Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pediatr Transplant ; 22(2)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29417689

RESUMEN

BOS is the pulmonary manifestation of cGvHD post-allogeneic HSCT. Survival and treatment of this often fatal complication have not improved over the last 20 years and there is no clear standard of care. For the past 10 years, BOS was treated in our center with monthly cycles of HDPS. We reviewed the outcomes of patients with post-HSCT BOS who met the diagnostic criteria for BOS as per the NIH consensus and were treated with at least one cycle of methylprednisolone at a dose of 10-30 mg/kg/d×3 d. We collected demographic and clinical data, responses to treatment and results of pulmonary function tests at several time points. Between January 2007 and January 2014, 12 patients were treated with HDPS for post-HSCT BOS. Five patients (42%) had a good response to treatment; four patients (33%) stabilized with moderate lung disease; and three patients (25%) progressed to end-stage disease. No significant acute side effects attributable to the HDPS treatment were identified. HDPS may be an effective treatment option for all but the most severely ill patients with post-HSCT BOS.


Asunto(s)
Antiinflamatorios/administración & dosificación , Bronquiolitis Obliterante/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Metilprednisolona/administración & dosificación , Adolescente , Antiinflamatorios/uso terapéutico , Bronquiolitis Obliterante/etiología , Niño , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Estudios de Seguimiento , Humanos , Inyecciones Intravenosas , Masculino , Metilprednisolona/uso terapéutico , Estudios Retrospectivos , Resultado del Tratamiento
2.
Am J Physiol Lung Cell Mol Physiol ; 308(4): L391-402, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25480331

RESUMEN

Rapid growth and formation of new gas exchange units (alveogenesis) are hallmarks of the perinatal lung. Bronchopulmonary dysplasia (BPD), common in very premature infants, is characterized by premature arrest of alveogenesis. Mesenchymal cells (fibroblasts) regulate both lung branching and alveogenesis through mesenchymal-epithelial interactions. Temporal or spatial deficiency of late-gestation lung 1/cysteine-rich secretory protein LD2 (LGL1/CRISPLD2), expressed in and secreted by lung fibroblasts, can impair both lung branching and alveogenesis (LGL1 denotes late gestation lung 1 protein; LGL1 denotes the human gene; Lgl1 denotes the mouse/rat gene). Absence of Lgl1 is embryonic lethal. Lgl1 levels are dramatically reduced in oxygen toxicity rat models of BPD, and heterozygous Lgl1(+/-) mice exhibit features resembling human BPD. To explore the role of LGL1 in mesenchymal-epithelial interactions in developing lung, we developed a doxycycline (DOX)-inducible RNA-mediated LGL1 knockdown cellular model in human fetal lung fibroblasts (MRC5(LGL1KD)). We assessed the impact of LGL1 on cell proliferation, cell migration, apoptosis, and wound healing. DOX-induced MRC5(LGL1KD) suppressed cell growth and increased apoptosis of annexin V(+) staining cells and caspase 3/7 activity. LGL1-conditioned medium increased migration of fetal rat primary lung epithelial cells and human airway epithelial cells. Impaired healing by MRC5(LGL1KD) cells of a wound model was attenuated by addition of LGL1-conditioned medium. Suppression of LGL1 was associated with dysregulation of extracellular matrix genes (downregulated MMP1, ColXVα1, and ELASTIN) and proapoptosis genes (upregulated BAD, BAK, CASP2, and TNFRSF1B) and inhibition of 44/42MAPK phosphorylation. Our findings define a role for LGL1 in fibroblast expansion and migration, epithelial cell migration, and mesenchymal-epithelial signaling, key processes in fetal lung development.


Asunto(s)
Apoptosis/fisiología , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Feto/embriología , Fibroblastos/metabolismo , Factores Reguladores del Interferón/metabolismo , Pulmón/embriología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Displasia Broncopulmonar/embriología , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patología , Moléculas de Adhesión Celular/genética , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/genética , Feto/citología , Fibroblastos/citología , Células HEK293 , Humanos , Factores Reguladores del Interferón/genética , Pulmón/citología , Ratones , Ratones Noqueados , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley , Mucosa Respiratoria/citología , Mucosa Respiratoria/embriología , Transducción de Señal/fisiología
3.
Respir Res ; 14: 138, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24344776

RESUMEN

BACKGROUND: Cystic fibrosis (CF) is a complex, multi-system, life-shortening, autosomal recessive disease most common among Caucasians. Pulmonary pathology, the major cause of morbidity and mortality in CF, is characterized by dysregulation of cytokines and a vicious cycle of infection and inflammation. This cycle causes a progressive decline in lung function, eventually resulting in respiratory failure and death. The Th17 immune response plays an active role in the pathogenesis of CF pulmonary pathology, but it is not known whether the pathophysiology of CF disease contributes to a heightened Th17 response or whether CF naïve CD4+ T lymphocytes (Th0 cells) intrinsically have a heightened predisposition to Th17 differentiation. METHODS: To address this question, Th0 cells were isolated from the peripheral blood of CF mice, human CF subjects and corresponding controls. Murine Th0 cells were isolated from single spleen cell suspensions using fluorescence-activated cell sorting. Lymphocytes from human buffy coats were isolated by gradient centrifugation and Th0 cells were further isolated using a human naïve T cell isolation kit. Th0 cells were then assessed for their capacity to differentiate along Th17, Th1 or Treg lineages in response to corresponding cytokine stimulation. The T cell responses of human peripheral blood cells were also assessed ex vivo using flow cytometry. RESULTS: Here we identify in both mouse and human CF an intrinsically enhanced predisposition of Th0 cells to differentiate towards a Th17 phenotype, while having a normal propensity for differentiation into Th1 and Treg lineages. Furthermore, we identify an active Th17 response in the peripheral blood of human CF subjects. CONCLUSIONS: We propose that these novel observations offer an explanation, at least in part, for the known increased Th17-associated inflammation of CF and the early signs of inflammation in CF lungs before any evidence of infection. Moreover, these findings point towards direct modulation of T cell responses as a novel potential therapeutic strategy for combating excessive inflammation in CF.


Asunto(s)
Linfocitos T CD4-Positivos/patología , Diferenciación Celular , Fibrosis Quística/patología , Fenotipo , Células Th17/patología , Adolescente , Adulto , Animales , Linfocitos T CD4-Positivos/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Células TH1/metabolismo , Células TH1/patología , Células Th17/metabolismo , Adulto Joven
4.
Am J Respir Cell Mol Biol ; 43(5): 599-606, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20042713

RESUMEN

Glucocorticoid (GC)-responsive epithelial-mesenchymal interactions regulate lung development. The GC receptor (GR) mediates GC signaling. Mice lacking GR in all tissues die at birth of respiratory failure. To determine the specific need for epithelial GR in lung development, we bred triple transgenic mice that carry SPC/rtTA, tet-O-Cre, and floxed, but not wild-type, GR genes. When exposed to doxycycline in utero, triple transgenic (GRepi⁻) mice exhibit a Cre-mediated recombination event that inactivates the floxed GR gene in airway epithelial cells. Immunofluorescence confirmed the elimination of GR in Cre-positive airway epithelial cells of late gestation GRepi⁻ mice. Embryonic Day 18.5 pups had a relatively immature appearance with increased lung cellularity and increased pools of glycogen in the epithelium. Postnatal Day 0.5 pups had decreased viability. We used quantitative RT-PCR to demonstrate that specific elimination of epithelial immunoreactive GR in GRepi⁻ mice is associated with reduced mRNA expression for surfactant proteins (SPs) A, B, C, and D; ß- and γ-ENaC; T1α; the 10-kD Clara cell protein (CCSP); and aquaporin 5 (AQP5). Western blots confirmed reduced levels of AQP5 protein. No reduction in the levels of the GR transport protein importin (IPO)-13 was observed. Our findings demonstrate a requirement for lung epithelial cell GR in normal lung development. We speculate that impaired epithelial differentiation, leading to decreased SPs, transepithelial Na, and liquid absorption at birth, may contribute to the reduced survival of newborn mice with suppressed lung epithelial GR.


Asunto(s)
Epitelio/metabolismo , Epitelio/patología , Pulmón/metabolismo , Pulmón/patología , Receptores de Glucocorticoides/deficiencia , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Doxiciclina/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Epitelio/efectos de los fármacos , Epitelio/embriología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Pulmón/embriología , Ratones , Ratones Noqueados , Especificidad de Órganos/efectos de los fármacos , Organogénesis/efectos de los fármacos , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Proteínas Asociadas a Surfactante Pulmonar/genética , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Análisis de Supervivencia , Factores de Transcripción/metabolismo
5.
Respir Res ; 11: 166, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21118573

RESUMEN

BACKGROUND: Among patients with cystic fibrosis (CF), females have worse pulmonary function and survival than males, primarily due to chronic lung inflammation and infection with Pseudomonas aeruginosa (P. aeruginosa). A role for gender hormones in the causation of the CF "gender gap" has been proposed. The female gender hormone 17ß-estradiol (E2) plays a complex immunomodulatory role in humans and in animal models of disease, suppressing inflammation in some situations while enhancing it in others. Helper T-cells were long thought to belong exclusively to either T helper type 1 (Th1) or type 2 (Th2) lineages. However, a distinct lineage named Th17 is now recognized that is induced by interleukin (IL)-23 to produce IL-17 and other pro-inflammatory Th17 effector molecules. Recent evidence suggests a central role for the IL-23/IL-17 pathway in the pathogenesis of CF lung inflammation. We used a mouse model to test the hypothesis that E2 aggravates the CF lung inflammation that occurs in response to airway infection with P. aeruginosa by a Th17-mediated mechanism. RESULTS: Exogenous E2 caused adult male CF mice with pneumonia due to a mucoid CF clinical isolate, the P. aeruginosa strain PA508 (PA508), to develop more severe manifestations of inflammation in both lung tissue and in bronchial alveolar lavage (BAL) fluid, with increased total white blood cell counts and differential and absolute cell counts of polymorphonuclear leukocytes (neutrophils). Inflammatory infiltrates and mucin production were increased on histology. Increased lung tissue mRNA levels for IL-23 and IL-17 were accompanied by elevated protein levels of Th17-associated pro-inflammatory mediators in BAL fluid. The burden of PA508 bacteria was increased in lung tissue homogenate and in BAL fluid, and there was a virtual elimination in lung tissue of mRNA for lactoferrin, an antimicrobial peptide active against P. aeruginosa in vitro. CONCLUSIONS: Our data show that E2 increases the severity of PA508 pneumonia in adult CF male mice, and suggest two potential mechanisms: enhancement of Th17-regulated inflammation and suppression of innate antibacterial defences. Although this animal model does not recapitulate all aspects of human CF lung disease, our present findings argue for further investigation of the effects of E2 on inflammation and infection with P. aeruginosa in the CF lung.


Asunto(s)
Fibrosis Quística/complicaciones , Estrógenos/efectos adversos , Neumonía Bacteriana/inducido químicamente , Neumonía Bacteriana/patología , Infecciones por Pseudomonas/inducido químicamente , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa , Animales , Fibrosis Quística/patología , Modelos Animales de Enfermedad , Masculino , Ratones
6.
Pediatr Res ; 67(4): 375-81, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20057335

RESUMEN

Alveolarization depends on circulating glucocorticoid (GC), retinoid (RA), and vitamin D (VitD). Bronchopulmonary dysplasia, a leading cause of neonatal morbidity, is associated with arrested alveolarization. In hyperoxia-exposed rats displaying features of bronchopulmonary dysplasia, reduced levels of late gestation lung 1 (Lgl1) normalize during recovery. We show that GC (100 nM) stimulates (7- to 115-fold) and VitD (100 microM) suppresses (twofold) Lgl1 expression. RA (all-trans/9-cis, 10 microM) effects are biphasic. From postnatal days 7-10, RA was stimulatory (twofold) at 24 h, after which effects were inhibitory (3- to 15-fold). Lgl1 promoter-luciferase reporter assays confirmed that these agents operated at the transcriptional level. Interestingly, the individual inhibitory effects of VitD and RA on GC induction of Lgl1 were abrogated when both agents were present, suggesting that steric hindrance may influence promoter accessibility. Analysis of the proximity (<50 base pairs) of binding sites for overlapping VitD and RA receptors to that of the GC receptor identified 81% of promoters in 66 genes (including Lgl1) important in human lung development compared with 48% in a random set of 1000 genes. Complex integration of the effects of GC, RA, and VitD on gene expression in the postnatal lung is likely to contribute to the timely advance of alveolarization without attendant inflammation.


Asunto(s)
Antineoplásicos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas/metabolismo , Alveolos Pulmonares/fisiología , Esteroides/farmacología , Tretinoina/farmacología , Vitamina D/farmacología , Animales , Sitios de Unión , Línea Celular , Femenino , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Embarazo , Regiones Promotoras Genéticas , Proteínas/genética , Alveolos Pulmonares/citología , Ratas , Ratas Sprague-Dawley , Transcripción Genética
7.
Respir Res ; 10: 83, 2009 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-19772569

RESUMEN

BACKGROUND: Neonatal lung injury, a leading cause of morbidity in prematurely born infants, has been associated with arrested alveolar development and is often accompanied by goblet cell hyperplasia. Genes that regulate alveolarization and inflammation are likely to contribute to susceptibility to neonatal lung injury. We previously cloned Lgl1, a developmentally regulated secreted glycoprotein in the lung. In rat, O2 toxicity caused reduced levels of Lgl1, which normalized during recovery. We report here on the generation of an Lgl1 knockout mouse in order to determine whether deficiency of Lgl1 is associated with arrested alveolarization and contributes to neonatal lung injury. METHODS: An Lgl1 knockout mouse was generated by introduction of a neomycin cassette in exon 2 of the Lgl1 gene. To evaluate the pulmonary phenotype of Lgl1+/- mice, we assessed lung morphology, Lgl1 RNA and protein, elastin fibers and lung function. We also analyzed tracheal goblet cells, and expression of mucin, interleukin (IL)-4 and IL-13 as markers of inflammation. RESULTS: Absence of Lgl1 was lethal prior to lung formation. Postnatal Lgl1+/- lungs displayed delayed histological maturation, goblet cell hyperplasia, fragmented elastin fibers, and elevated expression of TH2 cytokines (IL-4 and IL-13). At one month of age, reduced expression of Lgl1 was associated with elevated tropoelastin expression and altered pulmonary mechanics. CONCLUSION: Our findings confirm that Lgl1 is essential for viability and is required for developmental processes that precede lung formation. Lgl1+/- mice display a complex phenotype characterized by delayed histological maturation, features of inflammation in the post-natal period and altered lung mechanics at maturity. Lgl1 haploinsufficiency may contribute to lung disease in prematurity and to increased risk for late-onset respiratory disease.


Asunto(s)
Glicoproteínas/metabolismo , Células Caliciformes/metabolismo , Factores Inmunológicos/metabolismo , Pulmón/metabolismo , Ratones Noqueados/metabolismo , Mecánica Respiratoria , Animales , Células Cultivadas , Citocinas , Glicoproteínas/genética , Lesión Pulmonar , Ratones
8.
Respir Res ; 10: 77, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19698107

RESUMEN

BACKGROUND: A precise balance exists between the actions of endogenous glucocorticoids (GC) and retinoids to promote normal lung development, in particular during alveolarization. The mechanisms controlling this balance are largely unknown, but recent evidence suggests that midkine (MK), a retinoic acid-regulated, pro-angiogenic growth factor, may function as a critical regulator. The purpose of this study was to examine regulation of MK by GC and RA during postnatal alveolar formation in rats. METHODS: Newborn rats were treated with dexamethasone (DEX) and/or all-trans-retinoic acid (RA) during the first two weeks of life. Lung morphology was assessed by light microscopy and radial alveolar counts. MK mRNA and protein expression in response to different treatment were determined by Northern and Western blots. In addition, MK protein expression in cultured human alveolar type 2-like cells treated with DEX and RA was also determined. RESULTS: Lung histology confirmed that DEX treatment inhibited and RA treatment stimulated alveolar formation, whereas concurrent administration of RA with DEX prevented the DEX effects. During normal development, MK expression was maximal during the period of alveolarization from postnatal day 5 (PN5) to PN15. DEX treatment of rat pups decreased, and RA treatment increased lung MK expression, whereas concurrent DEX+RA treatment prevented the DEX-induced decrease in MK expression. Using human alveolar type 2 (AT2)-like cells differentiated in culture, we confirmed that DEX and cAMP decreased, and RA increased MK expression. CONCLUSION: We conclude that MK is expressed by AT2 cells, and is differentially regulated by corticosteroid and retinoid treatment in a manner consistent with hormonal effects on alveolarization during postnatal lung development.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Citocinas/metabolismo , Dexametasona/farmacología , Células Epiteliales/efectos de los fármacos , Alveolos Pulmonares/efectos de los fármacos , Tretinoina/farmacología , Factores de Edad , Proteínas Angiogénicas/genética , Animales , Animales Recién Nacidos , Northern Blotting , Western Blotting , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Citocinas/genética , Células Epiteliales/metabolismo , Humanos , Midkina , Alveolos Pulmonares/crecimiento & desarrollo , Alveolos Pulmonares/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
9.
Pediatr Pulmonol ; 42(6): 519-24, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17469152

RESUMEN

RATIONALE: There is no adequate explanation for gender-based differences in rates of mortality and of deterioration in pulmonary function in cystic fibrosis (CF) patients. One potential explanation is that gender hormones (sex steroids) may modulate the severity of CF lung disease, the principal cause of mortality in CF, by altering respiratory transepithelial ion transport. OBJECTIVE: To determine whether respiratory epithelial ion transport varied during the menstrual cycle of CF females. METHODS: The nasal transepithelial electrical potential difference (NPD) was determined as a measure of ion transport across human respiratory epithelium, coincident with measurements of endogenous serum hormone levels in the luteal and follicular phases of the menstrual cycle in CF females aged 16-22 years. RESULTS: The component of the NPD that is insensitive to the Na(+) transport blocker amiloride, but not the amiloride-sensitive component, changed in association with endogenous, menstrual cycle-induced changes in serum levels of progesterone and estrogen (P=0.02, n=7, paired t-test). Measurements using Cl(-) free perfusates suggested that the changes are not a result of Cl(-) conductance. CONCLUSIONS: Our results suggest that in CF respiratory epithelium amiloride-insensitive, but not amiloride-sensitive, ion transport is altered by female gender hormones in vivo. We speculate that amiloride-insensitive ion transport may contribute to the regulation of human airway surface fluid.


Asunto(s)
Amilorida/farmacología , Fibrosis Quística/fisiopatología , Potenciales de la Membrana/efectos de los fármacos , Ciclo Menstrual/fisiología , Mucosa Nasal/fisiología , Bloqueadores de los Canales de Sodio/farmacología , Adolescente , Adulto , Estudios de Casos y Controles , Fibrosis Quística/sangre , Estrógenos/sangre , Femenino , Fase Folicular/sangre , Humanos , Transporte Iónico/efectos de los fármacos , Transporte Iónico/fisiología , Fase Luteínica/sangre , Potenciales de la Membrana/fisiología , Ciclo Menstrual/sangre , Mucosa Nasal/citología , Mucosa Nasal/efectos de los fármacos , Progesterona/sangre , Factores Sexuales
10.
Physiol Rep ; 4(17)2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27597766

RESUMEN

Chronic lung disease of prematurity/bronchopulmonary dysplasia (BPD) is the leading cause of perinatal morbidity in developed countries. Inflammation is a prominent finding. Currently available interventions have associated toxicities and limited efficacy. While BPD often resolves in childhood, survivors of preterm birth are at risk for acquired respiratory disease in early life and are more likely to develop chronic obstructive pulmonary disease (COPD) in adulthood. We previously cloned Crispld2 (Lgl1), a glucocorticoid-regulated mesenchymal secretory protein that modulates lung branching and alveogenesis through mesenchymal-epithelial interactions. Absence of Crispld2 is embryonic lethal. Heterozygous Crispld2+/- mice display features of BPD, including distal airspace enlargement, disruption of elastin, and neonatal lung inflammation. CRISPLD2 also plays a role in human fetal lung fibroblast cell expansion, migration, and mesenchymal-epithelial signaling. This study assessed the effects of endogenous and exogenous CRISPLD2 on expression of proinflammatory mediators in human fetal and adult (normal and COPD) lung fibroblasts and epithelial cells. CRISPLD2 expression was upregulated in a lipopolysaccharide (LPS)-induced human fetal lung fibroblast line (MRC5). LPS-induced upregulation of the proinflammatory cytokines IL-8 and CCL2 was exacerbated in MRC5-CRISPLD2(knockdown) cells. siRNA suppression of endogenous CRISPLD2 in adult lung fibroblasts (HLFs) led to augmented expression of IL-8, IL-6, CCL2. LPS-stimulated expression of proinflammatory mediators by human lung epithelial HAEo- cells was attenuated by purified secretory CRISPLD2. RNA sequencing results from HLF-CRISPLD2(knockdown) suggest roles for CRISPLD2 in extracellular matrix and in inflammation. Our data suggest that suppression of CRISPLD2 increases the risk of lung inflammation in early life and adulthood.


Asunto(s)
Displasia Broncopulmonar/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Mediadores de Inflamación/antagonistas & inhibidores , Factores Reguladores del Interferón/metabolismo , Pulmón/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Adulto , Anciano , Animales , Displasia Broncopulmonar/embriología , Displasia Broncopulmonar/patología , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Proliferación Celular/fisiología , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/fisiología , Fibroblastos/citología , Fibroblastos/patología , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Factores Reguladores del Interferón/deficiencia , Factores Reguladores del Interferón/genética , Interleucina-6/metabolismo , Lipopolisacáridos/metabolismo , Pulmón/citología , Pulmón/embriología , Pulmón/patología , Masculino , Ratones , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Transducción de Señal
11.
Pediatr Pulmonol ; 39(2): 185-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15532092

RESUMEN

Recurrent respiratory papillomatosis is the most common neoplasm of the larynx in childhood. Extension into lung parenchyma occurs in less than 1% of patients and has a low risk of malignant transformation. Treatment options for intrapulmonary spread have shown limited success. We describe a case of recurrent respiratory papillomatosis with extensive parenchymal involvement and adenosquamous carcinoma in a 14-year-old girl.


Asunto(s)
Carcinoma Adenoescamoso/patología , Transformación Celular Neoplásica , Neoplasias Laríngeas/patología , Neoplasias Pulmonares/patología , Recurrencia Local de Neoplasia/patología , Papiloma/patología , Adolescente , Biopsia con Aguja , Carcinoma Adenoescamoso/diagnóstico por imagen , Carcinoma Adenoescamoso/terapia , Transformación Celular Neoplásica/patología , Terapia Combinada , Diagnóstico Diferencial , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Laríngeas/diagnóstico por imagen , Neoplasias Laríngeas/terapia , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/terapia , Papiloma/diagnóstico por imagen , Papiloma/terapia , Radiografía Torácica , Tomografía Computarizada por Rayos X
12.
Biochem J ; 376(Pt 1): 61-9, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12880386

RESUMEN

Secreted glycoproteins serve a variety of functions related to cell-cell communication in developmental systems. We cloned LGL1, a novel glucocorticoid-inducible gene in foetal lung, and described its temporal and spatial localization in the rat. Disruption of foetal mesenchyme-specific LGL1 expression using antisense oligodeoxynucleotides, which was associated with a 50% decrease in lgl1 protein levels, inhibited airway epithelial branching in foetal rat gestational day 13 lung buds in explant culture. These findings suggested that lgl1 functions as a secreted signalling molecule. We now provide evidence supporting a role for lgl1 in mesenchymal-epithelial interactions that govern lung organogenesis. Lgl1 is a secreted glycoprotein with a conserved N-terminal secretory signal peptide. Using dual immunofluorescence, intracellular lgl1 was found to co-localize with markers of the Golgi apparatus and endoplasmic reticulum, consistent with its association with secretory vesicles. Using pulse-chase studies, we show that lgl1 is a stable protein with a half-life of 11.5 h. Furthermore, at gestational days 20 and 21 (term=22), foetal distal lung epithelial cells import lgl1 protein. Taken together, our findings support distinct roles for lgl1 as a mediator of glucocorticoid-induced mesenchymal-epithelial interactions in early and late foetal lung organogenesis.


Asunto(s)
Glicoproteínas/metabolismo , Pulmón/embriología , Proteínas/metabolismo , Mucosa Respiratoria/embriología , Animales , Línea Celular , Células Cultivadas , Retículo Endoplásmico/química , Células Epiteliales/metabolismo , Edad Gestacional , Glicosilación , Aparato de Golgi/química , Humanos , Pulmón/citología , Pulmón/metabolismo , Mesodermo/fisiología , Morfogénesis , Señales de Clasificación de Proteína , Transporte de Proteínas , Proteínas/fisiología , Ratas , Ratas Wistar , Mucosa Respiratoria/metabolismo
13.
Pediatr Pulmonol ; 49(4): 309-17, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24339235

RESUMEN

Cystic fibrosis (CF) is a complex, multi-system, autosomal recessive disease predominantly affecting Caucasians that leads to vigorous airway inflammation and chronic respiratory infection, commonly with Pseudomonas aeruginosa. A variety of factors significantly modify the progression and severity of CF lung disease and the timing of the resulting mortality. We summarize here data indicating that there is in CF a female disadvantage in survival and morbidity, called the "CF gender gap". Although controversy exists regarding the nature and relative importance of the various contributing mechanisms involved, gender affects the progression of CF disease with respect to lung infection, decline in pulmonary function and nutritional status. These interrelated factors in turn have a negative impact on survival. This review will emphasize the increasing evidence that suggest a role for the effects of gender, and particularly the female sex hormone estrogen, on infection, inflammation and transepithelial ion transport, all major determinants of CF lung disease. Future elucidation of the pathophysiology of hormonal aggravation of CF lung disease may pave the way for novel therapeutic interventions. This, combined with the magnitude of the gender gap in CF mortality, strongly suggests that further work in this field is well justified.


Asunto(s)
Fibrosis Quística , Estrógenos/fisiología , Fibrosis Quística/complicaciones , Fibrosis Quística/etiología , Fibrosis Quística/mortalidad , Progresión de la Enfermedad , Femenino , Humanos , Inflamación/etiología , Masculino , Factores Sexuales , Tasa de Supervivencia
14.
Allergy Asthma Clin Immunol ; 10(1): 2, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24438707

RESUMEN

A girl was diagnosed with cystic fibrosis (CF) at birth, with repeatedly positive sweat tests and homozygous F508del mutations of her CF transmembrane conductance regulator (CFTR) gene. From an early age, her lung disease was more severe than her birth cohort peers despite aggressive treatment. At the age of 16 she was listed for lung transplantation, but prior to transplant was not on systemic corticosteroids or other immunosuppressive agents. In response to ex vivo stimulation, her pre-transplant peripheral blood T cells unexpectedly failed to produce detectable levels of IFN-γ, unlike cells from healthy controls or from another girl with CF and lung disease of comparable severity. Furthermore, naïve T cells freshly isolated from her peripheral blood showed a complete block of T cell differentiation into Th1, Th17 and Treg lineages, even in the presence of cytokines known to promote differentiation into the respective lineages. Her serology has been remarkably devoid of evidence of exposure to viruses that have been associated with T cell exhaustion. However, her freshly isolated naïve T cells showed sustained expression of markers of T cell exhaustion, which were further induced upon ex vivo stimulation, pointing to T cell exhaustion as the cause of the failure of naïve T cells to undergo differentiation in response to cytokine stimulation. Although excessive inflammation in CF lung can be both ineffective at clearing certain pathogens as well as destructive to the lung tissue itself, adequate inflammation is a component of an effective overall immune response to microbial pathogens. Our present findings suggest that intrinsic impairment of T cell differentiation may have contributed to the greater severity and more rapid progression of her CF lung disease than of the lung disease of most of her peers.

16.
Pediatr Pulmonol ; 43(2): 125-33, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18085690

RESUMEN

In order to better understand the regulation of lung maturation by glucocorticoid-glucocorticoid receptor signaling, we studied glucocorticoid receptor (GR) hypomorphic mice with a mixed C57Bl6/129 sv background, in which disruption of exon 2 of the GR gene produces an N-terminal truncated GR protein. Four groups of mice were compared: homozygous mice that die at birth (non-survivors), homozygous mice that survive the neonatal period (survivors), heterozygotes and wild-type mice. Newborn non-survivors had 50% thicker airspace walls and a 46% decrease in the formation of secondary crests (the beginning of alveolar secondary septation) compared to either survivor or wild-type littermates (n = 9 mice in each group). The lung tissue to airspace ratio in homozygous mice not expressing wild-type GR (non-survivor and survivor) was increased compared to heterozygotes and wild-type mice that do express wild-type GR (0.91 +/- 0.08 vs. 0.49 +/- 0.02, n = 4 in each of the four subgroups), suggesting that complete morphological maturation of the lung is dependent on effective glucocorticoid signaling through a fully functional GR. Moreover, the relatively mature lung morphology of survivor versus non-survivor newborns suggests that a partial reduction in mesenchymal thickness is compatible with capillary remodeling, alveolar septation, and viable respiratory function after birth. Our findings suggest that in mice homozygous for disrupted GR, the severity of newborn respiratory insufficiency correlates with the degree of lung structural immaturity.


Asunto(s)
Glucocorticoides/metabolismo , Pulmón/patología , Receptores de Glucocorticoides/genética , Insuficiencia Respiratoria/genética , Insuficiencia Respiratoria/patología , Animales , Animales Recién Nacidos , Capilares/patología , Células Endoteliales/patología , Glucocorticoides/genética , Heterocigoto , Homocigoto , Inmunohistoquímica , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL/genética , Músculo Liso/patología , Insuficiencia Respiratoria/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal
17.
Biol Neonate ; 90(1): 46-57, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16534186

RESUMEN

BACKGROUND: Congenital truncation of the glucocorticoid receptor (GR) is known to lead to lethal lung immaturity in newborn mice associated with increased lung cellularity (ratio of tissue to airspace) and, as we previously showed, prolonged expression of the retinoid-responsive growth factor midkine. OBJECTIVES: We sought to determine if these changes would be reversed by transgenic expression of GR exclusively in the distal airway epithelium. METHODS: Mice were generated with expression of transgenic rat (r) GR driven by the human (h) SP-C promoter, on a background of congenital GR truncation. RESULTS: Transgenic epithelial GR expression reduced lung cellularity and midkine expression to levels comparable to wild-type littermates. Nevertheless, the newborn transgenic mice still displayed respiratory failure. Moreover, epithelial expression of the GR transgene did not alter expression of a number of important markers of lung maturation. CONCLUSIONS: Our data demonstrating normalization of the lung tissue to airspace ratio in neonatal mice expressing transgenic GR in the distal airway epithelium is consistent with the concept that normal mesenchymal cell loss is due to GR-responsive stimulation from epithelial cells. However, we could find no evidence of altered apoptotic activity between the groups of mice. We speculate that correction of the severe neonatal lung phenotype of GR-deficient mice will require expression of normal GR in non-epithelial as well as epithelial tissues.


Asunto(s)
Citocinas/genética , Pulmón/fisiología , Péptidos/genética , Regiones Promotoras Genéticas , Receptores de Glucocorticoides/genética , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Secuencia de Bases , Cartilla de ADN , Regulación de la Expresión Génica , Genotipo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Pulmón/citología , Ratones , Ratones Noqueados , Ratones Transgénicos , Midkina , Proteína C Asociada a Surfactante Pulmonar , Ratas , Receptores de Glucocorticoides/deficiencia
18.
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
19.
Am J Respir Cell Mol Biol ; 28(2): 232-40, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12540491

RESUMEN

We previously described the cloning of the late gestation lung 1 gene (LGL1), a novel glucocorticoid-inducible gene expressed in the mesenchyme of fetal lung. We report here evidence for a role of the LGL1 gene product (lgl1) in fetal rat lung airway branching morphogenesis, temporal and spatial localization of LGL1 mRNA and lgl1 protein in fetal rat lung, and a correction of the previously published LGL1 sequence. Both the mRNA and protein were detected during fetal lung development. LGL1 mRNA was detected from gestational Day 12 by reverse transcriptase-polymerase chain reaction, and from Day 13 by in situ hybridization. lgl1 protein was detected from Day 18 by Western analysis and from Day 16 by immunohistochemistry. The types of cells expressing LGL1 mRNA and lgl1 protein were assessed by immunohistochemical staining of adjacent serial tissue sections for markers of mesenchymal (vimentin) and smooth muscle (alpha-actin) cells. As gestation advanced, increasing amounts of mRNA and protein were expressed in these cells. In support of a role for lgl1 in airway branching morphogenesis, antisense (but neither sense nor scrambled) oligodeoxynucleotides directed against LGL1 inhibited airway branching in fetal rat lung buds in explant culture, in a dose- and time-dependent manner. The levels of lgl1 protein and LGL1 mRNA expression were decreased in those explants that had inhibited airway branching, compared with the uninhibited controls. Our findings suggest that lgl1 plays an important role in fetal airway branching morphogenesis.


Asunto(s)
Pulmón/embriología , Pulmón/metabolismo , Oligodesoxirribonucleótidos Antisentido/genética , Oligodesoxirribonucleótidos Antisentido/farmacología , Proteínas/antagonistas & inhibidores , Proteínas/genética , Animales , Secuencia de Bases , Técnicas de Cultivo , Inmunohistoquímica , Hibridación in Situ , Pulmón/efectos de los fármacos , Morfogénesis/efectos de los fármacos , Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
20.
Am J Respir Cell Mol Biol ; 31(4): 377-81, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15381503

RESUMEN

Cystic fibrosis (CF) lung disease is characterized by chronic neutrophilic inflammation and infection. Effective management of airway inflammation could complement other therapies for the treatment of CF. Recent progress has been made in understanding the signaling pathways regulating inflammatory cytokines in the lung. Here we examine the mechanisms responsible for inflammation in the CF lung, and discuss potential therapeutic strategies targeting inflammation.


Asunto(s)
Antibacterianos/uso terapéutico , Antiinflamatorios no Esteroideos/uso terapéutico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Fibrosis Quística/fisiopatología , Fibrosis Quística/terapia , Mediadores de Inflamación/fisiología , Pulmón/fisiopatología , Animales , Fibrosis Quística/complicaciones , Humanos , Pulmón/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA