Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros











Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L208-L216, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27913427

RESUMEN

Bronchopulmonary dysplasia (BPD) is a chronic lung injury characterized by impaired alveologenesis that may persist into adulthood. Rat models of BPD using varying degrees of hyperoxia to produce injury either cause early mortality or spontaneously recover following removal of the inciting stimulus, thus limiting clinical relevance. We sought to refine an established rat model induced by exposure to 60% O2 from birth by following hyperoxia with intermittent hypoxia (IH). Rats exposed from birth to air or 60% O2 until day 14 were recovered in air with or without IH (FIO2 = 0.10 for 10 min every 6 h) until day 28 Animals exposed to 60% O2 and recovered in air had no evidence of abnormal lung morphology on day 28 or at 10-12 wk. In contrast, 60% O2-exposed animals recovered in IH had persistently increased mean chord length, more dysmorphic septal crests, and fewer peripheral arteries. Recovery in IH also increased pulmonary vascular resistance, Fulton index, and arterial wall thickness. IH-mediated abnormalities in lung structure (but not pulmonary hypertension) persisted when reexamined at 10-12 wk, accompanied by increased pulmonary vascular reactivity and decreased exercise tolerance. Increased mean chord length secondary to IH was prevented by treatment with a peroxynitrite decomposition catalyst [5,10,15,20-Tetrakis(4-sulfonatophenyl)-21H,23H-porphyrin iron (III) chloride, 30 mg/kg/day, days 14-28], an effect accompanied by fewer inflammatory cells. We conclude that IH during recovery from hyperoxia-induced injury prevents recovery of alveologenesis and leads to changes in lung and pulmonary vascular function lasting into adulthood, thus more closely mimicking contemporary BPD.


Asunto(s)
Displasia Broncopulmonar/complicaciones , Displasia Broncopulmonar/patología , Hiperoxia/complicaciones , Hipoxia/complicaciones , Lesión Pulmonar/complicaciones , Alveolos Pulmonares/crecimiento & desarrollo , Alveolos Pulmonares/patología , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Catálisis , Modelos Animales de Enfermedad , Femenino , Hiperoxia/patología , Hipertensión Pulmonar/complicaciones , Hipoxia/patología , Lesión Pulmonar/patología , Masculino , Metaloporfirinas/farmacología , Ácido Peroxinitroso/metabolismo , Condicionamiento Físico Animal , Neumonía/complicaciones , Ratas Sprague-Dawley
2.
Am J Physiol Lung Cell Mol Physiol ; 309(1): L53-62, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26136527

RESUMEN

Neutrophil (PMNL) influx precedes lung macrophage (LM) influx into the lung following exposure of newborn pups to 60% O2. We hypothesized that PMNL were responsible for the signals leading to LM influx. This was confirmed when inhibition of PMNL influx with a CXC chemokine receptor-2 antagonist, SB-265610, also prevented the 60% O2-dependent LM influx, LM-derived nitrotyrosine formation, and pruning of small arterioles. Exposure to 60% O2 was associated with increased lung contents of neutrophil elastase and α-elastin, a marker of denatured elastin, and a decrease in elastin fiber density. This led us to speculate that neutrophil elastase-induced elastin fragments were the chemokines that led to a LM influx into the 60% O2-exposed lung. Inhibition of neutrophil elastase with sivelestat or elafin attenuated the LM influx. Sivelestat also attenuated the 60% O2-induced decrease in elastin fiber density. Daily injections of pups with an antibody to α-elastin prevented the 60% O2-dependent LM influx, impaired alveologenesis, and impaired small vessel formation. This suggests that neutrophil elastase inhibitors may protect against neonatal lung injury not only by preventing structural elastin degradation, but also by blocking elastin fragment-induced LM influx, thus preventing tissue injury from LM-derived peroxynitrite formation.


Asunto(s)
Elastina/metabolismo , Elastasa de Leucocito/metabolismo , Macrófagos/inmunología , Neutrófilos/inmunología , Oxígeno/toxicidad , Animales , Animales Recién Nacidos , Movimiento Celular/inmunología , Elafina/farmacología , Elastina/inmunología , Femenino , Glicina/análogos & derivados , Glicina/farmacología , Elastasa de Leucocito/antagonistas & inhibidores , Pulmón/patología , Lesión Pulmonar/inmunología , Exposición Materna , Oxígeno/farmacología , Ácido Peroxinitroso/biosíntesis , Compuestos de Fenilurea/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Interleucina-8B/antagonistas & inhibidores , Sulfonamidas/farmacología , Triazoles/farmacología , Remodelación Vascular
3.
Free Radic Biol Med ; 80: 1-11, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25514442

RESUMEN

Postnatal alveolarization is regulated by a number of growth factors, including insulin-like growth factor-I (IGF-I) acting through the insulin-like growth factor receptor-1 (IGF-R1). Exposure of the neonatal rat lung to 60% O2 for 14 days results in impairments of lung cell proliferation, secondary crest formation, and alveologenesis. This lung injury is mediated by peroxynitrite and is prevented by treatment with a peroxynitrite decomposition catalyst. We hypothesized that one of the mechanisms by which peroxynitrite induces lung injury in 60% O2 is through nitration and inactivation of critical growth factors or their receptors. Increased nitration of both IGF-I and IGF-R1 was evident in 60% O2-exposed lungs, which was reversible by concurrent treatment with a peroxynitrite decomposition catalyst. Increased nitration of the IGF-R1 was associated with its reduced activation, as assessed by IGF-R1 phosphotyrosine content. IGF-I displacement binding plots were conducted in vitro using rat fetal lung distal epithelial cells which respond to IGF-I by an increase in DNA synthesis. When IGF-I was nitrated to a degree similar to that observed in vivo there was minimal, if any, effect on IGF-I displacement binding. In contrast, nitrating cell IGF-R1 to a similar degree to that observed in vivo completely prevented specific binding of IGF-I to the IGF-R1, and attenuated an IGF-I-mediated increase in DNA synthesis. Additionally, we hypothesized that peroxynitrite also impairs alveologenesis by being an upstream regulator of the growth inhibitor, TGFß1. That 60% O2-induced impairment of alveologenesis was mediated in part by TGFß1 was confirmed by demonstrating an improvement in secondary crest formation when 60% O2-exposed pups received concurrent treatment with the TGFß1 activin receptor-like kinase, SB 431542. That the increased TGFß1 content in lungs of pups exposed to 60% O2 was regulated by peroxynitrite was confirmed by its attenuation by concurrent treatment with a peroxynitrite decomposition catalyst. We conclude that peroxynitrite contributes to the impaired alveologenesis observed following the exposure of neonatal rats to 60% O2 both by preventing binding of IGF-I to the IGF-R1, secondary to nitration of the IGF-R1, and by causing an up-regulation of the growth inhibitor, TGFß1.


Asunto(s)
Células Epiteliales/metabolismo , Hiperoxia/metabolismo , Lesión Pulmonar/metabolismo , Alveolos Pulmonares/metabolismo , Receptores de Somatomedina/metabolismo , Factor de Crecimiento Transformador beta1/agonistas , Animales , Animales Recién Nacidos , Benzamidas/farmacología , ADN/biosíntesis , Dioxoles/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Regulación de la Expresión Génica , Hiperoxia/complicaciones , Hiperoxia/tratamiento farmacológico , Hiperoxia/patología , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Metaloporfirinas/farmacología , Nitratos/metabolismo , Ácido Peroxinitroso/antagonistas & inhibidores , Ácido Peroxinitroso/metabolismo , Cultivo Primario de Células , Unión Proteica , Alveolos Pulmonares/crecimiento & desarrollo , Alveolos Pulmonares/patología , Ratas , Ratas Sprague-Dawley , Receptores de Somatomedina/genética , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
4.
Pediatr Pulmonol ; 49(10): 991-1002, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24273102

RESUMEN

RATIONALE: Use of the anti-inflammatory agent dexamethasone in premature infants with bronchopulmonary dysplasia has been curtailed, and no alternative anti-inflammatory agents are approved for this use. Our objective was to use a neonatal rat model of bronchopulmonary dysplasia to determine if an highly selective cyclooxygenase-2 inhibitor, 5,5-dimethyl-3-(3-fluorophenyl)4-(4-methylsulfonyl)phenyl-2(5H)-furanone (DFU; 10 µg/g body weight), could prevent inflammatory cell influx and protect against lung injury. METHODS: Neonatal rats exposed to air or 60% O2 for 14 days from birth either received daily i.p. injections of (i) vehicle or DFU or (ii) vehicle or an EP(1) receptor antagonist, SC-19220. RESULTS: DFU attenuated the lung macrophage and neutrophil influx, prevented interstitial thickening and prevented the loss of peripheral blood vessels induced by 60% O2 , but did not protect against the variance in alveolar diameter induced by 60% O2 . Exposure to 60% O2 caused both an increase in lung prostaglandin E2 content and a reduction in lung mesenchymal cell mass which was reversed by DFU. Prostaglandin E2 binding to the EP(1) receptor inhibited DNA synthesis in cultures of lung fibroblasts in a dose dependent fashion. Treatment with SC-19220 attenuated the reduction in lung mesenchymal mass observed following exposure of rat pups to 60% O2 . CONCLUSIONS: An highly selective cyclooxygenase-2 inhibitor is an effective anti-inflammatory substitute for dexamethasone for preventing phagocyte influx into the neonatal lung during 60% O2 -mediated lung injury, and can modify the severity of that injury.


Asunto(s)
Displasia Broncopulmonar/tratamiento farmacológico , Inhibidores de la Ciclooxigenasa 2/farmacología , Furanos/farmacología , Oxígeno/administración & dosificación , Animales , Animales Recién Nacidos , Dinoprostona/metabolismo , Pulmón/metabolismo , Ratas Sprague-Dawley
5.
Am J Physiol Lung Cell Mol Physiol ; 303(1): L75-87, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22582114

RESUMEN

Bleomycin-induced lung injury is characterized in the neonatal rat by inflammation, arrested lung growth, and pulmonary hypertension (PHT), as observed in human infants with severe bronchopulmonary dysplasia. Inhalation of CO(2) (therapeutic hypercapnia) has been described to limit cytokine production and to have anti-inflammatory effects on the injured lung; we therefore hypothesized that therapeutic hypercapnia would prevent bleomycin-induced lung injury. Spontaneously breathing rat pups were treated with bleomycin (1 mg/kg/d ip) or saline vehicle from postnatal days 1-14 while being continuously exposed to 5% CO(2) (Pa(CO(2)) elevated by 15-20 mmHg), 7% CO(2) (Pa(CO(2)) elevated by 35 mmHg), or normocapnia. Bleomycin-treated animals exposed to 7%, but not 5%, CO(2), had significantly attenuated lung tissue macrophage influx and PHT, as evidenced by normalized pulmonary vascular resistance and right ventricular systolic function, decreased right ventricular hypertrophy, and attenuated remodeling of pulmonary resistance arteries. The level of CO(2) neither prevented increased tissue neutrophil influx nor led to improvements in decreased lung weight, septal thinning, impaired alveolarization, or decreased numbers of peripheral arteries. Bleomycin led to increased expression and content of lung tumor necrosis factor (TNF)-α, which was found to colocalize with tissue macrophages and to be attenuated by exposure to 7% CO(2). Inhibition of TNF-α signaling with the soluble TNF-2 receptor etanercept (0.4 mg/kg ip from days 1-14 on alternate days) prevented bleomycin-induced PHT without decreasing tissue macrophages and, similar to CO(2), had no effect on arrested alveolar development. Our findings are consistent with a preventive effect of therapeutic hypercapnia with 7% CO(2) on bleomycin-induced PHT via attenuation of macrophage-derived TNF-α. Neither tissue macrophages nor TNF-α appeared to contribute to arrested lung development induced by bleomycin. That 7% CO(2) normalized pulmonary vascular resistance and right ventricular function without improving inhibited airway and vascular development suggests that vascular hypoplasia does not contribute significantly to functional changes of PHT in this model.


Asunto(s)
Hipercapnia/fisiopatología , Hipertensión Pulmonar/prevención & control , Macrófagos/metabolismo , Alveolos Pulmonares/fisiopatología , Arteria Pulmonar/fisiopatología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Animales Recién Nacidos/metabolismo , Animales Recién Nacidos/fisiología , Bleomicina/toxicidad , Dióxido de Carbono/administración & dosificación , Dióxido de Carbono/sangre , Hipercapnia/sangre , Hipercapnia/inducido químicamente , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/metabolismo , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/fisiopatología , Inflamación/metabolismo , Inflamación/fisiopatología , Pulmón/metabolismo , Pulmón/fisiopatología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/fisiopatología , Lesión Pulmonar/prevención & control , Neutrófilos/metabolismo , Alveolos Pulmonares/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Ratas , Función Ventricular Derecha/efectos de los fármacos
6.
Am J Physiol Lung Cell Mol Physiol ; 300(4): L534-47, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21239531

RESUMEN

Survivors of moderate-to-severe bronchopulmonary dysplasia have impaired alveologenesis lasting at least into early adult life. The mechanisms underlying this long-term effect are unknown. We hypothesized that short-term inhibition of growth factor-mediated early alveolar formation would result in a long-term impairment of subsequent alveologenesis. Neonatal rats were injected daily with the platelet-derived growth factor (PDGF) receptor antagonist, imatinib mesylate, from day 1-7 of life, to inhibit the early alveolar formation occurring by in-growth of secondary crests into precursor saccules. The pups were then allowed to recover for 7, 14, 21, or 58 days. In imatinib-treated pups, DNA synthesis in total lung cells, and specifically in cells of secondary crests, was reduced at day 8 of life, had rebounded on day 14 of life but was then again reduced by day 28 of life. At day 8 of life, imatinib-treated pups had impaired alveologenesis as reflected by a decrease in secondary crests, an increase in alveolar size, and an overall decrease in both estimated alveolar number and generations compared with age-matched controls. No meaningful recovery was observed, even after a 21- or 58-day recovery period. The lungs of imatinib-treated pups had increased fibulin-5 content and an abnormal deposition of elastin. We conclude that reduced signaling through the PDGF pathways, at an early stage of alveologenesis, can result in long-lasting changes in lung architecture. A likely mechanism is through impaired formation of the elastin scaffold required for alveolarization.


Asunto(s)
Organogénesis/efectos de los fármacos , Piperazinas/farmacología , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/crecimiento & desarrollo , Pirimidinas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Benzamidas , Western Blotting , Peso Corporal/efectos de los fármacos , Bromodesoxiuridina/metabolismo , Recuento de Células , Densitometría , Elastina/metabolismo , Mesilato de Imatinib , Tamaño de los Órganos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Piperazinas/administración & dosificación , Proteínas Proto-Oncogénicas c-kit/metabolismo , Alveolos Pulmonares/patología , Pirimidinas/administración & dosificación , Ratas , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Tiempo
7.
Am J Physiol Lung Cell Mol Physiol ; 300(3): L319-29, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21148793

RESUMEN

During early postnatal alveolar formation, the lung tissue of rat pups undergoes a physiological remodeling involving apoptosis of distal lung cells. Exposure of neonatal rats to severe hyperoxia (≥95% O(2)) both arrests lung growth and results in increased lung cell apoptosis. In contrast, exposure to moderate hyperoxia (60% O(2)) for 14 days does not completely arrest lung cell proliferation and is associated with parenchymal thickening. On the basis of similarities in lung architecture observed following either exposure to 60% O(2), or pharmacological inhibition of physiological apoptosis, we hypothesized that exposure to 60% O(2) would result in an inhibition of physiological lung cell apoptosis. Consistent with this hypothesis, we observed that the parenchymal thickening induced by exposure to 60% O(2) was associated with decreased numbers of apoptotic cells, increased expressions of the antiapoptotic regulator Bcl-xL, and the putative antiapoptotic protein survivin, and decreased expressions of the proapoptotic cleaved caspases-3 and -7. In summary, exposure of the neonatal rat lung to moderate hyperoxia results in an inhibition of physiological apoptosis, which contributes to the parenchymal thickening observed in the resultant lung injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Oxígeno/farmacología , Transducción de Señal/efectos de los fármacos , Aire , Animales , Animales Recién Nacidos , Western Blotting , Caspasa 3/metabolismo , Recuento de Células , Muerte Celular/efectos de los fármacos , Femenino , Inmunohistoquímica , Pulmón/efectos de los fármacos , Pulmón/enzimología , Pulmón/patología , Ratas , Ratas Sprague-Dawley , Estaurosporina/farmacología
8.
Free Radic Res ; 45(3): 359-65, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21034359

RESUMEN

Neonatal lung injury has been induced experimentally by infusion of multivitamin-containing light-exposed parenteral nutrition (PN) solutions. The objective was to explore the role of ascorbate in toxic effects of light-exposed PN on primary cultured foetal rat lung epithelial cells. Hydroperoxides were measured in 3% amino acid solutions at baseline, immediately after addition of either multivitamins or ascorbate alone (400 µg/mL) and again after a 24-h period of exposure to (or protection from) ambient light. Cellular toxicity was assessed by [C(14)]adenine release. Multivitamins or ascorbate alone increased hydroperoxides in PN, which was attenuated by light protection. Light-exposed PN containing multivitamins was more toxic to cells than baseline or light-protected PN. Exposure to ascorbate at concentrations both lower (< 5 µg/mL) and higher (> 1000 µg/mL) than normally contained in PN-induced oxidant-mediated cell death, as indicated by protective effects of hydroperoxide and hydroxyl radical scavengers. This study concludes that ascorbate generates toxic amounts of peroxide in PN solutions. The types and physiological importance of hydroperoxides induced by pro-oxidant effects of ascorbate require further evaluation in vivo.


Asunto(s)
Ácido Ascórbico/toxicidad , Células Epiteliales/efectos de los fármacos , Pulmón/citología , Soluciones para Nutrición Parenteral/toxicidad , Animales , Células Cultivadas , Depuradores de Radicales Libres/farmacología , Peróxido de Hidrógeno/farmacología , Luz , Ratas , Ratas Sprague-Dawley
9.
Free Radic Biol Med ; 49(7): 1182-91, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20647043

RESUMEN

Exposure of newborn rats to 60% O2 for 14days results in a chronic neonatal lung injury characterized by parenchymal thickening, impaired alveolarization, evidence of pulmonary hypertension, and pulmonary vascular pruning. The contribution of peroxynitrite to this injury was assessed by treating pups with a peroxynitrite decomposition catalyst, 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinato iron(III) chloride (FeTPPS), at 30microg/g/day. Body and lung weights and postfixation lung volumes were all slightly, but significantly, reduced by exposure to 60% O2 and this was attenuated by a concurrent FeTPPS intervention. The FeTPPS intervention had no impact on increased neutrophil or macrophage influx into the lung, but attenuated 60% O2-induced reductions in the lung contents of vascular endothelial-derived growth factor, its receptor-2, and angiopoietin and increases in 8-isoprostane and preproendothelin-1. The 60% O2-induced parenchymal thickening and impairment of alveologenesis, as well as vascular pruning and peripheral vessel medial wall thickening, were attenuated by FeTPPS, despite a persistent inflammatory cell influx. Pups exposed to 60% O2 and treated with FeTPPS had enhanced alveolar formation relative to control pups. We conclude that peroxynitrite plays a critical role in the development of chronic neonatal lung injury.


Asunto(s)
Lesión Pulmonar/prevención & control , Pulmón/metabolismo , Metaloporfirinas/administración & dosificación , Ácido Peroxinitroso/metabolismo , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Animales , Animales Recién Nacidos , Catálisis , Hipertensión Pulmonar , Inmunohistoquímica , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/fisiopatología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Metaloporfirinas/química , Neovascularización Fisiológica/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Oxígeno/administración & dosificación , Ácido Peroxinitroso/química , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Pediatr Res ; 67(2): 177-82, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19858775

RESUMEN

Rho-kinase (ROCK) inhibitors prevent pulmonary hypertension (PHT) in adult rodents, but little is known about their effects on the neonatal lung. Our objective was to examine the effects of ROCK inhibition on chronic hypoxia (CH)-induced PHT and abnormal lung structure in the neonatal rat. Pups were exposed to air or CH from postnatal d 1-14 while receiving Y-27632 (5 or 10 mg x kg(-1) x d(-1)), fasudil (20 mg x kg(-1) x d(-1)), or saline intraperitoneally. Relative to air, CH-exposed pups had increased pulmonary vascular resistance, right ventricular hypertrophy, arterial medial wall thickening, and abnormal distal airway morphology characterized by septal thinning and decreased secondary septation. Treatment with 10 mg/kg Y-27632 or fasudil attenuated the structural and hemodynamic changes of PHT while having no effect on septal thinning or inhibited secondary septation. In addition, Y-27632 (10 mg/kg) and fasudil augmented CH-induced somatic growth restriction. Pulmonary arteries of CH-exposed pups had increased ROCK activity, up-regulated expression of PDGF-BB and increased smooth muscle DNA synthesis, all of which were attenuated by treatment with 10 mg/kg Y-27632. Systemically administered ROCK inhibitors prevented PHT in the CH-exposed neonatal rat but at the cost of inhibited somatic growth. Limiting effects on vascular remodeling likely resulted, in major part, from attenuated vascular PDGF-BB/beta-receptor signaling.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Amidas/farmacología , Antihipertensivos/farmacología , Hiperoxia/tratamiento farmacológico , Hipertensión Pulmonar/prevención & control , Pulmón/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Animales , Animales Recién Nacidos , Becaplermina , Peso Corporal , Enfermedad Crónica , Replicación del ADN , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hiperoxia/complicaciones , Hiperoxia/enzimología , Hiperoxia/patología , Hiperoxia/fisiopatología , Hipertensión Pulmonar/enzimología , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/enzimología , Hipertrofia Ventricular Derecha/etiología , Hipertrofia Ventricular Derecha/prevención & control , Pulmón/irrigación sanguínea , Pulmón/enzimología , Pulmón/patología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Tamaño de los Órganos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/enzimología , Arteria Pulmonar/patología , Ratas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Resistencia Vascular/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo
12.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L920-30, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19749000

RESUMEN

Permissive hypercapnia, achieved using low tidal volume ventilation, has been an effective protective strategy in patients with acute respiratory distress syndrome. To date, no such protective effect has been demonstrated for the chronic neonatal lung injury, bronchopulmonary dysplasia. The objective of our study was to determine whether evolving chronic neonatal lung injury, using a rat model, is resistant to the beneficial effects of hypercapnia or simply requires a less conservative approach to hypercapnia than that applied clinically to date. Neonatal rats inhaled air or 60% O2 for 14 days with or without 5.5% CO2. Lung parenchymal neutrophil and macrophage numbers were significantly increased by hyperoxia alone, which was associated with interstitial thickening and reduced secondary crest formation. The phagocyte influx, interstitial thickening, and impaired alveolar formation were significantly attenuated by concurrent hypercapnia. Hyperoxic pups that received 5.5% CO2 had a significant increase in alveolar number relative to air-exposed pups. Increased tyrosine nitration, a footprint for peroxynitrite-mediated reactions, arteriolar medial wall thickening, and both reduced small peripheral pulmonary vessel number and VEGF and angiopoietin-1 (Ang-1) expression, which were observed with hyperoxia, was attenuated by concurrent hypercapnia. We conclude that evolving chronic neonatal lung injury in a rat model is responsive to the beneficial effects of hypercapnia. Inhaled 5.5% CO2 provided a significant degree of protection against parenchymal and vascular injury in an animal model of chronic neonatal lung injury likely due, at least in part, to its inhibition of a phagocyte influx.


Asunto(s)
Vasos Sanguíneos/fisiología , Hipercapnia/complicaciones , Hipercapnia/fisiopatología , Lesión Pulmonar/fisiopatología , Lesión Pulmonar/terapia , Actinas/metabolismo , Aire , Animales , Animales Recién Nacidos , Análisis de los Gases de la Sangre , Vasos Sanguíneos/efectos de los fármacos , Dióxido de Carbono/farmacología , Elastina/metabolismo , Técnica del Anticuerpo Fluorescente , Frecuencia Cardíaca/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/patología , Lesión Pulmonar/complicaciones , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Tamaño de los Órganos/efectos de los fármacos , Peroxidasa/metabolismo , Ratas , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo , Factor de von Willebrand/metabolismo
13.
Pediatr Res ; 66(3): 260-5, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19542903

RESUMEN

IL-1 beta, a proinflammatory cytokine, may contribute to the development of the chronic neonatal lung injury, bronchopulmonary dysplasia. Chronic neonatal lung injury was induced in rats, by exposure to 60% O2 for 14 d from birth, to determine whether pulmonary IL-1 expression was up-regulated and, if so, whether a daily s.c. IL-1 receptor antagonist injections would be protective. Exposure to 60% O2 for 14 d caused pulmonary neutrophil and macrophage influx, increased tissue fraction and tyrosine nitration, reduced VEGF-A and angiopoietin-1 expression, and reduced small vessel (20-65 microm) and alveolar numbers. Lung IL-1 alpha and -1 beta contents were increased after a 4-d exposure to 60% O2. IL-1 receptor antagonist treatment attenuated the 60% O2-dependent neutrophil influx, the increased tissue fraction, and the reduced alveolar number. Treatment did not restore VEGF-A or angiopoietin-1 expression and only partially attenuated the reduced vessel number in 60% O2-exposed pups. It also caused a paradoxical increase in macrophage influx and a reduction in small vessels in air-exposed pups. We conclude that antagonism of IL-1-mediated effects can, in major part, protect against lung injury in a rat model of 60% O2-induced chronic neonatal lung injury.


Asunto(s)
Lesión Pulmonar/etiología , Oxígeno/efectos adversos , Receptores de Interleucina-1/metabolismo , Angiopoyetina 1/metabolismo , Animales , Animales Recién Nacidos , Peso Corporal , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-1beta/metabolismo , Pulmón/citología , Pulmón/metabolismo , Pulmón/patología , Macrófagos/citología , Macrófagos/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Tamaño de los Órganos , Fagocitos/metabolismo , Ratas , Tirosina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
Cell Res ; 18(6): 649-63, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18475289

RESUMEN

Airway inflammation is the hallmark of many respiratory disorders, such as asthma and cystic fibrosis. Changes in airway gene expression triggered by inflammation play a key role in the pathogenesis of these diseases. Genetic linkage studies suggest that ESE-2 and ESE-3, which encode epithelium-specific Ets-domain-containing transcription factors, are candidate asthma susceptibility genes. We report here that the expression of another member of the Ets family transcription factors ESE-1, as well as ESE-3, is upregulated by the inflammatory cytokines interleukin-1beta (IL-1beta) and tumor necrosis factor-alpha (TNF-alpha) in bronchial epithelial cell lines. Treatment of these cells with IL-1beta and TNF-alpha resulted in a dramatic increase in mRNA expression for both ESE-1 and ESE-3. We demonstrate that the induced expression is mediated by activation of the transcription factor NF-kappaB. We have characterized the ESE-1 and ESE-3 promoters and have identified the NF-kappaB binding sequences that are required for the cytokine-induced expression. In addition, we also demonstrate that ESE-1 upregulates ESE-3 expression and downregulates its own induction by cytokines. Finally, we have shown that in Elf3 (homologous to human ESE-1) knockout mice, the expression of the inflammatory cytokine interleukin-6 (IL-6) is downregulated. Our findings suggest that ESE-1 and ESE-3 play an important role in airway inflammation.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Epiteliales/metabolismo , Epitelio/metabolismo , Proteínas Proto-Oncogénicas/genética , Sistema Respiratorio/metabolismo , Sistema Respiratorio/patología , Factores de Transcripción/genética , Animales , Secuencia de Bases , Línea Celular , Citocinas/farmacología , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Epitelio/efectos de los fármacos , Humanos , Inflamación/genética , Mediadores de Inflamación/farmacología , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Especificidad de Órganos/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ets , Sistema Respiratorio/efectos de los fármacos , Eliminación de Secuencia , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
15.
Pediatr Res ; 63(3): 232-8, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18091341

RESUMEN

Mesenchymal cell-derived FGF-7 (fibroblast growth factor-7) induces proliferation in both epithelial and endothelial cells. We found FGF-7 to be expressed in the lungs of neonatal rats from birth to d 14 of age. A role for FGF-7 in early postnatal lung growth and alveolar formation, by an action on type II pneumocytes, has been excluded by the work of others. However, a role through an action of FGF-7 on other cell types has not been excluded. We used intraperitoneal injections of neutralizing antibodies on d 3, 4, and 5 of life to inhibit binding of FGF-7 to its receptors, and assessed alveolar formation on d 6 of life. This intervention inhibited DNA synthesis in, and number of, alveoli-forming secondary crests, resulting in a significantly reduced alveolar number. This failure of alveolar formation was associated with a reduction in the number of small blood vessels in the lung periphery. We conclude that FGF-7, most likely through its effect on the vascular bed, is required for normal early postnatal alveolar formation from secondary crests.


Asunto(s)
Células Endoteliales/metabolismo , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Alveolos Pulmonares/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Animales , Animales Recién Nacidos , Anticuerpos/administración & dosificación , Especificidad de Anticuerpos , Proliferación Celular , Replicación del ADN , Factor 7 de Crecimiento de Fibroblastos/inmunología , Factor de Crecimiento de Hepatocito/metabolismo , Inyecciones Intraperitoneales , Alveolos Pulmonares/irrigación sanguínea , Alveolos Pulmonares/citología , Alveolos Pulmonares/crecimiento & desarrollo , Ratas , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
Am J Physiol Lung Cell Mol Physiol ; 292(3): L725-41, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17114281

RESUMEN

Classical tissue recombination experiments have reported that at early gestation both tracheal and distal lung epithelium have the plasticity to respond to mesenchymal signals. Herein we examined the role of epithelial-mesenchymal interactions in maintaining epithelial differentiation at late (E19-E21, term = 22 days) fetal gestation in the rat. Isolated distal lung epithelial cells were recombined with mesenchymal cells from lung, skin, and intestine, and the homotypic or heterotypic recombinant cell aggregates were cultured for up to 5 days. Recombining lung epithelial cells with mesenchyme from various sources induced a morphological pattern that was specific to the type of inducing mesenchyme. In situ analysis of surfactant protein (SP)-C, SP-B, and Clara cell secretory protein (CCSP) expression, as well as SP-C and CCSP promoter transactivation experiments, revealed that distal lung epithelium requires lung mesenchyme to maintain the alveolar, but not bronchiolar, phenotype. Incubation of lung recombinants with an anti-FGF7 antibody resulted in a partial inhibition of mesenchyme-induced SP-C promoter transactivation. Immunoreactivity for Delta and Lunatic fringe, components of the Notch pathway that regulates cell differentiation, was downregulated in the heterotypic recombinants. In contrast, Hes1 mRNA expression was increased in these recombinants. Cumulatively, these results suggest that at late fetal gestation, distal lung epithelial cells are not fully committed to a specific phenotype and still have the plasticity to respond to various signals. Their alveolar phenotype is likely maintained by Notch/Notch ligand interactions and mesenchymal factors, including FGF7.


Asunto(s)
Células Epiteliales/metabolismo , Desarrollo Fetal , Pulmón/embriología , Mesodermo/metabolismo , Animales , Células Cultivadas , Células Epiteliales/ultraestructura , Factor 7 de Crecimiento de Fibroblastos/genética , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Técnicas para Inmunoenzimas , Hibridación in Situ , Pulmón/citología , Pulmón/ultraestructura , Masculino , Mesodermo/citología , Mesodermo/ultraestructura , Fenotipo , Surfactantes Pulmonares/metabolismo , Sondas ARN , Ratas , Ratas Wistar , Uteroglobina/genética , Uteroglobina/metabolismo
17.
Pediatr Res ; 60(3): 299-303, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16923948

RESUMEN

Inflammation contributes greatly to the pathogenesis of bronchopulmonary dysplasia. In previous studies, we showed that blocking neutrophil influx by treatment with SB265610, a selective CXCR2 antagonist, could partly reduce superoxide accumulation and preserve alveolar development in 60% O(2)-exposed newborn rats. The purpose of this study was to further investigate the role of neutrophils in the formation of reactive oxygen and nitrogen species mediating hyperoxia-impaired lung development. We found that hydroxyl radical formation and lipid peroxidation in rat lungs were significantly increased during 60% O(2) exposure. These increases were attenuated by the administration of SB265610. In addition, SB265610 largely inhibited protein nitration induced by hyperoxia. SB265610 partly prevented the hyperoxia-enhanced bronchoalveolar lavage (BAL) protein content in 60% O(2)-exposed animals. Our results demonstrate that neutrophils have a pivotal role in hydroxyl radical formation, lipid peroxidation and protein nitration. Taken together with our previous studies, the present findings show that blocking neutrophil influx protects alveolar development and improves lung function in part by preventing reactive oxygen/nitrogen species accumulation.


Asunto(s)
Radical Hidroxilo/antagonistas & inhibidores , Radical Hidroxilo/metabolismo , Hiperoxia/metabolismo , Pulmón/metabolismo , Oxígeno/toxicidad , Receptores de Interleucina-8B/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Dinoprost/análogos & derivados , Dinoprost/antagonistas & inhibidores , Neutrófilos/metabolismo , Ratas , Especies de Nitrógeno Reactivo/antagonistas & inhibidores , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Receptores de Interleucina-8B/fisiología , Tirosina/análogos & derivados , Tirosina/antagonistas & inhibidores , Tirosina/metabolismo
18.
Am J Respir Crit Care Med ; 174(5): 581-9, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16728710

RESUMEN

RATIONALE: Fibroblast growth factor receptor-1alpha(IIIc) [FGF-R1alpha(IIIc)] regulates recovery of neonatal rat lung growth, after 95% oxygen-mediated growth arrest. Its role in normal postnatal alveologenesis is unknown. OBJECTIVE: To determine if FGF-R1alpha(IIIc) regulates normal postnatal alveologenesis. METHODS: Truncated soluble FGF-R1alpha(IIIc) or neutralizing antibodies to FGF-1 or FGF-2 were injected intraperitoneally into 3-d-old rats. The pups were killed at Day 7 for studies of alveolar development. MEASUREMENTS AND MAIN RESULTS: Injected, truncated soluble FGF-R1alpha(IIIc) inhibited phosphorylation of the endogenous FGF-R1, and downstream pathway, and paradoxically increased lung DNA content and tissue fraction while inhibiting lung cell DNA synthesis. The increase in tissue thickness was due to reduced apoptosis, as indicated by reductions in cleaved effector caspases 3 and 7. Inhibition of the intrinsic apoptosis pathway was suggested by decreases in the proapoptotic protein Bax and mitochondrial cytochrome c release, and an increase in the antiapoptotic protein Bcl-x(L). Injected antibodies to FGF-1 and FGF-2 had no effect on DNA synthesis, but both increased Bcl-x(L) content and decreased cytochrome c release and cleaved caspase-7 protein expression. However, only injection of the antibody to FGF-2 replicated the increased tissue fraction and inhibited apoptosis observed with the injection of truncated soluble FGF-R1alpha(IIIc). CONCLUSIONS: Inhibition of ligand binding, most likely of FGF-2, to the FGF-R1alpha(IIIc) inhibits normal postnatal lung cell apoptosis.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/fisiología , Alveolos Pulmonares/crecimiento & desarrollo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis/metabolismo , Citocromos c/metabolismo , ADN/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Ratas , Receptor fas/metabolismo
19.
Pediatr Res ; 59(3): 389-95, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16492977

RESUMEN

Bronchopulmonary dysplasia (BPD), a major cause of morbidity in premature infants, is characterized by arrest of lung growth and inhibited alveologenesis. We had earlier cloned late-gestation lung 1 (LGL1), a glucocorticoid (GC)-induced, developmentally regulated gene in lung mesenchyme, and showed that reduced levels of late-gestation lung 1 protein (lgl1) inhibit lung branching. Maximal fetal expression of LGL1 is concordant with the onset of alveolar septation, suggesting an additional role for lgl1 in alveologenesis. At postnatal d 7, during the period of maximal septation in postnatal rat lung, lgl1 concentrates at the tips of budding secondary alveolar septa. We studied two models of impaired postnatal alveologenesis generated by exposure of newborn rats to 60% O2 for 2 wk or 95% O2 for 1 wk. A profound decrease of lgl1 expression with oxygen exposure was observed in both animal models. Animals exposed to 95% O2 for 1 wk recovered in air over a 3-wk period, associated with normalization of lgl1 levels. Changes in lung levels of alpha-actin (a marker of myofibroblast differentiation associated with alveologenesis) and the mesenchymal marker vimentin were significant but less marked. Our findings support a role for lgl1 in postnatal lung development. We speculate that deficiency of lgl1 contributes to the arrested alveolar partitioning observed in BPD and that recovery is associated with normalization of lgl1 levels.


Asunto(s)
Aire , Displasia Broncopulmonar/fisiopatología , Pulmón , Oxígeno/toxicidad , Proteínas/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Pulmón/anatomía & histología , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Pulmón/patología , Embarazo , Proteínas/genética , Ratas , Ratas Sprague-Dawley , Vimentina/genética , Vimentina/metabolismo
20.
Am J Respir Crit Care Med ; 172(7): 907-14, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15994466

RESUMEN

RATIONALE: Our core hypothesis is that growth factors that have dysregulated expression during experimental neonatal lung injury are likely to be involved in normal postnatal lung growth and alveologenesis. OBJECTIVES: To determine if hepatocyte growth factor (HGF) is upregulated in neonatal lung injury and is essential for postnatal alveologenesis. METHODS: A neonatal lung injury, in which there were patchy areas of interstitial thickening with a relative increase in the proportion of epithelial cells, was induced in newborn rats by exposing them to 60% oxygen for 14 days. Air-exposed pups had binding of endogenous HGF to its natural receptor, c-Met, inhibited by the intraperitoneal injection of either neutralizing antibody to HGF, or a truncated soluble c-Met receptor. MEASUREMENTS AND MAIN RESULTS: The 60% oxygen-mediated lung injury was associated with increased lung mRNAs for hepatocyte growth factor and c-Met, relative to air-exposed control lungs, at Day 7 after birth. After exposure to 60% oxygen, immunoreactive HGF was increased at Days 4 and 7, and immunoreactive c-Met was increased at Day 14. In air-exposed pups, intraperitoneal injections of neutralizing antibody to HGF inhibited DNA synthesis in alveoli-forming secondary crests, and reduced the number of alveoli in 6-day-old pups. Intraperitoneal injections of a truncated soluble c-Met receptor inhibited DNA synthesis in secondary crests in 4-day-old air-exposed rat pups. CONCLUSIONS: HGF and its c-Met receptor are required for normal postnatal alveolar formation from secondary crests, and are upregulated during 60% oxygen-induced neonatal lung injury.


Asunto(s)
Factor de Crecimiento de Hepatocito/fisiología , Alveolos Pulmonares/fisiología , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/fisiopatología , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Recién Nacido , Proteínas Proto-Oncogénicas c-met/metabolismo , Ratas , Regulación hacia Arriba/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA