Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Int J Mol Sci ; 25(2)2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38279237

ABSTRACT

Amidst increasing concern about antibiotic resistance resulting from the overuse of antibiotics, there is a growing interest in exploring alternative agents. One such agent is citric acid, an organic compound commonly used for various applications. Our research findings indicate that the inclusion of citric acid can have several beneficial effects on the tight junctions found in the mouse intestine. Firstly, the study suggests that citric acid may contribute to weight gain by stimulating the growth of intestinal epithelial cells (IE-6). Citric acid enhances the small intestinal villus-crypt ratio in mice, thereby promoting intestinal structural morphology. Additionally, citric acid has been found to increase the population of beneficial intestinal microorganisms, including Bifidobacterium and Lactobacillus. It also promotes the expression of important protein genes such as occludin, ZO-1, and claudin-1, which play crucial roles in maintaining the integrity of the tight junction barrier in the intestines. Furthermore, in infected IEC-6 cells with H9N2 avian influenza virus, citric acid augmented the expression of genes closely associated with the influenza virus infection. Moreover, it reduces the inflammatory response caused by the viral infection and thwarted influenza virus replication. These findings suggest that citric acid fortifies the intestinal tight junction barrier, inhibits the replication of influenza viruses targeting the intestinal tract, and boosts intestinal immune function.


Subject(s)
Influenza A Virus, H9N2 Subtype , Influenza, Human , Animals , Mice , Humans , Citric Acid/pharmacology , Citric Acid/metabolism , Influenza, Human/metabolism , Intestines/microbiology , Intestinal Mucosa/metabolism , Tight Junctions/metabolism , Immunity
2.
Virol J ; 20(1): 76, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37085816

ABSTRACT

BACKGROUND: H9N2 virus is mainly transmitted through the respiratory mucosal pathway, so mucosal immunity is considered to play a good role in controlling avian influenza infection. It is commonly accepted that no adequate mucosal immunity is achieved by inactivated vaccines, which was widely used to prevent and control avian influenza virus infection. Thus, an improved vaccine to induce both mucosal immunity and systemic immunity is urgently required to control H9N2 avian influenza outbreaks in poultry farms. METHODS: In this study, we constructed a novel Lactococcus lactis (L. lactis) strain expressing a recombinant fusion protein consisting of the HA1 proteins derived from an endemic H9N2 virus strain and chicken IgY Fc fragment. We evaluated the immunogenicity and protective efficacy of this recombinant L. lactis HA1-Fc strain. RESULTS: Our data demonstrated that chickens immunized with L. lactis HA1-Fc strain showed significantly increased levels of serum antibodies, mucosal secretory IgA, T cell-mediated immune responses, and lymphocyte proliferation. Furthermore, following challenge with H9N2 avian influenza virus, chickens immunized with L. lactis HA1-Fc strain showed reduced the weight loss, relieved clinical symptoms, and decreased the viral titers and the pathological damage in the lung. Moreover, oropharyngeal and cloacal shedding of the H9N2 influenza virus was detected in chicken immunized with L. lactis HA1-Fc after infection, the results showed the titer was low and reduced quickly to reach undetectable levels at 7 days after infection. CONCLUSION: Our data showed that the recombinant L. lactis HA1-Fc strain could induce protective mucosal and systemic immunity, and this study provides a theoretical basis for improving immune responses to prevent and control H9N2 virus infection.


Subject(s)
Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Lactococcus lactis , Animals , Chickens , Influenza A Virus, H9N2 Subtype/genetics , Influenza in Birds/prevention & control , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Immunity, Mucosal , Influenza Vaccines/genetics , Vaccination , Antibodies, Viral
3.
J Gen Virol ; 102(6)2021 06.
Article in English | MEDLINE | ID: mdl-34061017

ABSTRACT

Avian H9N2 influenza viruses in East Asia are genetically diversified and multiple genotypes (A-W) have been established in poultry. Genotype S strains are currently the most prevalent strains, have caused many human infections and pose a public health threat. In this study, human adaptation mutations in the PB2 polymerase in genotype S strains were identified by database screening. Several PB2 double mutations were identified that acted cooperatively to produce higher genotype S virus polymerase activity and replication in human cells than in avian cells and to increase viral growth and virulence in mice. These mutations were chronologically and phylogenetically clustered in a new group within genotype S viruses. Most of the relevant human virus isolates carry the PB2-A588V mutation together with another PB2 mutation (i.e. K526R, E627V or E627K), indicating a host adaptation advantage for these double mutations. The prevalence of PB2 double mutations in human H9N2 virus isolates has also been found in genetically related human H7N9 and H10N8 viruses. These results suggested that PB2 double mutations in viruses in the field acted cooperatively to increase human adaptation of the currently prevalent H9N2 genotype S strains. This may have contributed to the recent surge of H9N2 infections and may be applicable to the human adaptation of several other avian influenza viruses. Our study provides a better understanding of the human adaptation pathways of genetically related H9N2, H7N9 and H10N8 viruses in nature.


Subject(s)
Host Adaptation , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/physiology , Influenza, Human/virology , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Animals , Birds , Cell Line , Genes, Viral , Genotype , HEK293 Cells , Humans , Influenza A Virus, H9N2 Subtype/classification , Influenza A Virus, H9N2 Subtype/isolation & purification , Influenza in Birds/virology , Mice , Mice, Inbred BALB C , Models, Molecular , Mutation , Orthomyxoviridae Infections/virology , Phylogeny , Poultry , RNA-Dependent RNA Polymerase/chemistry , Reassortant Viruses/genetics , Viral Proteins/chemistry , Viral Zoonoses , Virulence/genetics
4.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32641475

ABSTRACT

Some avian influenza (AI) viruses have a deletion of up to 20 to 30 amino acids in their neuraminidase (NA) stalk. This has been associated with changes in virus replication and host range. Currently prevalent H9N2 AI viruses have only a 2- or 3-amino-acid deletion, and such deletions were detected in G1 and Y280 lineage viruses, respectively. The effect of an NA deletion on the H9N2 phenotype has not been fully elucidated. In this study, we isolated G1 mutants that carried an 8-amino-acid deletion in their NA stalk. To systematically analyze the effect of NA stalk length and concomitant (de)glycosylation on G1 replication and host range, we generated G1 viruses that had various NA stalk lengths and that were either glycosylated or not glycosylated. The stalk length was correlated with NA sialidase activity, using low-molecular-weight substrates, and with virus elution efficacy from erythrocytes. G1 virus replication in avian cells and eggs was positively correlated with the NA stalk length but was negatively correlated in human cells and mice. NA stalk length modulated G1 virus entry into host cells, with shorter stalks enabling more efficient G1 entry into human cells. However, with a hemagglutinin (HA) with a higher α2,6-linked sialylglycan affinity, the effect of NA stalk length on G1 virus infection was reversed, with shorter NA stalks reducing virus entry into human cells. These results indicate that a balance between HA binding affinity and NA sialidase activity, modulated by NA stalk length, is required for optimal G1 virus entry into human airway cells.IMPORTANCE H9N2 avian influenza (AI) virus, one of the most prevalent AI viruses, has caused repeated poultry and human infections, posing a huge public health risk. The H9N2 virus has diversified into multiple lineages, with the G1 lineage being the most prevalent worldwide. In this study, we isolated G1 variants carrying an 8-amino-acid deletion in their NA stalk, which is, to our knowledge, the longest deletion found in H9N2 viruses in the field. The NA stalk length was found to modulate G1 virus entry into host cells, with the effects being species specific and dependent on the corresponding HA binding affinity. Our results suggest that, in nature, H9N2 G1 viruses balance their HA and NA functions by the NA stalk length, leading to the possible association of host range and virulence in poultry and mammals during the evolution of G1 lineage viruses.


Subject(s)
Gene Expression Regulation, Viral , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H9N2 Subtype/genetics , Influenza in Birds/virology , Neuraminidase/genetics , Orthomyxoviridae Infections/virology , Amino Acid Sequence , Animals , Chickens , Genotype , Glycosylation , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinins , Host Specificity , Host-Pathogen Interactions/genetics , Humans , Influenza A Virus, H9N2 Subtype/metabolism , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza in Birds/genetics , Influenza in Birds/metabolism , Influenza in Birds/pathology , Mice , Neuraminidase/metabolism , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Phenotype , Phylogeny , Receptors, Virus , Sequence Deletion , Structure-Activity Relationship , Virulence , Virus Internalization , Virus Replication
5.
Microb Pathog ; 150: 104645, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33285220

ABSTRACT

Influenza virus is responsible for significant morbidity and mortality worldwide. Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is the major cause of death in influenza virus infected patients. Recent studies indicated that active glucagon like peptide-1 (GLP-1) encoded by glucagon (GCG) gene exerts anti-inflammatory functions. The aim of this study was to determine the potential role of active GLP-1 in H9N2 influenza virus-induced ALI/ARDS in mice. First, we uncovered that GCG mRNA expression levels and GCG precursor protein levels were significantly increased, but total GLP-1 and active GLP-1 levels were decreased in the lungs of H9N2-infected mice. Next, liraglutide, an active GLP-1 analogue, was used to treat infected mice and to observe its effects on H9N2 virus-induced ALI. Liraglutide treatment ameliorated the declined body weight, decreased food intake and mortality observed in infected mice. It also alleviated the severity of lung injury, including lowering lung index, decreasing inflammatory cell infiltration and lowing total protein levels in bronchoalveolar lavage fluid (BALF). In addition, liraglutide did not influence viral titers in infected lungs, but decreased the levels of interleukin-1ß, interleukin-6 and tumor necrosis factor-α in BALF. These results indicated that liraglutide alleviated H9N2 virus-induced ALI in mice most likely due to lower levels of pro-inflammatory cytokines.


Subject(s)
Acute Lung Injury , Influenza A Virus, H9N2 Subtype , Acute Lung Injury/drug therapy , Animals , Bronchoalveolar Lavage Fluid , Glucagon-Like Peptide 1 , Humans , Liraglutide/pharmacology , Lung , Mice
6.
Virol J ; 18(1): 22, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33461581

ABSTRACT

BACKGROUND: Oxidative stress is an important pathogenic factor in influenza A virus infection. It has been found that reactive oxygen species induced by the H9N2 influenza virus is associated with viral replication. However, the mechanisms involved remain to be elucidated. METHODS: In this study, the role of autophagy was investigated in H9N2 influenza virus-induced oxidative stress and viral replication in A549 cells. Autophagy induced by H9N2 was inhibited by an autophagy inhibitor or RNA interference, the autophagy level, viral replication and the presence of oxidative stress were detected by western blot, TCID50 assay, and Real-time PCR. Then autophagy and oxidative stress were regulated, and viral replication was determined. At last, the Akt/TSC2/mTOR signaling pathways was detected by western blot. RESULTS: Autophagy was induced by the H9N2 influenza virus and the inhibition of autophagy reduced the viral titer and the expression of nucleoprotein and matrix protein. The blockage of autophagy suppressed the H9N2 virus-induced increase in the presence of oxidative stress, as evidenced by decreased reactive oxygen species production and malonaldehyde generation, and increased superoxide dismutase 1 levels. The changes in the viral titer and NP mRNA level caused by the antioxidant, N-acetyl-cysteine (NAC), and the oxidizing agent, H2O2, confirmed the involvement of oxidative stress in the control of viral replication. NAC plus transfection with Atg5 siRNA significantly reduced the viral titer and oxidative stress compared with NAC treatment alone, which confirmed that autophagy was involved in the replication of H9N2 influenza virus by regulating oxidative stress. Our data also revealed that autophagy was induced by the H9N2 influenza virus through the Akt/TSC2/mTOR pathway. The activation of Akt or the inhibition of TSC2 suppressed the H9N2 virus-induced increase in the level of LC3-II, restored the decrease in the expression of phospho-pAkt, phospho-mTOR and phospho-pS6 caused by H9N2 infection, suppressed the H9N2-induced increase in the presence of oxidative stress, and resulted in a decrease in the viral titer. CONCLUSION: Autophagy is involved in H9N2 virus replication by regulating oxidative stress via the Akt/TSC2/mTOR signaling pathway. Thus, autophagy maybe a target which may be used to improve antiviral therapeutics.


Subject(s)
Alveolar Epithelial Cells/virology , Autophagy/genetics , Gene Expression Regulation , Influenza A Virus, H9N2 Subtype/physiology , Orthomyxoviridae Infections/veterinary , Oxidative Stress/genetics , Virus Replication , A549 Cells , Animals , Humans , Influenza A Virus, H9N2 Subtype/pathogenicity , Signal Transduction , Swine
7.
J Gen Virol ; 100(9): 1273-1281, 2019 09.
Article in English | MEDLINE | ID: mdl-31305236

ABSTRACT

Adaptation of PB2 protein is important for the establishment of avian influenza viruses in mammalian hosts. Here, we identify I292V as the prevalent mutation in PB2 of circulating avian H9N2 and pandemic H1N1 viruses. The same dominant PB2 mutation is also found in most human isolates of emergent avian H7N9 and H10N8 viruses. In human cells, PB2-292V in H9N2 virus has the combined ability of conferring higher viral polymerase activity and stronger attenuation of IFN-ß induction than that of its predecessor PB2-292I. IFN-ß attenuation is accompanied by higher binding affinity of PB2-292V for host mitochondrial antiviral signalling protein, an important intermediary protein in the induction of IFN-ß. In the mouse in vivo model, PB2-292V mutation increases H9N2 virus replication with ensuing increase in disease severity. Collectively, PB2-292V is a new mammalian adaptive marker that promotes H9N2 virus replication in mammalian hosts with the potential to improve transmission from birds to humans.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Influenza A Virus, H9N2 Subtype/physiology , Influenza in Birds/virology , Interferon-beta/metabolism , Viral Proteins/metabolism , Adaptation, Physiological/genetics , Animals , Chickens , DNA-Directed DNA Polymerase/genetics , Female , Gene Expression Regulation, Enzymologic , HEK293 Cells , Humans , Influenza A Virus, H1N1 Subtype , Influenza, Human/virology , Interferon-beta/genetics , Mice , Mice, Inbred BALB C , Mutation , Species Specificity , Viral Proteins/genetics
8.
J Med Virol ; 91(4): 711-716, 2019 04.
Article in English | MEDLINE | ID: mdl-30560545

ABSTRACT

To understand the evolution and molecular characteristics of Jiangxi H9N2 viruses, we isolated 17 viruses in 2011 and analyzed their characteristics. Phylogenetic analyses revealed that their hemagglutinin genes originate from JS/1/00-like sublineage, neuraminidase genes originate from BJ/94-like sublineage, PB1, PA, NP, and NS genes all come from SH/F/98-like sublineage, PB2 genes originate from ST/163/04-like sublineage, while M genes come from G1-like sublineage. Genotype analysis showed that our isolates were classified as genotype 57. Molecular analyses indicated that our strains contained specific sites characteristic of low-pathogenic viruses. The current study once again highlights the necessity for continued surveillance of novel H9N2 viruses.


Subject(s)
Evolution, Molecular , Genotype , Influenza A Virus, H9N2 Subtype/classification , Influenza A Virus, H9N2 Subtype/isolation & purification , Influenza in Birds/virology , Animals , China , Influenza A Virus, H9N2 Subtype/genetics , Phylogeny , Poultry , Viral Proteins/genetics
9.
Br Poult Sci ; 60(5): 493-498, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31116018

ABSTRACT

1. Infectious bursal disease virus (IBDV) causes immunosuppression in chickens, increasing their susceptibility to other infectious diseases and resulting in vaccination failure. Here, we investigated the immune-depressing effect of IBDV on H9N2 avian influenza viral infection in the broiler chickens. 2. For this purpose, chickens were divided into four groups. In group A, chickens were inoculated with IBDV at 21 days of age and H9N2 avian influenza virus (AIV) 5 days later. Groups B and C only received AIV at 26 days of age and IBDV at 21 days, respectively. The control group (D) were inoculated with normal saline at the same times. Tissue samples from different organs were collected on the days 1, 3, 6, 9, and 12 after H9N2 infection. 3. Macroscopic observation showed IBD lesions in groups A and C, including swollen bursa with the presence of gelatinous exudates, haemorrhages in the thigh muscle, edema, and nephritis. 4. Reverse Transcription-PCR was used to study H9N2 AIV dissemination, and qRT-PCR to determine viral genome copy number in different organs. A considerable titre of AIV was found in the trachea, lungs, cecal tonsils, spleens, and feces of infected chickens. The genome copy number of the virus in the trachea and lungs of group A was significantly higher than that in group B on the first day after inoculation. But in the other days post inoculation, RT-PCR did not detect the AIV genome in group A. Although there might have been some immunosuppression in group A, IBDV could interfere with AIV replication in the chickens of this group. 5. In conclusion, we propose that pre-exposure to IBDV at 3 weeks of age reduces the replication and shedding of H9N2 in broiler chicken.


Subject(s)
Birnaviridae Infections/veterinary , Chickens , Infectious bursal disease virus/physiology , Influenza A Virus, H9N2 Subtype/physiology , Influenza in Birds/virology , Poultry Diseases , Virus Shedding/physiology , Animals , Birnaviridae Infections/virology , Coinfection/veterinary , Coinfection/virology , Random Allocation
10.
BMC Vet Res ; 14(1): 71, 2018 Mar 06.
Article in English | MEDLINE | ID: mdl-29510698

ABSTRACT

BACKGROUND: Vaccines constitute a unique selective pressure, different from natural selection, drives the evolution of influenza virus. In this study, A/Chicken/Shanghai/F/1998 (H9N2) was continually passaged in specific pathogen-free embryonated chicken eggs with or without selective pressures from antibodies induced by homologous maternal antibodies. Genetic mutations, antigenic drift, replication, and pathogenicity of the passaged virus were evaluated. RESULTS: Antigenic drift of the passaged viruses occurred in the 47th generation (vF47) under selective pressure on antibodies and in the 52nd generation (nF52) without selective pressure from antibodies. Seven mutations were observed in the vF47 virus, with three in PB2 and four in HA, whereas 12 mutations occurred in the nF52 virus, with three in PB2, two in PB1, four in HA, one in NP, one in NA, and one in NS. Remarkably, the sequences of the HA segment from vF47 were 100% homologous with those of the nF52 virus. Both the vF47 and nF52 viruses showed enhanced replication compared to the parental virus F/98, but higher levels of replication and pathogenicity were displayed by nF52 than by vF47. An inactive vaccine derived from the parental virus F/98 did not confer protection against challenges by either the vF47 or nF52 virus, but inactive vaccines derived from the vF47 or nF52 virus were able to provide protection against a challenge using F/98. CONCLUSION: Taken together, the passage of H9N2 viruses with or without selective pressure of the antibodies induced by homologous maternal antibodies showed genetic variation, enhanced replication, and variant antigenicity. Selective pressure of the antibody does not seem to play a key role in antigenic drift in the egg model but may impact the genetic variation and replication ability of H9N2 viruses. These results improve understanding of the evolution of the H9N2 influenza virus and may aid in selecting appropriate vaccine seeds.


Subject(s)
Chick Embryo/virology , Evolution, Molecular , Influenza A Virus, H9N2 Subtype/metabolism , Influenza Vaccines/immunology , Influenza in Birds/virology , Animals , Antibodies, Viral/immunology , Chickens/virology , Influenza A Virus, H9N2 Subtype/immunology , Influenza in Birds/immunology , Selection, Genetic , Serial Passage/veterinary
11.
Br Poult Sci ; 59(2): 160-165, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29148834

ABSTRACT

1. The aim of this study was to determine the most likely time interval after infection with influenza virus H9N2 for co-infection with Escherichia coli to cause colibacillosis, the importance of lung load of E. coli and the involvement of respiratory phagocytes. 2. Specific pathogen free chickens were inoculated intranasally with 106EID50 of influenza virus or uninfected. After specified time intervals, 107 CFU E. coli or phosphate-buffered saline was inoculated. The presence of lesions, the number of respiratory phagocytes in the respiratory lavage fluid and the E. coli load in the lung were determined after different time intervals. 3. Compared with the number of lesions in chickens receiving only E. coli inoculation, the number lesions in co-infected chickens were increased at 0- and 3-d time intervals, but reduced in the groups at 6- and 9-d intervals between co-infection. 4. At 1-3 d after E. coli inoculation, the number of lesions chickens was correlated with the number of respiratory phagocytes harvested and related to the E. coli load in the lungs at 5 d. 5. These results suggest that the lesions caused by E. coli in chickens were increased within a 0-3 d interval following H9N2 virus inoculation and that this effect is related to the number of respiratory phagocytes.


Subject(s)
Chickens , Coinfection/veterinary , Escherichia coli Infections/veterinary , Influenza in Birds/pathology , Poultry Diseases/pathology , Animals , Bacterial Load/veterinary , Coinfection/microbiology , Coinfection/pathology , Coinfection/virology , Escherichia coli/physiology , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli Infections/virology , Influenza A Virus, H9N2 Subtype/physiology , Influenza in Birds/microbiology , Influenza in Birds/virology , Lung/microbiology , Lung/pathology , Lung/virology , Phagocytes/immunology , Poultry Diseases/microbiology , Poultry Diseases/virology , Random Allocation , Time Factors
12.
Virol J ; 14(1): 213, 2017 11 03.
Article in English | MEDLINE | ID: mdl-29100522

ABSTRACT

BACKGROUND: Endothelial cells are believed to play an important role in response to virus infection. Our previous microarray analysis showed that H9N2 virus infection and inactivated viral particle inoculation increased the expression of interferon-inducible transmembrane protein 1 (IFITM1) in human umbilical vein endothelial cells (HUVECs). In present study, we deeply investigated the expression patterns of IFITM1 and IFITM1-mediated antiviral response induced by H9N2 virus infection and inactivated viral particle inoculation in HUVECs. Epithelial cells that are considered target cells of the influenza virus were selected as a reference control. METHODS: First, we quantified the expression levels of IFITM1 in HUVECs induced by H9N2 virus infection or viral particle inoculation using quantitative real-time PCR and western blot. Second, we observed whether hemagglutinin or neuraminidase affected IFITM1 expression in HUVECs. Finally, we investigated the effect of induced-IFITM1 on the antiviral state in HUVECs by siRNA and activation plasmid transfection. RESULTS: Both H9N2 virus infection and viral particle inoculation increased the expression of IFITM1 without elevating the levels of interferon-ɑ/ß in HUVECs. HA or NA protein binding alone is not sufficient to increase the levels of IFITM1 and interferon-ɑ/ß in HUVECs. IFITM1 induced by viral particle inoculation significantly decreased the virus titers in culture supernatants of HUVECs. CONCLUSIONS: Our results showed that inactivated viral particle inoculation increased the expression of IFITM1 at mRNA and protein levels. Moreover, the induction of IFITM1 expression mediated the antiviral state in HUVECs.


Subject(s)
Antigens, Differentiation/metabolism , Antiviral Agents/metabolism , Human Umbilical Vein Endothelial Cells/virology , Influenza A Virus, H9N2 Subtype/immunology , Virion/immunology , Antigens, Differentiation/genetics , Cell Line , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/immunology , Influenza A Virus, H9N2 Subtype/genetics , Interferon-alpha/metabolism , Interferon-beta/metabolism , RNA, Small Interfering/genetics , Virion/genetics , Virus Inactivation
13.
Avian Pathol ; 45(6): 630-639, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27215697

ABSTRACT

SD/818 and SD/196 are H9N2 influenza virus strains isolated from chickens from the same farm at different times that exhibited similar genetic evolution. However, strain SD/818 exhibited higher pathogenicity in chickens than strain SD/196 and other H9N2 influenza virus epidemic strains from China. The expression of cytokines is an important host defence mechanism following viral infection and their intensity is a major determinant of viral pathogenicity. To elucidate the mechanism underlying the increased pathogenicity of strain SD/818 from the host's perspective, viral replication and cytokine expression were dynamically studied using real-time quantitative reverse transcription PCR in chickens infected with strain SD/818 compared with chickens infected with strain SD/196 in this study. The results showed that the replication of strain SD/818 and the expressions of IL-1ß, IL-6, TNF-α, IFN-α and IFN-ß induced by strain SD/818 were higher than those induced by strain SD/196 in the chicken host system. Expression of these cytokines in chickens coincided with or followed virus replication. These results suggested that high-level viral replication and pro-inflammatory cytokine expression (but not decreased type I IFN expression) were associated with the higher pathogenicity of strain SD/818 in chickens.


Subject(s)
Chickens/immunology , Cytokines/metabolism , Influenza A Virus, H9N2 Subtype/immunology , Influenza in Birds/immunology , Poultry Diseases/immunology , Animals , Chick Embryo , Chickens/virology , China , Cytokines/genetics , Female , Host-Pathogen Interactions , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza A Virus, H9N2 Subtype/physiology , Influenza in Birds/virology , Poultry Diseases/virology , Species Specificity , Specific Pathogen-Free Organisms , Virus Replication
14.
Pathol Int ; 66(12): 678-686, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27910264

ABSTRACT

H9N2 avian influenza virus causes sporadic human infection. Since humans do not possess acquired immunity specific to this virus, we examined the pathogenicity of an H9N2 virus isolated from a human and then analyzed protective effects of a vaccine in cynomolgus macaques. After intranasal challenge with A/Hong Kong/1073/1999 (H9N2) (HK1073) isolated from a human patient, viruses were isolated from nasal and tracheal swabs in unvaccinated macaques with mild fever and body weight loss. A formalin-inactivated H9N2 whole particle vaccine derived from our virus library was subcutaneously inoculated to macaques. Vaccination induced viral antigen-specific IgG and neutralization activity in sera. After intranasal challenge with H9N2, the virus was detected only the day after inoculation in the vaccinated macaques. Without vaccination, many bronchus-associated lymphoid tissues (BALTs) were formed in the lungs after infection, whereas the numbers of BALTs were smaller and the cytokine responses were weaker in the vaccinated macaques than those in the unvaccinated macaques. These findings indicate that the H9N2 avian influenza virus HK1073 is pathogenic in primates but seems to cause milder symptoms than does H7N9 influenza virus as found in our previous studies and that a formalin-inactivated H9N2 whole particle vaccine induces protective immunity against H9N2 virus.


Subject(s)
Influenza A Virus, H9N2 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Animals , Antibodies, Viral/blood , Bronchi/pathology , Lymphoid Tissue/pathology , Macaca fascicularis , Orthomyxoviridae Infections/virology , Vaccination , Vaccines, Inactivated/immunology
15.
Virology ; 589: 109927, 2024 01.
Article in English | MEDLINE | ID: mdl-37951087

ABSTRACT

The reassortment between avian H9N2 and Eurasian avian-like (EA) H1N1 viruses may have potentially changed from avian-to-mammals adaptation. This study generated 20 reassortant viruses with the introduction of H1N1/2009 internal genes from EA H1N1 virus into H9N2 virus. 12 of these recovered the replication capability both in the lungs and turbinate samples. 10 of 12 obtained PA gene segments from the ribonucleoprotein (RNP) complexes of the EA H1N1 virus, and 3 exhibited extreme virulence. Specially, the combination of PB2, PA and NP genes could overcome the species-specific restriction in human cells. Analysis of the polymerase activities found that introduction of the PA gene resulted in increased polymerase activity. These findings indicated that RNP complexes from EA H1N1 virus could confer an adaptation advantage and high compatibility to avian H9N2 virus. This raises new concerns for public health due to the possible coexistence of H9N2 and EA H1N1 viruses in dogs.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H9N2 Subtype , Influenza, Human , Orthomyxoviridae Infections , Animals , Swine , Dogs , Humans , Mice , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H1N1 Subtype/genetics , Reassortant Viruses/genetics , Virulence/genetics , Birds , Ribonucleoproteins/genetics , Orthomyxoviridae Infections/veterinary , Virus Replication , Mammals
16.
Vet Microbiol ; 285: 109875, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37729705

ABSTRACT

Global poultry production is still severely affected by H9N2 avian influenza virus (AIV), and the development of a novel universal AIV vaccine is still urgently needed. Neuraminidase (NA) has recently been shown to be an efficient conserved protective antigen. In this study, we fused the extracellular region of the NA gene with a ferritin cassette (pYL281), which resulted in self-assembled 24-mer nanoparticles with the NA protein displayed outside the nanoparticles. In addition, a chicken dendritic cell-targeting nanobody-phage74 was also inserted ahead of the NA protein to yield pYL294. Incubation with chicken bone marrow-derived dendritic cells (chBMDCs) showed that the DC-targeting nanoparticles purified from the pYL294 strain significantly increased the maturation of chBMDCs, as shown by increased levels of CCL5, CCR7, CD83 and CD86 compared with nontargeting proteins. Then, a chicken study was performed using Salmonella oral administration together with intranasal boost with purified proteins. Compared with the other groups, oral immunization with Salmonella harboring pYL294 followed by intranasal boost with purified DC-targeting nanoparticles dramatically increased the humoral IgY and mucosal IgA antibody response, as well as increased the cellular immune response, as shown by elevated splenic lymphocyte proliferation and intracellular mRNA levels of IL-4 and IFN-γ. Finally, sequential immunization with DC-targeting nanoparticles showed increased protection against G57 subtype H9N2 virus challenge compared with other groups, as shown by significantly decreased virus RNA copy numbers in oropharyngeal washes (Days 3, 5 and 7 post challenge) and cloacal washes (Day 7), significantly decreased lung virus titers on Day 5 post challenge and increased body weight gains during the challenge.


Subject(s)
Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Influenza, Human , Single-Domain Antibodies , Animals , Humans , Influenza A Virus, H9N2 Subtype/genetics , Chickens , Immunization/veterinary , Influenza in Birds/prevention & control , Dendritic Cells
17.
Microbiome ; 11(1): 163, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37496083

ABSTRACT

BACKGROUND: Antibiotics are widely used for prophylactic therapy and for improving the growth performance of chicken. The problem of bacterial drug resistance caused by antibiotic abuse has previously attracted extensive attention; however, the influence of early-day use of prophylactic antibiotics on the gut microflora and on the disease resistance ability in chicks has not been explored. Here, we comprehensively evaluate the growth performance, gut microbial dynamics, level of antibiotic resistance genes (ARGs) in the gut microbial community, and resistance to H9N2 avian influenza virus (AIV) in chickens following long-term and short-term early-day prophylactic antibiotic treatment. RESULTS: Unexpectedly, long-term prophylactic enrofloxacin treatment slowed the growth rate of chickens, whereas short-term antibiotics treatments were found to increase the growth rate, but these changes were not statistically significant. Strikingly, expansions of Escherichia-Shigella populations were observed in early-life prophylactic antibiotics-treated groups of chickens, which is in contrast to the general perception that antibiotics should control their pathogenicity in chicks. The gut microbiota composition of chickens treated long term with antibiotics or received early-day antibiotics treatment tend to be more dramatically disturbed compared to the gut microbiome of chickens treated with antibiotics for a short term at a later date, especially after H9N2 AIV infection. CONCLUSIONS: Our data provide evidence that early-day and long-term antibiotic treatments have a more adverse effect on the intestinal microbiome of chickens, compared to short-term late age antibiotic treatment. Furthermore, our metagenomic data reveal that both long-term and short-term antibiotic treatment increase the relative abundance of ARGs. Our findings highlight the adverse effects of prophylactic antibiotic treatment and provide a theoretical basis for the cautious administration of antibiotics in food-producing animal management. Video Abstract.


Subject(s)
Gastrointestinal Microbiome , Influenza A Virus, H9N2 Subtype , Microbiota , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Gastrointestinal Microbiome/genetics , Influenza A Virus, H9N2 Subtype/genetics , Chickens/microbiology
18.
Viral Immunol ; 35(5): 365-374, 2022 06.
Article in English | MEDLINE | ID: mdl-35527642

ABSTRACT

The effectiveness of inactivated H9N2 influenza vaccines is doubtful due to changes in antigenic regions of the virus hemagglutinin (HA) protein. One strategy for the development of the efficacious vaccine is the use of nanoparticles that display more immunogenic regions of the influenza virus. In this study, chitosan (CS)-based nanoparticles were developed as a delivery system for intranasal immunization using recombinant H9N2 virus HA1 and nucleoprotein (NP), for the induction of humoral and cellular responses. CS-HA1 and CS-NP nanoparticles were prepared by the ionic gelation method and characterized for their physicochemical properties and shape. The immunogenicity and the protective efficacy were evaluated by measuring antibody titers, T cell proliferation response, CD4+/CD8+ ratio, and quantitative real-time RT-PCR following intranasal administration of the prepared nanoparticles alone or in combination in chickens compared to an inactivated H9N2 vaccine. The average size, surface charge, and spherical structure of the synthesized nanoparticles showed high quality. Serologic analysis revealed that the immunization of inactivated vaccine groups resulted in strong influenza antibodies, which were significantly (p < 0.05) higher compared to the other groups. The vaccinated chickens with CS-HA1+CS-NP developed higher specific anti-influenza antibodies than in those vaccinated with each of rHA1 and rNP. Administration of a combination of the protein-based nanoparticles has stimulated the activation of both CD4+ and CD8+T cells and induced a significantly higher T cell proliferation. The viral shedding was significantly lower in CS-HA1+CS-NP and inactivated vaccine groups compared with other challenged groups. The data demonstrate the potential of CS-HA1+CS-NP nanoparticles for eliciting specific influenza antibodies and conferring protection in chickens.


Subject(s)
Chitosan , Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Influenza, Human , Nanoparticles , Animals , Antibodies, Viral , Chickens , Humans , Immunity , Nanoparticles/chemistry , Nucleoproteins , Vaccines, Inactivated
19.
Transbound Emerg Dis ; 69(2): 669-684, 2022 Mar.
Article in English | MEDLINE | ID: mdl-33566453

ABSTRACT

H9N2 influenza virus has been reported worldwide for several decades, and it has evolved into multiple genotypes among domestic poultry. However, the study involving ecology and evolution of low pathogenic avian influenza virus H9N2 in wild birds in China is limited. Here, we carried out surveillance of avian influenza virus H9N2 in wild birds along with the East Asian-Australian migratory flyway in China in 2017. To estimate the prevalence of H9N2 avian virus in wild birds, information on exposure of wild bird populations to H9N2 viruses using serology, in addition to virology, would greatly improve monitoring capabilities. In this study, we also present serological data of H9N2 among wild birds in China during 2013-2016. We report the identification of poultry-derived H9N2 isolates from asymptomatic infected multispecies wild birds such as Common kestrel (Falco tinnunculus), Northern goshawk (Accipiter gentilis), Little owl (Athene noctua) and Ring-necked Pheasant (Phasianus colchicus) in North China in June 2017. Phylogenetic analysis demonstrated that Tianjin H9N2 isolates belong to the G81 and carry internal genes highly homologous to human H10N8 and H7N9. The isolates could directly infect mice without adaptation but were restricted to replicate in the respiratory system. Glycan-binding preference analyses suggested that the H9N2 isolates have acquired a binding affinity for the human-like receptor. Notably, results from transmission experiment in guinea pigs and ferrets demonstrated the wild birds-derived H9N2 influenza virus exhibits efficient transmission phenotypes in mammalian models via respiratory droplets. Our results indicate that the H9N2 AIVs continued to circulate extensively in wild bird populations and migratory birds play an important role in the spread and genetic diversification of H9N2 AIVs. The pandemic potential of H9N2 viruses demonstrated by aerosol transmission in mammalian models via respiratory droplets highlights the importance of monitoring influenza viruses in these hosts.


Subject(s)
Influenza A Virus, H7N9 Subtype , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Rodent Diseases , Animals , Australia , Birds , China/epidemiology , Ferrets , Guinea Pigs , Humans , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H9N2 Subtype/genetics , Mammals , Mice , Phylogeny , Poultry , Respiratory Aerosols and Droplets
20.
Vet Microbiol ; 246: 108747, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32605760

ABSTRACT

H9N2 avian influenza viruses (AIVs) can cross species barriers and expand from birds tomammals and humans. It usually leads to economic loss for breeding farms and poses a serious threat to human health.This study investigated the molecular characteristics of H9N2 AIV isolated from a racing pigeon and its pathogenesis in BALB/c mice and pigeons. Phylogenetic analysis indicated that the H9N2 virus belonged to the Ck/BJ/94-like lineage, and acquired multiple specific amino acid substitutions that might contribute to viral transmission from birds to mammals and humans. A pathogenesis study showed that both mice and pigeons infected with H9N2 virus showed clinical signs and mortality. The H9N2 viruses efficiently replicated in mice and pigeons. In our study, high levels of viral shedding were detected in pigeons, but the infection was not transmitted to co-housed pigeons. Histopathological examination revealed the presence of inflammatory responses in the infected mice and pigeons. Immunohistochemical analysis showed the presence of H9N2 virus in multiple organs of the infected mice and pigeons. Moreover, the infected mice and pigeons demonstrated significant cytokine/chemokine production. Our results showed that the H9N2 virus can infect mice and pigeons, and can not be transmitted between pigeons through direct contact.


Subject(s)
Columbidae/virology , Genome, Viral , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza in Birds/virology , Amino Acid Substitution , Animals , Chemokines/immunology , Cytokines/immunology , Female , Influenza A Virus, H9N2 Subtype/isolation & purification , Influenza in Birds/transmission , Mice , Mice, Inbred BALB C , Mutation , Phylogeny , Specific Pathogen-Free Organisms , Virus Replication , Virus Shedding
SELECTION OF CITATIONS
SEARCH DETAIL