Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Int J Pharm ; 660: 124318, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38852750

ABSTRACT

Avian influenza virus subtype H9N2 has the ability to infect birds and humans, further causing significant losses to the poultry industry and even posing a great threat to human health. Oral vaccine received particular interest for preventing majority infection due to its ability to elicit both mucosal and systemic immune responses, but their development is limited by the bad gastrointestinal (GI) environment, compact epithelium and mucus barrier, and the lack of effective mucosal adjuvants. Herein, we developed the dendritic fibrous nano-silica (DFNS) grafted with Cistanche deserticola polysaccharide (CDP) nanoparticles (CDP-DFNS) as an adjuvant for H9N2 vaccine. Encouragingly, CDP-DFNS facilitated the proliferation of T and B cells, and further induced the activation of T lymphocytes in vitro. Moreover, CDP-DFNS/H9N2 significantly promoted the antigen-specific antibodies levels in serum and intestinal mucosal of chickens, indicating the good ability to elicit both systemic and mucosal immunity. Additional, CDP-DFNS facilitate the activation of CD4 + and CD8 + T cells both in spleen and intestinal mucosal, and the indexes of immune organs. This study suggested that CDP-DFNS may be a new avenue for development of oral vaccine against pathogens that are transmitted via mucosal route.


Subject(s)
Adjuvants, Immunologic , Chickens , Immunity, Mucosal , Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Nanoparticles , Polysaccharides , Silicon Dioxide , Animals , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/drug effects , Polysaccharides/administration & dosage , Polysaccharides/pharmacology , Polysaccharides/chemistry , Polysaccharides/immunology , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Nanoparticles/administration & dosage , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Immunity, Mucosal/drug effects , Influenza in Birds/prevention & control , Influenza in Birds/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Administration, Oral , Intestinal Mucosa/immunology , Intestinal Mucosa/drug effects , Antibodies, Viral/blood , Antibodies, Viral/immunology
2.
Molecules ; 29(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731436

ABSTRACT

In our research, we explored a natural substance called Oxymatrine, found in a traditional Chinese medicinal plant, to fight against a common bird flu virus known as H9N2. This virus not only affects birds but can also pose a threat to human health. We focused on how this natural compound can help in stopping the virus from spreading in cells that line the lungs of birds and potentially humans. Our findings show that Oxymatrine can both directly block the virus and boost the body's immune response against it. This dual-action mechanism is particularly interesting because it indicates that Oxymatrine might be a useful tool in developing new ways to prevent and treat this type of bird flu. Understanding how Oxymatrine works against the H9N2 virus could lead to safer and more natural ways to combat viral infections in animals and humans, contributing to the health and well-being of society. The H9N2 Avian Influenza Virus (AIV) is a persistent health threat because of its rapid mutation rate and the limited efficacy of vaccines, underscoring the urgent need for innovative therapies. This study investigated the H9N2 AIV antiviral properties of Oxymatrine (OMT), a compound derived from traditional Chinese medicine, particularly focusing on its interaction with pulmonary microvascular endothelial cells (PMVECs). Employing an array of in vitro assays, including 50% tissue culture infectious dose, Cell Counting Kit-8, reverse transcription-quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, and Western blot, we systematically elucidated the multifaceted effects of OMT. OMT dose-dependently inhibited critical antiviral proteins (PKR and Mx1) and modulated the expression of type I interferons and key cytokines (IFN-α, IFN-ß, IL-6, and TNF-α), thereby affecting TLR3 signaling and its downstream elements (NF-κB and IRF-3). OMT's antiviral efficacy extended beyond TLR3-mediated responses, suggesting its potential as a versatile antiviral agent. This study not only contributes to the growing body of research on the use of natural compounds as antiviral agents but also underscores the importance of further investigating the broader application of OMT for combating viral infections.


Subject(s)
Antiviral Agents , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Matrines , Signal Transduction , Toll-Like Receptor 3 , Animals , Dogs , Humans , Antiviral Agents/pharmacology , Influenza A Virus, H9N2 Subtype/drug effects , Influenza in Birds/drug therapy , Influenza in Birds/immunology , Madin Darby Canine Kidney Cells , Signal Transduction/drug effects , Toll-Like Receptor 3/metabolism
3.
Jpn J Infect Dis ; 75(4): 398-402, 2022 Jul 22.
Article in English | MEDLINE | ID: mdl-34980710

ABSTRACT

The circulation of avian influenza A viruses in poultry is a public health concern due to the potential transmissibility and severity of these viral infections. Monitoring the susceptibility of these viruses to antivirals is important for developing measures to strengthen the level of preparedness against influenza pandemics. However, drug susceptibility information on these viruses is limited. Here, we determined the susceptibilities of avian influenza A(H5N1), A(H5N2), A(H5N8), A(H7N7), A(H7N9), A(H9N1), and A(H9N2) viruses isolated in Japan to the antivirals approved for use there: an M2 inhibitor (amantadine), neuraminidase inhibitors (oseltamivir, peramivir, zanamivir, and laninamivir) and RNA polymerase inhibitors (baloxavir and favipiravir). Genotypic methods that detect amino acid substitutions associated with antiviral resistance and phenotypic methods that assess phenotypic viral susceptibility to drugs have revealed that these avian influenza A viruses are susceptible to neuraminidase and RNA polymerase inhibitors. These results suggest that neuraminidase and RNA polymerase inhibitors currently approved in Japan could be a treatment option against influenza A virus infections in humans.


Subject(s)
Drug Resistance, Viral , Influenza in Birds , Influenza, Human , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , DNA-Directed RNA Polymerases , Drug Resistance, Viral/genetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N2 Subtype/drug effects , Influenza A Virus, H7N7 Subtype/drug effects , Influenza A Virus, H7N9 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/drug effects , Influenza in Birds/epidemiology , Influenza in Birds/virology , Influenza, Human/epidemiology , Influenza, Human/virology , Japan/epidemiology , Neuraminidase/genetics , Neuraminidase/metabolism , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Poultry
4.
BMC Complement Med Ther ; 21(1): 289, 2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34836523

ABSTRACT

BACKGROUND: H9N2 avian influenza viruses (AIVs) infect avian and mammalian hosts and provide internal genes for new emerging highly pathogenic avian viruses that cause severe pneumonia with high mortality, for which few medications are available. Arctiin, a bioactive lignan glycoside, has been reported to possess multiple pharmacological properties. However, the effect of arctiin on H9N2 virus infection is unclear. In the current study, we analyzed the effect of arctiin on H9N2 virus infection and the underlying molecular mechanism in vitro. METHODS: The antiviral effect against H9N2 virus was determined by plaque reduction assay (PRA) and progeny virus reduction assay. We employed MTT assay, qRT-PCR, ELISA, immunofluorescence and Western blotting to better understand the anti-inflammatory effect and corresponding mechanism of arctiin on H9N2 virus-infected cells. RESULTS: The results showed that arctiin had antiviral activity against H9N2 virus. Arctiin treatment reduced H9N2 virus-triggered proinflammatory cytokines, such as IL-6, and TNF-α. Moreover, arctiin significantly suppressed H9N2 virus-mediated expression of COX-2 and PGE2. Furthermore, we found that arctiin inhibited H9N2 virus-mediated activation of RIG-I/JNK MAPK signaling. Interestingly, arctiin treatment obviously reversed H9N2 virus-induced reduction of Nrf2, increased the nuclear translocation of Nrf2, and upregulated Nrf2 signaling target genes (HO-1 and SOD2). Zinc protoporphyrin (Znpp)-an HO-1 inhibitor-weakened the inhibitory effect of arctiin on H9N2 virus-induced RIG-I/JNK MAPK and proinflammatory mediators. CONCLUSION: Taken together, these results suggested that the anti-inflammatory effects of arctiin on H9N2 virus infection may be due to the activation of Nrf2/HO-1 and blocked RIG-I/JNK MAPK signaling; thus, arctiin may be a promising agent for prevention and treatment of H9N2 virus infections.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Furans/pharmacology , Glucosides/pharmacology , Heme Oxygenase-1/metabolism , Influenza A Virus, H9N2 Subtype/drug effects , NF-E2-Related Factor 2/metabolism , Plant Extracts/pharmacology , A549 Cells , Animals , Anti-Inflammatory Agents/chemistry , China , Dogs , Furans/chemistry , Glucosides/chemistry , Humans , Madin Darby Canine Kidney Cells , Molecular Structure
5.
Mol Immunol ; 140: 106-119, 2021 12.
Article in English | MEDLINE | ID: mdl-34678620

ABSTRACT

The recent advances in our understanding of the host factors in orchestrating qualitatively different immune responses against influenza Type A virus (IAV) have changed the perception of conventional approaches for controlling avian influenza virus (AIV) infection in chickens. Given that infection-induced pathogenicity and replication of influenza virus largely rely on regulating host immune responses, immunoregulatory cytokine profiles often determine the disease outcomes. However, in contrast to the function of other inflammatory cytokines, interleukin-17A (IL-17A) has been described as a 'double-edged sword', indicating that in addition to antiviral host responses, IL-17A has a distinct role in promoting viral infection. Therefore, in the present study, we investigated the chicken IL-17A mediated antiviral immune effects on IAVs infection in primary chicken embryo fibroblasts cells (CEFs). To this end, we first bioengineered a food-grade Lactic Acid Producing Bacteria (LAB), Lactococcus lactis (L. lactis), secreting bioactive recombinant chicken IL-17A (sChIL-17A). Next, the functionality of sChIL-17A was confirmed by transcriptional upregulation of several genes associated with antiviral host responses, including granulocyte-monocyte colony-stimulating factor (GM-CSF) (CSF3 in the chickens), interleukin-6 (IL-6), interferon-α (IFN-α), -ß and -γ genes in primary CEFs cells. Consistent with our hypothesis that such a pro-inflammatory state may translate to immunoprotection against IAVs infection, we observed that sChIL-17A pre-treatment could significantly limit the viral replication and protect the primary CEFs cells against two heterotypic IAVs such as A/turkey/Wisconsin/1/1966(H9N2) and A/PR/8/1934(H1N1). Together, the data presented in this work suggest that exogenous application of sChIL-17A secreted by modified LAB vector may represent an alternative strategy for improving antiviral immunity against avian influenza virus infection in chickens.


Subject(s)
Bioengineering , Cytoprotection , Fibroblasts/virology , Genetic Vectors/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H9N2 Subtype/physiology , Interleukin-17/pharmacology , Lactobacillales/genetics , Animals , Cell Death/drug effects , Cells, Cultured , Chick Embryo , Chickens/virology , Cytopathogenic Effect, Viral/drug effects , Cytoprotection/drug effects , Dogs , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Profiling , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H9N2 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/genetics , Influenza in Birds/immunology , Influenza in Birds/virology , Interleukin-17/genetics , Madin Darby Canine Kidney Cells , Nisin/pharmacology , Phenotype , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
6.
Trop Biomed ; 38(3): 353-359, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34508343

ABSTRACT

Curcumin is a potent antimicrobial herb used traditionally as a spice in culinary. This study was designed to evaluate the antiviral effects of curcuma longa extract against H9 influenza virus. A total of 60 embryonated eggs were equally divided into 5 groups with 12 eggs in each group. Group 1 (G1) served as uninfected negative control. Whereas Group 2 (G2) was kept as positive control infected with known virus @ 0.2 ml with 10-9.3 EID50. Group 3 (G3) was offered Curcuma longa @ 0.2 mg/0.2 ml and H9N2 virus (@ 0.2 ml with 10-9.3 EID50. Similarly, Group 4 (G4) was inoculated with extract of Curcuma longa @ 0.2 mg/0.2 ml per egg, whereas Group 5 (G5) was given Ribazole @ 0.2 ml/ egg. The crude extract and virus were administered on the 15th day of incubation and were checked after every 24 hours up to 96th hour post inoculation by random selection of 3 eggs. Death and survival rate were noted in all groups. Gross and histopathological lesions were also observed. Results revealed that Curcuma longa extract had significantly (p<0.05) reduced the mortality rate of embryos infected with H9N2 virus. In G3, increased lymphocytes and mild fatty changes were seen in liver. Whereas, mature RBCs, plasma cells and some lymphoblast's were observed in Spleen. Similarly, the bursa follicles were with lymphocytic aggregation. The G4 (Curcuma longa) and G5 (Ribazole) were with maximum embryo survival after 48 and 72 h post inoculation. This study revealed potential antiviral activity of Curcuma longa against H9N2 influenza viruses and can be opted as alternative to antibiotics and antiviral drugs to minimize the antimicrobial resistance in human and animal population.


Subject(s)
Antiviral Agents , Curcuma , Influenza A Virus, H9N2 Subtype , Plant Extracts/pharmacology , Animals , Antiviral Agents/pharmacology , Chick Embryo , Curcuma/chemistry , Influenza A Virus, H9N2 Subtype/drug effects
7.
Virol J ; 18(1): 171, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34412671

ABSTRACT

BACKGROUND: Diallyl trisulfide (DATS) is a garlic-derived organosulfur compound. As it has been shown to have anti-viral activity, we hypothesized that it may alleviate infections caused by H9N2 avian influenza virus (AIV), which is prevalent in poultry with pandemic potential. METHODS: Human lung A549 epithelial cells were treated with three different concentrations of DATS 24 h before (pre-treatment) or one hour after (post-treatment) H9N2 AIV infection. Culture supernatants were collected 24 h and 48 h post-infection and analyzed for viral titers and levels of inflammatory and anti-viral immune responses. For in vivo experiments, BABL/c mice were administered daily by intraperitoneal injection with DATS (30 mg/kg) for 2 weeks starting 1 day after H9N2 AIV infection. Clinical signs, lung pathology, and inflammatory and anti-viral immune responses were assessed 2, 4, and 6 days after infection. RESULTS: Both pre-treatment and post-treatment of A549 cells with DATS resulted in reduced viral loads, increased expression of anti-viral genes (RIG-I, IRF-3, and interferon-ß), and decreased expression of inflammatory cytokines (TNF-α and IL-6). These effects were also observed in H9N2 AIV-infected mice treated with DATS. Such treatment also reduced lung edema and inflammation in mice. CONCLUSIONS: Results suggest that DATS has anti-viral activity against H9N2 AIV and may be used as an alternative treatment for influenza virus infection.


Subject(s)
Allyl Compounds/pharmacology , Antiviral Agents/pharmacology , Influenza A Virus, H9N2 Subtype , Orthomyxoviridae Infections/drug therapy , Sulfides/pharmacology , A549 Cells , Animals , Chickens , Humans , Influenza A Virus, H9N2 Subtype/drug effects , Influenza in Birds/drug therapy , Mice
8.
Sci Rep ; 11(1): 9427, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33941825

ABSTRACT

Influenza viruses cause significant morbidity and mortality worldwide. Long-term or frequent use of approved anti-influenza agents has resulted in drug-resistant strains, thereby necessitating the discovery of new drugs. In this study, we found aprotinin, a serine protease inhibitor, as an anti-influenza candidate through screening of compound libraries. Aprotinin has been previously reported to show inhibitory effects on a few influenza A virus (IAV) subtypes (e.g., seasonal H1N1 and H3N2). However, because there were no reports of its inhibitory effects on the other types of influenza viruses, we investigated the inhibitory effects of aprotinin in vitro on a wide range of influenza viruses, including avian and oseltamivir-resistant influenza virus strains. Our cell-based assay showed that aprotinin had inhibitory effects on seasonal human IAVs (H1N1 and H3N2 subtypes), avian IAVs (H5N2, H6N5, and H9N2 subtypes), an oseltamivir-resistant IAV, and a currently circulating influenza B virus. We have also confirmed its activity in mice infected with a lethal dose of influenza virus, showing a significant increase in survival rate. Our findings suggest that aprotinin has the capacity to inhibit a wide range of influenza virus subtypes and should be considered for development as a therapeutic agent against influenza.


Subject(s)
Antiviral Agents/pharmacology , Aprotinin/pharmacology , Drug Evaluation, Preclinical , Orthomyxoviridae Infections/drug therapy , Serine Proteinase Inhibitors/pharmacology , Animals , Cell Line , Dogs , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H5N2 Subtype/drug effects , Influenza A Virus, H5N2 Subtype/growth & development , Influenza A Virus, H9N2 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/growth & development , Influenza B virus/drug effects , Influenza B virus/growth & development , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL
9.
Sci Rep ; 10(1): 16631, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33024252

ABSTRACT

The aim of this study was to test in vitro the ability of a mixture of citrus extract, maltodextrin, sodium chloride, lactic acid and citric acid (AuraShield L) to inhibit the virulence of infectious bronchitis, Newcastle disease, avian influenza, porcine reproductive and respiratory syndrome (PRRS) and bovine coronavirus viruses. Secondly, in vivo, we have investigated its efficacy against infectious bronchitis using a broiler infection model. In vitro, these antimicrobials had expressed antiviral activity against all five viruses through all phases of the infection process of the host cells. In vivo, the antimicrobial mixture reduced the virus load in the tracheal and lung tissue and significantly reduced the clinical signs of infection and the mortality rate in the experimental group E2 receiving AuraShield L. All these effects were accompanied by a significant reduction in the levels of pro-inflammatory cytokines and an increase in IgA levels and short chain fatty acids (SCFAs) in both trachea and lungs. Our study demonstrated that mixtures of natural antimicrobials, such AuraShield L, can prevent in vitro viral infection of cell cultures. Secondly, in vivo, the efficiency of vaccination was improved by preventing secondary viral infections through a mechanism involving significant increases in SCFA production and increased IgA levels. As a consequence the clinical signs of secondary infections were significantly reduced resulting in recovered production performance and lower mortality rates in the experimental group E2.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/drug therapy , Coronavirus, Bovine/drug effects , Epithelial Cells/drug effects , Infectious bronchitis virus/drug effects , Influenza A Virus, H9N2 Subtype/drug effects , Newcastle disease virus/drug effects , Porcine respiratory and reproductive syndrome virus/drug effects , Poultry Diseases/drug therapy , Animals , Cell Line , Chick Embryo , Chickens , Coronavirus Infections/virology , Disease Models, Animal , Epithelial Cells/virology , Humans , Influenza in Birds/metabolism , Influenza in Birds/virology , Influenza, Human/metabolism , Influenza, Human/virology , Newcastle Disease/metabolism , Newcastle Disease/virology , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine Reproductive and Respiratory Syndrome/virology , Poultry Diseases/virology , Swine
10.
Viruses ; 12(10)2020 10 08.
Article in English | MEDLINE | ID: mdl-33050000

ABSTRACT

Janus kinase (JAK) inhibitors have been developed as novel immunomodulatory drugs and primarily used for treating rheumatoid arthritis and other inflammatory diseases. Recent studies have suggested that this category of anti-inflammatory drugs could be potentially useful for the control of inflammation "storms" in respiratory virus infections. In addition to their role in regulating immune cell functions, JAK1 and JAK2 have been recently identified as crucial cellular factors involved in influenza A virus (IAV) replication and could be potentially targeted for antiviral therapy. Gingerenone A (Gin A) is a compound derived from ginger roots and a dual inhibitor of JAK2 and p70 S6 kinase (S6K1). Our present study aimed to determine the antiviral activity of Gin A on influenza A virus (IAV) and to understand its mechanisms of action. Here, we reported that Gin A suppressed the replication of three IAV subtypes (H1N1, H5N1, H9N2) in four cell lines. IAV replication was also inhibited by Ruxolitinib (Rux), a JAK inhibitor, but not by PF-4708671, an S6K1 inhibitor. JAK2 overexpression enhanced H5N1 virus replication and attenuated Gin A-mediated antiviral activity. In vivo experiments revealed that Gin A treatment suppressed IAV replication in the lungs of H5N1 virus-infected mice, alleviated their body weight loss, and prolonged their survival. Our study suggests that Gin A restricts IAV replication by inhibiting JAK2 activity; Gin A could be potentially useful for the control of influenza virus infections.


Subject(s)
Antiviral Agents/pharmacology , Diarylheptanoids/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/drug effects , Janus Kinase 2/antagonists & inhibitors , A549 Cells , Animals , Cell Line , Dogs , Female , HEK293 Cells , Humans , Imidazoles/pharmacology , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H9N2 Subtype/growth & development , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Nitriles , Piperazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Virus Replication/drug effects
11.
Virology ; 550: 51-60, 2020 11.
Article in English | MEDLINE | ID: mdl-32882637

ABSTRACT

Influenza virus neuraminidase (NA) contains a universally conserved epitope (NAe, NA222-230). However, no studies have reported vaccines targeting this NA conserved epitope and inducing antibodies recognizing NAe. The extracellular domain of M2 (M2e) is considered as an attractive target for a universal influenza vaccine. We generated recombinant influenza H1N1 viruses expressing conserved epitopes in hemagglutinin (HA) molecules: NAe (NAe-HA) or M2e (M2e-HA) within the HA head domain. Inactivated recombinant NAe-HA and M2e-HA viruses were more effective in inducing IgG antibodies specific for an inserted conserved epitope than live recombinant virus. Recombinant inactivated M2e-HA virus vaccination induced cross protection against H3N2 virus with less weight loss compared to NAe-HA and was more effective in inducing humoral and cellular M2e immune responses. This study provides insight into developing recombinant influenza virus vaccines compatible with current platforms to induce antibody responses to conserved poorly immunogenic epitopes.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/biosynthesis , Neuraminidase/immunology , Orthomyxoviridae Infections/prevention & control , Reassortant Viruses/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/biosynthesis , Cross Protection , Epitopes/chemistry , Epitopes/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H9N2 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Mice , Mice, Inbred BALB C , Neuraminidase/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Reassortant Viruses/drug effects , Reassortant Viruses/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccination/methods , Vaccines, Inactivated , Viral Matrix Proteins/genetics
12.
Vet Microbiol ; 248: 108803, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32827926

ABSTRACT

The H9N2 subtype avian influenza virus (AIV) is one of the most prevalent AIV subtypes that can be found throughout most countries. Currently, due to the neglect of low pathogenic avian influenza virus (LPAIV) and monotonous control technique, an expanding H9N2 virus epizootic have been arisen and causes great economic losses in the poultry industry. Therefore, novel anti-influenza drugs are necessary for the prevention and control of H9N2 AIV. Our previous studies have found that Taishan Pinus massoniana pollen polysaccharides (TPPPS) have antiviral effects, but whether they can inhibit the H9N2 AIV remains unclear. Here, we further investigated the effects of TPPPS on the H9N2 virus and its underlying mechanisms of action. We found that TPPPS significantly inhibited the replication of the H9N2 virus in a dose-dependent manner, especially during the period of virus adsorption in vitro. Transmission electron microscopy demonstrated that TPPPS reduce infection by interfering with virus entry into host cells rather than by interacting with the H9N2 virus particles. A fluorescence quantitative PCR (qPCR) assay and an animal experiment were performed to evaluate the anti-viral effect of TPPPS in vivo. As expected, the lungs of chickens treated with TPPPS had fewer lesions and lower virus contents compared with the PBS group. In addition, pre-treatment with TPPPS clearly enhanced host disease resistance and delayed infection by the H9N2 virus. Taken together, our results reveal that TPPPS suppress H9N2 virus replication both in vitro and in vivo and therefore shows promising as an anti-AIV agent.


Subject(s)
Antiviral Agents/therapeutic use , Influenza A Virus, H9N2 Subtype/drug effects , Pinus/chemistry , Pollen/chemistry , Polysaccharides/therapeutic use , Administration, Oral , Animals , Antibodies, Viral/blood , Chickens/virology , Dogs , Influenza in Birds/drug therapy , Influenza in Birds/prevention & control , Madin Darby Canine Kidney Cells , Poultry Diseases/drug therapy , Poultry Diseases/prevention & control , Poultry Diseases/virology , Viral Load , Virus Internalization/drug effects , Virus Replication/drug effects
13.
J Ethnopharmacol ; 259: 112945, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32389854

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Angelica dahurica (Hoffm.) Benth. & Hook.f. ex Franch. & Sav. (Umbelliferae family) is an herbaceous, perennial plant native to northern and eastern Asia. The root of A. dahurica has traditionally been used under the name "Bai Zhi" as a medicinal plant for colds, dizziness, ulcers, and rheumatism. Moreover, it is also an important ingredient of various prescriptions, such as Gumiganghwal-Tang, for the common cold and influenza. AIM OF THE STUDY: Even though various biological activities of the root of A. dahurica have been reported along with its chemical components, the detailed mechanism of how it exerts anti-influenza activity at the compound level has not been studied. Therefore, we investigated the anti-influenza properties of furanocoumarins purified by bioactivity-guided isolation. MATERIALS AND METHODS: Bioactivity-guided isolation from a 70% EtOH extract of the root of A. dahurica was performed to produce four active furanocoumarins. The inhibition of cytopathic effects (CPEs) was evaluated to ascertain the antiviral activity of these compounds against influenza A (H1N1 and H9N2) viruses. The most potent compound was subjected to detailed mechanistic studies such as the inhibition of viral protein synthesis, CPE inhibition in different phases of the viral replication cycle, neuraminidase (NA) inhibition, antiapoptotic activity using flow cytometry, and immunofluorescence. RESULTS: The bioactivity-guided isolation produced four active furanocoumarins, isoimperatorin (1), oxypeucedanin (2), oxypeucedanin hydrate (3) and imperatorin (4) from the n-BuOH fraction. Among them, compound 2 (followed by compounds 1, 4 and 3) showed a significant CPE inhibition effect, which was stronger than that of the positive control ribavirin, against both H1N1 and H9N2 with an EC50 (µM) of 5.98 ± 0.71 and 4.52 ± 0.39, respectively. Compound 2 inhibited the synthesis of NA and nucleoprotein (NP) in a dose-dependent manner. In the time course assays, the cytopathic effects of influenza A-infected MDCK cells were reduced by 80-90% when treated with compound 2 for 1 and 2 h after infection and declined drastically 3 h after infection. The level of viral NA and NP production was markedly reduced to less than 20% for both proteins in compound 2 (20 µM)-treated cells compared to untreated cells at 2 h after infection. In the molecular docking analysis, compound 2 showed a stronger binding affinity for the C-terminus of polymerase acidic protein (PAC; -36.28 kcal/mol) than the other two polymerase subunits. Compound 2 also exerted an antiapoptotic effect on virus infected cells and significantly inhibited the mRNA expression of caspase-3 and Bax. CONCLUSION: Our results suggest that compound 2 might exert anti-influenza A activity via the inhibition of the early phase of the viral replication cycle, not direct neutralization of surface proteins, such as hemagglutinin and NA, and abnormal apoptosis induced by virus infection. Taken together, these findings suggest that furanocoumarins predominant in A. dahurica play a pivotal role in its antiviral activity. These findings can also explain the reasons for the ethnopharmacological uses of this plant as an important ingredient in many antiviral prescriptions in traditional Chinese medicine (TCM).


Subject(s)
Angelica , Antiviral Agents/pharmacology , Epithelial Cells/drug effects , Furocoumarins/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Plant Extracts/pharmacology , Angelica/chemistry , Animals , Antiviral Agents/isolation & purification , Apoptosis/drug effects , Cytopathogenic Effect, Viral/drug effects , Dogs , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Furocoumarins/isolation & purification , Host Microbial Interactions , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H9N2 Subtype/growth & development , Influenza A Virus, H9N2 Subtype/metabolism , Madin Darby Canine Kidney Cells , Molecular Docking Simulation , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Plant Extracts/isolation & purification , Plant Roots , Virus Replication/drug effects
14.
Vet Res ; 51(1): 68, 2020 May 24.
Article in English | MEDLINE | ID: mdl-32448402

ABSTRACT

Low-pathogenicity avian influenza H9N2 remains an endemic disease worldwide despite continuous vaccination, indicating the need for an improved vaccine strategy. Bacillus subtilis (B. subtilis), a gram-positive and endospore-forming bacterium, is a non-pathogenic species that has been used in probiotic formulations for both animals and humans. The objective of the present study was to elucidate the effect of B. subtilis spores as adjuvants in chickens administered inactivated avian influenza virus H9N2. Herein, the adjuvanticity of B. subtilis spores in chickens was demonstrated by enhancement of H9N2 virus-specific IgG responses. B. subtilis spores enhanced the proportion of B cells and the innate cell population in splenocytes from chickens administered both inactivated H9N2 and B. subtilis spores (Spore + H9N2). Furthermore, the H9N2 and spore administration induced significantly increased expression of the pro-inflammatory cytokines IL-1ß and IL-6 compared to that in the H9N2 only group. Additionally, total splenocytes from chickens immunized with inactivated H9N2 in the presence or absence of B. subtilis spores were re-stimulated with inactivated H9N2. The subsequent results showed that the extent of antigen-specific CD4+ and CD8+ T cell proliferation was higher in the Spore + H9N2 group than in the group administered only H9N2. Taken together, these data demonstrate that B. subtilis spores, as adjuvants, enhance not only H9N2 virus-specific IgG but also CD4+ and CD8+ T cell responses, with an increase in pro-inflammatory cytokine production. This approach to vaccination with inactivated H9N2 together with a B. subtilis spore adjuvant in chickens produces a significant effect on antigen-specific antibody and T cell responses against avian influenza virus.


Subject(s)
Adjuvants, Immunologic/pharmacology , B-Lymphocytes/immunology , Bacillus subtilis/chemistry , Chickens , Influenza A Virus, H9N2 Subtype/drug effects , Influenza in Birds/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Viral/immunology , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Influenza A Virus, H9N2 Subtype/immunology , Poultry Diseases/immunology , Spores, Bacterial/chemistry
15.
Nanomedicine ; 27: 102209, 2020 07.
Article in English | MEDLINE | ID: mdl-32305593

ABSTRACT

Biodegradable nanomaterials can protect antigens from degradation, promote cellular absorption, and enhance immune responses. We constructed a eukaryotic plasmid [pCAGGS-opti441-hemagglutinin (HA)] by inserting the optimized HA gene fragment of H9N2 AIV into the pCAGGS vector. The pCAGGS-opti441-HA/DGL was developed through packaging the pCAGGS-opti441-HA with dendrigraft poly-l-lysines (DGLs). DGL not only protected the pCAGGS-opti441-HA from degradation, but also exhibited high transfection efficiency. Strong cellular immune responses were induced in chickens immunized with the pCAGGS-opti441-HA/DGL. The levels of IFN-γ and IL-2, and lymphocyte transformation rate of the vaccinated chickens increased at the third week post the immunization. For the vaccinated chickens, T lymphocytes were activated and proliferated, the numbers of CD3+CD4+ and CD4+/CD8+ increased, and the chickens were protected completely against H9N2 AIV challenge. This study provides a method for the development of novel AIV vaccines, and a theoretical basis for the development of safe and efficient gene delivery carriers.


Subject(s)
Antibodies, Viral/immunology , Influenza Vaccines/pharmacology , Influenza in Birds/drug therapy , Vaccines, DNA/pharmacology , Animals , Antibodies, Viral/pharmacology , Antibody Formation/drug effects , Antibody Formation/immunology , Chickens/immunology , Chickens/virology , Influenza A Virus, H9N2 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza Vaccines/chemistry , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza in Birds/immunology , Influenza in Birds/virology , Polylysine/chemistry , Polylysine/pharmacology , Vaccines, DNA/chemistry , Vaccines, DNA/immunology
16.
J Nanobiotechnology ; 18(1): 54, 2020 Mar 24.
Article in English | MEDLINE | ID: mdl-32209114

ABSTRACT

BACKGROUND: Influenza viruses (IVs) have become increasingly resistant to antiviral drugs that target neuraminidase and matrix protein 2 due to gene mutations that alter their drug-binding target protein regions. Consequently, almost all recent IV pandemics have exhibited resistance to commercial antiviral vaccines. To overcome this challenge, an antiviral target is needed that is effective regardless of genetic mutations. MAIN BODY: In particular, hemagglutinin (HA), a highly conserved surface protein across many IV strains, could be an effective antiviral target as it mediates binding of IVs with host cell receptors, which is crucial for membrane fusion. HA has 6 disulfide bonds that can easily bind with the surfaces of gold nanoparticles. Herein, we fabricated porous gold nanoparticles (PoGNPs) via a surfactant-free emulsion method that exhibited strong affinity for disulfide bonds due to gold-thiol interactions, and provided extensive surface area for these interactions. A remarkable decrease in viral infectivity was demonstrated by increased cell viability results after exposing MDCK cells to various IV strains (H1N1, H3N2, and H9N2) treated with PoGNP. Most of all, the viability of MDCK cells infected with all IV strains increased to 96.8% after PoGNP treatment of the viruses compared to 33.9% cell viability with non-treated viruses. Intracellular viral RNA quantification by real-time RT-PCR also confirmed that PoGNP successfully inhibited viral membrane fusion by blocking the viral entry process through conformational deformation of HA. CONCLUSION: We believe that the technique described herein can be further developed for PoGNP-utilized antiviral protection as well as metal nanoparticle-based therapy to treat viral infection. Additionally, facile detection of IAV can be achieved by developing PoGNP as a multiplatform for detection of the virus.


Subject(s)
Antiviral Agents/pharmacology , Gold/pharmacology , Influenza A virus/drug effects , Metal Nanoparticles/chemistry , Animals , Dogs , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/genetics , Influenza A virus/genetics , Madin Darby Canine Kidney Cells , Membrane Fusion , Porosity , RNA, Viral/analysis , RNA, Viral/genetics , Virus Internalization
17.
Viruses ; 12(2)2020 02 18.
Article in English | MEDLINE | ID: mdl-32085430

ABSTRACT

Influenza A viruses (IAV) have been a major public health threat worldwide, and options for antiviral therapy become increasingly limited with the emergence of drug-resisting virus strains. New and effective anti-IAV drugs, especially for highly pathogenic influenza, with different modes of action, are urgently needed. The influenza virus glycoprotein hemagglutinin (HA) plays critical roles in the early stage of virus infection, including receptor binding and membrane fusion, making it a potential target for the development of anti-influenza drugs. In this study, we show that OA-10, a newly synthesized triterpene out of 11 oleanane-type derivatives, exhibited significant antiviral activity against four different subtypes of IAV (H1N1, H5N1, H9N2 and H3N2) replications in A549 cell cultures with EC50 ranging from 6.7 to 19.6 µM and a negligible cytotoxicity (CC50 > 640 µM). It inhibited acid-induced hemolysis in a dose-dependent manner, with an IC50 of 26 µM, and had a weak inhibition on the adsorption of H5 HA to chicken erythrocytes at higher concentrations (≥40 µM). Surface plasmon resonance (SPR) analysis showed that OA-10 interacted with HA in a dose-dependent manner with the equilibrium dissociation constants (KD) of the interaction of 2.98 × 10-12 M. Computer-aided molecular docking analysis suggested that OA-10 might bind to the cavity in HA stem region which is known to undergo significant rearrangement during membrane fusion. Our results demonstrate that OA-10 inhibits H5N1 IAV replication mainly by blocking the conformational changes of HA2 subunit required for virus fusion with endosomal membrane. These findings suggest that OA-10 could serve as a lead for further development of novel virus entry inhibitors to prevent and treat IAV infections.


Subject(s)
Antiviral Agents/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/drug effects , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , Virus Internalization/drug effects , A549 Cells , Animals , Antiviral Agents/chemistry , Chickens , Erythrocytes/drug effects , Hemolysis/drug effects , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/drug effects , Influenza A virus/physiology , Inhibitory Concentration 50 , Molecular Docking Simulation
18.
Trop Biomed ; 37(4): 1129-1140, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33612765

ABSTRACT

Herbal medicines are becoming more popular and acceptable day by day due to their effectiveness, limited side effects, and cost-effectiveness. Cholistani plants are reported as a rich source of antibacterial, antifungal, antiprotozoal, antioxidant, and anticancer agents. The current study has evaluated antiviral potential of selected Cholistani plants. The whole plants were collected, ground and used in extract formation with n-hexane, ethyl acetate and n-butanol. All the extracts were concentrated by using a rotary evaporator and concentrate was finally dissolved in an appropriate vol of the same solvent. All of the extracts were tested for their antiviral potential by using 9-11 days old chick embryonated eggs. Each extract was tested against the Avian Influenza virus H9N2 strain (AIV), New Castle Disease virus Lasoota strain (NDV), Infectious bronchitis virus (IBV) and an Infectious bursal disease virus (IBDV). Hemagglutination test (HA) and Indirect Hemagglutination (IHA) tests were performed for different viruses. The overall order of the antiviral potential of Cholistani plants against viruses was NDV>IBV>IBDV>AIV. In terms of antiviral activity from extracts, the order of activity was n-butanol>ethyl acetate>n-hexane. The medicinal plants Achyranthes aspera, Neuroda procumbens, Panicum antidotale, Ochthochloa compressa and Suaeda fruticose were very effective against all four poultry viruses through their extracts. The low IC50 values of these extracts confirm the high antiviral potential against these viruses. It is worth to mention that Achyranthes aspera was found positive against IBDV through all its extracts which overcome the problem of unavailability of any known drug against IBDV. In short, the study proved that Cholistani plants are rich source of antiviral agent and their extracts can be used as good source of antiviral drugs both in crude and in purified form.


Subject(s)
Antiviral Agents/pharmacology , Infectious bronchitis virus/drug effects , Infectious bursal disease virus/drug effects , Influenza A Virus, H9N2 Subtype/drug effects , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Animals , Chick Embryo , Hemagglutination Tests , Pakistan , Phytochemicals/pharmacology , Poultry Diseases/virology
19.
Acta Vet Hung ; 67(4): 610-618, 2019 12.
Article in English | MEDLINE | ID: mdl-31842597

ABSTRACT

The 3D8 single-chain variable fragment (scFv) is a mini-antibody sequence with independent nuclease activity that shows antiviral effects against all types of viruses in chickens and mice. In this study, chickens were treated daily with an oral dose of 109 CFU Lactobacillus paracasei (L. paracasei) expressing either a secreted or anchored 3D8 scFv for three weeks. After L. paracasei administration, the chickens were challenged with avian influenza virus (AIV). From each experimental group, three chickens were directly infected with 100 µL of 107.5 EID50/mL H9N2 AIV and seven chickens were indirectly challenged through contact transmission. oropharyngeal and cloacal swab samples were collected at 3, 5, 7, and 9 days post-inoculation (dpi) from AIV-challenged chickens, AIV Shedding titres were measured by quantitative real-time PCR. Contact transmission in the chickens that were fed 3D8 scFv-secreting L. paracasei showed a significant reduction in viral shedding when compared with other groups. These results suggest that L. paracasei secreting 3D8 provides a basis for the development of ingestible antiviral probiotics with activity against AIV.


Subject(s)
Chickens , Influenza in Birds/drug therapy , Lacticaseibacillus paracasei/chemistry , Poultry Diseases/drug therapy , Probiotics/administration & dosage , Animals , Influenza A Virus, H9N2 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/physiology , Influenza in Birds/virology , Lacticaseibacillus paracasei/genetics , Poultry Diseases/virology , Virus Shedding/drug effects
20.
Microb Pathog ; 137: 103779, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31600542

ABSTRACT

Antiviral susceptibility screening of avian influenza (AI) H9N2 viruses is crucial considering their role at the animal-human interface and potential to cause human infections. The Matrix 2 (M2) inhibitors (amantadine and rimantadine) have been used for prophylaxis and treatment of influenza A virus infections, however, resistance to these drugs has been widely reported. Information about amantadine susceptibility of H9N2 viruses from India is scanty. Matrix genes of 48H9N2 viruses isolated from India during 2009-2017 were sequenced and M2 trans-membrane region sequences were screened for mutations which are known to confer resistance to amantadine namely, L26F, V27A, A30 T/V, S31N and G34E. All the viruses isolated during the year 2009 were sensitive to amantadine. However, resistance started to appear since the year 2010 and all the viruses isolated from the year 2015 onwards showed presence of molecular markers conferring resistance to amantadine. Majority of the resistant viruses exhibited S31 N mutation. Four isolates showed presence of V27A + S31 N dual mutations. Comparison of the M2 sequences from other Asian countries showed different patterns of amantadine resistance wherein phylogenetic analysis of the M genes of the strains from Pakistan formed a separate cluster. In conclusion, the present study reports prevalence and gradual increase of amantadine resistance among AI H9N2 viruses in India, emphasizing the importance of the antiviral surveillance.


Subject(s)
Amantadine/pharmacology , Antiviral Agents/pharmacology , Drug Resistance, Viral , Influenza A Virus, H9N2 Subtype/drug effects , Influenza in Birds/virology , Poultry Diseases/virology , Animals , Birds , Chickens , Genetic Markers/drug effects , India , Influenza A Virus, H9N2 Subtype/classification , Influenza A Virus, H9N2 Subtype/isolation & purification , Influenza A Virus, H9N2 Subtype/physiology , Phylogeny , Quail , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...