Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Lung ; 197(3): 361-369, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31028466

RESUMEN

PURPOSE: Vascular endothelial cells demonstrate severe injury in sepsis, and a reduction in endothelial inflammation would be beneficial. Inter-α-Inhibitor (IαI) is a family of abundant plasma proteins with anti-inflammatory properties and has been investigated in human and animal sepsis with encouraging results. We hypothesized that IαI may protect endothelia from sepsis-related inflammation. METHODS: IαI-deficient or sufficient mice were treated with endotoxin or underwent complement-induced lung injury. VCAM-1 and ICAM-1 expression was measured in blood and lung as marker of endothelial activation. Human endothelia were exposed to activated complement C5a with or without IαI. Blood from human sepsis patients was examined for VCAM-1 and ICAM-1 and levels were correlated with blood levels of IαI. RESULTS: IαI-deficient mice showed increased endothelial activation in endotoxin/sepsis- and complement-induced lung injury models. In vitro, levels of endothelial pro-inflammatory cytokines and cell growth factors induced by activated complement C5a were significantly decreased in the presence of IαI. This effect was associated with decreased ERK and NFκB activation. IαI levels were inversely associated with VCAM-1 and ICAM-1 levels in a human sepsis cohort. CONCLUSIONS: IαI ameliorates endothelial inflammation and may be beneficial as a treatment of sepsis.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , alfa-Globulinas/inmunología , Células Endoteliales/inmunología , Endotelio Vascular/inmunología , Pulmón/inmunología , Sepsis/inmunología , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , alfa-Globulinas/deficiencia , alfa-Globulinas/metabolismo , alfa-Globulinas/farmacología , Animales , Complemento C5a/inmunología , Complemento C5a/farmacología , Modelos Animales de Enfermedad , Selectina E/inmunología , Selectina E/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotoxinas/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas In Vitro , Inflamación , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Sistema de Señalización de MAP Quinasas , Ratones , FN-kappa B/efectos de los fármacos , FN-kappa B/inmunología , FN-kappa B/metabolismo , Sepsis/genética , Sepsis/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología , Molécula 1 de Adhesión Celular Vascular/metabolismo
2.
J Biol Chem ; 292(51): 20845-20858, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29122888

RESUMEN

Exposure to pollutants, such as ozone, exacerbates airway inflammation and hyperresponsiveness (AHR). TNF-stimulated gene 6 (TSG-6) is required to transfer inter-α-inhibitor heavy chains (HC) to hyaluronan (HA), facilitating HA receptor binding. TSG-6 is necessary for AHR in allergic asthma, because it facilitates the development of a pathological HA-HC matrix. However, the role of TSG-6 in acute airway inflammation is not well understood. Here, we hypothesized that TSG-6 is essential for the development of HA- and ozone-induced AHR. TSG-6-/- and TSG-6+/+ mice were exposed to ozone or short-fragment HA (sHA), and AHR was assayed via flexiVent. The AHR response to sHA was evaluated in the isolated tracheal ring assay in tracheal rings from TSG-6-/- or TSG-6+/+, with or without the addition of exogenous TSG-6, and with or without inhibitors of Rho-associated, coiled-coil-containing protein kinase (ROCK), ERK, or PI3K. Smooth-muscle cells from mouse tracheas were assayed in vitro for signaling pathways. We found that TSG-6 deficiency protects against AHR after ozone (in vivo) or sHA (in vitro and in vivo) exposure. Moreover, TSG-6-/- tracheal ring non-responsiveness to sHA was reversed by exogenous TSG-6 addition. sHA rapidly activated RhoA, ERK, and Akt in airway smooth-muscle cells, but only in the presence of TSG-6. Inhibition of ROCK, ERK, or PI3K/Akt blocked sHA/TSG-6-mediated AHR. In conclusion, TSG-6 is necessary for AHR in response to ozone or sHA, in part because it facilitates rapid formation of HA-HC complexes. The sHA/TSG-6 effect is mediated by RhoA, ERK, and PI3K/Akt signaling.


Asunto(s)
alfa-Globulinas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Ácido Hialurónico/metabolismo , Hipersensibilidad Respiratoria/etiología , Hipersensibilidad Respiratoria/metabolismo , Contaminantes Atmosféricos/toxicidad , alfa-Globulinas/química , Animales , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Modelos Animales de Enfermedad , Ácido Hialurónico/química , Técnicas In Vitro , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Modelos Biológicos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Ozono/toxicidad , Transducción de Señal/efectos de los fármacos , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
3.
Am J Physiol Lung Cell Mol Physiol ; 315(5): L787-L798, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30188746

RESUMEN

Allergic asthma is a major cause of morbidity in both pediatric and adult patients. Recent research has highlighted the role of hyaluronan (HA), an extracellular matrix glycosaminoglycan, in asthma pathogenesis. Experimental allergic airway inflammation and clinical asthma are associated with an increase of shorter fragments of HA (sHA), which complex with inter-α-inhibitor heavy chains (HCs) and induce inflammation and airway hyperresponsiveness (AHR). Importantly, the effects of sHA can be antagonized by the physiological counterpart high molecular weight HA (HMWHA). We used a mouse model of house dust mite-induced allergic airway inflammation and demonstrated that instilled HMWHA ameliorated allergic airway inflammation and AHR, even when given after the establishment of allergic sensitization and after challenge exposures. Furthermore, instilled HMWHA reduced the development of HA-HC complexes and the activation of Rho-associated, coiled-coil containing protein kinase 2. We conclude that airway application of HMWHA is a potential treatment for allergic airway inflammation.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Modelos Animales de Enfermedad , Ácido Hialurónico/administración & dosificación , Inflamación/prevención & control , Pyroglyphidae/patogenicidad , Hipersensibilidad Respiratoria/prevención & control , Animales , Femenino , Inflamación/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Peso Molecular , Hipersensibilidad Respiratoria/etiología
4.
Respir Res ; 17(1): 123, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27716205

RESUMEN

Cystic fibrosis (CF) is a chronic inflammatory disease that is affecting thousands of patients worldwide. Adjuvant anti-inflammatory treatment is an important component of cystic fibrosis treatment, and has shown promise in preserving lung function and prolonging life expectancy. Inhaled high molecular weight hyaluronan (HMW-HA) is reported to improve tolerability of hypertonic saline and thus increase compliance, and has been approved in some European countries for use as an adjunct to hypertonic saline treatment in cystic fibrosis. However, there are theoretical concerns that HMW-HA breakdown products may be pro-inflammatory. In this clinical pilot study we show that sputum cytokines in CF patients receiving HMW-HA are not increased, and therefore HMW-HA does not appear to adversely affect inflammatory status in CF airways.


Asunto(s)
Fibrosis Quística/tratamiento farmacológico , Ácido Hialurónico/administración & dosificación , Neumonía/tratamiento farmacológico , Solución Salina Hipertónica/administración & dosificación , Administración por Inhalación , Adolescente , Niño , Fibrosis Quística/diagnóstico , Fibrosis Quística/inmunología , Citocinas/inmunología , Femenino , Humanos , Ácido Hialurónico/efectos adversos , Ácido Hialurónico/química , Masculino , Peso Molecular , Proyectos Piloto , Neumonía/diagnóstico , Neumonía/inmunología , Solución Salina Hipertónica/efectos adversos , Esputo/inmunología , Resultado del Tratamiento , Adulto Joven
5.
Am J Physiol Lung Cell Mol Physiol ; 308(9): L891-903, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25747964

RESUMEN

Chlorine (Cl2) inhalation induces severe oxidative lung injury and airway hyperresponsiveness (AHR) that lead to asthmalike symptoms. When inhaled, Cl2 reacts with epithelial lining fluid, forming by-products that damage hyaluronan, a constituent of the extracellular matrix, causing the release of low-molecular-weight fragments (L-HA, <300 kDa), which initiate a series of proinflammatory events. Cl2 (400 ppm, 30 min) exposure to mice caused an increase of L-HA and its binding partner, inter-α-trypsin-inhibitor (IαI), in the bronchoalveolar lavage fluid. Airway resistance following methacholine challenge was increased 24 h post-Cl2 exposure. Intratracheal administration of high-molecular-weight hyaluronan (H-HA) or an antibody against IαI post-Cl2 exposure decreased AHR. Exposure of human airway smooth muscle (HASM) cells to Cl2 (100 ppm, 10 min) or incubation with Cl2-exposed H-HA (which fragments it to L-HA) increased membrane potential depolarization, intracellular Ca(2+), and RhoA activation. Inhibition of RhoA, chelation of intracellular Ca(2+), blockade of cation channels, as well as postexposure addition of H-HA, reversed membrane depolarization in HASM cells. We propose a paradigm in which oxidative lung injury generates reactive species and L-HA that activates RhoA and Ca(2+) channels of airway smooth muscle cells, increasing their contractility and thus causing AHR.


Asunto(s)
Asma/tratamiento farmacológico , Hiperreactividad Bronquial/tratamiento farmacológico , Ácido Hialurónico/uso terapéutico , Lesión Pulmonar/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , alfa-Globulinas/antagonistas & inhibidores , alfa-Globulinas/biosíntesis , alfa-Globulinas/inmunología , Animales , Hiperreactividad Bronquial/inmunología , Pruebas de Provocación Bronquial , Líquido del Lavado Bronquioalveolar/citología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio , Canales de Calcio/metabolismo , Células Cultivadas , Cloro/toxicidad , Activación Enzimática , Matriz Extracelular , Inflamación , Potenciales de la Membrana/efectos de los fármacos , Cloruro de Metacolina/toxicidad , Ratones , Ratones Endogámicos C57BL , Contracción Muscular/efectos de los fármacos , Miocitos del Músculo Liso , Técnicas de Placa-Clamp , Especies Reactivas de Oxígeno/metabolismo , Tráquea/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
6.
Am J Respir Cell Mol Biol ; 50(5): 953-62, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24303840

RESUMEN

Increased activity of lung epithelial sodium channels (ENaCs) contributes to the pathophysiology of cystic fibrosis (CF) by increasing the rate of epithelial lining fluid reabsorption. Inter-α-inhibitor (IαI), a serum protease inhibitor, may decrease ENaC activity by preventing its cleavage by serine proteases. High concentrations of IαI were detected in the bronchoalveolar lavage fluid (BALF) of children with CF and lower airway diseases. IαI decreased amiloride-sensitive (IENaC) but not cAMP-activated Cl(-) currents across confluent monolayers of rat ATII, and mouse nasal epithelial cells grew in primary culture by 45 and 25%, respectively. Changes in IENaC by IαI in ATII cells were accompanied by increased levels of uncleaved (immature) surface α-ENaC. IαI increased airway surface liquid depth overlying murine nasal epithelial cells to the same extent as amiloride, consistent with ENaC inhibition. Incubation of lung slices from C57BL/6, those lacking phenylalanine at position 508 (∆F508), or CF transmembrane conductance regulator knockout mice with IαI for 3 hours decreased the open probability of their ENaC channels by 50%. ∆F508 mice had considerably higher levels the amiloride-sensitive fractions of ENaC nasal potential difference (ENaC-NPD) than wild-type littermates and only background levels of IαI in their BALF. A single intranasal instillation of IαI decreased their ENaC-NPD 24 hours later by 25%. In conclusion, we show that IαI is present in the BALF of children with CF, is an effective inhibitor of ENaC proteolysis, and decreases ENaC activity in lung epithelial cells of ∆F508 mice.


Asunto(s)
alfa-Globulinas/metabolismo , Células Epiteliales/metabolismo , Agonistas del Canal de Sodio Epitelial/metabolismo , Canales Epiteliales de Sodio/metabolismo , Animales , Líquido del Lavado Bronquioalveolar , Células Cultivadas , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/metabolismo , Ratas , Xenopus laevis/metabolismo
7.
Am J Physiol Lung Cell Mol Physiol ; 306(11): L1045-55, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24748604

RESUMEN

Epithelial injury is often detected in lung allografts, however, its relation to rejection pathogenesis is unknown. We hypothesized that sterile epithelial injury can lead to alloimmune activation in the lung. We performed adoptive transfer of mismatched splenocytes into recombinant activating gene 1 (Rag1)-deficient mice to induce an alloimmune status and then exposed these mice to naphthalene to induce sterile epithelial injury. We evaluated lungs for presence of alloimmune lung injury, endoplasmic reticulum (ER) stress, and hyaluronan expression, examined the effect of ER stress induction on hyaluronan expression and lymphocyte trapping by bronchial epithelia in vitro, and examined airways from patients with bronchiolitis obliterans syndrome and normal controls histologically. We found that Rag1-deficient mice that received mismatched splenocytes and naphthalene injection displayed bronchial epithelial ER stress, peribronchial hyaluronan expression, and lymphocytic bronchitis. Bronchial epithelial ER stress led to the expression of lymphocyte-trapping hyaluronan cables in vitro. Blockade of hyaluronan binding ameliorated naphthalene-induced lymphocytic bronchitis. ER stress was present histologically in >40% of bronchial epithelia of BOS patients and associated with subepithelial hyaluronan deposition. We conclude that sterile bronchial epithelial injury in the context of alloimmunity can lead to sustained ER stress and promote allograft rejection through hyaluronan expression.


Asunto(s)
Bronquiolitis Obliterante/metabolismo , Células Epiteliales/inmunología , Ácido Hialurónico/metabolismo , Linfocitos/inmunología , Aloinjertos/inmunología , Animales , Bronquios/patología , Bronquiolitis Obliterante/inmunología , Células Cultivadas , Técnicas de Cocultivo , Estrés del Retículo Endoplásmico , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Humanos , Hialuronano Sintasas , Trasplante de Pulmón , Linfocitos/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mucosa Respiratoria/patología , Tenascina/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba
8.
Am J Respir Crit Care Med ; 181(7): 666-75, 2010 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-20007931

RESUMEN

RATIONALE: Ozone is a common environmental air pollutant that contributes to hospitalizations for respiratory illness. The mechanisms, which regulate ozone-induced airway hyperresponsiveness, remain poorly understood. We have previously reported that toll-like receptor 4 (TLR4)-deficient animals are protected against ozone-induced airway hyperresponsiveness (AHR) and that hyaluronan (HA) mediates ozone-induced AHR. However, the relation between TLR4 and hyaluronan in the airway response to ozone remains unexplored. OBJECTIVES: We hypothesized that HA acts as an endogenous TLR4 ligand for the development of AHR after ozone-induced environmental airway injury. METHODS: TLR4-deficient and wild-type C57BL/6 mice were exposed to either inhaled ozone or intratracheal HA and the inflammatory and AHR response was measured. MEASUREMENTS AND MAIN RESULTS: TLR4-deficient mice have similar levels of cellular inflammation, lung injury, and soluble HA levels as those of C57BL/6 mice after inhaled ozone exposure. However, TLR4-deficient mice are partially protected from AHR after ozone exposure as well as after direct intratracheal instillation of endotoxin-free low molecular weight HA. Similar patterns of TLR4-dependent cytokines were observed in the bronchial alveolar lavage fluid after exposure to either ozone or HA. Exposure to ozone increased immunohistological staining of TLR4 on lung macrophages. Furthermore, in vitro HA exposure of bone marrow-derived macrophages induced NF-kappaB and production of a similar pattern of proinflammatory cytokines in a manner dependent on TLR4. CONCLUSIONS: Our observations support the observation that extracellular matrix HA contributes to ozone-induced airways disease. Furthermore, our results support that TLR4 contributes to the biological response to HA by mediating both the production of proinflammatory cytokines and the development of ozone-induced AHR.


Asunto(s)
Hiperreactividad Bronquial/inducido químicamente , Hiperreactividad Bronquial/metabolismo , Ácido Hialurónico/metabolismo , Ozono/administración & dosificación , Receptor Toll-Like 4/metabolismo , Administración por Inhalación , Animales , Hiperreactividad Bronquial/patología , Líquido del Lavado Bronquioalveolar/química , Citocinas/biosíntesis , Ácido Hialurónico/administración & dosificación , Mediadores de Inflamación/metabolismo , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ozono/toxicidad , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Receptor Toll-Like 4/deficiencia
9.
Biomarkers ; 15(6): 488-97, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20500019

RESUMEN

We observed naphthyl-keratin adducts and dose-related metabolic enzyme induction at the mRNA level in reconstructed human epidermis in vitro after exposure to naphthalene. Immunofluorescence detection of 2-naphthyl-keratin-1 adducts confirmed the metabolism of naphthalene and adduction of keratin. We also observed naphthyl-keratin adducts in dermal tape-strip samples collected from naphthalene-exposed workers at levels ranging from 0.004 to 6.104 pmol adduct microg(-1) keratin. We have demonstrated the ability of the human skin to metabolize naphthalene and to form naphthyl-keratin adducts both in vitro and in vivo. The results indicate the potential use of keratin adducts as biomarkers of dermal exposure.


Asunto(s)
Epidermis/efectos de los fármacos , Epidermis/metabolismo , Queratinas/metabolismo , Naftalenos/farmacología , Biomarcadores , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Ensayo de Inmunoadsorción Enzimática , Epidermis/química , Técnica del Anticuerpo Fluorescente , Humanos , Hidrocarburos/química , Inmunohistoquímica , Exposición Profesional/análisis , ARN/biosíntesis , Piel/química , Piel/metabolismo
10.
Elife ; 92020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31989925

RESUMEN

Lung disease causes significant morbidity and mortality, and is exacerbated by environmental injury, for example through lipopolysaccharide (LPS) or ozone (O3). Toll-like receptors (TLRs) orchestrate immune responses to injury by recognizing pathogen- or danger-associated molecular patterns. TLR4, the prototypic receptor for LPS, also mediates inflammation after O3, triggered by endogenous hyaluronan. Regulation of TLR4 signaling is incompletely understood. TLR5, the flagellin receptor, is expressed in alveolar macrophages, and regulates immune responses to environmental injury. Using in vivo animal models of TLR4-mediated inflammations (LPS, O3, hyaluronan), we show that TLR5 impacts the in vivo response to LPS, hyaluronan and O3. We demonstrate that immune cells of human carriers of a dominant negative TLR5 allele have decreased inflammatory response to O3 exposure ex vivo and LPS exposure in vitro. Using primary murine macrophages, we find that TLR5 physically associates with TLR4 and biases TLR4 signaling towards the MyD88 pathway. Our results suggest an updated paradigm for TLR4/TLR5 signaling.


Immune cells in the lung help guard against infections. On the surface of these cells are proteins called TLR receptors that recognize dangerous molecules or DNA from disease-causing microbes such as bacteria. When the immune cells detect these invaders, the TLR receptors spring into action and trigger an inflammatory response to destroy the microbes. This inflammation usually helps the lung clear infections. But it can also be harmful and damage the lung, for example when inflammation is caused by non-infectious substances such as pollutants in the atmosphere. There are several TLR receptors that each recognize a specific molecule. In 2010, researchers showed that the receptor TLR4 is responsible for causing inflammation in the lung after exposure to pollution. Another receptor called TLR5 also helps activate the immune response in the lung. But it was unclear whether this receptor also plays a role in pollution-linked lung damage. Now, Hussain, Johnson, Sciurba et al. ­ including one of the researchers involved in the 2010 study ­ have investigated the role of TLR5 in immune cells from the lungs of humans and mice. The experiments showed that TLR5 works together with TLR4 and helps trigger an inflammatory response to both pollutants and bacteria. Hussain et al. found that people lacking a working TLR5 receptor (which make up 3­10% of the population) are less likely to experience lung inflammation when exposed to pollution or bacterial proteins that activate TLR4. These findings suggest that people without TLR5 may be protected from pollution-induced lung injury. Further research into the role of TLR5 could help develop genetic tests for identifying people who are more sensitive to damage from pollution. This information could then be used to determine the likelihood of a patient experiencing certain lung diseases.


Asunto(s)
Lesión Pulmonar , Factor 88 de Diferenciación Mieloide , Transducción de Señal , Receptor Toll-Like 4 , Receptor Toll-Like 5 , Animales , Células Cultivadas , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Polimorfismo de Nucleótido Simple/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 5/genética , Receptor Toll-Like 5/metabolismo
11.
Methods Mol Biol ; 1809: 301-314, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29987796

RESUMEN

Ozone is a major pollutant in the air we breathe, and elevated levels lead to significant morbidity and mortality. As the climate warms, levels of ozone are predicted to increase. Accordingly, studies to assess the mechanisms of ozone-induced lung diseases are paramount. This chapter describes mouse models of ozone exposure and methods for assessing the effects of ozone in the lungs. These include bronchoalveolar lavage, necropsy, and measurement of lung function. Lavage allows for assessment of cell infiltration, cytokine production, tissue damage and capillary leakage in the airspaces. Necropsy provides tissue for gene expression, histology, and protein assessment in the whole lung. Lung physiology is used to assess airway hyperresponsiveness, tissue and total lung resistance, compliance, and elastance.


Asunto(s)
Lesión Pulmonar/etiología , Ozono/efectos adversos , Animales , Lavado Broncoalveolar , Exposición a Riesgos Ambientales , Lesión Pulmonar/diagnóstico , Lesión Pulmonar/metabolismo , Ratones , Pruebas de Función Respiratoria
12.
Environ Health Perspect ; 120(6): 857-64, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22391508

RESUMEN

BACKGROUND: Individual genetic variation that results in differences in systemic response to xenobiotic exposure is not accounted for as a predictor of outcome in current exposure assessment models. OBJECTIVE: We developed a strategy to investigate individual differences in single-nucleotide polymorphisms (SNPs) as genetic markers associated with naphthyl-keratin adduct (NKA) levels measured in the skin of workers exposed to naphthalene. METHODS: The SNP-association analysis was conducted in PLINK using candidate-gene analysis and genome-wide analysis. We identified significant SNP-NKA associations and investigated the potential impact of these SNPs along with personal and workplace factors on NKA levels using a multiple linear regression model and the Pratt index. RESULTS: In candidate-gene analysis, a SNP (rs4852279) located near the CYP26B1 gene contributed to the 2-naphthyl-keratin adduct (2NKA) level. In the multiple linear regression model, the SNP rs4852279, dermal exposure, exposure time, task replacing foam, age, and ethnicity all were significant predictors of 2NKA level. In genome-wide analysis, no single SNP reached genome-wide significance for NKA levels (all p ≥ 1.05 × 10(-5)). Pathway and network analyses of SNPs associated with NKA levels were predicted to be involved in the regulation of cellular processes and homeostasis. CONCLUSIONS: These results provide evidence that a quantitative biomarker can be used as an intermediate phenotype when investigating the association between genetic markers and exposure-dose relationship in a small, well-characterized exposed worker population.


Asunto(s)
Aductos de ADN/metabolismo , Variación Genética/genética , Naftalenos/toxicidad , Exposición Profesional/estadística & datos numéricos , Polimorfismo de Nucleótido Simple/genética , Piel/metabolismo , Estudios de Asociación Genética , Estudio de Asociación del Genoma Completo , Humanos , Queroseno/análisis , Queroseno/toxicidad , Modelos Lineales , Naftoles
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA