Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Gastroenterology ; 134(7): 2091-100, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18466903

RESUMEN

BACKGROUNDS & AIMS: ATP8B1 is a phosphatidylserine flippase in the canalicular membrane; patients with mutations in ATP8B1 develop severe chronic (PFIC1) or periodic (BRIC1) cholestatic liver disease. We have observed that Atp8b1 deficiency leads to enhanced biliary cholesterol excretion. It has been established that biliary cholesterol excretion depends on transport by the heterodimer Abcg5/Abcg8. We hypothesized that the increased cholesterol output was due to enhanced extraction from the altered canalicular membrane rather than to higher Abcg5/Abcg8 activity. We therefore studied the relation between Abcg5/Abcg8 expression and biliary cholesterol excretion in mice lacking Atp8b1, Abcg8, or both (GF mice). METHODS: Bile formation was studied in LXR agonist-fed wild-type mice as well as mice lacking Atp8b1 or Abcg8, or in GF mice upon infusion of taurocholate. Bile samples were analyzed for cholesterol, bile salt, phospholipids, and ectoenzyme content. RESULTS: LXR agonist increased Abcg5/8 expression, and this was accompanied by increased biliary cholesterol output in both wild-type and Atp8b1(G308V/G308V) mice. However, Atp8b1(G308V/G308V) mice maintained higher cholesterol output. Although in Abcg8(-/-) mice biliary cholesterol output was severely reduced, GF mice displayed high biliary cholesterol output, which was comparable with wild-type mice. Bile of both Atp8b1(G308V/G308V) and GF mice displayed elevated levels of phosphatidylserine and sphingomyelin, indicating membrane stress. CONCLUSIONS: Our data demonstrate that the increased biliary cholesterol excretion in Atp8b1-deficient mice is independent of Abcg5/8 activity. This implicates that Atp8b1 deficiency leads to a decrease in the detergent resistance and subsequent nonspecific extraction of cholesterol from the canalicular membrane by bile salts.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfatasas/metabolismo , Bilis/metabolismo , Colesterol/metabolismo , Lipoproteínas/metabolismo , Hígado/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8 , Transportadoras de Casetes de Unión a ATP/genética , Adenosina Trifosfatasas/deficiencia , Adenosina Trifosfatasas/genética , Animales , Bilis/enzimología , Ácidos y Sales Biliares/sangre , Canalículos Biliares/metabolismo , Membrana Celular/metabolismo , Proteínas de Unión al ADN/agonistas , Proteínas de Unión al ADN/metabolismo , Hidrocarburos Fluorados , Lipoproteínas/deficiencia , Lipoproteínas/genética , Hígado/efectos de los fármacos , Hígado/enzimología , Receptores X del Hígado , Masculino , Fluidez de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Nucleares Huérfanos , Proteínas de Transferencia de Fosfolípidos , Fosfolípidos/metabolismo , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo , Sulfonamidas/farmacología , Factores de Tiempo , Regulación hacia Arriba
2.
Shock ; 42(1): 60-4, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24667617

RESUMEN

Innate immunity pathways are found to play an important role in ventilator-induced lung injury. We analyzed pulmonary expression of Toll-like receptor 2 (TLR2) in humans and mice and determined the role of TLR2 in the pathogenesis of ventilator-induced lung injury in mice. Toll-like receptor 2 gene expression was analyzed in human bronchoalveolar lavage fluid (BALF) cells and murine lung tissue after 5 h of ventilation. In addition, wild-type (WT) and TLR2 knockout (KO) mice were ventilated with either lower tidal volumes (VT) of 7 mL/kg with positive end-expiratory pressure (PEEP) or higher VT of 15 mL/kg without PEEP for 5 h. Spontaneously breathing mice served as controls. Total protein and immunoglobulin M levels in BALF, neutrophil influx into the alveolar compartment, and interleukin 6 (IL-6), IL-1ß, and keratinocyte-derived chemokine concentrations in lung tissue homogenates were measured. We observed enhanced TLR2 gene expression in BALF cells of ventilated patients and in lung tissue of ventilated mice. In WT mice, ventilation with higher VT without PEEP resulted in lung injury and inflammation with higher immunoglobulin M levels, neutrophil influx, and levels of inflammatory mediators compared with controls. In TLR2 KO mice, neutrophil influx and IL-6, IL-1ß, and keratinocyte-derived chemokine were enhanced by this ventilation strategy. Ventilation with lower VT with PEEP only increased neutrophil influx and was similar in WT and TLR2 KO mice. In summary, injurious ventilation enhances TLR2 expression in lungs. Toll-like receptor 2 deficiency does not protect lungs from ventilator-induced lung injury. In contrast, ventilation with higher VT without PEEP aggravates inflammation in TLR2 KO mice.


Asunto(s)
Respiración Artificial/métodos , Receptor Toll-Like 2/deficiencia , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Dióxido de Carbono/sangre , Regulación de la Expresión Génica , Humanos , Pulmón/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno/sangre , Presión Parcial , Respiración con Presión Positiva/efectos adversos , ARN Mensajero/genética , Respiración Artificial/efectos adversos , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 2/fisiología , Lesión Pulmonar Inducida por Ventilación Mecánica/etiología , Lesión Pulmonar Inducida por Ventilación Mecánica/genética
3.
Intensive Care Med Exp ; 2(1): 22, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26215707

RESUMEN

BACKGROUND: Mechanical ventilation (MV) can cause ventilator-induced lung injury (VILI). The innate immune response mediates this iatrogenic inflammatory condition. The receptor for advanced glycation end products (RAGE) is a multiligand receptor that can amplify immune and inflammatory responses. We hypothesized that RAGE signaling contributes to the pro-inflammatory state induced by MV. METHODS: RAGE expression was analyzed in lung brush and lavage cells obtained from ventilated patients and lung tissue of ventilated mice. Healthy wild-type (WT) and RAGE knockout (KO) mice were ventilated with relatively low (approximately 7.5 ml/kg) or high (approximately 15 ml/kg) tidal volume. Positive end-expiratory pressure was set at 2 cm H2O during both MV strategies. Also, WT and RAGE KO mice with lipopolysaccharide (LPS)-induced lung injury were ventilated with the above described ventilation strategies. In separate experiments, the contribution of soluble RAGE, a RAGE isoform that may function as a decoy receptor, in ventilated RAGE KO mice was investigated. Lung wet-to-dry ratio, cell and neutrophil influx, cytokine and chemokine concentrations, total protein levels, soluble RAGE, and high-mobility group box 1 (HMGB1) presence in lung lavage fluid were analyzed. RESULTS: MV was associated with increased RAGE mRNA levels in both human lung brush samples and lung tissue of healthy mice. In healthy high tidal volume-ventilated mice, RAGE deficiency limited inflammatory cell influx. Other VILI parameters were not affected. In our second set of experiments where we compared RAGE KO and WT mice in a 2-hit model, we observed higher pulmonary cytokine and chemokine levels in RAGE KO mice undergoing LPS/high tidal volume MV as compared to WT mice. Third, in WT mice undergoing the LPS/high tidal volume MV, we observed HMGB1 presence in lung lavage fluid. Moreover, MV increased levels of soluble RAGE in lung lavage fluid, with the highest levels found in LPS/high tidal volume-ventilated mice. Administration of soluble RAGE to LPS/high tidal volume-ventilated RAGE KO mice attenuated the production of inflammatory mediators. CONCLUSIONS: RAGE was not a crucial contributor to the pro-inflammatory state induced by MV. However, the presence of sRAGE limited the production of pro-inflammatory mediators in our 2-hit model of LPS and high tidal volume MV.

4.
Shock ; 40(4): 334-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23856920

RESUMEN

BACKGROUND: Use of aspirin (acetylsalicylic acid [ASA]) was found to improve outcome in animal models of acute lung injury (ALI) or its more severe form, acute respiratory distress syndrome. In patients with acute respiratory distress syndrome, data indicating a protective effect of ASA are less convincing. We hypothesize that ASA in a high dose is superior to low-dose ASA in preventing lung injury. Also, the effect on lung injury of inhibiting platelet activation by clopidogrel was investigated. METHODS: Acute lung injury was induced by intranasal instillation of 10 µg lipopolysaccharide (LPS). Before LPS, BALB/c mice were pretreated with either high dose of ASA (100 µg/g intraperitoneally, low-dose ASA (12.5 µg/g i.p), clopidogrel (50 µg/g i.p), or clopidogrel in combination with low dose of ASA. Controls received vehicle or LPS without intervention. Five hours after LPS, bronchoalveolar lavage fluid (BALF) and plasma were obtained. MEASUREMENTS AND MAIN RESULTS: All treatment regimens reduced neutrophil influx in the BALF compared with LPS controls (high-dose ASA 75% ± 2% [mean ± SD], low-dose ASA 86% ± 3%, clopidogrel 82% ± 1%, and low-dose ASA-clopidogrel 82% ± 3% vs. LPS control 88% ± 2%; P ≤ 0.05). High-dose ASA reduced BALF levels of protein compared with LPS controls (median [interquartile range], 0.2 [15] vs. 75 [20] pg/mL; P < 0.01), to a greater extent than after low-dose ASA (48 [32] pg/mL), clopidogrel (37 [23] pg/mL), or low-dose ASA-clopidogrel (57 [8] pg/mL). CONCLUSIONS: High-dose ASA is superior to low-dose ASA, clopidogrel, and to a combination of clopidogrel and low-dose ASA in attenuating LPS-induced lung injury in mice, suggesting high-dose ASA to be the antiplatelet therapy of choice in further research on preventing ALI.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Aspirina/administración & dosificación , Líquido del Lavado Bronquioalveolar/citología , Ticlopidina/análogos & derivados , Animales , Plaquetas/efectos de los fármacos , Clopidogrel , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos BALB C , Infiltración Neutrófila/efectos de los fármacos , Ticlopidina/administración & dosificación
5.
Crit Care Res Pract ; 2013: 435236, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23691294

RESUMEN

Background. Mechanical ventilation (MV) has the potential to initiate ventilator-induced lung injury (VILI). The pathogenesis of VILI has been primarily studied in animal models using more or less injurious ventilator settings. However, we speculate that duration of MV also influences severity and character of VILI. Methods. Sixty-four healthy C57Bl/6 mice were mechanically ventilated for 5 or 12 hours, using lower tidal volumes with positive end-expiratory pressure (PEEP) or higher tidal volumes without PEEP. Fifteen nonventilated mice served as controls. Results. All animals remained hemodynamically stable and survived MV protocols. In both MV groups, PaO2 to FiO2 ratios were lower and alveolar cell counts were higher after 12 hours of MV compared to 5 hours. Alveolar-capillary permeability was increased after 12 hours compared to 5 hours, although differences did not reach statistical significance. Lung levels of inflammatory mediators did not further increase over time. Only in mice ventilated with increased strain, lung compliance declined and wet to dry ratio increased after 12 hours of MV compared to 5 hours. Conclusions. Deleterious effects of MV are partly dependent on its duration. Even lower tidal volumes with PEEP may initiate aspects of VILI after 12 hours of MV.

6.
PLoS One ; 8(7): e68694, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874727

RESUMEN

BACKGROUND: Bacterial products add to mechanical ventilation in enhancing lung injury. The role of endogenous triggers of innate immunity herein is less well understood. S100A8/A9 proteins are released by phagocytes during inflammation. The present study investigates the role of S100A8/A9 proteins in ventilator-induced lung injury. METHODS: Pulmonary S100A8/A9 levels were measured in samples obtained from patients with and without lung injury. Furthermore, wild-type and S100A9 knock-out mice, naive and with lipopolysaccharide-induced injured lungs, were randomized to 5 hours of spontaneously breathing or mechanical ventilation with low or high tidal volume (VT). In addition, healthy spontaneously breathing and high VT ventilated mice received S100A8/A9, S100A8 or vehicle intratracheal. Furthermore, the role of Toll-like receptor 4 herein was investigated. RESULTS: S100A8/A9 protein levels were elevated in patients and mice with lung injury. S100A8/A9 levels synergistically increased upon the lipopolysaccharide/high VT MV double hit. Markers of alveolar barrier dysfunction, cytokine and chemokine levels, and histology scores were attenuated in S100A9 knockout mice undergoing the double-hit. Exogenous S100A8/A9 and S100A8 induced neutrophil influx in spontaneously breathing mice. In ventilated mice, these proteins clearly amplified inflammation: neutrophil influx, cytokine, and chemokine levels were increased compared to ventilated vehicle-treated mice. In contrast, administration of S100A8/A9 to ventilated Toll-like receptor 4 mutant mice did not augment inflammation. CONCLUSION: S100A8/A9 proteins increase during lung injury and contribute to inflammation induced by HVT MV combined with lipopolysaccharide. In the absence of lipopolysaccharide, high levels of extracellular S100A8/A9 still amplify ventilator-induced lung injury via Toll-like receptor 4.


Asunto(s)
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/fisiopatología , Animales , Líquido del Lavado Bronquioalveolar/química , Calgranulina B/efectos adversos , Calgranulina B/genética , Humanos , Ratones , Ratones Noqueados , Estadísticas no Paramétricas , Receptor Toll-Like 4/genética
7.
PLoS One ; 7(11): e50559, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226314

RESUMEN

INTRODUCTION: Uric acid released from injured tissue is considered a major endogenous danger signal and local instillation of uric acid crystals induces acute lung inflammation via activation of the NLRP3 inflammasome. Ventilator-induced lung injury (VILI) is mediated by the NLRP3 inflammasome and increased uric acid levels in lung lavage fluid are reported. We studied levels in human lung injury and the contribution of uric acid in experimental VILI. METHODS: Uric acid levels in lung lavage fluid of patients with acute lung injury (ALI) were determined. In a different cohort of cardiac surgery patients, uric acid levels were correlated with pulmonary leakage index. In a mouse model of VILI the effect of allopurinol (inhibits uric acid synthesis) and uricase (degrades uric acid) pre-treatment on neutrophil influx, up-regulation of adhesion molecules, pulmonary and systemic cytokine levels, lung pathology, and regulation of receptors involved in the recognition of uric acid was studied. In addition, total protein and immunoglobulin M in lung lavage fluid and pulmonary wet/dry ratios were measured as markers of alveolar barrier dysfunction. RESULTS: Uric acid levels increased in ALI patients. In cardiac surgery patients, elevated levels correlated significantly with the pulmonary leakage index. Allopurinol or uricase treatment did not reduce ventilator-induced inflammation, IκB-α degradation, or up-regulation of NLRP3, Toll-like receptor 2, and Toll-like receptor 4 gene expression in mice. Alveolar barrier dysfunction was attenuated which was most pronounced in mice pre-treated with allopurinol: both treatment strategies reduced wet/dry ratio, allopurinol also lowered total protein and immunoglobulin M levels. CONCLUSIONS: Local uric acid levels increase in patients with ALI. In mice, allopurinol and uricase attenuate ventilator-induced alveolar barrier dysfunction.


Asunto(s)
Alopurinol/farmacología , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/fisiopatología , Urato Oxidasa/farmacología , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Lesión Pulmonar Inducida por Ventilación Mecánica/fisiopatología , Lesión Pulmonar Aguda/metabolismo , Adulto , Alopurinol/uso terapéutico , Animales , Líquido del Lavado Bronquioalveolar , Permeabilidad Capilar/efectos de los fármacos , Proteínas Portadoras/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas I-kappa B/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Inhibidor NF-kappaB alfa , Proteína con Dominio Pirina 3 de la Familia NLR , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Urato Oxidasa/uso terapéutico , Ácido Úrico/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA