Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
J Microbiol Biotechnol ; 34(3): 735-745, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-37915251

ABSTRACT

Avian influenza is a serious threat to both public health and the poultry industry worldwide. This respiratory virus can be combated by eliciting robust immune responses at the site of infection through mucosal immunization. Recombinant probiotics, specifically lactic acid bacteria, are safe and effective carriers for mucosal vaccines. In this study, we engineered recombinant fusion protein by fusing the hemagglutinin 1 (HA1) subunit of the A/Aquatic bird/Korea/W81/2005 (H5N2) with the Bacillus subtilis poly γ-glutamic acid synthetase A (pgsA) at the surface of Lactobacillus casei (pgsA-HA1/L. casei). Using subcellular fractionation and flow cytometry we confirmed the surface localization of this fusion protein. Mucosal administration of pgsA-HA1/L. casei in mice resulted in significant levels of HA1-specific serum IgG, mucosal IgA and neutralizing antibodies against the H5N2 virus. Additionally, pgsA-HA1/L. casei-induced systemic and local cell-mediated immune responses specific to HA1, as evidenced by an increased number of IFN-γ and IL-4 secreting cells in the spleens and higher levels of IL-4 in the local lymphocyte supernatants. Finally, mice inoculated with pgsA-HA1/L. casei were protected against a 10LD50 dose of the homologous mouse-adapted H5N2 virus. These results suggest that mucosal immunization with L. casei displaying HA1 on its surface could be a potential strategy for developing a mucosal vaccine against other H5 subtype viruses.


Subject(s)
Influenza A Virus, H5N2 Subtype , Influenza A virus , Influenza Vaccines , Lacticaseibacillus casei , Animals , Mice , Lacticaseibacillus casei/genetics , Interleukin-4 , Administration, Mucosal , Immunity , Administration, Oral
2.
Molecules ; 26(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34070099

ABSTRACT

Wild ginseng has better pharmacological effects than cultivated ginseng. However, its industrialization is limited by the inability to grow wild ginseng on a large scale. Herein, we demonstrate how to optimize ginseng production through cultivation, and how to enhance the concentrations of specific ginsenosides through fermentation. In the study, we also evaluated the ability of fermented cultured wild ginseng root extract (HLJG0701-ß) to inhibit acetylcholinesterase (AChE), as well as its neuroprotective effects and antioxidant activity. In invitro tests, HLJG0701-ß inhibited AChE activity and exerted neuroprotective and antioxidant effects (showing increased catalyst activity but decreased reactive oxygen species concentration). In invivo tests, after HLJG0701-ß was orally administered at doses of 0, 125, 250, and 500 mg/kg in an animal model of memory impairment, behavioral evaluation (Morris water maze test and Y-maze task test) was performed. The levels of AChE, acetylcholine (ACh), blood catalase (CAT), and malondialdehyde (MDA) in brain tissues were measured. The results showed that HLJG0701-ß produced the best results at a dose of 250 mg/kg or more. The neuroprotective mechanism of HLJG0701-ß was determined to involve the inhibition of AChE activity and a decrease in oxidative stress. In summary, both invitro and invivo tests confirmed that HJG0701-ß administration can lead to memory improvement.


Subject(s)
Antioxidants/pharmacology , Fermentation , Neuroprotective Agents/pharmacology , Panax/chemistry , Plant Extracts/pharmacology , Plant Roots/chemistry , Acetylcholine/metabolism , Acetylcholinesterase/metabolism , Animals , Brain/metabolism , Catalase/blood , Catalase/metabolism , Cholinesterase Inhibitors/pharmacology , Disease Models, Animal , Female , Galactose , Ginsenosides/pharmacology , Male , Malondialdehyde/blood , Mice , Morris Water Maze Test , Ovariectomy , Reactive Oxygen Species/metabolism , Scopolamine
3.
PLoS Negl Trop Dis ; 15(3): e0009170, 2021 03.
Article in English | MEDLINE | ID: mdl-33651791

ABSTRACT

In Korea, typhoid fever is a rare disease due to improved living standards. However, typhoid fever remains a major burden in developing countries and regions, such as India and Southeast Asia. In this study, we isolated Salmonella Typhi (S. Typhi) from eight patients with typhoid fever who were travelers returning from India. The strains isolated were characterized by antimicrobial susceptibility profiling and whole-genome sequencing (WGS) analysis. All strains were resistant to nalidixic acid and azithromycin. Among them, four isolates were highly resistant to ciprofloxacin (MIC ≥32 µg/ml); these strains have not been confirmed in Korea PulseNet DB. According to WGS, the ciprofloxacin-resistant strains belong to the global dominant multidrug-resistant (MDR) haplotype H58 (SNP glpA C1047T, SptP protein Q185* (premature stop codon)) and do not harbor the MDR plasmid. H58-associated SNPs in membrane and metabolism genes, including yhdA, yajI, hyaE, tryE, rlpB and metH, are present. Additionally, phylogenetic analysis assigned the H58 strains to sublineage II, whereas the non-H58 strains are closely related to haplotype H50. The presence of high-level ciprofloxacin-resistant S. Typhi haplotype H58 in Korea was first confirmed as due to influx from overseas via travelers. This study provides information about intercontinental drug-resistant transmission between countries and suggests that travelers need to be careful about personal hygiene.


Subject(s)
Drug Resistance, Multiple, Bacterial , Salmonella typhi/classification , Salmonella typhi/drug effects , Typhoid Fever/microbiology , Anti-Bacterial Agents/pharmacology , Ciprofloxacin/pharmacology , Haplotypes , Humans , India , Microbial Sensitivity Tests , Republic of Korea/epidemiology , Salmonella typhi/genetics , Salmonella typhi/isolation & purification , Travel-Related Illness , Typhoid Fever/epidemiology
4.
EMBO J ; 39(21): e105139, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32935379

ABSTRACT

NF-κB essential modulator (NEMO) is a key regulatory protein that functions during NF-κB- and interferon-mediated signaling in response to extracellular stimuli and pathogen infections. Tight regulation of NEMO is essential for host innate immune responses and for maintenance of homeostasis. Here, we report that the E3 ligase MARCH2 is a novel negative regulator of NEMO-mediated signaling upon bacterial or viral infection. MARCH2 interacted directly with NEMO during the late phase of infection and catalyzed K-48-linked ubiquitination of Lys326 on NEMO, which resulted in its degradation. Deletion of MARCH2 resulted in marked resistance to bacterial/viral infection, along with increased innate immune responses both in vitro and in vivo. In addition, MARCH2-/- mice were more susceptible to LPS challenge due to massive production of cytokines. Taken together, these findings provide new insight into the molecular regulation of NEMO and suggest an important role for MARCH2 in homeostatic control of innate immune responses.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism , Animals , Cell Line , Female , Gene Deletion , Humans , Immunity, Innate/physiology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , NF-kappa B/metabolism , Signal Transduction/genetics , Transcriptome , Ubiquitin-Protein Ligases/genetics , Ubiquitination
5.
J Clin Med ; 9(7)2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32708872

ABSTRACT

OBJECTIVES: To investigate antibody production in asymptomatic and mild COVID-19 patients. METHODS: Sera from asymptomatic to severe COVID-19 patients were collected. Microneutralization (MN), fluorescence immunoassay (FIA), and enzyme-linked immunosorbent assay (ELISA) were performed. RESULTS: A total of 70 laboratory-confirmed COVID-19 patients were evaluated, including 15 asymptomatic/anosmia, 49 mild symptomatic, and 6 pneumonia patients. The production of the neutralizing antibody was observed in 100% of pneumonia, 93.9% of mild symptomatic, and 80.0% of asymptomatic/anosmia groups. All the patients in the pneumonia group showed high MN titer (≥1:80), while 36.7% of mild symptomatic and 20.0% of asymptomatic/anosmia groups showed high titer (p < 0.001). Anti-SARS-CoV-2 antibodies could be more sensitively detected by FIA IgG (98.8%) and ELISA (97.6%) in overall. For the FIA IgG test, all patients in the pneumonia group exhibited a high COI value (≥15.0), while 89.8% of mild symptomatic and 73.3% of asymptomatic/anosmia groups showed a high value (p = 0.049). For the ELISA test, all patients in the pneumonia group showed a high optical density (OD) ratio (≥3.0), while 65.3% of mild symptomatic and 53.3% of asymptomatic/anosmia groups showed a high ratio (p = 0.006). CONCLUSIONS: Most asymptomatic and mild COVID-19 patients produced the neutralizing antibody, although the titers were lower than pneumonia patients. ELISA and FIA sensitively detected anti-SARS-CoV-2 antibodies.

6.
Mol Cells ; 42(10): 721-728, 2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31600868

ABSTRACT

Non-structural protein 1 (NS1) of influenza virus has been shown to inhibit the innate immune response by blocking the induction of interferon (IFN). In this study, we isolated two single-stranded RNA aptamers specific to NS1 with K d values of 1.62 ± 0.30 nM and 1.97 ± 0.27 nM, respectively, using a systematic evolution of ligand by exponential enrichment (SELEX) procedure. The selected aptamers were able to inhibit the interaction of NS1 with tripartite motif-containing protein 25 (TRIM25), and suppression of NS1 enabled retinoic acid inducible gene I (RIG-I) to be ubiquitinated regularly by TRIM25. Additional luciferase reporter assay and quantitative real-time PCR (RT-PCR) experiments demonstrated that suppression of NS1 by the selected aptamers induced IFN production. It is noted that viral replication was also inhibited through IFN induction in the presence of the selected aptamers. These results suggest that the isolated aptamers are strongly expected to be new therapeutic agents against influenza infection.


Subject(s)
Aptamers, Nucleotide/metabolism , DEAD Box Protein 58/metabolism , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Viral Nonstructural Proteins/metabolism , Animals , HEK293 Cells , Humans , Interferons/metabolism , Mice , Protein Binding , RAW 264.7 Cells , Receptors, Immunologic , Virus Replication
7.
PLoS Pathog ; 15(8): e1008004, 2019 08.
Article in English | MEDLINE | ID: mdl-31412082

ABSTRACT

Fas-associated factor 1 is a death-promoting protein that induces apoptosis by interacting with the Fas receptor. Until now, FAF1 was reported to interact potentially with diverse proteins and to function as a negative and/or positive regulator of several cellular possesses. However, the role of FAF1 in defense against bacterial infection remains unclear. Here, we show that FAF1 plays a pivotal role in activating NADPH oxidase in macrophages during Listeria monocytogenes infection. Upon infection by L. monocytogenes, FAF1 interacts with p67phox (an activator of the NADPH oxidase complex), thereby facilitating its stabilization and increasing the activity of NADPH oxidase. Consequently, knockdown or ectopic expression of FAF1 had a marked effect on production of ROS, proinflammatory cytokines, and antibacterial activity, in macrophages upon stimulation of TLR2 or after infection with L. monocytogenes. Consistent with this, FAF1gt/gt mice, which are knocked down in FAF1, showed weaker inflammatory responses than wild-type mice; these weaker responses led to increased replication of L. monocytogenes. Collectively, these findings suggest that FAF1 positively regulates NADPH oxidase-mediated ROS production and antibacterial defenses.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis Regulatory Proteins/physiology , Immunity, Innate/immunology , Inflammation/immunology , Listeriosis/immunology , Macrophages/immunology , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Animals , Cytokines/metabolism , Inflammation/metabolism , Inflammation/microbiology , Listeria monocytogenes/immunology , Listeriosis/metabolism , Listeriosis/microbiology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , NADPH Oxidases/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Signal Transduction
8.
Cell Rep ; 27(7): 2105-2118.e5, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31091449

ABSTRACT

Small heterodimer partner (SHP) is an orphan nuclear receptor that acts as a transcriptional co-repressor by interacting with nuclear receptors and transcription factors. Although SHP plays a negative regulatory function in various signaling pathways, its role in virus infection has not been studied. Here, we report that SHP is a potent negative regulator of the virus-mediated type I IFN signaling that maintains homeostasis within the antiviral innate immune system. Upon virus infection, SHP interacts specifically with CREB-binding protein (CBP) in the nucleus, thereby obstructing CBP/ß-catenin interaction competitively. Consequently, SHP-deficient cells enhance antiviral responses, including transcription of the type I IFN gene, upon virus infection. Furthermore, SHP-deficient mice show higher levels of IFN production and are more resistant to influenza A virus infection. Our results suggest that SHP is a nuclear regulator that blocks transcription of the type I IFN gene to inhibit excessive innate immune responses.


Subject(s)
Cell Nucleus/immunology , Immunity, Innate , Membrane Proteins/immunology , Phosphoproteins/immunology , Receptors, Cytoplasmic and Nuclear/immunology , Virus Diseases/immunology , Animals , Cell Nucleus/genetics , Cell Nucleus/virology , Membrane Proteins/genetics , Mice , Mice, Knockout , Phosphoproteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Virus Diseases/genetics
9.
Nutrients ; 11(5)2019 May 20.
Article in English | MEDLINE | ID: mdl-31137483

ABSTRACT

Wild ginseng is known to contain additional physiologically and pharmacologically active substances than common ginseng. The utilization of this herb can be maximized by altering its composition via tissue culture generating adventitious roots. We enriched the content of specific ginsenosides and investigated their role in ameliorating memory impairment. Cultured wild ginseng root was subjected to extraction, steaming, and fermentation using Pediococcus pentosaceus HLJG0702 to enhance the levels of ginsenosides Rg5 /Rk1. The analysis of product, HLJG0701, confirmed target ginsenosides. We analyzed the inhibitory effect of ginsenoside Rg5/Rk1, HLJG0701 and the raw material on acetylcholinesterase. Further, we performed Morris water maze, Y-maze, and passive avoidance tasks with mice exhibiting memory deficit induced by scopolamine, and we analyzed the concentrations of acetylcholinesterase and acetylcholine in their brains. Studies showed that the levels of ginsenosides Rg5 /Rk1, not found in the raw material, were enhanced in HLJG0701. Ginsenosides and HLJG0701 significantly inhibited acetylcholinesterase unlike the raw material. In all behavioral tasks, HLJG0701 showed memory improvement. It reduced acetylcholinesterase, whereas, it preserved acetylcholine in brain. In conclusion, cultured wild ginseng root extract fermented by P. pentosaceus HLJG0702 contains the distinctive ginsenosides Rg5/Rk1, which may ameliorate memory impairment via inhibition of acetylcholinesterase resulting in increased acetylcholine levels in the brain.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Cholinesterase Inhibitors/pharmacology , Ginsenosides/pharmacology , Memory Disorders/prevention & control , Memory/drug effects , Panax/metabolism , Pediococcus pentosaceus/metabolism , Plant Extracts/pharmacology , Acetylcholinesterase/metabolism , Animals , Avoidance Learning/drug effects , Brain/enzymology , Brain/physiopathology , Cholinesterase Inhibitors/isolation & purification , Disease Models, Animal , Fermentation , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Ginsenosides/isolation & purification , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Memory Disorders/physiopathology , Memory Disorders/psychology , Mice, Inbred C57BL , Panax/microbiology , Plant Extracts/isolation & purification , Plant Roots/metabolism , Plant Roots/microbiology , Scopolamine
10.
J Virol ; 93(2)2019 01 15.
Article in English | MEDLINE | ID: mdl-30355684

ABSTRACT

Tryptophanyl-tRNA synthetase (WRS) is one of the aminoacyl-tRNA synthetases (ARSs) that possesses noncanonical functions. Full-length WRS is released during bacterial infection and primes the Toll-like receptor 4 (TLR4)-myeloid differentiation factor 2 (MD2) complex to elicit innate immune responses. However, the role of WRS in viral infection remains unknown. Here, we show that full-length WRS is secreted by immune cells in the early phase of viral infection and functions as an antiviral cytokine. Treatment of cells with recombinant WRS protein promotes the production of inflammatory cytokines and type I interferons (IFNs) and curtails virus replication in THP-1 and Raw264.7 cells but not in TLR4-/- or MD2-/- bone marrow-derived macrophages (BMDMs). Intravenous and intranasal administration of recombinant WRS protein induces an innate immune response and blocks viral replication in vivo These findings suggest that secreted full-length WRS has a noncanonical role in inducing innate immune responses to viral infection as well as to bacterial infection.IMPORTANCE ARSs are essential enzymes in translation that link specific amino acids to their cognate tRNAs. In higher eukaryotes, some ARSs possess additional, noncanonical functions in the regulation of cell metabolism. Here, we report a novel noncanonical function of WRS in antiviral defense. WRS is rapidly secreted in response to viral infection and primes the innate immune response by inducing the secretion of proinflammatory cytokines and type I IFNs, resulting in the inhibition of virus replication both in vitro and in vivo Thus, we consider WRS to be a member of the antiviral innate immune response. The results of this study enhance our understanding of host defense systems and provide additional information on the noncanonical functions of ARSs.


Subject(s)
Rhabdoviridae Infections/immunology , Tryptophan-tRNA Ligase/genetics , Tryptophan-tRNA Ligase/metabolism , Vesiculovirus/pathogenicity , Administration, Intranasal , Administration, Intravenous , Animals , Cell Line , Cytokines/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate , Interferon Type I/metabolism , Mice , RAW 264.7 Cells , Rhabdoviridae Infections/genetics , THP-1 Cells , Tryptophan-tRNA Ligase/administration & dosage , Vesiculovirus/immunology
11.
Vaccine ; 37(3): 484-493, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30502069

ABSTRACT

Because H5N1 influenza viruses continuously threaten the public health, the WHO has prepared various clades of H5N1 mock-up vaccines as one of the measures for pandemic preparedness. The recent worldwide outbreak of H5Nx virus which belongs to clade 2.3.4.4 and of which H5N6 subtype belongs and already caused human infection also increases the need of pandemic vaccine for such novel emerging viruses. In this study, we evaluated the protective efficacy and immunogenicity of an egg-based and inactivated whole-virus H5N8 (IDCDC-RG43A) developed by CDC containing HA and NA gene of the parent virus A/gyrfalcon/Washington/41088-6/2014. Mice vaccinated two times elicited low to moderate antibody titer in varying amount of antigen doses against the homologous H5N8 vaccine virus and heterologous intra-clade 2.3.4.4 H5N6 (A/Sichuan/26221/2014) virus. Mice immunized with at least 3.0 µg/dose of IDCDC-RG43A with aluminum hydroxide adjuvant were completely protected from lethal challenge with the mouse-adapted H5N8 (A/Environment/Korea/ma468/2015, maH5N8) as well as cleared the viral replication in tissues including lung, brain, spleen, and kidney. Vaccinated ferrets induced high antibody titers against clade 2.3.4.4 H5N8/H5N6 viruses and the antibody showed high cross-reactivity to clade 2.2 H5N1 but not to clade 1 and 2.3.4 viruses as measured by hemagglutinin inhibition and serum neutralization assays. Furthermore, administration of the vaccine in ferrets resulted in attenuation of clinical disease signs and virus spread to peripheral organs including lung, spleen, and kidney from high dose challenge with maH5N8 virus. The protective and immunogenic characteristic of the candidate vaccine are essential attributes to be considered for further clinical trials as a pre-pandemic vaccine for a potential pandemic virus.


Subject(s)
Antibodies, Neutralizing/blood , Immunogenicity, Vaccine , Influenza A Virus, H5N8 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Pandemics/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/blood , Cross Protection , Disease Models, Animal , Drug Evaluation, Preclinical , Ferrets , Influenza Vaccines/administration & dosage , Mice
14.
Arch Virol ; 163(8): 2073-2083, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29619599

ABSTRACT

Enterovirus 71 (EV71) is the major causative agent of hand-foot-and-mouth disease (HFMD) and many neurological manifestations. Recently, this virus has become a serious concern because of consecutive epidemics in the Asia-Pacific region. However, no effective vaccine for EV71 has been discovered except two EV71 vaccines which are being used in local communities of China. To develop a safe and efficient EV71 vaccine candidate, we generated inactivated EV71 and evaluated its efficacy with γ-PGA/Chitosan nanoparticles (PC NPs), which are safe, biodegradable and effective as an adjuvant. The subcutaneous administration of inactivated EV71 with PC NPs adjuvant induces higher levels of virus-specific humoral (IgG, IgG1, and IgG2a) and cell-mediated immune responses (IFN-γ and IL-4). Additionally, inactivated EV71 with PC NPs adjuvant induces significantly higher virus neutralizing antibody responses compared to the virus only group, and resulted in a long lasting immunity without any noticeable side effects. Together, our findings demonstrate that PC NPs are safe and effective immunogenic adjuvants which may be promising candidates in the development of more efficacious EV71 vaccines.


Subject(s)
Enterovirus A, Human/immunology , Hand, Foot and Mouth Disease/prevention & control , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neutralizing , Antibodies, Viral/immunology , Chitosan/administration & dosage , Chitosan/analogs & derivatives , Chitosan/immunology , Enterovirus A, Human/genetics , Female , Hand, Foot and Mouth Disease/immunology , Hand, Foot and Mouth Disease/virology , Humans , Immunity, Humoral , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Polyglutamic Acid/administration & dosage , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
15.
Emerg Microbes Infect ; 7(1): 29, 2018 Mar 14.
Article in English | MEDLINE | ID: mdl-29535296

ABSTRACT

Highly pathogenic avian influenza (HPAI) A(H5N6) and A(H5N8) virus infections resulted in the culling of more than 37 million poultry in the Republic of Korea during the 2016/17 winter season. Here we characterize two representative viruses, A/Environment/Korea/W541/2016 [Em/W541(H5N6)] and A/Common Teal/Korea/W555/2017 [CT/W555(H5N8)], and evaluate their zoonotic potential in various animal models. Both Em/W541(H5N6) and CT /W555(H5N8) are novel reassortants derived from various gene pools of wild bird viruses present in migratory waterfowl arising from eastern China. Despite strong preferential binding to avian virus-type receptors, the viruses were able to grow in human respiratory tract tissues. Em/W541(H5N6) was found to be highly pathogenic in both chickens and ducks, while CT/W555(H5N8) caused lethal infections in chickens but did not induce remarkable clinical illness in ducks. In mice, both viruses appeared to be moderately pathogenic and displayed limited tissue tropism relative to HPAI H5N1 viruses. Em/W541(H5N6) replicated to moderate levels in the upper respiratory tract of ferrets and was detected in the lungs, brain, spleen, liver, and colon. Unexpectedly, two of three ferrets in direct contact with Em/W541(H5N6)-infected animals shed virus and seroconverted at 14 dpi. CT/W555(H5N8) was less pathogenic than the H5N6 virus in ferrets and no transmission was detected. Given the co-circulation of different, phenotypically distinct, subtypes of HPAI H5Nx viruses for the first time in South Korea, detailed virologic investigations are imperative given the capacity of these viruses to evolve and cause human infections.


Subject(s)
Influenza A Virus, H5N8 Subtype/pathogenicity , Influenza A virus/pathogenicity , Influenza in Birds/virology , Animal Migration , Animals , Animals, Wild/virology , Chickens , China , Ducks , Ferrets , Influenza A Virus, H5N8 Subtype/genetics , Influenza A Virus, H5N8 Subtype/isolation & purification , Influenza A Virus, H5N8 Subtype/physiology , Influenza A virus/genetics , Influenza A virus/isolation & purification , Influenza A virus/physiology , Influenza in Birds/epidemiology , Influenza in Birds/pathology , Influenza in Birds/physiopathology , Poultry Diseases/epidemiology , Poultry Diseases/pathology , Poultry Diseases/physiopathology , Poultry Diseases/virology , Reassortant Viruses/classification , Reassortant Viruses/genetics , Reassortant Viruses/isolation & purification , Reassortant Viruses/pathogenicity , Republic of Korea/epidemiology , Seasons , Virulence , Virus Replication
16.
Virulence ; 9(1): 133-148, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28873012

ABSTRACT

Recently identified highly pathogenic avian influenza (HPAI) H5N8 viruses (clade 2.3.4.4) are relatively low to moderately pathogenic in mammalian hosts compared with HPAI H5N1 viruses. In this study, we generated reassortant viruses comprised of A/MD/Korea/W452/2014(H5N8) with substitution of individual genes from A/EM/Korea/W149/2006(H5N1) to understand the contribution of each viral gene to virulence in mammals. Substituting the PB2 gene segment or the NA gene segment of the H5N8 virus by that from the H5N1 virus resulted in significantly enhanced pathogenicity compared with the parental H5N8 virus in mice. Of note, substitution of the PB2 gene segment of the H5N8 virus by that from the H5N1 virus resulted in a 1000-fold increase in virulence for mice compared with the parental virus (MLD50 decreased from 105.8 to 102.5 EID50). Further, the W452W149PB2 virus also induced the highest virus titers in lungs at all time points and the highest levels of inflammatory cytokine responses among all viruses tested. This high virulence phenotype was also confirmed by high viral titers in the respiratory tracts of infected ferrets. Further, a mini-genome assay revealed that W452W149PB2 has significantly increased polymerase activity (p < 0.001). Taken together, our study demonstrates that a single gene substitution from other avian influenza viruses can alter the pathogenicity of recent H5N8 viruses, and therefore emphasizes the need for intensive monitoring of reassortment events among co-circulating avian and mammalian viruses.


Subject(s)
Disease Models, Animal , Influenza A virus/pathogenicity , Orthomyxoviridae Infections/pathology , RNA-Dependent RNA Polymerase/genetics , Reassortant Viruses/pathogenicity , Viral Proteins/genetics , Virulence/genetics , Animals , Chemokines/immunology , Coinfection , Cytokines/immunology , Dogs , Ferrets , HEK293 Cells , Humans , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N8 Subtype/genetics , Influenza A Virus, H5N8 Subtype/pathogenicity , Influenza A virus/genetics , Influenza A virus/physiology , Lung/immunology , Lung/pathology , Lung/virology , Madin Darby Canine Kidney Cells , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Reassortant Viruses/genetics , Viral Tropism
17.
J Virol ; 92(1)2018 01 01.
Article in English | MEDLINE | ID: mdl-29046464

ABSTRACT

Several subtypes of avian influenza viruses (AIVs) are emerging as novel human pathogens, and the frequency of related infections has increased in recent years. Although neuraminidase (NA) inhibitors (NAIs) are the only class of antiviral drugs available for therapeutic intervention for AIV-infected patients, studies on NAI resistance among AIVs have been limited, and markers of resistance are poorly understood. Previously, we identified unique NAI resistance substitutions in AIVs of the N3, N7, and N9 NA subtypes. Here, we report profiles of NA substitutions that confer NAI resistance in AIVs of the N4, N5, N6, and N8 NA subtypes using gene-fragmented random mutagenesis. We generated libraries of mutant influenza viruses using reverse genetics (RG) and selected resistant variants in the presence of the NAIs oseltamivir carboxylate and zanamivir in MDCK cells. In addition, two substitutions, H274Y and R292K (N2 numbering), were introduced into each NA gene for comparison. We identified 37 amino acid substitutions within the NA gene, 16 of which (4 in N4, 4 in N5, 4 in N6, and 4 in N8) conferred resistance to NAIs (oseltamivir carboxylate, zanamivir, or peramivir) as determined using a fluorescence-based NA inhibition assay. Substitutions conferring NAI resistance were mainly categorized as either novel NA subtype specific (G/N147V/I, A246V, and I427L) or previously reported in other subtypes (E119A/D/V, Q136K, E276D, R292K, and R371K). Our results demonstrate that each NA subtype possesses unique NAI resistance markers, and knowledge of these substitutions in AIVs is important in facilitating antiviral susceptibility monitoring of NAI resistance in AIVs.IMPORTANCE The frequency of human infections with avian influenza viruses (AIVs) has increased in recent years. Despite the availability of vaccines, neuraminidase inhibitors (NAIs), as the only available class of drugs for AIVs in humans, have been constantly used for treatment, leading to the inevitable emergence of drug-resistant variants. To screen for substitutions conferring NAI resistance in AIVs of N4, N5, N6, and N8 NA subtypes, random mutations within the target gene were generated, and resistant viruses were selected from mutant libraries in the presence of individual drugs. We identified 16 NA substitutions conferring NAI resistance in the tested AIV subtypes; some are novel and subtype specific, and others have been previously reported in other subtypes. Our findings will contribute to an increased and more comprehensive understanding of the mechanisms of NAI-induced inhibition of influenza virus and help lead to the development of drugs that bind to alternative interaction motifs.


Subject(s)
Drug Resistance, Viral/genetics , Influenza in Birds/virology , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Orthomyxoviridae/enzymology , Acids, Carbocyclic , Amino Acid Substitution , Animals , Antiviral Agents/pharmacology , Birds , Cyclopentanes/pharmacology , Dogs , Enzyme Inhibitors , Guanidines/pharmacology , Humans , Influenza in Birds/drug therapy , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mutagenesis , Neuraminidase/chemistry , Neuraminidase/classification , Orthomyxoviridae/drug effects , Orthomyxoviridae/genetics , Oseltamivir/analogs & derivatives , Oseltamivir/pharmacology , Reverse Genetics , Zanamivir/pharmacology
18.
J Clin Virol ; 94: 63-66, 2017 09.
Article in English | MEDLINE | ID: mdl-28759774

ABSTRACT

BACKGROUND: Hepatitis A virus (HAV), a major cause of acute hepatitis, has had the highest occurrence among group 1 nationally notifiable infectious diseases in Korea since 2010.Recently,the annual increase in the HAV infection rate among young adults has become a public health concern. OBJECTIVES: The aim of this study was to describe an outbreak of acute hepatitis in a residential facility in April 2015 and to identify potential sources of this outbreak. STUDY DESIGN: Sera from all exposed residents were tested for anti-HAV IgM or IgG antibodies by ELISA. Clinical (sera and stool) and environmental samples were screened for the presence of HAV RNA using one-step RT-PCR and nested PCR. The VP3-VP1 regions of HAV were analyzed using the BLAST database and MEGA7 software. RESULTS: Of the 82 persons in the facility, 12 (14.6%, including 10 residents and 2 health care workers) were diagnosed with hepatitis A. Clinical symptoms were evident in 9 individuals, one of whom died, and the remaining four patients were asymptomatic. Traceback investigation revealed that HAV-RNA (genotype IA) was detected in the patients' stools and the groundwater used in the facility. CONCLUSIONS: We described an HAV outbreak in a facility for the disabled due to using a water supply that was mixed with contaminated groundwater. Therefore, HAV vaccination and periodic water inspections in group facilities should be emphasized to prevent HAV infection.


Subject(s)
Disease Outbreaks/statistics & numerical data , Hepatitis A virus/genetics , Hepatitis A/epidemiology , Water Microbiology , Adolescent , Adult , Genotype , Hepatitis A/immunology , Hepatitis A/transmission , Hepatitis A Antibodies/blood , Hepatitis A virus/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Phylogeny , Republic of Korea/epidemiology , Water Supply , Young Adult
19.
Sci Rep ; 7(1): 4875, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28687749

ABSTRACT

The antiviral activities of synthesized Kα2-helix peptide, which was derived from the viral FLICE-like inhibitor protein (vFLIP) of Kaposi's sarcoma-associated herpesvirus (KSHV), against influenza A virus (IAV) were investigated in vitro and in vivo, and mechanisms of action were suggested. In addition to the robust autophagy activity of the Kα2-helix peptide, the present study showed that treatment with the Kα2 peptide fused with the TAT peptide significantly inhibited IAV replication and transmission. Moreover, TAT-Kα2 peptide protected the mice, that were challenged with lethal doses of highly pathogenic influenza A H5N1 or H1N1 viruses. Mechanistically, we found that TAT-Kα2 peptide destabilized the viral membranes, depending on their lipid composition of the viral envelop. In addition to IAV, the Kα2 peptide inhibited infections with enveloped viruses, such as Vesicular Stomatitis Virus (VSV) and Respiratory Syncytial Virus (RSV), without cytotoxicity. These results suggest that TAT-Kα2 peptide is a potential antiviral agent for controlling emerging or re-emerging enveloped viruses, particularly diverse subtypes of IAVs.


Subject(s)
Antiviral Agents/metabolism , Influenza A virus/drug effects , Oligopeptides/metabolism , Viral Proteins/metabolism , Virus Replication/drug effects , Animals , Antiviral Agents/isolation & purification , Disease Models, Animal , Dogs , Influenza A virus/physiology , Lung/virology , Madin Darby Canine Kidney Cells , Mice, Inbred BALB C , Oligopeptides/isolation & purification , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/physiology , Survival Analysis , Treatment Outcome , Vesiculovirus/drug effects , Vesiculovirus/physiology , Viral Load , Virus Internalization/drug effects , Virus Release/drug effects
20.
Infect Genet Evol ; 53: 56-67, 2017 09.
Article in English | MEDLINE | ID: mdl-28477974

ABSTRACT

During the outbreaks of highly pathogenic avian influenza (HPAI) H5N6 viruses in 2016 in South Korea, novel H5N8 viruses were also isolated from migratory birds. Phylogenetic analysis revealed that the HA gene of these H5N8 viruses belonged to clade 2.3.4.4, similarly to recent H5Nx viruses, and originated from A/Brk/Korea/Gochang1/14(H5N8), a minor lineage of H5N8 that appeared in 2014 and then disappeared. At least four reassortment events occurred with different subtypes (H5N8, H7N7, H3N8 and H10N7) and a chicken challenge study revealed that they were classified as HPAI viruses according to OIE criteria.


Subject(s)
Disease Outbreaks/veterinary , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H5N8 Subtype/genetics , Influenza in Birds/epidemiology , Phylogeny , Reassortant Viruses/genetics , Animals , Animals, Wild , Birds/virology , Chickens , Genotype , Influenza A Virus, H10N7 Subtype/classification , Influenza A Virus, H10N7 Subtype/genetics , Influenza A Virus, H10N7 Subtype/isolation & purification , Influenza A Virus, H3N8 Subtype/classification , Influenza A Virus, H3N8 Subtype/genetics , Influenza A Virus, H3N8 Subtype/isolation & purification , Influenza A Virus, H5N8 Subtype/classification , Influenza A Virus, H5N8 Subtype/isolation & purification , Influenza A Virus, H7N7 Subtype/classification , Influenza A Virus, H7N7 Subtype/genetics , Influenza A Virus, H7N7 Subtype/isolation & purification , Influenza in Birds/virology , Phylogeography , Reassortant Viruses/classification , Reassortant Viruses/isolation & purification , Republic of Korea/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...