HEV case with onset of disease close in time and geography, although no direct epidemiological link could be established. Despite liverwurst and liver pate undergo heat treatment (information dd. Mid 2017) that may be sufficient to inactivate HEV, persons at risk, including Dutch transplant recipients, have been advised to avoid the consumption of raw liver as well as liverwurst and liver pate.


Subject(s)
Hepatitis E virus/isolation & purification , Liver/virology , Meat Products/virology , RNA, Viral/analysis , Red Meat/virology , Animals , Food Microbiology/methods , Genotype , Hepatitis E/transmission , Hepatitis E/virology , Hepatitis E virus/genetics , Humans , RNA, Viral/genetics , Sus scrofa , Swine , Swine Diseases/transmission , Swine Diseases/virology
8.
Int J Food Microbiol ; 257: 225-231, 2017 Sep 18.
Article in English | MEDLINE | ID: mdl-28692890

ABSTRACT

The aim of the present study was to investigate whether the use of porcine blood(products) in food could be a risk for a hepatitis E virus (HEV) infection. HEV RNA was detected in 33/36 batches of (non-heated) liquid products and in 7/24 spray dried powder products. Contamination levels varied among the products, but were highest in liquid whole blood, plasma and fibrinogen reaching levels of 2.2×102 to 2.8×102 HEV genome copies per 0.2g. Sequence analyses revealed genotype 3 strains, of which two were 100% (493nt) identical to recently diagnosed HEV cases, although no direct epidemiological link was established. The industry provided information on processing of blood products in (ready-to-eat)-meat. From this, it was concluded that blood products as an ingredient of processed meat may not be sufficiently heated prior to consumption, and therefore could be a vehicle for transmission.


Subject(s)
Food Contamination/analysis , Hepatitis E/transmission , Hepatitis E/veterinary , Meat Products/virology , Meat/analysis , Swine Diseases/transmission , Animals , Blood-Borne Pathogens , Female , Genotype , Hepatitis E/virology , Hepatitis E virus/genetics , Humans , Netherlands , RNA, Viral/analysis , Swine , Swine Diseases/virology
9.
Antiviral Res ; 133: 106-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27485294

ABSTRACT

It has been generally accepted that hydantoin [5-(3,4-dichlorophenyl)methylhydantoin] exerts its anti-enterovirus activity by solely inhibiting viral assembly. However, we here show that hydantoin inhibits enteroviral RNA synthesis as well as subgenomic replication in a dose-dependent manner. We demonstrate that inhibition of RNA synthesis is the predominant mechanism of action at relatively high concentrations of hydantoin. However, at lower concentrations inhibition of viral morphogenesis is the main mechanism of action. Thus, hydantoin inhibits enteroviral replication by two distinct mechanisms.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus/drug effects , Hydantoins/pharmacology , Humans , RNA, Viral , Virus Assembly/drug effects , Virus Replication/drug effects , Virus Replication/genetics
10.
Methods Mol Biol ; 1387: 325-38, 2016.
Article in English | MEDLINE | ID: mdl-26983743

ABSTRACT

The Global Polio Eradication Initiative, launched in 1988, had as its goal the eradication of polio worldwide by the year 2000 through large-scale vaccinations campaigns with the live attenuated oral PV vaccine (OPV) (Griffiths et al., Biologicals 34:73-74, 2006). Despite substantial progress, polio remains endemic in several countries and new imported cases are reported on a regular basis ( http://www.polioeradication.org/casecount.asp ).It was recognized by the poliovirus research community that developing antivirals against poliovirus would be invaluable in the post-OPV era. Here, we describe three methods essential for the identification of selective inhibitors of poliovirus replication and for determining their mode of action by time-of-drug-addition studies as well as by the isolation of compound-resistant poliovirus variants.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery/methods , Poliomyelitis/drug therapy , Poliovirus/drug effects , Virus Replication/drug effects , Antiviral Agents/therapeutic use , Disease Outbreaks , Drug Resistance, Viral , Genotype , Global Health , Humans , Microbial Sensitivity Tests/methods , Poliomyelitis/epidemiology , Poliovirus/genetics , Poliovirus/physiology
11.
Eur J Med Chem ; 111: 84-94, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-26854380

ABSTRACT

Very recently, we demonstrated that N(6)-isopentenyladenosine, a cytokinin nucleoside, exerts a potent and selective antiviral effect on the replication of human enterovirus 71. The present study is devoted to the structure optimization of another natural compound: N(6)-benzyladenosine. We mainly focused on the exploration of the size and nature of the linker between the adenine and the phenyl ring, as well as on the necessity of the D-ribose residue. More than 30 analogues of N(6)-benzyladenosine were prepared and their antiviral properties were evaluated. Two main methodologies were used for preparation: N(6)-acetyl-2',3',5'-tri-O-acetyladenosine can be regioselectively alkylated either by alkyl halides under base promoted conditions or by alcohols in Mitsunobu reactions. After deacylation with 4 M PrNH2 in MeOH at room temperature for one day, the desired products were obtained in overall high yields. Analysis of the structure-activity relationship clearly shows that the optimal size of the linker is limited to 2 or 3 atoms (compounds 4-7). 2'-Deoxyadenosine derivatives did not elicit any inhibitory or cytotoxic effect, while 5'-deoxynucleosides still induced some cell protective antiviral activity. Based on these observations, it can be hypothesized that there may be another mechanism that is at the base of the antiviral activity of these compounds against enterovirus 71 besides a possible 5'-triphosphorylation followed by a putative inhibitory effect on RNA synthesis.


Subject(s)
Adenosine/analogs & derivatives , Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Adenosine/chemical synthesis , Adenosine/chemistry , Adenosine/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
12.
PLoS Pathog ; 11(10): e1005165, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26485389

ABSTRACT

The replication of enterovirus 71 (EV71) and coxsackievirus A16 (CVA16), which are the major cause of hand, foot and mouth disease (HFMD) in children, can be inhibited by the capsid binder GPP3. Here, we present the crystal structure of CVA16 in complex with GPP3, which clarifies the role of the key residues involved in interactions with the inhibitor. Based on this model, in silico docking was performed to investigate the interactions with the two next-generation capsid binders NLD and ALD, which we show to be potent inhibitors of a panel of enteroviruses with potentially interesting pharmacological properties. A meta-analysis was performed using the available structural information to obtain a deeper insight into those structural features required for capsid binders to interact effectively and also those that confer broad-spectrum anti-enterovirus activity.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus A, Human/chemistry , Enterovirus A, Human/drug effects , Enterovirus A, Human/ultrastructure , Models, Molecular , Animals , Capsid/metabolism , Capsid Proteins/metabolism , Cell Line , Coxsackievirus Infections/prevention & control , Crystallography, X-Ray , Humans
13.
Eur Heart J ; 36(42): 2909-19, 2015 11 07.
Article in English | MEDLINE | ID: mdl-26206211

ABSTRACT

AIMS: Viral myocarditis (VM) is an important cause of heart failure and sudden cardiac death in young healthy adults; it is also an aetiological precursor of dilated cardiomyopathy. We explored the role of the miR-221/-222 family that is up-regulated in VM. METHODS AND RESULTS: Here, we show that microRNA-221 (miR-221) and miR-222 levels are significantly elevated during acute VM caused by Coxsackievirus B3 (CVB3). Both miRs are expressed by different cardiac cells and by infiltrating inflammatory cells, but their up-regulation upon myocarditis is mostly exclusive for the cardiomyocyte. Systemic inhibition of miR-221/-222 in mice increased cardiac viral load, prolonged the viraemic state, and strongly aggravated cardiac injury and inflammation. Similarly, in vitro, overexpression of miR-221 and miR-222 inhibited enteroviral replication, whereas knockdown of this miR-cluster augmented viral replication. We identified and confirmed a number of miR-221/-222 targets that co-orchestrate the increased viral replication and inflammation, including ETS1/2, IRF2, BCL2L11, TOX, BMF, and CXCL12. In vitro inhibition of IRF2, TOX, or CXCL12 in cardiomyocytes significantly dampened their inflammatory response to CVB3 infection, confirming the functionality of these targets in VM and highlighting the importance of miR-221/-222 as regulators of the cardiac response to VM. CONCLUSIONS: The miR-221/-222 cluster orchestrates the antiviral and inflammatory immune response to viral infection of the heart. Its inhibition increases viral load, inflammation, and overall cardiac injury upon VM.


Subject(s)
Coxsackievirus Infections/virology , MicroRNAs/physiology , Myocarditis/virology , Animals , Coxsackievirus Infections/immunology , Humans , Immunity, Cellular/immunology , Macrophages/immunology , Male , Mice, Inbred C3H , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Myocarditis/immunology , Myocytes, Cardiac/immunology , T-Lymphocytes/immunology , Up-Regulation , Viral Load/immunology , Virus Replication/immunology
14.
Eur J Med Chem ; 90: 406-13, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25461889

ABSTRACT

In this study, we demonstrate that N(6)-isopentenyladenosine, which essentially is a plant cytokinin-like compound, exerts a potent and selective antiviral effect on the replication of human enterovirus 71 with an EC50 of 1.0 ± 0.2 µM and a selectivity index (SI) of 5.7. The synthesis of analogs with modification of the N(6)-position did not result in a lower EC50 value. However, in particular with the synthesis of N(6)-(5-hexene-2-yne-1-yl)adenosine (EC50 = 4.3 ± 1.5 µM), the selectivity index was significantly increased: because of a reduction in the adverse effect of this compound on the host cells, an SI > 101 could be calculated. With this study, we for the first time provide proof that a compound class that is based on the plant cytokinin skeleton offers an interesting starting point for the development of novel antivirals against mammalian viruses, in the present context in particular against enterovirus 71.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Isopentenyladenosine/pharmacology , Plants/chemistry , Virus Replication/drug effects , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Humans , Isopentenyladenosine/chemical synthesis , Isopentenyladenosine/chemistry , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
15.
Antimicrob Agents Chemother ; 58(11): 6990-2, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25199773

ABSTRACT

Antivirals against enterovirus 71 (EV71) are urgently needed. We demonstrate that the novel enteroviral protease inhibitor (PI) SG85 and capsid binder (CB) vapendavir efficiently inhibit the in vitro replication of 21 EV71 strains/isolates that are representative of the different genogroups A, B, and C. The PI rupintrivir, the CB pirodavir, and the host-targeting compound enviroxime, which were included as reference compounds, also inhibited the replication of all isolates. Remarkably, the CB compound pleconaril was devoid of any anti-EV71 activity. An in silico docking study revealed that pleconaril-unlike vapendavir and pirodavir-lacks essential binding interactions with the viral capsid. Vapendavir and SG85 (or analogues) should be further explored for the treatment of EV71 infections. The data presented here may serve as a reference when developing yet-novel inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Protease Inhibitors/pharmacology , Virus Replication/drug effects , Benzimidazoles/pharmacology , Capsid/drug effects , Capsid Proteins/metabolism , Drug Resistance, Viral/genetics , Enterovirus A, Human/classification , Enterovirus A, Human/isolation & purification , Enterovirus Infections/drug therapy , Enterovirus Infections/virology , Isoxazoles/pharmacology , Molecular Docking Simulation , Oximes , Phenylalanine/analogs & derivatives , Piperidines/pharmacology , Protein Binding , Pyridazines/pharmacology , Pyrrolidinones/pharmacology , Sulfonamides , Valine/analogs & derivatives
16.
Antiviral Res ; 110: 1-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25043639

ABSTRACT

A novel small molecule, H1PVAT, was identified as a potent and selective inhibitor of the in vitro replication of all three poliovirus serotypes, whereas no activity was observed against other enteroviruses. Time-of-drug-addition studies revealed that the compound interfered with an early stage of virus replication. Four independently-selected H1PVAT-resistant virus variants uniformly carried the single amino acid substitution I194F in the VP1 capsid protein. Poliovirus type 1 strain Sabin, reverse-engineered to contain this substitution, proved to be completely insensitive to the antiviral effect of H1PVAT and was cross-resistant to the capsid-binding inhibitors V-073 and pirodavir. The VP1 I194F mutant had a smaller plaque phenotype than wild-type virus, and the amino acid substitution rendered the virus more susceptible to heat inactivation. Both for the wild-type and VP1 I194F mutant virus, the presence of H1PVAT increased the temperature at which the virus was inactivated, providing evidence that the compound interacts with the viral capsid, and that capsid stabilization and antiviral activity are not necessarily correlated. Molecular modeling suggested that H1PVAT binds with high affinity in the pocket underneath the floor of the canyon that is involved in receptor binding. Introduction of the I194F substitution in the model of VP1 induced a slight concerted rearrangement of the core ß-barrel in this pocket, which disfavors binding of the compound. Taken together, the compound scaffold, to which H1PVAT belongs, may represent another promising class of poliovirus capsid-binding inhibitors next to V-073 and pirodavir. Potent antivirals against poliovirus will be essential in the poliovirus eradication end-game.


Subject(s)
Antiviral Agents/pharmacology , Capsid Proteins/antagonists & inhibitors , Poliomyelitis/drug therapy , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Virus Replication/drug effects , Amino Acid Substitution/genetics , Animals , Base Sequence , Binding Sites , Capsid/drug effects , Capsid Proteins/genetics , Cell Line, Tumor , Chlorocebus aethiops , Drug Resistance, Viral , HeLa Cells , Humans , Models, Molecular , Piperidines/pharmacology , Poliovirus/drug effects , Poliovirus/genetics , Pyridazines/pharmacology , RNA, Viral/genetics , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL
...