Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
1.
Respir Investig ; 61(2): 270-283, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36543714

ABSTRACT

Respiratory viruses like rhinovirus, influenza virus, respiratory syncytial virus, and coronavirus cause several respiratory diseases, such as bronchitis, pneumonia, pulmonary fibrosis, and coronavirus disease 2019, and exacerbate bronchial asthma, chronic obstructive pulmonary disease, bronchiectasis, and diffuse panbronchiolitis. The production of inflammatory mediators and mucin and the accumulation of inflammatory cells have been reported in patients with viral infection-induced respiratory diseases. Interleukin (IL)-1ß, IL-6, IL-8, tumor necrosis factor-α, granulocyte-macrophage colony-stimulating factor, and regulated on activation normal T-cell expressed and secreted are produced in the cells, including human airway and alveolar epithelial cells, partly through the activation of toll-like receptors, nuclear factor kappa B and p44/42 mitogen-activated protein kinase. These mediators are associated with the development of viral infection-induced respiratory diseases through the induction of inflammation and injury in the airway and lung, airway remodeling and hyperresponsiveness, and mucus secretion. Medications used to treat respiratory diseases, including corticosteroids, long-acting ß2-agonists, long-acting muscarinic antagonists, mucolytic agents, antiviral drugs for severe acute respiratory syndrome coronavirus 2 and influenza virus, macrolides, and Kampo medicines, reduce the production of viral infection-induced mediators, including cytokines and mucin, as determined in clinical, in vivo, or in vitro studies. These results suggest that the anti-inflammatory effects of these medications on viral infection-induced respiratory diseases may be associated with clinical benefits, such as improvements in symptoms, quality of life, and mortality rate, and can prevent hospitalization and the exacerbation of chronic obstructive pulmonary disease, bronchial asthma, bronchiectasis, and diffuse panbronchiolitis.


Subject(s)
Asthma , Bronchiectasis , COVID-19 , Pulmonary Disease, Chronic Obstructive , Virus Diseases , Humans , Quality of Life , Asthma/drug therapy , Pulmonary Disease, Chronic Obstructive/drug therapy , Virus Diseases/drug therapy , Anti-Inflammatory Agents/therapeutic use , Mucins/therapeutic use
2.
Front Pharmacol ; 12: 687818, 2021.
Article in English | MEDLINE | ID: mdl-34531740

ABSTRACT

Rhinovirus (RV) is a primary etiologic agent of common cold that can subsequently acutely exacerbate bronchial asthma or chronic obstructive pulmonary disease. Kakkonto (Ge-gen-tang in Chinese), one of the most frequently prescribed traditional Japanese (Kampo) medicines, is used for treating common cold, shoulder stiffness, or inflammatory diseases of the upper body. Previous experimental studies have indicated that kakkonto exerts antiviral and anti-inflammatory effects on the influenza virus and the human respiratory syncytial virus. However, there is a lack of reports investigating the efficacy of kakkonto in RV infection. Hence, the aim of the current study was to investigate the effects of kakkonto on RV infection of human nasal epithelial (HNE) cells. HNE cells obtained via endoscopic sinus surgery were cultured and infected with RV14, with or without kakkonto treatment. The supernatants from the cells were collected, and the RV14 titer and cytokine levels were assessed. Reverse transcription-polymerase chain reaction was performed to determine the amount of viral RNA, while the level of nuclear factor kappa B (NF-κB) subunits in the nucleus was assessed by enzyme-linked immunosorbent assay. Although kakkonto treatment did not reduce RV14 titer or RNA levels, indicating that it did not inhibit RV14 proliferation, it was found to reduce the production of specific pro-inflammatory cytokines, including interleukin (IL)-8, tumor necrosis factor (TNF)-α, and monocyte chemotactic protein-1 (MCP-1). Unlike that observed with the kakkonto extract, none of the crude drugs contained in kakkonto reduced IL-8 level. Furthermore, though kakkonto treatment significantly reduced p50 levels, it did not impact the p65 subunit of NF-κB. These results indicated that kakkonto can inhibit inflammation caused by RV infection and may exert an immunomodulatory effect on HNE cells. This is the first report to elucidate the effects of kakkonto extract on RV infection in primary cultures of HNE cells, providing evidence that kakkonto may act as an effective therapy for RV infection and subsequent airway inflammation.

3.
Virus Res ; 304: 198548, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34425162

ABSTRACT

Rhinovirus species C (RV-C) causes more severe asthma attacks than other rhinovirus species. However, the modulation of RV-C replication by drugs has not been well studied. Primary human nasal epithelial (HNE) cells cultured on filter membranes with air-liquid interface methods were infected with RV-C03, and the levels of RV-C03 RNA collected from the airway surface liquid (ASL) of HNE cells were measured with a SYBR Green assay. Pretreatment of HNE cells with the specific vacuolar H+-ATPase inhibitor bafilomycin A1 reduced the RV-C03 RNA levels in the ASL; inflammatory cytokines, including interleukin (IL)-1ß, IL-6 and IL-8, in the supernatant; the mRNA expression of the RV-C receptor cadherin-related family member 3 (CDHR3) in the cells; and the number of acidic endosomes where RV-B RNA enters the cytoplasm. The levels of RV-C03 RNA in the ASL obtained from HNE cells with the CDHR3 rs6967,330 G/A genotype tended to be higher than those obtained from HNE cells with the G/G genotype. Pretreatment with the Na+/H+ exchanger inhibitor ethyl-isopropyl amiloride or either of the macrolides clarithromycin or EM900 also reduced RV-C03 RNA levels in the ASL and the number of acidic endosomes in HNE cells. In addition, significant levels of RV-A16, RV-B14 and RV-C25 RNA were detected in the ASL, and bafilomycin A1 also decreased the RV-C25 RNA levels. These findings suggest that bafilomycin A1 may reduce the release of RV-Cs and inflammatory cytokines from human airway epithelial cells. RV-Cs may be sensitive to drugs, including bafilomycin A1, that increase endosomal pH, and CDHR3 may mediate virus entry through receptor-mediated endocytosis in human airway epithelial cells.


Subject(s)
Picornaviridae Infections , Protons , Adenosine Triphosphatases/metabolism , Cadherin Related Proteins , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Cytokines/metabolism , Enterovirus , Epithelial Cells , Humans , Macrolides , Membrane Proteins/genetics , RNA/metabolism , Rhinovirus , Virus Replication
4.
J Virol ; 95(21): e0129621, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34406864

ABSTRACT

Influenza C virus (ICV) has only one kind of spike protein, the hemagglutinin-esterase (HE) glycoprotein. HE functions similarly to hemagglutinin (HA) and neuraminidase of the influenza A and B viruses (IAV and IBV, respectively). It has a monobasic site, which is cleaved by some host enzymes. The cleavage is essential to activating the virus, but the enzyme or enzymes in the respiratory tract have not been identified. This study investigated whether the host serine proteases, transmembrane protease serine S1 member 2 (TMPRSS2) and human airway trypsin-like protease (HAT), which reportedly cleave HA of IAV/IBV, are involved in HE cleavage. We established TMPRSS2- and HAT-expressing MDCK cells (MDCK-TMPRSS2 and MDCK-HAT). ICV showed multicycle replication with HE cleavage without trypsin in MDCK-TMPRSS2 cells as well as IAV did. The HE cleavage and multicycle replication did not appear in MDCK-HAT cells infected with ICV without trypsin, while HA cleavage and multistep growth of IAV appeared in the cells. Amino acid sequences of the HE cleavage site in 352 ICV strains were completely preserved. Camostat and nafamostat suppressed the growth of ICV and IAV in human nasal surface epithelial (HNE) cells. Therefore, this study revealed that, at least, TMPRSS2 is involved in HE cleavage and suggested that nafamostat could be a candidate for therapeutic drugs for ICV infection. IMPORTANCE Influenza C virus (ICV) is a pathogen that causes acute respiratory illness, mostly in children, but there are no anti-ICV drugs. ICV has only one kind of spike protein, the hemagglutinin-esterase (HE) glycoprotein on the virion surface, which possesses receptor-binding, receptor-destroying, and membrane fusion activities. The HE cleavage is essential for the virus to be activated, but the enzyme or enzymes in the respiratory tract have not been identified. This study revealed that transmembrane protease serine S1 member 2 (TMPRSS2), and not human airway trypsin-like protease (HAT), is involved in HE cleavage. This is a novel study on the host enzymes involved in HE cleavage, and the result suggests that the host enzymes, such as TMPRSS2, may be a target for therapeutic drugs of ICV infection.


Subject(s)
Gammainfluenzavirus/enzymology , Gammainfluenzavirus/metabolism , Hemagglutinins, Viral/metabolism , Influenza, Human/virology , Orthomyxoviridae Infections/virology , Serine Endopeptidases/metabolism , Viral Fusion Proteins/metabolism , Amino Acid Sequence , Animals , Antiviral Agents/pharmacology , Benzamidines/pharmacology , Cell Line , Cell Line, Tumor , Cells, Cultured , Dogs , Esters/pharmacology , Guanidines/pharmacology , Host Microbial Interactions , Humans , Madin Darby Canine Kidney Cells , Trypsin/metabolism , Viral Proteins/metabolism
5.
J Med Virol ; 93(6): 3484-3495, 2021 06.
Article in English | MEDLINE | ID: mdl-33247612

ABSTRACT

The effects of the clinically used protease inhibitor nafamostat on influenza virus replication have not been well studied. Primary human tracheal (HTE) and nasal (HNE) epithelial cells were pretreated with nafamostat and infected with the 2009 pandemic [A/Sendai-H/108/2009/(H1N1) pdm09] or seasonal [A/New York/55/2004(H3N2)] influenza virus. Pretreatment with nafamostat reduced the titers of the pandemic and seasonal influenza viruses and the secretion of inflammatory cytokines, including interleukin-6 and tumor necrosis factor-α, in the supernatants of the cells infected with the pandemic influenza virus. HTE and HNE cells exhibited mRNA and/or protein expression of transmembrane protease serine 2 (TMPRSS2), TMPRSS4, and TMPRSS11D. Pretreatment with nafamostat reduced cleavage of the precursor protein HA0 of the pandemic influenza virus into subunit HA1 in HTE cells and reduced the number of acidic endosomes in HTE and HNE cells where influenza virus RNA enters the cytoplasm. Additionally, nafamostat (30 mg/kg/day, intraperitoneal administration) reduced the levels of the pandemic influenza virus [A/Hyogo/YS/2011 (H1N1) pdm09] in mouse lung washes. These findings suggest that nafamostat may inhibit influenza virus replication in human airway epithelial cells and mouse lungs and reduce infection-induced airway inflammation by modulating cytokine production.


Subject(s)
Benzamidines/pharmacology , Benzamidines/therapeutic use , Epithelial Cells/drug effects , Guanidines/pharmacology , Guanidines/therapeutic use , Lung/drug effects , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/therapeutic use , Virus Replication/drug effects , Animals , Cells, Cultured , Cytokines/analysis , Cytokines/immunology , Epithelial Cells/virology , Female , Humans , Lung/virology , Male , Mice , Mice, Inbred BALB C , Nose/cytology , Trachea/cytology
6.
Article in English | MEDLINE | ID: mdl-32368031

ABSTRACT

Background: Different characteristics of patients with chronic obstructive pulmonary disease (COPD) between Western and Japanese populations have been reported. Risk factors for COPD exacerbation have been reported in Western countries but have not been studied in Japan. Patients and Methods: We retrospectively examined risk factors for COPD exacerbation. A total of 156 Japanese patients were enrolled, and the records of 136 patients were analyzed. Results: In the exacerbation group (n=60), body mass index, forced vital capacity (FVC), forced expiratory volume in one second (FEV1), the FEV1/FVC ratio (FEV1/FVC), the percent predicted values of FEV1 (%FEV1), and serum total protein (TP) and albumin concentrations were lower, and age, mortality rate, frequency of common cold and pneumonia, COPD severity rankings, modified Medical Research Council (mMRC) dyspnea score, and proportions of patients with severe emphysema (>50% of low attenuation area) and receiving long-term oxygen therapy were higher than those in the nonexacerbation group (n=76). However, the proportion of patients with a greater number of eosinophils (≥200/µL and/or ≥2%) and the exhaled nitric oxide concentration did not differ between the two groups. In the univariate analysis, the risk factors for exacerbation were age; long-term oxygen therapy; low FVC, FEV1, FEV1/FVC and %FEV1; high COPD severity ranking and mMRC score; severe emphysema; hypoproteinemia (<6.5 g/dL); hypoalbuminemia (<3.5 g/dL); leukocytosis; lymphocytopenia; and anemia. In the multivariate analysis, the risk factors were hypoalbuminemia, hypoproteinemia and low FEV1. Additionally, in patients in the exacerbation-induced mortality subgroup, age, exacerbation frequency, mMRC score and the proportion of patients with lymphocytopenia were higher, and FVC, %FVC, FEV1, serum TP concentration and the lymphocyte number were lower than those in the exacerbation survival subgroup. Conclusion: Malnutrition, airflow limitation and severe emphysema were risks for exacerbation and mortality associated with infection in Japanese patients with COPD.


Subject(s)
Emphysema , Malnutrition , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Forced Expiratory Volume , Humans , Japan/epidemiology , Malnutrition/diagnosis , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/therapy , Pulmonary Emphysema/diagnosis , Pulmonary Emphysema/epidemiology , Retrospective Studies , Severity of Illness Index , Vital Capacity
7.
Tohoku J Exp Med ; 251(1): 27-30, 2020 05.
Article in English | MEDLINE | ID: mdl-32448818

ABSTRACT

The number of patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly increased, although the WHO declared a pandemic. However, drugs that function against SARS-CoV-2 have not been established. SARS-CoV-2 has been suggested to bind angiotensin-converting enzyme 2, the receptor of the SARS coronavirus. SARS coronavirus and coronavirus 229E, the cause of the common cold, replicate through cell-surface and endosomal pathways using a protease, the type II transmembrane protease. To examine the effects of protease inhibitors on the replication of coronavirus 229E, we pretreated primary cultures of human nasal epithelial (HNE) cells with camostat or nafamostat, each of which has been used for the treatment of pancreatitis and/or disseminated intravascular coagulation. HNE cells were then infected with coronavirus 229E, and viral titers in the airway surface liquid of the cells were examined. Pretreatment with camostat (0.1-10 µg/mL) or nafamostat (0.01-1 µg/mL) reduced the titers of coronavirus 229E. Furthermore, a significant amount of type II transmembrane protease protein was detected in the airway surface liquid of HNE cells. Additionally, interferons have been reported to have antiviral effects against SARS coronavirus. The additive effects of interferons on the inhibitory effects of other candidate drugs to treat SARS-CoV-2 infection, such as lopinavir, ritonavir and favipiravir, have also been studied. These findings suggest that protease inhibitors of this type may inhibit coronavirus 229E replication in human airway epithelial cells at clinical concentrations. Protease inhibitors, interferons or the combination of these drugs may become candidate drugs to inhibit the replication of SARS-CoV-2.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus 229E, Human/drug effects , Coronavirus Infections/drug therapy , Gabexate/analogs & derivatives , Guanidines/pharmacology , Pneumonia, Viral/drug therapy , Protease Inhibitors/pharmacology , Virus Replication/drug effects , Benzamidines , Betacoronavirus/drug effects , COVID-19 , Cells, Cultured , Coronavirus 229E, Human/enzymology , Coronavirus 229E, Human/physiology , Culture Media, Conditioned , Epithelial Cells/virology , Esters , Gabexate/pharmacology , Humans , Nasal Mucosa/cytology , Pandemics , Primary Cell Culture , SARS-CoV-2 , Serine Endopeptidases/physiology , Spike Glycoprotein, Coronavirus/metabolism , Viral Load
8.
Respir Investig ; 58(3): 155-168, 2020 May.
Article in English | MEDLINE | ID: mdl-32094077

ABSTRACT

BACKGROUND: Coronavirus 229E (HCoV-229E), one of the causes of the common cold, exacerbates chronic obstructive pulmonary disease (COPD) and bronchial asthma. Long-acting muscarinic antagonists and ß2-agonists and inhaled corticosteroids inhibit the exacerbation of COPD and bronchial asthma caused by infection with viruses, including HCoV-229E. However, the effects of these drugs on HCoV-229E replication and infection-induced inflammation in the human airway are unknown. METHODS: Primary human nasal (HNE) and tracheal (HTE) epithelial cell cultures were infected with HCoV-229E. RESULTS: Pretreatment of HNE and HTE cells with glycopyrronium or formoterol decreased viral RNA levels and/or titers, the expression of the HCoV-229E receptor CD13, the number and fluorescence intensity of acidic endosomes where HCoV-229E RNA enters the cytoplasm, and the infection-induced production of cytokines, including IL-6, IL-8, and IFN-ß. Treatment of the cells with the CD13 inhibitor 2'2'-dipyridyl decreased viral titers. Pretreatment of the cells with a combination of three drugs (glycopyrronium, formoterol, and budesonide) exerted additive inhibitory effects on viral titers and cytokine production. Pretreatment of HNE cells with glycopyrronium or formoterol reduced the susceptibility to infection, and pretreatment with the three drugs inhibited activation of nuclear factor-kappa B p50 and p65 proteins. Pretreatment with formoterol increased cAMP levels and treatment with cAMP decreased viral titers, CD13 expression, and the fluorescence intensity of acidic endosomes. CONCLUSIONS: These findings suggest that glycopyrronium, formoterol, and a combination of glycopyrronium, formoterol, and budesonide inhibit HCoV-229E replication partly by inhibiting receptor expression and/or endosomal function and that these drugs modulate infection-induced inflammation in the airway.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Budesonide/pharmacology , Coronavirus/physiology , Cytokines/metabolism , Epithelial Cells/metabolism , Epithelial Cells/virology , Formoterol Fumarate/pharmacology , Glycopyrrolate/pharmacology , Muscarinic Antagonists/pharmacology , Nasal Mucosa/cytology , Trachea/cytology , Virus Replication/drug effects , CD13 Antigens/metabolism , Cells, Cultured , Humans
9.
Intern Med ; 59(2): 181-192, 2020.
Article in English | MEDLINE | ID: mdl-31941868

ABSTRACT

Objective Pneumonia develops in bedridden patients, even in those receiving oral care, and malnutrition is associated with the development of pneumonia. We examined the effects of nutritional treatment on the prevention of pneumonia. Patients and Methods We retrospectively examined the effects of nutritional treatment on the prevention of pneumonia by analyzing the records of bedridden patients (n=68; mean age: 68.0 years) who stayed in a hospital for 2 years or longer. Results Among the analyzed patients, pneumonia developed in 52 (76%) patients, and the mean frequency of pneumonia was 1.6 times per year during the first year of stay. In a multivariate analysis, the serum albumin level at admission in the pneumonia group was lower than that in the non-pneumonia group. The frequency of pneumonia during the second year of stay was lower than that during the first year of stay. Serum levels of albumin and total protein (TP) at one year after admission were higher than those at admission in all analyzed patients, and in all patients (n=52) and elderly (≥65 years) patients (n=31) in the pneumonia group. The proportions of patients with hypoalbuminemia (<3.5 g/dL) and hypoproteinemia (<6.5 g/dL) at one year after admission were lower than those at admission. The increases in the proportions of patients presenting a reduced frequency of pneumonia were correlated with increases in the proportions of patients presenting increased levels of albumin and/or TP. Conclusion Nutritional treatment may reduce the frequency of pneumonia by improving malnutrition in bedridden patients receiving oral care.


Subject(s)
Bedridden Persons , Malnutrition/prevention & control , Nutritional Support/methods , Pneumonia, Bacterial/prevention & control , Aged , Female , Hospitalization , Humans , Hypoalbuminemia/etiology , Hypoproteinemia/etiology , Male , Malnutrition/diet therapy , Multivariate Analysis , Pneumonia, Bacterial/diet therapy , Retrospective Studies
10.
Arch Pharm Res ; 43(5): 526-539, 2020 May.
Article in English | MEDLINE | ID: mdl-28861755

ABSTRACT

Rhinoviral infection is associated with an increased risk of asthma attacks. The macrolide clarithromycin decreases cytokine production in nasopharyngeal aspirates from patients with wheezing, but the effects of macrolides on cytokine production in nasal epithelial cells obtained from asthmatic subjects remain unclear. Here, human nasal epithelial cells were infected with type-14 rhinovirus (RV14), a major RV group. Titers and RNA of RV14 and cytokine concentrations, including IL-1ß and IL-6, were higher in the supernatants of the cells obtained from subjects with bronchial asthma (asthmatic group) than in those from the non-asthmatic group. Pretreatment with clarithromycin decreased RV14 titers, viral RNA and cytokine concentrations, and susceptibility to RV14 infection. Pretreatment with clarithromycin also decreased IL-33 production, which was detected after infection. Pretreatment with clarithromycin decreased the expression of intercellular adhesion molecule-1, the receptor for RV14, after infection, the number and fluorescence intensity of the acidic endosomes through which RV RNA enters the cytoplasm, and the activation of nuclear factor kappa-B proteins in nuclear extracts. These findings suggested that RV replication and cytokine production may be enhanced in nasal epithelial cells obtained from subjects with bronchial asthma and may be modulated by clarithromycin.


Subject(s)
Antiviral Agents/pharmacology , Asthma/drug therapy , Clarithromycin/pharmacology , Cytokines/biosynthesis , Epithelial Cells/drug effects , Rhinovirus/drug effects , Asthma/metabolism , Cells, Cultured , Epithelial Cells/metabolism , Female , Humans , Interleukin-33/antagonists & inhibitors , Interleukin-33/biosynthesis , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Interleukin-8/antagonists & inhibitors , Interleukin-8/biosynthesis , Male , Middle Aged , Virus Replication/drug effects
11.
Tohoku J Exp Med ; 247(4): 215-222, 2019 04.
Article in English | MEDLINE | ID: mdl-30971638

ABSTRACT

Carbon monoxide (CO) and nitric oxide (NO) exhibit physiological properties that include the activation of guanylate cyclase. NO inhibits replication of rhinovirus (RV), a major cause of the common cold and exacerbation of bronchial asthma and chronic obstructive pulmonary disease. However, the anti-rhinoviral effects of CO remain unclear. This study investigated whether the exogenous application of low-dose CO could inhibit RV replication in human alveolar and airway epithelial cells. A549 human lung carcinoma cells with alveolar epithelial features and primary cultures of human tracheal epithelial (HTE) cells were pretreated with CO (100 ppm) and infected with a major group RV, type 14 RV (RV14). CO exposure reduced RV14 titers in the supernatants and RV RNA levels in A549 and HTE cells. The treatment with a guanylate cyclase inhibitor, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, reversed the inhibitory effects of CO exposure on RV14 replication in A549 cells. Pretreatment of A549 cells with 8-Br-cGMP, a cell-permeable cGMP analog, caused the decrease in RV14 replication, while CO exposure increased cGMP production. CO exposure also increased the expression levels of interferon (IFN)-γ mRNA and protein. In contrast, pretreatment with CO did not increase DNA fragmentation and did not reduce the expression of intercellular adhesion molecule-1, the RV14 receptor, or the number of acidic endosomes, through which RV RNA enters the cytoplasm. These findings suggest that low-dose CO may decrease RV14 replication in alveolar and airway epithelial cells. IFN-γ production, which is induced by CO exposure via guanylate cyclase activation-mediated cGMP production, may be involved in RV14 replication inhibition.


Subject(s)
Carbon Monoxide/pharmacology , Epithelial Cells/virology , Pulmonary Alveoli/virology , Rhinovirus/physiology , Virus Replication/drug effects , A549 Cells , Acids , Cyclic GMP/antagonists & inhibitors , Cyclic GMP/biosynthesis , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Endosomes/drug effects , Endosomes/metabolism , Epithelial Cells/drug effects , Guanylate Cyclase/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/biosynthesis , Pulmonary Alveoli/drug effects , Rhinovirus/drug effects
12.
Heliyon ; 5(2): e01149, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30839917

ABSTRACT

High temperature reduces influenza viral replication; however, the treatment of fevers is thought to be necessary to improve patients' conditions. We examined the effects of high temperature on viral replication and infection-induced damage to human tracheal epithelial cells. Cell viability and dome formation were reduced, the number of detached cells was increased and lactate dehydrogenase (LDH) levels tended to be increased from 72 h to 120 h in uninfected cells cultured at 40 °C. Long-term (72 h and/or 120 h) exposure to high temperatures (39 °C and/or 40 °C) decreased RNA levels and/or viral titers of eight influenza virus strains. Cell viability and dome formation were reduced, and the number of detached cells and LDH levels were increased to a similar extent after infection with the A/H1N1 pdm 2009 virus at 37 °C and 40 °C. High temperature increased the endosomal pH, where the viral RNA enters the cytoplasm, in uninfected cells. High temperature reduced the production of IL-6, which mediate viral replication processes, and IL-1ß and IL-8 in uninfected and infected cells. Based on these findings, high temperature may cause similar levels of airway cell damage after infection to cells exposed normal temperatures, although high temperature reduces viral replication by affecting the function of acidic endosomes and inhibiting IL-6-mediated processes.

13.
Respir Res ; 19(1): 116, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29895305

ABSTRACT

Figure 2 of this original publication was incorrectly formatted. The updated Fig. 2 is published in this correction article [1].

14.
Article in English | MEDLINE | ID: mdl-29629340

ABSTRACT

Cleavage and activation of hemagglutinin (HA) by trypsin-like proteases in influenza A virus (IAV) are essential prerequisites for its successful infection and spread. In host cells, some transmembrane serine proteases such as TMPRSS2, TMPRSS4 and HAT, along with plasmin in the bloodstream, have been reported to cleave the HA precursor (HA0) molecule into its active forms, HA1 and HA2. Some trypsinogens can also enhance IAV proliferation in some cell types (e.g., rat cardiomyoblasts). However, the precise activation mechanism for this process is unclear, because the expression level of the physiological activator of the trypsinogens, the TMPRSS15 enterokinase, is expected to be very low in such cells, with the exception of duodenal cells. Here, we show that at least two variant enterokinases are expressed in various human cell lines, including A549 lung-derived cells. The exogenous expression of these enterokinases was able to enhance the proliferation of IAV in 293T human kidney cells, but the proliferation was reduced by knocking down the endogenous enterokinase in A549 cells. The enterokinase was able to enhance HA processing in the cells, which activated trypsinogen in vitro and in the IAV-infected cells also. Therefore, we conclude that enterokinase plays a role in IAV infection and proliferation by activating trypsinogen to process viral HA in human cell lines.


Subject(s)
Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/enzymology , Trypsin/metabolism , Cell Line , Enzyme Activation , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/genetics , Influenza, Human/virology , Protein Processing, Post-Translational , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Trypsin/genetics
15.
Respir Res ; 19(1): 52, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29587772

ABSTRACT

BACKGROUND: Interleukin-33 (IL-33) is a cytokine belonging to the IL-1 family, and its possible involvement in the pathophysiology of COPD and viral-induced exacerbations has been demonstrated. IL-33 has been shown to be increased in the airway epithelial cells from COPD patients, but the regulating mechanism of IL-33 expression in airway epithelial cells remains largely unknown. In the current study, we examined whether oxidative stress, which participates in the pathogenesis of COPD, affects the expression of IL-33 in airway epithelial cells and also evaluated the effect during viral infection. METHODS: The involvement of oxidative stress in the expression of IL-33, and its signal pathway was examined after stimulation with hydrogen peroxide (H2O2), with or without stimulation by polyinosinic-polycytidylic acid [poly (I:C)], a synthetic analogue of dsRNA that mimics viral infection, or rhinovirus infection in NCI-H292 cells and primary human bronchial epithelial cells (HBECs). In addition, the effect of antioxidant, N-acetylcysteine (NAC) in the expression of IL-33 was compared between HBECs from healthy subjects and those from COPD patients. RESULTS: Treatment with H2O2 significantly potentiated IL-33 expression in NCI-H292 cells, and the potentiation was reversed by NAC treatment. Mitogen-activated protein kinase (MAPK) inhibitors, but not nuclear factor-kappa B inhibitors, also significantly decreased the H2O2-potentiated IL-33 expression. In addition, H2O2 significantly potentiated the poly (I:C)- or rhinovirus-stimulated IL-33 expression. In HBECs from healthy subjects, H2O2-potentiated IL-33 expression and its reversal by NAC was also confirmed. Under the condition without H2O2-stimulation, treatment with NAC significantly decreased the expression of IL-33 in HBECs from COPD patients, but not in those from healthy subjects. CONCLUSIONS: These results demonstrate that oxidative stress involves in the expression of IL-33 in airway epithelial cells via MAPK signal pathway and it augments IL-33 expression during viral infection. This mechanism may participate in the regulation of IL-33 expression in airway epithelial cells in COPD and the viral-induced exacerbations. Modulation of this pathway could become a therapeutic target for viral-induced exacerbations of COPD.


Subject(s)
Interleukin-33/biosynthesis , Oxidative Stress/physiology , Respiratory Mucosa/metabolism , Aged , Antiviral Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Female , Gene Expression , Humans , Hydrogen Peroxide/toxicity , Interleukin-33/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Middle Aged , Oxidative Stress/drug effects , Poly I-C/toxicity , Respiratory Mucosa/drug effects , Respiratory Mucosa/virology , Rhinovirus/drug effects , Rhinovirus/physiology
16.
Geriatr Gerontol Int ; 18(5): 714-722, 2018 May.
Article in English | MEDLINE | ID: mdl-29380508

ABSTRACT

AIM: Pneumonia develops in bedridden patients even when they are receiving oral care. However, the pneumonia risk in bedridden patients remains unclear, and no screening tool has been developed to assess this risk by using daily hospital data. METHODS: We retrospectively examined pneumonia risk factors by analyzing the records of 102 bedridden patients receiving oral care. RESULTS: Body mass index, peripheral blood hemoglobin, and serum concentrations of total protein, albumin, total cholesterol and uric acid in the pneumonia group (n = 51; mean age 73.4 years) were lower than those in the non-pneumonia group (n = 51; mean age 68.1 years). In the univariate analysis, body mass index; leukocytosis; high C-reactive protein; low levels of hemoglobin, total protein and albumin (<3.5 g/dL); and urine bacteria were associated with the development of pneumonia. Furthermore, in the multivariate analysis, low levels of albumin and urine bacteria were independently associated with pneumonia. We developed a bedridden patient pneumonia risk (BPPR) score using these two risk factors to assess pneumonia risk. We applied scores of zero (0) or one (1) according to the absence or presence of the two risk factors and summed the scores in each patient. The proportion of pneumonia patients increased with increasing BPPR score when the patients were divided into three groups - low, moderate and high risk - according to the BPPR score (0, 1 or 2, respectively). CONCLUSIONS: Malnutrition, urinary tract infection-induced inflammation and anemia were associated with pneumonia in bedridden patients. BPPR scoring might be useful for assessing pneumonia risk and managing affected patients. Geriatr Gerontol Int 2018; 18: 714-722.


Subject(s)
Bedridden Persons/statistics & numerical data , Dental Care/statistics & numerical data , Mass Screening/methods , Pneumonia/epidemiology , Aged , Humans , Inflammation/etiology , Malnutrition/epidemiology , Retrospective Studies , Risk Factors , Urinary Tract Infections/complications
17.
Immun Inflamm Dis ; 4(2): 166-181, 2016 06.
Article in English | MEDLINE | ID: mdl-27957326

ABSTRACT

Increased viral replication and cytokine production may be associated with the pathogenesis of asthma attacks in rhinovirus (RV) infections. However, the association between increased RV replication and enhanced expression of intercellular adhesion molecule-1 (ICAM-1), a receptor for a major RV group, in airway epithelial cells has remained unclear. Furthermore, the inhibitory effects of mucolytics, which have clinical benefits in asthmatic subjects, are uncertain. Human nasal epithelial (HNE) cells were infected with type 14 rhinovirus (RV14), a major RV group. RV14 titers and cytokine concentrations, including interleukin (IL)-6 and IL-8, in supernatants, RV14 RNA replication and susceptibility to RV14 infection were higher in HNE cells obtained from subjects in the allergic group (allergic subjects) than in those from subjects in the non-allergic group (non-allergic subjects). ICAM-1 expression and the number and fluorescence intensity of acidic endosomes from which RV14 RNA enters the cytoplasm were higher in HNE cells from allergic subjects, though substantial amounts of interferon (IFN)-γ and IFN-λ were not detected in the supernatant. The abundance of p50 and p65 subunits of transcription factor nuclear factor kappa B (NF-κB) in nuclear extracts of the cells from allergic subjects was higher compared to non-allergic subjects, and an inhibitor of NF-κB, caffeic acid phenethyl ester, reduced the fluorescence intensity of acidic endosomes as well as RV titers and RNA. Furthermore, a mucolytic agent, L-carbocisteine, reduced RV14 titers and RNA levels, cytokine release, ICAM-1 expression, the fluorescence intensity of acidic endosomes, and NF-κB activation. The increased RV14 replication observed in HNE cells from allergic subjects might be partly associated with enhanced ICAM-1 expression and decreased endosomal pH through NF-κB activation. L-Carbocisteine inhibits RV14 infection by reducing ICAM-1 and acidic endosomes and may, therefore, modulate airway inflammation caused by RV infection in allergic subjects.


Subject(s)
Hypersensitivity/virology , Intercellular Adhesion Molecule-1/metabolism , Nasal Mucosa/immunology , Rhinovirus/immunology , Cells, Cultured , Humans , Nasal Mucosa/metabolism , RNA, Viral , Trachea , Virus Replication
18.
Respir Res ; 17(1): 121, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27677339

ABSTRACT

BACKGROUND: In response to tissue damage or inflammation, adenosine-5'-triphosphate (ATP) is released into the extracellular compartment and has been demonstrated to augment inflammation via purinergic P2 receptors (P2Rs). Recently, ATP has been shown to be increased in the airways of COPD patients. In the present study, we examined the possible involvement of extracellular ATP in airway mucus hypersecretion during viral-induced COPD exacerbations. METHODS: The involvement of extracellular ATP in the release of a major airway mucin, MUC5AC, and its signal pathway was examined after stimulation with polyinosine-polycytidylic acid [poly(I:C)], a synthetic analog of dsRNA to mimic viral infection, and rhinovirus (RV) infection in NCI-H292 cells and differentiated airway epithelial cells from COPD patients. RESULTS: Treatment with poly(I:C) significantly increased the amount of extracellular ATP and induced MUC5AC release in NCI-H292 cells. Pre-treatment with a pannexin channel inhibitor, carbenoxolone (CBX), reduced the amount of extracellular ATP and suppressed MUC5AC release from poly(I:C)-treated cells. Pre-treatment with the P2R antagonist suramin significantly reduced the expression and release of MUC5AC. The inhibitory effects of CBX and suramin on the release of ATP and/or MUC5AC were replicated with RV infection. Pre-treatment with suramin also significantly reduced the expression and amount of extracellular EGFR ligands and the phosphorylation of EGFR and ERK in poly(I:C)-treated cells. In addition, pre-treatment with a P2Y2 receptor siRNA significantly suppressed the poly(I:C)-potentiated EGFR ligands and MUC5AC release. After poly(I:C) stimulation, the expression of MUC5AC in the differentiated cells from COPD patients was significantly higher than those from healthy subjects and the values of MUC5AC expression were inversely related with forced expiratory volume in 1 s (FEV1) % predicted. The inhibitory effects of CBX and suramin on poly(I:C)-potentiated MUC5AC expression were confirmed in differentiated airway epithelium from COPD patients. CONCLUSIONS: These results demonstrate that dsRNA induces the release of ATP via pannexin channel and that the extracellular ATP is involved in the expression and release of MUC5AC, mainly via P2Y2R, in an autocrine manner. Modulation of this pathway could be a therapeutic target for viral-induced mucus hypersecretion in COPD exacerbations.

19.
Nihon Rinsho ; 74(5): 833-8, 2016 May.
Article in Japanese | MEDLINE | ID: mdl-27254955

ABSTRACT

Mucolytic agents and macrolides have been used in the treatment of patients with chronic obstructive pulmonary disease (COPD). These drugs improve symptoms, such as sputum, and quality of life in COPD patients and reduce the frequency of COPD exacerbation. Mucolytic agents have various biological effects, such as reduction of mucin production, improvement of goblet cell hyperplasia and mucociliary transport, reduction of airway inflammation, and anti-oxidant and anti-viral effects. Similarly, in addition to antimicrobial effects, macrolides have biological effects, including reduction of mucin production and airway inflammation induced by airway infection, improvement of mucociliary transport, and anti-bacterial and anti-viral effects. These biological effects may be associated with clinical benefits of mucolvtic agents and macrolides in the treatment of COPD patients.


Subject(s)
Expectorants/therapeutic use , Macrolides/therapeutic use , Pulmonary Disease, Chronic Obstructive/drug therapy , Anti-Bacterial Agents , Anti-Inflammatory Agents, Non-Steroidal , Antioxidants , Antiviral Agents , Clarithromycin/pharmacology , Clarithromycin/therapeutic use , Disease Progression , Drug Therapy, Combination , Erythromycin/pharmacology , Erythromycin/therapeutic use , Expectorants/pharmacology , Goblet Cells/pathology , Humans , Hyperplasia/drug therapy , Macrolides/pharmacology , Mucins/metabolism , Mucociliary Clearance/drug effects , Prospective Studies , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Quality of Life , Retrospective Studies , Treatment Outcome
20.
Respir Investig ; 54(2): 98-108, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26879479

ABSTRACT

BACKGROUND: Receptor for advanced glycation end products (RAGE) is abundantly expressed on alveolar epithelial cells (AECs) and participates in innate immune responses such as apoptosis and inflammation. However, it is unclear whether RAGE-mediated apoptosis of AECs is associated with hyperoxia-induced lung injury. METHODS: We used wild-type and RAGE-knockout C57BL6/J mice in this study. In addition, we developed bone marrow chimeric mouse models expressing RAGE on hematopoietic or non-hematopoietic cells, including lung parenchymal cells, and compared survival ratios and changes in the permeability of the alveolar-capillary barrier after hyperoxia exposure. Further, we prepared single cell suspensions of lung cells and evaluated the apoptosis of AECs or microvascular endothelial cells (MVECs) by using a combination of antibodies and JC-1 dye. We also examined whether RAGE inhibition decreased hyperoxia-induced apoptosis of human lung epithelial cells in vitro. RESULTS: After hyperoxia exposure, mice expressing RAGE on lung cells showed lower survival rate and increased alveolar-capillary permeability than mice expressing RAGE on hematopoietic cells. RAGE-expressing AECs showed significantly higher apoptosis than RAGE-knockout AECs after in vivo hyperoxia exposure. The level of hyperoxia-induced apoptosis was not different in MVECs. However, RAGE-null lung epithelial cells showed lower apoptosis than RAGE-expressing cells in vitro. CONCLUSION: These results indicated that RAGE on AECs mainly contributed to hyperoxia-induced lung injury and alveolar-capillary barrier disruption.


Subject(s)
Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Hypoxia/complications , Lung Injury/etiology , Lung Injury/pathology , Receptor for Advanced Glycation End Products/metabolism , Animals , Apoptosis , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Male , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL