Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Pediatr Transplant ; 22(2)2018 03.
Article in English | MEDLINE | ID: mdl-29417689

ABSTRACT

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


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Bronchiolitis Obliterans/drug therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Methylprednisolone/administration & dosage , Adolescent , Anti-Inflammatory Agents/therapeutic use , Bronchiolitis Obliterans/etiology , Child , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Injections, Intravenous , Male , Methylprednisolone/therapeutic use , Retrospective Studies , Treatment Outcome
2.
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
3.
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
4.
Allergy Asthma Clin Immunol ; 10(1): 2, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24438707

ABSTRACT

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

5.
Pediatr Pulmonol ; 49(4): 309-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24339235

ABSTRACT

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


Subject(s)
Cystic Fibrosis , Estrogens/physiology , Cystic Fibrosis/complications , Cystic Fibrosis/etiology , Cystic Fibrosis/mortality , Disease Progression , Female , Humans , Inflammation/etiology , Male , Sex Factors , Survival Rate
6.
Respir Res ; 14: 138, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24344776

ABSTRACT

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


Subject(s)
CD4-Positive T-Lymphocytes/pathology , Cell Differentiation , Cystic Fibrosis/pathology , Phenotype , Th17 Cells/pathology , Adolescent , Adult , Animals , CD4-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cells, Cultured , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Disease Models, Animal , Female , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th1 Cells/metabolism , Th1 Cells/pathology , Th17 Cells/metabolism , Young Adult
7.
Respir Res ; 11: 166, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21118573

ABSTRACT

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


Subject(s)
Cystic Fibrosis/complications , Estrogens/adverse effects , Pneumonia, Bacterial/chemically induced , Pneumonia, Bacterial/pathology , Pseudomonas Infections/chemically induced , Pseudomonas Infections/pathology , Pseudomonas aeruginosa , Animals , Cystic Fibrosis/pathology , Disease Models, Animal , Male , Mice
8.
Am J Respir Cell Mol Biol ; 43(5): 599-606, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20042713

ABSTRACT

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


Subject(s)
Epithelium/metabolism , Epithelium/pathology , Lung/metabolism , Lung/pathology , Receptors, Glucocorticoid/deficiency , Animals , Animals, Newborn , Biomarkers/metabolism , Doxycycline/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Epithelium/drug effects , Epithelium/embryology , Gene Expression Regulation, Developmental/drug effects , Lung/embryology , Mice , Mice, Knockout , Organ Specificity/drug effects , Organogenesis/drug effects , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Pulmonary Surfactant-Associated Proteins/genetics , Pulmonary Surfactant-Associated Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Survival Analysis , Transcription Factors/metabolism
9.
Pediatr Res ; 67(4): 375-81, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20057335

ABSTRACT

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


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation/drug effects , Proteins/metabolism , Pulmonary Alveoli/physiology , Steroids/pharmacology , Tretinoin/pharmacology , Vitamin D/pharmacology , Animals , Binding Sites , Cell Line , Female , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Pregnancy , Promoter Regions, Genetic , Proteins/genetics , Pulmonary Alveoli/cytology , Rats , Rats, Sprague-Dawley , Transcription, Genetic
10.
Respir Res ; 10: 83, 2009 Sep 21.
Article in English | MEDLINE | ID: mdl-19772569

ABSTRACT

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


Subject(s)
Glycoproteins/metabolism , Goblet Cells/metabolism , Immunologic Factors/metabolism , Lung/metabolism , Mice, Knockout/metabolism , Respiratory Mechanics , Animals , Cells, Cultured , Cytokines , Glycoproteins/genetics , Lung Injury , Mice
11.
Respir Res ; 10: 77, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19698107

ABSTRACT

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


Subject(s)
Angiogenic Proteins/metabolism , Cytokines/metabolism , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Pulmonary Alveoli/drug effects , Tretinoin/pharmacology , Age Factors , Angiogenic Proteins/genetics , Animals , Animals, Newborn , Blotting, Northern , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Cytokines/genetics , Epithelial Cells/metabolism , Humans , Midkine , Pulmonary Alveoli/growth & development , Pulmonary Alveoli/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
12.
Pediatr Pulmonol ; 43(2): 125-33, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18085690

ABSTRACT

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


Subject(s)
Glucocorticoids/metabolism , Lung/pathology , Receptors, Glucocorticoid/genetics , Respiratory Insufficiency/genetics , Respiratory Insufficiency/pathology , Animals , Animals, Newborn , Capillaries/pathology , Endothelial Cells/pathology , Glucocorticoids/genetics , Heterozygote , Homozygote , Immunohistochemistry , Lung/metabolism , Mice , Mice, Inbred C57BL/genetics , Muscle, Smooth/pathology , Respiratory Insufficiency/metabolism , Severity of Illness Index , Signal Transduction
13.
Pediatr Pulmonol ; 42(6): 519-24, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17469152

ABSTRACT

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


Subject(s)
Amiloride/pharmacology , Cystic Fibrosis/physiopathology , Membrane Potentials/drug effects , Menstrual Cycle/physiology , Nasal Mucosa/physiology , Sodium Channel Blockers/pharmacology , Adolescent , Adult , Case-Control Studies , Cystic Fibrosis/blood , Estrogens/blood , Female , Follicular Phase/blood , Humans , Ion Transport/drug effects , Ion Transport/physiology , Luteal Phase/blood , Membrane Potentials/physiology , Menstrual Cycle/blood , Nasal Mucosa/cytology , Nasal Mucosa/drug effects , Progesterone/blood , Sex Factors
14.
Biol Neonate ; 90(1): 46-57, 2006.
Article in English | MEDLINE | ID: mdl-16534186

ABSTRACT

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


Subject(s)
Cytokines/genetics , Lung/physiology , Peptides/genetics , Promoter Regions, Genetic , Receptors, Glucocorticoid/genetics , Animals , Animals, Genetically Modified , Animals, Newborn , Base Sequence , DNA Primers , Gene Expression Regulation , Genotype , Humans , Intercellular Signaling Peptides and Proteins , Lung/cytology , Mice , Mice, Knockout , Mice, Transgenic , Midkine , Pulmonary Surfactant-Associated Protein C , Rats , Receptors, Glucocorticoid/deficiency
15.
Pediatr Res ; 59(3): 389-95, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16492977

ABSTRACT

Bronchopulmonary dysplasia (BPD), a major cause of morbidity in premature infants, is characterized by arrest of lung growth and inhibited alveologenesis. We had earlier cloned late-gestation lung 1 (LGL1), a glucocorticoid (GC)-induced, developmentally regulated gene in lung mesenchyme, and showed that reduced levels of late-gestation lung 1 protein (lgl1) inhibit lung branching. Maximal fetal expression of LGL1 is concordant with the onset of alveolar septation, suggesting an additional role for lgl1 in alveologenesis. At postnatal d 7, during the period of maximal septation in postnatal rat lung, lgl1 concentrates at the tips of budding secondary alveolar septa. We studied two models of impaired postnatal alveologenesis generated by exposure of newborn rats to 60% O2 for 2 wk or 95% O2 for 1 wk. A profound decrease of lgl1 expression with oxygen exposure was observed in both animal models. Animals exposed to 95% O2 for 1 wk recovered in air over a 3-wk period, associated with normalization of lgl1 levels. Changes in lung levels of alpha-actin (a marker of myofibroblast differentiation associated with alveologenesis) and the mesenchymal marker vimentin were significant but less marked. Our findings support a role for lgl1 in postnatal lung development. We speculate that deficiency of lgl1 contributes to the arrested alveolar partitioning observed in BPD and that recovery is associated with normalization of lgl1 levels.


Subject(s)
Air , Bronchopulmonary Dysplasia/physiopathology , Lung , Oxygen/toxicity , Proteins/metabolism , Actins/genetics , Actins/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Female , Humans , Infant, Newborn , Lung/anatomy & histology , Lung/growth & development , Lung/metabolism , Lung/pathology , Pregnancy , Proteins/genetics , Rats , Rats, Sprague-Dawley , Vimentin/genetics , Vimentin/metabolism
16.
Pediatr Pulmonol ; 39(2): 185-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15532092

ABSTRACT

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


Subject(s)
Carcinoma, Adenosquamous/pathology , Cell Transformation, Neoplastic , Laryngeal Neoplasms/pathology , Lung Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Papilloma/pathology , Adolescent , Biopsy, Needle , Carcinoma, Adenosquamous/diagnostic imaging , Carcinoma, Adenosquamous/therapy , Cell Transformation, Neoplastic/pathology , Combined Modality Therapy , Diagnosis, Differential , Female , Follow-Up Studies , Humans , Laryngeal Neoplasms/diagnostic imaging , Laryngeal Neoplasms/therapy , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/therapy , Papilloma/diagnostic imaging , Papilloma/therapy , Radiography, Thoracic , Tomography, X-Ray Computed
17.
Am J Respir Crit Care Med ; 170(11): 1188-96, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15347560

ABSTRACT

The lungs of newborn rats exposed to 60% oxygen for 14 days develop an injury that shares morphologic similarities to human bronchopulmonary dysplasia (BPD). Neutrophil influx into the lung, as part of an inflammatory response, may play a pivotal role in the development of BPD. A neutrophil chemokine, cytokine-induced neutrophil chemoattractant-1, which signals through the neutrophil CXC chemokine receptor-2, is increased in the lung tissue of newborn rats exposed to 60% oxygen. The purpose of this study was to explore the role of neutrophils in the rat model of BPD by inhibiting neutrophil influx using SB265610, a selective CXC chemokine receptor-2 antagonist. SB265610, administered to 60% oxygen-exposed newborn rats from birth to 14 days, completely inhibited neutrophil influx. It also attenuated increased production of reactive oxygen species in newborn rat lung tissue after exposure to 60% oxygen for 4 days. Lung morphometric analysis revealed that 60% oxygen for 14 days, when accompanied by treatment with SB265610 to prevent neutrophil accumulation, increased alveolar formation over that seen in newborn rats exposed to air. These data suggest that exposure of the neonatal lung to moderate hyperoxia may enhance postnatal lung growth, provided postnatal pulmonary inflammation is suppressed.


Subject(s)
Lung Diseases/physiopathology , Lung/growth & development , Neutrophil Infiltration/immunology , Oxygen/pharmacology , Receptors, Interleukin-8B/antagonists & inhibitors , Animals , Animals, Newborn , Bronchopulmonary Dysplasia , Disease Models, Animal , Humans , Infant, Newborn , Lung/drug effects , Lung/immunology , Lung Diseases/immunology , Oxygen/immunology , Rats , Reactive Oxygen Species , Receptors, Interleukin-8B/immunology
18.
Am J Respir Cell Mol Biol ; 31(4): 377-81, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15381503

ABSTRACT

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


Subject(s)
Anti-Bacterial Agents/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Cystic Fibrosis/physiopathology , Cystic Fibrosis/therapy , Inflammation Mediators/physiology , Lung/physiopathology , Animals , Cystic Fibrosis/complications , Humans , Lung/immunology
19.
Am J Respir Cell Mol Biol ; 30(3): 350-9, 2004 Mar.
Article in English | MEDLINE | ID: mdl-12871852

ABSTRACT

To investigate molecular mechanisms of lung organogenesis, we searched for glucocorticoid-inducible genes in developing lung. We cloned LGL2, a developmentally and hormonally regulated gene in fetal lung (Zhang, C., N. B. Sweezey, S. Gagnon, B. Muskat, D. Koehler, M. Post, and F. Kaplan. 2000. A novel karyopherin-beta homolog is developmentally and hormonally regulated in fetal lung. Am. J. Respir. Cell Mol. Biol. 22:451-459). A comparison of lgl2 protein to sequences in the genome database suggested that lgl2 is a nuclear transport receptor. We report on the functional characterization of lgl2 as an importin beta protein and on the developmental regulation of its nucleocytoplasmic shuttling in fetal lung. We investigated the subcellular localization and Ran-binding properties of lgl2 and its N- and C-terminal regions. We used fluorescence recovery after photobleaching and fluorescence loss in photobleaching to study nucleocytoplasmic shuttling of lgl2. We showed that N-terminal lgl2 supports shuttling at a reduced rate. We showed that the nucleocytoplasmic distribution of lgl2 favors the nucleus in fetal lung and that lgl2 enters the nucleus much more rapidly at fetal Day 18 than at Day 21. Total nuclear recovery of lgl2 was dramatically different at the two time points. Early in development, nuclear import of transcription factors in response to hormones and growth agonists regulates prominent signal transduction pathways that govern lung organogenesis. We speculate that lgl2 may be one important modulator of this process.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Expression Regulation, Developmental/physiology , Lung/embryology , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , Animals , Biological Transport , Embryonic and Fetal Development , HeLa Cells , Humans , Lung/physiology , Nuclear Localization Signals , Rats , Rats, Wistar , Signal Transduction , Subcellular Fractions , beta Karyopherins/genetics , ran GTP-Binding Protein/metabolism
20.
Biochem J ; 376(Pt 1): 61-9, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-12880386

ABSTRACT

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


Subject(s)
Glycoproteins/metabolism , Lung/embryology , Proteins/metabolism , Respiratory Mucosa/embryology , Animals , Cell Line , Cells, Cultured , Endoplasmic Reticulum/chemistry , Epithelial Cells/metabolism , Gestational Age , Glycosylation , Golgi Apparatus/chemistry , Humans , Lung/cytology , Lung/metabolism , Mesoderm/physiology , Morphogenesis , Protein Sorting Signals , Protein Transport , Proteins/physiology , Rats , Rats, Wistar , Respiratory Mucosa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...