Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 315
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Genet ; 2(1): 13-20, 1992 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-1284640

RESUMEN

Human cystic fibrosis transmembrane conductance regulator (CFTR) was expressed in transgenic mice under the control of transcriptional elements derived from the human surfactant protein C (SP-C) gene. The hCFTR mRNA was expressed in lungs and testes: in the lung, we found hCFTR mRNA in bronchiolar and alveolar epithelial cells, and CFTR protein in respiratory epithelial cells. While the level of expression of hCFTR mRNA varied, hCFTR mRNA and protein were detected in pulmonary epithelial cells of several lines. Lung weight, morphology, somatic growth and reproductive capacity were not altered by expression hCFTR in lung and testes of the transgenics. Our findings suggest that hCFTR can be safely transferred to lung epithelial cells for CF therapy.


Asunto(s)
Pulmón/metabolismo , Proteínas de la Membrana/genética , Animales , Fibrosis Quística/genética , Fibrosis Quística/terapia , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Epitelio/metabolismo , Expresión Génica , Terapia Genética , Humanos , Inmunohistoquímica , Hibridación in Situ , Masculino , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Transgénicos , Proteolípidos/genética , Surfactantes Pulmonares/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Distribución Tisular
2.
Int J Pharm ; 585: 119387, 2020 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-32473376

RESUMEN

Preterm infants, particularly those who born between 23 and 28 weeks' gestation, suffer from a very high incidence of respiratory distress syndrome (RDS) related to pulmonary immaturity and inability to make Pulmonary Surfactant (PS). These infants are supported by the use of oxygen, ventilators, and routine administration of surfactant replacement. The currently commercial surfactant replacement therapies do not contain hydrophilic surfactant proteins such as Surfactant Protein D (SP-D). These proteins have a key role in the innate lung host defense, thus the development of a sustained release vehicle that provides SP-D for long periods in preterm infants' lungs would exploit the therapeutic potential of SP-D and other pulmonary medications. The proposed SP-D delivery system is based on nanoparticles (NPs) composed of poly (lactic acid-co-glycolic acid) (PLGA), a biodegradable, FDA approved biopolymer. The resulted NPs were spherical with high Zeta potential value, were not toxic to A-549 lungs cells, and did not induce any inflammatory response in mouse's lungs for short and long-term periods. Moreover, SP-D released from NPs showed biological activity for several days and in vivo release experiment of SP-D loaded NPs revealed that SP-D was released from NPs in mouse lungs with different NPs delivery doses.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Proteína D Asociada a Surfactante Pulmonar/administración & dosificación , Síndrome de Dificultad Respiratoria del Recién Nacido/tratamiento farmacológico , Animales , Línea Celular , Química Farmacéutica/métodos , Portadores de Fármacos/uso terapéutico , Estabilidad de Medicamentos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Recién Nacido , Recien Nacido Prematuro , Ratones , Ratones Endogámicos C57BL , Tamaño de la Partícula , Proteína D Asociada a Surfactante Pulmonar/uso terapéutico
3.
Science ; 266(5191): 1705-8, 1994 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-7527588

RESUMEN

Cystic fibrosis (CF) is caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR). A potential animal model of CF, the CFTR-/- mouse, has had limited utility because most mice die from intestinal obstruction during the first month of life. Human CFTR (hCFTR) was expressed in CFTR-/- mice under the control of the rat intestinal fatty acid-binding protein gene promoter. The mice survived and showed functional correction of ileal goblet cell and crypt cell hyperplasia and cyclic adenosine monophosphate-stimulated chloride secretion. These results support the concept that transfer of the hCFTR gene may be a useful strategy for correcting physiologic defects in patients with CF.


Asunto(s)
Fibrosis Quística/terapia , Terapia Genética , Mucosa Intestinal/patología , Proteínas de la Membrana/genética , Proteínas de Neoplasias , Proteínas del Tejido Nervioso , Proteínas Supresoras de Tumor , Animales , Secuencia de Bases , Proteínas Portadoras/genética , Cloruros/metabolismo , Colforsina/farmacología , Colon/química , Colon/patología , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Modelos Animales de Enfermedad , Proteína de Unión a los Ácidos Grasos 7 , Proteínas de Unión a Ácidos Grasos , Expresión Génica , Humanos , Mucosa Intestinal/química , Mucosa Intestinal/metabolismo , Intestino Delgado/química , Intestino Delgado/patología , Proteínas de la Membrana/análisis , Proteínas de la Membrana/fisiología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , ARN Mensajero/análisis , ARN Mensajero/genética , Ratas , Proteínas Recombinantes/biosíntesis
4.
Science ; 264(5159): 713-6, 1994 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-8171324

RESUMEN

The in vivo function of murine granulocyte-macrophage colony-stimulating factor (GM-CSF) was investigated in mice, carrying a null allele of the GM-CSF gene, that were generated by gene targeting techniques in embryonic stem cells. Although steady-state hematopoiesis was unimpaired in homozygous mutant animals, all animals developed the progressive accumulation of surfactant lipids and proteins in the alveolar space, the defining characteristic of the idiopathic human disorder pulmonary alveolar proteinosis. Extensive lymphoid hyperplasia associated with lung airways and blood vessels was also found, yet no infectious agents could be detected. These results demonstrate that GM-CSF is not an essential growth factor for basal hematopoiesis and reveal an unexpected, critical role for GM-CSF in pulmonary homeostasis.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Pulmón/patología , Proteinosis Alveolar Pulmonar/patología , Alveolos Pulmonares/metabolismo , Surfactantes Pulmonares/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Hematopoyesis , Homeostasis , Humanos , Hiperplasia , Ratones , Ratones Endogámicos C57BL , Mutación , Proteolípidos/metabolismo , Proteinosis Alveolar Pulmonar/metabolismo , Alveolos Pulmonares/patología , Proteínas Asociadas a Surfactante Pulmonar
5.
J Clin Invest ; 97(3): 649-55, 1996 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-8609219

RESUMEN

Mutation of the granulocyte-macrophage colony-stimulating factor (GM-CSF) gene by homologous recombination caused alveolar proteinosis in mice. To further discern the role of GM-CSF in surfactant homeostasis, the synthesis of GM-CSF was directed to the respiratory epithelium of GM-CSF-hull mutant mice (GM-/-) with a chimeric gene expressing GM-CSF under the control of the promoter from the human surfactant protein-C (SP-C) gene. Transgenic mice bearing the SP-C-GM-CSF construct (SP-C-GM+) were bred to GM-/- mice resulting in complete correction of alveolar proteinosis in bitransgenic GM-/-, SP-C-GM+ mice. No effects of the transgene were found outside the lung. GM-CSF was increased in bronchoalveolar lavage fluid of the bitransgenic mice. Surfactant proteins-A and -B and phospholipid in bronchoalveolar lavage fluid were normalized in the GM-/-, SP-C-GM+ mice. SP-A, -B, and -C mRNAs were unaltered in lungs from GM-CSF-deficient and -replete mice. Expression of GM-CSF in respiratory epithelial cells of transgenic mice restores surfactant homeostasis in GM-/- mice. From these findings, we conclude that GM-CSF regulates the clearance or catabolism rather than synthesis of surfactant proteins and lipids.


Asunto(s)
Terapia Genética/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Proteinosis Alveolar Pulmonar/terapia , Animales , Modelos Animales de Enfermedad , Epitelio/metabolismo , Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Pulmón/anatomía & histología , Ratones , Ratones Mutantes , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteolípidos/genética , Proteinosis Alveolar Pulmonar/genética , Surfactantes Pulmonares/genética , Surfactantes Pulmonares/metabolismo , Distribución Tisular
6.
J Clin Invest ; 96(1): 250-9, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7542280

RESUMEN

The murine TNF-alpha gene was expressed under the control of the human surfactant protein SP-C promoter in transgenic mice. A number of the SP-C TNF-alpha mice died at birth or after a few weeks with very severe lung lesions. Surviving mice transmitted a pulmonary disease to their offspring, the severity and evolution of which was related to the level of TNF-alpha mRNA in the lung; TNF-alpha RNA was detected in alveolar epithelium, presumably in type II epithelial cells. In a longitudinal study of two independent mouse lines, pulmonary pathology, at 1-2 mo of age, consisted of a leukocytic alveolitis with a predominance of T lymphocytes. Leukocyte infiltration was associated with endothelial changes and increased levels of mRNA for the endothelial adhesion molecule VCAM-1. In the following months, alveolar spaces enlarged in association with thickening of the alveolar walls due to an accumulation of desmin-containing fibroblasts, collagen fibers, and lymphocytes. Alveolar surfaces were lined by regenerating type II epithelial cells, and alveolar spaces contained desquamating epithelial cells in places. Platelet trapping in the damaged alveolar capillaries was observed. Pulmonary pathology in the SP-C TNF-alpha mice bears a striking resemblance to human idiopathic pulmonary fibrosis, in which increased expression of TNF-alpha in type II epithelial cells has also been noted. These mice provide a valuable animal model for understanding the pathogenesis of pulmonary fibrosis and exploring possible therapeutic approaches.


Asunto(s)
Modelos Animales de Enfermedad , Fibrosis Pulmonar/etiología , Factor de Necrosis Tumoral alfa/genética , Animales , Secuencia de Bases , Moléculas de Adhesión Celular/genética , Femenino , Inmunohistoquímica , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Datos de Secuencia Molecular , ARN Mensajero/análisis , Factor de Crecimiento Transformador beta/genética , Molécula 1 de Adhesión Celular Vascular
7.
J Clin Invest ; 103(4): 563-9, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10021465

RESUMEN

Granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-targeted mice (GM-/-) cleared group B streptococcus (GBS) from the lungs more slowly than wild-type mice. Expression of GM-CSF in the respiratory epithelium of GM-/- mice improved bacterial clearance to levels greater than that in wild-type GM+/+ mice. Acute aerosolization of GM-CSF to GM+/+ mice significantly enhanced clearance of GBS at 24 hours. GBS infection was associated with increased neutrophilic infiltration in lungs of GM-/- mice, while macrophage infiltrates predominated in wild-type mice, suggesting an abnormality in macrophage clearance of bacteria in the absence of GM-CSF. While phagocytosis of GBS was unaltered, production of superoxide radicals and hydrogen peroxide was markedly deficient in macrophages from GM-/- mice. Lipid peroxidation, assessed by measuring the isoprostane 8-iso-PGF2alpha, was decreased in the lungs of GM-/- mice. GM-CSF plays an important role in GBS clearance in vivo, mediated in part by its role in enhancing superoxide and hydrogen peroxide production and bacterial killing by alveolar macrophages.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Pulmón/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Administración por Inhalación , Animales , Líquido del Lavado Bronquioalveolar , Citocinas/metabolismo , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Susceptibilidad a Enfermedades , F2-Isoprostanos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Pulmón/microbiología , Pulmón/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Nitritos/metabolismo , Fagocitosis , Infecciones Estreptocócicas/microbiología , Superóxidos/metabolismo
8.
J Clin Invest ; 98(4): 914-22, 1996 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-8770862

RESUMEN

The A/Japan/57 influenza hemagglutinin (HA) was expressed in BALB/c mice under the transcriptional control of the surfactant protein C (SP-C) promoter, resulting in expression of HA in type II alveolar epithelial cells, as well as low level variable expression in other tissues, including the thymus in some of the founder lines. Transgenic animals were able to recover from infection with A/Japan/57 influenza, and they were able to mount antibody responses to A/Japan/57 HA in titers similar to wild type. We therefore tested their CD4+ T lymphocyte responses to HA and found them to be similar to wild type responses. However, CD8+ T cells from A/Japan/57-infected transgenic animals were unable to express cytolytic activity against target cells expressing the A/Japan/57 HA. The CD8+ T cell tolerance was also extremely specific, since transgenics immunized with an influenza strain containing a single amino acid substitution in a dominant HA epitope were able to mount full cytolytic responses to that epitope, but not the wild-type epitope. Adoptive transfer of CD8+ T cell clones into transgenic animals resulted extensive interstitial pneumonitis that was antigen-specific and associated with significant morbidity and mortality. We conclude that a lung-specific transgene may lead to specific CD8+ T cell tolerance, with CD4+ T cell and B cell reactivity to the antigen, and that CD4+ T cell reactivity may remain intact to an antigen expressed in the thymus, even when CD8+ T cell tolerance exists. This observation may have profound implications concerning immune-mediated lung diseases, particularly those mediated by CD4+ T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Tolerancia Inmunológica , Transgenes/genética , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Linfocitos B/inmunología , Supresión Clonal , Citotoxicidad Inmunológica , Genes Virales , Hemaglutininas Virales/inmunología , Activación de Linfocitos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Péptidos/química , Péptidos/inmunología , Alveolos Pulmonares/inmunología , Bazo/citología , Proteínas Estructurales Virales/genética
9.
J Clin Invest ; 88(6): 1976-81, 1991 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1752956

RESUMEN

Acute Respiratory Distress Syndrome (ARDS) is characterized by lung injury and damage to the alveolar type II cells. This study sought to determine if endogenous surfactant is altered in ARDS. Bronchoalveolar lavage was performed in patients at-risk to develop ARDS (AR, n = 20), with ARDS (A, n = 66) and in normal subjects (N, n = 29). The crude surfactant pellet was analyzed for total phospholipids (PL), individual phospholipids, SP-A, SP-B, and minimum surface tension (STmin). PL was decreased in both AR and A (3.48 +/- 0.61 and 2.47 +/- 0.40 mumol/ml, respectively) compared to N (7.99 +/- 0.60 mumol/ml). Phosphatidylcholine was decreased in A (62.64 +/- 2.20% PL) compared to N (76.27 +/- 2.05% PL). Phosphatidylglycerol was 11.58 +/- 1.21% PL in N and was decreased to 6.48 +/- 1.43% PL in A. SP-A was 123.64 +/- 20.66 micrograms/ml in N and was decreased to 49.28 +/- 21.68 micrograms/ml in AR and to 29.88 +/- 8.49 micrograms/ml in A. SP-B was 1.28 +/- 0.33 micrograms/ml in N and was decreased to 0.57 +/- 0.24 micrograms/ml in A. STmin was increased in AR (15.1 +/- 2.53 dyn/cm) and A (29.04 +/- 2.05 dyn/cm) compared to N (7.44 +/- 1.61 dyn/cm). These data demonstrate that the chemical composition and functional activity of surfactant is altered in ARDS. Several of these alterations also occur in AR, suggesting that these abnormalities occur early in the disease process.


Asunto(s)
Surfactantes Pulmonares/análisis , Síndrome de Dificultad Respiratoria/metabolismo , Enfermedad Aguda , Adulto , Líquido del Lavado Bronquioalveolar/química , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fosfolípidos/análisis , Riesgo , Tensión Superficial
10.
J Clin Invest ; 86(6): 1954-60, 1990 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2123888

RESUMEN

Tumor necrosis factor-alpha (TNF-alpha) decreased the expression of pulmonary surfactant proteins SP-A and SP-B in human pulmonary adenocarcinoma cell lines. The effect of TNF alpha on SP-A content and mRNA in the pulmonary adenocarcinoma cell line, H441-4, was concentration and time dependent. TNF alpha decreased the cellular content of SP-A to less than 10% of control 48 h after addition. TNF alpha decreased de novo synthesis of SP-A and decreased the accumulation of SP-A in media. SP-A mRNA was decreased within 12 h of addition of TNF alpha, with nearly complete loss of SP-A mRNA observed after 24 h. Inhibitory effects of TNF alpha on SP-A mRNA were dose-related with nearly complete inhibition of SP-A mRNA caused by 25 ng/ml TNF alpha. The effects of TNF alpha on SP-A were distinct from the effects of interferon gamma which increased SP-A content approximately twofold in H441-4 cells. TNF alpha also decreased the content of SP-B mRNA. In contrast to the inhibitory effect of TNF alpha on SP-A and SP-B mRNA, TNF alpha increased mRNA encoding human manganese superoxide dismutase (Mn-SOD). TNF alpha did not inhibit growth, alter cell viability or beta-actin mRNA in either cell line. These in vitro studies demonstrate the marked pretranslational inhibitory effects of the cytokine, TNF alpha, on the expression of pulmonary surfactant proteins, SP-A and SP-B. The results support the concept that macrophage-derived cytokines may control surfactant protein expression.


Asunto(s)
Pulmón/metabolismo , Proteolípidos/metabolismo , Surfactantes Pulmonares/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Northern Blotting , Epitelio/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Técnicas In Vitro , Interferón gamma/farmacología , Pulmón/citología , Proteolípidos/genética , Proteína A Asociada a Surfactante Pulmonar , Proteínas Asociadas a Surfactante Pulmonar , Surfactantes Pulmonares/genética , ARN Mensajero/genética , Superóxido Dismutasa/genética , Células Tumorales Cultivadas
11.
J Clin Invest ; 94(5): 2028-35, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7962549

RESUMEN

We produced transgenic mice which overexpress human IL-6 in the airway epithelial cells. Transgenic mice develop a mononuclear cell infiltrate adjacent to large and mid-sized airways. Immunohistochemistry reveals these cells to be predominantly CD4+ cells, MHC class II+ cells, and B220+ cells. Transgenic mice and nontransgenic mice had similar baseline respiratory system resistance (0.47 +/- 0.06 vs 0.43 +/- 0.04 cmH2O/ml per s at 9 wk of age, P = NS and 0.45 +/- 0.07 vs 0.43 +/- 0.09 cmH2O/ml per s at 17 wk of age, P = NS). Transgenic mice, however, required a significantly higher log dose of methacholine to produce a 100% increase in respiratory system resistance as compared with non-transgenic littermates (1.34 +/- 0.24 vs 0.34 +/- 0.05 mg/ml, P < or = 0.01). We conclude that the expression of human IL-6 in the airways of transgenic mice results in a CD4+, MHC class II+, B220+ lymphocytic infiltrate surrounding large and mid-sized airways that does not alter basal respiratory resistance, but does diminish airway reactivity to methacholine. These findings demonstrate an uncoupling of IL-6-induced airway lymphocytic inflammation and airway hyperresponsiveness and suggest that some forms of airway inflammation may serve to restore altered airway physiology.


Asunto(s)
Hiperreactividad Bronquial/etiología , Interleucina-6/fisiología , Pulmón/patología , Resistencia de las Vías Respiratorias , Animales , Inflamación/patología , Interleucina-6/genética , Cloruro de Metacolina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos
12.
J Clin Invest ; 93(4): 1691-9, 1994 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8163670

RESUMEN

Increased production of EGF or TGF-alpha by the respiratory epithelial cells has been associated with the pathogenesis of various forms of lung injury. Growth factors and cytokines are thought to act locally, via paracrine and autocrine mechanisms, to stimulate cell proliferation and matrix deposition by interstitial lung cells resulting in pulmonary fibrosis. To test whether TGF-alpha mediates pulmonary fibrotic responses, we have generated transgenic mice expressing human TGF-alpha under control of regulatory regions of the human surfactant protein C (SP-C) gene. Human TGF-alpha mRNA was expressed in pulmonary epithelial cells in the lungs of the transgenic mice. Adult mice bearing the SP-C-TGF-alpha transgene developed severe pulmonary fibrosis. Fibrotic lesions were observed in peribronchial, peribronchiolar, and perivascular regions, as well as subjacent to pleural surfaces. Lesions consisted of fibrous tissue that included groups of epithelial cells expressing endogenous SP-C mRNA, consistent with their identification as distal respiratory epithelial cells. Peripheral fibrotic regions consisted of thickened pleura associated with extensive collagen deposition. Alveolar architecture was disrupted in the transgenic mice with loss of alveoli in the lung parenchyma. Pulmonary epithelial cell expression of TGF-alpha in transgenic mice disrupts alveolar morphogenesis and produces fibrotic lesions mediated by paracrine signaling between respiratory epithelial and interstitial cells of the lung.


Asunto(s)
Pulmón/metabolismo , Fibrosis Pulmonar/etiología , Factor de Crecimiento Transformador alfa/fisiología , Animales , Colágeno/metabolismo , Elastina/metabolismo , Epitelio/metabolismo , Femenino , Pulmón/patología , Masculino , Ratones , Ratones Transgénicos , Alveolos Pulmonares/patología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , ARN Mensajero/análisis , Factor de Crecimiento Transformador alfa/biosíntesis , Factor de Crecimiento Transformador alfa/genética
13.
J Clin Invest ; 103(5): 637-47, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10074480

RESUMEN

It is well established that maternal smoking during pregnancy is a leading preventable cause of low birth weight and prematurity. Less appreciated is that maternal smoking during pregnancy is also associated with alterations in pulmonary function at birth and greater incidence of respiratory illnesses after birth. To determine if this is the direct result of nicotine interacting with nicotinic cholinergic receptors (nAChRs) during lung development, rhesus monkeys were treated with 1 mg/kg/day of nicotine from days 26 to 134 of pregnancy. Nicotine administration caused lung hypoplasia and reduced surface complexity of developing alveoli. Immunohistochemistry and in situ alpha-bungarotoxin (alphaBGT) binding showed that alpha7 nAChRs are present in the developing lung in airway epithelial cells, cells surrounding large airways and blood vessels, alveolar type II cells, free alveolar macrophages, and pulmonary neuroendocrine cells (PNEC). As detected both by immunohistochemistry and by alphaBGT binding, nicotine administration markedly increased alpha7 receptor subunit expression and binding in the fetal lung. Correlating with areas of increased alpha7 expression, collagen expression surrounding large airways and vessels was significantly increased. Nicotine also significantly increased numbers of type II cells and neuroendocrine cells in neuroepithelial bodies. These findings demonstrate that nicotine can alter fetal monkey lung development by crossing the placenta to interact directly with nicotinic receptors on non-neuronal cells in the developing lung, and that similar effects likely occur in human infants whose mothers smoke during pregnancy.


Asunto(s)
Pulmón/embriología , Pulmón/metabolismo , Exposición Materna , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Receptores Nicotínicos/biosíntesis , Animales , Desarrollo Embrionario y Fetal/efectos de los fármacos , Femenino , Haplorrinos , Humanos , Pulmón/patología , Embarazo
14.
Mol Cell Biol ; 14(9): 5671-81, 1994 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8065304

RESUMEN

We used the lung epithelial cell-specific surfactant protein B (SPB) gene promoter as a model with which to investigate mechanisms involved in transcriptional control of lung-specific genes. In a previous study, we showed that the SPB promoter specifically activated expression of a linked reporter gene in the continuous H441 lung cell line and that H441 nuclear proteins specifically protected a region of this promoter from bp -111 to -73. In this study, we further show that this region is a complex binding site for thyroid transcription factor 1 (TTF-1) and hepatocyte nuclear factor 3 (HNF-3). Whereas TTF-1 bound two highly degenerate and closely spaced sites, HNF-3 proteins bound a TGT3 motif (TGTTTGT) that is also found in several liver-specific gene regulatory regions, where it appears to be a weak affinity site for HNF-3. Point mutations of these binding sites eliminated factor binding and resulted in significant decreases in transfected SPB promoter activity. In addition, we developed a cotransfection assay and showed that a family of lung-specific gene promoters that included the SPB, SPC, SPA, and Clara cell secretory protein (CCSP) gene promoters were specifically activated by cotransfected TTF-1. We conclude that TTF-1 and HNF-3 are major activators of lung-specific genes and propose that these factors are involved in a general mechanism of lung-specific gene transcription. Importantly, these data also show that common factors are involved in organ-specific gene expression along the mammalian foregut axis.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Pulmón/fisiología , Proteínas Nucleares/fisiología , Regiones Promotoras Genéticas , Proteolípidos/genética , Surfactantes Pulmonares/genética , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Sitios de Unión , Factor Nuclear 3-alfa del Hepatocito , Factor Nuclear 3-beta del Hepatocito , Técnicas In Vitro , Hígado/fisiología , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética , Glándula Tiroides/fisiología , Factor Nuclear Tiroideo 1 , Activación Transcripcional
15.
Mol Cell Biol ; 13(7): 3860-71, 1993 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8321193

RESUMEN

We have shown that a large fragment (-2339 to +57) from the rat CC10 gene directed lung-specific expression of a reporter construct in transgenic animals. Upon transfection, a smaller fragment (-165 to +57) supported reporter gene expression exclusively in the Clara cell-like NCI-H441 cell line, suggesting that a Clara cell-specific transcriptional element resided on this fragment (B. R. Stripp, P. L. Sawaya, D. S. Luse, K. A. Wikenheiser, S. E. Wert, J. A. Huffman, D. L. Lattier, G. Singh, S. L. Katyal, and J. A. Whitsett, J. Biol. Chem. 267:14703-14712, 1992). The interactions of nuclear proteins with a particular segment of the CC10 promoter which extends from 79 to 128 bp upstream of the CC10 transcription initiation site (CC10 region I) have now been studied. This sequence can stimulate both in vitro transcription in H441 nuclear extract and transient expression of reporter constructs in H441 cells. Electrophoretic mobility shift assays using extracts from H441, HeLa, rat liver, and fetal sheep lung cells were used to demonstrate that members of the AP-1, octamer, and HNF-3 families bind to CC10 region I. Transcription factors from H441 cells which are capable of binding to CC10 region I are either absent in HeLa, rat liver, and fetal sheep lung extracts or enriched in H441 extracts relative to extracts from non-Clara cells.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Proteínas/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factores de Transcripción/metabolismo , Uteroglobina , Animales , Secuencia de Bases , Línea Celular , ADN , Células HeLa , Factor Nuclear 3-alfa del Hepatocito , Factor C1 de la Célula Huésped , Humanos , Datos de Secuencia Molecular , Factor 1 de Transcripción de Unión a Octámeros , Ratas , Ratas Sprague-Dawley , Ovinos , Transfección
16.
Mol Cell Biol ; 16(7): 3626-36, 1996 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8668179

RESUMEN

Thyroid transcription factor 1 (TTF-1), hepatocyte nuclear factor 3alpha (HNF-3alpha), and HNF-3beta regulate the transcription of genes expressed in the respiratory epithelium. To test whether members of the HNF-3/forkhead family influence TTF-1 gene expression, deletion constructs containing the 5' region of the human TTF-1 gene were transfected into immortalized mouse lung epithelial (MLE) cells. DNase I protection and electrophoretic mobility shift assays identified elements in the 5' region of the TTF-1 gene that bound MLE cell nuclear proteins consistent with the binding of HNF-3 to sites at positions -135 to -124 and -14 to -3. In MLE cells, TTF-1-luciferase reporter constructs were activated by cotransfection with HNF-3beta, activated to a lesser extent by HNF-3alpha, but not activated by HFH-8. HNF-3alpha. and HFH-8 inhibited the activation of TTF-1-luciferase by HNF-3beta. Site-specific mutagenesis of each of the HNF-3 binding sites in the human TTF-1 gene inhibited the binding of MLE cell nuclear proteins and inhibited transactivation of the TTF-1-luciferase constructs after cotransfection with HNF-3beta. Immunohistochemical staining demonstrated that both HNF-3beta and TTF-1 were detected in bronchiolar and alveolar type II cells in the human lung. Modulation of TTF-1 gene expression by members of the HNF-3/forkhead family members may provide a mechanism by which distinct HNF-3/forkhead family members influence respiratory epithelial cell gene expression and cell differentiation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Pulmón/metabolismo , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/metabolismo , Transcripción Genética , Activación Transcripcional , Células 3T3 , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Secuencia de Consenso , ADN/química , ADN/metabolismo , Proteínas de Unión al ADN/biosíntesis , Células Epiteliales , Epitelio/metabolismo , Factor Nuclear 3-alfa del Hepatocito , Humanos , Inmunohistoquímica , Recién Nacido , Luciferasas/biosíntesis , Pulmón/citología , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Mapeo Restrictivo , Eliminación de Secuencia , Factor Nuclear Tiroideo 1 , Transfección
17.
J Perinatol ; 26(6): 371-4, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16724078

RESUMEN

Two surviving female infants, born from a triplet pregnancy at 30 weeks gestation, were noted to have severe osteopenia and multiple fractures diagnosed at 20 days of age. Their mother had been treated for preterm labor with intravenous magnesium sulfate from week 22 until their birth at 30 weeks gestation. At birth, the triplets exhibited craniotabes with enlarged fontanelles and sutures. All developed Respiratory Distress Syndrome (RDS) and the two surviving infants required prolonged respiratory support. Serum calcium and phosphate levels were normal and alkaline phosphatase levels were increased. The infants were treated with supplements of calcium and phosphorous, with resultant healing of the multiple fractures without deformity. Fetal magnesium toxicity impairs bone mineralization and can lead to serious bone demineralization that may cause fractures in the newborn period that complicate recovery from respiratory disease. Early recognition and treatment may minimize complications related to osteopenia caused by fetal magnesium toxicity.


Asunto(s)
Desmineralización Ósea Patológica/inducido químicamente , Feto/efectos de los fármacos , Fracturas Óseas/inducido químicamente , Magnesio/envenenamiento , Adulto , Desmineralización Ósea Patológica/diagnóstico por imagen , Resultado Fatal , Femenino , Fémur/diagnóstico por imagen , Fracturas Óseas/diagnóstico por imagen , Humanos , Recién Nacido , Inyecciones Intravenosas , Sulfato de Magnesio/administración & dosificación , Sulfato de Magnesio/envenenamiento , Sulfato de Magnesio/uso terapéutico , Masculino , Trabajo de Parto Prematuro/tratamiento farmacológico , Embarazo , Radiografía , Costillas/diagnóstico por imagen , Tibia/diagnóstico por imagen , Tocolíticos/administración & dosificación , Tocolíticos/envenenamiento , Tocolíticos/uso terapéutico , Trillizos
18.
J Natl Cancer Inst ; 71(4): 779-86, 1983 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-6312152

RESUMEN

beta-Adrenergic receptors were demonstrated in membrane preparations from 6 human Ewing's sarcomas and compared to those from 46 other pediatric cancers with the use of the beta-adrenergic antagonist (-)-(3H)dihydroalprenolol [(-)[3H]DHA]. In contrast to the high numbers of receptor sites found in Ewing's sarcomas (55-640 fmol x mg-1 protein; dissociation constant Kd, 1-2 nM), other childhood cancers (neuroblastoma, rhabdomyosarcoma, brain tumors, lymphoma, osteosarcoma, hepatoblastoma, yolk sac, and Wilms' tumor) contained in general fewer beta-adrenergic receptor sites. Characteristics of (-)-[3H]DHA binding were therefore more fully characterized in the Ewing's tumors. Competition of (-)-[3H]DHA binding by classical catecholamine agonists, as well as by subtype selective agents metoprolol and zinterol, demonstrated the presence of a homogeneous population of beta 1-adrenergic sites in several Ewing's tumors. Adenylate cyclase activity in all Ewing's sarcomas was enhanced by GTP and NaF. However, in spite of high numbers of beta-adrenergic receptors, (-)-isoproterenol was not very effective in the activation of adenylate cyclase activity in several of the Ewing's tumors tested. Neither guanyl-5'-yl-imidophosphate nor GTP altered agonist potency for the receptor site in these catecholamine-insensitive tumors. Hill coefficients obtained from the competition experiments with (-)-isoproterenol (in the presence or absence of guanine nucleotide) were approximately 1.0. These uncoupled receptors were resistant to N-ethylmaleimide denaturation and were densensitized only 50% during culture in the presence of (-)-isoproterenol. Thus Ewing's sarcomas are relatively rich in beta-adrenergic sites, and several tumors appear to have a coupling lesion involving guanine nucleotide-dependent regulatory protein interaction with beta-adrenergic receptors and adenylate cyclase, similar in phenotype to that described in the (unc) variant of S49 mouse lymphoma.


Asunto(s)
Neoplasias Óseas/metabolismo , Receptores Adrenérgicos beta/análisis , Sarcoma de Ewing/metabolismo , Adenilil Ciclasas/metabolismo , Antagonistas Adrenérgicos beta , Animales , Unión Competitiva , Línea Celular , Niño , Dihidroalprenolol/farmacología , Etanolaminas/farmacología , Humanos , Metoprolol/farmacología , Ratones , Ratones Desnudos , Neuroblastoma , Osteosarcoma
19.
Cancer Res ; 52(19): 5342-52, 1992 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-1394139

RESUMEN

A model of pulmonary adenocarcinomas was produced in transgenic mice harboring a chimeric gene comprising the SV40 large T antigen under the control of a transcriptional region derived from the human surfactant protein C (SP-C) gene. Transgenic mice succumbed with pulmonary tumors within 4-5 months of age. By histology, the tumors were adenocarcinomas with lepidic, papillary, and solid growth patterns that were indistinguishable from adenocarcinomas occurring in humans. Immunocytochemistry demonstrated the lack of staining for neuroendocrine markers, consistent with the identification of the tumors as non-small cell rather than small cell carcinomas. The presence of SV40 large T mRNA in the lung and tumors was detected by in situ hybridization and Northern blot analysis. Exogenous SV40 large T mRNA and endogenous CC10 (a nonciliated respiratory epithelial cell marker) and SP-C (a Type II alveolar cell marker) mRNAs were expressed at variable levels in the lung tumors. SV40 large T mRNA and CC10 mRNA were detected in the majority of tumors, while SP-C mRNA was detected less frequently. The heterogeneity of bronchiolar and alveolar cell markers in the tumors from the transgenic mice supports the concept that tumorigenesis was initiated in distinct subsets of epithelial cells that produce characteristic adenocarcinomas of the lung.


Asunto(s)
Adenocarcinoma/genética , Antígenos Transformadores de Poliomavirus/genética , Genes Reguladores/genética , Neoplasias Pulmonares/genética , Proteolípidos/genética , Surfactantes Pulmonares/genética , Adenocarcinoma/inmunología , Animales , Biomarcadores de Tumor/análisis , Northern Blotting , Quimera/genética , Modelos Animales de Enfermedad , Epitelio/fisiología , Expresión Génica/genética , Humanos , Pulmón/fisiología , Neoplasias Pulmonares/inmunología , Ratones , Ratones Transgénicos , Hibridación de Ácido Nucleico , ARN Mensajero/genética
20.
Cancer Res ; 50(17): 5481-7, 1990 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-2386953

RESUMEN

Clara cells and type II pneumocytes are the progenitor cells of the bronchioles and alveoli, respectively. These peripheral airway cells (PAC) contain characteristic cytoplasmic structures and express surfactant associated proteins. PAC cell markers are expressed by many pulmonary adenocarcinomas having papillary and/or lepidic growth patterns, which are characteristics of the bronchioloalveolar and papillary subtypes. We investigated the expression of PAC markers in a panel of 41 lung cancer cell lines. Ultrastructural studies demonstrated the presence of cytoplasmic structures characteristic of Clara cells or of type II pneumocytes in 9 of 34 (26%) non-small cell lung cancer cell lines, including 7 of 17 (41%) adenocarcinomas, one squamous cell carcinoma, and one large cell carcinoma. Of interest, the cytoplasmic structures were present in 5 of 6 (83%) cell lines initiated from papillolepidic adenocarcinomas. In addition, we examined the lines for expression of the surfactant associated proteins SP-A, SP-B, and SP-C. Eight of the nine cell lines containing cytoplasmic inclusions characteristic of PAC cells also expressed protein and/or RNA of SP-A, the major surfactant associated protein. Five of these lines expressed SP-B RNA (either constitutively or after dexamethasone induction), while a single line expressed SP-C only after dexamethasone induction. None of six small cell lung cancer cell lines examined expressed any of the PAC markers. Thus, PAC markers are expressed frequently (but not exclusively) in pulmonary adenocarcinoma cell lines, especially in those initiated from tumors having papillolepidic growth patterns. The establishment and identification of multiple cell lines expressing PAC features provide an important new resource for biological and preclinical therapeutic studies.


Asunto(s)
Adenocarcinoma/patología , Carcinoma/patología , Neoplasias Pulmonares/patología , Proteolípidos/análisis , Surfactantes Pulmonares/análisis , Sistema Respiratorio/patología , Células Tumorales Cultivadas/citología , Adulto , Animales , Carcinoma/clasificación , Línea Celular , Sondas de ADN , Dexametasona/farmacología , Glicoproteínas/análisis , Humanos , Neoplasias Pulmonares/ultraestructura , Ratones , Ratones Desnudos , Proteolípidos/genética , Proteína A Asociada a Surfactante Pulmonar , Proteínas Asociadas a Surfactante Pulmonar , Surfactantes Pulmonares/genética , Sistema Respiratorio/citología , Trasplante Heterólogo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA