Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Dev Dyn ; 244(7): 827-38, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25904058

ABSTRACT

BACKGROUND: Lung maturation can be disrupted through pro-inflammatory processes including intra-uterine amniotic infection, mechanical ventilation, or oxidative stress. Lincr, originally identified as a gene induced in the lung by lipopolysaccharide (LPS), is also expressed in the developing lung. The Lung-inducible Neuralized-related C3HC4 RING domain (LINCR) protein is structurally related to Drosophila Neuralized, a regulator of the developmentally important Notch signaling pathway. LINCR is expressed in alveolar epithelial type II cells in the mature lung, and its expression is markedly increased by LPS and inflammatory cytokines. To test the hypothesis that targeted overexpression of LINCR in lung epithelium would interfere with normal lung development, we generated double transgenic mice that conditionally overexpress LINCR in lung epithelium under the control of doxycycline. RESULTS: Single transgenic controls and double transgenic mice not treated with doxycycline were unaffected, but double transgenic mice exposed to doxycycline starting at embryonic day 6 developed markedly hypoplastic lungs with decreased numbers of alveoli and large cysts lined with a proximalized and poorly differentiated epithelium expressing Hairy/Enhancer of Split 1, an effector of Notch signaling. The phenotype was similar to that caused by overexpression of activated Notch1 in lung epithelium. CONCLUSIONS: LINCR may exert its effects on distal lung development in this model through activation of the Notch signaling pathway.


Subject(s)
Gene Expression Regulation , Lung Diseases/embryology , Lung/embryology , Receptor, Notch1/biosynthesis , Respiratory Mucosa/embryology , Ubiquitin-Protein Ligases , Animals , Lung/pathology , Lung Diseases/genetics , Lung Diseases/pathology , Mice , Mice, Transgenic , Receptor, Notch1/genetics , Respiratory Mucosa/pathology , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
2.
Exp Lung Res ; 41(2): 103-14, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25517107

ABSTRACT

PURPOSE/AIM: The alveolar epithelium participates in host defense through inflammatory pathways that activate NF-κB. Lung infections involving endotoxins trigger acute respiratory distress syndrome (ARDS) in adult and pediatric patients. The purpose of this study was to test the hypothesis that overexpression of NF-κB would worsen and conditional deletion of NF-κB signaling would improve endotoxin-induced lung inflammation using transgenic mouse models. MATERIALS AND METHODS: Two previously described transgenic mouse models were used in which overexpression of the RelA/p65 subunit of NF-κB was targeted to the lung epithelium using an SPC promoter (SPC-RelA) and conditional deletion of the IKKƟ molecule involved in NF-κB signaling was targeted to the lung epithelium using Nkx2.1(Cre) (Nkx2.1(Cre);IKKƟ(F/F)). Adult transgenic and control mice were injected with intratracheal lipopolysaccharide (LPS) or saline followed by lung harvest at 48 h. Collected tissue included whole lungs from transgenic and control mice which was processed for analysis of BAL, lung histology, chemokine expression, and markers of cell apoptosis as well as collection of freshly isolated AECII cells from wild type mice for additional chemokine and apoptotic marker analysis. RESULTS: SPC-RelA mice showed significant increases in lung inflammation and injury following LPS injection with increased neutrophil recruitment as compared to wild type and saline treated controls. In contrast, Nkx2.1(Cre); IKKƟ(F/F) mice showed markedly decreased lung inflammation and injury with decreased neutrophil recruitment as compared to controls. In both models, lung inflammation was associated with increased cell apoptosis and these findings were confirmed in freshly isolated AECII cells in wild type mice following LPS injection. CONCLUSIONS: Overexpression of NF-κB targeted to the lung epithelium worsened lung inflammation and injury in response to LPS exposure while conditional deletion of NF-κB signaling reduced lung inflammation. Lung inflammation and injury were associated with increased cell apoptosis.


Subject(s)
Endotoxins/adverse effects , NF-kappa B/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pulmonary Alveoli/metabolism , Signal Transduction/physiology , Animals , Apoptosis/physiology , Chemokines/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Alveoli/pathology , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology
3.
Cell Mol Life Sci ; 71(18): 3439-63, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24676717

ABSTRACT

The dynamic nature of chromatin and its myriad modifications play a crucial role in gene regulation (expression and repression) during development, cellular survival, homeostasis, ageing, and apoptosis/death. Histone 3 lysine 4 methylation (H3K4 methylation) catalyzed by H3K4 specific histone methyltransferases is one of the more critical chromatin modifications that is generally associated with gene activation. Additionally, the deposition of H3 variant(s) in conjunction with H3K4 methylation generates an intricately reliable epigenetic regulatory circuit that guides transcriptional activity in normal development and homeostasis. Consequently, alterations in this epigenetic circuit may trigger disease development. The mechanistic relationship between H3 variant deposition and H3K4 methylation during normal development has remained foggy. However, recent investigations in the field of chromatin dynamics in various model organisms, tumors, cancer tissues, and cell lines cultured without and with therapeutic agents, as well as from model reconstituted chromatins reveal that there may be different subsets of chromatin assemblage with specific patterns of histone replacement executing similar functions. In this light, we attempt to explain the intricate control system that maintains chromatin structure and dynamics during normal development as well as during tumor development and cancer progression in this review. Our focus is to highlight the contribution of H3K4 methylation-histone variant crosstalk in regulating chromatin architecture and subsequently its function.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Gene Expression Regulation , Histones/metabolism , Models, Genetic , Neoplasms/genetics , Amino Acid Sequence , Chromatin/ultrastructure , Epigenomics , Genetic Variation , Genome , Humans , Methylation , Molecular Sequence Data , Sequence Alignment
4.
Pediatr Res ; 75(3): 395-402, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24321990

ABSTRACT

BACKGROUND: Caffeine is a nonspecific adenosine receptor antagonist used in premature neonates to treat apnea of prematurity. While its use may reduce the incidence of bronchopulmonary dysplasia (BPD), the precise mechanisms remain unknown. Evidence of increased adenosine levels are noted in chronic lung diseases including tracheal aspirates of infants with BPD. Utilizing a well-characterized newborn mouse model of alveolar hypoplasia, we hypothesized that hyperoxia-induced alveolar inflammation and hypoplasia is associated with alterations in the adenosine signaling pathway. METHODS: Newborn murine pups were exposed to a 14-d period of hyperoxia and daily caffeine administration followed by a 14-d recovery period in room air. Lungs were collected at both time points for bronchoalveolar lavage (BAL) analysis as well as histopathology and mRNA and protein expression. RESULTS: Caffeine treatment increased inflammation and worsened alveolar hypoplasia in hyperoxia-exposed newborn mice. These changes were associated with decreased alveolar type II (ATII) cell numbers, increased cell apoptosis, and decreased expression of A2A receptors. Following discontinuation of caffeine and hyperoxia, lung histology returned to baseline levels comparable to hyperoxia exposure alone. CONCLUSION: Results of this study suggest a potentially adverse role of caffeine on alveolar development in a murine model of hyperoxia-induced alveolar hypoplasia.


Subject(s)
Apoptosis/drug effects , Caffeine/pharmacology , Pulmonary Alveoli/drug effects , Purinergic P1 Receptor Antagonists/pharmacology , Signal Transduction/physiology , Adenosine/metabolism , Animals , Animals, Newborn , Bronchoalveolar Lavage , Caffeine/administration & dosage , In Situ Nick-End Labeling , Mice , Pulmonary Alveoli/cytology , Pulmonary Alveoli/physiopathology , Purinergic P1 Receptor Antagonists/administration & dosage , Receptor, Adenosine A2A/metabolism , Signal Transduction/drug effects
5.
Am J Respir Cell Mol Biol ; 48(2): 179-87, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23087051

ABSTRACT

Infants born with intrauterine growth retardation (IUGR) are at increased risk of adverse pulmonary outcomes at birth, including meconium aspiration and persistent pulmonary hypertension. Preterm infants with IUGR are at especially high risk of developing bronchopulmonary dysplasia (BPD), a disease hallmarked by alveolar hypoplasia. Although vitamin A supplementation has been shown to decrease the incidence of BPD or death in preterm very low birth weight infants, its potential to reduce BPD or death in preterm infants with IUGR remains unknown. We used a well-characterized rat model of caloric restriction to mimic IUGR and determine the impact of IUGR on lung development. We hypothesized that retinoic acid treatment would preserve alveolar formation through increases in key signaling molecules of the retinoic acid signaling pathway. Our results showed that alveolar hypoplasia caused by caloric restriction can be reversed with refeeding, and that retinoic acid prevents the alveolar hypoplasia coincident with the increased expression of elastin and retinoic acid receptor-α and decreased transforming growth factor-Ɵ activity in developing rat lungs. These findings suggest that alveolar hypoplasia attributable to caloric restriction is reversible, and raises the possibility that retinoic acid therapy may prove a useful strategy to prevent adverse pulmonary sequelae such as BPD in preterm infants with IUGR.


Subject(s)
Caloric Restriction , Hyperplasia/prevention & control , Lung/embryology , Maternal Exposure , Pulmonary Alveoli/drug effects , Tretinoin/pharmacology , Animals , Elastin/metabolism , Female , Pregnancy , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Rats , Rats, Sprague-Dawley , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Signal Transduction , Transforming Growth Factor beta/metabolism
6.
Am J Perinatol ; 30(9): 771-80, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23329565

ABSTRACT

OBJECTIVE: Preterm infants with intrauterine growth restriction are at increased risk of respiratory distress syndrome and bronchopulmonary dysplasia (BPD). A randomized clinical trial by the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Neonatal Research Network demonstrated that vitamin A supplementation in extremely low-birth-weight (ELBW) preterm infants requiring early respiratory support decreased the risk of developing BPD. STUDY DESIGN: A subgroup analysis of small-for-gestational-age (SGA) infants from the original NICHD trial was performed to test the hypothesis that in infants requiring early respiratory support, vitamin A supplementation decreases the relative risk of BPD or death in premature SGA infants to a greater extent than in gestational age-equivalent vitamin A-treated appropriate-for-gestational-age (AGA) infants. RESULTS: Although vitamin A supplementation significantly increased serum retinol concentrations in AGA ELBW infants (median [5th percentile, 95th percentile]: 16.3 [-7.0, 68.8] versus 2.4 [-13.9, 55.1]; p < 0.001), no increases were noted in SGA ELBW infants. CONCLUSIONS: Given the limited power of this analysis due to a low number of SGA infants, these data did not provide evidence to support the hypothesis that vitamin A supplementation in preterm SGA infants requiring early respiratory support decreases the relative risk of BPD or death as compared with preterm AGA infants.


Subject(s)
Fetal Growth Retardation/drug therapy , Infant, Extremely Low Birth Weight , Infant, Small for Gestational Age , Premature Birth/drug therapy , Vitamin A/therapeutic use , Vitamins/therapeutic use , Bronchopulmonary Dysplasia/epidemiology , Bronchopulmonary Dysplasia/prevention & control , Female , Fetal Growth Retardation/mortality , Fetal Growth Retardation/therapy , Humans , Infant, Extremely Low Birth Weight/blood , Infant, Newborn , Infant, Small for Gestational Age/blood , Male , Premature Birth/mortality , Premature Birth/therapy , Respiration, Artificial , Vitamin A/blood
7.
Am J Respir Cell Mol Biol ; 44(6): 804-12, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20693404

ABSTRACT

Alveolar formation is hallmarked by the transition of distal lung saccules into gas exchange units through the emergence of secondary crests and an exponential increase in surface area. Several cell types are involved in this complex process, including families of epithelial cells that differentiate into alveolar type I and II cells. Subsets of cells expressing Clara cell secretory protein (CCSP) have been identified in both lung and bone marrow compartments, and are described as a progenitor/stem cell pool involved in airway regeneration and alveolar homeostasis. Whether these cells also participate in alveolar formation during postnatal development remains unknown. Based on their regenerative capacity, we asked whether these cells participate in alveogenesis. We used a previously described transgenic mouse model (CCSP-tk) in which Ganciclovir exposure selectively depletes all cells with CCSP promoter activity through intracellular generation of a toxic metabolite of thymidine kinase. Our results showed that Ganciclovir treatment in newborn CCtk mice depleted this cell population in lung airways and bone marrow, and was associated with alveolar hypoplasia and respiratory failure. Hypoplastic lungs had fewer alveolar type I and II cells, with impaired secondary crest formation and decreased vascular endothelial growth factor expression in distal airways. These findings are consistent with a model in which a unique population of cells with CCSP promoter activity that expresses vascular endothelial growth factor participates in alveolar development.


Subject(s)
Epithelial Cells/metabolism , Gene Expression Regulation , Promoter Regions, Genetic , Pulmonary Alveoli/growth & development , Uteroglobin/genetics , Animals , Ganciclovir/pharmacology , Lung/metabolism , Mice , Mice, Transgenic , Naphthalenes/pharmacology , Stem Cells/cytology , Thymidine Kinase/metabolism , Vascular Endothelial Growth Factor A/metabolism
8.
Respir Res ; 12: 134, 2011 Oct 10.
Article in English | MEDLINE | ID: mdl-21985298

ABSTRACT

BACKGROUND: Alveolar septation marks the beginning of the transition from the saccular to alveolar stage of lung development. Inflammation can disrupt this process and permanently impair alveolar formation resulting in alveolar hypoplasia as seen in bronchopulmonary dysplasia in preterm newborns. NF-κB is a transcription factor central to multiple inflammatory and developmental pathways including dorsal-ventral patterning in fruit flies; limb, mammary and submandibular gland development in mice; and branching morphogenesis in chick lungs. We have previously shown that epithelial overexpression of NF-κB accelerates lung maturity using transgenic mice. The purpose of this study was to test our hypothesis that targeted deletion of NF-κB signaling in lung epithelium would impair alveolar formation. METHODS: We generated double transgenic mice with lung epithelium-specific deletion of IKKƟ, a known activating kinase upstream of NF-κB, using a cre-loxP transgenic recombination strategy. Lungs of resulting progeny were analyzed at embryonic and early postnatal stages to determine specific effects on lung histology, and mRNA and protein expression of relevant lung morphoreulatory genes. Lastly, results measuring expression of the angiogenic factor, VEGF, were confirmed in vitro using a siRNA-knockdown strategy in cultured mouse lung epithelial cells. RESULTS: Our results showed that IKKƟ deletion in the lung epithelium transiently decreased alveolar type I and type II cells and myofibroblasts and delayed alveolar formation. These effects were mediated through increased alveolar type II cell apoptosis and decreased epithelial VEGF expression. CONCLUSIONS: These results suggest that epithelial NF-κB plays a critical role in early alveolar development possibly through regulation of VEGF.


Subject(s)
Apoptosis Regulatory Proteins/deficiency , Gene Deletion , Gene Expression Regulation, Developmental , I-kappa B Kinase/deficiency , Pulmonary Alveoli/growth & development , Respiratory Mucosa/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Apoptosis Regulatory Proteins/genetics , I-kappa B Kinase/genetics , Lung/embryology , Lung/growth & development , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myofibroblasts/metabolism , Myofibroblasts/pathology , Pulmonary Alveoli/embryology , Pulmonary Alveoli/pathology , Respiratory Mucosa/embryology , Respiratory Mucosa/growth & development , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/physiology
9.
Pediatr Res ; 69(5 Pt 1): 371-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21270677

ABSTRACT

Alveolar development comprises the transition of lung architecture from saccules to gas-exchange units during late gestation and early postnatal development. Exposure to hyperoxia disrupts developmental signaling pathways and causes alveolar hypoplasia as seen in bronchopulmonary dysplasia affecting preterm human newborns. Expanding literature suggests that epigenetic changes caused by environmental triggers during development may lead to heritable changes in gene expression. Given recent data on altered histone deacetylase (HDAC) activity in lungs of humans and animal models with airspace enlargement/emphysema, we hypothesized that alveolar hypoplasia from hyperoxia exposure in neonatal mice is a consequence of cell cycle arrest and reduced HDAC activity and up-regulation of the cyclin-dependent kinase inhibitor, p21. We exposed newborn mice to hyperoxia and compared lung morphologic and epigenetic changes to room air controls. Furthermore, we pretreated a subgroup of animals with the macrolide antibiotic azithromycin (AZM), known to possess antiinflammatory properties. Our results showed that hyperoxia exposure resulted in alveolar hypoplasia and was associated with decreased HDAC1 and HDAC2 and increased p53 and p21 expression. Furthermore, AZM did not confer protection against hyperoxia-induced alveolar changes. These findings suggest that alveolar hypoplasia caused by hyperoxia is mediated by epigenetic changes affecting cell cycle regulation/senescence during lung development.


Subject(s)
Bronchopulmonary Dysplasia/enzymology , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Hyperoxia/enzymology , Pulmonary Alveoli/enzymology , Animals , Animals, Newborn , Anti-Inflammatory Agents/pharmacology , Azithromycin/pharmacology , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/physiopathology , Cell Proliferation , Cellular Senescence/genetics , Disease Models, Animal , Down-Regulation , Epigenesis, Genetic , Humans , Hyperoxia/genetics , Hyperoxia/pathology , Hyperoxia/physiopathology , Infant, Newborn , Mice , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/growth & development , Pulmonary Alveoli/pathology , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Up-Regulation
10.
Am J Respir Crit Care Med ; 180(8): 701-12, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19574443

ABSTRACT

RATIONALE: Pten is a tumor-suppressor gene involved in stem cell homeostasis and tumorigenesis. In mouse, Pten expression is ubiquitous and begins as early as 7 days of gestation. Pten(-/-) mouse embryos die early during gestation indicating a critical role for Pten in embryonic development. OBJECTIVES: To test the role of Pten in lung development and injury. METHODS: We conditionally deleted Pten throughout the lung epithelium by crossing Pten(flox/flox) with Nkx2.1-cre driver mice. The resulting Pten(Nkx2.1-cre) mutants were analyzed for lung defects and response to injury. MEASUREMENTS AND MAIN RESULTS: Pten(Nkx2.1-cre) embryonic lungs showed airway epithelial hyperplasia with no branching abnormalities. In adult mice, Pten(Nkx2.1-cre) lungs exhibit increased progenitor cell pools composed of basal cells in the trachea, CGRP/CC10 double-positive neuroendocrine cells in the bronchi, and CC10/SPC double-positive cells at the bronchioalveolar duct junctions. Pten deletion affected differentiation of various lung epithelial cell lineages, with a decreased number of terminally differentiated cells. Over time, Pten(Nxk2.1-cre) epithelial cells residing in the bronchioalveolar duct junctions underwent proliferation and formed uniform masses, supporting the concept that the cells residing in this distal niche may also be the source of procarcinogenic stem cells. Finally, increased progenitor cells in all the lung compartments conferred an overall selective advantage to naphthalene injury compared with wild-type control mice. CONCLUSIONS: Pten has a pivotal role in lung stem cell homeostasis, cell differentiation, and consequently resistance to lung injury.


Subject(s)
Cell Differentiation/genetics , Epithelial Cells/physiology , Gene Deletion , Lung Injury/genetics , Lung/embryology , PTEN Phosphohydrolase/genetics , Animals , Homeostasis , Mice , Mice, Mutant Strains , Respiratory Mucosa/cytology , Stem Cells
11.
Gene ; 393(1-2): 70-80, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17350185

ABSTRACT

Bone Morphogenetic Proteins are key signaling molecules in vertebrate development. Little is known about Bmp gene regulation in any organ. In Drosophila, the Bmp gene, dpp is regulated by Dorsal, the invertebrate homologue of Rel-NF-kB. In this study we examined whether TNF-alpha, which activates NF-kB, can regulate Bmp4 gene expression. TNF-alpha reduced Bmp4 mRNA in lung adenocarcinoma A549 cells and repressed transcriptional activity of the human Bmp4 promoter in a dose-dependent manner. Similar repression was observed when the Bmp4 promoter was co-transfected with a p65 (RelA) expression vector in the absence of TNF-alpha treatment, suggesting that RelA mediates the effect of TNF-alpha. In support of this finding, the repressor effect of TNF-alpha on Bmp4 was abrogated by a co-transfected dominant negative mutant of IkB (S32A/S36A). The human Bmp4 promoter contains 3 putative consensus binding sites for NF-kB. Surprisingly, only one of the latter binding sites was capable of binding NF-kB. Repressor effect of NF-kB was not dependent on any of the three binding sites, but localized to a 122 bp fragment which bound both RelA and SP1. SP1 stimulated transcription, whereas increasing doses of RelA opposed this effect. In vivo, TNF-alpha inhibited branching morphogenesis and LacZ gene expression in Bmp4-lacz transgenic lungs. These data support a model in which TNF-alpha-induced RelA interacts with SP1 to bring about transcriptional repression of Bmp4 gene. These findings provide a mechanistic paradigm for interactions between mediators of inflammation and morphogenesis with relevant implications for normal lung development and pathogenesis of disease.


Subject(s)
Bone Morphogenetic Proteins/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Lung/cytology , Lung/metabolism , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Base Pairing/genetics , Base Sequence , Binding Sites , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/metabolism , Cell Line, Tumor , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung/drug effects , Lung/embryology , Mice , Molecular Sequence Data , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion , Transcription Factor RelA/metabolism
12.
J Clin Invest ; 110(11): 1703-16, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12464676

ABSTRACT

Mortality related to adult respiratory distress syndrome (ARDS) ranges from 35% to 65%. Lung-protective ventilator strategies can reduce mortality during ARDS. The protective strategies limit tidal volumes and peak pressures while maximizing positive end-expiratory pressure. The efficacy of this approach is due to a reduction of shear-stress of the lung and release of inflammatory mediators. Ventilator-induced lung injury (VILI) is characterized by inflammation. The specific mechanism(s) that recruit leukocytes during VILI have not been elucidated. Because the murine CXC chemokines KC/CXCL1 and MIP-2/CXCL2/3, via CXCR2, are potent neutrophil chemoattractants, we investigated their role in a murine model of VILI. We compared two ventilator strategies in C57BL/6 mice: high peak pressure and high stretch (high peak pressure/stretch) versus low peak pressure/stretch for 6 hours. Lung injury and neutrophil sequestration from the high-peak pressure/stretch group were greater than those from the low-peak pressure/stretch group. In addition, lung expression of KC/CXCL1 and MIP-2/CXCL2/3 paralleled lung injury and neutrophil sequestration. Moreover, in vivo inhibition of CXCR2/CXC chemokine ligand interactions led to a marked reduction in neutrophil sequestration and lung injury. These findings were confirmed using CXCR2(-/-) mice. Together these experiments support the notion that increased expression of KC/CXCL1 and MIP-2/CXCL2/3 and their interaction with CXCR2 are important in the pathogeneses of VILI.


Subject(s)
Lung Injury , Lung/pathology , Receptors, Interleukin-8B/physiology , Respiration, Artificial/adverse effects , Animals , Chemokines, CXC/physiology , Humans , Ligands , Mice , Models, Animal , Peroxidase/analysis , Phosphorylation
13.
FEBS Lett ; 553(1-2): 33-8, 2003 Oct 09.
Article in English | MEDLINE | ID: mdl-14550542

ABSTRACT

Double-stranded (ds) RNA is a biologically active component of many viruses including rhinoviruses infecting the upper respiratory tract. Mucus production is a common symptom of such infections. Here, we show that mucin, the glycoprotein subunit of mucus gels, is transcriptionally upregulated in an NF-kappaB- and p38-dependent manner when homogeneous cultures of epithelial cells are exposed to dsRNA. Furthermore, upstream of p38 in this system, dsRNA stimulates the extracellular release of ATP and activation of cell surface ATP receptors, which are G protein-coupled. This results in the stimulation of phospholipase C and protein kinase C. These findings suggest that ATP receptor antagonists could be used to modulate mucus production induced by virus.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/metabolism , Mucins/genetics , RNA, Double-Stranded/pharmacology , RNA, Viral/pharmacology , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Cell Line , Enzyme Activation/drug effects , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Mucin-2 , Mucins/biosynthesis , NF-kappa B/metabolism , Protein Kinase C/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/drug effects , Type C Phospholipases/metabolism , eIF-2 Kinase/metabolism , p38 Mitogen-Activated Protein Kinases
14.
Dev Dyn ; 237(2): 328-38, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18161062

ABSTRACT

Lung maturation is hallmarked by the appearance of surfactant-producing alveoli during transition from the saccular to alveolar stage of lung development. Inflammation can disrupt this process and accelerate lung maturity following intrauterine amniotic infection (chorioamnionitis). Nuclear factor kB (NF-kB) is a transcription factor central to multiple inflammatory and developmental pathways, including dorsal-ventral patterning in fruit flies, limb and mammary and submandibular gland development in mice, and branching morphogenesis in chick lungs. Given its shared role in inflammation and developmental signaling, we hypothesized that overexpression of NF-kB targeted to the lung epithelium would exert maturational effects on alveolar development. We generated transgenic mice with lung-specific overexpression of the RelA subunit of NF-kB using a surfactant protein C promoter construct. Our results showed that RelA overexpression in the lung yields increased alveolar type I and type II cells. These findings are consistent with a model whereby NF-kB may induce maturation of lung development through decreased apoptosis of epithelial cells.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Lung/embryology , Morphogenesis/physiology , Transcription Factor RelA/metabolism , Animals , Apoptosis/physiology , Aquaporin 5/metabolism , Blotting, Northern , Blotting, Western , Bromodeoxyuridine , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Intercellular Signaling Peptides and Proteins , Lung/ultrastructure , Mice , Mice, Transgenic , Microscopy, Electron, Scanning , Nuclear Proteins/metabolism , Peptides/metabolism , Promoter Regions, Genetic/genetics , Protein C/genetics , Pulmonary Surfactant-Associated Protein C , Thyroid Nuclear Factor 1 , Transcription Factors/metabolism
15.
Clin Immunol ; 120(1): 45-56, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16473552

ABSTRACT

Co-aggregation of FcepsilonRI with FcgammaRIIb can block FcepsilonRI-mediated reactivity and Fc gamma:allergen chimeric proteins, by co-crosslinking FcgammaRIIb to allergen-specific IgE bound to the FcepsilonRI can block allergen-specific reactivity. We evaluated whether a human cat chimeric fusion protein (GFD) composed of part of the human Ig G1 Fc fused to the major cat allergen (Fel d1) would function as allergen immunotherapy while not inducing acute allergic reactivity in mice sensitized to Fel d1. Injection of GFD 6 h prior to Fel d1 challenge acutely blocked systemic and skin reactivity to Fel d1 challenge while mice given subcutaneous immunotherapy with GFD at days 37, 38, and 39 showed inhibition of systemic, lung, and cutaneous reactivity to Fel d1 2 weeks later. GFD immunotherapy did not induce systemic reactivity. Overall, the Fcgamma-Fel d1 chimeric fusion protein blocked Fel d1-induced IgE-mediated reactivity but did not induce in vivo mediator release on its own; suggesting that this approach using allergen combined with Fc gamma1 so as to achieve inhibitory signaling may provide an enhanced form of allergen immunotherapy.


Subject(s)
Glycoproteins/immunology , Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/prevention & control , Immunotherapy/methods , Receptors, IgG/immunology , Recombinant Fusion Proteins/immunology , Animals , Bronchial Provocation Tests , Bronchoalveolar Lavage Fluid/cytology , Cats , Cell Count , Glycoproteins/genetics , Histocytochemistry , Humans , Immunoglobulin E/blood , Immunoglobulin G/blood , Lung/immunology , Male , Mice , Mice, Inbred BALB C , Receptors, IgG/genetics , Recombinant Fusion Proteins/genetics , Skin/immunology , Specific Pathogen-Free Organisms
16.
Pediatr Res ; 59(4 Pt 2): 84R-93R, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16549554

ABSTRACT

Tissues have the capacity to maintain a homeostatic balance between wear-and-tear and regeneration. Repair of non-lethal injury also activates cell proliferation to repopulate the injured sites with appropriate cell types and to restore function. Although controversial, the source of the material appears to be at least partly from pools of unique, multipotent stem cells that reside in specialized locations referred to as "niches." Molecular interactions between the niche and the intracellular factors within stem cells are crucial in maintaining stem cell functions, particularly the balance between self-renewal and differentiation. Many of the mediators of the stem cell-niche interactions are similar or identical to those that control developmental pathways during organogenesis. In this review, we present a systematic discussion and evaluation of the relevant literature with a focused emphasis on three primary signaling pathways, WNT, SHH and BMP with potentially overlapping roles during both development and stem cell maintenance.


Subject(s)
Lung/cytology , Stem Cells/cytology , Humans , Lung/growth & development , Morphogenesis
17.
J Inflamm (Lond) ; 2(1): 4, 2005 05 28.
Article in English | MEDLINE | ID: mdl-15921526

ABSTRACT

BACKGROUND: Respiratory viral infections are characterized by the infiltration of leukocytes, including activated neutrophils into the lung that can lead to sustained lung injury and potentially contribute to chronic lung disease. Specific mechanisms recruiting neutrophils to the lung during virus-induced lung inflammation and injury have not been fully elucidated. Since CXCL1 and CXCL2/3, acting through CXCR2, are potent neutrophil chemoattractants, we investigated their role in dsRNA-induced lung injury, where dsRNA (Poly IC) is a well-described synthetic agent mimicking acute viral infection. METHODS: We used 6-8 week old female BALB/c mice to intratracheally inject either single-stranded (ssRNA) or double-stranded RNA (dsRNA) into the airways. The lungs were then harvested at designated timepoints to characterize the elicited chemokine response and resultant lung injury following dsRNA exposure as demonstrated qualititatively by histopathologic analysis, and quantitatively by FACS, protein, and mRNA analysis of BAL fluid and tissue samples. We then repeated the experiments by first pretreating mice with an anti-PMN or corresponding control antibody, and then subsequently pretreating a separate cohort of mice with an anti-CXCR2 or corresponding control antibody prior to dsRNA exposure. RESULTS: Intratracheal dsRNA led to significant increases in neutrophil infiltration and lung injury in BALB/c mice at 72 h following dsRNA, but not in response to ssRNA (Poly C; control) treatment. Expression of CXCR2 ligands and CXCR2 paralleled neutrophil recruitment to the lung. Neutrophil depletion studies significantly reduced neutrophil infiltration and lung injury in response to dsRNA when mice were pretreated with an anti-PMN monoclonal Ab. Furthermore, inhibition of CXCR2 ligands/CXCR2 interaction by pretreating dsRNA-exposed mice with an anti-CXCR2 neutralizing Ab also significantly attenuated neutrophil sequestration and lung injury. CONCLUSION: These findings demonstrate that CXC chemokine ligand/CXCR2 biological axis is critical during the pathogenesis of dsRNA-induced lung injury relevant to acute viral infections.

18.
Pediatr Res ; 58(5): 919-26, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16183824

ABSTRACT

The histologic phenotype of bronchopulmonary dysplasia (BPD) is characterized by decreased alveolization and is preceded by infiltration of activated neutrophils into the lung that can lead to sustained lung injury and potential interruption of normal lung development. Potential pathogens triggering early neutrophil influx include either prenatal or postnatal exposure to bacteria or viruses. Specific mechanisms recruiting neutrophils to the lung and subsequently decreasing alveolization during virus-induced lung inflammation and injury have not been fully elucidated. Because CXC chemokines, such as CXCL1 and CXCL2/3 acting through their putative receptor, CXCR2, are potent neutrophil chemoattractants, the authors investigated their role in dsRNA-induced lung injury and decreased alveolization, in which dsRNA (poly IC) is a well-described synthetic agent mimicking acute viral infection. Intratracheal dsRNA led to significant increases in neutrophil infiltration and lung injury at 72 hours and to decreased alveolization at 5 days after dsRNA exposure in newborn (10 days old) BALB/c mice, when compared with controls treated and not treated with ssRNA (poly C). Expression of CXCL1 and CXCR2 paralleled neutrophil recruitment to the lung and preceded the decrease in alveolization. Inhibition of CXCR2/CXCR2 ligand interaction by pretreating dsRNA-exposed mice with an anti-CXCR2 neutralizing antibody significantly attenuated neutrophil sequestration and lung injury, and preserved normal alveolization. These findings demonstrate that the CXCR2/CXCR2 ligand biologic axis plays an important role during the pathogenesis of dsRNA-induced lung injury and decreased alveolization and may be relevant to the pathogenesis of BPD.


Subject(s)
Pneumonia/pathology , Pulmonary Alveoli/pathology , RNA, Double-Stranded/administration & dosage , Receptors, Interleukin-8B/physiology , Animals , Base Sequence , Bronchoalveolar Lavage Fluid , DNA Primers , Enzyme-Linked Immunosorbent Assay , Ligands , Mice , Mice, Inbred BALB C , Pneumonia/chemically induced , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8B/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL