Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Clin Invest ; 129(7): 2904-2919, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31162135

ABSTRACT

Bronchopulmonary dysplasia (BPD) remains a major respiratory illness in extremely premature infants. The biological mechanisms leading to BPD are not fully understood, although an arrest in lung development has been implicated. The current study aimed to investigate the occurrence of autophagy in the developing mouse lung and its regulatory role in airway branching and terminal sacculi formation. We found 2 windows of epithelial autophagy activation in the developing mouse lung, both resulting from AMPK activation. Inhibition of AMPK-mediated autophagy led to reduced lung branching in vitro. Conditional deletion of beclin 1 (Becn1) in mouse lung epithelial cells (Becn1Epi-KO), either at early (E10.5) or late (E16.5) gestation, resulted in lethal respiratory distress at birth or shortly after. E10.5 Becn1Epi-KO lungs displayed reduced airway branching and sacculi formation accompanied by impaired vascularization, excessive epithelial cell death, reduced mesenchymal thinning of the interstitial walls, and delayed epithelial maturation. E16.5 Becn1Epi-KO lungs had reduced terminal air sac formation and vascularization and delayed distal epithelial differentiation, a pathology similar to that seen in infants with BPD. Taken together, our findings demonstrate that intrinsic autophagy is an important regulator of lung development and morphogenesis and may contribute to the BPD phenotype when impaired.


Subject(s)
Autophagic Cell Death , Bronchopulmonary Dysplasia/embryology , Lung/embryology , Organogenesis , Animals , Beclin-1/genetics , Beclin-1/metabolism , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/pathology , Lung/pathology , Mice , Mice, Knockout
2.
Paediatr Respir Rev ; 31: 82-88, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31103368

ABSTRACT

Birth prior to term interrupts the normal development of the respiratory system and consequently results in poor respiratory outcomes that persist throughout childhood. The mechanisms underpinning these poor respiratory outcomes are not well understood, but intrinsic abnormalities within the airway epithelium may be a contributing factor. Current evidence suggests that the airway epithelium is both structurally and functionally abnormal after preterm birth, with reports of epithelial thickening and goblet cell hyperplasia in addition to increased inflammation and apoptosis in the neonatal intensive care unit. However, studies focusing on the airway epithelium are limited and many questions remain unanswered; including whether abnormalities are a direct result of interrupted development, a consequence of exposure to inflammatory stimuli in the perinatal period or a combination of the two. In addition, the difficulty of accessing airway tissue has resulted in the majority of evidence being collected in the pre-surfactant era which may not reflect contemporary preterm birth. This review examines the consequences of preterm birth on the airway epithelium and explores the clinical relevance of currently available models whilst highlighting the need to develop a clinically relevant in vitro model to help further our understanding of the airway epithelium in preterm birth.


Subject(s)
Apoptosis , Bronchopulmonary Dysplasia/embryology , Inflammation , Premature Birth , Respiratory Mucosa/embryology , Bronchopulmonary Dysplasia/immunology , Bronchopulmonary Dysplasia/metabolism , Chorioamnionitis/immunology , Chorioamnionitis/metabolism , Female , Goblet Cells/pathology , Humans , Hyperplasia , Infant, Newborn , Infant, Premature , Infections/immunology , Infections/metabolism , Intensive Care Units, Neonatal , Lung Injury/etiology , Lung Injury/immunology , Lung Injury/metabolism , Oxygen Inhalation Therapy/adverse effects , Positive-Pressure Respiration/adverse effects , Pregnancy , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Resuscitation/adverse effects
3.
Am J Physiol Lung Cell Mol Physiol ; 315(3): L348-L359, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29722560

ABSTRACT

Intrauterine growth restriction (IUGR) in premature newborns increases the risk for bronchopulmonary dysplasia, a chronic lung disease characterized by disrupted pulmonary angiogenesis and alveolarization. We previously showed that experimental IUGR impairs angiogenesis; however, mechanisms that impair pulmonary artery endothelial cell (PAEC) function are uncertain. The NF-κB pathway promotes vascular growth in the developing mouse lung, and we hypothesized that IUGR disrupts NF-κB-regulated proangiogenic targets in fetal PAEC. PAECs were isolated from the lungs of control fetal sheep and sheep with experimental IUGR from an established model of chronic placental insufficiency. Microarray analysis identified suppression of NF-κB signaling and significant alterations in extracellular matrix (ECM) pathways in IUGR PAEC, including decreases in collagen 4α1 and laminin α4, components of the basement membrane and putative NF-κB targets. In comparison with controls, immunostaining of active NF-κB complexes, NF-κB-DNA binding, baseline expression of NF-κB subunits p65 and p50, and LPS-mediated inducible activation of NF-κB signaling were decreased in IUGR PAEC. Although pharmacological NF-κB inhibition did not affect angiogenic function in IUGR PAEC, angiogenic function of control PAEC was reduced to a similar degree as that observed in IUGR PAEC. These data identify reductions in endothelial NF-κB signaling as central to the disrupted angiogenesis observed in IUGR, likely by impairing both intrinsic PAEC angiogenic function and NF-κB-mediated regulation of ECM components necessary for vascular development. These data further suggest that strategies that preserve endothelial NF-κB activation may be useful in lung diseases marked by disrupted angiogenesis such as IUGR.


Subject(s)
Bronchopulmonary Dysplasia , Endothelial Cells , Fetal Growth Retardation , NF-kappa B p50 Subunit/metabolism , Pulmonary Artery , Signal Transduction , Transcription Factor RelA/metabolism , Animals , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/physiopathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Fetal Growth Retardation/physiopathology , Lipopolysaccharides/toxicity , Pregnancy , Pulmonary Artery/embryology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Sheep
4.
Am J Respir Crit Care Med ; 197(6): 776-787, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29268623

ABSTRACT

RATIONALE: Pregnancies complicated by antenatal stress, including preeclampsia (PE) and chorioamnionitis (CA), increase the risk for bronchopulmonary dysplasia (BPD) in preterm infants, but biologic mechanisms linking prenatal factors with BPD are uncertain. Levels of sFlt-1 (soluble fms-like tyrosine kinase 1), an endogenous antagonist to VEGF (vascular endothelial growth factor), are increased in amniotic fluid and maternal blood in PE and associated with CA. OBJECTIVES: Because impaired VEGF signaling has been implicated in the pathogenesis of BPD, we hypothesized that fetal exposure to sFlt-1 decreases lung growth and causes abnormal lung structure and pulmonary hypertension during infancy. METHODS: To test this hypothesis, we studied the effects of anti-sFlt-1 monoclonal antibody (mAb) treatment on lung growth in two established antenatal models of BPD that mimic PE and CA induced by intraamniotic (i.a.) injections of sFlt-1 or endotoxin, respectively. In experimental PE, mAb was administered by three different approaches, including antenatal treatment by either i.a. instillation or maternal uterine artery infusion, or by postnatal intraperitoneal injections. RESULTS: With each strategy, mAb therapy improved infant lung structure as assessed by radial alveolar count, vessel density, right ventricular hypertrophy, and lung function. As found in the PE model, the adverse lung effects of i.a. endotoxin were also reduced by antenatal or postnatal mAb therapy. CONCLUSIONS: We conclude that treatment with anti-sFlt-1 mAb preserves lung structure and function and prevents right ventricular hypertrophy in two rat models of BPD of antenatal stress and speculate that early mAb therapy may provide a novel strategy for the prevention of BPD.


Subject(s)
Bronchopulmonary Dysplasia/physiopathology , Endothelium, Vascular/growth & development , Lung/growth & development , Pulmonary Alveoli/growth & development , Vascular Endothelial Growth Factor Receptor-1/therapeutic use , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/embryology , Disease Models, Animal , Endothelium, Vascular/embryology , Female , Humans , Lung/embryology , Pregnancy , Pulmonary Alveoli/embryology , Rats , Rats, Sprague-Dawley
6.
Physiol Rep ; 4(17)2016 09.
Article in English | MEDLINE | ID: mdl-27597766

ABSTRACT

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.


Subject(s)
Bronchopulmonary Dysplasia/metabolism , Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , Fibroblasts/metabolism , Inflammation Mediators/antagonists & inhibitors , Interferon Regulatory Factors/metabolism , Lung/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Adult , Aged , Animals , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/pathology , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Proliferation/physiology , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/physiology , Fibroblasts/cytology , Fibroblasts/pathology , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Lung/cytology , Lung/embryology , Lung/pathology , Male , Mice , Middle Aged , Pulmonary Disease, Chronic Obstructive/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction
7.
J Immunol ; 196(8): 3411-20, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26951798

ABSTRACT

Inflammation in the developing preterm lung leads to disrupted airway morphogenesis and chronic lung disease in human neonates. However, the molecular mechanisms linking inflammation and the pathways controlling airway morphogenesis remain unclear. In this article, we show that IL-1ß released by activated fetal lung macrophages is the key inflammatory mediator that disrupts airway morphogenesis. In mouse lung explants, blocking IL-1ß expression, posttranslational processing, and signaling protected the formation of new airways from the inhibitory effects ofEscherichia coliLPS. Consistent with a critical role for IL-1ß, mice expressing a gain-of-functionNlrp3allele and subsequent overactive inflammasome activity displayed abnormal saccular-stage lung morphogenesis and died soon after birth. Although the early-stage fetal lung appeared capable of mounting an NF-κB-mediated immune response, airway formation became more sensitive to inflammation later in development. This period of susceptibility coincided with higher expression of multiple inflammasome components that could increase the ability to release bioactive IL-1ß. Macrophages fromNlrp3gain-of-function mice also expressed higher levels of more mature cell surface markers, additionally linking inflammasome activation with macrophage maturation. These data identify developmental expression of the inflammasome and IL-1ß release by fetal lung macrophages as key mechanisms and potential therapeutic targets for neonatal lung disease.


Subject(s)
Inflammasomes/immunology , Inflammation/immunology , Interleukin-1beta/immunology , Lung/embryology , Macrophages/immunology , Animals , Bronchopulmonary Dysplasia/embryology , Carrier Proteins/metabolism , Disease Models, Animal , Interleukin-1beta/biosynthesis , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction/immunology
8.
Am J Physiol Lung Cell Mol Physiol ; 309(9): L1018-26, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26342089

ABSTRACT

Vitamin D [vit D; 1,25-(OH)2D] treatment improves survival and lung alveolar and vascular growth in an experimental model of bronchopulmonary dysplasia (BPD) after antenatal exposure to endotoxin (ETX). However, little is known about lung-specific 1,25-(OH)2D3 regulation during development, especially regarding maturational changes in lung-specific expression of the vitamin D receptor (VDR), 1α-hydroxylase (1α-OHase), and CYP24A1 during late gestation and the effects of antenatal ETX exposure on 1,25-(OH)2D3 metabolism in the lung. We hypothesized that vit D regulatory proteins undergo maturation regulation in the late fetal and early neonatal lung and that prenatal exposure to ETX impairs lung growth partly through abnormal endogenous vit D metabolism. Normal fetal rat lungs were harvested between embryonic day 15 and postnatal day 14. Lung homogenates were assayed for VDR, 1α-OHase, and CYP24A1 protein contents by Western blot analysis. Fetal rats were injected on embryonic day 20 with intra-amniotic ETX, ETX + 1,25-(OH)2D3, or saline and delivered 2 days later. Pulmonary artery endothelial cells (PAECs) from fetal sheep were assessed for VDR, 1α-OHase, and CYP24A1 expression after treatment with 25-(OH)D3, 1,25-(OH)2D3, ETX, ETX + 25-(OH)D3, or ETX + 1,25-(OH)2D3. We found that lung VDR, 1α-OHase, and CYP2741 protein expression dramatically increase immediately before birth (P < 0.01 vs. early fetal values). Antenatal ETX increases CYP24A1 expression (P < 0.05) and decreases VDR and 1α-OHase expression at birth (P < 0.001), but these changes are prevented with concurrent vit D treatment (P < 0.001). ETX-induced reduction of fetal PAEC growth and tube formation and lung 1α-OHase expression are prevented by vit D treatment (P < 0.001). We conclude that lung VDR, 1α-OHase, and CYP24A1 protein content markedly increase before birth and that antenatal ETX disrupts lung vit D metabolism through downregulation of VDR and increased vit D catabolic enzyme expression, including changes in developing endothelium. We speculate that endogenous vitamin D metabolism modulates normal fetal lung development and that prenatal disruption of vit D signaling may contribute to impaired postnatal lung growth at least partly through altered angiogenic signaling.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/biosynthesis , Endothelial Cells/metabolism , Endotoxins/toxicity , Gene Expression Regulation, Developmental/drug effects , Lung/embryology , Receptors, Calcitriol/biosynthesis , Animals , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/pathology , Calcifediol/metabolism , Endothelial Cells/pathology , Lung/pathology , Pulmonary Artery/embryology , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Sheep , Vitamin D3 24-Hydroxylase/biosynthesis
9.
Am J Respir Cell Mol Biol ; 53(1): 60-73, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25387348

ABSTRACT

DNA methylation, a major epigenetic mechanism, may regulate coordinated expression of multiple genes at specific time points during alveolar septation in lung development. The objective of this study was to identify genes regulated by methylation during normal septation in mice and during disordered septation in bronchopulmonary dysplasia. In mice, newborn lungs (preseptation) and adult lungs (postseptation) were evaluated by microarray analysis of gene expression and immunoprecipitation of methylated DNA followed by sequencing (MeDIP-Seq). In humans, microarray gene expression data were integrated with genome-wide DNA methylation data from bronchopulmonary dysplasia versus preterm and term lung. Genes with reciprocal changes in expression and methylation, suggesting regulation by DNA methylation, were identified. In mice, 95 genes with inverse correlation between expression and methylation during normal septation were identified. In addition to genes known to be important in lung development (Wnt signaling, Angpt2, Sox9, etc.) and its extracellular matrix (Tnc, Eln, etc.), genes involved with immune and antioxidant defense (Stat4, Sod3, Prdx6, etc.) were also observed. In humans, 23 genes were differentially methylated with reciprocal changes in expression in bronchopulmonary dysplasia compared with preterm or term lung. Genes of interest included those involved with detoxifying enzymes (Gstm3) and transforming growth factor-ß signaling (bone morphogenetic protein 7 [Bmp7]). In terms of overlap, 20 genes and three pathways methylated during mouse lung development also demonstrated changes in methylation between preterm and term human lung. Changes in methylation correspond to altered expression of a number of genes associated with lung development, suggesting that DNA methylation of these genes may regulate normal and abnormal alveolar septation.


Subject(s)
Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/metabolism , DNA Methylation , Gene Expression Regulation, Developmental , Pulmonary Alveoli/embryology , Pulmonary Alveoli/metabolism , Adult , Animals , Bronchopulmonary Dysplasia/pathology , Epigenesis, Genetic , Female , Humans , Male , Mice , Pulmonary Alveoli/pathology
10.
Am J Physiol Lung Cell Mol Physiol ; 308(4): L391-402, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25480331

ABSTRACT

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.


Subject(s)
Apoptosis/physiology , Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Fetus/embryology , Fibroblasts/metabolism , Interferon Regulatory Factors/metabolism , Lung/embryology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Cell Adhesion Molecules/genetics , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Fetus/cytology , Fibroblasts/cytology , HEK293 Cells , Humans , Interferon Regulatory Factors/genetics , Lung/cytology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Respiratory Mucosa/cytology , Respiratory Mucosa/embryology , Signal Transduction/physiology
11.
Free Radic Biol Med ; 75: 167-77, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25106706

ABSTRACT

The most common form of newborn chronic lung disease, bronchopulmonary dysplasia (BPD), is thought to be caused by oxidative disruption of lung morphogenesis, which results in decreased pulmonary vasculature and alveolar simplification. Although cellular redox status is known to regulate cellular proliferation and differentiation, redox-sensitive pathways associated with these processes in developing pulmonary epithelium are unknown. Redox-sensitive pathways are commonly regulated by cysteine thiol modifications. Therefore two thiol oxidoreductase systems, thioredoxin and glutathione, were chosen to elucidate the roles of these pathways on cell death. Studies herein indicate that thiol oxidation contributes to cell death through impaired activity of glutathione-dependent and thioredoxin (Trx) systems and altered signaling through redox-sensitive pathways. Free thiol content decreased by 71% with hyperoxic (95% oxygen) exposure. Increased cell death was observed during oxygen exposure when either the Trx or the glutathione-dependent system was pharmacologically inhibited with aurothioglucose (ATG) or buthionine sulfoximine, respectively. However, inhibition of the Trx system yielded the smallest decrease in free thiol content (1.44% with ATG treatment vs 21.33% with BSO treatment). Although Trx1 protein levels were unchanged, Trx1 function was impaired during hyperoxic treatment as indicated by progressive cysteine oxidation. Overexpression of Trx1 in H1299 cells utilizing an inducible construct increased cell survival during hyperoxia, whereas siRNA knockdown of Trx1 during oxygen treatment reduced cell viability. Overall, this indicated that a comparatively small pool of proteins relies on Trx redox functions to mediate cell survival in hyperoxia, and the protective functions of Trx1 are progressively lost by its oxidative inhibition. To further elucidate the role of Trx1, potential Trx1 redox protein-protein interactions mediating cytoprotection and cell survival pathways were determined by utilizing a substrate trap (mass action trapping) proteomics approach. With this method, known Trx1 targets were detected, including peroxiredoxin-1as well as novel targets, including two HSP90 isoforms (HSP90AA1 and HSP90AB1). Reactive cysteines within the structure of HSP90 are known to modulate its ATPase-dependent chaperone activity through disulfide formation and S-nitrosylation. Whereas HSP90 expression is unchanged at the protein level during hyperoxic exposure, siRNA knockdown significantly increased hyperoxic cell death by 2.5-fold, indicating cellular dependence on HSP90 chaperone functions in response to hyperoxic exposure. These data support the hypothesis that hyperoxic impairment of Trx1 has a negative impact on HSP90-oxidative responses critical to cell survival, with potential implications for pathways implicated in lung development and the pathogenesis of BPD.


Subject(s)
Glutathione/metabolism , Hyperoxia/metabolism , Peroxiredoxins/metabolism , Thioredoxins/metabolism , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/pathology , Buthionine Sulfoximine/pharmacology , Cell Line, Tumor , Cell Survival , Glutathione/biosynthesis , HSP90 Heat-Shock Proteins/metabolism , Humans , Lung/blood supply , Lung/embryology , Oxidation-Reduction , Oxygen/pharmacology , RNA Interference , RNA, Small Interfering , Signal Transduction , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/biosynthesis , Thioredoxins/genetics
12.
Lik Sprava ; (9-10): 113-8, 2014.
Article in Ukrainian | MEDLINE | ID: mdl-26492786

ABSTRACT

The analysis of history in 116 premature infants, of which 74 children formed a new form of bronchopulmionary dysplasia. The analysis revealed that predictors of the formation of new forms of bronchopulmonary dysplasia are the pregnant woman, accompanied by hypoxia (jeopardy throughout pregnancy, fetoplacental insufficiency, hypotension, pregnancy, anemia) or infectious inflammation (presence of IgG to a pregnant ureaplasma, chorioamnionitis, polyhydramnios).


Subject(s)
Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/etiology , Pregnancy Complications , Bronchopulmonary Dysplasia/diagnosis , Bronchopulmonary Dysplasia/epidemiology , Child, Preschool , Chorioamnionitis/diagnosis , Chorioamnionitis/epidemiology , Female , Humans , Infant , Infant, Premature , Placental Insufficiency/diagnosis , Placental Insufficiency/epidemiology , Pregnancy , Pregnancy Complications/diagnosis , Pregnancy Complications/epidemiology , Pregnancy Complications, Cardiovascular/diagnosis , Pregnancy Complications, Cardiovascular/epidemiology , Pregnancy Complications, Infectious/diagnosis , Pregnancy Complications, Infectious/epidemiology , Prognosis
13.
Arch Dis Child Fetal Neonatal Ed ; 97(1): F8-F17, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21697236

ABSTRACT

OBJECTIVE: To conduct a systematic review of the association between chorioamnionitis (CA) and bronchopulmonary dysplasia (BPD) in preterm infants. METHODS: The authors searched Medline, Embase, CINAHL, Science Citation Index and PubMed, reviewed reference lists and contacted the primary authors of relevant studies. Studies were included if they had a comparison group, examined preterm or low birthweight infants, and provided primary data. Two reviewers independently screened the search results, applied inclusion criteria and assessed methodological quality. One reviewer extracted data and a second reviewer checked data extraction. Studies were combined with an OR using a random effects model. Meta-regression was used to explore potential confounders. RESULTS: 3587 studies were identified; 59 studies (15 295 patients) were included. The pooled unadjusted OR showed that CA was significantly associated with BPD (OR 1.89, 95% CI 1.56 to 2.3). Heterogeneity was substantial (I(2)=66.2%) and may be partially explained by the type of CA. Infants exposed to CA were significantly younger and lighter at birth. The pooled adjusted OR was 1.58 (95% CI 1.11 to 2.24); heterogeneity was substantial (I(2)=65.1%) which may be due to different variables being controlled in each study. There was strong evidence of publication bias which suggests potential overestimation of the measure of association between CA and BPD. CONCLUSIONS: Unadjusted and adjusted analyses showed that CA was significantly associated with BPD; however, the adjusted results were more conservative in the magnitude of association. The authors found strong evidence of publication bias. Despite a large body of evidence, CA cannot be definitively considered a risk factor for BPD.


Subject(s)
Bronchopulmonary Dysplasia/embryology , Chorioamnionitis/epidemiology , Birth Weight , Bronchopulmonary Dysplasia/epidemiology , Confounding Factors, Epidemiologic , Female , Gestational Age , Humans , Infant, Newborn , Infant, Premature , Pregnancy , Risk Factors
15.
Am J Physiol Lung Cell Mol Physiol ; 298(5): L637-45, 2010 May.
Article in English | MEDLINE | ID: mdl-20172951

ABSTRACT

The fetal lung vasculature forms in tandem with developing airways. Whereas saccular airway morphogenesis is arrested in bronchopulmonary dysplasia (BPD), the potential vascular phenotype in BPD at this stage of development is less well-understood. As inflammation increases the risk of BPD and induces arrest of saccular airway morphogenesis, we tested the effects of Escherichia coli LPS on fetal mouse lung vascular development. Injecting LPS into the amniotic fluid of Tie2-lacZ endothelial reporter mice at embryonic day 15 stimulated angiogenesis in the saccular stage fetal lung mesenchyme. LPS also increased the number of endothelial cells in saccular stage fetal mouse lung explants. Inflammation appeared to directly promote vascular development, as LPS stimulated pulmonary microvascular endothelial cell angiogenesis, cell migration, and proliferation in vitro. Whereas LPS did not increase expression of VEGF, angiopoietin-1 (Ang-1), Tie2, fetal liver kinase-1 (Flk-1), fms-like tyrosine kinase-1 (Flt-1), PDGFA, PDGFB, heparin-binding EGF-like growth factor (HB-EGF), or connective tissue growth factor (CTGF), LPS did stimulate the production of the angiogenic CC chemokines macrophage inflammatory protein-1α (MIP-1α) and monocyte chemoattractant protein-1 (MCP-1). Both MIP-1α and MCP-1 increased angiogenesis in fetal mouse lung explants. In addition, inhibitory antibodies against MIP-1α and MCP-1 blocked the effects of LPS on fetal lung vascular development, suggesting these chemokines are downstream mediators of LPS-induced angiogenesis. We speculate that an inflammation-mediated surge in angiogenesis could lead to formation of aberrant alveolar capillaries in the lungs of patients developing BPD.


Subject(s)
Chemokines, CC/metabolism , Chorioamnionitis/immunology , Lung/blood supply , Lung/embryology , Neovascularization, Physiologic , Animals , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/immunology , Case-Control Studies , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/pharmacology , Chemokine CCL3/antagonists & inhibitors , Chemokine CCL3/pharmacology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/physiology , Female , Humans , Immunity, Innate , Infant , Infant, Newborn , Lipopolysaccharides/pharmacology , Lung/abnormalities , Lung/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/immunology , Pregnancy
17.
J Histochem Cytochem ; 52(9): 1231-40, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15314090

ABSTRACT

We studied cell-specific protein expression of all the major antioxidant enzymes (AOEs) and related proteins, such as copper-zinc superoxide dismutase (CuZnSOD), manganese SOD (MnSOD), extracellular SOD (ECSOD), catalase, the heavy and light chains of gamma-glutamylcysteine synthetase (gamma-GCS-l and gamma-GCS-h, also called glutamate cysteine ligase), the rate-limiting enzyme in glutathione synthesis, hemeoxygenase-1 (HO-1), and thioredoxin (Trx), in developing human lung, respiratory distress syndrome, and bronchopulmonary dysplasia by immunohistochemistry. Generally, after 17 weeks of gestational age, MnSOD was predominantly expressed in bronchial epithelium, alveolar epithelium, and macrophages, CuZnSOD was expressed in bronchial epithelium, ECSOD was expressed in bronchial epithelium, vascular endothelium, and the extracellular matrix, catalase was expressed in bronchial epithelium and alveolar macrophages, gamma-GCS-h was expressed in bronchial epithelium and endothelium, and gamma-GCS-l was expressed in bronchial epithelium. Trx was restricted to bronchial epithelium and to a lesser extent to alveolar macrophages, and HO-1 found in alveolar macrophages. Basically, the expression of these enzymes was similar in normal and diseased lung. It can be concluded that various AOEs and related proteins differ in their distribution and expression in lung before term, but generally it seems that infants are better adapted to high oxygen tension than might be expected.


Subject(s)
Antioxidants/metabolism , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/enzymology , Lung/embryology , Lung/enzymology , Respiratory Distress Syndrome, Newborn/embryology , Respiratory Distress Syndrome, Newborn/enzymology , Bronchopulmonary Dysplasia/pathology , Catalase/metabolism , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , Humans , Infant , Infant, Newborn , Lung/pathology , Male , Membrane Proteins , Pregnancy , Pregnancy Outcome , Respiratory Distress Syndrome, Newborn/pathology , Superoxide Dismutase/metabolism , Thioredoxins/metabolism
18.
Pediatrics ; 111(4 Pt 1): 766-76, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12671110

ABSTRACT

OBJECTIVE: Infants with chronic lung disease (CLD) have an arrest of primary and secondary septation. We hypothesized that this may be related to damage or abnormal development of lung collagen secondary to positive pressure ventilation. Our aims were to identify the sites and quantity of collagen in control infants 22 to 72 weeks' postconceptional age and compare these with infants with various degrees of severity of CLD. METHODS: The controls were 22 to 42 weeks' gestation (n = 30), received minimal ventilator care, and died within 48 hours of birth, plus 5 term infants who died at 43 to 72 weeks' postconceptional age from nonpulmonary causes. Infants who were 23 to 30 weeks' gestation, were at risk for CLD, and lived 5 to 94 days (n = 33) were separated into 3 groups on the basis of respiratory score (score group; the integrated area under the curve of the average daily fraction of inspired oxygen x mean airway pressure [cm H2O] over the number of days lived). The score groups, <20, 20 to 69, and 70 to 500, related clinically to mild to moderate and severe lung disease. The lungs were tracheally perfused and formalin fixed. Total lung volume was determined by water displacement. The paraffin-embedded lung blocks were sectioned 5 micro m thick, stained with Gomori's reticulum stain, hematoxylin and eosin, and immunohistochemically for collagen IV. The parenchyma was point-counted, and the volume density of collagen was measured. The chord diameter of the peripheral airway saccules and alveoli was measured. Descriptive collagen data were assessed on en face 40- micro m-thick sections through the alveolar or saccular walls on all infants at risk for CLD and in selected controls. RESULTS: In the controls, the volume density of collagen decreased from a maximum of 9% at 22 weeks to 5% at term and 72 weeks. With Scores < or =69, the fraction of collagen was similar to controls, but in infants with scores 70 to 500, it was increased relative to controls. However, when collagen was expressed as the volume density of interstitial tissue, ie, excluding parenchymal air space, it increased from a low of 5% at 22 weeks to 25% at 72 weeks. In infants with scores 70 to 500, 79% of infants had collagens greater than controls. Saccular and alveolar diameter increased from 40 micro m at 23 weeks to 100 microm at 72 weeks. Most infants with severe CLD (scores > or =70) had diameters more than twice that of controls at the same age. The total lung parenchymal collagen had a similar pattern as the volume density of collagen in interstitial tissue, increasing from 0.4 cm3 at 23 weeks to 9.7 cm3 at 72 weeks in the controls. Eighty-five percent of infants with scores 70 to 500 had total parenchymal collagen greater than the 95% confidence interval of the controls. With en face sections, a fine collagen mesh was seen at 23 weeks, which progressively increased in fiber size and quantity until 72 weeks. With severe CLD, the secondary collagen fibers in the saccular wall were thickened, tortuous, and disorganized relative to same-aged controls. Under 30 weeks, in the controls, the interstitium contained a wide, delicate network of interconnected collagen fibers. After positive pressure ventilation, some saccules markedly increased their diameter, which compressed and obliterated the interstitial network. In contrast with severe CLD, the interstitium was wide, with coarse wavy collagen fibers. CONCLUSIONS: Parenchymal collagen increases throughout development. Before 30 weeks, there is a delicate complex interstitial collagen network, which may be important for primary septation and subsequent normal development. Positive pressure ventilation, if excessive, and depending on lung maturity and disease state, over a short time can severely compress the interstitium and damage this collagen network and prevent normal primary septation and arrest or distort future lung development. With severe CLD, distal air space diameter increases. There is a failure of primary and secondary septation, arrested lung development and remodeling, with thickened cnt and remodeling, with thickened collagenous saccular walls, and a wide interstitium with increased quantity and size of collagen fibers that can affect the mechanics of ventilation. We conclude that normal lung development is dependent on a normal interstitium and, perhaps, collagen architecture and that origins of CLD begin early in the course of positive pressure ventilation.


Subject(s)
Collagen/metabolism , Lung Diseases/embryology , Lung Diseases/metabolism , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/mortality , Child , Child, Preschool , Chronic Disease , Collagen/chemistry , Collagen/immunology , Collagen Type IV/immunology , Collagen Type IV/metabolism , Humans , Immunohistochemistry , Infant , Infant, Newborn , Infant, Premature , Lung/abnormalities , Lung/embryology , Lung/metabolism , Lung/pathology , Lung Diseases/mortality , Lung Diseases/pathology , Lung Volume Measurements/methods , Positive-Pressure Respiration/adverse effects , Positive-Pressure Respiration/methods , Respiratory Distress Syndrome, Newborn/therapy
19.
J Anat ; 201(4): 325-34, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12430957

ABSTRACT

In the adult lung the pulmonary arteries run alongside the airways and the pulmonary veins show a similar branching pattern to the arteries, though separated from them. During early fetal development the airways act as a template for pulmonary blood vessel development in that the vessels form by vasculogenesis around the branching airways. In later lung development the capillary bed is essential for alveolar formation. This paper reviews evidence for the interaction of the airways and blood vessels in both normal and abnormal lung development.


Subject(s)
Lung/embryology , Mammals/embryology , Pulmonary Artery/embryology , Pulmonary Veins/embryology , Animals , Bronchial Arteries/embryology , Bronchopulmonary Dysplasia/embryology , Gestational Age , Humans , Infant, Newborn , Infant, Premature , Lung/abnormalities , Organogenesis/physiology
20.
Am J Respir Crit Care Med ; 165(4): 463-9, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11850337

ABSTRACT

Intratracheal endotoxin causes acute inflammation in the adult lung, and injurious styles of mechanical ventilation can result in systemic inflammation derived from the lungs. We asked how ventilated premature and near-term lungs responded to intratracheal endotoxin and if systemic inflammation occurred. Lambs delivered at 130 d gestational age (GA) were treated with surfactant or surfactant plus endotoxin (0.1 mg/kg or 10 mg/kg) (Escherichia coli, serotype O55:B5) and were ventilated for 6 h. Both endotoxin doses resulted in impaired gas exchange and systemic inflammation in the preterm lambs. Lambs at 141 d GA (term 146 d) were given either 10 mg/kg intratracheal endotoxin, 10 mg/kg endotoxin plus high tidal volume ventilation for the first 30 min of life, or 5 microg/kg endotoxin given intravenously. Endotoxin alone (10 mg/kg) caused lung inflammation but no systemic effects after 6 h of ventilation. Lambs given 10 mg/kg endotoxin plus high tidal volume ventilation or 5 microg/kg endotoxin intravenously had decreased gas exchange and systemic inflammation. Endotoxin was detected in the plasma of lambs at 130 d GA but not at 141 d GA. Inflammation in the lungs was more severe in preterm animals. Mechanical ventilation of the endotoxin-exposed preterm lung resulted in systemic effects at a low endotoxin dose and without high tidal volume ventilation.


Subject(s)
Bronchopulmonary Dysplasia/immunology , Cytokines/blood , Neutrophils/metabolism , Systemic Inflammatory Response Syndrome/immunology , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/pathology , Endotoxins/pharmacology , Gestational Age , Humans , Infant, Newborn , Lung/drug effects , Lung/embryology , Lung/immunology , Random Allocation , Respiration, Artificial/adverse effects , Sheep , Systemic Inflammatory Response Syndrome/embryology , Systemic Inflammatory Response Syndrome/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...