Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 285
Filter
1.
J Biol Chem ; 300(4): 107168, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38490434

ABSTRACT

Lipids have been previously implicated in the lifecycle of neuroinvasive viruses. However, the role of lipids in programmed cell death and the relationship between programmed cell death and lipid droplets (LDs) in neuroinvasive virus infection remains unclear. Here, we found that the infection of neuroinvasive virus, such as rabies virus and encephalomyocarditis virus could enhance the LD formation in N2a cells, and decreasing LDs production by targeting diacylglycerol acyltransferase could suppress viral replication. The lipidomics analysis revealed that arachidonic acid (AA) was significantly increased after reducing LD formation by restricting diacylglycerol acyltransferase, and AA was further demonstrated to induce ferroptosis to inhibit neuroinvasive virus replication. Moreover, lipid peroxidation and viral replication inhibition could be significantly alleviated by a ferroptosis inhibitor, ferrostatin-1, indicating that AA affected neuroinvasive virus replication mainly through inducing ferroptosis. Furthermore, AA was demonstrated to activate the acyl-CoA synthetase long-chain family member 4-lysophosphatidylcholine acyltransferase 3-cytochrome P450 oxidoreductase axis to induce ferroptosis. Our findings highlight novel cross-talks among viral infection, LDs, and ferroptosis for the first time, providing a potential target for antiviral drug development.


Subject(s)
Arachidonic Acid , Ferroptosis , Lipid Droplets , Virus Replication , Ferroptosis/drug effects , Lipid Droplets/metabolism , Lipid Droplets/drug effects , Animals , Virus Replication/drug effects , Mice , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Encephalomyocarditis virus/drug effects , Diacylglycerol O-Acyltransferase/metabolism , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Lipid Peroxidation/drug effects , Coenzyme A Ligases/metabolism , Cell Line, Tumor , Humans
2.
BMC Vet Res ; 17(1): 318, 2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34587973

ABSTRACT

BACKGROUND: Encephalomyocarditis virus (EMCV) infection can cause reproductive failure in sows and acute myocarditis and sudden death in piglets. It has caused huge economic losses to the global pig industry and that is why it is necessary to develop effective new treatment compounds. Zedoary turmeric oil has been used for treating myocarditis. Curcumol extracted from the roots of curcuma is one of the main active ingredient of zedoary turmeric oil. The anti-EMCV activity of curcumol along with the molecular mechanisms involved with a focus on IFN-ß signaling pathway was investigated in this study. METHOD: 3-(4,5-dimethyithiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to determine the maximum non-toxic concentration (MNTC), 50% cytotoxic concentration (CC50), maximum inhibition rate (MIR) and 50% effective concentration (EC50) against EMCV. Through EMCV load, the anti-viral effect of curcumol was quantitatively determined using real-time quantitative PCR (qPCR). The effect of curcumol on the expression of IFN-ß was investigated using real-time quantitative PCR and ELISA. Western blot was used to determine the amounts of MDA5, MAVS, TANK, IRF3 and P-IRF3 proteins in human embryonic kidney 293 T (HEK-293 T) cells infected with EMCV. RESULTS: The results of MTT showed that compared with the ribavirin positive control group, the maximum inhibition ratio (MIR) of curcumol was greater but the selection index (SI) value was much smaller than that of ribavirin. The results of qPCR showed that curcumol and ribavirin significantly reduced the replication of EMCV in HEK-293 T cells. The curcumol (0.025 mg/mL) treatment has significantly increased IFN-ß mRNA expression in the EMCV-infected HEK-293 T cells while ribavirin treatment did not. The results of ELISA showed that curcumol (0.025 mg/mL and 0.0125 mg/mL) has significantly increased the expression of IFN-ß protein in EMCV-infected HEK-293 T cells. The results of Western blot showed that curcumol can inhibit the degradation of TANK protein mediated by EMCV and promote the expression of MDA5 and P-IRF3, while the protein expression level of MAVS and IRF3 remain unchanged. CONCLUSION: Curcumol has biological activity against EMCV which we suggest that IFN-ß signaling pathway is one of its mechanisms.


Subject(s)
Antiviral Agents/pharmacology , Encephalomyocarditis virus/drug effects , Sesquiterpenes/pharmacology , Cardiovirus Infections/drug therapy , Cardiovirus Infections/virology , HEK293 Cells , Humans , Interferon-beta/drug effects , Interferon-beta/metabolism , Ribavirin/pharmacology , Sesquiterpenes/toxicity , Signal Transduction/drug effects , Virus Replication/drug effects
3.
PLoS Pathog ; 14(10): e1007307, 2018 10.
Article in English | MEDLINE | ID: mdl-30308076

ABSTRACT

As antimicrobial signalling molecules, type III or lambda interferons (IFNλs) are critical for defence against infection by diverse pathogens, including bacteria, fungi and viruses. Counter-intuitively, expression of one member of the family, IFNλ4, is associated with decreased clearance of hepatitis C virus (HCV) in the human population; by contrast, a natural frameshift mutation that abrogates IFNλ4 production improves HCV clearance. To further understand how genetic variation between and within species affects IFNλ4 function, we screened a panel of all known extant coding variants of human IFNλ4 for their antiviral potential and identify three that substantially affect activity: P70S, L79F and K154E. The most notable variant was K154E, which was found in African Congo rainforest 'Pygmy' hunter-gatherers. K154E greatly enhanced in vitro activity in a range of antiviral (HCV, Zika virus, influenza virus and encephalomyocarditis virus) and gene expression assays. Remarkably, E154 is the ancestral residue in mammalian IFNλ4s and is extremely well conserved, yet K154 has been fixed throughout evolution of the hominid genus Homo, including Neanderthals. Compared to chimpanzee IFNλ4, the human orthologue had reduced activity due to amino acid K154. Comparison of published gene expression data from humans and chimpanzees showed that this difference in activity between K154 and E154 in IFNλ4 correlates with differences in antiviral gene expression in vivo during HCV infection. Mechanistically, our data show that the human-specific K154 negatively affects IFNλ4 activity through a novel means by reducing its secretion and potency. We thus demonstrate that attenuated activity of IFNλ4 is conserved among humans and postulate that differences in IFNλ4 activity between species contribute to distinct host-specific responses to-and outcomes of-infection, such as HCV infection. The driver of reduced IFNλ4 antiviral activity in humans remains unknown but likely arose between 6 million and 360,000 years ago in Africa.


Subject(s)
Antiviral Agents/therapeutic use , Cardiovirus Infections/drug therapy , Hepatitis C/drug therapy , Interleukins/genetics , Polymorphism, Single Nucleotide , Zika Virus Infection/drug therapy , Animals , Biological Evolution , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , Cells, Cultured , Encephalomyocarditis virus/drug effects , Encephalomyocarditis virus/isolation & purification , Gene Expression Regulation , Hepacivirus/drug effects , Hepacivirus/isolation & purification , Hepatitis C/genetics , Hepatitis C/virology , Humans , Pan troglodytes , Species Specificity , Zika Virus/drug effects , Zika Virus/isolation & purification , Zika Virus Infection/genetics , Zika Virus Infection/virology
4.
Biotechnol Appl Biochem ; 65(3): 446-454, 2018 May.
Article in English | MEDLINE | ID: mdl-29072333

ABSTRACT

α1-Antitrypsin (AAT) is widely used to treat patients with congenital AAT deficiency. Cohn Fraction IV (Cohn F IV) is normally discarded during the manufacturing process of albumin but contains approximately 33% of plasma AAT. We established a new process for large-scale purification of AAT from it. liquid chromatography-electrospray ionization-tandem mass spectrometry and high-performance liquid chromatography were applied for qualitative identification and composition analysis, respectively. Stabilizers were optimized for AAT activity protection during lyophilization and dry-heat. Virus inactivation by dry-heat and solvent/detergent (S/D) was validated on a range of viruses. AAT with purity of 95.54%, specific activity of 3,938.5 IU/mg, and yield of 26.79%, was achieved. More than 95% activity was reserved after S/D. More than 96% activity was obtained after lyophilization or dry-heat. After S/D, pseudorabies virus (PRV) and vesicular stomatitis virus (VSV) were inactivated below detectable level within 1 H. Virus titer reductions of more than 5.50 log10 and 5.38 log10 were achieved for PRV and VSV, respectively. Porcine parvovirus and encephalomyocarditis virus were inactivated by 3.17 log10 and 5.88 log10 reduction after dry-heat. The advantages of this process, including suitability for large-scale production, high purity, better utilization of human plasma, viral safety, commercial and inexpensive chromatography medium, may facilitate its further application.


Subject(s)
Blood Proteins/chemistry , Detergents/pharmacology , Hot Temperature , Solvents/pharmacology , Virus Inactivation/drug effects , alpha 1-Antitrypsin/isolation & purification , Animals , Cell Line , Detergents/chemistry , Encephalomyocarditis virus/drug effects , Herpesvirus 1, Suid/drug effects , Parvovirus/drug effects , Solvents/chemistry , Swine , Vesicular stomatitis Indiana virus/drug effects , alpha 1-Antitrypsin/chemistry
5.
Pol J Microbiol ; 66(1): 131-134, 2017 Mar 30.
Article in English | MEDLINE | ID: mdl-29359700

ABSTRACT

We compared the susceptibility to viral infection of splenocytes, isolated from young versus old CBA mice, and evaluated the antiviral actions of lactoferrin in splenocytes infected with Encephalomyocarditis virus (EMCV). Recombinant mouse lactoferrin (rmLF) and bovine lactoferrin (bLF) were used. There were no differences in the susceptibility to EMCV infection in the studied age categories. Both types of lactoferrins were protective in young and old mice. The study confirmed the undisturbed viral resistance in old mice and the protective actions of lactoferrin in viral infection. The antiviral action of the homologous mouse lactoferrin was demonstrated for the first time.


Subject(s)
Aging/immunology , Encephalomyocarditis virus/physiology , Lactoferrin/pharmacology , Spleen/cytology , Spleen/virology , Animals , Antiviral Agents , Cattle , Cells, Cultured , Encephalomyocarditis virus/drug effects , Mice , Mice, Inbred CBA , Recombinant Proteins
6.
Antimicrob Agents Chemother ; 60(10): 6402-6, 2016 10.
Article in English | MEDLINE | ID: mdl-27480860

ABSTRACT

Encephalomyocarditis virus (EMCV), like hepatitis C virus (HCV), requires phosphatidylinositol 4-kinase IIIα (PI4KA) for genome replication. Here, we demonstrate that tyrphostin AG1478, a known epidermal growth factor receptor (EGFR) inhibitor, also inhibits PI4KA activity, both in vitro and in cells. AG1478 impaired replication of EMCV and HCV but not that of an EMCV mutant previously shown to escape PI4KA inhibition. This work uncovers novel cellular and antiviral properties of AG1478, a compound previously regarded only as a cancer chemotherapy agent.


Subject(s)
1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , Antiviral Agents/pharmacology , Encephalomyocarditis virus/drug effects , Enzyme Inhibitors/pharmacology , Hepacivirus/drug effects , Quinazolines/pharmacology , Tyrphostins/pharmacology , 1-Phosphatidylinositol 4-Kinase/metabolism , Dose-Response Relationship, Drug , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/physiology , HeLa Cells/drug effects , HeLa Cells/virology , Hepacivirus/physiology , Humans , Molecular Targeted Therapy/methods , Mutation , Virus Replication/drug effects
7.
J Interferon Cytokine Res ; 36(3): 192-203, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26824268

ABSTRACT

Human serum albumin (HSA)-free formulation of Escherichia coli-derived human interferon beta (IFNß-1b) with a high percentage of monomeric protein and low immunogenicity is developed and characterized in the current study. UV spectroscopy, fluorescence spectroscopy, dynamic light scattering, sodium dodecyl sulfate polyacrylamide gel electrophoresis, Western blotting, Micro-Flow Imaging, resonant mass measurement, size exclusion, and reversed-phase high performance liquid chromatographies were applied to assess the effect of excipients on the stability of IFNß-1b to establish a HSA-free formulation. The antiviral activity of IFNß-1b was evaluated using human lung carcinoma cell line. Immune tolerant mice to hIFNß were used to assess the immunogenicity of the HSA-free formulated IFNß-1b in comparison to Betaferon drug product and Avonex drug substance as standards through IgG titering of plasma. HSA-free formulated IFNß-1b, including 200 mM L-arginine, 200 mM trehalose, and 0.1% n-dodecyl ß-D-maltoside in 10 mM sodium acetate buffer, pH 7.4, showed the highest biological activity. The stability of IFNß-1b in the HSA-free formulation was monitored for 3 weeks at 4°C and 37°C with relative humidity of 10% and 75%, respectively. Protein aggregation and immunogenicity in transgenic mice were decreased in the HSA-free formulated IFNß-1b compared to Betaferon. The stability, biological activity, and immunogenicity of the HSA-free formulation and Betaferon were evaluated. Incubation of formulations at 4°C and 37°C for 3 weeks showed that the HSA-free formulated IFNß-1b was more stable and less immunogenic in transgenic FVB/N mice. Low immunogenicity and the absence of HSA, which reduces the potential risk of viral infection (eg, HIV and HCV), are promising for clinical studies.


Subject(s)
Antibodies, Viral/blood , Antiviral Agents/pharmacology , Encephalomyocarditis virus/drug effects , Interferon beta-1b/pharmacology , A549 Cells , Animals , Antiviral Agents/immunology , Antiviral Agents/isolation & purification , Cloning, Molecular , Encephalomyocarditis virus/growth & development , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Immune Tolerance , Interferon beta-1b/biosynthesis , Interferon beta-1b/isolation & purification , Mice , Mice, Transgenic , Protein Stability , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
8.
J Interferon Cytokine Res ; 36(3): 180-91, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26700737

ABSTRACT

Type I interferons (IFNs) exhibit broad-spectrum antiviral activity, with potential utility against emerging acute virus infections that pose a threat to global health. Recombinant IFN-αs that have been approved for clinical use require cold storage and are administered through intramuscular or subcutaneous injection, features that are problematic for global distribution, storage, and administration. Cognizant that the biological potency of an IFN-α subtype is determined by its binding affinity to the type I IFN receptor, IFNAR, we identified a panel of small molecule nonpeptide compounds using an in silico screening strategy that incorporated specific structural features of amino acids in the receptor-binding domains of the most potent IFN-α, IFN alfacon-1. Hit compounds were selected based on ease of synthesis and formulation properties. In preliminary biological assays, we provide evidence that these compounds exhibit antiviral activity. This proof-of-concept study validates the strategy of in silico design and development for IFN mimetics.


Subject(s)
Antiviral Agents/pharmacology , Encephalomyocarditis virus/drug effects , Interferon-alpha/chemistry , Peptidomimetics/pharmacology , Receptor, Interferon alpha-beta/agonists , Small Molecule Libraries/pharmacology , Antiviral Agents/chemical synthesis , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , B-Lymphocytes/virology , Cell Line, Tumor , Computer Simulation , Drug Design , Encephalomyocarditis virus/growth & development , Gene Expression , High-Throughput Screening Assays , Humans , Ligands , Models, Molecular , Peptidomimetics/chemical synthesis , Protein Structure, Secondary , Receptor, Interferon alpha-beta/chemistry , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Recombinant Proteins/chemistry , Small Molecule Libraries/chemical synthesis , Structure-Activity Relationship , User-Computer Interface
9.
PLoS One ; 10(7): e0130797, 2015.
Article in English | MEDLINE | ID: mdl-26158644

ABSTRACT

Type I interferons are multi-potent cytokines that serve as first line of defense against viruses and other pathogens, posses immunomudolatory functions and elicit a growth inhibitory response. In recent years it has been shown that interferons are also detrimental, for example in lupus, AIDS, tuberculosis and cognitive decline, highlighted the need to develop interferon antagonists. We have previously developed the antagonist IFN-1ant, with much reduced binding to the IFNAR1 receptor and enhanced binding to IFNAR2. Here, we further tune the IFN-1ant by producing three additional antagonists based on IFN-1ant but with altered activity profiles. We show that in all three cases the antiproliferative activity of interferons is blocked and the induction of gene transcription of immunomudolatory and antiproliferative associated genes are substantially decreased. Conversely, each of the new antagonists elicits a different degree of antiviral response, STAT phosphorylation and related gene induction. Two of the new antagonists promote decreased activity in relation to the original IFN-1ant, while one of them promotes increased activity. As we do not know the exact causes of the detrimental effects of IFNs, the four antagonists that were produced and analyzed provide the opportunity to investigate the extent of antagonistic and agonistic activity optimal for a given condition.


Subject(s)
Interferon Type I/genetics , Mutant Proteins/genetics , Mutation , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Encephalomyocarditis virus/drug effects , Gene Expression/drug effects , Humans , Interferon Type I/antagonists & inhibitors , Interferon Type I/metabolism , Mice, Inbred C57BL , Mutant Proteins/metabolism , Mutant Proteins/pharmacology , Phosphorylation/drug effects , Protein Binding/drug effects , Receptor, Interferon alpha-beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Vesicular stomatitis Indiana virus/drug effects
10.
Int J Biol Macromol ; 79: 459-68, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26003302

ABSTRACT

In the report, three bioactive fractions from Cerrena unicolor: laccase (LAC), endopolysaccharides (c-EPL), and low molecular weight (ex-LMS) were tested for the first time towards their antiviral, immunostimulatory, cytotoxic and antiproliferative effect. The immunomodulatory activity was studied by means of THP-1-derived macrophages able to synthesize and secrete IL-6 and TNF-α. We used cervical carcinoma cell lines SiHa (ATCC, HTB-35) and CaSki (ATCC, CRL 1550) to determine antitumor activity and human skin fibroblasts (HSF) as a control. SiHa and L929 cell lines were used in the antiviral activity assay to propagate HHV-1 and EMCV, respectively. LAC was the most active against HSV at an early stage of viral replication, whereas the activity of laccase against EMCV was evident after incubation of the virus with LAC before and after the adsorption step. Moreover, the investigations showed that the fungal c-EPL fraction stimulated the production and secretion of TNF-α and IL-6 by THP-1-derived macrophages up to a level of 2000 pg/ml and 400 pg/ml, respectively. It was indicated for the first time that the LAC and ex-LMS fractions exhibited anticancer activity. This resulted from their cytotoxic or antiproliferative action against the investigated tumor cells at concentrations above 250 µg/ml and 10 µg/ml, respectively.


Subject(s)
Antineoplastic Agents/isolation & purification , Antiviral Agents/isolation & purification , Fungal Polysaccharides/isolation & purification , Fungal Proteins/isolation & purification , Immunologic Factors/isolation & purification , Laccase/isolation & purification , Polyporaceae/chemistry , Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Encephalomyocarditis virus/drug effects , Encephalomyocarditis virus/physiology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Fibroblasts/virology , Fungal Polysaccharides/pharmacology , Fungal Proteins/pharmacology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Humans , Immunologic Factors/pharmacology , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Laccase/pharmacology , Macrophages/drug effects , Macrophages/pathology , Macrophages/virology , Polyporaceae/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Virus Replication/drug effects
11.
J Pept Sci ; 21(7): 554-60, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25810135

ABSTRACT

Interferon-alpha (IFNα) is a cytokine that orchestrates innate and adaptive immune responses and potently inhibits proliferation of normal and tumor cells. These properties have warranted the use of IFNα in clinical practice for the treatment of several viral infections and malignancies. However, overexpression of IFNα leads to immunopathology observed in the context of chronic viral infections and autoimmune conditions. Thus, it is desirable to develop therapeutic approaches that aim at suppressing excessive IFNα production. To that end, artificial evolution of peptides from phage display libraries represents a strategy that seeks to disrupt the interaction between IFNα and its cell surface receptor and thus inhibit the ensuing biological effects. Mirror-image phage display that screens peptide libraries against the D-enantiomer is particularly attractive because it allows for identification of proteolysis-resistant D-peptide inhibitors. This approach, however, relies on the availability of chemically synthesized D-IFNα composed entirely of D-amino acids. Here, we describe the synthesis and biological properties of IFNα2b of 165 amino acid residues produced by native chemical ligation, which represents an important first step toward the discovery of D-peptide antagonists with potential therapeutic applications.


Subject(s)
Interferon-alpha/chemical synthesis , Peptide Library , Peptides/chemical synthesis , Solid-Phase Synthesis Techniques/methods , Amino Acid Sequence , Cell Line, Tumor , Cell Proliferation/drug effects , Disulfides/chemistry , Encephalomyocarditis virus/drug effects , Encephalomyocarditis virus/growth & development , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/virology , Humans , Interferon alpha-2 , Interferon-alpha/pharmacology , Models, Molecular , Molecular Sequence Data , Peptides/pharmacology , Primary Cell Culture , Protein Folding , Recombinant Proteins/chemical synthesis , Recombinant Proteins/pharmacology , Stereoisomerism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
12.
J Interferon Cytokine Res ; 35(5): 359-66, 2015 May.
Article in English | MEDLINE | ID: mdl-25517543

ABSTRACT

The oligoadenylate synthetase (OAS) proteins are traditionally considered intracellular antiviral proteins that mediate antiviral activity through the synthesis of 2'-5'-linked oligoadenylates and subsequent activation of the endoribonuclease RNase L. However, we have recently demonstrated that exogenous recombinant OAS1 is taken up by cells and reduces viral replication both in cell culture and in vivo, independent of RNase L. These results demonstrate a novel paracrine antiviral activity of OAS working in parallel with the classical RNase L pathway. In this study, we investigate the uptake kinetics of recombinant porcine OAS1 and show that it is rapidly and efficiently internalized in a manner that can be blocked by heparin. Heparin, furthermore, abolishes the antiviral activity of OAS1, demonstrating the requirement of the intracellular localization of OAS1 to inhibit the virus. In addition, we demonstrate that exogenous OAS1 affects an early step of the viral replication cycle.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Virus Replication , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/pharmacology , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Encephalomyocarditis virus/drug effects , Encephalomyocarditis virus/physiology , Extracellular Space , HeLa Cells , Heparin/metabolism , Humans , Protein Binding , Protein Transport , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Swine , Vero Cells , Virus Replication/drug effects
13.
Antibiot Khimioter ; 59(3-4): 16-21, 2014.
Article in Russian | MEDLINE | ID: mdl-25300117

ABSTRACT

Substances with gender action on immunity were detected in water soluble hydrolised matter from reptile carcases. The gender action was shown on isolated blood neutrophils, whole blood and in vivo by the antiviral activity on experimental animals, contaminated with three types of viruses: Herpes simplex type 1, the virus of encephalomyocarditis and the virus of hepatitis of mice. The possible mechanism of the inhibitory action on the male immunity was associated with the protein kinase cascade, including protein kinase C, activated by phorbolmyristate in the cells of the immune system.


Subject(s)
Complex Mixtures/pharmacology , Immunity, Innate/drug effects , NADPH Oxidases/antagonists & inhibitors , Neutrophils/drug effects , Protein Kinase C/antagonists & inhibitors , Viral Load/drug effects , Animals , Cardiovirus Infections/drug therapy , Cardiovirus Infections/virology , Complex Mixtures/isolation & purification , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Encephalomyocarditis virus/drug effects , Encephalomyocarditis virus/physiology , Female , Hepatitis, Viral, Animal/drug therapy , Hepatitis, Viral, Animal/virology , Herpes Simplex/drug therapy , Herpes Simplex/virology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Humans , Male , Mice , Murine hepatitis virus/drug effects , Murine hepatitis virus/physiology , NADPH Oxidases/metabolism , Neutrophils/cytology , Neutrophils/immunology , Protein Kinase C/metabolism , Reptiles/metabolism , Sex Factors
14.
AAPS PharmSciTech ; 15(6): 1619-29, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25142823

ABSTRACT

Understanding the mechanism of aggregation of a therapeutic protein would not only ease the manufacturing processing but could also lead to a more stable finished product. Aggregation of recombinant interferon (IFNß-1b) was studied by heating, oxidizing, or seeding of unformulated monomeric solution. The formation of aggregates was monitored by dynamic light scattering (DLS) and UV spectroscopy. The autocatalytic monomer loss model was used to fit the data on aggregation rates. The influence of pre-nucleation on aggregation step was demonstrated by inducing the liquid samples containing a monomer form of folded IFNß-1b by heat and also an oxidizing agent. Results tend to suggest that the nucleus includes a single protein molecule which has been probably deformed. Seeding tests showed that aggregation of IFNß-1b was probably initiated when 1.0% (w/w) of monomers converted to nucleus form. Chemiluminescence spectroscopy analysis of the sample indicated the generation of 3.0 µM of hydrogen peroxide (H2O2) during nucleation stage of IFNß-1b aggregation. Arginine with a concentration of 200 mM was sufficient to suppress aggregation of IFNß-1b by decreasing the rate of pre-nucleation step. We proposed the formation of pre-nucleus structures prior to nucleation as the mechanism of aggregation of IFNß-1b. Furthermore, we have showed the positive anti-aggregation effect of arginine on pre-nucleation step.


Subject(s)
Antiviral Agents/chemistry , Arginine/chemistry , Excipients/chemistry , Interferon-beta/chemistry , Antiviral Agents/pharmacology , Cell Line, Tumor , Cytopathogenic Effect, Viral/drug effects , Encephalomyocarditis virus/drug effects , Humans , Hydrogen Peroxide/chemistry , Interferon beta-1b , Interferon-beta/pharmacology , Kinetics , Light , Models, Chemical , Oxidation-Reduction , Protein Aggregates , Protein Folding , Recombinant Proteins/chemistry , Scattering, Radiation , Spectrophotometry, Ultraviolet , Technology, Pharmaceutical/methods
15.
J Virol ; 88(19): 11091-107, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25008939

ABSTRACT

UNLABELLED: Few drugs targeting picornaviruses are available, making the discovery of antivirals a high priority. Here, we identified and characterized three compounds from a library of kinase inhibitors that block replication of poliovirus, coxsackievirus B3, and encephalomyocarditis virus. Using an in vitro translation-replication system, we showed that these drugs inhibit different stages of the poliovirus life cycle. A4(1) inhibited both the formation and functioning of the replication complexes, while E5(1) and E7(2) were most effective during the formation but not the functioning step. Neither of the compounds significantly inhibited VPg uridylylation. Poliovirus resistant to E7(2) had a G5318A mutation in the 3A protein. This mutation was previously found to confer resistance to enviroxime-like compounds, which target a phosphatidylinositol 4-kinase IIIß (PI4KIIIß)-dependent step in viral replication. Analysis of host protein recruitment showed that E7(2) reduced the amount of GBF1 on the replication complexes; however, the level of PI4KIIIß remained intact. E7(2) as well as another enviroxime-like compound, GW5074, interfered with viral polyprotein processing affecting both 3C- and 2A-dependent cleavages, and the resistant G5318A mutation partially rescued this defect. Moreover, E7(2) induced abnormal recruitment to membranes of the viral proteins; thus, enviroxime-like compounds likely severely compromise the interaction of the viral polyprotein with membranes. A4(1) demonstrated partial protection from paralysis in a murine model of poliomyelitis. Multiple attempts to isolate resistant mutants in the presence of A4(1) or E5(1) were unsuccessful, showing that effective broad-spectrum antivirals could be developed on the basis of these compounds. IMPORTANCE: Diverse picornaviruses can trigger multiple human maladies, yet currently, only hepatitis A virus and poliovirus can be controlled with vaccination. The development of antipicornavirus therapeutics is also facing significant difficulties because these viruses readily generate resistance to compounds targeting either viral or cellular factors. Here, we describe three novel compounds that effectively block replication of distantly related picornaviruses with minimal toxicity to cells. The compounds prevent viral RNA replication after the synthesis of the uridylylated VPg primer. Importantly, two of the inhibitors are strongly refractory to the emergence of resistant mutants, making them promising candidates for further broad-spectrum therapeutic development. Evaluation of one of the compounds in an in vivo model of poliomyelitis demonstrated partial protection from the onset of paralysis.


Subject(s)
Antiviral Agents/pharmacology , Poliomyelitis/drug therapy , Poliovirus/drug effects , Small Molecule Libraries/pharmacology , Viral Proteins/antagonists & inhibitors , Virus Replication/drug effects , 1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , 1-Phosphatidylinositol 4-Kinase/genetics , 1-Phosphatidylinositol 4-Kinase/metabolism , Animals , Antiviral Agents/chemistry , Cell-Free System , Disease Models, Animal , Encephalomyocarditis virus/drug effects , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/metabolism , Enterovirus B, Human/drug effects , Enterovirus B, Human/genetics , Enterovirus B, Human/metabolism , Gene Expression Regulation, Viral , HeLa Cells , Humans , Mice , Mutation , Poliomyelitis/virology , Poliovirus/genetics , Poliovirus/growth & development , Polyproteins/antagonists & inhibitors , Polyproteins/genetics , Polyproteins/metabolism , Signal Transduction , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Viral Proteins/genetics , Viral Proteins/metabolism
16.
PLoS One ; 9(4): e94491, 2014.
Article in English | MEDLINE | ID: mdl-24722666

ABSTRACT

Most antiviral treatment options target the invading pathogen and unavoidably encounter loss of efficacy as the pathogen mutates to overcome replication restrictions. A good strategy for circumventing drug resistance, or for pathogens without treatment options, is to target host cell proteins that are utilized by viruses during infection. The small molecule WP1130 is a selective deubiquitinase inhibitor shown previously to successfully reduce replication of noroviruses and some other RNA viruses. In this study, we screened a library of 31 small molecule derivatives of WP1130 to identify compounds that retained the broad-spectrum antiviral activity of the parent compound in vitro but exhibited improved drug-like properties, particularly increased aqueous solubility. Seventeen compounds significantly reduced murine norovirus infection in murine macrophage RAW 264.7 cells, with four causing decreases in viral titers that were similar or slightly better than WP1130 (1.9 to 2.6 log scale). Antiviral activity was observed following pre-treatment and up to 1 hour postinfection in RAW 264.7 cells as well as in primary bone marrow-derived macrophages. Treatment of the human norovirus replicon system cell line with the same four compounds also decreased levels of Norwalk virus RNA. No significant cytotoxicity was observed at the working concentration of 5 µM for all compounds tested. In addition, the WP1130 derivatives maintained their broad-spectrum antiviral activity against other RNA viruses, Sindbis virus, LaCrosse virus, encephalomyocarditis virus, and Tulane virus. Thus, altering structural characteristics of WP1130 can maintain effective broad-spectrum antiviral activity while increasing aqueous solubility.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Nitriles/pharmacology , Pyridines/pharmacology , Small Molecule Libraries/pharmacology , Ubiquitin-Specific Proteases/antagonists & inhibitors , Virus Replication/drug effects , Animals , Antiviral Agents/chemistry , Cell Line , Cyanoacrylates , Encephalomyocarditis virus/drug effects , Encephalomyocarditis virus/physiology , Enzyme Inhibitors/chemistry , Host-Pathogen Interactions , Humans , La Crosse virus/drug effects , La Crosse virus/physiology , Macrophages/drug effects , Macrophages/virology , Mice , Nitriles/chemistry , Norovirus/drug effects , Norovirus/physiology , Norwalk virus/drug effects , Norwalk virus/physiology , Primary Cell Culture , Pyridines/chemistry , Sindbis Virus/drug effects , Sindbis Virus/physiology , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Ubiquitin-Specific Proteases/metabolism
17.
Elife ; 3: e01535, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24550253

ABSTRACT

The RIG-I-like receptors RIG-I, LGP2, and MDA5 initiate an antiviral response that includes production of type I interferons (IFNs). The nature of the RNAs that trigger MDA5 activation in infected cells remains unclear. Here, we purify and characterise LGP2/RNA complexes from cells infected with encephalomyocarditis virus (EMCV), a picornavirus detected by MDA5 and LGP2 but not RIG-I. We show that those complexes contain RNA that is highly enriched for MDA5-stimulatory activity and for a specific sequence corresponding to the L region of the EMCV antisense RNA. Synthesis of this sequence by in vitro transcription is sufficient to generate an MDA5 stimulatory RNA. Conversely, genomic deletion of the L region in EMCV generates viruses that are less potent at stimulating MDA5-dependent IFN production. Thus, the L region antisense RNA of EMCV is a key determinant of innate immunity to the virus and represents an RNA that activates MDA5 in virally-infected cells. DOI: http://dx.doi.org/10.7554/eLife.01535.001.


Subject(s)
DEAD-box RNA Helicases/metabolism , Encephalomyocarditis virus/metabolism , RNA Helicases/metabolism , RNA, Antisense/metabolism , RNA, Viral/metabolism , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , DEAD-box RNA Helicases/genetics , Encephalomyocarditis virus/drug effects , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/immunology , Gene Expression Regulation, Viral , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Influenza A virus/genetics , Influenza A virus/metabolism , Interferon-Induced Helicase, IFIH1 , Interferons/genetics , Interferons/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , RNA Helicases/genetics , RNA, Antisense/genetics , RNA, Viral/genetics , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Transfection , Vero Cells , Virus Replication
18.
Assay Drug Dev Technol ; 11(6): 355-66, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23906347

ABSTRACT

The use of small molecules to modulate cellular processes is a powerful approach to investigate gene function as a complement to genetic approaches. The discovery and characterization of compounds that modulate translation initiation, the rate-limiting step of protein synthesis, is important both to provide tool compounds to explore this fundamental biological process and to further evaluate protein synthesis as a therapeutic target. While most messenger ribonucleic acids (mRNAs) recruit ribosomes via their 5' cap, some viral and cellular mRNAs initiate protein synthesis via an alternative "cap-independent" mechanism utilizing internal ribosome entry sites (IRES) elements, which are complex mRNA secondary structures, localized within the 5' nontranslated region of the mRNA upstream of the AUG start codon. This report describes the design of a functional, high throughput screen of small molecules miniaturized into a 1,536-well format and performed using the luciferase reporter gene under control of the viral Cardiovirus encephalomyocarditis virus (EMCV) IRES element to identify nontoxic compounds modulating translation initiated from the EMCV IRES. One activating compound, validated in a dose response manner, has previously been shown to bind the glucocorticoid receptor (GR). Subsequent testing of additional GR modulators further supported this as the possible mechanism of action. Detailed characterization of this compound activity supported the notion that this was due to an effect at the level of translation.


Subject(s)
Encephalomyocarditis virus/drug effects , Protein Biosynthesis/drug effects , Receptors, Glucocorticoid/drug effects , Ribosomes/virology , Virus Internalization/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Design , Encephalomyocarditis virus/physiology , High-Throughput Screening Assays , Humans , Ligands , Receptors, Glucocorticoid/physiology
19.
PLoS One ; 8(5): e63940, 2013.
Article in English | MEDLINE | ID: mdl-23696859

ABSTRACT

The type III interferons (IFNs), comprising IFN-λ1, IFN-λ2, and IFN-λ3, behave similarly to IFN-α in eliciting antiviral, antitumor, and immune-modulating activities. Due to their more restricted cellular targets, IFN-λs are attractive as potential alternatives to existing therapeutic regimens based on IFN-αs. We have applied the DOCK-AND-LOCK™ method to improve the anti-proliferative potency of IFN-λ1 up to 1,000-fold in targeted cancer cell lines by tethering stabilized Fab dimers, derived from hRS7 (humanized anti-Trop-2), hMN-15 (humanized anti-CEACAM6), hL243 (humanized anti-HLA-DR), and c225 (chimeric anti-EGFR), to IFN-λ1 site-specifically, resulting in novel immunocytokines designated (E1)-λ1, (15)-λ1, (C2)-λ1, and (c225)-λ1, respectively. Targeted delivery of IFN-λ1 via (15)-λ1 or (c225)-λ1 to respective antigen-expressing cells also significantly increased antiviral activity when compared with non-targeting (C2)-λ1, as demonstrated in human lung adenocarcinoma cell line A549 by (15)-λ1 against encephalomyocarditis virus (EC50 = 22.2 pM versus 223 pM), and in human hepatocarcinoma cell line Huh-7 by (c225)-λ1 against hepatitis C virus (EC50 = 0.56 pM versus 91.2 pM). These promising results, which are attributed to better localization and stronger binding of IFN-λ1 to antibody-targeted cells, together with the favorable pharmacokinetic profile of (E1)-λ1 in mice (T(1/2) = 8.6 h), support further investigation of selective prototypes as potential antiviral and antitumor therapeutic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Immunoglobulin Fab Fragments/chemistry , Interferons/pharmacology , Animals , Antineoplastic Agents/chemistry , Antiviral Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Electrophoresis, Polyacrylamide Gel , Encephalomyocarditis virus/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Hepacivirus/drug effects , Humans , Interferons/chemistry , Mice , Mice, Nude
20.
J Biol Regul Homeost Agents ; 27(4): 1001-9, 2013.
Article in English | MEDLINE | ID: mdl-24382181

ABSTRACT

Type III interferons (IFN-lambda) are the most recently discovered members of IFN family. Synergism between different IFN types is well established, but for type I and type III IFNs no conclusive evidence has been reported so far. Possible synergism/antagonism between IFN-alpha and IFN-lambda in the inhibition of virus replication (EMCV, WNV lineage 1 and 2, CHIKV and HSV-1), and in the activation of intracellular pathways of IFN response (MxA and 2'-5' OAS) was evaluated in different cell lines (Vero E6, A549 and Wish cells). The antiviral potency of IFN-lambda1 and -l2 was lower than that of IFN-alpha. When IFN-alpha and -lambda were used together, the Combination Index (CI) for virus inhibition was greater than 1 virtually for all virus/host cell systems, indicating antagonistic effect. Antagonism between IFN-alpha and -l was also observed for the induction of mRNA for both MxA and 2'-5'OAS. Elucidating the interplay between IFN-alpha and -lambda may help to better understand innate defence mechanisms against viral infections, including the molecular mechanisms underlying the influence of IL-28B polymorphisms in the response to HCV and other viral infections.


Subject(s)
2',5'-Oligoadenylate Synthetase/genetics , Antiviral Agents/antagonists & inhibitors , Interferon-alpha/antagonists & inhibitors , Interferons/pharmacology , Interleukins/pharmacology , Animals , Chlorocebus aethiops , Encephalomyocarditis virus/drug effects , Humans , Myxovirus Resistance Proteins/genetics , Vero Cells , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...