Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Type of study
Publication year range
1.
Virus Res ; 297: 198383, 2021 05.
Article in English | MEDLINE | ID: mdl-33705798

ABSTRACT

Slightly acidic hypochlorous acid waters (SAHWs) with pH of 5.2-5.8 containing different concentrations of free available chlorine - 62, 119, 220, 300, and 540 ppm (SAHW-62, -119, -220, -300, and -540, respectively) - were evaluated for their virucidal activity toward a low pathogenic H7N1 avian influenza virus (AIV) and an infectious bronchitis virus (IBV) in suspension, abiotic carrier, and direct spray tests, with the presence of organic materials. In the carrier test, the dropping and wiping techniques were performed toward viruses on carriers. In the suspension test, SAHW-62 could decrease the viral titer of both AIV and IBV by more than 1000 times within 30 s. With the dropping technique, IBV on carriers showed high resistance to SAHW, while AIV on plastic carrier was inactivated to an effective level (≧3 log virus reduction) within 1 min. With the wiping technique, SAHW-62 could inactivate both AIV and IBV on wiped plastic carriers to an effective level within 30 s. However, SAHW-220 could not inactivate IBV in the wiping rayon sheet to an effective level. In the direct spray test, sprayed SAHW-300 within 10 min, and SAHW-540 within 20 min, inactivated AIV and IBV on the rayon sheets to undetectable level, respectively. Our study indicates that the usage of wipes with SAHW could remove viruses from plastic carriers, while viruses remained in the wipes. Besides, a small volume of sprayed SAHW was effective against the viruses on the rayon sheets for daily cleaning in the application area. The findings we obtained concerning IBV might basically be applicable in relation to SARS-CoV-2, given the resemblance between the two viruses.


Subject(s)
Antiviral Agents/pharmacology , Disinfectants/pharmacology , Hypochlorous Acid/pharmacology , Infectious bronchitis virus/drug effects , Influenza A Virus, H7N1 Subtype/drug effects , Animals , Chickens , Coronavirus Infections/prevention & control , Dogs , Ducks , Hepatocytes , Influenza in Birds/prevention & control , Madin Darby Canine Kidney Cells
2.
J Vet Med Sci ; 80(4): 574-577, 2018 Apr 18.
Article in English | MEDLINE | ID: mdl-29540633

ABSTRACT

A quaternary ammonium compound (QAC) was evaluated for its virucidal efficacies with food additive grade calcium hydroxide (FdCa(OH)2). When the QAC was diluted 1:500 (QACx500) with redistilled water (dW2), it inactivated avian influenza virus (AIV) within 30 sec at 25°C, while at 2°C, it required 1 hr for inactivation. When FdCa(OH)2 powder was added to QACx500 at a final concentration of 0.17%, the mixture, namely Mix500, inactivated AIV within 3 min at 2°C. After contamination with 1% fetal bovine serum (FBS), Mix500 inactivated AIV within 2 hr at 2°C, but QACx500 did not. These results indicate synergistic effects of the QAC and FdCa(OH)2 solutions on virucidal activity.


Subject(s)
Calcium Hydroxide/pharmacology , Infectious bursal disease virus/drug effects , Influenza A Virus, H7N1 Subtype/drug effects , Newcastle disease virus/drug effects , Quaternary Ammonium Compounds/pharmacology , Antiviral Agents , Disinfectants , Quaternary Ammonium Compounds/chemistry , Virus Inactivation
3.
J Vet Med Sci ; 80(4): 568-573, 2018 Apr 18.
Article in English | MEDLINE | ID: mdl-29434116

ABSTRACT

An acidic agent, potassium monopersulfate (PMPS), was evaluated for bactericidal and virucidal effects against Salmonella Infantis (SI), Escherichia coli, rifampicin-resistant Salmonella Infantis (SI-rif), Newcastle disease virus (NDV), and avian influenza virus (AIV), in the absence or presence of organic materials. In addition, inactivation activity toward a virus on virus-spiked clothes was also examined. PMPS could inactivate SI, E. coli, and SI-rif even in the presence of organic materials under various concentrations and exposure/contact time conditions. PMPS could also inactivate NDV and AIV. In addition, PMPS could inactivate AIV on a virus-spiked rayon sheet. In conclusion, the present study showed that PMPS has good antimicrobial properties against SI, E. coli, SI-rif, NDV, and AIV when used at the optimal dosage and exposure timing. These results suggest that PMPS could be used as an alternative disinfectant for biosecurity enhancement in animal farms or hospitals.


Subject(s)
Clothing , Escherichia coli/drug effects , Influenza A Virus, H7N1 Subtype/drug effects , Newcastle disease virus/drug effects , Potassium Compounds/pharmacology , Salmonella/drug effects , Sulfates/pharmacology , Animals , Disinfectants/pharmacology , Virus Inactivation/drug effects
4.
Avian Dis ; 62(4): 355-363, 2018 12 01.
Article in English | MEDLINE | ID: mdl-31119919

ABSTRACT

The virucidal efficacies of a 0.2% food additive-grade calcium hydroxide [FdCa(OH)2] solution, a quaternary ammonium compound (QAC) diluted at 1:500 (QACx500), and their mixture [Mix500; FdCa(OH)2 powder added at a final concentration of 0.2% to QACx500] were investigated as fomites for avian influenza virus (AIV) and Newcastle disease virus (NDV) on abiotic carriers (steel, rubber, and plastic) at two different temperatures (room temperature [RT; 25 ± 2 C] and 2 C). These viruses were seeded on coupons (5 cm×5 cm) of rubber, steel, or plastic with 5% fetal bovine serum. After complete drying, the coupons were covered with the test solutions at RT or 2 C. After fixed incubation periods, viruses were recovered from the coupons and titrated. At RT, Mix500 required a short time (3 min) to inactivate AIV and NDV to effective levels (≥3 log virus reduction) on rubber, steel, and plastic carriers compared with QAC or FdCa(OH)2. At low temperature, QACx500 inactivated AIV on steel and plastic carriers to effective levels within 60 min, whereas Mix500 did so within 10 min. QACx500 and FdCa(OH)2 solutions could inactivate NDV on steel and plastic carriers within 20 and 10 min, respectively, and Mix500 could do so within 3 min. Viruses on the carriers required longer incubation periods for inactivation at 2 C than at 25 C. These results demonstrate desirable synergistic virucidal effects of Mix500 for important poultry viruses on abiotic carriers, while indicating high applicability within poultry farming.


Eficacia virucida de un compuesto de amonio cuaternario con hidróxido de calcio con grado aditivo alimentario contra el virus de la influenza aviar y el virus de la enfermedad de Newcastle en vehículo abióticos. Las eficacias virucidas de una solución de hidróxido de calcio [FdCa(OH)2] con grado aditivo alimentario al 0.2%, de un compuesto de amonio cuaternario (QAC) diluido a 1:500 (QACx500) y su mezcla [Mix500; FdCa(OH)2 en polvo agregado en una concentración final de 0.2% en QACx500] se investigaron en forma de fómites para el virus de la influenza aviar (AIV) y para el virus de la enfermedad de Newcastle (NDV) en vehículos abióticos (acero, hule y plástico) a dos temperaturas diferentes (temperatura ambiente [RT; 25 ± 2 C] y 2 C). Estos virus se sembraron en placas (5 cm x 5 cm) de hule, acero o plástico con suero bovino fetal al 5%. Después del secado completo, los cupones se cubrieron con las soluciones bajo ensayo a temperatura ambiente o 2 C. Después de los períodos de incubación fijos, los virus se recuperaron de las placas y se titularon. A temperatura ambiente, la mezcla Mix500 requirió un corto tiempo (tres minutos) para desactivar a los virus de influenza y de Newcastle a niveles efectivos (reducción igual o mayor de tres logaritmos en el título viral) en las placa de hule, acero y plástico en comparación con el tratamiento QAC o FdCa(OH)2. A baja temperatura, el tratamiento QACx500 inactivó al virus de la influenza en placas de acero y plástico a niveles efectivos dentro de 60 minutos, mientras que el tratamiento Mix500 lo hizo en 10 minutos. Las soluciones QACx500 y FdCa(OH)2 pudieron inactivar al virus de Newcastle en las placas de acero y plástico dentro de 20 y 10 minutos, respectivamente, y el tratamiento Mix500 pudo hacerlo dentro de tres minutos. Los virus en los vehículos requirieron períodos de incubación más prolongados para la inactivación a 2 C que a 25 C. Estos resultados demuestran los efectos virucidas sinérgicos deseables del tratamiento Mix500 para virus aviares importantes en vehículos abióticos, además de que indican una alta aplicabilidad dentro de la avicultura.


Subject(s)
Calcium Hydroxide/pharmacology , Fomites/virology , Influenza A Virus, H7N1 Subtype/drug effects , Newcastle disease virus/drug effects , Quaternary Ammonium Compounds/pharmacology , Animals , Calcium Hydroxide/chemistry , Disinfectants/chemistry , Disinfectants/pharmacology , Plastics , Quaternary Ammonium Compounds/chemistry , Rubber , Steel , Temperature
5.
Biomaterials ; 138: 22-34, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28550754

ABSTRACT

Inhibition of influenza A virus infection by multivalent sialic acid inhibitors preventing viral hemagglutinin binding to host cells of the respiratory tract is a promising strategy. However, optimal geometry and optimal ligand presentation on multivalent scaffolds for efficient inhibition both in vitro and in vivo application are still unclear. Here, by comparing linear and dendritic polyglycerol sialosides (LPGSA and dPGSA) we identified architectural requirements and optimal ligand densities for an efficient multivalent inhibitor of influenza virus A/X31/1 (H3N2). Due to its large volume, the LPGSA at optimal ligand density sterically shielded the virus significantly better than the dendritic analog. A statistical mechanics model rationalizes the relevance of ligand density, morphology, and the size of multivalent scaffolds for the potential to inhibit virus-cell binding. Optimized LPGSA inhibited virus infection at IC50 in the low nanomolar nanoparticle concentration range and also showed potent antiviral activity against two avian influenza strains A/Mallard/439/2004 (H3N2) and A/turkey/Italy/472/1999 (H7N1) post infection. In vivo application of inhibitors clearly confirmed the higher inhibition potential of linear multivalent scaffolds to prevent infection. The optimized LPGSA did not show any acute toxicity, and was much more potent than the neuraminidase inhibitor oseltamivir carboxylate in vivo. Combined application of the LPGSA and oseltamivir carboxylate revealed a synergistic inhibitory effect and successfully prevented influenza virus infection in mice.


Subject(s)
Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H7N1 Subtype/drug effects , Influenza in Birds/drug therapy , Influenza, Human/drug therapy , Analysis of Variance , Animals , Antiviral Agents/chemistry , Cell Line , Disease Models, Animal , Dogs , Drug Synergism , Enzyme Inhibitors/pharmacology , Glycerol/chemistry , Humans , Inhibitory Concentration 50 , Madin Darby Canine Kidney Cells , Mice , Models, Statistical , Nanoparticles/chemistry , Oseltamivir/analogs & derivatives , Oseltamivir/pharmacology , Polymers/chemistry , Poultry , Sialic Acids
6.
Antiviral Res ; 139: 41-48, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28012921

ABSTRACT

High morbidity and mortality associated with human cases of highly pathogenic avian influenza (HPAI) viruses, including H5N1 influenza virus, have been reported. The purpose of the present study was to evaluate the antiviral effects of peramivir against HPAI viruses. In neuraminidase (NA) inhibition and virus replication inhibition assays, peramivir showed strong inhibitory activity against H5N1, H7N1 and H7N7 HPAI viruses with sub-nanomolar activity in enzyme assays. In H5N1 viruses containing the NA H275Y mutation, the antiviral activity of peramivir against the variant was lower than that against the wild-type. Evaluation of the in vivo antiviral activity showed that a single intravenous treatment of peramivir (10 mg/kg) prevented lethality in mice infected with wild-type H5N1 virus and also following infection with H5N1 virus with the H275Y mutation after a 5 day administration of peramivir (30 mg/kg). Furthermore, mice injected with peramivir showed low viral titers and low levels of proinflammatory cytokines in the lungs. These results suggest that peramivir has therapeutic activity against HPAI viruses even if the virus harbors the NA H275Y mutation.


Subject(s)
Antiviral Agents/therapeutic use , Cyclopentanes/therapeutic use , Guanidines/therapeutic use , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/genetics , Neuraminidase/genetics , Orthomyxoviridae Infections/drug therapy , Acids, Carbocyclic , Animals , Antiviral Agents/administration & dosage , Cyclopentanes/administration & dosage , Cytokines/immunology , Disease Models, Animal , Guanidines/administration & dosage , Humans , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H7N1 Subtype/drug effects , Influenza A Virus, H7N1 Subtype/enzymology , Influenza A Virus, H7N7 Subtype/drug effects , Influenza A Virus, H7N7 Subtype/enzymology , Influenza, Human/drug therapy , Lung/immunology , Lung/virology , Mice , Mutation , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/virology , Viral Load/drug effects , Virus Replication/drug effects
7.
Antiviral Res ; 120: 89-100, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26022200

ABSTRACT

Antiviral medication is used for the treatment of severe influenza infections, of which the neuraminidase inhibitors (NAIs) are the most effective drugs, approved so far. Here, we investigated the antiviral efficacy of the peptidomimetic furin inhibitor MI-701 in combination with oseltamivir carboxylate and ribavirin against the infection of highly pathogenic avian influenza viruses (HPAIV) that are activated by the host protease furin. Cell cultures infected with the strains A/Thailand/1(KAN-1)/2004 (H5N1) and A/FPV/Rostock/1934 (H7N1) were treated with each agent alone, or in double and triple combinations. MI-701 alone achieved a concentration-dependent reduction of virus propagation. Double treatment of MI-701 with oseltamivir carboxylate and triple combination with ribavirin showed synergistic inhibition and a pronounced delay of virus propagation. MI-701 resistant mutants were not observed. Emergence of NA mutation H275Y conferring high oseltamivir resistance was significantly delayed in the presence of MI-701. Our data indicate that combination with a potent furin inhibitor significantly enhances the therapeutic efficacy of conventional antivirals drugs against HPAIV infection.


Subject(s)
Antiviral Agents/metabolism , Furin/antagonists & inhibitors , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H7N1 Subtype/drug effects , Oseltamivir/metabolism , Peptidomimetics/metabolism , Ribavirin/metabolism , Animals , Dogs , Drug Resistance, Viral , Drug Synergism , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H7N1 Subtype/growth & development , Madin Darby Canine Kidney Cells , Microbial Sensitivity Tests , Mutant Proteins/genetics , Mutation, Missense , Neuraminidase/genetics , Viral Proteins/genetics
8.
Antimicrob Agents Chemother ; 59(2): 1061-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25451059

ABSTRACT

The emergence of drug-resistant influenza A virus (IAV) strains represents a serious threat to global human health and underscores the need for novel approaches to anti-influenza chemotherapy. Combination therapy with drugs affecting different IAV targets represents an attractive option for influenza treatment. We have previously shown that the thiazolide anti-infective nitazoxanide (NTZ) inhibits H1N1 IAV replication by selectively blocking viral hemagglutinin maturation. Herein we investigate the anti-influenza activity of NTZ against a wide range of human and avian IAVs (H1N1, H3N2, H5N9, H7N1), including amantadine-resistant and oseltamivir-resistant strains, in vitro. We also investigate whether therapy with NTZ in combination with the neuraminidase inhibitors oseltamivir and zanamivir exerts synergistic, additive, or antagonistic antiviral effects against influenza viruses. NTZ was effective against all IAVs tested, with 50% inhibitory concentrations (IC50s) ranging from 0.9 to 3.2 µM, and selectivity indexes (SIs) ranging from >50 to >160, depending on the strain and the multiplicity of infection (MOI). Combination therapy studies were performed in cell culture-based assays using A/Puerto Rico/8/1934 (H1N1), A/WSN/1933 (H1N1), or avian A/chicken/Italy/9097/1997 (H5N9) IAVs; dose-effect analysis and synergism/antagonism quantification were performed using isobologram analysis according to the Chou-Talalay method. Combination index (CI) analysis indicated that NTZ and oseltamivir combination treatment was synergistic against A/Puerto Rico/8/1934 (H1N1) and A/WSN/1933 (H1N1) IAVs, with CI values ranging between 0.39 and 0.63, independently of the MOI used. Similar results were obtained when NTZ was administered in combination with zanamivir (CI=0.3 to 0.48). NTZ-oseltamivir combination treatment was synergistic also against the avian A/chicken/Italy/9097/1997 (H5N9) IAV (CI=0.18 to 0.31). Taken together, the results suggest that regimens that combine neuraminidase inhibitors and nitazoxanide exert synergistic anti-influenza effects.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/pathogenicity , Neuraminidase/antagonists & inhibitors , Thiazoles/pharmacology , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A Virus, H7N1 Subtype/drug effects , Influenza A Virus, H7N1 Subtype/pathogenicity , Influenza A virus/drug effects , Nitro Compounds , Oseltamivir/pharmacology , Zanamivir/pharmacology
9.
J Vet Med Sci ; 77(2): 211-5, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25421399

ABSTRACT

Hypochlorous acid (HOCl) solutions were evaluated for their virucidal ability against a low pathogenic avian influenza virus (AIV), H7N1. HOCl solutions containing 50, 100 and 200 ppm chlorine (pH 6) or their sprayed solutions (harvested in dishes placed at 1 or 30 cm distance between the spray nozzle and dish) were mixed with the virus with or without organic materials (5% fetal bovine serum: FBS). Under plain diluent conditions (without FBS), harvested solutions of HOCl after spraying could decrease the AIV titer by more than 1,000 times, to an undetectable level (< 2.5 log10TCID50/ml) within 5 sec, with the exception of the 50 ppm solution harvested after spraying at the distance of 30 cm. Under the dirty conditions (in the presence of 5% FBS), they lost their virucidal activity. When HOCl solutions were sprayed directly on the virus on rayon sheets for 10 sec, the solutions of 100 and 200 ppm could inactivate AIV immediately after spraying, while 50 ppm solution required at least 3 min of contact time. In the indirect spray form, after 10 sec of spraying, the lids of the dishes were opened to expose the virus on rayon sheets to HOCl. In this form, the 200 ppm solution inactivated AIV within 10 min of contact, while 50 and 100 ppm could not inactivate it. These data suggest that HOCl can be used in spray form to inactivate AIV at the farm level.


Subject(s)
Disinfectants/pharmacology , Hypochlorous Acid/pharmacology , Influenza A Virus, H7N1 Subtype/drug effects , Aerosols , Surface Properties
10.
Article in English | MEDLINE | ID: mdl-25151089

ABSTRACT

Influenza A viruses (IAV) have the potential to cause devastating pandemics. In recent years, the emergence of new avian strains able to infect humans represents a serious threat to global human health. The increase in drug-resistant IAV strains underscores the need for novel approaches to anti-influenza chemotherapy. Herein we show that prostaglandin-A1 (PGA1) possesses antiviral activity against avian IAV, including H5N9, H7N1 and H1N1 strains, acting at a level different from the currently available anti-influenza drugs. PGA1 acts at postentry level, causing dysregulation of viral protein synthesis and preventing virus-induced disassembly of host microtubular network and activation of pro-inflammatory factor NF-κB. The antiviral activity is dependent on the presence of a cyclopentenone ring structure and is associated with activation of a cytoprotective heat shock response in infected cells. The results suggest that cyclopentenone prostanoids or prostanoids-derived molecules may represent a new tool to combat avian influenza virus infection.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , NF-kappa B/drug effects , Prostaglandins A/pharmacology , Viral Proteins/biosynthesis , Virus Replication/drug effects , Animals , Cell Line , Chickens , Dogs , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H7N1 Subtype/drug effects , Influenza A Virus, H7N1 Subtype/physiology , Influenza A virus/physiology , Madin Darby Canine Kidney Cells , NF-kappa B/metabolism , Pulmonary Alveoli
11.
Eur J Med Chem ; 83: 601-8, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-24997292

ABSTRACT

Triterpene derivatives with an α,ß-alkenenitrile moiety in the five-membered ring A have been synthesized by nitrile anion cyclizations of 1-cyano-2,3-secotriterpenoids. Oxime-containing precursors, 2,3-secointermediates and five-membered ring A products of cyclizations were screened for in vitro antiviral activity against enveloped viruses - influenza A virus and human immunodeficiency virus type I (HIV-1). Lupane ketoxime and the 2,3-secolupane C-3 aldoxime which possess antiviral activities against both influenza A virus (EC50 12.9-18.2 µM) and HIV-1 (EC50 0.06 µM) were the most promising compounds.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Terpenes/chemistry , Terpenes/pharmacology , Cyclization , HIV-1/drug effects , Influenza A Virus, H7N1 Subtype/drug effects , Models, Molecular , Molecular Conformation , Oximes/chemistry
12.
Bioorg Med Chem ; 20(6): 2152-7, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22342267

ABSTRACT

Evidences of oseltamivir resistant influenza patients raised the need of novel neuraminidase inhibitors. In this study, five oseltamivir analogs PMC-31-PMC-36, synthesised according to the outcomes of a rational design analysis aimed to investigate the effects of substitution at the 5-amino and 4-amido groups of oseltamivir on its antiviral activity, were screened for their inhibition against neuraminidase N1 and N3. The enzymes used as models were from the avian influenza A H7N1 and H7N3 viruses. The neuraminidase inhibition assay was carried out by using recombinant species obtained from a baculovirus expression system and the fluorogenic substrate MUNANA. The assay was validated by using oseltamivir carboxylate as a reference inhibitor. Among the tested compounds, PMC-36 showed the highest inhibition on N1 with an IC(50) of 14.6±3.0nM (oseltamivir 25±4nM), while PMC-35 showed a significant inhibitory effect on N3 with an IC(50) of 0.1±0.03nM (oseltamivir 0.2±0.02nM). The analysis of the inhibitory properties of this panel of compounds allowed a preliminary assessment of a structure-activity relationship for the modification of the 4-amido and 5-amino groups of oseltamivir carboxylate. The substitution of the acetamido group in the oseltamivir structure with a 2-butenylamido moiety reduced the observed activity, while the introduction of a propenylamido group was well tolerated. Substitution of the free 5-amino group of oseltamivir carboxylate with an azide, decreased the activity against both N1 and N3. When these structural changes were both introduced, a dramatic reduction of activity was observed for both N1 and N3. The alkylation of the free 5-amino group in oseltamivir carboxylate introducing an isopropyl group seemed to increase the inhibitory effect for both N1 and N3 neuraminidases, displaying a more pronounced effect against N1.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Influenza A virus/enzymology , Influenza in Birds/drug therapy , Neuraminidase/antagonists & inhibitors , Oseltamivir/analogs & derivatives , Oseltamivir/pharmacology , Animals , Antiviral Agents/chemical synthesis , Binding Sites , Birds/virology , Influenza A Virus, H7N1 Subtype/chemistry , Influenza A Virus, H7N1 Subtype/drug effects , Influenza A Virus, H7N1 Subtype/enzymology , Influenza A Virus, H7N3 Subtype/chemistry , Influenza A Virus, H7N3 Subtype/drug effects , Influenza A Virus, H7N3 Subtype/enzymology , Influenza A virus/chemistry , Influenza A virus/drug effects , Influenza in Birds/enzymology , Models, Molecular , Neuraminidase/chemistry , Neuraminidase/metabolism , Oseltamivir/chemical synthesis
13.
J Virol ; 85(4): 1554-62, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21123387

ABSTRACT

Influenza A viruses constitute a major and ongoing global public health concern. Current antiviral strategies target viral gene products; however, the emergence of drug-resistant viruses highlights the need for novel antiviral approaches. Cleavage of the influenza virus hemagglutinin (HA) by host cell proteases is crucial for viral infectivity and therefore presents a potential drug target. Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO) are single-stranded-DNA-like antisense agents that readily enter cells and can act as antisense agents by sterically blocking cRNA. Here, we evaluated the effect of PPMO targeted to regions of the pre-mRNA or mRNA of the HA-cleaving protease TMPRSS2 on proteolytic activation and spread of influenza viruses in human Calu-3 airway epithelial cells. We found that treatment of cells with a PPMO (T-ex5) designed to interfere with TMPRSS2 pre-mRNA splicing resulted in TMPRSS2 mRNA lacking exon 5 and consequently the expression of a truncated and enzymatically inactive form of TMPRSS2. Altered splicing of TMPRSS2 mRNA by the T-ex5 PPMO prevented HA cleavage in different human seasonal and pandemic influenza A viruses and suppressed viral titers by 2 to 3 log(10) units, strongly suggesting that TMPRSS2 is responsible for HA cleavage in Calu-3 airway cells. The data indicate that PPMO provide a useful reagent for investigating HA-activating proteases and may represent a promising strategy for the development of novel therapeutics to address influenza infections.


Subject(s)
Epithelial Cells/virology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/drug effects , Influenza A virus/pathogenicity , Metalloendopeptidases/metabolism , Morpholines/pharmacology , Serine Endopeptidases/metabolism , Animals , Bronchi/cytology , Cell Line , Cells, Cultured , Chick Embryo , Dogs , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H7N1 Subtype/drug effects , Influenza A Virus, H7N1 Subtype/enzymology , Influenza A virus/enzymology , Morpholinos , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...