Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
IJID Reg ; 8(Suppl): S1-S7, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37799539

ABSTRACT

Objectives: To determine the lineage distribution of the virus during the first wave of the pandemic in North Sumatra, Indonesia. Methods: A total of 20 samples with positive results based on reverse transcription-polymerase chain reaction were selected for virus culture and then performed whole-genome sequence analysis using next-generation sequencing which was applied by the Illumina MiSeq instrument. Results: Whole-genome sequence analysis revealed that the majority of our samples belong to lineages B.1.468 (n = 10), B.1 (n = 5), B.1.1 (n = 2), B.1.1.398 (n = 2), and B.6 (n = 1). Other unique amino acid mutations found in our samples were present in A58T on non-structural protein (NSP3) (70%), P323L on NSP12 (95%), Q57H on NS3 protein (75%), and D614G on S (100%). Conclusion: The SARS-CoV-2 lineage B.1.468 may be the main virus variant circulating in North Sumatra at the beginning of the emergence of COVID-19 cases in this province.

2.
Int J Mol Sci ; 24(8)2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37108459

ABSTRACT

Acute respiratory distress syndrome is an acute respiratory failure caused by cytokine storms; highly pathogenic influenza A virus infection can induce cytokine storms. The innate immune response is vital in this cytokine storm, acting by activating the transcription factor NF-κB. Tissue injury releases a danger-associated molecular pattern that provides positive feedback for NF-κB activation. Exogenous mesenchymal stem cells can also modulate immune responses by producing potent immunosuppressive substances, such as prostaglandin E2. Prostaglandin E2 is a critical mediator that regulates various physiological and pathological processes through autocrine or paracrine mechanisms. Activation of prostaglandin E2 results in the accumulation of unphosphorylated ß-catenin in the cytoplasm, which subsequently reaches the nucleus to inhibit the transcription factor NF-κB. The inhibition of NF-κB by ß-catenin is a mechanism that reduces inflammation.


Subject(s)
Influenza, Human , Respiratory Distress Syndrome , Humans , NF-kappa B/metabolism , Dinoprostone , beta Catenin/metabolism , Cytokine Release Syndrome , Wnt Signaling Pathway , Immunity, Innate
3.
PeerJ ; 11: e14917, 2023.
Article in English | MEDLINE | ID: mdl-36846456

ABSTRACT

Background: Influenza A viruses are a major pathogen that causes significant clinical and economic harm to many animals. In Indonesia, the highly pathogenic avian influenza (HPAI) H5N1 virus has been endemic in poultry since 2003 and has caused sporadic deadly infections in humans. The genetic bases that determine host range have not yet been fully elucidated. We analyzed the whole-genome sequence of a recent H5 isolate to reveal the evolution toward its mammalian adaptation. Methods: We determined the whole-genome sequence of A/chicken/East Java/Av1955/2022 (hereafter, "Av1955") from a healthy chicken in April 2022 and conducted phylogenetic and mutational analysis. Results: Phylogenetic analysis revealed that Av1955 belonged to the H5N1 clade 2.3.2.1c (Eurasian lineage). The six gene segments (PB1, PB2, HA, NP, NA, and NS) out of the eight segments derived from viruses of H5N1 Eurasian lineage, one (PB2) from the H3N6 subtype and the remaining one (M) from the H5N1 clade 2.1.3.2b (Indonesian lineage). The donor of the PB2 segment was a reassortant among three viruses of H5N1 Eurasian and Indonesian lineages and the H3N6 subtype. The HA amino acid sequence contained multiple basic amino acids at the cleavage site. Mutation analysis revealed that Av1955 possessed the maximal number of mammalian adaptation marker mutations. Conclusions: Av1955 was a virus of H5N1 Eurasian lineage. The HA protein contains an HPAI H5N1-type cleavage site sequence, while the virus was isolated from a healthy chicken suggesting its low pathogenicity nature. The virus has increased mammalian adaptation markers by mutation and intra- and inter-subtype reassortment, gathering gene segments possessing the most abundant maker mutations among previously circulating viruses. The increasing mammalian adaptation mutation in avian hosts suggests that they might be adaptive to infection in mammalian and avian hosts. It highlights the importance of genomic surveillance and adequate control measures for H5N1 infection in live poultry markets.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza A virus , Influenza in Birds , Humans , Animals , Influenza in Birds/epidemiology , Chickens , Influenza A Virus, H5N1 Subtype/genetics , Indonesia , Phylogeny , Influenza A virus/genetics , Poultry , Mammals
5.
Sci Rep ; 12(1): 4835, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35318357

ABSTRACT

To halt the pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), governments around the world have imposed policies, such as lockdowns, mandatory mask wearing, and social distancing. The application of disinfecting materials in shared public facilities can be an additional measure to control the spread of the virus. Copper is a prominent material with antibacterial and antiviral effects. In this study, we synthesized copper nanoparticles (CuNPs) as a surface coating agent and assessed their antiviral activity against SARS-CoV-2. CuNPs with a mean size of 254 nm in diameter were synthesized from copper sulfate as a source and were predominantly composed of copper oxide. The synthesized CuNPs were mixed with resin-based paint (CuNP/paint) and sprayed on the surface of stainless steel remnants. SARS-CoV-2 lost 97.8% infectivity on the CuNP/paint-coated surface after 30 min of exposure and more than 99.995% infectivity after 1 h of exposure. The inactivation rate was approximately 36-fold faster than that on the paint alone-coated and uncoated surfaces. The CuNP/paint-coated surface showed powerful inactivation of SARS-CoV-2 infectivity, although further study is needed to elucidate the inactivation mechanisms. Applications of CuNP/paint coatings to public or hospital facilities and other commonly touched areas are expected to be beneficial.


Subject(s)
COVID-19 , Nanoparticles , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Communicable Disease Control , Copper , Humans , SARS-CoV-2
6.
Hum Genomics ; 15(1): 29, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001248

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19) is a global health problem that causes millions of deaths worldwide. The clinical manifestation of COVID-19 widely varies from asymptomatic infection to severe pneumonia and systemic inflammatory disease. It is thought that host genetic variability may affect the host's response to the virus infection and thus cause severity of the disease. The SARS-CoV-2 virus requires interaction with its receptor complex in the host cells before infection. The transmembrane protease serine 2 (TMPRSS2) has been identified as one of the key molecules involved in SARS-CoV-2 virus receptor binding and cell invasion. Therefore, in this study, we investigated the correlation between a genetic variant within the human TMPRSS2 gene and COVID-19 severity and viral load. RESULTS: We genotyped 95 patients with COVID-19 hospitalised in Dr Soetomo General Hospital and Indrapura Field Hospital (Surabaya, Indonesia) for the TMPRSS2 p.Val160Met polymorphism. Polymorphism was detected using a TaqMan assay. We then analysed the association between the presence of the genetic variant and disease severity and viral load. We did not observe any correlation between the presence of TMPRSS2 genetic variant and the severity of the disease. However, we identified a significant association between the p.Val160Met polymorphism and the SARS-CoV-2 viral load, as estimated by the Ct value of the diagnostic nucleic acid amplification test. Furthermore, we observed a trend of association between the presence of the C allele and the mortality rate in patients with severe COVID-19. CONCLUSION: Our data indicate a possible association between TMPRSS2 p.Val160Met polymorphism and SARS-CoV-2 infectivity and the outcome of COVID-19.


Subject(s)
COVID-19/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide , SARS-CoV-2/isolation & purification , Serine Endopeptidases/genetics , Adult , Alleles , COVID-19/diagnosis , COVID-19/virology , Cross-Sectional Studies , Female , Gene Frequency , Genotype , Humans , Indonesia , Male , Middle Aged , SARS-CoV-2/physiology , Viral Load/genetics
7.
BMC Infect Dis ; 20(1): 823, 2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33176722

ABSTRACT

BACKGROUND: The highly pathogenic avian influenza A/H5N1 virus is one of the causative agents of acute lung injury (ALI) with high mortality rate. Studies on therapeutic administration of bone marrow-derived mesenchymal stem cells (MSCs) in ALI caused by the viral infection have been limited in number and have shown conflicting results. The aim of the present investigation is to evaluate the therapeutic potential of MSC administration in A/H5N1-caused ALI, using a mouse model. METHODS: MSCs were prepared from the bone marrow of 9 to 12 week-old BALB/c mice. An H5N1 virus of A/turkey/East Java/Av154/2013 was intranasally inoculated into BALB/c mice. On days 2, 4, and 6 after virus inoculation, MSCs were intravenously administered into the mice. To evaluate effects of the treatment, we examined for lung alveolar protein as an indicator for lung injury, PaO2/FiO2 ratio for lung functioning, and lung histopathology. Expressions of NF-κB, RAGE (transmembrane receptor for damage associated molecular patterns), TNFα, IL-1ß, Sftpc (alveolar cell type II marker), and Aqp5+ (alveolar cell type I marker) were examined by immunohistochemistry. In addition, body weight, virus growth in lung and brain, and duration of survival were measured. RESULTS: The administration of MSCs lowered the level of lung damage in the virus-infected mice, as shown by measuring lung alveolar protein, PaO2/FiO2 ratio, and histopathological score. In the MSC-treated group, the expressions of NF-κB, RAGE, TNFα, and IL-1ß were significantly suppressed in comparison with a mock-treated group, while those of Sftpc and Aqp5+ were enhanced. Body weight, virus growth, and survival period were not significantly different between the groups. CONCLUSION: The administration of MSCs prevented further lung injury and inflammation, and enhanced alveolar cell type II and I regeneration, while it did not significantly affect viral proliferation and mouse morbidity and mortality. The results suggested that MSC administration was a promissing strategy for treatment of acute lung injuries caused by the highly pathogenic avian influenza A/H5N1 virus, although further optimization and combination use of anti-viral drugs will be obviously required to achieve the goal of reducing mortality.


Subject(s)
Acute Lung Injury/etiology , Acute Lung Injury/surgery , Influenza A Virus, H5N1 Subtype , Mesenchymal Stem Cell Transplantation , Orthomyxoviridae Infections/complications , Pneumonia/etiology , Pneumonia/surgery , Acute Lung Injury/prevention & control , Acute Lung Injury/virology , Animals , Cytokines/metabolism , Disease Models, Animal , Lung/metabolism , Lung/virology , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/virology , Pneumonia/prevention & control , Pneumonia/virology , Treatment Outcome
8.
Jpn J Infect Dis ; 73(5): 336-342, 2020 Sep 24.
Article in English | MEDLINE | ID: mdl-32350224

ABSTRACT

In Indonesia, the highly pathogenic avian influenza A/H5N1 virus has become endemic and has been linked with direct transmission to humans. From 2013 to 2014, we isolated avian influenza A/H5N1 and A/H3N6 viruses from poultry in Indonesia. This study aimed to reveal their pathogenicity in mammals using a mouse model. Three of the isolates, Av154 of A/H5N1 clade 2.3.2.1c, Av240 of A/H5N1 clade 2.1.3.2b, and Av39 of A/H3N6, were inoculated into BALB/c mice. To assess morbidity and mortality, we measured body weight daily and monitored survival for 20 d. Av154- and Av240-infected mice lost 25% of their starting body weight by day 7, while Av39-infected mice did not. Most of the Av154-infected mice died on day 8, while the majority of the Av240-infected mice survived until day 20. A 50% mouse lethal dose was calculated to be 2.0 × 101 50% egg infectious doses for Av154, 1.1 × 105 for Av240 and > 3.2 × 106 for Av39. The Av154 virus was highly virulent and lethal in mice without prior adaptation, suggesting its high pathogenic potential in mammals. The Av240 virus was highly virulent but modestly lethal, whereas the Av39 virus was neither virulent nor lethal. Several mammalian adaptive markers of amino acid residues were associated with the highly virulent and lethal phenotypes of the Av154 virus.


Subject(s)
Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/virology , Influenza, Human/virology , Poultry/virology , Amino Acid Sequence , Animals , Body Weight , Female , Humans , Indonesia , Influenza A virus/classification , Influenza A virus/pathogenicity , Influenza in Birds/mortality , Influenza, Human/mortality , Mice , Mice, Inbred BALB C , Models, Animal , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Phenotype , Virulence
9.
Microbiol Resour Announc ; 8(17)2019 Apr 25.
Article in English | MEDLINE | ID: mdl-31023807

ABSTRACT

We isolated an avian influenza A/H9N2 virus from an apparently healthy chicken at a live-poultry market in January 2018. This is the first report of a whole-genome sequence of A/H9N2 virus in Indonesia. Phylogenetic analyses indicated that intrasubtype reassortment of genome segments is involved in the genesis of the A/H9N2 virus.

10.
Arch Virol ; 163(8): 2099-2109, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29633076

ABSTRACT

We evaluated the anti-influenza-virus effects of Melia components and discuss the utility of these components. The effects of leaf components of Melia azedarach L. on viruses were examined, and plaque inhibition tests were performed. The in vivo efficacy of M. azedarach L. was tested in a mouse model. Leaf components of Melia azedarach L. markedly inhibited the growth of various influenza viruses. In an initial screening, multiplication and haemagglutination (HA) activities of H1N1, H3N2, H5, and B influenza viruses were inactivated by the liquid extract of leaves of M. azedarach L. (MLE). Furthermore, plaque inhibition titres of H1N1, H3N2, and B influenza viruses treated with MLE ranged from 103.7 to 104.2. MLE possessed high plaque-inhibitory activity against pandemic avian H5N1, H7N9, and H9N2 vaccine candidate strains, with a plaque inhibition titre of more than 104.2. Notably, the buoyant density decreased from 1.175 to 1.137 g/cm3, and spikeless particles appeared. We identified four anti-influenza virus substances: pheophorbide b, pheophorbide a, pyropheophorbide a, and pheophytin a. Photomorphogenesis inside the envelope may lead to removal of HA and neuraminidase spikes from viruses. Thus, MLE could efficiently remove floating influenza virus in the air space without toxicity. Consistent with this finding, intranasal administration of MLE in mice significantly decreased the occurrence of pneumonia. Additionally, leaf powder of Melia (MLP) inactivated influenza viruses and viruses in the intestines of chickens. MLE and MLP may have applications as novel, safe biological disinfectants for use in humans and poultry.


Subject(s)
Antiviral Agents/administration & dosage , Influenza A virus/drug effects , Influenza A virus/growth & development , Influenza B virus/drug effects , Influenza B virus/growth & development , Influenza in Birds/drug therapy , Melia azedarach/chemistry , Plant Extracts/administration & dosage , Animals , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Chick Embryo , Chickens , Female , Influenza A virus/genetics , Influenza A virus/metabolism , Influenza B virus/genetics , Influenza B virus/metabolism , Influenza in Birds/virology , Mice , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Leaves/chemistry , Poultry Diseases/virology
11.
J Infect Dis ; 214(12): 1929-1936, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27923953

ABSTRACT

BACKGROUND: In Indonesia, highly pathogenic avian influenza A(H5N1) virus has become endemic in poultry and has caused sporadic deadly infections in human. Since 2012, we have conducted fixed-point surveillance of avian influenza viruses at a live-poultry market in East Java, Indonesia. In this study, we examined the seroprevalence of avian influenza A(H5N1) virus infection among market workers. METHODS: Sera were collected from 101 workers in early 2014 and examined for antibody activity against avian A(H5N1) Eurasian lineage virus by a hemagglutination-inhibition (HI) assay. RESULTS: By the HI assay, 84% of the sera tested positive for antibody activity against the avian virus. Further analysis revealed that the average HI titer in 2014 was 2.9-fold higher than in 2012 and that seroconversion occurred in 44% of paired sera (11 of 25) between 2012 and 2014. A medical history survey was performed in 2016; responses to questionnaires indicated that none of workers had had severe acute respiratory illness during 2013. CONCLUSIONS: This study provides evidence of a high prevalence of avian A(H5N1) virus infection in 2013 among workers at a live-poultry market. However, because no instances of hospitalizations were reported, we can conclude the virus did not manifest any clinical symptoms in workers.


Subject(s)
Animal Husbandry , Antibodies, Viral/blood , Asymptomatic Infections/epidemiology , Influenza A Virus, H5N1 Subtype/immunology , Influenza, Human/epidemiology , Occupational Exposure , Animals , Hemagglutination Inhibition Tests , Humans , Indonesia/epidemiology , Poultry , Seroepidemiologic Studies
12.
Cell Mol Life Sci ; 72(2): 357-66, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25001578

ABSTRACT

Influenza is a serious respiratory disease among immunocompromised individuals, such as the elderly, and its prevention is an urgent social issue. Influenza viruses rely on neuraminidase (NA) activity to release progeny viruses from infected cells and spreading the infection. NA is, therefore, an important target of anti-influenza drugs. A causal relationship between bacteria and influenza virus infection has not yet been established, however, a positive correlation between them has been reported. Thus, in this study, we examined the biological effects of oral mitis group streptococci, which are predominant constituents of human oral florae, on the release of influenza viruses. Among them, Streptococcus oralis ATCC 10557 and Streptococcus mitis ATCC 6249 were found to exhibit NA activity and their culture supernatants promoted the release of influenza virus and cell-to-cell spread of the infection. In addition, culture supernatants of these NA-producing oral bacteria increased viral M1 protein expression levels and cellular ERK activation. These effects were not observed with culture supernatants of Streptococcus sanguinis ATCC 10556 which lacks the ability to produce NA. Although the NA inhibitor zanamivir suppressed the release of progeny viruses from the infected cells, the viral release was restored upon the addition of culture supernatants of NA-producing S. oralis ATCC 10557 or S. mitis ATCC 6249. These findings suggest that an increase in the number of NA-producing oral bacteria could elevate the risk of and exacerbate the influenza infection, hampering the efficacy of viral NA inhibitor drugs.


Subject(s)
Antiviral Agents/pharmacology , Influenza, Human/drug therapy , Influenza, Human/microbiology , Neuraminidase/metabolism , Streptococcus mitis/enzymology , Streptococcus oralis/enzymology , Zanamivir/pharmacology , Analysis of Variance , Blotting, Western , Fluorescent Antibody Technique, Indirect , Humans , Zanamivir/antagonists & inhibitors
13.
PLoS One ; 8(12): e81396, 2013.
Article in English | MEDLINE | ID: mdl-24339924

ABSTRACT

Discrimination of closely related strains is a key issue, particularly for infectious diseases whose incidence fluctuates according to variations in the season and evolutionary changes. Among infectious diseases, influenza viral infections are a worldwide cause of pandemic disease and mortality. With the emergence of different influenza strains, it is vital to develop a method using antibodies that can differentiate between viral types and subtypes. Ideally, such a system would also be user friendly. In this study, a polyclonal antibody generated against A/Udorn/307/1972 (H3N2) was used as a probe to distinguish between influenza H3N2 viruses based on the interaction between the antibody and hemagglutinin, demonstrating its applicability for viral discrimination. Clear discrimination was demonstrated using an evanescent-field-coupled waveguide-mode sensor, which has appealing characteristics over other methods in the viewpoint of improving the sensitivity, measurement time, portability and usability. Further supporting evidence was obtained using enzyme-linked immunosorbent assays, hemagglutination-inhibition assays, and infectivity neutralization assays. The results obtained indicate that the polyclonal antibody used here is a potential probe for distinguishing influenza viruses and, with the aid of a handheld sensor it could be used for influenza surveillance.


Subject(s)
Antibodies, Viral/immunology , Antibody Specificity , Biosensing Techniques/methods , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/isolation & purification , Amino Acid Sequence , Antibodies, Viral/chemistry , Enzyme-Linked Immunosorbent Assay , Gold/chemistry , Hemagglutination Inhibition Tests , Metal Nanoparticles/chemistry , Molecular Sequence Data , Neutralization Tests , Species Specificity
14.
PLoS One ; 8(7): e69121, 2013.
Article in English | MEDLINE | ID: mdl-23874887

ABSTRACT

Gold nanoparticles were conjugated to an antibody (immuno-AuNP) against A/Udorn/307/1972 (H3N2) influenza virus to detect viruses on a sensing plate designed for an evanescent field-coupled waveguide-mode sensor. Experiments were conducted using human influenza A/H3N2 strains, and immuno-AuNP could detect 8×10(5) PFU/ml (40 pg/µl) intact A/Udorn/307/1972 and 120 pg/µl A/Brisbane/10/2007. Furthermore, increased signal magnitude was achieved in the presence of non-ionic detergent, as the virtual detection level was increased to 8×10(4) PFU/ml A/Udorn/307/1972. Immuno-AuNPs were then complexed with viruses to permit direct observation, and they formed a ring of confined nanodots on the membrane of both intact and detergent-treated viruses as directly visualized by scanning electron microscopy. With this complex the detection limit was improved further to 8×10(3) PFU/ml on anti-rabbit IgG immobilized sensing plate. These strategies introduce methods for observing trapped intact viruses on the sensing plates generated for optical systems.


Subject(s)
Gold/administration & dosage , Immunoconjugates/administration & dosage , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/diagnosis , Metal Nanoparticles/administration & dosage , Animals , Antibodies/immunology , Dogs , Humans , Immunoconjugates/immunology , Influenza, Human/immunology , Madin Darby Canine Kidney Cells , Microscopy, Electron, Scanning/methods
15.
J Mol Graph Model ; 44: 161-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23831996

ABSTRACT

Influenza hemagglutinin (HA) consists of a fibrous globular stem (HA2) inserted into the viral membrane supporting a globular head (HA1). HA1 receptor-binding has been hypothesized to be structurally correlated to the HA2 B-loop, however, this was never fully understood. Here, we elucidated the structural relationship between the HA2 B-loop and the HA1 receptor-binding site (RBS). Throughout this study, we analyzed 2486 H1N1 HA homology models obtained from human, swine and avian strains during 1976-2012. Quality of all homology models were verified before further analyses. We established that amino acid residue 882 is putatively strain-conserved and differs in the human (K882), swine (H882) and avian (N882) strains. Moreover, we observed that the amino acid at residue 882 and, similarly, its orientation has the potential to influence the HA1 RBS diameter measurements which we hypothesize may consequentially affect influenza H1N1 viral infectivity, immune escape, transmissibility, and evolution.


Subject(s)
Binding Sites , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Models, Molecular , Receptors, Virus/chemistry , Amino Acids/chemistry , Animals , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H1N1 Subtype , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Conformation , Quantitative Structure-Activity Relationship , Receptors, Virus/metabolism
16.
J Biosci Bioeng ; 116(5): 620-3, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23742748

ABSTRACT

A protein that specifically targets lipid droplets (LDs) was created by connecting two domains of nonstructural protein 4B containing amphipathic helices from hepatitis C virus. We demonstrated its direct targeting and accumulation to the LD surface by time-lapse live cell imaging, comparable to those observed with adipose differentiation-related protein.


Subject(s)
Hepacivirus/chemistry , Lipids , Proteins/chemistry , Proteins/metabolism , Cell Line , Cell Survival , Humans , Membrane Proteins/metabolism , Microscopy, Confocal , Perilipin-2 , Protein Structure, Tertiary , Proteins/genetics , Substrate Specificity , Time Factors , Viral Nonstructural Proteins/chemistry
17.
Biol Pharm Bull ; 36(7): 1152-8, 2013.
Article in English | MEDLINE | ID: mdl-23628892

ABSTRACT

Pyrrole-imidazole (PI) polyamides are a novel group of gene-silencing compounds, which bind to a minor groove of double stranded (ds)DNA in a sequence-specific manner. To explore the RNA binding properties of PI polyamides targeting rat transforming growth factor-ß1 (TGF-ß1 Polyamide) and influenza A virus (PA polyamide), we designed dsRNAs with an identical sequence to the target DNA and analyzed RNA binding properties of the polyamide. Biacore assay showed fast binding of TGF-ß1 Polyamide to the dsRNA, whereas mismatch polyamide did not bind to the dsRNA. Dissociation equilibrium constant (KD) value was 6.7×10(-7) of the target dsRNA. These results indicate that PI polyamide could bind to RNA with a 2 log lower binding affinity than its DNA-binding affinity. We designed a PI polyamide targeting the panhandle stem region of influenza A virus. KD value of the PI polyamide to dsRNA targeting influenza A virus was 4.6×10(-7). Gel-shift assay showed that TGF-ß1 and PA polyamides bound to the appropriate dsDNA, whereas these PI polyamides did not show obvious gel-shift with the appropriate dsRNA. Structural modeling suggests that PI polyamide binds to the appropriate B-form dsDNA in the minor groove, whereas it does not fit in the minor groove to dsRNA. Thus PI polyamides have a lower binding affinity with target dsRNA than they do with dsDNA. The distinct binding properties of PI polyamides to dsRNA and dsDNA may be associated with differences of secondary structure and chemical binding properties between target RNA and DNA.


Subject(s)
Gene Silencing/drug effects , Imidazoles/pharmacology , Influenza A virus/chemistry , Nylons/pharmacology , Pyrroles/pharmacology , RNA, Double-Stranded/chemistry , Transforming Growth Factor beta1/chemistry , Animals , DNA/chemistry , DNA/genetics , Electrophoretic Mobility Shift Assay , Influenza A virus/genetics , Molecular Docking Simulation , Nucleic Acid Conformation , RNA, Double-Stranded/genetics , Rats , Surface Plasmon Resonance , Transforming Growth Factor beta1/genetics
18.
J Ethnopharmacol ; 146(3): 866-72, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23439031

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The fruits of Chaenomeles sinensis Koehne (Chinese quince) are distributed throughout China and Japan. It has traditionally been known to have a therapeutic effect against respiratory symptoms caused by infectious diseases. AIM OF THE STUDY: The polyphenol-rich extract, CSD3, from Chaenomeles sinensis has previously been shown to neutralize influenza virus infectivity. The aim of this study was to clarify which step(s) in the replication cycle in vitro were inhibited. MATERIALS AND METHODS: We examined cell-binding, hemagglutination and hemolytic activities and infectivity of A/Udorn/72(H3N2) virus after pre-treatment with CSD3. We also investigated the time course of synthesis for viral mRNA, cRNA, and vRNA in Madin-Darby canine kidney epithelial cells (MDCK) cells infected with CSD3-treated virus. Finally, we studied the effect of CSD3-treatment on the ultrastructure of the influenza virion. RESULTS: Pre-treatment with CSD3 mildly reduced cell-binding, hemagglutination and hemolytic activities. These activities were reduced by 70% to be equivalent to 30% of the control at 1µg/ml. CSD3 severely reduced infectivity to 1% of the control at 1µg/ml. Primary transcription in MDCK cells infected with CSD3 (1µg/ml)-treated virus was decreased to about 1% of that in cells infected with mock-treated virus. Synthesis of viral cRNA, vRNA and secondary mRNA was also severely decreased. Electron microscopy revealed that the integrity of the virus envelope was damaged by CSD3 and was permeable to uranyl acetate. CONCLUSIONS: The main target step(s) of CSD3 in the replication cycle is after cell-binding but before or at primary transcription. Involvement of the increased permeability of virus envelope as the inhibition mechanism was proposed. CSD3 could be useful in preventing influenza virus infection, and be employed as a lozenge or mouthwash for daily use.


Subject(s)
Antiviral Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Influenza A Virus, H3N2 Subtype/drug effects , Polyphenols/pharmacology , Rosaceae/chemistry , Transcription, Genetic/drug effects , Animals , Antiviral Agents/isolation & purification , Cell Culture Techniques , Chickens , Dogs , Drugs, Chinese Herbal/isolation & purification , Epithelial Cells/drug effects , Epithelial Cells/virology , Erythrocytes/drug effects , Erythrocytes/virology , Hemagglutination, Viral/drug effects , Hemolysis/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/ultrastructure , Madin Darby Canine Kidney Cells , Microscopy, Electron, Transmission , Polyphenols/isolation & purification , RNA, Viral/biosynthesis , RNA, Viral/genetics , Virion/ultrastructure , Virus Replication/drug effects
19.
PLoS One ; 8(2): e55874, 2013.
Article in English | MEDLINE | ID: mdl-23409074

ABSTRACT

We isolated and characterized two human monoclonal antibodies to the envelope E2 protein of hepatitis C virus (HCV). Lymphoblastoid cell lines stably producing antibodies were obtained by immortalizing peripheral blood mononuclear cells of a patient with chronic hepatitis C using Epstein-Barr virus. Screening for antibody-positive clones was carried out by immunofluorescence with Huh7 cells expressing the E2 protein of HCV strain H (genotype 1a) isolated from the same patient. Isotype of resulting antibodies, #37 and #55, was IgG1/kappa and IgG1/lambda, respectively. Epitope mapping revealed that #37 and #55 recognize conformational epitopes spanning amino acids 429 to 652 and 508 to 607, respectively. By immunofluorescence using virus-infected Huh7.5 cells as targets both antibodies were reactive with all of the nine different HCV genotypes/subtypes tested. The antibodies showed a different pattern of immuno-staining; while #37 gave granular reactions mostly located in the periphery of the nucleus, #55 gave diffuse staining throughout the cytoplasm. Both antibodies were shown by immuno-gold electron microscopy to bind to intact viral particles. In a neutralization assay (focus-forming unit reduction using chimeric infectious HCV containing structural proteins derived from genotypes 1a, 1b, 2a, 2b, 3a, 4a, 5a, 6a, and 7a), #55 inhibited the infection of all HCV genotypes tested but genotype 7a to a lesser extent. #37 did not neutralize any of these viruses. As a broadly cross-neutralizing human antibody, #55 may be useful for passive immunotherapy of HCV infection.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Hepacivirus/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Antibodies, Blocking/immunology , Antibodies, Blocking/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Neutralizing , Antibodies, Viral/chemistry , Antibodies, Viral/genetics , Antibody Affinity/immunology , Antigens, Viral/chemistry , Antigens, Viral/immunology , Antigens, Viral/metabolism , Cell Line, Transformed , Cross Reactions/immunology , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Genotype , Hepacivirus/genetics , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Lymphocytes/immunology , Lymphocytes/metabolism , Molecular Sequence Data , Neutralization Tests , Protein Transport , Sequence Alignment , Viral Envelope Proteins/metabolism , Virion/immunology , Virion/ultrastructure
20.
PLoS One ; 7(9): e45371, 2012.
Article in English | MEDLINE | ID: mdl-23028967

ABSTRACT

Influenza virus neuraminidase (NA) cleaves terminal sialic acid residues on oligosaccharide chains that are receptors for virus binding, thus playing an important role in the release of virions from infected cells to promote the spread of cell-to-cell infection. In addition, NA plays a role at the initial stage of viral infection in the respiratory tract by degrading hemagglutination inhibitors in body fluid which competitively inhibit receptor binding of the virus. Current first line anti-influenza drugs are viral NA-specific inhibitors, which do not inhibit bacterial neuraminidases. Since neuraminidase producing bacteria have been isolated from oral and upper respiratory commensal bacterial flora, we posited that bacterial neuraminidases could decrease the antiviral effectiveness of NA inhibitor drugs in respiratory organs when viral NA is inhibited. Using in vitro models of infection, we aimed to clarify the effects of bacterial neuraminidases on influenza virus infection in the presence of the NA inhibitor drug zanamivir. We found that zanamivir reduced progeny virus yield to less than 2% of that in its absence, however the yield was restored almost entirely by the exogenous addition of bacterial neuraminidase from Streptococcus pneumoniae. Furthermore, cell-to-cell infection was severely inhibited by zanamivir but restored by the addition of bacterial neuraminidase. Next we examined the effects of bacterial neuraminidase on hemagglutination inhibition and infectivity neutralization activities of human saliva in the presence of zanamivir. We found that the drug enhanced both inhibitory activities of saliva, while the addition of bacterial neuraminidase diminished this enhancement. Altogether, our results showed that bacterial neuraminidases functioned as the predominant NA when viral NA was inhibited to promote the spread of infection and to inactivate the neutralization activity of saliva. We propose that neuraminidase from bacterial flora in patients may reduce the efficacy of NA inhibitor drugs during influenza virus infection. (295 words).


Subject(s)
Antiviral Agents/pharmacology , Bacterial Proteins/pharmacology , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Neuraminidase/pharmacology , Orthomyxoviridae/drug effects , Virus Replication/drug effects , Animals , Cell Line , Dogs , Streptococcus pneumoniae/enzymology , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , Zanamivir/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL