Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Virol ; 95(21): e0129621, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34406864

RESUMO

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.


Assuntos
Gammainfluenzavirus/enzimologia , Gammainfluenzavirus/metabolismo , Hemaglutininas Virais/metabolismo , Influenza Humana/virologia , Infecções por Orthomyxoviridae/virologia , Serina Endopeptidases/metabolismo , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Antivirais/farmacologia , Benzamidinas/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Cães , Ésteres/farmacologia , Guanidinas/farmacologia , Interações entre Hospedeiro e Microrganismos , Humanos , Células Madin Darby de Rim Canino , Tripsina/metabolismo , Proteínas Virais/metabolismo
2.
J Med Virol ; 93(6): 3484-3495, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33247612

RESUMO

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.


Assuntos
Benzamidinas/farmacologia , Benzamidinas/uso terapêutico , Células Epiteliais/efeitos dos fármacos , Guanidinas/farmacologia , Guanidinas/uso terapêutico , Pulmão/efeitos dos fármacos , Inibidores de Serina Proteinase/farmacologia , Inibidores de Serina Proteinase/uso terapêutico , Replicação Viral/efeitos dos fármacos , Animais , Células Cultivadas , Citocinas/análise , Citocinas/imunologia , Células Epiteliais/virologia , Feminino , Humanos , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nariz/citologia , Traqueia/citologia
3.
Tohoku J Exp Med ; 251(1): 27-30, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32448818

RESUMO

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.


Assuntos
Antivirais/farmacologia , Coronavirus Humano 229E/efeitos dos fármacos , Infecções por Coronavirus/tratamento farmacológico , Gabexato/análogos & derivados , Guanidinas/farmacologia , Pneumonia Viral/tratamento farmacológico , Inibidores de Proteases/farmacologia , Replicação Viral/efeitos dos fármacos , Benzamidinas , Betacoronavirus/efeitos dos fármacos , COVID-19 , Células Cultivadas , Coronavirus Humano 229E/enzimologia , Coronavirus Humano 229E/fisiologia , Meios de Cultivo Condicionados , Células Epiteliais/virologia , Ésteres , Gabexato/farmacologia , Humanos , Mucosa Nasal/citologia , Pandemias , Cultura Primária de Células , SARS-CoV-2 , Serina Endopeptidases/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Carga Viral
4.
Tohoku J Exp Med ; 247(4): 215-222, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30971638

RESUMO

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.


Assuntos
Monóxido de Carbono/farmacologia , Células Epiteliais/virologia , Alvéolos Pulmonares/virologia , Rhinovirus/fisiologia , Replicação Viral/efeitos dos fármacos , Células A549 , Ácidos , GMP Cíclico/antagonistas & inibidores , GMP Cíclico/biossíntese , Fragmentação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Células Epiteliais/efeitos dos fármacos , Guanilato Ciclase/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Interferon gama/biossíntese , Alvéolos Pulmonares/efeitos dos fármacos , Rhinovirus/efeitos dos fármacos
5.
Respir Res ; 19(1): 116, 2018 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-29895305

RESUMO

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

6.
Respir Res ; 19(1): 52, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29587772

RESUMO

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.


Assuntos
Interleucina-33/biossíntese , Estresse Oxidativo/fisiologia , Mucosa Respiratória/metabolismo , Idoso , Antivirais/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Feminino , Expressão Gênica , Humanos , Peróxido de Hidrogênio/toxicidade , Interleucina-33/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo/efeitos dos fármacos , Poli I-C/toxicidade , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/virologia , Rhinovirus/efeitos dos fármacos , Rhinovirus/fisiologia
7.
Respir Res ; 17(1): 121, 2016 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-27677339

RESUMO

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.

8.
J Pathol ; 235(4): 632-45, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25348279

RESUMO

Chronic inflammation induces lymphangiogenesis and blood vessel remodelling. Since aged pneumonia patients often have repeated episodes of aspiration pneumonia, the pathogenesis may involve chronic inflammation. For lymphangiogenesis, VEGFR-3 and its ligand VEGF-C are key factors. No previous studies have examined chronic inflammation or vascular changes in aspiration pneumonia or its mouse models. In lung inflammation, little is known about the effect of blocking VEGFR-3 on lung lymphangiogenesis and, moreover, its effect on the disease condition. This study aimed to establish a mouse model of aspiration pneumonia, examine the presence of chronic inflammation and vascular changes in the model and in patients, and evaluate the effect of inhibiting VEGFR-3 on the lymphangiogenesis and disease condition in this model. To induce aspiration pneumonia, we repeated inoculation of pepsin at low pH and LPS into mice for 21-28 days, durations in which bronchioalveolar lavage and plasma leakage in the lung suggested the presence of exaggerated inflammation. Conventional and immunohistochemical analysis of tracheal whole mounts suggested the presence of chronic inflammation, lymphangiogenesis, and blood vessel remodelling in the model. Quantitative RT-PCR of the trachea and lung suggested the involvement of lymphangiogenic factor VEGF-C, VEGFR-3, and pro-inflammatory cytokines. In the lung, the aspiration model showed the presence of chronic inflammation and exaggerated lymphangiogenesis. Treatment with the VEGFR inhibitor axitinib or the VEGFR-3 specific inhibitor SAR131675 impaired lymphangiogenesis in the lung and improved oxygen saturation in the aspiration model. Since the lung is the main site of aspiration pneumonia, the changes were intensive in the lung and mild in the trachea. Human lung samples also showed the presence of chronic inflammation and exaggerated lymphangiogenesis, suggesting the relevance of the model to the disease. These results suggest lymphatics in the lung as a new target of analysis and therapy in aspiration pneumonia.


Assuntos
Imidazóis/farmacologia , Indazóis/farmacologia , Pulmão/efeitos dos fármacos , Linfangiogênese/efeitos dos fármacos , Vasos Linfáticos/efeitos dos fármacos , Naftiridinas/farmacologia , Pneumonia Aspirativa/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Autopsia , Axitinibe , Doença Crônica , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/metabolismo , Pulmão/enzimologia , Pulmão/imunologia , Pulmão/fisiopatologia , Vasos Linfáticos/enzimologia , Vasos Linfáticos/imunologia , Vasos Linfáticos/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Pneumonia Aspirativa/enzimologia , Pneumonia Aspirativa/genética , Pneumonia Aspirativa/imunologia , Pneumonia Aspirativa/fisiopatologia , Fatores de Tempo , Fator C de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
9.
Nihon Rinsho ; 74(5): 833-8, 2016 May.
Artigo em Japonês | MEDLINE | ID: mdl-27254955

RESUMO

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.


Assuntos
Expectorantes/uso terapêutico , Macrolídeos/uso terapêutico , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Antibacterianos , Anti-Inflamatórios não Esteroides , Antioxidantes , Antivirais , Claritromicina/farmacologia , Claritromicina/uso terapêutico , Progressão da Doença , Quimioterapia Combinada , Eritromicina/farmacologia , Eritromicina/uso terapêutico , Expectorantes/farmacologia , Células Caliciformes/patologia , Humanos , Hiperplasia/tratamento farmacológico , Macrolídeos/farmacologia , Mucinas/metabolismo , Depuração Mucociliar/efeitos dos fármacos , Estudos Prospectivos , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Qualidade de Vida , Estudos Retrospectivos , Resultado do Tratamento
10.
J Med Virol ; 87(1): 25-34, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24838828

RESUMO

Defining the effects of neuraminidase inhibitors on influenza virus infection may provide important information for the treatment of patients. The effects of neuraminidase inhibitors have been examined using various methods, including viral release from kidney cells. However, the effects of neuraminidase inhibitors on viral release from primary cultures of human tracheal epithelial cells, which retain functions of the original tissues, have not been studied. The effects of neuraminidase inhibitors on the replication of the pandemic influenza virus [A/Sendai-H/N0633/2009 (H1N1) pdm09] and the seasonal influenza virus [A/Sendai-H/216/2009 (H1N1)] that was isolated during the 2008-2009 season were examined. The virus stocks were generated by infecting tracheal cells with the pandemic or seasonal influenza virus. Four types of inhibitors (oseltamivir, zanamivir, laninamivir, and peramivir) reduced pandemic viral titers and concentrations of the cytokines interleukin-6 and tumor necrosis factor-α in supernatants and viral RNA in cells. However, oseltamivir did not reduce seasonal viral titers, cytokine concentrations and viral RNA, and the 50% inhibitory concentration (IC50 ) of oseltamivir for neuraminidase activity in the seasonal virus was 300-fold higher than that observed for the pandemic influenza virus. The seasonal influenza virus had an oseltamivir-resistant genotype. The magnitude of the IC50 values of the neuraminidase inhibitors for the seasonal influenza virus was inversely related to the magnitude of the inhibitory effects on viral release. These methods for measuring the release of virus and inflammatory cytokines from primary cultures of human tracheal epithelium may provide useful information regarding the effects of neuraminidase inhibitors on influenza viruses.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Oseltamivir/farmacologia , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Feminino , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Concentração Inibidora 50 , Masculino , Mucosa Respiratória , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
11.
Pulm Pharmacol Ther ; 33: 66-74, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26166259

RESUMO

BACKGROUND: Serine proteases act through the proteolytic cleavage of the hemagglutinin (HA) of influenza viruses for the entry of influenza virus into cells, resulting in infection. However, the inhibitory effects of serine protease inhibitors on influenza virus infection of human airway epithelial cells, and on their production of inflammatory cytokines are unclear. METHODS: Primary cultures of human tracheal epithelial cells were treated with four types of serine protease inhibitors, including camostat, and infected with A/Sendai-H/108/2009/(H1N1) pdm09 or A/New York/55/2004(H3N2). RESULTS: Camostat reduced the amounts of influenza viruses in the supernatants and viral RNA in the cells. It reduced the cleavage of an influenza virus precursor protein, HA0, into the subunit HA1. Camostat also reduced the concentrations of the cytokines interleukin (IL)-6 and tumor necrosis factor (TNF)-α in the supernatants. Gabexate and aprotinin reduced the viral titers and RNA levels in the cells, and aprotinin reduced the concentrations of TNF-α in the supernatants. The proteases transmembrane protease serine S1 member (TMPRSS) 2 and HAT (human trypsin-like protease: TMPRSS11D), which are known to cleave HA0 and to activate the virus, were detected at the cell membrane and in the cytoplasm. mRNA encoding TMPRSS2, TMPRSS4 and TMPRSS11D was detectable in the cells, and the expression levels were not affected by camostat. CONCLUSIONS: These findings suggest that human airway epithelial cells express these serine proteases and that serine protease inhibitors, especially camostat, may reduce influenza viral replication and the resultant production of inflammatory cytokines possibly through inhibition of activities of these proteases.


Assuntos
Gabexato/análogos & derivados , Influenza Humana/tratamento farmacológico , Inibidores de Serina Proteinase/farmacologia , Replicação Viral/efeitos dos fármacos , Idoso , Animais , Aprotinina/farmacologia , Células Cultivadas , Cães , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Ésteres , Feminino , Gabexato/farmacologia , Guanidinas , Humanos , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Vírus da Influenza A Subtipo H3N2/isolamento & purificação , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Masculino , Pessoa de Meia-Idade , RNA Viral/metabolismo , Traqueia/citologia , Traqueia/virologia
12.
Exp Lung Res ; 41(8): 422-34, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26151196

RESUMO

AIM: Surfactant protein-C (SP-C) of alveolar epithelial type II cells (ATII) plays a key role in maintaining alveolar integrity and repair. Mutations or decreased expression of SFTPC, the gene encoding SP-C, causes ATII injury and aberrant repair of the lung tissue to develop pulmonary fibrosis. Histone deacetylases (HDACs) epigenetically remove acetyl groups from acetylated histones and regulate transcription. HDAC inhibitors attenuated epithelial-to-mesenchymal transition (EMT) and fibrotic disorders. The aim of this study is to investigate whether Trichostatin A (TSA), a pan-HDAC inhibitor, epigenetically exerts a protective effect on ATII against fibrotic changes via the restoration of SFTPC expression. MATERIALS AND METHODS: We treated A549 cells with TGF-ß1 to induce EMT, followed by TSA treatment. We evaluated SFTPC mRNA, histone acetylation levels in the SFTPC gene promoter region, and pro-SP-C protein. C57BL6/J mice were treated with intratracheal bleomycin instillation followed by TSA administration. Histological changes and Sftpc mRNA expression in isolated ATII were evaluated. RESULTS: TGF-ß1 treatment decreased SFTPC mRNA in A549 cells. TSA restored SFTPC mRNA, and increased histone H4 acetylation in the SFTPC promoter region in vitro. The administration of TSA partially attenuated BLM-induced pulmonary fibrosis and increased the Sftpc mRNA expression in isolated ATII from bleomycin-treated lungs in vivo. CONCLUSIONS: Decreased expression of SFTPC by TGF-ß1 treatment was restored by TSA via hyperacetylation of histone H4 in the promoter region. TSA partially attenuated pulmonary fibrosis and increased Sftpc mRNA in ATII. Our findings suggest that the epigenetic restoration of SP-C would be a therapeutic target for pulmonary fibrosis.


Assuntos
Bleomicina/farmacologia , Células Epiteliais/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Alvéolos Pulmonares/efeitos dos fármacos , Fibrose Pulmonar/tratamento farmacológico , Proteína C Associada a Surfactante Pulmonar/metabolismo , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Animais , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Ácidos Hidroxâmicos/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
13.
Tohoku J Exp Med ; 237(1): 45-50, 2015 09.
Artigo em Inglês | MEDLINE | ID: mdl-26346967

RESUMO

Ebola virus disease (EVD) has been a great concern worldwide because of its high mortality. EVD usually manifests with fever, diarrhea and vomiting, as well as disseminated intravascular coagulation (DIC). To date, there is neither a licensed Ebola vaccine nor a promising therapeutic agent, although clinical trials are ongoing. For replication inside the cell, Ebola virus (EBOV) must undergo the proteolytic processing of its surface glycoprotein in the endosome by proteases including cathepsin B (CatB), followed by the fusion of the viral membrane and host endosome. Thus, the proteases have been considered as potential targets for drugs against EVD. However, no protease inhibitor has been presented as effective clinical drug against it. A synthetic serine protease inhibitor, nafamostat mesilate (NM), reduced the release of CatB from the rat pancreas. Furthermore, it has anticoagulant activities, such as inhibition of the factor VIIa complex, and has been used for treating DIC in Japan. Thus, NM could be considered as a drug candidate for the treatment of DIC induced by EBOV infection, as well as for the possible CatB-related antiviral action. Moreover, the drug has a history of large-scale production and clinical use, and the issues of safety and logistics might have been cleared. We advocate in vitro and in vivo experiments using active EBOV to examine the activities of NM against the infection and the DIC induced by the infection. In addition, we suggest trials for comparison among anti-DIC drugs including the NM in EVD patients, in parallel with the experiments.


Assuntos
Antivirais/uso terapêutico , Ebolavirus/efeitos dos fármacos , Guanidinas/uso terapêutico , Doença pelo Vírus Ebola/tratamento farmacológico , Inibidores de Serina Proteinase/uso terapêutico , Animais , Benzamidinas , Coagulação Intravascular Disseminada/tratamento farmacológico , Coagulação Intravascular Disseminada/etiologia , Ebolavirus/enzimologia , Fator VIIa/antagonistas & inibidores , Doença pelo Vírus Ebola/complicações , Humanos , Hiperpotassemia/etiologia , Hiperpotassemia/terapia , Hiponatremia/etiologia , Hiponatremia/terapia , Ratos , Serina Proteases/metabolismo , Replicação Viral/efeitos dos fármacos
14.
Am J Physiol Lung Cell Mol Physiol ; 306(9): L855-65, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24583878

RESUMO

Expression of c-Kit and its ligand, stem cell factor (SCF), in developing human lung tissue was investigated by immunohistochemistry. Twenty-eight human fetal lungs [age range 13 to 38 gestational wk (GW)] and 12 postnatal lungs (age range 1-79 yr) were evaluated. We identified c-Kit(+) cells in the lung mesenchyme as early as 13 GW. These mesenchymal c-Kit(+) cells in the lung did not express mast cell tryptase or α-smooth muscle actin. However, these cells did express CD34, VEGFR2, and Tie-2, indicating their endothelial lineage. Three-dimensional reconstructions of confocal laser scanning images revealed that c-Kit(+) cells displayed a closed-end tube formation that did not contain hematopoietic cells. From the pseudoglandular phase to the canalicular phase, c-Kit(+) cells appeared to continuously proliferate, to connect with central pulmonary vessels, and finally, to develop the lung capillary plexus. The spatial distribution of c-Kit- and SCF-positive cells was also demonstrated, and these cells were shown to be in close association. Our results suggest that c-Kit expression in early fetal lungs marks a progenitor population that is restricted to endothelial lineage. This study also suggests the potential involvement of c-Kit signaling in lung vascular development.


Assuntos
Endotélio Vascular/metabolismo , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Pulmão/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células-Tronco/metabolismo , Adolescente , Idoso , Células Cultivadas , Criança , Pré-Escolar , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Humanos , Técnicas Imunoenzimáticas , Lactente , Pulmão/citologia , Pulmão/imunologia , Microscopia Confocal , Microscopia de Fluorescência , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-kit/imunologia , Células-Tronco/citologia , Células-Tronco/imunologia
15.
EMBO Rep ; 12(4): 358-64, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21399623

RESUMO

Clearance of apoptotic cells is necessary for tissue development, homeostasis and resolution of inflammation. The uptake of apoptotic cells is initiated by an 'eat-me' signal, such as phosphatidylserine, on the cell surface and phagocytes recognize the signal by using specific receptors. In this study, we show that the soluble form of the receptor for advanced glycation end products (RAGE) binds to phosphatidylserine as well as to the apoptotic thymocytes. RAGE-deficient (Rage(-/-)) alveolar macrophages showed impaired phagocytosis of apoptotic thymocytes and defective clearance of apoptotic neutrophils in Rage(-/-) mice. Our results indicate that RAGE functions as a phosphatidylserine receptor and assists in the clearance of apoptotic cells.


Assuntos
Fagocitose/fisiologia , Fosfatidilserinas/metabolismo , Receptores Imunológicos/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Citometria de Fluxo , Transferência Ressonante de Energia de Fluorescência , Macrófagos/metabolismo , Camundongos , Camundongos Mutantes , Microscopia Confocal , Fagocitose/genética , Fosfatidilserinas/genética , Ligação Proteica/genética , Ligação Proteica/fisiologia , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Ressonância de Plasmônio de Superfície
16.
Exp Lung Res ; 39(4-5): 155-61, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23550836

RESUMO

Circulating endothelial microparticles (EMPs) are membrane vesicles that are shed into the blood stream from activated or apoptotic endothelial cells. We previously reported that circulating EMP numbers significantly increased in stable chronic obstructive pulmonary disease (COPD) patients and during exacerbation compared with healthy control subjects. However, different types of circulating EMPs with distinct time profiles were detectable during exacerbations. We hypothesized that the released EMP subtypes correlated with differences in the inflammatory stimuli and the endothelial cell type. We compared the EMP subtypes from human aortic endothelial cells (Aortic ECs) and human lung microvascular endothelial cells (Pulmonary microvascular ECs) released in response to various stimuli, including proinflammatory cytokines (TNFα), oxidative stress (H2O2), and cigarette smoke extracts (CSE) in vitro. We defined circulating EMPs by the expression of endothelial antigens: CD144(+) MPs (VE-cadherin EMPs), CD31(+)/CD41(-) MPs (PECAM EMPs), CD62E(+) MPs (E-selectin EMPs), and CD146(+) MPs (MCAM EMPs). E-selectin EMPs were released from both pulmonary microvascular and aortic ECs in response to TNFα but not to H2O2 or CSE stimulation. The amount of MCAM EMPs released from pulmonary microvascular ECs differed significantly between the cells stimulated with H2O2 and those stimulated with CSE. VE-cadherin EMPs were only released from aortic ECs, whereas PECAM EMPs were released exclusively from pulmonary microvascular ECs. The EMP subtypes released differ in vitro among TNFα, H2O2, and CSE stimulation as well as between pulmonary microvascular and aortic ECs. The differences in circulating EMP subtypes may reflect a condition or site of endothelial injury and may serve as markers for endothelial damage in COPD patients.


Assuntos
Aorta/metabolismo , Micropartículas Derivadas de Células/metabolismo , Células Endoteliais/metabolismo , Pulmão/irrigação sanguínea , Microvasos/metabolismo , Aorta/citologia , Aorta/efeitos dos fármacos , Aorta/imunologia , Biomarcadores/metabolismo , Micropartículas Derivadas de Células/efeitos dos fármacos , Micropartículas Derivadas de Células/imunologia , Células Cultivadas , Citocinas/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Humanos , Mediadores da Inflamação/farmacologia , Potencial da Membrana Mitocondrial , Microvasos/citologia , Microvasos/efeitos dos fármacos , Microvasos/imunologia , Oxidantes/toxicidade , Estresse Oxidativo , Fumaça/efeitos adversos , Fumar/efeitos adversos
17.
Respir Investig ; 61(2): 270-283, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36543714

RESUMO

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.


Assuntos
Asma , Bronquiectasia , COVID-19 , Doença Pulmonar Obstrutiva Crônica , Viroses , Humanos , Qualidade de Vida , Asma/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Viroses/tratamento farmacológico , Anti-Inflamatórios/uso terapêutico , Mucinas/uso terapêutico
18.
Am J Respir Cell Mol Biol ; 46(4): 422-30, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22033268

RESUMO

A variety of lung diseases, such as pulmonary emphysema and idiopathic pulmonary fibrosis, develop in the lung alveoli. Multiple cell types are localized in the alveoli, including epithelial, mesenchymal, and endothelial cells. These resident cells participate in the pathogenesis of lung disease in various ways. To elaborate clearly on the mechanisms of these pathologic processes, cell type-specific analyses of lung disease are required. However, no method exists for individually isolating the different types of cells found in the alveoli. We report on the development of a FACS-based method for the direct isolation of individual cell types from the adult human distal lung. We obtained human lung tissue from lung resections, and prepared single-cell suspension. After depleting CD45-positive cells, a combination of antibodies against epithelial cell adhesion molecule (EpCAM), T1α, and vascular endothelial (VE)-cadherin as used to delineate alveolar cell types. Alveolar Type II cells were highly purified in the EpCAM(hi)/T1α(-) subset, whereas the EpCAM(+)/T1α(-/low) subset contained a mixed epithelial population consisting of alveolar Type I and bronchiolar epithelial cells. The EpCAM(-)/T1α(-) subset included both microvascular endothelial and mesenchymal cells, and these were separated by immunoreactivity to VE-cadherin. Lymphatic endothelial cells existed in the EpCAM(-)/T1α(hi) subset. Isolated cells were viable, and further cell culture studies could be performed. These results suggest that this novel method enables the isolation of different cellular components from normal and diseased lungs, and is capable of elucidating phenotypes specific to certain alveolar cell types indicative of lung disease.


Assuntos
Separação Celular/métodos , Citometria de Fluxo/métodos , Pulmão/citologia , Adulto , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Caderinas/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Molécula de Adesão da Célula Epitelial , Humanos , Pulmão/metabolismo , Glicoproteínas de Membrana , Proteínas de Membrana/metabolismo , Mesoderma/citologia , Alvéolos Pulmonares/citologia
19.
Antimicrob Agents Chemother ; 56(8): 4052-61, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22585227

RESUMO

Respiratory virus infections, including infections with rhinoviruses (RVs), are related to exacerbations of chronic obstructive pulmonary disease (COPD). A new quinolone antibiotic, levofloxacin (LVFX), has been used to treat bacterial infections that cause COPD exacerbations as well as bacterial infections that are secondary to viral infection in COPD patients. However, the inhibitory effects of LVFX on RV infection and RV infection-induced airway inflammation have not been studied. We examined the effects of LVFX on type 14 rhinovirus (RV14) (a major human RV) infection of human tracheal epithelial cells pretreated with LVFX. LVFX pretreatment reduced the RV14 titer, the level of cytokines in the supernatant, the amount of RV14 RNA in the cells after RV14 infection, and the cells' susceptibility to RV14 infection. LVFX pretreatment decreased the mRNA level of intercellular adhesion molecule 1 (ICAM-1), a receptor for RV14, in the cells and the concentration of the soluble form of ICAM-1 in the supernatant before RV14 infection. LVFX pretreatment also decreased the number and the fluorescence intensity of the acidic endosomes from which RV14 RNA enters the cytoplasm. LVFX pretreatment inhibited the activation of nuclear factor κB proteins, including p50 and p65, in nuclear extracts. LVFX pretreatment did not reduce the titers of RV2 (a minor human RV) but reduced the titers of RV15 (a major human RV). These results suggest that LVFX inhibits major-group rhinovirus infections in part by reducing ICAM-1 expression levels and the number of acidic endosomes. LVFX may also modulate airway inflammation in rhinoviral infections.


Assuntos
Molécula 1 de Adesão Intercelular/metabolismo , Levofloxacino , Ofloxacino/farmacologia , Receptores Virais/metabolismo , Mucosa Respiratória/virologia , Rhinovirus/efeitos dos fármacos , Traqueia/virologia , Antibacterianos/farmacologia , Células Cultivadas , Citocinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Humanos , Molécula 1 de Adesão Intercelular/genética , NF-kappa B/metabolismo , Infecções por Picornaviridae/tratamento farmacológico , Infecções por Picornaviridae/virologia , RNA Viral/biossíntese , Mucosa Respiratória/citologia , Rhinovirus/genética , Traqueia/citologia
20.
Thorax ; 67(12): 1067-74, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22843558

RESUMO

RATIONALE: The influence of COPD exacerbation on the endothelium is not completely understood. Circulating endothelial microparticles (EMPs) are membrane vesicles in circulating blood that are shed by activated or apoptotic endothelial cells. OBJECTIVE: To compare EMP numbers in stable COPD patients with those during and after exacerbation. METHODS: We examined the EMP numbers in 80 stable COPD patients, 27 patients with exacerbated COPD, and 20 healthy non-COPD volunteers. EMPs were defined as CD144+ MPs (VE-cadherin EMPs), CD31+/CD41- MPs (PECAM EMPs), CD146 MPs (MCAM EMPs) and CD62E+ EMPs (E-selectin EMPs) as analysed by FACS. Von Willebrand factor (vWF) expression was utilised to identify the origins of the EMPs. RESULTS: VE-cadherin, PECAM and E-selectin EMP numbers were significantly higher in the stable COPD patients than in the non-COPD volunteers, and they were significantly higher in the patients with exacerbated COPD than in the stable COPD patients. The majority of these increased EMPs were vWF-negative, indicating a pulmonary capillary origin. Baseline E-selectin EMP levels were significantly higher in COPD patients who experienced frequent exacerbations than in those who did not have frequent exacerbations (p<0.001). Twenty-eight days after the onset of exacerbation, E-selectin EMP levels returned to those observed in stable COPD patients, whereas PECAM EMP levels remained high. MCAM EMP numbers were not elevated in stable or exacerbated-COPD patients. CONCLUSIONS: Endothelial damage, mainly in pulmonary capillaries, occurs during exacerbation and continues even after clinical symptoms disappear. Higher baseline E-selectin EMP levels may indicate COPD patients who are susceptible to exacerbation.


Assuntos
Biomarcadores/sangue , Micropartículas Derivadas de Células , Endotélio Vascular/metabolismo , Doença Pulmonar Obstrutiva Crônica/sangue , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Idoso , Antígenos CD/sangue , Antígeno CD146/sangue , Caderinas/sangue , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , Selectina E/sangue , Feminino , Humanos , Masculino , Molécula-1 de Adesão Celular Endotelial a Plaquetas/sangue , Glicoproteína IIb da Membrana de Plaquetas/sangue , Análise de Regressão , Testes de Função Respiratória , Fatores de Risco , Estatísticas não Paramétricas , Fator de von Willebrand/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA