Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
PLoS One ; 16(9): e0257784, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34582497

RESUMEN

Drug repurposing has the potential to bring existing de-risked drugs for effective intervention in an ongoing pandemic-COVID-19 that has infected over 131 million, with 2.8 million people succumbing to the illness globally (as of April 04, 2021). We have used a novel `gene signature'-based drug repositioning strategy by applying widely accepted gene ranking algorithms to prioritize the FDA approved or under trial drugs. We mined publically available RNA sequencing (RNA-Seq) data using CLC Genomics Workbench 20 (QIAGEN) and identified 283 differentially expressed genes (FDR<0.05, log2FC>1) after a meta-analysis of three independent studies which were based on severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) infection in primary human airway epithelial cells. Ingenuity Pathway Analysis (IPA) revealed that SARS-CoV-2 activated key canonical pathways and gene networks that intricately regulate general anti-viral as well as specific inflammatory pathways. Drug database, extracted from the Metacore and IPA, identified 15 drug targets (with information on COVID-19 pathogenesis) with 46 existing drugs as potential-novel candidates for repurposing for COVID-19 treatment. We found 35 novel drugs that inhibit targets (ALPL, CXCL8, and IL6) already in clinical trials for COVID-19. Also, we found 6 existing drugs against 4 potential anti-COVID-19 targets (CCL20, CSF3, CXCL1, CXCL10) that might have novel anti-COVID-19 indications. Finally, these drug targets were computationally prioritized based on gene ranking algorithms, which revealed CXCL10 as the common and strongest candidate with 2 existing drugs. Furthermore, the list of 283 SARS-CoV-2-associated proteins could be valuable not only as anti-COVID-19 targets but also useful for COVID-19 biomarker development.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos/métodos , SARS-CoV-2/genética , Antivirales/uso terapéutico , Evaluación Preclínica de Medicamentos/métodos , Células Epiteliales/efectos de los fármacos , Epitelio/efectos de los fármacos , Humanos , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología , Sistema Respiratorio/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad
2.
mBio ; 12(4): e0097021, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34340553

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic has caused significant morbidity and mortality on a global scale. The etiologic agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), initiates host cell entry when its spike protein (S) binds to its receptor, angiotensin-converting enzyme 2 (ACE2). In airway epithelia, the spike protein is cleaved by the cell surface protease TMPRSS2, facilitating membrane fusion and entry at the cell surface. This dependence on TMPRSS2 and related proteases suggests that protease inhibitors might limit SARS-CoV-2 infection in the respiratory tract. Here, we tested two serine protease inhibitors, camostat mesylate and nafamostat mesylate, for their ability to inhibit entry of SARS-CoV-2 and that of a second pathogenic coronavirus, Middle East respiratory syndrome coronavirus (MERS-CoV). Both camostat and nafamostat reduced infection in primary human airway epithelia and in the Calu-3 2B4 cell line, with nafamostat exhibiting greater potency. We then assessed whether nafamostat was protective against SARS-CoV-2 in vivo using two mouse models. In mice sensitized to SARS-CoV-2 infection by transduction with human ACE2, intranasal nafamostat treatment prior to or shortly after SARS-CoV-2 infection significantly reduced weight loss and lung tissue titers. Similarly, prophylactic intranasal treatment with nafamostat reduced weight loss, viral burden, and mortality in K18-hACE2 transgenic mice. These findings establish nafamostat as a candidate for the prevention or treatment of SARS-CoV-2 infection and disease pathogenesis. IMPORTANCE The causative agent of COVID-19, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), requires host cell surface proteases for membrane fusion and entry into airway epithelia. We tested the hypothesis that inhibitors of these proteases, the serine protease inhibitors camostat and nafamostat, block infection by SARS-CoV-2. We found that both camostat and nafamostat reduce infection in human airway epithelia, with nafamostat showing greater potency. We then asked whether nafamostat protects mice against SARS-CoV-2 infection and subsequent COVID-19 lung disease. We performed infections in mice made susceptible to SARS-CoV-2 infection by introducing the human version of ACE2, the SARS-CoV-2 receptor, into their airway epithelia. We observed that pretreating these mice with nafamostat prior to SARS-CoV-2 infection resulted in better outcomes, in the form of less virus-induced weight loss, viral replication, and mortality than that observed in the untreated control mice. These results provide preclinical evidence for the efficacy of nafamostat in treating and/or preventing COVID-19.


Asunto(s)
Benzamidinas/farmacología , Ésteres/farmacología , Guanidinas/farmacología , SARS-CoV-2/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Tratamiento Farmacológico de COVID-19
3.
Aging (Albany NY) ; 12(22): 22425-22444, 2020 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-33221759

RESUMEN

With the current COVID-19 pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there is an urgent need for new therapies and prevention strategies that can help curtail disease spread and reduce mortality. The inhibition of viral entry and thus spread is a plausible therapeutic avenue. SARS-CoV-2 uses receptor-mediated entry into a human host via the angiotensin-converting enzyme 2 (ACE2), which is expressed in lung tissue as well as the oral and nasal mucosa, kidney, testes and gastrointestinal tract. The modulation of ACE2 levels in these gateway tissues may be an effective strategy for decreasing disease susceptibility. Cannabis sativa, especially those high in the anti-inflammatory cannabinoid cannabidiol (CBD), has been found to alter gene expression and inflammation and harbour anti-cancer and anti-inflammatory properties. However, its effects on ACE2 expression remain unknown. Working under a Health Canada research license, we developed over 800 new C. sativa cultivars and hypothesized that high-CBD C. sativa extracts may be used to down-regulate ACE2 expression in target COVID-19 tissues. Using artificial 3D human models of oral, airway and intestinal tissues, we identified 13 high-CBD C. sativa extracts that decrease ACE2 protein levels. Some C. sativa extracts down-regulate serine protease TMPRSS2, another critical protein required for SARS-CoV-2 entry into host cells. While our most effective extracts require further large-scale validation, our study is important for future analyses of the effects of medical cannabis on COVID-19. The extracts of our most successful novel high-CBD C. sativa lines, pending further investigation, may become a useful and safe addition to the prevention/treatment of COVID-19 as an adjunct therapy.


Asunto(s)
Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , COVID-19/prevención & control , Cannabis/química , Extractos Vegetales/farmacología , SARS-CoV-2/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/epidemiología , COVID-19/virología , Cannabidiol/farmacología , Cannabidiol/uso terapéutico , Simulación por Computador , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/virología , Modelos Anatómicos , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/metabolismo , Mucosa Bucal/virología , Pandemias/prevención & control , Extractos Vegetales/química , Extractos Vegetales/uso terapéutico , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Internalización del Virus/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
4.
mSphere ; 5(3)2020 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-32461278

RESUMEN

Zinc supplementation in cell culture has been shown to inhibit various viruses, like herpes simplex virus, rotavirus, severe acute respiratory syndrome (SARS) coronavirus, rhinovirus, and respiratory syncytial virus (RSV). However, whether zinc plays a direct antiviral role in viral infections and whether viruses have adopted strategies to modulate zinc homeostasis have not been investigated. Results from clinical trials of zinc supplementation in infections indicate that zinc supplementation may be beneficial in a pathogen- or disease-specific manner, further underscoring the importance of understanding the interaction between zinc homeostasis and virus infections at the molecular level. We investigated the effect of RSV infection on zinc homeostasis and show that RSV infection in lung epithelial cells leads to modulation of zinc homeostasis. The intracellular labile zinc pool increases upon RSV infection in a multiplicity of infection (MOI)-dependent fashion. Small interfering RNA (siRNA)-mediated knockdown of the ubiquitous zinc uptake transporter ZIP1 suggests that labile zinc levels are increased due to the increased uptake by RSV-infected cells as an antiviral response. Adding zinc to culture medium after RSV infection led to significant inhibition of RSV titers, whereas depletion of zinc by a zinc chelator, N,N,N',N'-tetrakis(2-pyridinylmethyl)-1,2-ethanediamine (TPEN) led to an increase in RSV titers. The inhibitory effect of zinc was specific, as other divalent cations had no effect on RSV titers. Both RSV infection and zinc chelation by TPEN led to reactive oxygen species (ROS) induction, whereas addition of zinc blocked ROS induction. These results suggest a molecular link between RSV infection, zinc homeostasis, and oxidative-stress pathways and provide new insights for developing strategies to counter RSV infection.IMPORTANCE Zinc deficiency rates in developing countries range from 20 to 30%, and zinc supplementation trials have been shown to correct clinical manifestations attributed to zinc deficiency, but the outcomes in the case of respiratory infections have been inconsistent. We aimed at understanding the role of zinc homeostasis in respiratory syncytial virus (RSV) infection. Infection of lung epithelial cell lines or primary small-airway epithelial cells led to an increase in labile zinc pools, which was due to increased uptake of zinc. Zinc supplementation inhibited RSV replication, whereas zinc chelation had an opposing effect, leading to increases in RSV titers. Increases in labile zinc in RSV-infected cells coincided with induction of reactive oxygen species (ROS). Both zinc depletion and addition of exogenous ROS led to enhanced RSV infection, whereas addition of the antioxidant inhibited RSV, suggesting that zinc is part of an interplay between RSV-induced oxidative stress and the host response to maintain redox balance.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitial Respiratorio Humano/metabolismo , Replicación Viral/efectos de los fármacos , Zinc/metabolismo , Zinc/farmacología , Células A549 , Adolescente , Proteínas de Transporte de Catión/genética , Línea Celular , Niño , Preescolar , Células Epiteliales/metabolismo , Etilenodiaminas/farmacología , Femenino , Interacciones Huésped-Patógeno , Humanos , Pulmón/citología , Pulmón/metabolismo , Masculino , Estrés Oxidativo/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología
5.
Sci Rep ; 9(1): 4787, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30886217

RESUMEN

Pollens are well-known triggers of respiratory allergies and asthma. The pollen burden in today's ambient air is constantly increasing due to rising climate change and air pollution. How pollens interact with the respiratory mucosa remains largely unknown due to a lack of representative model systems. We here demonstrate how pollen proteases of Kentucky bluegrass, white birch and hazel selectively destroy integrity and anchorage of columnar respiratory epithelial cells, but not of basal cells, in both ex vivo respiratory mucosal explants and in vitro primary equine respiratory epithelial cells (EREC). In turn, this pollen protease-induced damage to respiratory epithelial cell anchorage resulted in increased infection by the host-specific and ancestral alphaherpesvirus equine herpesvirus type 1 (EHV1). Pollen proteases of all three plant species were characterized by zymography and those of white birch were fully identified for the first time as serine proteases of the subtilase family and meiotic prophase aminopeptidase 1 using mass spectrometry-based proteomics. Together, our findings demonstrate that pollen proteases selectively and irreversibly damage integrity and anchorage of columnar respiratory epithelial cells. In turn, alphaherpesviruses benefit from this partial loss-of-barrier function, resulting in increased infection of the respiratory epithelium.


Asunto(s)
Infecciones por Herpesviridae/etiología , Proteínas de Plantas/metabolismo , Polen/toxicidad , Mucosa Respiratoria/virología , Serina Proteasas/metabolismo , Animales , Betula , Células Cultivadas , Corylus , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Herpesvirus Équido 1/patogenicidad , Caballos , Poaceae , Polen/enzimología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología
6.
Mol Med Rep ; 19(4): 3330-3336, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30816467

RESUMEN

Respiratory syncytial virus (RSV) infections are associated with significant morbidity and mortality. Inflammation is mediated by cytokine secretion from RSV­infected airway epithelial cells. Grape seed proanthocyanidin extract (GSPE) exhibits potent antioxidant capacity, as well as anti­bacterial, anti­viral, anti­carcinogenic, anti­inflammatory and anti­allergic actions. However, few studies have explored the anti­inflammatory effects of GSPE on airway epithelial cells infected with RSV. Airway epithelial A549 cells were pretreated with GSPE and its effects on cytokine production during RSV infection were investigated. A549 cells were infected with RSV, with or without GSPE pretreatment, and cultured for 24, 48 and 72 h. The expression of interleukin (IL)­1ß, IL­6 and IL­8, were measured by reverse transcription­quantitative polymerase chain reaction, ELISA and western blotting. RSV infection induced significant increases in proinflammatory cytokine expression. However, GSPE pretreatment decreased the mRNA and protein expression levels of IL­1ß, IL­6 and IL­8. GSPE regulated the immune response by reducing the RSV­induced transcription of proinflammatory cytokines in airway epithelial cells, suggesting that GSPE helps to prevent RSV­induced airway disease.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Extracto de Semillas de Uva/farmacología , Proantocianidinas/farmacología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Virus Sincitiales Respiratorios/fisiología , Células A549 , Supervivencia Celular/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Células Epiteliales/virología , Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Mucosa Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología
7.
J Antimicrob Chemother ; 73(7): 1823-1829, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29596680

RESUMEN

Objectives: We report the use of reconstituted 3D human airway epithelium cells (HuAECs) of bronchial origin in an air-liquid interface to study respiratory syncytial virus (RSV) infection and to assess the efficacy of RSV inhibitors in (pre-)clinical development. Methods: HuAECs were infected with RSV-A Long strain (0.01 CCID50/cell, where CCID50 represents 50% cell culture infectious dose in HEp2 cells) on the apical compartment of the culture. At the time of infection or at 1 or 3 days post-infection, selected inhibitors were added and refreshed daily on the basal compartment of the culture. Viral shedding was followed up by apical washes collected daily and quantifying viral RNA by RT-qPCR. Results: RSV-A replicates efficiently in HuAECs and viral RNA is shed for weeks after infection. RSV infection reduces the ciliary beat frequency of the ciliated cells as of 4 days post-infection, with complete ciliary dyskinesia observed by day 10. Treatment with RSV fusion inhibitors resulted in an antiviral effect only when added at the time of infection. In contrast, the use of replication inhibitors (both nucleoside and non-nucleoside) elicited a marked antiviral effect even when the start of treatment was delayed until 1 day or even 3 days after infection. Levels of the inflammation marker RANTES (mRNA) increased ∼200-fold in infected, untreated cultures (at 3 weeks post-infection), but levels were comparable to those of uninfected cultures in the presence of PC786, an RSV replication inhibitor, suggesting that an efficient antiviral treatment might inhibit virus-induced inflammation in this model. Conclusions: Overall, HuAECs offer a robust and physiologically relevant model to study RSV replication and to assess the efficacy of antiviral compounds.


Asunto(s)
Antivirales/farmacología , Mucosa Respiratoria/virología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Benzamidas , Benzazepinas , Técnicas de Cultivo de Célula , Evaluación Preclínica de Medicamentos , Células Epiteliales/virología , Humanos , Técnicas de Cultivo de Órganos , ARN Viral/genética , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitial Respiratorio Humano/genética , Compuestos de Espiro/farmacología
8.
Br J Pharmacol ; 175(12): 2520-2534, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29579332

RESUMEN

BACKGROUND AND PURPOSE: Effective anti-respiratory syncytial virus (RSV) agents are still not available for clinical use. Current major targets are virus surface proteins, such as a fusion protein involved in viral entry, but agents effective after RSV infection is established are required. Here we have investigated the effects of late therapeutic intervention with a novel inhaled RSV polymerase inhibitor, PC786, on RSV infection in human airway epithelium. EXPERIMENTAL APPROACH: Air liquid interface-cultured bronchial or small airway epithelium was infected with RSVA2. PC786 was applied apically or basolaterally once daily following peak virus load on Day 3 post inoculation. Apical wash was collected daily for determination of viral burden by PCR and plaque assay (primary endpoints) and biomarker analyses. The effects were compared with those of ALS-8112, an anti-RSV nucleoside analogue, and GS-5806, a fusion-protein inhibitor, which were treated basolaterally. KEY RESULTS: Late intervention with GS-5806 did not show significant anti-viral effects, but PC786 produced potent, concentration-dependent inhibition of viral replication with viral load falling below detectable limits 3 days after treatment commenced in airway epithelium. These effects were superior to those of ALS-8112. PC786 showed inhibitory activities against RSV-induced increases of CCL5, IL-6, double-strand DNA and mucin. The effects of PC786 were also confirmed in small airway epithelium. CONCLUSION AND IMPLICATIONS: Late therapeutic intervention with the RSV polymerase inhibitor, PC786, reduced the viral burden quickly in human airway epithelium. Thus, PC786 demonstrates the potential to be an effective therapeutic agent to treat active RSV infection.


Asunto(s)
Antivirales/farmacología , Epitelio/efectos de los fármacos , Mucosa Respiratoria/efectos de los fármacos , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Compuestos de Espiro/farmacología , Antivirales/química , Benzamidas , Benzazepinas , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , ARN Polimerasas Dirigidas por ADN/metabolismo , Relación Dosis-Respuesta a Droga , Epitelio/metabolismo , Epitelio/virología , Células HeLa , Humanos , Pruebas de Sensibilidad Microbiana , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Compuestos de Espiro/química , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
9.
Nutrients ; 7(2): 1021-36, 2015 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-25658239

RESUMEN

Ginseng has been used in humans for thousands of years but its effects on viral infection have not been well understood. We investigated the effects of red ginseng extract (RGE) on respiratory syncytial virus (RSV) infection using in vitro cell culture and in vivo mouse models. RGE partially protected human epithelial (HEp2) cells from RSV-induced cell death and viral replication. In addition, RGE significantly inhibited the production of RSV-induced pro-inflammatory cytokine (TNF-α) in murine dendritic and macrophage-like cells. More importantly, RGE intranasal pre-treatment prevented loss of mouse body weight after RSV infection. RGE treatment improved lung viral clearance and enhanced the production of interferon (IFN-γ) in bronchoalveolar lavage cells upon RSV infection of mice. Analysis of cellular phenotypes in bronchoalveolar lavage fluids showed that RGE treatment increased the populations of CD8+ T cells and CD11c+ dendritic cells upon RSV infection of mice. Taken together, these results provide evidence that ginseng has protective effects against RSV infection through multiple mechanisms, which include improving cell survival, partial inhibition of viral replication and modulation of cytokine production and types of immune cells migrating into the lung.


Asunto(s)
Antivirales/farmacología , Células Epiteliales/virología , Panax , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitiales Respiratorios/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/administración & dosificación , Líquido del Lavado Bronquioalveolar/microbiología , Antígeno CD11c/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/virología , Femenino , Humanos , Interferón gamma/metabolismo , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/virología , Factor de Necrosis Tumoral alfa/metabolismo , Carga Viral/efectos de los fármacos
10.
Int J Mol Med ; 34(1): 183-90, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24756136

RESUMEN

Panax ginseng has been known to have a number of immuno-modulatory effects. In this study, we investigated whether Panax Korean red ginseng extract (KRGE) has in vitro and in vivo antiviral effects on respiratory syncytial virus (RSV) infection. KRGE improved the survival of human lung epithelial cells against RSV infection and inhibited RSV replication. In addition, KRGE treatment suppressed the expression of RSV-induced inflammatory cytokine genes (IL-6 and IL-8) and the formation of reactive oxygen species in epithelial cell cultures. Oral administration of mice with KRGE resulted in lowering lung viral loads after RSV infection. Additionally, the in vivo effects of KRGE showed an enhanced level of interferon-γ (IFN-γ) producing dendritic cells subsequent to RSV infection. Taken together, these results suggested that KRGE has antiviral activity against RSV infection.


Asunto(s)
Antivirales/farmacología , Panax/química , Extractos Vegetales/farmacología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitiales Respiratorios/efectos de los fármacos , Administración Oral , Animales , Antivirales/aislamiento & purificación , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/virología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/virología , Femenino , Interacciones Huésped-Patógeno , Humanos , Interferón gamma/biosíntesis , Interferón gamma/metabolismo , Interleucina-6/biosíntesis , Interleucina-6/metabolismo , Interleucina-8/biosíntesis , Interleucina-8/metabolismo , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Extractos Vegetales/aislamiento & purificación , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/fisiología , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
11.
PLoS One ; 9(1): e86755, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24475177

RESUMEN

Vitamin D has been linked to reduced risk of viral respiratory illness. We hypothesized that vitamin D could directly reduce rhinovirus (RV) replication in airway epithelium. Primary human bronchial epithelial cells (hBEC) were treated with vitamin D, and RV replication and gene expression were evaluated by quantitative PCR. Cytokine/chemokine secretion was measured by ELISA, and transepithelial resistance (TER) was determined using a voltohmmeter. Morphology was examined using immunohistochemistry. Vitamin D supplementation had no significant effects on RV replication, but potentiated secretion of CXCL8 and CXCL10 from infected or uninfected cells. Treatment with vitamin D in the form of 1,25(OH)2D caused significant changes in cell morphology, including thickening of the cell layers (median of 46.5 µm [35.0-69.0] vs. 30 µm [24.5-34.2], p<0.01) and proliferation of cytokeratin-5-expressing cells, as demonstrated by immunohistochemical analysis. Similar effects were seen for 25(OH)D. In addition to altering morphology, higher concentrations of vitamin D significantly upregulated small proline-rich protein (SPRR1ß) expression (6.3 fold-induction, p<0.01), suggestive of squamous metaplasia. Vitamin D treatment of hBECs did not alter repair of mechanically induced wounds. Collectively, these findings indicate that vitamin D does not directly affect RV replication in airway epithelial cells, but can influence chemokine synthesis and alters the growth and differentiation of airway epithelial cells.


Asunto(s)
Calcitriol/farmacología , Células Epiteliales/efectos de los fármacos , Rhinovirus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Bronquios/efectos de los fármacos , Bronquios/patología , Bronquios/virología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Quimiocina CXCL10/biosíntesis , Quimiocina CXCL10/metabolismo , Proteínas Ricas en Prolina del Estrato Córneo/agonistas , Proteínas Ricas en Prolina del Estrato Córneo/genética , Proteínas Ricas en Prolina del Estrato Córneo/metabolismo , Células Epiteliales/patología , Células Epiteliales/ultraestructura , Células Epiteliales/virología , Expresión Génica , Humanos , Interleucina-8/biosíntesis , Interleucina-8/metabolismo , Queratina-5/agonistas , Queratina-5/genética , Queratina-5/metabolismo , Cultivo Primario de Células , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Rhinovirus/fisiología , Cicatrización de Heridas/efectos de los fármacos
12.
Am J Respir Cell Mol Biol ; 50(2): 389-97, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24053146

RESUMEN

Recent studies have revealed that the human and nonrodent mammalian airway mucosa contains an oxidative host defense system. This three-component system consists of the hydrogen peroxide (H2O2)-producing enzymes dual oxidase (Duox)1 and Duox2, thiocyanate (SCN(-)), and secreted lactoperoxidase (LPO). The LPO-catalyzed reaction between H2O2 and SCN(-) yields the bactericidal hypothiocyanite (OSCN(-)) in airway surface liquid (ASL). Although SCN(-) is the physiological substrate of LPO, the Duox/LPO/halide system can generate hypoiodous acid when the iodide (I(-)) concentration is elevated in ASL. Because hypoiodous acid, but not OSCN(-), inactivates respiratory syncytial virus (RSV) in cell culture, we used a lamb model of RSV to test whether potassium iodide (KI) could enhance this system in vivo. Newborn lambs received KI by intragastric gavage or were left untreated before intratracheal inoculation of RSV. KI treatment led to a 10-fold increase in ASL I(-) concentration, and this I(-) concentration was approximately 30-fold higher than that measured in the serum. Also, expiratory effort, gross lung lesions, and pulmonary expression of an RSV antigen and IL-8 were reduced in the KI-treated lambs as compared with nontreated control lambs. Inhibition of LPO activity significantly increased lesions, RSV mRNA, and antigen. Similar experiments in 3-week-old lambs demonstrated that KI administration was associated with reduced gross lesions, decreased RSV titers in bronchoalveolar lavage fluid, and reduced RSV antigen expression. Overall, these data indicate that high-dose KI supplementation can be used in vivo to lessen the severity of RSV infections, potentially through the augmentation of mucosal oxidative defenses.


Asunto(s)
Yoduro de Potasio/farmacología , Mucosa Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Animales , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Humanos , Lactoperoxidasa/metabolismo , Yoduro de Potasio/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Ovinos , Tiocianatos/metabolismo
13.
Br J Nutr ; 101(4): 533-40, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18631417

RESUMEN

Long-chain n-3 PUFA (LCn-3PUFA) including DHA and EPA, are known to decrease inflammation by inhibiting arachidonic acid (AA) metabolism to eicosanoids, decreasing the production of pro-inflammatory cytokines and reducing immune cell function. The aim of this study was to determine if EPA and DHA reduced the release of inflammatory mediators from airway epithelial cells infected with rhinovirus (RV). Airway epithelial cells (Calu-3) were incubated with EPA, DHA and AA for 24 h, followed by rhinovirus infection for 48 h. IL-6, IL-8 and interferon-gamma-induced protein-10 (IP-10) released by cells were measured using ELISA. Viral replication was measured by serial titration assays. The fatty acid content of cells was analysed using GC. Cellular viability was determined by visual inspection of cells and lactate dehydrogenase release. DHA (400 microm) resulted in a significant 16% reduction in IL-6 release after RV-43 infection, 29% reduction in IL-6 release after RV-1B infection, 28% reduction in IP-10 release after RV-43 infection and 23 % reduction in IP-10 release after RV-1B infection. Cellular DHA content negatively correlated with IL-6 and IP-10 release. None of the fatty acids significantly modified rhinovirus replication. DHA supplementation resulted in increased cellular content of DHA at the cost of AA, which may explain the decreased inflammatory response of cells. EPA and AA did not change the release of inflammatory biomarkers significantly. It is concluded that DHA has a potential role in suppressing RV-induced airway inflammation.


Asunto(s)
Antiinflamatorios/uso terapéutico , Células Epiteliales/virología , Ácidos Grasos Omega-3/uso terapéutico , Infecciones por Picornaviridae/tratamiento farmacológico , Mucosa Respiratoria/virología , Rhinovirus , Análisis de Varianza , Ácido Araquidónico/metabolismo , Línea Celular , Quimiocina CXCL10/análisis , Ácidos Docosahexaenoicos/análisis , Ácidos Docosahexaenoicos/farmacología , Relación Dosis-Respuesta a Droga , Ácido Eicosapentaenoico/análisis , Ácido Eicosapentaenoico/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Humanos , Interleucina-6/análisis , Interleucina-8/análisis , Infecciones por Picornaviridae/inmunología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA