Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Physiol Genomics ; 54(10): 389-401, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36062885

RESUMEN

Military Deployment to Southwest Asia and Afghanistan and exposure to toxic airborne particulates have been associated with an increased risk of developing respiratory disease, collectively termed deployment-related respiratory diseases (DRRDs). Our knowledge about how particulates mediate respiratory disease is limited, precluding the appropriate recognition or management. Central to this limitation is the lack of understanding of how exposures translate into dysregulated cell identity with dysregulated transcriptional programs. The small airway epithelium is involved in both the pathobiology of DRRD and fine particulate matter deposition. To characterize small airway epithelial cell epigenetic and transcriptional responses to Afghan desert particulate matter (APM) and investigate the functional interactions of transcription factors that mediate these responses, we applied two genomics assays, the assay for transposase accessible chromatin with sequencing (ATAC-seq) and Precision Run-on sequencing (PRO-seq). We identified activity changes in a series of transcriptional pathways as candidate regulators of susceptibility to subsequent insults, including signal-dependent pathways, such as loss of cytochrome P450 or P53/P63, and lineage-determining transcription factors, such as GRHL2 loss or TEAD3 activation. We further demonstrated that TEAD3 activation was unique to APM exposure despite similar inflammatory responses when compared with wood smoke particle exposure and that P53/P63 program loss was uniquely positioned at the intersection of signal-dependent and lineage-determining transcriptional programs. Our results establish the utility of an integrated genomics approach in characterizing responses to exposures and identifying genomic targets for the advanced investigation of the pathogenesis of DRRD.


Asunto(s)
Células Epiteliales Alveolares , Material Particulado , Factores de Transcripción , Afganistán , Células Epiteliales Alveolares/metabolismo , Cromatina/metabolismo , Epigénesis Genética , Genómica/métodos , Despliegue Militar , Material Particulado/toxicidad , Enfermedades Respiratorias/epidemiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transposasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
2.
Chem Res Toxicol ; 34(12): 2408-2423, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34808040

RESUMEN

Approximately 3 million United States military personnel and contractors were deployed to Southwest Asia and Afghanistan over the past two decades. After returning to the United States, many developed persistent respiratory symptoms, including those due to asthma, rhinosinusitis, bronchiolitis, and others, which we collectively refer to as deployment-related lung diseases (DRLD). The mechanisms of different DRLD have not been well defined. Limited studies from us and others suggest that multiple factors and biological signaling pathways contribute to the onset of DRLD. These include, but are not limited to, exposures to high levels of particulate matter (PM) from sandstorms, burn pit combustion products, improvised explosive devices, and diesel exhaust particles. Once inhaled, these hazardous substances can activate lung immune and structural cells to initiate numerous cell-signaling pathways such as oxidative stress, Toll-like receptors, and cytokine-driven cell injury (e.g., interleukin-33). These biological events may lead to a pro-inflammatory response and airway hyperresponsiveness. Additionally, exposures to PM and other environmental hazards may predispose military personnel and contractors to more severe disease due to the interactions of those hazardous materials with subsequent exposures to allergens and cigarette smoke. Understanding how airborne exposures during deployment contribute to DRLD may identify effective targets to alleviate respiratory diseases and improve quality of life in veterans and active duty military personnel.


Asunto(s)
Enfermedades Pulmonares/inducido químicamente , Material Particulado/efectos adversos , Afganistán , Humanos , Irak , Personal Militar
3.
Toxicol Appl Pharmacol ; 404: 115186, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32777237

RESUMEN

Increased symptoms of asthma-like respiratory illnesses have been reported in soldiers returning from tours of duty in Afghanistan. Inhalation of desert particulate matter (PM) may contribute to this deployment-related lung disease (DRLD), but little is known about disease mechanisms. The IL-33 signaling pathway, including its receptor ST2, has been implicated in the pathogenesis of lung diseases including asthma, but its role in PM-mediated airway dysfunction has not been studied. The goal of this study was to investigate whether IL-33/ST2 signaling contributes to airway dysfunction in preclinical models of lung exposure to Afghanistan PM (APM). Wild-type (WT) and ST2 knockout (KO) mice on the BALB/C background were oropharyngeally instilled with a single dose of saline or 50 µg of APM in saline. Airway hyperresponsiveness (AHR) and inflammation were assessed after 24 h. In WT mice, a single APM exposure induced AHR and neutrophilic inflammation. Unlike the WT mice, ST2 KO mice that lack the receptor for IL-33 did not demonstrate AHR although airway neutrophilic inflammation was comparable to the WT mice. Oropharyngeal delivery of a soluble ST2 decoy receptor in APM-exposed WT mice significantly blocked AHR. Additional data in mouse tracheal epithelial cell and lung macrophage cultures demonstrated a role of APM-induced IL-33/ST2 signaling in suppression of regulator of G protein signaling 2 (RGS2), a gene known to protect against bronchoconstriction. We present for the first time that APM may increase AHR, one of the features of asthma, in part through the IL-33/ST2/RGS2 pathway.


Asunto(s)
Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Enfermedades Pulmonares/inducido químicamente , Material Particulado/toxicidad , Afganistán , Animales , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-33/genética , Macrófagos/efectos de los fármacos , Ratones , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Tamaño de la Partícula , Alveolos Pulmonares/citología , Transducción de Señal/efectos de los fármacos
4.
J Biol Chem ; 291(24): 12673-12687, 2016 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-27076634

RESUMEN

Antagonism of pro-inflammatory transcription factors by monomeric glucocorticoid receptor (GR) has long been viewed as central to glucocorticoid (GC) efficacy. However, the mechanisms and targets through which GCs exert therapeutic effects in diseases such as asthma remain incompletely understood. We previously defined a surprising cooperative interaction between GR and NF-κB that enhanced expression of A20 (TNFAIP3), a potent inhibitor of NF-κB. Here we extend this observation to establish that A20 is required for maximal cytokine repression by GCs. To ascertain the global extent of GR and NF-κB cooperation, we determined genome-wide occupancy of GR, the p65 subunit of NF-κB, and RNA polymerase II in airway epithelial cells treated with dexamethasone, TNF, or both using chromatin immunoprecipitation followed by deep sequencing. We found that GR recruits p65 to dimeric GR binding sites across the genome and discovered additional regulatory elements in which GR-p65 cooperation augments gene expression. GR targets regulated by this mechanism include key anti-inflammatory and injury response genes such as SERPINA1, which encodes α1 antitrypsin, and FOXP4, an inhibitor of mucus production. Although dexamethasone treatment reduced RNA polymerase II occupancy of TNF targets such as IL8 and TNFAIP2, we were unable to correlate specific binding sequences for GR or occupancy patterns with repressive effects on transcription. Our results suggest that cooperative anti-inflammatory gene regulation by GR and p65 contributes to GC efficacy, whereas tethering interactions between GR and p65 are not universally required for GC-based gene repression.


Asunto(s)
Antiinflamatorios/farmacología , Células Epiteliales/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Factor de Transcripción ReIA/metabolismo , Western Blotting , Línea Celular , Células Cultivadas , Dexametasona/farmacología , Células Epiteliales/metabolismo , Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Humanos , Unión Proteica/efectos de los fármacos , Interferencia de ARN , ARN Polimerasa II/metabolismo , Receptores de Glucocorticoides/genética , Sistema Respiratorio/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción ReIA/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
5.
Inflamm Res ; 66(8): 691-700, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28451734

RESUMEN

OBJECTIVE: To evaluate the effects of MUC18 on IL-13-mediated airway inflammatory responses in human airway epithelial cells and in mice. MATERIALS: Primary normal human tracheobronchial epithelial (HTBE) cells, wild-type (WT) and Muc18 knockout (KO) mice, and mouse tracheal epithelial cells (mTECs) were utilized. TREATMENT: Cultured HTBE cells treated with MUC18 siRNA or MUC18 expressing lentivirus were incubated with IL-13 (10 ng/mL) for 24 h. Mice were intranasally instilled with 500 ng of IL-13 for 3 days. mTECs were treated with IL-13 (10 ng/mL) for 3 days. METHODS: PCR was used to measure mRNA expression. Western Blot and ELISAs were used to quantify protein expression. Cytospins of bronchoalveolar lavage (BAL) cells were used to obtain leukocyte differentials. RESULTS: MUC18 siRNA reduced IL-13-mediated eotaxin-3 (183 ± 44 vs. 380 ± 59 pg/mL, p < 0.05), while MUC18 overexpression increased IL-13-mediated eotaxin-3 (95 ± 3 vs. 58 ± 3 pg/mL, p < 0.05) in HTBE cells. IL-13-treated Muc18 KO mice had a lower percentage of neutrophils in BAL than WT mice (25 ± 3 vs. 35 ± 3%, p = 0.0565). CONCLUSIONS: These results implicate MUC18 as a potential enhancer of airway inflammation in a type 2 cytokine (e.g., IL-13) milieu.


Asunto(s)
Antígeno CD146/inmunología , Citocinas/inmunología , Inflamación/inmunología , Sistema Respiratorio/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Antígeno CD146/genética , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Humanos , Ratones Noqueados , ARN Interferente Pequeño/genética , Sistema Respiratorio/citología
6.
Toxicol Sci ; 182(2): 297-309, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34051097

RESUMEN

Upon returning from deployment to Afghanistan, a substantial number of U.S. military personnel report deployment-related lung disease (DRLD) symptoms, including those consistent with an asthma-like airways disease. DRLD is thought to be caused by prolonged inhalation of toxic desert particulate matter, which can persist in the postdeployment setting such as exposure to common household allergens. The goal of this study was to define the transcriptomic responses of lung leukocytes of mice exposed to Afghanistan desert particulate matter (APM) and house dust mite (HDM). C57BL/6 mice (n = 15/group) were exposed to filtered air or aerosolized APM for 12 days, followed by intranasal PBS or HDM allergen challenges for 24 h. Bronchoalveolar lavage (BAL) cells were collected for single-cell RNA sequencing (scRNAseq), and assessment of inflammation and airway hyper-responsiveness. Unsupervised clustering of BAL cell scRNAseq data revealed a unique monocyte population induced only by both APM and allergen treatments. This population of monocytes is characterized by the expression of genes involved in allergic asthma, including Alox15. We validated Alox15 expression in monocytes via immunostaining of lung tissue. APM pre-exposure, followed by the HDM challenge, led to significantly increased total respiratory system resistance compared with filtered air controls. Using this mouse model to mimic DRLD, we demonstrated that inhalation of airborne PM during deployment may prime airways to be more responsive to allergen exposure after returning home, which may be linked to dysregulated immune responses such as induction of a unique lung monocyte population.


Asunto(s)
Alérgenos , Material Particulado , Afganistán , Alérgenos/toxicidad , Animales , Lavado Broncoalveolar , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Pulmón , Ratones , Ratones Endogámicos C57BL , Monocitos , Material Particulado/toxicidad , Análisis de Secuencia de ARN
7.
JCI Insight ; 4(8)2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30996135

RESUMEN

Bacterial permeability family member A1 (BPIFA1), also known as short palate, lung, and nasal epithelium clone 1 (SPLUNC1), is a protein involved in the antiinflammatory response. The goal of this study was to determine whether BPIFA1 expression in asthmatic airways is regulated by genetic variations, altering epithelial responses to type 2 cytokines (e.g., IL-13). Nasal epithelial cells from patients with mild to severe asthma were collected from the National Heart, Lung, and Blood Institute Severe Asthma Research Program centers, genotyped for rs750064, and measured for BPIFA1. To determine the function of rs750064, cells were cultured at air-liquid interface and treated with IL-13 with or without recombinant human BPIFA1 (rhBPIFA1). Noncultured nasal cells with the rs750064 CC genotype had significantly less BPIFA1 mRNA expression than the CT and TT genotypes. Cultured CC versus CT and TT cells without stimulation maintained less BPIFA1 expression. With IL-13 treatment, CC genotype cells secreted more eotaxin-3 than CT and TT genotype cells. Also, rhBPIFA1 reduced IL-13-mediated eotaxin-3. BPIFA1 mRNA levels negatively correlated with serum IgE and fractional exhaled nitric oxide. Baseline FEV1% levels were lower in the asthma patients with the CC genotype (n = 1,016). Our data suggest that less BPIFA1 in asthma patients with the CC allele may predispose them to greater eosinophilic inflammation, which could be attenuated by rhBPIFA1 protein therapy.


Asunto(s)
Asma/genética , Células Epiteliales/inmunología , Regulación de la Expresión Génica/inmunología , Glicoproteínas/genética , Fosfoproteínas/genética , Transducción de Señal/inmunología , Adolescente , Adulto , Anciano , Alelos , Asma/diagnóstico , Asma/tratamiento farmacológico , Asma/inmunología , Células Cultivadas , Quimiocina CCL26/inmunología , Quimiocina CCL26/metabolismo , Niño , Eosinófilos/inmunología , Células Epiteliales/patología , Femenino , Volumen Espiratorio Forzado , Predisposición Genética a la Enfermedad , Glicoproteínas/metabolismo , Glicoproteínas/farmacología , Glicoproteínas/uso terapéutico , Humanos , Interleucina-13/inmunología , Interleucina-13/metabolismo , Masculino , Persona de Mediana Edad , Mucosa Nasal/citología , Mucosa Nasal/inmunología , Fosfoproteínas/metabolismo , Fosfoproteínas/farmacología , Fosfoproteínas/uso terapéutico , Polimorfismo de Nucleótido Simple , Cultivo Primario de Células , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
8.
Toxicol Sci ; 166(2): 345-353, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30169750

RESUMEN

Since the start of Afghanistan combat operations in 2001, there has been an increase in complaints of respiratory illnesses in deployed soldiers with no previous history of lung disorders. It is postulated that deployment-related respiratory illnesses are the result of inhalation of desert particulate matter (PM) potentially acting in combination with exposure to other pro-inflammatory compounds. Why some, but not all, soldiers develop respiratory diseases remains unclear. Our goal was to investigate if human airway epithelial cells primed with IL-13, a type 2 inflammatory cytokine, demonstrate stronger pro-inflammatory responses to Afghanistan desert PM (APM). Primary human brushed bronchial epithelial cells from non-deployed, healthy subjects were exposed to APM, both with and without IL-13 pretreatment. APM exposure in conjunction with IL-13 resulted in significantly increased expression of IL-8, a pro-inflammatory cytokine involved in neutrophil recruitment and activation. Furthermore, expression of TLR2 mRNA was increased after combined IL-13 and APM exposure. siRNA-mediated TLR2 knockdown dampened IL-8 production after exposure to APM with IL-13. APM with IL-13 treatment increased IRAK-1 (a downstream signaling molecule of TLR2 signaling) activation, while IRAK-1 knockdown effectively eliminated the IL-8 response to APM and IL-13. Our data suggest that APM exposure may promote neutrophilic inflammation in airways with a type 2 cytokine milieu.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Interleucina-13/farmacología , Material Particulado/envenenamiento , Afganistán , Anciano , Enfermedades Bronquiales/inducido químicamente , Enfermedades Bronquiales/metabolismo , Células Cultivadas , Citocinas , Células Epiteliales/inmunología , Femenino , Voluntarios Sanos , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-13/metabolismo , Interleucina-8/metabolismo , Masculino , Persona de Mediana Edad , ARN Interferente Pequeño , Transducción de Señal , Receptor Toll-Like 2/metabolismo
9.
Innate Immun ; 23(2): 206-215, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28120642

RESUMEN

Myeloid cells such as macrophages are critical to innate defense against infection. IL-1 receptor-associated kinase M (IRAK-M) is a negative regulator of TLR signaling during bacterial infection, but the role of myeloid cell IRAK-M in bacterial infection is unclear. Our goal was to generate a novel conditional knockout mouse model to define the role of myeloid cell IRAK-M during bacterial infection. Myeloid cell-specific IRAK-M knockout mice were generated by crossing IRAK-M floxed mice with LysM-Cre knock-in mice. The resulting LysM-Cre+/IRAK-Mfl/wt and control (LysM-Cre-/IRAK-Mfl/wt) mice were intranasally infected with Pseudomonas aeruginosa (PA). IRAK-M deletion, inflammation, myeloperoxidase (MPO) activity and PA load were measured in leukocytes, bronchoalveolar lavage (BAL) fluid and lungs. PA killing assay with BAL fluid was performed to determine mechanisms of IRAK-M-mediated host defense. IRAK-M mRNA and protein levels in alveolar and lung macrophages were significantly reduced in LysM-Cre+/IRAK-Mfl/wt mice compared with control mice. Following PA infection, LysM-Cre+/IRAK-Mfl/wt mice have enhanced lung neutrophilic inflammation, including MPO activity, but reduced PA load. The increased lung MPO activity in LysM-Cre+/IRAK-Mfl/wt mouse BAL fluid reduced PA load. Generation of IRAK-M conditional knockout mice will enable investigators to determine precisely the function of IRAK-M in myeloid cells and other types of cells during infection and inflammation.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Enfermedades Pulmonares/inmunología , Macrófagos/fisiología , Neutrófilos/fisiología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Animales , Bacteriólisis/genética , Movimiento Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Quinasas Asociadas a Receptores de Interleucina-1/genética , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peroxidasa/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-27354786

RESUMEN

Human rhinovirus (HRV) infections target airway epithelium and are the leading cause of acute exacerbations of COPD. Cigarette smoke (CS) increases the severity of viral infections, but there is no effective therapy for HRV infection. We determined whether α1-antitrypsin (A1AT) reduces HRV-16 infection in CS-exposed primary human airway epithelial cells. Brushed bronchial epithelial cells from normal subjects and patients diagnosed with COPD were cultured at air-liquid interface to induce mucociliary differentiation. These cells were treated with A1AT or bovine serum albumin for 2 hours and then exposed to air or whole cigarette smoke (WCS) with or without HRV-16 (5×10(4) 50% Tissue Culture Infective Dose [TCID50]/transwell) infection for 24 hours. WCS exposure significantly increased viral load by an average of fivefold and decreased the expression of antiviral genes interferon-λ1, OAS1, and MX1. When A1AT was added to WCS-exposed cells, viral load significantly decreased by an average of 29-fold. HRV-16 infection significantly increased HRV-16 receptor intercellular adhesion molecule-1 messenger RNA expression in air-exposed cells, which was decreased by A1AT. A1AT-mediated reduction of viral load was not accompanied by increased epithelial antiviral gene expression or by inhibiting the activity of 3C protease involved in viral replication or maturation. Our findings demonstrate that A1AT treatment prevents a WCS-induced increase in viral load and for the first time suggest a therapeutic effect of A1AT on HRV infection.


Asunto(s)
Antivirales/farmacología , Bronquios/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Infecciones por Picornaviridae/prevención & control , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Mucosa Respiratoria/efectos de los fármacos , Infecciones del Sistema Respiratorio/prevención & control , Rhinovirus/efectos de los fármacos , Humo/efectos adversos , Fumar/efectos adversos , alfa 1-Antitripsina/farmacología , Anciano , Bronquios/enzimología , Bronquios/patología , Bronquios/virología , Estudios de Casos y Controles , Células Cultivadas , Células Epiteliales/enzimología , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Interacciones Huésped-Patógeno , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Persona de Mediana Edad , Infecciones por Picornaviridae/enzimología , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/enzimología , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Infecciones del Sistema Respiratorio/enzimología , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología , Rhinovirus/patogenicidad , Carga Viral
11.
J Clin Cell Immunol ; 7(6)2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28191362

RESUMEN

OBJECTIVE: Excessive airway inflammation is seen in chronic obstructive pulmonary disease (COPD) patients experiencing acute exacerbations, which are often associated with human rhinovirus (HRV) infection. Alpha-1 antitrypsin (A1AT) has anti-inflammatory function in endothelial cells and monocytes, but its anti-inflammatory effect has not been investigated in COPD airway epithelial cells. We determined A1AT's anti-inflammatory function in COPD airway epithelial cells and the underlying mechanisms such as the role of caspase-1. METHODS: Brushed bronchial epithelial cells from COPD and normal subjects were cultured at air-liquid interface and treated with A1AT or bovine serum albumin (BSA, control) two hours prior to whole cigarette smoke (WCS) or air exposure, followed by HRV-16 infection. After 24 hours of viral infection, cell supernatants were collected for measuring IL-8, and cells were examined for caspase-1. The in vivo anti-inflammatory function of A1AT was determined by infecting mice intranasally with HRV-1B followed by aerosolized A1AT or BSA. RESULTS: A1AT significantly reduced WCS and HRV-16-induced IL-8 production in normal and COPD airway epithelial cells. COPD cells are less sensitive to A1AT's anti-inflammatory effect than normal cells. A1AT exerted the anti-inflammatory function in part via reducing caspase-1 in normal cells, but not in COPD cells. In mice, A1AT significantly reduced HRV-1B induced lung neutrophilic inflammation. CONCLUSIONS: A1AT exerts an anti-inflammatory effect in cigarette smoke-exposed and HRV-infected human airway epithelial cells, which may be related to its inhibitory effect on caspase-1 activity.

12.
PLoS One ; 11(10): e0163927, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27701461

RESUMEN

BACKGROUND: MUC18 is upregulated in the lungs of asthma and COPD patients. It has been shown to have pro-inflammatory functions in cultured human airway epithelial cells during viral infections and in mice during lung bacterial infections. However, the in vivo role of MUC18 in the context of viral infections remains poorly understood. The goal of this study is to define the in vivo function of MUC18 during respiratory rhinovirus infection. METHODS: Muc18 wild-type (WT) and knockout (KO) mice were infected with human rhinovirus 1B (HRV-1B) and sacrificed after 1 day to determine the inflammatory and antiviral responses. To examine the direct effects of Muc18 on viral infection, tracheal epithelial cells isolated from WT and KO mice were grown under air-liquid interface and infected with HRV-1B. Finally, siRNA mediated knockdown of MUC18 was performed in human airway epithelial cells (AECs) to define the impact of MUC18 on human airway response to HRV-1B. RESULTS: Both viral load and neutrophilic inflammation were significantly decreased in Muc18 KO mice compared to WT mice. In the in vitro setting, viral load was significantly lower and antiviral gene expression was higher in airway epithelial cells of Muc18 KO mice than the WT mice. Furthermore, in MUC18 knockdown human AECs, viral load was decreased and antiviral gene expression was increased compared to controls. CONCLUSIONS: Our study is the first to demonstrate MUC18's pro-inflammatory and pro-viral function in an in vivo mouse model of rhinovirus infection.


Asunto(s)
Infecciones por Picornaviridae/metabolismo , Rhinovirus/fisiología , Animales , Antígeno CD146/genética , Antígeno CD146/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/virología , Tráquea/citología , Tráquea/metabolismo , Tráquea/virología , Carga Viral
13.
PLoS One ; 10(10): e0141232, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26509529

RESUMEN

Cystic fibrosis (CF) is a genetic disease with many airway pathological features, including aberrant epithelial sodium channel (ENaC) function, persistent Pseudomonas aeruginosa (PA) infection and neutrophil-dominant inflammation. PA infection in CF airways is difficult to treat due to antibiotic resistance and other factors. Recently, α1-antitrypsin (A1AT) have been shown to be effective to reduce CF airway PA infection. However, there is a dearth of studies about the mechanisms underlying A1AT's therapeutic effects. The goal of our study is to provide an animal model of A1AT therapy in CF lungs. ENaC transgenic mice with PA infection were used as a CF-like model. Mice were intratracheally treated with PA or saline (control) in a fibrin plug. Two hours after PA infection, aerosolized A1AT were delivered to mouse lungs once daily. At day 1 and day 3 post PA infection, lung inflammation, PA load as well as host defence protein short palate, lung, and nasal epithelium clone 1 (SPLUNC1) were measured. At day 1 post PA infection when A1AT was delivered once to ENaC transgenic mouse lungs, A1AT did not reduce lung inflammation (e.g., neutrophils) and PA load. However, at day 3 post PA infection when ENaC transgenic mice received three repeated A1AT treatments, a significant decrease in airspace inflammation and PA load was observed. Although A1AT prevented the loss of SPLUNC1 in bronchoalveolar lavage fluid of PA-infected wild-type mice, it did not restore SPLUNC1 levels in ENaC transgenic mice. Our current study has provided a valid and quick A1AT therapeutic model in CF-like lungs that may serve as a platform for future mechanistic studies about how A1AT exerts beneficial effects in human CF patients.


Asunto(s)
Antibacterianos/uso terapéutico , Canales Epiteliales de Sodio/metabolismo , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/patogenicidad , alfa 1-Antitripsina/uso terapéutico , Animales , Canales Epiteliales de Sodio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Pseudomonas/genética
14.
J Clin Cell Immunol ; 5(5)2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25478290

RESUMEN

OBJECTIVE: MUC18 or CD146, a transmembrane glycoprotein, is mainly expressed by endothelial cells and smooth muscle cells where it serves as a cell-cell adhesion molecule. We have found MUC18 up-regulation in airway epithelial cells of patients with asthma and chronic obstructive pulmonary disease (COPD). However, the function of MUC18 in airway epithelial cells remains unclear. In the present study, we tested the hypothesis that MUC18 exerts a pro-inflammatory function during stimulation with a viral mimic polyI:C or human rhinovirus infection. METHODS: Normal human primary airway epithelial cells were transduced with lentivirus encoding MUC18 cDNA to over-express MUC18 or with GFP (control), and treated with polyI:C or HRV for detection of pro-inflammatory cytokine IL-8 and anti-viral gene IFN-ß. Additionally, we performed cell culture of human lung epithelial cell line NCIH292 cells to determine the mechanisms of MUC18 function. RESULTS: We found that MUC18 over-expression promoted IL-8 production, while it inhibited IFN-ß expression following polyI:C stimulation or HRV infection. Increased phosphorylation of MUC18 serines was observed in MUC18 over-expressing cells. Reduction of MUC18 serine phosphorylation by inhibiting ERK activity was associated with less production of IL-8 following polyI:C stimulation. CONCLUSIONS: Our results for the first time demonstrate MUC18's pro-inflammatory and anti-viral function in human airway epithelial cells.

15.
Eur J Endocrinol ; 169(4): 453-62, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23904282

RESUMEN

OBJECTIVE: Chemerin is an adipocyte-secreted hormone and has recently been associated with obesity and the metabolic syndrome. Although studies in rodents have outlined the aspects of chemerin's function and expression, its physiology and expression patterns are still to be elucidated in humans. METHODS: To evaluate for any day/night variation in chemerin secretion, we analyzed hourly serum samples from six females in the fed state. To examine whether energy deprivation affects chemerin levels, and whether this could be mediated through leptin, we analyzed samples from the same subjects in the fasting state while administering either placebo or leptin. To evaluate for any potential dose-effect relationship between leptin and chemerin, we administered increasing metreleptin doses to five females. A tissue array was used to study the expression of chemerin in different human tissues. Ex vivo treatment of human fat explants from three subjects with leptin was carried out to evaluate for any direct effect of leptin on adipocyte chemerin secretion. RESULTS: Chemerin does not display a day/night variation, while acute energy deprivation resulted in a significant drop in circulating chemerin levels by ∼42%. The latter was unaltered by metreleptin administration, and leptin administration did not affect the secretion of chemerin by human adipose tissue studied ex vivo. Chemerin was expressed primarily in the pancreas and liver. Chemerin receptor showed increased expression in the lymph nodes and the spleen. CONCLUSIONS: We outline for the first time chemerin expression and physiology in humans, which are different from those in mice.


Asunto(s)
Quimiocinas/genética , Ritmo Circadiano/fisiología , Hígado/fisiología , Páncreas/fisiología , Receptores de Quimiocina/genética , Adulto , Animales , Quimiocinas/sangre , Estudios Cruzados , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Ayuno/fisiología , Femenino , Humanos , Insulina/sangre , Péptidos y Proteínas de Señalización Intercelular , Leptina/administración & dosificación , Leptina/análogos & derivados , Leptina/sangre , Ratones , ARN Mensajero/metabolismo , Receptores de Quimiocina/metabolismo , Adulto Joven
16.
PLoS One ; 8(9): e72858, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24023786

RESUMEN

CONTEXT AND OBJECTIVE: The myokine irisin has been proposed to regulate energy homeostasis. Little is known about its association with metabolic parameters and especially with parameters influencing pathways of lipid metabolism. In the context of a clinical trial, an exploratory post hoc analysis has been performed in healthy subjects to determine whether simvastatin and/or ezetimibe influence serum irisin levels. The direct effects of simvastatin on irisin were also examined in primary human skeletal muscle cells (HSKMCs). DESIGN AND PARTICIPANTS: A randomized, parallel 3-group study was performed in 72 men with mild hypercholesterolemia and without apparent cardiovascular disease. Each group of 24 subjects received a 14-day treatment with either simvastatin 40 mg, ezetimibe 10 mg, or their combination. RESULTS: Baseline irisin concentrations were not significantly correlated with age, BMI, estimated GFR, thyroid parameters, glucose, insulin, lipoproteins, non-cholesterol sterols, adipokines, inflammation markers and various molecular markers of cholesterol metabolism. Circulating irisin increased significantly in simvastatin-treated but not in ezetimibe-treated subjects. The changes were independent of changes in LDL-cholesterol and were not correlated with changes in creatine kinase levels. In HSKMCs, simvastatin significantly increased irisin secretion as well as mRNA expression of its parent peptide hormone FNDC5. Simvastatin significantly induced cellular reactive oxygen species levels along with expression of pro- and anti-oxidative genes such as Nox2, and MnSOD and catalase, respectively. Markers of cellular stress such as atrogin-1 mRNA and Bax protein expression were also induced by simvastatin. Decreased cell viability and increased irisin secretion by simvastatin was reversed by antioxidant mito-TEMPO, implying in part that irisin is secreted as a result of increased mitochondrial oxidative stress and subsequent myocyte damage. CONCLUSIONS: Simvastatin increases irisin concentrations in vivo and in vitro. It remains to be determined whether this increase is a result of muscle damage or a protective mechanism against simvastatin-induced cellular stress. TRIAL REGISTRATION: ClinicalTrials.gov NCT00317993 NCT00317993.


Asunto(s)
Fibronectinas/sangre , Hipolipemiantes/farmacología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Adulto , Azetidinas/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ezetimiba , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/citología , Simvastatina/farmacología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA