Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
ACS Appl Bio Mater ; 5(11): 5174-5180, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36240051

ABSTRACT

The COVID-19 pandemic has increased public health vigilance worldwide. The coronavirus (SARS-CoV-2) can spread via aerosols, and droplet-borne viruses remain viable on nonliving surfaces for long duration. Hence, effective antiviral coatings are highly useful in eliminating viral persistence on nonliving surfaces. Although innovative antiviral coatings have been designed, conventional procedures for antiviral assays are generally laborious, time-consuming, and have a high limit of detection. In the present study, we report a rapid and highly sensitive method for evaluating antiviral coatings by measuring the luciferase activity derived from recombinant Sendai virus (SeV). The physicochemical characteristics of SeV, which has a single-stranded RNA genome encapsulated within a lipid envelope, allow us to exploit it as an indicator of the physicochemical potential of coating materials against enveloped RNA viruses in general. We demonstrate that SeV-based assay systems allow for the rapid and quantitative evaluation of the surface coatings composed of iodine solubilized in polyvinyl acetate. Additionally, we have investigated the effect of mucins, the dominant protein component of saliva, on the antiviral activity of surface coatings. The presence of mucins in the SeV suspension considerably rescues luciferase activity at the viral-surface interface, presumably due to mucin-mediated viral protection. Our findings provide insights into a procedure capable of the rapid evaluation and optimization of surface coatings, and suggest an important role of the mucin in the valid evaluation of antiviral agents.


Subject(s)
Antiviral Agents , Sendai virus , Antiviral Agents/pharmacology , Luciferases , Mucins , Sendai virus/drug effects
2.
FEBS Open Bio ; 11(5): 1452-1464, 2021 05.
Article in English | MEDLINE | ID: mdl-33822489

ABSTRACT

Human pathogenic RNA viruses are threats to public health because they are prone to escaping the human immune system through mutations of genomic RNA, thereby causing local outbreaks and global pandemics of emerging or re-emerging viral diseases. While specific therapeutics and vaccines are being developed, a broad-spectrum therapeutic agent for RNA viruses would be beneficial for targeting newly emerging and mutated RNA viruses. In this study, we conducted a screen of repurposed drugs using Sendai virus (an RNA virus of the family Paramyxoviridae), with human-induced pluripotent stem cells (iPSCs) to explore existing drugs that may present anti-RNA viral activity. Selected hit compounds were evaluated for their efficacy against two important human pathogens: Ebola virus (EBOV) using Huh7 cells and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) using Vero E6 cells. Selective estrogen receptor modulators (SERMs), including raloxifene, exhibited antiviral activities against EBOV and SARS-CoV-2. Pioglitazone, a PPARγ agonist, also exhibited antiviral activities against SARS-CoV-2, and both raloxifene and pioglitazone presented a synergistic antiviral effect. Finally, we demonstrated that SERMs blocked entry steps of SARS-CoV-2 into host cells. These findings suggest that the identified FDA-approved drugs can modulate host cell susceptibility against RNA viruses.


Subject(s)
Antiviral Agents/pharmacology , Drug Repositioning , RNA Viruses/drug effects , RNA, Viral/antagonists & inhibitors , SARS-CoV-2/drug effects , Animals , Cell Line , Chlorocebus aethiops , Drug Repositioning/methods , Ebolavirus/drug effects , Ebolavirus/physiology , Humans , Induced Pluripotent Stem Cells/virology , Microbial Sensitivity Tests/methods , Pioglitazone/pharmacology , RNA Viruses/physiology , Raloxifene Hydrochloride/pharmacology , SARS-CoV-2/physiology , Selective Estrogen Receptor Modulators/pharmacology , Sendai virus/drug effects , Sendai virus/physiology , Vero Cells , COVID-19 Drug Treatment
3.
Lasers Med Sci ; 36(4): 743-750, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32592133

ABSTRACT

To enhance the potency of photosensitizer, we developed a novel photosensitizer, Laserphyrin®-HVJ-E (L-HVJ-E), by incorporating talaporfin sodium (Laserphyrin®, Meiji Seika Pharma) into hemagglutinating virus of Japan envelope (HVJ-E). In this study, we examined the optimal Laserphyrin® concentration for preparation of Laserphyrin®-HVJ-E which had photocytotoxicity and maintained direct cytotoxicity derived from HVJ-E. Then, potency of Laserphyrin®-HVJ-E and Laserphyrin® were compared in vitro using castration-resistant prostate cancer cell line (PC-3). A laser diode (L660P120, Thorlabs, USA) with a wavelength of 664 nm was used for light activation of Laserphyrin®, which corresponds to an absorption peak of Laserphyrin® and provides a high therapeutic efficiency. The photocytotoxicity and direct cytotoxicity of Laserphyrin®-HVJ-E prepared using various Laserphyrin® concentrations were evaluated using PC-3 cell in vitro. We categorized the treatment groups as Group 1: 50 µL of D-MEM treatment group, Group 2: HVJ-E treatment group, Group 3: Laserphyrin®-HVJ-E treatment group, and Group 4: Laserphyrin® treatment group. Group 3 was subjected to different concentrations of Laserphyrin®-HVJ-E suspension, and all groups were subjected to different incubation periods (24, 48 h), (30 min, 1 h, or 3 h,) respectively, without and after PDT. Laserphyrin®-HVJ-E prepared using 15 mM Laserphyrin® had high photocytotoxicity and maintained HVJ-E's ability to induce direct cytotoxicity. Therapeutic effect of Laserphyrin®-HVJ-E was substantially equivalent to that of Laserphyrin® alone even at half Laserphyrin® concentration. By utilizing Laserphyrin®-HVJ-E, PDT could be performed with lower Laserphyrin® concentration. In addition, Laserphyrin®-HVJ-E showed higher potency than Laserphyrin® by combining cytotoxicities of HVJ-E and PDT.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Photochemotherapy , Porphyrins/therapeutic use , Prostatic Neoplasms/drug therapy , Virion/physiology , Animals , Antineoplastic Agents/therapeutic use , Humans , Lasers, Semiconductor , Male , PC-3 Cells , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Sendai virus/drug effects
4.
Am J Respir Cell Mol Biol ; 63(6): 758-766, 2020 12.
Article in English | MEDLINE | ID: mdl-32853024

ABSTRACT

Viral pneumonias remain global health threats, as exemplified in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, requiring novel treatment strategies both early and late in the disease process. We have reported that mice treated before or soon after infection with a combination of inhaled Toll-like receptor (TLR) 2/6 and 9 agonists (Pam2-ODN) are broadly protected against microbial pathogens including respiratory viruses, but the mechanisms remain incompletely understood. The objective of this study was to validate strategies for immune modulation in a preclinical model of viral pneumonia and determine their mechanisms. Mice were challenged with the Sendai paramyxovirus in the presence or absence of Pam2-ODN treatment. Virus burden and host immune responses were assessed to elucidate Pam2-ODN mechanisms of action and to identify additional opportunities for therapeutic intervention. Enhanced survival of Sendai virus pneumonia with Pam2-ODN treatment was associated with reductions in lung virus burden and with virus inactivation before internalization. We noted that mortality in sham-treated mice corresponded with CD8+ T-cell lung inflammation on days 11-12 after virus challenge, after the viral burden had declined. Pam2-ODN blocked this injurious inflammation by minimizing virus burden. As an alternative intervention, depleting CD8+ T cells 8 days after viral challenge also decreased mortality. Stimulation of local innate immunity within the lungs by TLR agonists early in disease or suppression of adaptive immunity by systemic CD8+ T-cell depletion late in disease improves outcomes of viral pneumonia in mice. These data reveal opportunities for targeted immunomodulation to protect susceptible human subjects.


Subject(s)
Immunity, Innate/immunology , Lipopeptides/pharmacology , Pneumonia, Viral/drug therapy , Pneumonia/prevention & control , Respirovirus Infections/drug therapy , Sendai virus/drug effects , Viral Load/drug effects , Animals , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Female , Immunity, Innate/drug effects , Lung/drug effects , Lung/immunology , Lung/virology , Mice , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/pathology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Respirovirus Infections/immunology , Respirovirus Infections/virology , Sendai virus/immunology
5.
Virol J ; 16(1): 150, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31791359

ABSTRACT

BACKGROUND: Commercially available antiviral drugs, when used in the treatment of viral infections, do not always result in success. This is an urgent problem currently that needs to be addressed because several viruses including influenza and paramyxoviruses are acquiring multi-drug resistance. A potential solution for this emerging issue is to create new antiviral drugs from available compounds of natural products. It is known that the majority of drugs have been developed using compounds derived from actinomycetes, which are naturally occurring gram-positive bacteria. The purpose of this study was to investigate the antiviral properties of extremophilic actinomycetes extracts from strains that were isolated from extreme environments in Kazakhstan. METHODS: Five strains of extremophilic actinomycetes isolated from the unique ecosystems of Kazakhstan were extracted and tested for antiviral activity against influenza viruses (strains H7N1, H5N3, H1N1 and H3N2) and paramyxoviruses (Sendai Virus and Newcastle Disease Virus). The antiviral activity of these selected extracts was tested by checking their effect on hemagglutination and neuraminidase activities of the studied viruses. Additionally, actinomycetes extracts were compared with commercially available antiviral drugs and some plant preparations that have been shown to exhibit antiviral properties. RESULTS: The main findings show that extracts from strains K-192, K-340, K-362, K-522 and K525 showed antiviral activities when tested using influenza viruses, Sendai Virus, and Newcastle Disease Virus. These activities were comparable to those shown by Rimantadine and Tamiflu drugs, and "Virospan" and "Flavovir" plant preparations. CONCLUSIONS: We identified several extracts with antiviral activities against several strains of influenza viruses and paramyxoviruses. Our research findings can be applied towards characterization and development of new antiviral drugs from the active actinomycetes extracts.


Subject(s)
Actinobacteria/chemistry , Antiviral Agents/pharmacology , Biological Products/pharmacology , Influenza A virus/drug effects , Actinobacteria/isolation & purification , Animals , Antiviral Agents/isolation & purification , Biological Products/isolation & purification , Complex Mixtures/isolation & purification , Complex Mixtures/pharmacology , Hemagglutination , Kazakhstan , Microbial Sensitivity Tests , Neuraminidase/analysis , Newcastle disease virus/drug effects , Sendai virus/drug effects
6.
J Biol Chem ; 293(27): 10561-10573, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29802199

ABSTRACT

Host recognition of intracellular viral RNA and subsequent induction of cytokine signaling are tightly regulated at the cellular level and are a target for manipulation by viruses and therapeutics alike. Here, we characterize chromosome 6 ORF 106 (C6orf106) as an evolutionarily conserved inhibitor of the innate antiviral response. C6orf106 suppresses the synthesis of interferon (IFN)-α/ß and proinflammatory tumor necrosis factor (TNF) α in response to the dsRNA mimic poly(I:C) and to Sendai virus infection. Unlike canonical inhibitors of antiviral signaling, C6orf106 blocks interferon-regulatory factor 3 (IRF3) and, to a lesser extent, NF-κB activity without modulating their activation, nuclear translocation, cellular expression, or degradation. Instead, C6orf106 interacts with IRF3 and inhibits IRF3 recruitment to type I IFN promoter sequences while also reducing the nuclear levels of the coactivator proteins p300 and CREB-binding protein (CBP). In summary, we have defined C6orf106 as a negative regulator of antiviral immunity that blocks IRF3-dependent cytokine production via a noncanonical and poorly defined mechanism. This work presents intriguing implications for antiviral immunity, autoimmune disorders, and cancer.


Subject(s)
Antiviral Agents/pharmacology , Immunity, Innate/immunology , Interferon Regulatory Factor-3/antagonists & inhibitors , Neoplasm Proteins/pharmacology , Respirovirus Infections/prevention & control , Sendai virus/immunology , Animals , Antiviral Agents/administration & dosage , Chlorocebus aethiops , Gene Expression Regulation , HeLa Cells , Humans , Immunity, Innate/drug effects , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/administration & dosage , Respirovirus Infections/immunology , Respirovirus Infections/virology , Sendai virus/drug effects , Signal Transduction , Vero Cells
7.
Sci Adv ; 2(7): e1501889, 2016 07.
Article in English | MEDLINE | ID: mdl-27419230

ABSTRACT

Cytosolic RNA sensing is a prerequisite for initiation of innate immune response against RNA viral pathogens. Signaling through RIG-I (retinoic acid-inducible gene I)-like receptors (RLRs) to TBK1 (Tank-binding kinase 1)/IKKε (IκB kinase ε) kinases is transduced by mitochondria-associated MAVS (mitochondrial antiviral signaling protein). However, the precise mechanism of how MAVS-mediated TBK1/IKKε activation is strictly controlled still remains obscure. We reported that protein phosphatase magnesium-dependent 1A (PPM1A; also known as PP2Cα), depending on its catalytic ability, dampened the RLR-IRF3 (interferon regulatory factor 3) axis to silence cytosolic RNA sensing signaling. We demonstrated that PPM1A was an inherent partner of the TBK1/IKKε complex, targeted both MAVS and TBK1/IKKε for dephosphorylation, and thus disrupted MAVS-driven formation of signaling complex. Conversely, a high level of MAVS can dissociate the TBK1/PPM1A complex to override PPM1A-mediated inhibition. Loss of PPM1A through gene ablation in human embryonic kidney 293 cells and mouse primary macrophages enabled robustly enhanced antiviral responses. Consequently, Ppm1a(-/-) mice resisted to RNA virus attack, and transgenic zebrafish expressing PPM1A displayed profoundly increased RNA virus vulnerability. These findings identify PPM1A as the first known phosphatase of MAVS and elucidate the physiological function of PPM1A in antiviral immunity on whole animals.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytosol/metabolism , Protein Phosphatase 2C/genetics , Protein Serine-Threonine Kinases/metabolism , RNA/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Genetically Modified/metabolism , CRISPR-Cas Systems/genetics , Cell Line , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/virology , HEK293 Cells , Humans , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Protein Phosphatase 2C/antagonists & inhibitors , Protein Phosphatase 2C/metabolism , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism , Sendai virus/drug effects , Sendai virus/pathogenicity , Sendai virus/physiology , Vesiculovirus/drug effects , Vesiculovirus/pathogenicity , Vesiculovirus/physiology , Zebrafish/growth & development , Zebrafish/metabolism
8.
Virology ; 485: 340-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26331681

ABSTRACT

Vesicular stomatitis virus (VSV) is an effective oncolytic virus against most human pancreatic ductal adenocarcinoma (PDAC) cell lines. However, some PDAC cell lines are highly resistant to oncolytic VSV-ΔM51 infection. To better understand the mechanism of resistance, we tested a panel of 16 small molecule inhibitors of different cellular signaling pathways, and identified TPCA-1 (IKK-ß inhibitor) and ruxolitinib (JAK1/2 inhibitor), as strong enhancers of VSV-ΔM51 replication and virus-mediated oncolysis in all VSV-resistant PDAC cell lines. Both TPCA-1 and ruxolitinib similarly inhibited STAT1 and STAT2 phosphorylation and decreased expression of antiviral genes MxA and OAS. Moreover, an in situ kinase assay provided biochemical evidence that TPCA-1 directly inhibits JAK1 kinase activity. Together, our data demonstrate that TPCA-1 is a unique dual inhibitor of IKK-ß and JAK1 kinase, and provide a new evidence that upregulated type I interferon signaling plays a major role in resistance of pancreatic cancer cells to oncolytic viruses.


Subject(s)
Amides/pharmacology , I-kappa B Kinase/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/virology , Protein Kinase Inhibitors/pharmacology , Thiophenes/pharmacology , Vesicular stomatitis Indiana virus/physiology , Cell Line, Tumor , Cytopathogenic Effect, Viral/drug effects , Humans , Interferon Type I/metabolism , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Myxovirus Resistance Proteins/genetics , Nitriles , Oncolytic Viruses/drug effects , Oncolytic Viruses/physiology , Pyrazoles/pharmacology , Pyrimidines , STAT Transcription Factors/metabolism , Sendai virus/drug effects , Sendai virus/physiology , Signal Transduction/drug effects , Vesicular stomatitis Indiana virus/drug effects , Virus Replication/drug effects
9.
Org Lett ; 17(12): 3098-101, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26068271

ABSTRACT

Two new heterodimeric sesquiterpenes, sterhirsutins C (1) and D (2), along with eight new sesquiterpenoid derivatives, sterhirsutins E--L (3-10), were isolated from the culture of Stereum hirsutum. The absolute configuration of 1 was assigned by a single-crystal X-ray diffraction experiment. Compounds 1 and 2 possessed an unprecedented chemical skeleton with a 5/5/5/6/9/4 fused ring system. Compound 10 is the first sesquiterpene coupled with a xanthine moiety. Compounds 1-10 showed cytotoxicity against K562 and HCT116 cell lines. Compound 9 induced autophagy in HeLa cells. Compound 5 inhibited the activation of IFNß promoter in Sendai virus infected cells.


Subject(s)
Antineoplastic Agents/chemistry , Basidiomycota/chemistry , Fungi/chemistry , HCT116 Cells/chemistry , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Interferon-beta/chemistry , Sendai virus/chemistry , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Xanthine/chemistry , Antineoplastic Agents/pharmacology , Crystallography, X-Ray , HCT116 Cells/drug effects , HeLa Cells , Humans , Interferon-beta/pharmacology , K562 Cells , Magnetic Resonance Spectroscopy , Molecular Structure , Sendai virus/drug effects , Tibet
10.
J Mol Cell Biol ; 6(2): 154-63, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24755855

ABSTRACT

RIG-I is a pivotal cytoplasmic sensor that recognizes different species of viral RNAs. This recognition leads to activation of the transcription factors NF-κB and IRF3, which collaborate to induce type I interferons (IFNs) and innate antiviral response. In this study, we identified the TRIM family protein TRIM4 as a positive regulator of RIG-I-mediated IFN induction. Overexpression of TRIM4 potentiated virus-triggered activation of IRF3 and NF-κB, as well as IFN-ß induction, whereas knockdown of TRIM4 had opposite effects. Mechanistically, TRIM4 associates with RIG-I and targets it for K63-linked polyubiquitination. Our findings demonstrate that TRIM4 is an important regulator of the virus-induced IFN induction pathways by mediating RIG-I for K63-linked ubiquitination.


Subject(s)
Carrier Proteins/metabolism , DEAD-box RNA Helicases/metabolism , Interferon-beta/pharmacology , Lysine/metabolism , Sendai virus/physiology , Ubiquitination/drug effects , Cell Line , DEAD Box Protein 58 , Gene Knockdown Techniques , HEK293 Cells , Humans , Polyubiquitin/metabolism , Protein Binding/drug effects , Receptors, Immunologic , Sendai virus/drug effects , Signal Transduction/drug effects , Transcription Factors/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism
11.
J Biol Chem ; 288(12): 8061-8073, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23362274

ABSTRACT

Influenza is a severe disease in humans and animals with few effective therapies available. All strains of influenza virus are prone to developing drug resistance due to the high mutation rate in the viral genome. A therapeutic agent that targets a highly conserved region of the virus could bypass resistance and also be effective against multiple strains of influenza. Influenza uses many individually weak ligand binding interactions for a high avidity multivalent attachment to sialic acid-bearing cells. Polymerized sialic acid analogs can form multivalent interactions with influenza but are not ideal therapeutics due to solubility and toxicity issues. We used liposomes as a novel means for delivery of the glycan sialylneolacto-N-tetraose c (LSTc). LSTc-bearing decoy liposomes form multivalent, polymer-like interactions with influenza virus. Decoy liposomes competitively bind influenza virus in hemagglutination inhibition assays and inhibit infection of target cells in a dose-dependent manner. Inhibition is specific for influenza virus, as inhibition of Sendai virus and respiratory syncytial virus is not observed. In contrast, monovalent LSTc does not bind influenza virus or inhibit infectivity. LSTc decoy liposomes prevent the spread of influenza virus during multiple rounds of replication in vitro and extend survival of mice challenged with a lethal dose of virus. LSTc decoy liposomes co-localize with fluorescently tagged influenza virus, whereas control liposomes do not. Considering the conservation of the hemagglutinin binding pocket and the ability of decoy liposomes to form high avidity interactions with influenza hemagglutinin, our decoy liposomes have potential as a new therapeutic agent against emerging influenza strains.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , Influenza, Human/drug therapy , Polysaccharides/pharmacology , Sialic Acids/pharmacology , Animals , Antiviral Agents/administration & dosage , Cell Line , Chlorocebus aethiops , Dogs , Drug Evaluation, Preclinical , Epithelial Cells/drug effects , Epithelial Cells/virology , Female , Hemagglutination/drug effects , Humans , Influenza A virus/physiology , Liposomes , Mice , Mice, Inbred C57BL , Polysaccharides/administration & dosage , Rous sarcoma virus/drug effects , Sendai virus/drug effects , Sialic Acids/administration & dosage , Vero Cells , Virus Replication/drug effects
12.
Arch Virol ; 156(5): 793-801, 2011 May.
Article in English | MEDLINE | ID: mdl-21286764

ABSTRACT

Parainfluenza viruses are significant respiratory-tract pathogens that are notorious for infecting children. However, there are no clinical drugs to control the infection caused by these viruses. Sendai virus (SeV) belongs to the family Paramyxoviridae and causes fatal pneumonia in mice, its natural host. Baicalein is a flavonoid derived from the root of Scutellaria baicalensis, which is a traditional Chinese medicine that has been used for hundreds of years and has demonstrated a variety of biological activities. Our findings reveal that oral administration of baicalein to mice infected with Sendai virus results in a significant reduction in virus titers in the lungs and protection from death. The in vivo inhibitory effects of baicalein on Sendai virus are determined by baicalin in the serum. The mean IC(50) of baicalin was 0.71 µg/ml in an HA inhibition assay and 3.22 µg/ml in an NA inhibition assay. The mean IC(50) of baicalin in a CPE assay was measured to be 0.70 µg/ml, and significant inhibition was observed in a plaque assay at a concentration of 1.6 µg/ml baicalin in overlay medium, which suggests that baicalein is a potential anti-parainfluenzaviral agent in vivo.


Subject(s)
Antiviral Agents/administration & dosage , Flavanones/administration & dosage , Flavonoids/analysis , Hemagglutinins/metabolism , Neuraminidase/antagonists & inhibitors , Sendai virus/drug effects , Serum/chemistry , Administration, Oral , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Female , Flavanones/pharmacokinetics , Flavanones/pharmacology , Inhibitory Concentration 50 , Lung/virology , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Viral Load , Viral Plaque Assay
13.
PLoS One ; 5(6): e11265, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20582319

ABSTRACT

Ribavirin (RBV) is a synthetic nucleoside analog with broad spectrum antiviral activity. Although RBV is approved for the treatment of hepatitis C virus, respiratory syncytial virus, and Lassa fever virus infections, its mechanism of action and therapeutic efficacy remains highly controversial. Recent reports show that the development of cell-based resistance after continuous RBV treatment via decreased RBV uptake can greatly limit its efficacy. Here, we examined whether certain cell types are naturally resistant to RBV even without prior drug exposure. Seven different cell lines from various host species were compared for RBV antiviral activity against two nonsegmented negative-strand RNA viruses, vesicular stomatitis virus (VSV, a rhabdovirus) and Sendai virus (SeV, a paramyxovirus). Our results show striking differences between cell types in their response to RBV, ranging from virtually no antiviral effect to very effective inhibition of viral replication. Despite differences in viral replication kinetics for VSV and SeV in the seven cell lines, the observed pattern of RBV resistance was very similar for both viruses, suggesting that cellular rather than viral determinants play a major role in this resistance. While none of the tested cell lines was defective in RBV uptake, dramatic variations were observed in the long-term accumulation of RBV in different cell types, and it correlated with the antiviral efficacy of RBV. While addition of guanosine neutralized RBV only in cells already highly resistant to RBV, actinomycin D almost completely reversed the RBV effect (but not uptake) in all cell lines. Together, our data suggest that RBV may inhibit the same virus via different mechanisms in different cell types depending on the intracellular RBV metabolism. Our results strongly point out the importance of using multiple cell lines of different origin when antiviral efficacy and potency are examined for new as well as established drugs in vitro.


Subject(s)
Antiviral Agents/pharmacology , Ribavirin/pharmacology , Sendai virus/drug effects , Vesiculovirus/drug effects , Animals , Cell Line , Drug Resistance, Viral , Humans , Sendai virus/growth & development , Vesiculovirus/growth & development , Viral Plaque Assay
14.
J Theor Biol ; 265(4): 691-703, 2010 Aug 21.
Article in English | MEDLINE | ID: mdl-20553733

ABSTRACT

The interferon system provides a powerful and universal intracellular defense mechanism against viruses. As one part of their survival strategies, many viruses have evolved mechanisms to counteract the host type I interferon (IFN-alpha/beta) responses. In this study, we attempt to investigate virus- and double-strand RNA (dsRNA)-triggered type I IFN signaling pathways and understand the inhibition of IFN-alpha/beta induction by viral proteins using mathematical modeling and quantitative analysis. Based on available literature and our experimental data, we develop a mathematical model of virus- and dsRNA-triggered signaling pathways leading to type I IFN gene expression during the primary response, and use the genetic algorithm to optimize all rate constants in the model. The consistency between numerical simulation results and biological experimental data demonstrates that our model is reasonable. Further, we use the model to predict the following phenomena: (1) the dose-dependent inhibition by classical swine fever virus (CSFV) N(pro) or E(rns) protein is observed at a low dose and can reach a saturation above a certain dose, not an increase; (2) E(rns) and N(pro) have no synergic inhibitory effects on IFN-beta induction; (3) the different characters in an important transcription factor, phosphorylated IRF3 (IRF3p), are exhibited because N(pro) or E(rns) counteracted dsRNA- and virus-triggered IFN-beta induction by targeting the different molecules in the signaling pathways and (4) N(pro) inhibits the IFN-beta expression not only by interacting with IFR3 but also by affecting its complex with MITA. Our approaches help to gain insight into system properties and rational therapy design, as well as to generate hypotheses for further research.


Subject(s)
Interferon Type I/immunology , Models, Biological , Signal Transduction/immunology , Viral Proteins/antagonists & inhibitors , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line , Classical Swine Fever Virus/drug effects , Classical Swine Fever Virus/immunology , Computer Simulation , Interferon Regulatory Factors/metabolism , Interferon Type I/genetics , Interferon-beta/immunology , Plasmids/genetics , Poly I-C/pharmacology , RNA, Double-Stranded/metabolism , Sendai virus/drug effects , Sendai virus/immunology , Signal Transduction/drug effects , Swine , Time Factors , Viral Proteins/metabolism
15.
PLoS One ; 5(5): e10719, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20502666

ABSTRACT

One of the accessory proteins of Sendai virus (SeV), C, translated from an alternate reading frame of P/V mRNA has been shown to function at multiple stages of infection in cell cultures as well as in mice. C protein has been reported to counteract signal transduction by interferon (IFN), inhibit apoptosis induced by the infection, enhance the efficiency of budding of viral particles, and regulate the polarity of viral genome-length RNA synthesis to maximize production of infectious particles. In this study, we have generated a series of SeV recombinants containing substitutions of highly conserved, charged residues within the C protein, and characterized them together with previously-reported C'/C(-), 4C(-), and F170S recombinant viruses in infected cell cultures in terms of viral replication, cytopathogenicity, and antagonizing effects on host innate immunity. Unexpectedly, the amino acid substitutions had no or minimal effect on viral growth and viral RNA synthesis. However, all the substitutions of charged amino acids resulted in the loss of a counteracting effect against the establishment of an IFN-alpha-mediated anti-viral state. Infection by the virus (Cm2') containing mutations at K77 and D80 induced significant IFN-beta production, severe cytopathic effects, and detectable amounts of viral dsRNA production. In addition to the Cm2' virus, the virus containing mutations at E114 and E115 did not inhibit the poly(I:C)-triggered translocation of cellular IRF-3 to the nucleus. These results suggest that the C protein play important roles in viral escape from induction of IFN-beta and cell death triggered by infection by means of counteracting the pathway leading to activation of IRF-3 as well as of minimizing viral dsRNA production.


Subject(s)
Amino Acids/metabolism , Conserved Sequence , Immune Evasion/immunology , Immunity, Innate/immunology , Sendai virus/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytopathogenic Effect, Viral/drug effects , Genome, Viral/genetics , HeLa Cells , Humans , Immune Evasion/drug effects , Immunity, Innate/drug effects , Interferon Regulatory Factor-3/metabolism , Interferon-beta/biosynthesis , Kinetics , Mutation/genetics , Poly I-C/pharmacology , Protein Transport/drug effects , RNA, Double-Stranded/biosynthesis , Recombination, Genetic/drug effects , Recombination, Genetic/genetics , Respirovirus Infections/immunology , Respirovirus Infections/virology , Sendai virus/drug effects , Sendai virus/genetics , Signal Transduction/drug effects , Structure-Activity Relationship , Virion/genetics
16.
J Comb Chem ; 11(6): 1100-4, 2009.
Article in English | MEDLINE | ID: mdl-19754047

ABSTRACT

To identify novel potentially broad spectrum antiviral compounds against RNA viruses, we have developed the parallel synthesis of a structurally interesting class of 2-substituted-4,5-dihydroxypyrimidine-6-carboxamides. Variously 2-substituted-4,5-dihydroxypyrimidine-6-carboxylate methyl esters were initially prepared and were then diversified via a facile amidation reaction. This strategy affords libraries of thousands of diverse drug-like compounds for screening. Biological evaluation of a set of these compounds, via a small initial screen, identified antiviral compounds against a representative RNA virus (Sendai virus, a paramyxovirus). We provide details on the synthetic protocols and the in vitro antiviral activity studies, as part of our initial investigation of the resulting targeted libraries.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Combinatorial Chemistry Techniques/methods , Pyrimidines/chemistry , Pyrimidines/pharmacology , Sendai virus/drug effects , Small Molecule Libraries , Antiviral Agents/chemistry , Drug Evaluation, Preclinical , Microbial Sensitivity Tests , Molecular Structure , Stereoisomerism
17.
Antimicrob Agents Chemother ; 53(9): 3942-51, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19564364

ABSTRACT

Human parainfluenza virus type 3 (hPIV-3) is a major respiratory tract pathogen that affects young children, but no vaccines or antiviral drugs against it have yet been developed. We developed a mouse model to evaluate the efficacies of the novel parainfluenza virus hemagglutinin-neuraminidase (HN) inhibitors BCX 2798 and BCX 2855 against a recombinant Sendai virus (rSeV) in which the fusion (F) and HN surface glycoproteins (FHN) were replaced by those of hPIV-3 [rSeV(hPIV-3FHN)]. In the prophylaxis model, 129X1/SvJ mice were infected with a 90% or 20% lethal dose of the virus and were treated intranasally for 5 days with 10 mg/kg of body weight/day of either compound starting 4 h before infection. Prophylactic treatment of the mice with either compound did not prevent their death in a 90% lethality model of rSeV(hPIV-3FHN) infection. However, it significantly reduced the lung virus titers, the amount of weight lost, and the rate of mortality in mice infected with a 20% lethal virus dose. In the therapy model, mice were infected with a nonlethal dose of the virus (100 PFU/mouse) and were treated intranasally with 1 or 10 mg/kg/day of either compound for 5 days starting at 24 or 48 h postinfection. Treatment of the mice with either compound significantly reduced the virus titer in the lungs, subsequently causing a reduction in the number of immune cells and the levels of cytokines in the bronchoalveolar lavage fluid and histopathologic changes in the airways. Our results indicate that BCX 2798 and BCX 2855 are effective inhibitors of hPIV-3 HN in our mouse model and may be promising candidates for the prophylaxis and treatment of hPIV-3 infection in humans.


Subject(s)
Antiviral Agents/pharmacology , Azides/pharmacology , Hexuronic Acids/pharmacology , Parainfluenza Virus 3, Human/drug effects , Parainfluenza Virus 3, Human/pathogenicity , Sendai virus/drug effects , Sulfonamides/pharmacology , Animals , Cell Line , Cricetinae , Female , Haplorhini , Humans , Mice
18.
Mol Immunol ; 46(3): 393-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19038458

ABSTRACT

The innate cellular response to virus particle entry in non-immune cells requires the transcriptional activity of interferon regulatory factor 3 (IRF-3), but not production of type I interferon (IFN). Here, we characterize the IFN-independent innate cellular response to virus-derived stimuli in Vero cells, a monkey kidney epithelial cell line deficient for IFN production. We provide evidence that Vero cells are deficient in their ability to mount an IRF-3-dependent, IFN-independent antiviral response against either incoming virus particles or polyinosinic:polycytidylic acid (pIC), a dsRNA mimetic. We further demonstrate that abundance of IRF-3 protein is a determinant in the pIC-mediated antiviral signalling pathway. These observations further characterize the permissive nature of Vero cells to viral infection, and highlight the crucial involvement of IRF-3 in the innate antiviral response.


Subject(s)
Herpesvirus 1, Human/immunology , Interferon Regulatory Factor-3/immunology , Interferons/biosynthesis , Sendai virus/immunology , Animals , Chlorocebus aethiops , Cloning, Molecular , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/radiation effects , Humans , Poly I-C/pharmacology , Sendai virus/drug effects , Sendai virus/physiology , Sendai virus/radiation effects , Sequence Analysis, DNA , Ultraviolet Rays , Vero Cells , Virus Inactivation/drug effects , Virus Inactivation/radiation effects , Virus Replication/drug effects , Virus Replication/radiation effects
19.
Int J Pharm ; 361(1-2): 92-8, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18617337

ABSTRACT

Virus inactivation is a critical step in the manufacturing of recombinant therapeutic proteins, in particular antibodies, using mammalian expression systems. We have shown in the previous paper that arginine is effective in inactivation of herpes simplex virus type 1 (HSV-1) and influenza virus at low temperature under mildly acidic pH, i.e., above pH 4.0; above this pH, conformational changes of most antibodies are negligible. We have here extended virus inactivation study of arginine to other enveloped viruses, such as Sendai virus and Newcastle Disease Virus (NDV), and observed that arginine was ineffective against both viruses under the similar conditions, i.e., on ice and above pH 4.0. However, an arginine derivative, butyroyl-arginine, showed a strong virucidal potency against Sendai virus, leading to a 4log reduction in virus yield at pH 4.0, but not against NDV. In addition, although arginine and butyroyl-arginine were equally effective against influenza virus having a cleaved form of hemagglutinin spike proteins, only butyroyl-arginine was significantly effective against the same virus, but having an uncleaved hemagglutinin spike proteins. Furthermore, butyroyl-arginine was more effective than arginine against HSV-1 at pH 4.5; i.e., it has a broader pH spectrum than does arginine.


Subject(s)
Antiviral Agents/pharmacology , Arginine/analogs & derivatives , Arginine/pharmacology , Virus Inactivation/drug effects , Animals , Antiviral Agents/administration & dosage , Arginine/administration & dosage , Arginine/chemistry , Cell Line , Chlorocebus aethiops , Dogs , Hydrogen-Ion Concentration , Kidney , Newcastle disease virus/drug effects , Sendai virus/drug effects , Temperature , Vero Cells
20.
Med Microbiol Immunol ; 195(3): 151-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16465546

ABSTRACT

We prepared the chimeric recombinant Sendai virus [rSeV(Ppi)] by replacing the P gene of the Z strain with that of pi strain for analyzing the function of Ppi, Vpi and Cpi proteins. Intriguingly, HA production by rSeV(Ppi) is significantly lower at 38 degrees C than at 32 degrees C, showing that virus growth of rSeV(Ppi) is slightly suppressed at 38 degrees C. However, the main phenotypes of SeVpi, a marked temperature sensitivity as viral replication and an ability of establishing persistent infection, are not explained by the Ppi, Vpi and Cpi proteins. The V and C proteins form inclusion bodies in L929 cells infected with rSeV(Ppi) and incubated at 38 degrees C. L929 cells infected with rSeV(Ppi) and L929 cells stably expressing the Cpi protein show resistance to interferon-beta at 32 and 38 degrees C, indicating that the Cpi protein per se is not temperature-sensitive to inhibition of IFN signaling. The complete genome sequences of Sendai virus (SeV) pi and parent Nagoya strains were determined. Fifty nucleic acid substitutions are found in the genome sequence of SeV pi strain in comparison with Nagoya strain. There are three nucleic acid substitutions in the leader sequence, while the trailer, intergenic, gene-end and gene-start sequences of both strains are completely identical. Deletions and insertions of nucleotide are not found. There are 32 amino acid substitutions in Sendai virus pi strain. The specific amino acid substitutions unique to the SeVpi are 18. Information about the complete genome sequences of SeVpi strain is important to totally understand the persistent infection and lower pathogenicity of SeV.


Subject(s)
Hemagglutination/physiology , Sendai virus/metabolism , Viral Proteins/biosynthesis , Cell Line , HeLa Cells , Humans , Interferon-beta/pharmacology , Microbial Sensitivity Tests , Molecular Sequence Data , Recombination, Genetic , Sendai virus/drug effects , Sendai virus/genetics , Sendai virus/physiology , Temperature , Viral Fusion Proteins/metabolism , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...