Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Respir Cell Mol Biol ; 63(1): 67-78, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32101459

RESUMO

Epithelial dysfunction in the small airways may cause the development of emphysema in chronic obstructive pulmonary disease. C/EBPα (CCAAT/enhancer binding protein-α), a transcription factor, is required for lung maturation during development, and is also important for lung homeostasis after birth, including the maintenance of serine protease/antiprotease balance in the bronchiolar epithelium. This study aimed to show the roles of C/EBPα in the distal airway during chronic cigarette smoke exposure in mice and in the small airways in smokers. In a model of chronic smoke exposure using epithelial cell-specific C/EBPα-knockout mice, significant pathological phenotypes, such as higher protease activity, impaired ciliated cell regeneration, epithelial cell barrier dysfunction via reduced zonula occludens-1 (Zo-1), and decreased alveolar attachments, were found in C/EBPα-knockout mice compared with control mice. We found that Spink5 (serine protease inhibitor kazal-type 5) gene (encoding lymphoepithelial Kazal-type-related inhibitor [LEKTI], an anti-serine protease) expression in the small airways is a key regulator of protease activity in this model. Finally, we showed that daily antiprotease treatment counteracted the phenotypes of C/EBPα-knockout mice. In human studies, CEBPA (CCAAT/enhancer binding protein-α) gene expression in the lung was downregulated in patients with emphysema, and six smokers with centrilobular emphysema (CLE) showed a significant reduction in LEKTI in the small airways compared with 22 smokers without CLE. LEKTI downregulation in the small airways was associated with disease development during murine small airway injury and CLE in humans, suggesting that LEKTI might be a key factor linking small airway injury to the development of emphysema.


Assuntos
Pulmão/metabolismo , Pulmão/patologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patologia , Serina Proteases/metabolismo , Animais , Bronquíolos/metabolismo , Bronquíolos/patologia , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Inibidor de Serinopeptidase do Tipo Kazal 5/metabolismo , Fumar/metabolismo
2.
J Biol Chem ; 290(30): 18559-74, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26048993

RESUMO

Pulmonary surfactant, a mixture of proteins and phospholipids, plays an important role in facilitating gas exchange by maintaining alveolar stability. Saturated phosphatidylcholine (SatPC), the major component of surfactant, is synthesized both de novo and by the remodeling of unsaturated phosphatidylcholine (PC) by lyso-PC acyltransferase 1 (LPCAT1). After synthesis in the endoplasmic reticulum, SatPC is routed to lamellar bodies (LBs) for storage prior to secretion. The mechanism by which SatPC is transported to LB is not understood. The specificity of LPCAT1 for lyso-PC as an acyl acceptor suggests that formation of SatPC via LPCAT1 reacylation is a final step in SatPC synthesis prior to transport. We hypothesized that LPCAT1 forms a transient complex with SatPC and specific phospholipid transport protein(s) to initiate trafficking of SatPC from the endoplasmic reticulum to the LB. Herein we have assessed the ability of different StarD proteins to interact with LPCAT1. We found that LPCAT1 interacts with StarD10, that this interaction is direct, and that amino acids 79-271 of LPCAT1 and the steroidogenic acute regulatory protein-related lipid transfer (START) domain of START domain-containing protein 10 (StarD10) are sufficient for this interaction. The role of StarD10 in trafficking of phospholipid to LB was confirmed by the observation that knockdown of StarD10 significantly reduced transport of phospholipid to LB. LPCAT1 also interacted with one isoform of StarD7 but showed no interaction with StarD2/PC transfer protein.


Assuntos
1-Acilglicerofosfocolina O-Aciltransferase/metabolismo , Metabolismo dos Lipídeos , Fosfolipídeos/biossíntese , Fosfoproteínas/metabolismo , Alvéolos Pulmonares/metabolismo , 1-Acilglicerofosfocolina O-Aciltransferase/genética , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Humanos , Camundongos , Fosfatidilcolinas/biossíntese , Fosfatidilcolinas/metabolismo , Fosfolipídeos/metabolismo , Fosfoproteínas/genética , Mapas de Interação de Proteínas/genética , Alvéolos Pulmonares/citologia , Surfactantes Pulmonares/metabolismo
3.
Am J Respir Cell Mol Biol ; 50(4): 777-86, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24199692

RESUMO

Pulmonary fibrosis is caused by excessive proliferation and accumulation of stromal cells. Fibrocytes are bone marrow (BM)-derived cells that contribute to pathologic stromal cell accumulation in human lung disease. However, the cellular source for these stromal cells and the degree of fibrocyte contribution to pulmonary fibrosis remain unclear. To determine the etiology of stromal cell excess during pulmonary fibrosis, we measured fibrocytes during the progression of fibrosis in the transforming growth factor (TGF)-α transgenic mouse model. Lung epithelial-specific overexpression of TGF-α led to progressive pulmonary fibrosis associated with increased accumulation of fibrocytes in the fibrotic lesions. Although reconstitution of BM cells into TGF-α mice demonstrated accumulation of these cells in fibrotic lesions, the majority of the cells did not express α-smooth muscle actin, suggesting that fibrocytes did not transform into myofibroblasts. To explore the mechanisms of fibrocytes in pulmonary fibrogenesis, adoptive cell-transfer experiments were performed. Purified fibrocytes were transferred intravenously into TGF-α transgenic mice, and fibrosis endpoints were compared with controls. Analysis of lung histology and hydroxyproline levels demonstrated that fibrocyte transfers augment TGF-α-induced lung fibrosis. A major subset of TGF-α-induced fibrocytes expressed CD44 and displayed excessive invasiveness, which is attenuated in the presence of anti-CD44 antibodies. Coculture experiments of resident fibroblasts with fibrocytes demonstrated that fibrocytes stimulate proliferation of resident fibroblasts. In summary, fibrocytes are increased in the progressive, fibrotic lesions of TGF-α-transgenic mice and activate resident fibroblasts to cause severe lung disease.


Assuntos
Células da Medula Óssea/metabolismo , Movimento Celular , Proliferação de Células , Fibroblastos/metabolismo , Pulmão/metabolismo , Fibrose Pulmonar/metabolismo , Células Estromais/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Transferência Adotiva , Animais , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Progressão da Doença , Fibroblastos/patologia , Fibroblastos/transplante , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Receptores de Hialuronatos/metabolismo , Hidroxiprolina/metabolismo , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , Células Estromais/patologia , Células Estromais/transplante , Fatores de Tempo , Fator de Crescimento Transformador alfa/genética , Regulação para Cima
4.
Am J Physiol Lung Cell Mol Physiol ; 306(8): L726-35, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24508732

RESUMO

A number of growth factors and signaling pathways regulate matrix deposition and fibroblast proliferation in the lung. The epidermal growth factor receptor (EGFR) family of receptors and the transforming growth factor-ß (TGF-ß) family are active in diverse biological processes and are central mediators in the initiation and maintenance of fibrosis in many diseases. Transforming growth factor-α (TGF-α) is a ligand for the EGFR, and doxycycline (Dox)-inducible transgenic mice conditionally expressing TGF-α specifically in the lung epithelium develop progressive fibrosis accompanied with cachexia, changes in lung mechanics, and marked pleural thickening. Although recent studies demonstrate that EGFR activation modulates the fibroproliferative effects involved in the pathogenesis of TGF-ß induced pulmonary fibrosis, in converse, the direct role of EGFR induction of the TGF-ß pathway in the lung is unknown. The αvß6 integrin is an important in vivo activator of TGF-ß activation in the lung. Immunohistochemical analysis of αvß6 protein expression and bronchoalveolar analysis of TGF-ß pathway signaling indicates activation of the αvß6/TGF-ß pathway only at later time points after lung fibrosis was already established in the TGF-α model. To determine the contribution of the αvß6/TGF-ß pathway on the progression of established fibrotic disease, TGF-α transgenic mice were administered Dox for 4 wk, which leads to extensive fibrosis; these mice were then treated with a function-blocking anti-αvß6 antibody with continued administration of Dox for an additional 4 wk. Compared with TGF-α transgenic mice treated with control antibody, αvß6 inhibition significantly attenuated pleural thickening and altered the decline in lung mechanics. To test the effects of genetic loss of the ß6 integrin, TGF-α transgenic mice were mated with ß6-null mice and the degree of fibrosis was compared in adult mice following 8 wk of Dox administration. Genetic ablation of the ß6 integrin attenuated histological and physiological changes in the lungs of TGF-α transgenic mice although a significant degree of fibrosis still developed. In summary, inhibition of the ß6 integrin led to a modest, albeit significant, effect on pleural thickening and lung function decline observed with TGF-α-induced pulmonary fibrosis. These data support activation of the αvß6/TGF-ß pathway as a secondary effect contributing to TGF-α-induced pleural fibrosis and suggest a complex contribution of multiple mediators to the maintenance of progressive fibrosis in the lung.


Assuntos
Integrinas/antagonistas & inibidores , Fibrose Pulmonar/patologia , Fator de Crescimento Transformador alfa/farmacologia , Animais , Antibacterianos/toxicidade , Anticorpos Neutralizantes , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Lavagem Broncoalveolar , Colágeno , Doxiciclina/toxicidade , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Técnicas Imunoenzimáticas , Integrinas/genética , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/farmacologia , Uteroglobina/fisiologia
5.
Am J Respir Cell Mol Biol ; 49(3): 348-57, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23590306

RESUMO

Pulmonary surfactant levels within the alveoli are tightly regulated to maintain lung volumes and promote efficient gas exchange across the air/blood barrier. Quantitative and qualitative abnormalities in surfactant are associated with severe lung diseases in children and adults. Although the cellular and molecular mechanisms that control surfactant metabolism have been studied intensively, the critical molecular pathways that sense and regulate endogenous surfactant levels within the alveolus have not been identified and constitute a fundamental knowledge gap in the field. In this study, we demonstrate that expression of an orphan G protein-coupled receptor, GPR116, in the murine lung is developmentally regulated, reaching maximal levels 1 day after birth, and is highly expressed on the apical surface of alveolar type I and type II epithelial cells. To define the physiological role of GPR116 in vivo, mice with a targeted mutation of the Gpr116 locus, Gpr116(Δexon17), were generated. Gpr116(Δexon17) mice developed a profound accumulation of alveolar surfactant phospholipids at 4 weeks of age (12-fold) that was further increased at 20 weeks of age (30-fold). Surfactant accumulation in Gpr116(Δexon17) mice was associated with increased saturated phosphatidylcholine synthesis at 4 weeks and the presence of enlarged, lipid-laden macrophages, neutrophilia, and alveolar destruction at 20 weeks. mRNA microarray analyses indicated that P2RY2, a purinergic receptor known to mediate surfactant secretion, was induced in Gpr116(Δexon17) type II cells. Collectively, these data support the concept that GPR116 functions as a molecular sensor of alveolar surfactant lipid pool sizes by regulating surfactant secretion.


Assuntos
Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Macrófagos/metabolismo , Alvéolos Pulmonares/metabolismo , Surfactantes Pulmonares/metabolismo , Receptores Acoplados a Proteínas G/genética , Mucosa Respiratória/metabolismo , Animais , Líquido da Lavagem Broncoalveolar/química , Células Epiteliais/patologia , Éxons , Macrófagos/patologia , Camundongos , Camundongos Knockout , Fosfatidilcolinas/biossíntese , Alvéolos Pulmonares/patologia , Receptores Acoplados a Proteínas G/deficiência , Receptores Purinérgicos P2Y2/genética , Receptores Purinérgicos P2Y2/metabolismo , Mucosa Respiratória/patologia , Transdução de Sinais
6.
Dev Biol ; 362(1): 24-41, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22094019

RESUMO

Hypoxia inducible factor (HIF) 1a, EPAS1 and NEPAS are expressed in the embryonic mouse lung and each isoform exhibits distinct spatiotemporal expression patterns throughout morphogenesis. To further assess the role of the HIF1a isoform in lung epithelial cell differentiation and homeostasis, we created transgenic mice that express a constitutively active isoform of human HIF-1a (HIF-1a three point mutant (TPM)), in a doxycycline-dependent manner. Expression of HIF1a TPM in the developing pulmonary epithelium resulted in lung hypoplasia characterized by defective branching morphogenesis, altered cellular energetics and impaired epithelial maturation, culminating in neonatal lethality at birth from severe respiratory distress. Histological and biochemical analyses revealed expanded glycogen pools in the pulmonary epithelial cells at E18.5, concomitant with decreased pulmonary surfactant, suggesting a delay or an arrest in maturation. Importantly, these defects occurred in the absence of apoptosis or necrosis. In addition, sub-pleural hemorrhaging was evident as early as E14.5 in HIF1a TPM lungs, despite normal patterning of the blood vasculature, consistent with defects in endothelial barrier function. Epithelial expression of HIF1a TPM also resulted in increased VEGFA and VEGFC production, an increase in the number of lymphatic vessels and indirect activation of the multiple Notch pathway components in endothelial precursor cells. Collectively, these data indicate that HIF-1a protein levels in the pulmonary epithelium must be tightly controlled for proper development of the epithelial and mesenchymal compartments.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/embriologia , Linfangiogênese/fisiologia , Mucosa Respiratória/embriologia , Análise de Variância , Animais , Primers do DNA/genética , DNA Mitocondrial/genética , Doxiciclina , Vetores Genéticos/genética , Glicogênio/metabolismo , Soros Imunes/genética , Immunoblotting , Imuno-Histoquímica , Pulmão/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Fosfatidilcolinas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Mucosa Respiratória/metabolismo , Transgenes/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo
7.
Dev Biol ; 370(2): 198-212, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22885335

RESUMO

Respiratory epithelial cells are derived from cell progenitors in the foregut endoderm that subsequently differentiate into the distinct cell types lining the conducting and alveolar regions of the lung. To identify transcriptional mechanisms regulating differentiation and maintenance of respiratory epithelial cells, we conditionally deleted Foxm1 transcription factor from the conducting airways of the developing mouse lung. Conditional deletion of Foxm1 from Clara cells, controlled by the Scgb1a1 promoter, dramatically altered airway structure and caused peribronchial fibrosis, resulting in airway hyperreactivity in adult mice. Deletion of Foxm1 inhibited proliferation of Clara cells and disrupted the normal patterning of epithelial cell differentiation in the bronchioles, causing squamous and goblet cell metaplasia, and the loss of Clara and ciliated cells. Surprisingly, conducting airways of Foxm1-deficient mice contained highly differentiated cuboidal type II epithelial cells that are normally restricted to the alveoli. Lineage tracing studies showed that the ectopic alveolar type II cells in Foxm1-deficient airways were derived from Clara cells. Deletion of Foxm1 inhibited Sox2 and Scgb1a1, both of which are critical for differentiation and function of Clara cells. In co-transfection experiments, Foxm1 directly bound to and induced transcriptional activity of Scgb1a1 and Sox2 promoters. Foxm1 is required for differentiation and maintenance of epithelial cells lining conducting airways.


Assuntos
Diferenciação Celular , Proliferação de Células , Fatores de Transcrição Forkhead/metabolismo , Sistema Respiratório/citologia , Sistema Respiratório/crescimento & desenvolvimento , Animais , Células Epiteliais/citologia , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/genética , Deleção de Genes , Pulmão/citologia , Pulmão/crescimento & desenvolvimento , Camundongos , Alvéolos Pulmonares/citologia , Fatores de Transcrição SOXB1/metabolismo , Transcriptoma , Uteroglobina/metabolismo
8.
J Biol Chem ; 287(13): 10099-10114, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22267724

RESUMO

Pulmonary inflammation is associated with altered lipid synthesis and clearance related to diabetes, obesity, and various inherited metabolic disorders. In many tissues, lipogenesis is regulated at the transcriptional level by the activity of sterol-response element-binding proteins (SREBP). The role of SREBP activation in the regulation of lipid metabolism in the lung was assessed in mice in which both Insig1 and Insig2 genes, encoding proteins that bind and inhibit SREBPs in the endoplasmic reticulum, were deleted in alveolar type 2 cells. Although deletion of either Insig1 or Insig2 did not alter SREBP activity or lipid homeostasis, deletion of both genes (Insig1/2(Δ/Δ) mice) activated SREBP1, causing marked accumulation of lipids that consisted primarily of cholesterol esters and triglycerides in type 2 epithelial cells and alveolar macrophages. Neutral lipids accumulated in type 2 cells in association with the increase in mRNAs regulating fatty acid, cholesterol synthesis, and inflammation. Although bronchoalveolar lavage fluid phosphatidylcholine was modestly decreased, lung phospholipid content and lung function were maintained. Insig1/2(Δ/Δ) mice developed lung inflammation and airspace abnormalities associated with the accumulation of lipids in alveolar type 2 cells, alveolar macrophages, and within alveolar spaces. Deletion of Insig1/2 activated SREBP-enhancing lipogenesis in respiratory epithelial cells resulting in lipotoxicity-related lung inflammation and tissue remodeling.


Assuntos
Lipogênese , Proteínas de Membrana/metabolismo , Pneumonia/metabolismo , Alvéolos Pulmonares/metabolismo , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Animais , Ésteres do Colesterol/genética , Ésteres do Colesterol/metabolismo , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Pneumonia/genética , Pneumonia/patologia , Alvéolos Pulmonares/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Proteínas de Ligação a Elemento Regulador de Esterol/genética , Triglicerídeos/genética , Triglicerídeos/metabolismo
9.
Am J Respir Cell Mol Biol ; 47(4): 454-63, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22652201

RESUMO

Many transcription factors that regulate lung morphogenesis during development are reactivated to mediate repairs of the injured adult lung. We hypothesized that CCAAT/enhancer binding protein-α (C/EBPα), a transcription factor critical for perinatal lung maturation, regulates genes required for the normal repair of the bronchiolar epithelium after injury. Transgenic Cebpα(Δ/Δ) mice, in which Cebpa was conditionally deleted from Clara cells and Type II cells after birth, were used in this study. Airway injury was induced in mice by the intraperitoneal administration of naphthalene to ablate bronchiolar epithelial cells. Although the deletion of C/EBPα did not influence lung structure and function under unstressed conditions, C/EBPα was required for the normal repair of terminal bronchiolar epithelium after naphthalene injury. To identify cellular processes that are influenced by C/EBPα during repair, mRNA microarray was performed on terminal bronchiolar epithelial cells isolated by laser-capture microdissection. Normal repair of the terminal bronchiolar epithelium was highly associated with the mRNAs regulating antiprotease activities, and their induction required C/EBPα. The defective deposition of fibronectin in Cebpα(Δ/Δ) mice was associated with increased protease activity and delayed differentiation of FoxJ1-expressing ciliated cells. The fibronectin and ciliated cells were restored by the intratracheal treatment of Cebpα(Δ/Δ) mice with the serine protease inhibitor. In conclusion, C/EBPα regulates the expression of serine protease inhibitors that are required for the normal increase of fibronectin and the restoration of ciliated cells after injury. Treatment with serine protease inhibitor may aid in the recovery of injured bronchiolar epithelial cells, and prevent common chronic lung diseases.


Assuntos
Bronquíolos/patologia , Proteínas Estimuladoras de Ligação a CCAAT/fisiologia , Pulmão/patologia , Peptídeo Hidrolases/metabolismo , Proteínas Secretadas Inibidoras de Proteinases/metabolismo , Regeneração , Mucosa Respiratória/enzimologia , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/enzimologia , Lesão Pulmonar Aguda/patologia , Animais , Aprotinina/farmacologia , Bronquíolos/fisiologia , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem da Célula , Feminino , Fibronectinas/metabolismo , Regulação da Expressão Gênica , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Naftalenos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Secretadas Inibidoras de Proteinases/genética , Mucosa Respiratória/metabolismo , Inibidores de Serina Proteinase/farmacologia , Transcriptoma
10.
Am J Respir Cell Mol Biol ; 46(3): 380-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22021337

RESUMO

Pulmonary fibrosis remains a significant public health burden with no proven therapies. The mitogen-activated protein kinase (MAPK)/MAPK kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling cascade is a major pathway controlling cellular processes associated with fibrogenesis, including growth, proliferation, and survival. Activation of the MAPK/ERK pathway is detected in the lungs of human fibrosis samples; however, the effect of modulating the pathway in vivo is unknown. Overexpression of transforming growth factor (TGF)-α in the lung epithelium of transgenic mice causes a progressive pulmonary fibrosis associated with increased MEK/ERK activation localized primarily in mesenchymal cells. To determine the role of the MEK pathway in the induction of TGF-α-induced lung fibrosis, TGF-α was overexpressed for 4 weeks while mice were simultaneously treated with the specific MEK inhibitor, ARRY-142886 (ARRY). Treatment with ARRY prevented increases in lung cell proliferation and total lung collagen, attenuated production of extracellular matrix genes, and protected mice from changes in lung function. ARRY administered as a rescue treatment after fibrosis was already established inhibited fibrosis progression, as assessed by lung histology, changes in body weights, extracellular matrix gene expression, and lung mechanics. These findings demonstrate that MEK inhibition prevents progression of established fibrosis in the TGF-α model, and provides proof of concept of targeting the MEK pathway in fibrotic lung disease.


Assuntos
Benzimidazóis/farmacologia , Receptores ErbB/metabolismo , Pulmão/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Fibrose Pulmonar/prevenção & controle , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Regulação da Expressão Gênica , Humanos , Pulmão/enzimologia , Pulmão/patologia , Pulmão/fisiopatologia , Camundongos , Camundongos Transgênicos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação , Fibrose Pulmonar/enzimologia , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , Fibrose Pulmonar/fisiopatologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 302(4): L380-9, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22160306

RESUMO

The proinflammatory stimulus of chorioamnionitis is commonly associated with preterm delivery. Women at risk of preterm delivery receive antenatal glucocorticoids to functionally mature the fetal lung. However, the effects of the combined exposures of chorioamnionitis and antenatal glucocorticoids on the fetus are poorly understood. Time-mated ewes with singleton fetuses received an intra-amniotic injection of lipopolysaccharide (LPS) either preceding or following maternal intramuscular betamethasone 7 or 14 days before delivery, and the fetuses were delivered at 120 days gestational age (GA) (term = 150 days GA). Gestation matched controls received intra-amniotic and maternal intramuscular saline. Compared with saline controls, intra-amniotic LPS increased inflammatory cells in the bronchoalveolar lavage and myeloperoxidase, Toll-like receptor 2 and 4 mRNA, PU.1, CD3, and Foxp3-positive cells in the fetal lung. LPS-induced lung maturation measured as increased airway surfactant and improved lung gas volumes. Intra-amniotic LPS-induced inflammation persisted until 14 days after exposure. Betamethasone treatment alone induced modest lung maturation but, when administered before intra-amniotic LPS, suppressed lung inflammation. Interestingly, betamethasone treatment after LPS did not counteract inflammation but enhanced lung maturation. We conclude that the order of exposures of intra-amniotic LPS or maternal betamethasone had large effects on fetal lung inflammation and maturation.


Assuntos
Betametasona/uso terapêutico , Corioamnionite/tratamento farmacológico , Desenvolvimento Fetal/efeitos dos fármacos , Glucocorticoides/uso terapêutico , Pulmão/embriologia , Âmnio , Animais , Líquido da Lavagem Broncoalveolar/citologia , Corioamnionite/etiologia , Corioamnionite/imunologia , Citocinas/genética , Citocinas/metabolismo , Feminino , Maturidade dos Órgãos Fetais/efeitos dos fármacos , Maturidade dos Órgãos Fetais/imunologia , Expressão Gênica , Inflamação/tratamento farmacológico , Inflamação/imunologia , Lipopolissacarídeos/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/imunologia , Masculino , Acetato de Medroxiprogesterona/uso terapêutico , Peroxidase/metabolismo , Fosfatidilcolinas/metabolismo , Gravidez , Nascimento Prematuro/imunologia , Nascimento Prematuro/prevenção & controle , Proteína C Associada a Surfactante Pulmonar/genética , Proteína C Associada a Surfactante Pulmonar/metabolismo , Proteína D Associada a Surfactante Pulmonar/genética , Proteína D Associada a Surfactante Pulmonar/metabolismo , Distribuição Aleatória , Ovinos , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
12.
Pediatr Radiol ; 42(11): 1347-56, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22735927

RESUMO

BACKGROUND: Transporting premature infants from a neonatal intensive care unit (NICU) to a radiology department for MRI has medical risks and logistical challenges. OBJECTIVE: To develop a small 1.5-T MRI system for neonatal imaging that can be easily installed in the NICU and to evaluate its performance using a sheep model of human prematurity. MATERIALS AND METHODS: A 1.5-T MRI system designed for orthopedic use was adapted for neonatal imaging. The system was used for MRI examinations of the brain, chest and abdomen in 12 premature lambs during the first hours of life. Spin-echo, fast spin-echo and gradient-echo MR images were evaluated by two pediatric radiologists. RESULTS: All animals remained physiologically stable throughout the imaging sessions. Animals were imaged at two or three time points. Seven brain MRI examinations were performed in seven different animals, 23 chest examinations in 12 animals and 19 abdominal examinations in 11 animals. At each anatomical location, high-quality images demonstrating good spatial resolution, signal-to-noise ratio and tissue contrast were routinely obtained within 30 min using standard clinical protocols. CONCLUSION: Our preliminary experience demonstrates the feasibility and potential of the neonatal MRI system to provide state-of-the-art MRI capabilities within the NICU. Advantages include overall reduced cost and site demands, lower acoustic noise, improved ease of access and reduced medical risk to the neonate.


Assuntos
Unidades de Terapia Intensiva Neonatal , Terapia Intensiva Neonatal/métodos , Imageamento por Ressonância Magnética/instrumentação , Animais , Desenho de Equipamento , Análise de Falha de Equipamento , Estudos de Viabilidade , Feminino , Humanos , Recém-Nascido , Imageamento por Ressonância Magnética/métodos , Masculino , Projetos Piloto , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Ovinos
13.
Am J Physiol Lung Cell Mol Physiol ; 300(3): L498-505, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21216976

RESUMO

In the fetus, leptin in the circulation increases at late gestation and likely influences fetal organ development. Increased surfactant by leptin was previously demonstrated in vitro using fetal lung explant. We hypothesized that leptin treatment given to fetal sheep and pregnant mice might increase surfactant synthesis in the fetal lung in vivo. At 122-124 days gestational age (term: 150 days), fetal sheep were injected with 5 mg of leptin or vehicle using ultrasound guidance. Three and a half days after injection, preterm lambs were delivered, and lung function was studied during 30-min ventilation, followed by pulmonary surfactant components analyses. Pregnant A/J mice were given 30 or 300 mg of leptin or vehicle by intraperitoneal injection according to five study protocols with different doses, number of treatments, and gestational ages to treat. Surfactant components were analyzed in fetal lung 24 h after the last maternal treatment. Leptin injection given to fetal sheep increased fetal body weight. Control and leptin-treated groups were similar in lung function (preterm newborn lamb), surfactant components pool sizes (lamb and fetal mice), and expression of genes related to surfactant synthesis in the lung (fetal mice). Likewise, saturated phosphatidylcholine and phospholipid were normal in mice lungs with absence of circulating leptin (ob/ob mice) at all ages. These studies coincided in findings that neither exogenously given leptin nor deficiency of leptin influenced fetal lung maturation or surfactant pool sizes in vivo. Furthermore, the key genes critically required for surfactant synthesis were not affected by leptin treatment.


Assuntos
Feto/efeitos dos fármacos , Feto/metabolismo , Leptina/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/embriologia , Proteínas Associadas a Surfactantes Pulmonares/biossíntese , Ovinos/embriologia , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Leptina/administração & dosagem , Pulmão/anatomia & histologia , Pulmão/metabolismo , Camundongos , Camundongos Obesos , Tamanho do Órgão/efeitos dos fármacos , Fosfatidilcolinas/metabolismo , Gravidez , Proteínas Associadas a Surfactantes Pulmonares/genética , Proteínas Associadas a Surfactantes Pulmonares/metabolismo
14.
Am J Pathol ; 176(2): 679-86, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20042669

RESUMO

Transforming growth factor-alpha (TGFalpha) is a ligand for the epidermal growth factor receptor (EGFR). EGFR activation is associated with fibroproliferative processes in human lung disease and animal models of pulmonary fibrosis. EGFR signaling activates several intracellular signaling pathways including phosphatidylinositol 3'-kinase (PI3K). We previously showed that induction of lung-specific TGFalpha expression in transgenic mice caused progressive pulmonary fibrosis over a 4-week period. The increase in levels of phosphorylated Akt, detected after 1 day of doxycycline-induced TGFalpha expression, was blocked by treatment with the PI3K inhibitor, PX-866. Daily administration of PX-866 during TGFalpha induction prevented increases in lung collagen and airway resistance as well as decreases in lung compliance. Treatment of mice with oral PX-866 4 weeks after the induction of TGFalpha prevented additional weight loss and further increases in total collagen, and attenuated changes in pulmonary mechanics. These data show that PI3K is activated in TGFalpha/EGFR-mediated pulmonary fibrosis and support further studies to determine the role of PI3K activation in human lung fibrotic disease, which could be amenable to targeted therapy.


Assuntos
Gonanos/farmacologia , Gonanos/uso terapêutico , Inibidores de Fosfoinositídeo-3 Quinase , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/prevenção & controle , Fator de Crescimento Transformador alfa , Administração Oral , Animais , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Gonanos/administração & dosagem , Camundongos , Camundongos Transgênicos , Proteína Oncogênica v-akt/metabolismo , Fosforilação/efeitos dos fármacos , Uteroglobina/genética
15.
Am J Respir Crit Care Med ; 181(10): 1098-105, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20133924

RESUMO

RATIONALE: Premature newborns frequently require manual ventilation for resuscitation during which lung injury occurs. Although surfactant protein (SP)-D regulates pulmonary inflammation, SP-D levels are low in the preterm lung. Commercial surfactants for treatment of respiratory distress syndrome do not contain SP-D. OBJECTIVES: To determine whether addition of recombinant human SP-D (rhSP-D) to commercial surfactant influences lung inflammation in ventilated premature newborn lambs. METHODS: Prematurely delivered lambs (130 d gestation age) were resuscitated with 100% O(2) and peak inspiratory pressure 40 cm H(2)O for 20 minutes and then treated with Survanta or Survanta containing rhSP-D. Ventilation was then changed to regulate tidal volume at 8 to 9 ml/kg. At 5 hours of age lambs were killed for sample collection. MEASUREMENTS AND MAIN RESULTS: Sequential blood gas and tidal volume were similar in lambs treated with or without rhSP-D, indicating that lung immaturity and ventilatory stress used to support premature lambs were comparable between the two groups. Ventilation caused pulmonary inflammation in lambs treated with surfactant alone. In contrast, surfactant containing rhSP-D decreased neutrophil numbers in bronchoalveolar lavage fluid and decreased neutrophil elastase activity in lung tissue. IL-8 mRNA and IL-8 protein were significantly decreased in the +rhSP-D group lamb lungs, to 20% of those in controls. The addition of rhSP-D also rendered Survanta more resistant to plasma protein inhibition of surfactant function. CONCLUSIONS: Treatment with rhSP-D-containing surfactant inhibited lung inflammation and enhanced the resistance of surfactant to inhibition, supporting its potential usefulness for prevention of lung injury in the preterm newborn.


Assuntos
Pneumonia/prevenção & controle , Proteína D Associada a Surfactante Pulmonar/farmacologia , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle , Animais , Modelos Animais de Doenças , Feminino , Humanos , Pulmão/fisiopatologia , Masculino , Pneumonia/etiologia , Pneumonia/fisiopatologia , Proteínas Recombinantes/farmacologia , Respiração Artificial/efeitos adversos , Ovinos
16.
Am J Respir Crit Care Med ; 182(1): 49-61, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20224064

RESUMO

RATIONALE: Granulocyte/macrophage colony-stimulating factor (GM-CSF) autoantibodies (GMAb) are strongly associated with idiopathic pulmonary alveolar proteinosis (PAP) and are believed to be important in its pathogenesis. However, levels of GMAb do not correlate with disease severity and GMAb are also present at low levels in healthy individuals. OBJECTIVES: Our primary objective was to determine whether human GMAb would reproduce PAP in healthy primates. A secondary objective was to determine the concentration of GMAb resulting in loss of GM-CSF signaling in vivo (i.e., critical threshold). METHODS: Nonhuman primates (Macaca fascicularis) were injected with highly purified, PAP patient-derived GMAb in dose-ranging (2.2-50 mg) single and multiple administration studies, and after blocking antihuman immunoglobulin immune responses, in chronic administration studies maintaining serum levels greater than 40 microg/ml for up to 11 months. MEASUREMENTS AND MAIN RESULTS: GMAb blocked GM-CSF signaling causing (1) a milky-appearing bronchoalveolar lavage fluid containing increased surfactant lipids and proteins; (2) enlarged, foamy, surfactant-filled alveolar macrophages with reduced PU.1 and PPARgamma mRNA, and reduced tumor necrosis factor-alpha secretion; (3) pulmonary leukocytosis; (4) increased serum surfactant protein-D; and (5) impaired neutrophil functions. GM-CSF signaling varied inversely with GMAb concentration below a critical threshold of 5 microg/ml, which was similar in lungs and blood and to the value observed in patients with PAP. CONCLUSIONS: GMAb reproduced the molecular, cellular, and histopathologic features of PAP in healthy primates, demonstrating that GMAb directly cause PAP. These results have implications for therapy of PAP and help define the therapeutic window for potential use of GMAb to treat other disorders.


Assuntos
Autoanticorpos/efeitos adversos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Macrófagos Alveolares/imunologia , Proteinose Alveolar Pulmonar/imunologia , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Macaca fascicularis , Macrófagos Alveolares/diagnóstico por imagem , Proteinose Alveolar Pulmonar/patologia , Ultrassonografia
17.
Am J Physiol Lung Cell Mol Physiol ; 298(5): L646-59, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20190032

RESUMO

ATP-binding cassette A3 (ABCA3) is a lipid transport protein required for synthesis and storage of pulmonary surfactant in type II cells in the alveoli. Abca3 was conditionally deleted in respiratory epithelial cells (Abca3(Δ/Δ)) in vivo. The majority of mice in which Abca3 was deleted in alveolar type II cells died shortly after birth from respiratory distress related to surfactant deficiency. Approximately 30% of the Abca3(Δ/Δ) mice survived after birth. Surviving Abca3(Δ/Δ) mice developed emphysema in the absence of significant pulmonary inflammation. Staining of lung tissue and mRNA isolated from alveolar type II cells demonstrated that ∼50% of alveolar type II cells lacked ABCA3. Phospholipid content and composition were altered in lung tissue, lamellar bodies, and bronchoalveolar lavage fluid from adult Abca3(Δ/Δ) mice. In adult Abca3(Δ/Δ) mice, cells lacking ABCA3 had decreased expression of mRNAs associated with lipid synthesis and transport. FOXA2 and CCAAT enhancer-binding protein-α, transcription factors known to regulate genes regulating lung lipid metabolism, were markedly decreased in cells lacking ABCA3. Deletion of Abca3 disrupted surfactant lipid synthesis in a cell-autonomous manner. Compensatory surfactant synthesis was initiated in ABCA3-sufficient type II cells, indicating that surfactant homeostasis is a highly regulated process that includes sensing and coregulation among alveolar type II cells.


Assuntos
Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Transportadores de Cassetes de Ligação de ATP/genética , Células Epiteliais Alveolares/metabolismo , Proteínas Associadas a Surfactantes Pulmonares/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Células Epiteliais Alveolares/ultraestrutura , Animais , Animais Recém-Nascidos , Sequência de Bases , Modelos Animais de Doenças , Feminino , Expressão Gênica , Homeostase , Humanos , Recém-Nascido , Metabolismo dos Lipídeos , Camundongos , Microscopia Eletrônica de Transmissão , Gravidez , Enfisema Pulmonar/etiologia , Enfisema Pulmonar/genética , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patologia , Proteínas Associadas a Surfactantes Pulmonares/deficiência , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia
18.
Respir Res ; 11: 151, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-21034485

RESUMO

BACKGROUND: Increased pro-inflammatory cytokines in tracheal aspirates correlate with the development of BPD in preterm infants. Ventilation of preterm lambs increases pro-inflammatory cytokines and causes lung inflammation. OBJECTIVE: We tested the hypothesis that selective inhibitors of pro-inflammatory signaling would decrease lung inflammation induced by ventilation in preterm newborn lambs. We also examined if the variability in injury response was explained by variations in the endogenous surfactant pool size. METHODS: Date-mated preterm lambs (n = 28) were operatively delivered and mechanically ventilated to cause lung injury (tidal volume escalation to 15 mL/kg by 15 min at age). The lambs then were ventilated with 8 mL/kg tidal volume for 1 h 45 min. Groups of animals randomly received specific inhibitors for IL-8, IL-1, or NF-κB. Unventilated lambs (n = 7) were the controls. Bronchoalveolar lavage fluid (BALF) and lung samples were used to quantify inflammation. Saturated phosphatidylcholine (Sat PC) was measured in BALF fluid and the data were stratified based on a level of 5 µmol/kg (~8 mg/kg surfactant). RESULTS: The inhibitors did not decrease the cytokine levels or inflammatory response. The inflammation increased as Sat PC pool size in BALF decreased. Ventilated lambs with a Sat PC level > 5 µmol/kg had significantly decreased markers of injury and lung inflammation compared with those lambs with < 5 µmol/kg. CONCLUSION: Lung injury caused by high tidal volumes at birth were decreased when endogenous surfactant pool sizes were larger. Attempts to decrease inflammation by blocking IL-8, IL-1 or NF-κB were unsuccessful.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/fisiologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/prevenção & controle , Surfactantes Pulmonares/antagonistas & inibidores , Respiração Artificial/efeitos adversos , Fatores Etários , Animais , Animais Recém-Nascidos , Feminino , Lesão Pulmonar/etiologia , Ácidos Nicotínicos/farmacologia , Gravidez , Surfactantes Pulmonares/metabolismo , Distribuição Aleatória , Ovinos
19.
Am J Respir Crit Care Med ; 179(10): 955-61, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19234101

RESUMO

RATIONALE: Chorioamnionitis frequently associates with preterm delivery and increased amniotic fluid IL-1, and causes fetal lung and systemic inflammation. However, chorioamnionitis is also associated with a paradoxical reduction in the incidence of surfactant deficiency-related respiratory distress syndrome in preterm infants. OBJECTIVES: To identify the role of IL-1 signaling in the mediation of pulmonary and systemic inflammation and lung maturation in a fetal sheep model of lipopolysaccharide (LPS) induced chorioamnionitis. METHODS: After confirming the efficacy of recombinant human IL-1 receptor antagonist (rhIL-1ra), fetal sheep were exposed to intraamniotic (IA) injections of Escherichia coli LPS with or without prior IA injections of rhIL-1ra. Preterm lambs were delivered at 82% of term gestation. MEASUREMENTS AND MAIN RESULTS: rhIL-1ra decreased IA LPS-induced lung inflammation assessed by decreased lung neutrophil and monocyte influx, inducible nitric oxide synthase expression, lung IL-6 and IL-1beta mRNA expression, and airway myeloperoxidase concentrations. rhIL-1ra inhibited IA LPS-induced fetal systemic inflammation assessed by decreased plasma IL-8, protein carbonyls, blood neutrophilia, and the expression of serum amyloid A3 mRNA in the liver. rhIL-1ra also partially blocked the lung maturational effects of IA LPS. Therefore blockade of IL-1 signaling in the amniotic compartment inhibited fetal lung and systemic inflammation and lung maturation in response to LPS-induced chorioamnionitis. CONCLUSIONS: IL-1 plays a central role in the pathogenesis of chorioamnionitis-induced fetal inflammatory responses.


Assuntos
Corioamnionite/imunologia , Interleucina-1alfa/imunologia , Interleucina-1beta/imunologia , Pulmão/embriologia , Animais , Líquido da Lavagem Broncoalveolar/química , Corioamnionite/metabolismo , Corioamnionite/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Feto/efeitos dos fármacos , Feto/imunologia , Humanos , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1alfa/antagonistas & inibidores , Interleucina-1alfa/metabolismo , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/metabolismo , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/antagonistas & inibidores , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/patologia , Gravidez , Receptores de Interleucina-1/antagonistas & inibidores , Receptores de Interleucina-1/imunologia , Receptores de Interleucina-1/metabolismo , Proteínas Recombinantes/farmacologia , Ovinos , Transdução de Sinais
20.
Am J Respir Cell Mol Biol ; 41(4): 433-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19202005

RESUMO

Alveolar capillaries are located in close proximity to the alveolar epithelium and beneath the surfactant film. We hypothesized that the shape of alveolar capillaries and accompanying oxygenation are influenced by surfactant surface tension in the alveolus. To prove our hypothesis, surfactant surface tension was regulated by conditional expression of surfactant protein (SP)-B in Sftpb(-/-) mice, thereby inhibiting surface tension-lowering properties of surfactant in vivo within 24 hours after depletion of Sftpb. Minimum surface tension of isolated surfactant was increased and oxygen saturation was significantly reduced after 2 days of SP-B deficiency in association with deformation of alveolar capillaries. Intravascularly injected 3.2-mum-diameter microbeads through jugular vein were retained within narrowed pulmonary capillaries after reduction of SP-B. Ultrastructure studies demonstrated that the capillary protrusion typical of the normal alveolar-capillary unit was reduced in size, consistent with altered pulmonary blood flow. Pulmonary hypertension and intrapulmonary shunting are commonly associated with surfactant deficiency and dysfunction in neonates and adults with respiratory distress syndromes. Increased surfactant surface tension caused by reduction in SP-B induced narrowing of alveolar capillaries and oxygen desaturation, demonstrating an important role of surface tension-lowering properties of surfactant in the regulation of pulmonary vascular perfusion.


Assuntos
Alvéolos Pulmonares/irrigação sanguínea , Troca Gasosa Pulmonar/fisiologia , Proteína B Associada a Surfactante Pulmonar/fisiologia , Insuficiência Respiratória/etiologia , Tensão Superficial , Animais , Capilares/ultraestrutura , Doxiciclina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipóxia/etiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microesferas , Oxigênio/sangue , Pressão Parcial , Alvéolos Pulmonares/química , Alvéolos Pulmonares/fisiologia , Circulação Pulmonar/fisiologia , Proteína B Associada a Surfactante Pulmonar/deficiência , Proteína B Associada a Surfactante Pulmonar/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transgenes , Vasoconstrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA