Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
Pathogens ; 13(8)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39204284

ABSTRACT

This article deals with Central Nervous System (CNS) disorders of marine mammals as putative neuropathology and neuropathogenesis models for their human and, to some extent, their animal "counterparts" in a dual "One Health" and "Translational Medicine" perspective. Within this challenging context, special emphasis is placed upon Alzheimer's disease (AD), provided that AD-like pathological changes have been reported in the brain tissue of stranded cetacean specimens belonging to different Odontocete species. Further examples of potential comparative pathology interest are represented by viral infections and, in particular, by "Subacute Sclerosing Panencephalitis" (SSPE), a rare neurologic sequela in patients infected with Measles virus (MeV). Indeed, Cetacean morbillivirus (CeMV)-infected striped dolphins (Stenella coeruleoalba) may also develop a "brain-only" form of CeMV infection, sharing neuropathological similarities with SSPE. Within this framework, the global threat of the A(H5N1) avian influenza virus is another major concern issue, with a severe meningoencephalitis occurring in affected pinnipeds and cetaceans, similarly to what is seen in human beings. Finally, the role of Brucella ceti-infected, neurobrucellosis-affected cetaceans as putative neuropathology and neuropathogenesis models for their human disease counterparts is also analyzed and discussed. Notwithstanding the above, much more work is needed before drawing the conclusion marine mammal CNS disorders mirror their human "analogues".

2.
Microb Pathog ; 183: 106330, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37661072

ABSTRACT

The present study was aimed to investigate the role of cannibalism in transmission of H5N1 avian influenza virus to house crows (Corvus splendens). Four crows were intranasally inoculated with 108.0 EID50 (A/crow/India/01CA249/2021) H5N1 highly pathogenic avian influenza (HPAI) virus and were observed for 14 days for any overt signs of illness. Two of the infected crows showed signs of wing paralysis, incoordination, and torticollis. For cannibalism experiment, two crows showing clinical signs were euthanized on 14th day post-infection (dpi) and were kept in the isolator and four naïve healthy crows were introduced along with the euthanized crows. The viscera from the infected carcasses were eaten by all the four crows. Oropharyngeal and cloacal swabs were collected up to 14 days to assess virus excretion. All four crows showed clinical signs viz., dullness, reluctance to move with ruffled feathers on 6th day post cannibalism along with neurological signs including incoordination and paralysis of the wings. All the crows gradually recovered after showing clinical signs and were euthanized on 21st day of observation period. Virus excretion was observed from 3rd to 11th day post cannibalism through both oropharyngeal and cloacal routes with maximum shedding through oropharyngeal route. The virus was isolated from lungs and trachea of one the infected crows at 21st day after euthanasia. All the four crows seroconverted against H5N1 virus infection at 14th day post cannibalism. Our study confirms the transmission of H5N1 virus in crows through cannibalism and highlights how H5N1 virus might circulate in a crow colony once they become infected.


Subject(s)
Crows , Influenza A Virus, H5N1 Subtype , Influenza A virus , Influenza in Birds , Animals , Paralysis , Eating
3.
Sustain Cities Soc ; 70: 102887, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33816082

ABSTRACT

The fast spread of SARS-CoV-2 presented a worldwide challenge to public health, economy, and educational system, affecting wellbeing of human society. With high transmission rates, there are increasing evidences of COVID-19 spread via bioaerosols from an infected person. The current review was conducted to examine airborne pollen impact on COVID-19 transmission and to identify the major gaps for post-pandemic research. The study used all key terms to identify revenant literature and observation were collated for the current research. Based on existing literature, there is a potential association between pollen bioaerosols and COVID-19. There are few studies focusing the impact of airborne pollen on SARS-CoV-2, which could be useful to advance future research. Allergic rhinitis and asthma patients were found to have pre-modified immune activation, which could help to provide protection against COVID-19. However, does airborne pollen acts as a potent carrier for SARS-CoV-2 transport, dispersal and its proliferation still require multidisciplinary research. Further, a clear conclusion cannot be drawn due to limited evidence and hence more research is needed to show how pollen bioaerosols could affect virus survivals. The small but growing literature review focuses on searching for every possible answer to provide additional security layers to overcome near future corona-like infectious diseases.

4.
Front Microbiol ; 11: 553274, 2020.
Article in English | MEDLINE | ID: mdl-33250863

ABSTRACT

Mast cells play an important role in the pathogenesis of highly pathogenic H5N1 avian influenza virus (H5N1-HPAIV) infection. Defective viral particles (DPs) can interfere with the replication of infectious viruses and stimulate the innate immune response of host cells. However, DPs arising from mast cells during HPAIV replication and their potent antiviral actions has not been reported. Here, we showed that the human mastocytoma cell line, HMC-1, allowed for the productive replication of the H5N1-HPAIV. Compared with alveolar cell line A549, DPs were propagated preferentially and abundantly in mast cells following IAV infection, which can be attributed to the wide existence of Argonaute 2 (AGO2) in HMC-1 cells. In addition, DPs generated in H5N1-infected cells could provide great therapeutic protection on mice to fight against various influenza A viruses, which included not only homologous H5N1-HPAIV, but also heterologous H1N1, H3N2, H7N2, and H9N2. Importantly, DPs generated in H5N1-infected HMC-1 cells could diminish viral virulence in vivo and in vitro by triggering a robust antiviral response through type II interferon signaling pathways. This study is the first to illustrate the arising of DPs in H5N1-HPAIV infected mast cells and explore their favorable ability to protect mice from influenza A viruses infection, which provides a novel insight and valuable information for the progress of new strategies to fight influenza A viruses infection, especially highly pathogenic avian influenza virus infection by focusing on the DPs generated in mast cells.

5.
Vet Microbiol ; 250: 108838, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33045633

ABSTRACT

PA-X is a fusion protein of influenza virus which plays a crucial role in modulating influenza virus-induced host innate immune response and subsequent pathogenicity. However, the potential mechanism of PA-X regulation of the host innate immune response remains largely unknown. It is well known that NF-κB signal pathway is crucial for the immediate early step of immune responses activation, while the specific role of PA-X in NF-κB transcriptional activity is totally unknown. In this study, we initially showed that PA-X inhibits NF-κB transcription that stimulated by poly(I:C). We then further determined that the inhibitory effect on NF-κB activation mediated by PA-X was characterized by restricting NF-κB p65 nuclear translocation and nuclear NF-κB p65 activity but not by impeding the phosphorylation of NF-κB p65. Correspondingly, PA-X decreases the amount of NF-κB signaling pathway-associated genes, including TNF-α, Nos2, IL-6 and IL-2. Moreover, PA-X also suppresses both the mRNA and protein expression level of IFN-ß, suggesting the direct contribution of PA-X to the inhibition of NF-κB-regulated IFN-ß expression. Together, our study sheds light on the potential molecular mechanisms underlying the regulation of host NF-κB activity by PA-X and also identifies a novel functional role for PA-X in counteracting the host innate immune response. However, further exploration of the more elaborate mechanism of PA-X-mediated inhibition of NF-κB activity and the associated signaling pathway may help to elucidate its precise mechanism of evading and subverting the host immune response.


Subject(s)
Host Microbial Interactions/immunology , Immunity, Innate , Influenza A Virus, H5N1 Subtype/genetics , NF-kappa B/metabolism , Repressor Proteins/genetics , Signal Transduction , Viral Nonstructural Proteins/genetics , Animals , Cell Line , Chickens , Dogs , Gene Expression Regulation , HEK293 Cells , Humans , Influenza in Birds , Interferon-beta/metabolism , Madin Darby Canine Kidney Cells , Transcription, Genetic , Virus Replication , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
6.
Front Microbiol ; 11: 1246, 2020.
Article in English | MEDLINE | ID: mdl-32595623

ABSTRACT

The protein inhibitor of the activated STAT2 (PIAS2) has been implicated in many cellular processes and can also regulate viral replication in mammals. However, the role of PIAS2 in the highly pathogenic avian influenza virus (HPAIV) H5N1 replication in ducks is still unclear. Through liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay, we identified that duck PIAS2 (duPIAS2) was one protein that interacted with the nucleoprotein (NP) from the H5N1 HPAIV strain of DK212. Through confocal microscopy images and Co-IP assay, we confirmed NP could interact with duPIAS2. Overexpression of duPIAS2 in primary duck embryo fibroblast (DEF) cells was shown to promote DK212 replication, and knockdown of duPIAS2 could repress DK212 replication. We further found duPIAS2 could promote NP SUMOylation through duck SUMO1 (duSUMO1), and the potential SUMOylation sites of NP were at lysines 7, 48, and 87. Furthermore, duPIAS2 promoted the replication of DK212, here relying on the activity of its SUMO E3 ligase. Duck SENP1 (duSENP1), a deSUMOylation enzyme, could repress NP SUMOylation and also inhibit DK212 replication. Together, we identified duPIAS2 could interact with NP and that duPIAS2 promoted H5N1 HPAIV replication, which might be related to NP SUMOylation.

7.
J Interferon Cytokine Res ; 38(7): 290-297, 2018 07.
Article in English | MEDLINE | ID: mdl-30016179

ABSTRACT

The highly pathogenic H5N1 avian influenza virus (AIV) is widespread in waterfowl, causing enormous economic losses and posing a significant threat to public health. An increasing number of reagents have been identified to prevent the spread of influenza; however, there have been no reports on the anti-H5N1 effects of duck interferons, which exhibit antiviral activity against other viruses. Our aim was to investigate the antiviral effects of purified duck interferons. In this study, we successfully cloned and expressed duck interferon gamma (IFN-γ) in Escherichia coli. The antiviral effects of this recombinant duck IFN-γ (rDuIFN-γ) was assessed in vitro and in vivo. rDuIFN-γ displayed antiviral activity against vesicular stomatitis virus and AIV in duck embryo fibroblasts. Pretreating ducks with 3.4 × 104 U rDuIFN-γ also partially decreased mortality from 70% to 30% and delayed onset in 2-day-old Peking ducks. Virus titers in tissues and viral shedding decreased, and the expression of interferon-stimulated genes increased in brain and spleen in rDuIFN-γ-treated ducks. These results indicate that duck IFN-γ has the potential to inhibit viral replication in ducks.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H5N1 Subtype/drug effects , Interferon-gamma/pharmacology , Recombinant Proteins/pharmacology , Virus Replication/drug effects , Animals , Ducks , Influenza A Virus, H5N1 Subtype/growth & development , Microbial Sensitivity Tests
8.
J Interferon Cytokine Res ; 38(4): 145-152, 2018 04.
Article in English | MEDLINE | ID: mdl-29638205

ABSTRACT

The antiviral cytokine interferon-alpha (IFN-α) plays a critical role in the innate immune system. Previous studies have shown that recombinant chicken IFN-α inhibits avian influenza virus (AIV) replication in vivo; however, the antiviral effect of recombinant duck IFN-α (rDuIFN-α) on highly pathogenic AIV remains unknown. In this study, the duck IFN-α gene was cloned, expressed, and purified. The antiviral effects of the resulting rDuIFN-α were further evaluated in vitro and in vivo. Our results showed that rDuIFN-α inhibited the replication of vesicular stomatitis virus (VSV) and AIV in duck embryo fibroblasts in vitro, with antiviral activities against VSV and AIV of 2.1 × 105 and 4.1 × 105 U/mg, respectively. We next investigated the anti-H5N1 AIV effect of intramuscular injection of rDuIFN-α in vivo. rDuIFN-α reduced viral titers in the brains, lungs, and spleens of 2-day-old (2D) ducks compared with that in the virus-challenged control group, and pretreatment with rDuIFN-α reduced mortality from 60% to 10% in 2D ducks. Moreover, rDuIFN-α increased the expression of IFN-stimulated genes in the brains and spleens of 2D ducks. Our results demonstrate that rDuIFN-α blocks VSV and H5N1 influenza virus infection in vitro and exhibits antiviral effects against H5N1 influenza virus infection in 2D ducks.


Subject(s)
Antiviral Agents/pharmacology , Ducks , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/drug therapy , Influenza in Birds/virology , Interferon-alpha/pharmacology , Animals , Influenza A Virus, H5N1 Subtype/immunology , Influenza in Birds/immunology , Microbial Sensitivity Tests
9.
J Virol ; 92(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29563293

ABSTRACT

Transmission of avian influenza (AI) viruses to mammals involves phylogenetic bottlenecks that select small numbers of variants for transmission to new host species. However, little is known about the AI virus quasispecies diversity that produces variants for virus adaptation to humans. Here, we analyzed the hemagglutinin (HA) genetic diversity produced during AI H5N1 single-virus infection of primary human airway cells and characterized the phenotypes of these variants. During single-virus infection, HA variants emerged with increased fitness to infect human cells. These variants generally had decreased HA thermostability, an indicator of decreased transmissibility, that appeared to compensate for their increase in α2,6-linked sialic acid (α2,6 Sia) binding specificity and/or in the membrane fusion pH threshold, each of which is an advantageous mutational change for viral infection of human airway epithelia. An HA variant with increased HA thermostability also emerged but could not outcompete variants with less HA thermostability. These results provided data on HA quasispecies diversity in human airway cells.IMPORTANCE The diversity of the influenza virus quasispecies that emerges from a single infection is the starting point for viral adaptation to new hosts. A few studies have investigated AI virus quasispecies diversity during human adaptation using clinical samples. However, those studies could be appreciably affected by individual variability and multifactorial respiratory factors, which complicate identification of quasispecies diversity produced by selective pressure for increased adaptation to infect human airway cells. Here, we found that detectable HA genetic diversity was produced by H5N1 single-virus infection of human airway cells. Most of the HA variants had increased fitness to infect human airway cells but incurred a fitness cost of less HA stability. To our knowledge, this is the first report to characterize the adaptive changes of AI virus quasispecies produced by infection of human airway cells. These results provide a better perspective on AI virus adaptation to infect humans.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza, Human/transmission , Quasispecies/genetics , Receptors, Virus/metabolism , Respiratory Mucosa/cytology , Animals , Cell Line , Chlorocebus aethiops , Dogs , Genetic Variation/genetics , HEK293 Cells , Humans , Influenza A Virus, H5N1 Subtype/classification , Influenza, Human/pathology , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Receptors, Virus/genetics , Respiratory Mucosa/virology , Respiratory System/virology , Sialic Acids/metabolism , Vero Cells , Virus Attachment
10.
Clin Vaccine Immunol ; 24(12)2017 Dec.
Article in English | MEDLINE | ID: mdl-29021304

ABSTRACT

The kinetics, longevity, and breadth of antibodies to influenza virus neuraminidase (NA) in archival, sequential serum/plasma samples from influenza A virus (IAV) H5N1 infection survivors and from patients infected with the 2009 pandemic IAV (H1N1) virus were determined using an enzyme-linked lectin-based assay. The reverse-genetics-derived H4N1 viruses harboring a hemagglutinin (HA) segment from A/duck/Shan Tou/461/2000 (H4N9) and an NA segment derived from either IAV H5N1 clade 1, IAV H5N1 clade 2.3.4, the 2009 pandemic IAV (H1N1) (H1N1pdm), or A/Puerto Rico/8/1934 (H1N1) virus were used as the test antigens. These serum/plasma samples were also investigated by microneutralization (MN) and/or hemagglutination inhibition (HI) assays. Neuraminidase-inhibiting (NI) antibodies against N1 NA of both homologous and heterologous viruses were observed in H5N1 survivors and H1N1pdm patients. H5N1 survivors who were never exposed to H1N1pdm virus developed NI antibodies to H1N1pdm NA. Seroconversion of NI antibodies was observed in 65% of the H1N1pdm patients at day 7 after disease onset, but an increase in titer was not observed in serum samples obtained late in infection. On the other hand, an increase in seroconversion rate with the HI assay was observed in the follow-up series of sera obtained on days 7, 14, 28, and 90 after infection. The study also showed that NI antibodies are broadly reactive, while MN and HI antibodies are more strain specific.


Subject(s)
Antibodies, Viral/blood , Cross Reactions , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza, Human/immunology , Neuraminidase/immunology , Seroconversion , Viral Proteins/immunology , Adolescent , Adult , Child , Child, Preschool , Female , Hemagglutination Inhibition Tests/methods , Humans , Male , Neutralization Tests , Time Factors , Young Adult
11.
Proteomes ; 5(3)2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28895884

ABSTRACT

Septin forms a conserved family of cytoskeletal guanosine triphosphate (GTP) binding proteins that have diverse roles in protein scaffolding, vesicle trafficking, and cytokinesis. The involvement of septins in infectious viral disease pathogenesis has been demonstrated by the upregulation of SEPT5 protein and its mRNA in brain tissues of H5N1-infected chickens, thus, providing evidence for the potential importance of this protein in the pathogenesis of neurovirulence caused by the avian influenza virus. In this study, cloning, expression, and purification of Gallus gallus SEPT5 protein was performed in Escherichia coli. The SEPT5 gene was inserted into the pRSETB expression vector, transformed in the E. coli BL21 (DE3) strain and the expression of SEPT5 protein was induced by IPTG. The SEPT5 protein was shown to be authentic as it was able to be pulled down by a commercial anti-SEPT5 antibody in a co-immunoprecipitation assay. In vivo aggregation of the recombinant protein was limited by cultivation at a reduced temperature of 16 °C. Using co-immunoprecipitation techniques, the purified recombinant SEPT5 protein was used to pull down host's interacting or binding proteins, i.e., proteins of brains of chickens infected with the H5N1 influenza virus. Interacting proteins, such as CRMP2, tubulin proteins, heat-shock proteins and other classes of septins were identified using LCMS/MS. Results from this study suggest that the codon-optimized SEPT5 gene can be efficiently expressed in the E. coli bacterial system producing authentic SEPT5 protein, thus, enabling multiple host's proteins to interact with the SEPT5 protein.

12.
Poult Sci ; 94(8): 1836-42, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26069254

ABSTRACT

Ducks are a natural reservoir for H5N1 highly pathogenic avian influenza (HPAI) viruses, which produces a range of clinical outcomes from asymptomatic infections to severe disease with mortality. Vaccination against HPAI is one of the few methods available for controlling avian influenza virus (AIV) infection in domestic ducks; therefore, it is necessary to improve vaccine efficacy against HPAI in domestic ducks. However, few studies have focused on enhancing the immune response by testing alternative administration routes and adjuvants. While attempting to maximize the efficacy of a vaccine, it is important to select an appropriate vaccine delivery route and adjuvant to elicit an enhanced immune response. Although several studies have indicated that the vaccination of ducks against HPAI viruses has offered protection against lethal virus challenge, the immunogenicity of the vaccine still requires improvement. In this study, we characterized the immune response following a novel vaccination strategy against H5N1 HPAI virus in domestic ducks. Our novel intradermal delivery system and the application of the cytosine-phosphodiester-guanine (CpG) oligodeoxynucleotide (ODN) adjuvant allowed us to obtain information regarding the sustained vaccine immunity. Compared with the intramuscular route of vaccination, the intradermal route resulted in higher antibody titer as well as lower antibody deviation following secondary vaccination. In addition, the use of a CpG-ODN adjuvant had a dose-sparing effect on antibody titer. Furthermore, when a high dose of antigen was used, the CpG-ODN-adjuvanted vaccine maintained a high mean antibody titer. This data demonstrates that intradermal immunization combined with administration of CpG-ODN as an adjuvant may be a promising strategy for improving vaccine efficacy in domestic ducks.


Subject(s)
CpG Islands/immunology , Ducks/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza in Birds/prevention & control , Oligodeoxyribonucleotides/immunology , Adjuvants, Immunologic , Animals , Influenza Vaccines , Injections, Intradermal
13.
Poult Sci ; 94(6): 1170-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25840960

ABSTRACT

Laboratory of genetics and physiology 2 (LGP2) is an important intracellular receptor that recognizes viral RNAs in innate immunity. In this study, a novel LGP2 cDNA was identified from the spleen of a Muscovy duck (Cairina moschata). The deduced amino acid sequence of Muscovy duck LGP2 (MDLGP2) consisted of 675 amino acid residues. The peptide contained two main structure domains: six important motifs, including a DExD/H box for RNA helicase activity in the RNA helicase region located at the N-terminal region, and two Zn2+-binding regions with an RNA-binding loop in the C-terminus regulatory domain (CTD). The MdLGP2 mRNA was ubiquitously expressed in the tested tissues, with high expression levels in glandular stomach, colon, ileum, crop, and caecum tissues, and low expression levels in the brain, skin, and heart. The mRNA expression of MdLGP2 was significantly upregulated in the brain, spleen, and lungs of ducks in the early stages of postinfection with H5N1 highly pathogenic avian influenza virus (HPAIV). These results suggested that MdLGP2 was involved in the early stages of antiviral innate immune response in ducks after infection with H5N1 HPAIV. However, whether it plays a positive or negative regulatory role in the host antiviral response requires further investigation.


Subject(s)
Avian Proteins/genetics , Immunity, Innate , Influenza A Virus, H5N1 Subtype/physiology , Influenza in Birds/genetics , Influenza in Birds/immunology , Amino Acid Sequence , Animals , Avian Proteins/metabolism , Brain/immunology , Brain/metabolism , Cloning, Molecular , Ducks , Influenza in Birds/virology , Lung/immunology , Lung/metabolism , Molecular Sequence Data , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction/veterinary , Sequence Alignment/veterinary , Spleen/immunology , Spleen/metabolism
14.
Biosens Bioelectron ; 65: 211-9, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25461160

ABSTRACT

A switch of viral hemagglutinin receptor binding specificity from bird-type α2,3- to human-type α2,6-linked sialic acid is necessary for an avian influenza virus to become a pandemic virus. In this study, an easy-to-use strip test to detect receptor binding specificity of influenza virus was developed. A biotinylated anti-hemagglutinin antibody that bound a broad range of group 1 influenza A viruses and latex-conjugated α2,3 (blue) and α2,6 (red) sialylglycopolymers were used in an immunochromatographic strip test, with avidin and lectin immobilized on a nitrocellulose membrane at test and control lines, respectively. Accumulation of a sialylglycopolymer-virus-antibody complex at the test line was visualized by eye. The strip test could be completed in 30min and did not require special equipment or skills, thereby avoiding some disadvantages of current methods for analyzing receptor binding specificity of influenza virus. The strip test could detect the receptor binding specificity of a wide range of influenza viruses, as well as small increases in the binding affinity of variant H5N1 viruses to α2,6 sialylglycans at viral titers >128 hemagglutination units. The strip test results were in agreement with those of ELISA virus binding assays, with correlations >0.95. In conclusion, the immunochromatographic strip test developed in this study should be useful for monitoring potential changes in the receptor binding specificity of group 1 influenza A viruses in the field.


Subject(s)
Birds/virology , Chromatography, Affinity/instrumentation , Influenza A virus/isolation & purification , Influenza in Birds/diagnosis , Reagent Strips/analysis , Animals , Chromatography, Affinity/economics , Equipment Design , Hemagglutinin Glycoproteins, Influenza Virus/isolation & purification , Humans , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza, Human/diagnosis
15.
Chinese Pharmacological Bulletin ; (12): 647-654, 2015.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-464378

ABSTRACT

Aim To study the inhibitory activities of potential new anti-influenza virus agents,3-O-β-chaco-triosyl pentacyclic triterpenoids against the entry of H5N1influenza viruses.Methods Three target com-pounds were designed and synthesized structurally re-lated to the lead compound 3-O-β-chacotriosyl dioscin derivative (1 )with inhibitory activities against H5N1 influenza viruses.The inhibitory activities of these tar-get compounds were tested at a cellular level pseudo vi-rus system targeting H5N1 influenza viruse entry.Re-sults All the compounds 1 a,1 b and 1 c showed po-tent inhibitory activities against the entry of A/Thai-land/Kan353/2004 pseudo virus into the target cells, of which compound 1 b showed the best inhibitory activ-ity with an IC50 value of (1.25 ±0.22)μmol·L-1. Conclusion The SARs analysis of these compounds indicated that replacement of the aglycone moiety of compound 1 with pentacyclic triterpenoids could in-crease antiviral activity.Different types of pentacyclic triterpen as aglycone residue had the significant influ-ence on the inhibitory activity (1 b >1 c >1 a),sug-gesting ursane type of triterpenes was superior to the two other kinds of triterpenes as aglycone residue.

16.
World J Virol ; 3(4): 30-6, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25396120

ABSTRACT

AIM: To characterise neuraminidase (NA) substrate specificity of avian influenza H5N1 strains from humans and birds comparing to seasonal influenza virus. METHODS: Avian influenza H5N1 strains from humans and birds were recruited for characterising their NA substrate specificity by using a modified commercial fluorescence Amplex Red assay. This method can identify the preference of α2,6-linked sialic acid or α2,3-linked sialic acid. Moreover, to avoid the bias of input virus, reverse genetic virus using NA gene from human isolated H5N1 were generated and used to compare with the seasonal influenza virus. Lastly, the substrate specificity profile was further confirmed by high-performance liquid chromatography (HPLC) analysis of the enzymatic product. RESULTS: The H5N1 NA showed higher activity on α2,3-linked sialic acid than α2,6-linked (P < 0.0001). To compare the NA activity between the H5N1 and seasonal influenza viruses, reverse genetic viruses carrying the NA of H5N1 viruses and NA from a seasonal H3N2 virus was generated. In these reverse genetic viruses, the NA activity of the H5N1 showed markedly higher activity against α2,3-linked sialic acid than that of the H3N2 virus, whereas the activities on α2,6-linkage were comparable. Interestingly, NA from an H5N1 human isolate that was previously shown to have heamagglutinin (HA) with dual specificity showed reduced activity on α2,3-linkage. To confirm the substrate specificity profile, HPLC analytic of enzymatic product was performed. Similar to Amplex red assay, H5N1 virus showed abundant preference on α2,3-linked sialic acid. CONCLUSION: H5N1 virus maintains the avian specific NA and NA changes may be needed to accompany changes in HA receptor preference for the viral adaptation to humans.

17.
Vaccine ; 31(50): 5953-9, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24144474

ABSTRACT

The duck enteritis virus (DEV) may be a promising candidate viral vector for an aquatic poultry vaccination that can protect against multiple pathogens because it has a very large genome and a narrow host range. Recently, we described two DEV recombinants that contained deletions of the viral US2 or gIgE genes. The hemagglutinin (HA) gene of an H5N1-type highly pathogenic avian influenza virus (HPAIV) of goose origin was inserted into the deletion sites to construct two rDEVs expressing the AIV HA antigen. The resulting rDEV-ΔgIgE-HA or rDEV-ΔUS2-HA recombinant DEV viruses were used to infect duck embryo fibroblasts. Reverse transcription PCR, immunofluorescence and western blot analysis results indicated that rDEV-ΔgIgE-HA and rDEV-ΔUS2-HA were successfully expressed in duck embryo fibroblasts (DEFs). To investigate whether the HA gene could be stably maintained in the recombinant viruses, the viruses were passaged in DEFs 18 times. The HA gene in both recombinants could be detected by PCR amplification. The immunized four-week-old ducks induced specific antibodies against DEV and AIV HA and were protected against challenge infections with DEV AV1221 viruses.


Subject(s)
Drug Carriers , Genetic Vectors , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Mardivirus/genetics , Animals , Antibodies, Viral/blood , Cells, Cultured , Ducks , Fibroblasts/virology , Gene Expression Profiling , Genomic Instability , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza in Birds/immunology , Serial Passage , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
18.
Vet Microbiol ; 166(3-4): 386-93, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-23920409

ABSTRACT

Our previous studies have illustrated three strains of duck-origin H5N1 highly pathogenic avian influenza viruses (HPAIVs) had varying levels of pathogenicity in ducks (Sun et al., 2011). However, the host immune response of ducks infected with those of H5N1 HPAIVs was unclear. Here, we compared viral distribution and mRNA expression of immune-related genes in ducks following infection with the two HPAIV (A/Duck/Guangdong/212/2004, DK212 and A/Duck/Guangdong/383/2008, DK383). DK383 could replicate in the tested tissue of ducks (brain, spleen, lungs, cloacal bursa, kidney, and pancreas) more rapid and efficiently than DK212 at 1 and 2 days post-inoculation. Quantitative real-time PCR analysis showed that the expression levels of TLR3, IL-6, IL-8, and MHC class II in brains were higher than those of respective genes in lungs during the early stage of post infection. Furthermore, the expression levels of IL-6 and IL-8 in the brain of ducks following infection with DK383 were remarkably higher than those of ducks infected with DK212, respectively. Our results suggest that the shift in the H5N1 HPAIVs to increased virulence in ducks may be associated with efficient and rapid replication of the virus, accompanied by early destruction of host immune responses. These data are helpful to understand the underlying mechanism of the different outcome of H5N1 HPAIVs infection in ducks.


Subject(s)
Ducks/virology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/immunology , Poultry Diseases/immunology , Animals , Ducks/immunology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza in Birds/genetics , Influenza in Birds/pathology , Influenza in Birds/virology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-8/genetics , Interleukin-8/immunology , Lung/immunology , Lung/pathology , Poultry Diseases/pathology , Poultry Diseases/virology , Real-Time Polymerase Chain Reaction , Spleen/immunology , Spleen/pathology , Virulence
19.
Virologica Sinica ; (6): 425-431, 2010.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-402282

ABSTRACT

Chicken embryo fibroblasts(CEFs)are among the most commonly used cells for the study of interactions between chicken hosts and H5N1 avian influenza virus(AIV).In this study,the expression of eleven housekeeping genes typically used for the normalization of quantitative real-time PCR(QPCR)analysis in mammals were compared in CEFs infected with H5N1 AIV to determine the most reliable reference genes in this system.CEFs cultured from 10-day-old SPF chicken embryos were infected with 100 TCID50 of H5N1 AIV and harvested at 3,12,24 and 30 hours post-infection.The expression levels of the eleven reference genes in infected and uninfected CEFs were determined by real-time PCR.Based on expression stability and expression levels,our data suggest that the ribosomal protein L4(RPL4)and tyrosine 3-monooxygenase tryptophan 5-monooxygenase activation protein zeta polypeptide(YWHAZ)are the best reference genes to use in the study of host cell response to H5N1 AIV infection.However,for the study of replication levels of H5N1 AIV in CEFs,the β-actin gene(ACTB)and the ribosomal protein L4(RPL4)gene are the best references.

20.
Acta Laboratorium Animalis Scientia Sinica ; (6): 271-277,后插1, 2010.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-597433

ABSTRACT

Objective To test our hypothesis that sensitivity to avian influenza A(H5N1)virus varies among mouse strain backgrounds, we compared the pathogenicity of H5N1 viral infection in 5 mouse strains. Methods Onehundred-fifty mice from 2 inbred strains(BALB/c and C57BL/6), and 3 outbred stocks(ICR, NIH Swiss, and KM Swiss)were used. Thirty mice of each strain were subjected to an infected group(20 mice), in which mice were inoculated with 0. 1 mL(104.875 TCID50)of A/Goose/Guangdong/NH/2003(H5N1)virus intra-nasally; ten control mice received noninfectious allantoic fluid. Clinical signs were assessed daily for 14 days post-infection. Necropsy was performed on mice that died during the experiment and those euthanized at end of study. Tissue samples were collected for viral isolation and pathological analysis. Results H5N1 virus infection can cause respiratory illness in all 5 strains with severe or minor acute respiratory distress symptoms, but with different mortality rates: 70% in BALB/c; 50% in ICR; 40% in NIH Swiss; 25% in C57BL/6; and 10% in KM Swiss mice. Necrotizing interstitial pneumonia was found in all cases of death. The virus was isolated from the lungs of all infected dead mice. Conclusion A/Goose/Guangdong/NH/2003 (H5N1)virus can infect all mouse strains used in this study, and can cause clinical symptoms and pathological changes similar to those found in humans infected with HSN1 viruses. However, the pathogenicity of H5N1 viral infection varies significantly between the different mouse strains. Thus, in future study of H5N1 virus infections the mouse strain most relevant to their particular research purpose should be selected as animal model.

SELECTION OF CITATIONS
SEARCH DETAIL