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










Publication year range
1.
Chest ; 137(4): 969-73, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20371530

ABSTRACT

A man with usual interstitial pneumonia (age of onset 58 years) was previously found to have an Ile73Thr (I73T) surfactant protein C (SFTPC) mutation. Genomic DNA from the individual and two daughters (aged 39 and 43 years) was sequenced for the I73T mutation and variations in ATP-binding cassette A3 (ABCA3). All three had the I73T SFTPC mutation. The father and one daughter (aged 39 years) also had a transversion encoding an Asp123Asn (D123N) substitution in ABCA3. The daughters were evaluated by pulmonary function testing and high-resolution CT (HRCT). Neither daughter had evidence of disease, except for focal subpleural septal thickening on HRCT scan in one daughter (aged 39 years). This daughter underwent bronchoscopy with transbronchial biopsies revealing interstitial fibrotic remodeling. These findings demonstrate that subclinical fibrotic changes may be present in family members of patients with SFTPC mutation-associated interstitial lung disease and suggest that ABCA3 variants could affect disease pathogenesis.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Lung Diseases, Interstitial/genetics , Mutation/genetics , Pulmonary Surfactant-Associated Protein C/genetics , Adult , Female , Humans , Lung Diseases, Interstitial/diagnosis , Male , Middle Aged , Pedigree
2.
Am J Physiol Lung Cell Mol Physiol ; 298(5): L646-59, 2010 May.
Article in English | MEDLINE | ID: mdl-20190032

ABSTRACT

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


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Alveolar Epithelial Cells/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism , ATP-Binding Cassette Transporters/metabolism , Alveolar Epithelial Cells/ultrastructure , Animals , Animals, Newborn , Base Sequence , Disease Models, Animal , Female , Gene Expression , Homeostasis , Humans , Infant, Newborn , Lipid Metabolism , Mice , Microscopy, Electron, Transmission , Pregnancy , Pulmonary Emphysema/etiology , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Pulmonary Surfactant-Associated Proteins/deficiency , RNA, Messenger/genetics , RNA, Messenger/metabolism , Respiratory Distress Syndrome, Newborn/etiology
3.
Pediatr Res ; 65(4): 397-402, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19092718

ABSTRACT

Administration of retinoic acid (RA), the active metabolite of vitamin A, is linked to the stimulation of nephrogenesis. The aim of this study was to determine whether early postnatal administration of RA could enhance ongoing nephrogenesis in a baboon model of premature birth. Unbiased stereological methods were used to estimate kidney volume, renal corpuscle volume, and nephron number. The percentage of abnormal glomeruli and the number of glomerular generations was also determined in the kidneys of preterm control (n = 6) and preterm +RA (n = 6) animals that received 500 microg/kg/d of all-trans RA after premature delivery. There was no significant difference between the preterm control and the preterm +RA groups in kidney size, nephron number (preterm control: 329,924 +/- 41,752; preterm +RA: 354,041 +/- 52,095; p = 0.59), renal corpuscle volume, number of glomerular generations, or the percentage of abnormal glomeruli. The proportion of abnormal glomeruli did not appear to be linked to any elements of postnatal care examined. The results of this study indicate that early postnatal administration of RA is unable to stimulate nephrogenesis in the kidney of the preterm baboon. Encouragingly, it does not appear to have any adverse effects on kidney development.


Subject(s)
Kidney/drug effects , Nephrons/drug effects , Premature Birth , Tretinoin/administration & dosage , Vitamins/administration & dosage , Animals , Animals, Newborn , Birth Weight , Female , Gestational Age , Immunohistochemistry , In Situ Hybridization , Kidney/chemistry , Kidney/growth & development , Kidney Glomerulus/drug effects , Kidney Glomerulus/growth & development , Nephrons/chemistry , Nephrons/growth & development , Organ Size , Organogenesis/drug effects , Papio , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Podocytes/drug effects , Pregnancy , Vascular Endothelial Growth Factor A/analysis , WT1 Proteins/analysis
4.
J Biol Chem ; 284(6): 4018-30, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19074148

ABSTRACT

Pulmonary function after birth is dependent upon surfactant lipids that reduce surface tension in the alveoli. The sterol-responsive element-binding proteins (SREBPs) are transcription factors regulating expression of genes controlling lipid homeostasis in many tissues. To identify the role of SREBPs in the lung, we conditionally deleted the SREBP cleavage-activating protein gene, Scap, in respiratory epithelial cells (ScapDelta/Delta) in vivo. Prior to birth (E18.5), deletion of Scap decreased the expression of both SREBPs and a number of genes regulating fatty acid and cholesterol metabolism. Nevertheless, ScapDelta/Delta mice survived postnatally, surfactant and lung tissue lipids being substantially normalized in adult ScapDelta/Delta mice. Although phospholipid synthesis was decreased in type II cells from adult ScapDelta/Delta mice, lipid storage, synthesis, and transfer by lung lipofibroblasts were increased. mRNA microarray data indicated that SCAP influenced two major gene networks, one regulating lipid metabolism and the other stress-related responses. Deletion of the SCAP/SREBP pathway in respiratory epithelial cells altered lung lipid homeostasis and induced compensatory lipid accumulation and synthesis in lung lipofibroblasts.


Subject(s)
Fibroblasts/metabolism , Homeostasis/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism/physiology , Membrane Proteins/metabolism , Pulmonary Alveoli/metabolism , Animals , Fibroblasts/cytology , Gene Deletion , Gene Expression Regulation/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Transgenic , Phospholipids/genetics , Phospholipids/metabolism , Pulmonary Alveoli/cytology , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism
5.
Dev Biol ; 320(1): 149-60, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18544451

ABSTRACT

ELF5, an Ets family transcription factor found exclusively in epithelial cells, is expressed in the distal lung epithelium during embryogenesis, then becomes restricted to proximal airways at the end of gestation and postnatally. To test the hypothesis that ELF5 represses distal epithelial differentiation, we generated a transgenic mouse model in which a doxycycline inducible HA-tagged mouse Elf5 transgene was placed under the control of the lung epithelium-specific human SFTPC promoter. We found that expressing high levels of ELF5 during early lung development disrupted branching morphogenesis and produced a dilated epithelium. The effects of ELF5 on morphogenesis were stage-dependent, since inducing the transgene on E16.5 had no effect on branching. ELF5 reduced expression of the distal lung epithelial differentiation markers Erm, Napsa and Sftpc, and type II cell ultrastructural differentiation was immature. ELF5 overexpression did not induce the proximal airway epithelial markers Ccsp and Foxj1, but did induce expression of p63, a marker of basal cells in the trachea and esophagus. High ELF5 levels also induced the expression of genes found in other endodermal epithelia but not normally associated with the lung. These results suggest that precise levels of ELF5 regulate the specification and differentiation of epithelial cells in the lung.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Lung/cytology , Lung/embryology , Morphogenesis , Transcription Factors/metabolism , Animals , Cell Proliferation , DNA-Binding Proteins/genetics , Epithelium/embryology , Gene Expression Regulation, Developmental , Humans , Mice , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-ets/metabolism , Pulmonary Surfactant-Associated Protein C/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , Transcription Factors/genetics
6.
J Histochem Cytochem ; 55(1): 71-83, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16982851

ABSTRACT

ABCA3 is a member of the ATP-binding cassette (ABC) family of transport proteins and is required for perinatal respiratory adaptation. Monoclonal and polyclonal antibodies were generated against a recombinant human ABCA3 peptide and used to assess its expression in the developing lung and adult tissues. Immunostaining for ABCA3 was detected at highest levels in type II epithelial cells of the lung but was also noted in other organs including liver, stomach, kidney, adrenal, pancreas, trachea, and brain. In the fetal lung, ABCA3 staining and mRNA increased prior to birth. Like other surfactant protein genes, ABCA3 expression was induced by thyroid transcription factor-1 in vitro. ABCA3 was coexpressed with SP-B and proSP-C in type II epithelial cells. ABCA3 staining was detected surrounding large, intracellular organelles consistent with its association with lamellar bodies. In the human fetal lung, ABCA3 staining was not detected prior to 22-23 weeks of gestation, except in the presence of pulmonary inflammation. ABCA3 was detected in type II epithelial cells of the human lung from 28 weeks of gestation and thereafter. Postnatally, intense ABCA3 staining was observed in hyperplastic epithelial cells relining injured airways in infants with chronic lung disease. Localization and regulation of ABCA3 in the respiratory epithelium is consistent with its proposed role in surfactant homeostasis. The role of ABCA3 in extrapulmonary tissues and organs remains to be elucidated. This manuscript contains online supplemental material at (www.jhc.org). Please visit this article online to view these materials.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Lung/metabolism , ATP-Binding Cassette Transporters/genetics , Adult , Amino Acid Sequence , Animals , Bronchopulmonary Dysplasia/metabolism , Child , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Humans , Immunohistochemistry , Infant , Infant, Newborn , Lung/embryology , Lung/growth & development , Mice , Molecular Sequence Data , Organ Specificity , RNA, Messenger/biosynthesis , Retrospective Studies
7.
Exp Lung Res ; 32(8): 379-93, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17090478

ABSTRACT

The lipid-containing alveolar interstitial fibroblast (lipofibroblast) is known to be critically involved in rodent lung development, homeostasis, and injury/repair. However, there is lack of information on their presence and function in the human lung. Based on a number of morphological (lipid staining), molecular (presence of characteristic lipogenic and absence of myogenic markers), and functional (triglyceride uptake) characteristics that are the hallmarks of the rodent lung lipofibroblast, using human lung fibroblasts of embryonic (WI-38) and adult origin and lung tissue from human autopsy specimens, the authors for the first time clearly demonstrate the presence of lipofibroblasts in the human lung.


Subject(s)
Fibroblasts/cytology , Fibroblasts/metabolism , Pulmonary Alveoli/cytology , Triglycerides/pharmacokinetics , Actins/genetics , Adult , Azo Compounds , Biomarkers , Cells, Cultured , Coloring Agents , Fetus/cytology , Homeostasis/physiology , Humans , Inclusion Bodies/metabolism , PPAR gamma/genetics , Parathyroid Hormone-Related Protein/genetics , RNA, Messenger , Reverse Transcriptase Polymerase Chain Reaction
8.
Am J Respir Crit Care Med ; 173(3): 318-26, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16166622

ABSTRACT

RATIONALE: Bronchopulmonary dysplasia (BPD) continues to be a major morbidity in preterm infants. The lung pathology in BPD is characterized by impaired alveolar and capillary development. An imbalance between proteases and protease inhibitors in association with changes in lung elastic fibers has been implicated in the pathogenesis of BPD. OBJECTIVE: To investigate the expression and activity levels of papain-like lysosomal cysteine proteases, cathepsins B, H, K, L, S, and their inhibitors, cystatins B and C, in a baboon model of BPD. METHODS: Real-time reverse transcriptase-polymerase chain reaction, immunohistochemistry, immunoblotting, active site labeling of cysteine proteases, and in situ hybridization were performed. MEASUREMENTS AND MAIN RESULTS: The steady-state mRNA and protein levels of all cathepsins were significantly increased in the lung tissue of baboons with BPD. In contrast, the steady-state mRNA and protein levels of two major cysteine protease inhibitors, cystatin B and C, were unchanged. Correlating with these alterations, the activity of cysteine proteases in lung tissue homogenates and bronchoalveolar lavage fluid was significantly higher in the BPD group. The levels of cathepsin B, H, and S increased and cathepsin K decreased with advancing gestation. All cathepsins, except for cat K, were immunolocalized to macrophages in BPD. In addition, cathepsin H and cystatin B were colocalized in type 2 alveolar epithelial cells. Cathepsin L was detected in some bronchial epithelial, endothelial, and interstitial cells. Cathepsin K was localized to some perivascular cells by in situ hybridization. CONCLUSIONS: Cumulatively, these findings demonstrate an imbalance between cysteine proteases and their inhibitors in BPD.


Subject(s)
Bronchopulmonary Dysplasia/enzymology , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/metabolism , Animals , Animals, Newborn , Cathepsins/genetics , Cathepsins/metabolism , Cystatin B , Cystatin C , Cystatins/genetics , Cystatins/metabolism , Cysteine Endopeptidases/genetics , Cysteine Proteinase Inhibitors/genetics , Disease Models, Animal , Humans , Infant, Newborn , Papio , RNA, Messenger/metabolism
10.
Am J Physiol Lung Cell Mol Physiol ; 289(6): L971-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16040629

ABSTRACT

An activated form of beta-catenin [Catnb(Delta(ex3))] was expressed in respiratory epithelial cells of the developing lung. Although morphogenesis was not altered at birth, air space enlargement and epithelial cell dysplasia were observed in the early postnatal period and persisted into adulthood. The Catnb(Delta(ex3)) protein caused squamous, cuboidal, and goblet cell dysplasia in intrapulmonary conducting airways. Atypical epithelial cells that stained for surfactant pro protein C (pro-SP-C) and had morphological characteristics of alveolar type II cells were observed in bronchioles of the transgenic mice. Catnb(Delta(ex3)) inhibited expression of Foxa2 and caused goblet cell hyperplasia associated with increased staining for mucins and the MUC5A/C protein. In vitro, both wild type and activated beta-catenin negatively regulated the expression of the Foxa2 promoter. Catnb(Delta(ex3)) also caused pulmonary tumors in adult mice. Activation of beta-catenin caused ectopic differentiation of alveolar type II-like cells in conducting airways, goblet cell hyperplasia, and air space enlargement, demonstrating a critical role for the Wnt/beta-catenin signal transduction pathway in the differentiation of the respiratory epithelium in the postnatal lung.


Subject(s)
Cell Differentiation , Epithelial Cells/metabolism , Lung/embryology , Morphogenesis , beta Catenin/metabolism , Animals , Animals, Newborn , Cell Differentiation/genetics , Epithelial Cells/pathology , Gene Expression/genetics , Gene Expression Regulation, Developmental/genetics , Goblet Cells/cytology , Goblet Cells/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Intercellular Signaling Peptides and Proteins , Lung/cytology , Mice , Mice, Transgenic , Morphogenesis/genetics , Mucin 5AC , Mucins/metabolism , Peptides/metabolism , Protein Precursors/metabolism , Pulmonary Surfactant-Associated Protein C , Signal Transduction/genetics , Wnt Proteins/metabolism , beta Catenin/genetics
11.
Am J Physiol Lung Cell Mol Physiol ; 289(1): L34-43, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15764643

ABSTRACT

To determine potential relationships between transforming growth factor (TGF)-alpha and surfactant homeostasis, the metabolism, function, and composition of surfactant phospholipid and proteins were assessed in transgenic mice in which TGF-alpha was expressed in respiratory epithelial cells. Secretion of saturated phosphatidylcholine was decreased 40-60% by expression of TGF-alpha. Although SP-A, SP-B, and SP-C mRNA levels were unchanged by expression of TGF-alpha, SP-A and SP-B content in bronchoalveolar lavage fluid was decreased. The minimum surface tension of surfactant isolated from the transgenic mice was significantly increased. Incubation of cultured normal mice type II cells with TGF-alpha in vitro did not change secretion of surfactant phosphatidylcholine and SP-B, indicating that TGF-alpha does not directly influence surfactant secretion. Expression of a dominant negative (mutant) EGF receptor in the respiratory epithelium blocked the TGF-alpha-induced changes in lung morphology and surfactant secretion, indicating that EGF receptor signaling in distal epithelial cells was required for TGF-alpha effects on surfactant homeostasis. Because many epithelial cells were embedded in fibrotic lesions caused by TGF-alpha, changes in surfactant homeostasis may at least in part be influenced by tissue remodeling that results in decreased surfactant secretion. The number of nonembedded type II cells was decreased 30% when TGF-alpha was expressed during development and was increased threefold by TGF-alpha expression in adulthood, suggesting possible alteration of type II cells on surfactant metabolism in the adult lung. Abnormalities in surfactant function and decreased surfactant level in the airways may contribute to the pathophysiology induced by TGF-alpha in both the developing and adult lung.


Subject(s)
Epithelial Cells/metabolism , Homeostasis/physiology , Lung/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism , Transforming Growth Factor alpha/metabolism , Animals , Cells, Cultured , Epithelial Cells/ultrastructure , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation/genetics , Homeostasis/genetics , Lung/ultrastructure , Mice , Mice, Transgenic , Phosphatidylcholines/metabolism , Transforming Growth Factor alpha/genetics
12.
J Biol Chem ; 280(14): 13809-16, 2005 Apr 08.
Article in English | MEDLINE | ID: mdl-15668254

ABSTRACT

Foxa1 and Foxa2 are closely related family members of the Foxa group of transcription factors that are coexpressed in subsets of respiratory epithelial cells throughout lung morphogenesis. Shared patterns of expression, conservation of DNA binding, and transcriptional activation domains indicate that they may serve complementary functions in the regulation of gene expression during lung morphogenesis. Whereas branching morphogenesis of the fetal lung occurs normally in the Foxa2Delta/Delta and Foxa1-/- mice, deletion of both Foxa1 and Foxa2 (in Foxa2Delta/Delta, Foxa1-/- mice) inhibited cell proliferation, epithelial cell differentiation, and branching. Dilation of terminal lung tubules and decreased branching were observed as early as embryonic day 12.5. Foxa1 and Foxa2 regulated Shh (sonic hedgehog) and Shh-dependent genes in the respiratory epithelial cells that influenced the expression of genes in the pulmonary mesenchyme that are required for branching morphogenesis. Epithelial cell differentiation, as indicated by lack of expression of surfactant protein B, surfactant protein C, the Clara cell secretory protein, and Foxj1, was inhibited. Foxa family members regulate signaling and transcriptional programs required for morphogenesis and cell differentiation during formation of the lung.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Lung/embryology , Morphogenesis/physiology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Actins/metabolism , Animals , Cell Differentiation/physiology , DNA-Binding Proteins/genetics , Epithelial Cells/physiology , Epithelial Cells/ultrastructure , Glycoproteins/metabolism , Hedgehog Proteins , Hepatocyte Nuclear Factor 3-alpha , Hepatocyte Nuclear Factor 3-beta , Lung/anatomy & histology , Mice , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/genetics , Proto-Oncogene Proteins/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/embryology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Wnt Proteins
13.
Proc Natl Acad Sci U S A ; 101(40): 14449-54, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15452354

ABSTRACT

Toward the end of gestation in mammals, the fetal lung undergoes a process of differentiation that is required for transition to air breathing at birth. Respiratory epithelial cells synthesize the surfactant proteins and lipids that together form the pulmonary surfactant complex necessary for lung function. Failure of this process causes respiratory distress syndrome, a leading cause of perinatal death and morbidity in newborn infants. Here we demonstrate that expression of the forkhead gene Foxa2 in respiratory epithelial cells of the peripheral lung controls pulmonary maturation at birth. Newborn mice lacking Foxa2 expression in the lung develop severe pulmonary disease on the first day of life, with all of the morphological, molecular, and biochemical features of respiratory distress syndrome in preterm infants, including atelectasis, hyaline membranes, and the lack of pulmonary surfactant lipids and proteins. RNA microarray analysis at embryonic day 18.5 demonstrated that Foxa2-regulated expression of a group of genes mediating surfactant protein and lipid synthesis, host defense, and antioxidant production. Foxa2 regulates a complex pulmonary program of epithelial cell maturation required for transition to air breathing at birth.


Subject(s)
DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Parturition/physiology , Respiratory Physiological Phenomena , Transcription Factors/physiology , Animals , Animals, Newborn , Base Sequence , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Models, Animal , Fetus/physiology , Gene Expression Profiling , Hepatocyte Nuclear Factor 3-beta , Humans , Infant, Newborn , Lung/embryology , Lung/physiology , Lung/ultrastructure , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Electron , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Pulmonary Surfactants/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Respiratory Distress Syndrome, Newborn/etiology , Respiratory Distress Syndrome, Newborn/genetics , Respiratory Distress Syndrome, Newborn/physiopathology , Transcription Factors/deficiency , Transcription Factors/genetics
14.
Hum Pathol ; 35(5): 560-4, 2004 May.
Article in English | MEDLINE | ID: mdl-15138929

ABSTRACT

Forkhead box A2 (Foxa2) is a winged helix nuclear transcription protein that regulates the expression of genes that are critical to lung morphogenesis, differentiation, and function, including thyroid transcription factor-1, surfactant proteins, and Clara cell secretory protein. We examined the immunoreactivity of Foxa2 in paraffin sections of 75 lung tumors: 17 typical carcinoids, 2 atypical carcinoids, 4 large cell neuroendocrine (NE) carcinomas, 23 small cell carcinomas, 19 adenocarcinomas, 7 squamous cell carcinomas, and 3 (non-NE) large cell carcinomas, using a polyclonal rabbit Foxa2 antibody and a biotin-streptavidin detection system. In the adjacent lung, Foxa2 was detected in normal and hyperplastic type II cells. Foxa2 immunoreactivity was detected in 13 typical carcinoids (76%), 2 atypical carcinoids (100%), 2 large cell NE carcinomas (50%), 11 small cell carcinomas (48%), and 1 adenocarcinoma (5%). Squamous cell carcinomas and (non-NE) large cell carcinomas uniformly lacked Foxa2 staining. Expression of Foxa2 in the entire spectrum of NE lung tumors is another indication of differentiation shared by typical carcinoid, atypical carcinoid, large cell NE carcinoma, and small cell carcinoma.


Subject(s)
Biomarkers, Tumor/analysis , Lung Neoplasms/pathology , Neuroendocrine Tumors/pathology , Nuclear Proteins/biosynthesis , Transcription Factors/biosynthesis , Forkhead Transcription Factors , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Neuroendocrine Tumors/metabolism , Thyroid Nuclear Factor 1
15.
Development ; 131(4): 953-64, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14757645

ABSTRACT

The airways are lined by several distinct epithelial cells that play unique roles in pulmonary homeostasis; however, the mechanisms controlling their differentiation in health and disease are poorly understood. The winged helix transcription factor, FOXA2, is expressed in the foregut endoderm and in subsets of respiratory epithelial cells in the fetal and adult lung. Because targeted mutagenesis of the Foxa2 gene in mice is lethal before formation of the lung, its potential role in lung morphogenesis and homeostasis has not been determined. We selectively deleted Foxa2 in respiratory epithelial cells in the developing mouse lung. Airspace enlargement, goblet cell hyperplasia, increased mucin and neutrophilic infiltration were observed in lungs of the Foxa2-deleted mice. Experimental goblet cell hyperplasia caused by ovalbumin sensitization, interleukin 4 (IL4), IL13 and targeted deletion of the gene encoding surfactant protein C (SP-C), was associated with either absent or decreased expression of Foxa2 in airway epithelial cells. Analysis of lung tissue from patients with a variety of pulmonary diseases revealed a strong inverse correlation between FOXA2 and goblet cell hyperplasia. FOXA2 is required for alveolarization and regulates airway epithelial cell differentiation in the postnatal lung.


Subject(s)
DNA-Binding Proteins/metabolism , Goblet Cells/pathology , Nuclear Proteins/metabolism , Pulmonary Alveoli/embryology , Transcription Factors , Animals , DNA-Binding Proteins/genetics , Gene Deletion , Hepatocyte Nuclear Factor 3-beta , Humans , Immunohistochemistry , Interleukin-4/metabolism , Lung/abnormalities , Lung/embryology , Lung Diseases/genetics , Lung Diseases/metabolism , Mice , Mice, Inbred BALB C , Nuclear Proteins/genetics , Pulmonary Alveoli/abnormalities , STAT6 Transcription Factor , Trans-Activators/metabolism
16.
Am J Physiol Lung Cell Mol Physiol ; 285(6): L1246-54, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12909592

ABSTRACT

GATA-6, a member of a family of zinc finger transcription factors, is expressed in epithelial cells of the developing lung. To further assess the role of GATA-6 in lung morphogenesis, GATA-6 was expressed in respiratory epithelial cells of the developing mouse lung under control of the surfactant protein C promoter (hSP-CGATA-6 mice). Although GATA-6 did not alter lung morphology at embryonic day 18.5, defects in alveolar septation were observed early in the neonatal period, and air space enlargement persisted to adulthood. Airway resistance, airway elastance, tissue damping, and tissue elastance were significantly decreased, and lung volumes were significantly increased at 12 wk of age. Normal postnatal morphogenesis of the lung depends upon precise temporal-spatial regulation of GATA-6.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Pulmonary Alveoli/growth & development , Pulmonary Alveoli/physiology , Transcription Factors/genetics , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Division/physiology , DNA-Binding Proteins/metabolism , Female , GATA6 Transcription Factor , Humans , Mice , Mice, Transgenic , Pregnancy , Promoter Regions, Genetic , Pulmonary Alveoli/cytology , Pulmonary Circulation/physiology , Pulmonary Surfactant-Associated Protein C/genetics , RNA, Messenger/analysis , Respiratory Mucosa/cytology , Respiratory Mucosa/growth & development , Respiratory Mucosa/physiology , Transcription Factors/metabolism
17.
Am J Respir Crit Care Med ; 168(6): 685-91, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12829455

ABSTRACT

The pulmonary collectins surfactant protein (SP)-A and SP-D play important roles in innate lung defense, enhancing opsonization of microbes and limiting lung inflammatory responses. To quantify relationships among collectins, bacteria, and inflammation in early cystic fibrosis (CF) airway secretions, bronchoalveolar lavage fluids (BALFs) were collected from children undergoing clinically indicated bronchoscopy. Quantitative bacteriology, differential cell counts, and ELISA for SP-A and SP-D were assessed. Significantly increased numbers of neutrophils relative to bacteria were noted in BALF from CF compared with non-CF subjects. Although SP-A levels tended to be lower in CF compared with non-CF, this was only significant in the presence of bacterial infection. Among CF patients, SP-A concentrations in BALF were inversely related to inflammation, bacterial colony-forming units per milliliter, and age. SP-D levels were significantly decreased in CF patients, and SP-D was rarely detectable in the presence of infection. Among CF patients, SP-D correlated inversely with inflammation and bacterial colony-forming units per milliliter, and there was decreased immunostaining of BALF cells for SP-D in CF. Immunohistochemistry of CF autopsy lung sections for SP-A and SP-D confirmed their paucity at sites of infection and inflammation. We conclude that relative collectin deficiency occurs early in CF airways and is inversely related to inflammation in CF airways.


Subject(s)
Cystic Fibrosis/diagnosis , Pneumonia/diagnosis , Pulmonary Surfactant-Associated Protein A/analysis , Pulmonary Surfactant-Associated Protein D/analysis , Analysis of Variance , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Bronchoscopy , Child , Child, Preschool , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Infant , Inflammation Mediators/analysis , Male , Pneumonia/pathology , Probability , Prognosis , Sampling Studies , Sensitivity and Specificity , Severity of Illness Index
18.
J Biol Chem ; 278(16): 14291-8, 2003 Apr 18.
Article in English | MEDLINE | ID: mdl-12519727

ABSTRACT

SP-C-deficient (SP-C -/-) mice developed a severe pulmonary disorder associated with emphysema, monocytic infiltrates, epithelial cell dysplasia, and atypical accumulations of intracellular lipids in type II epithelial cells and alveolar macrophages. Whereas alveolar and tissue surfactant phospholipid pools were increased, levels of other surfactant proteins were not altered (SP-B) or were modestly increased (SP-A and SP-D). Analysis of pressure-volume curves and forced oscillatory dynamics demonstrated abnormal respiratory mechanics typical of emphysema. Lung disease was progressive, causing weight loss and cardiomegaly. Extensive alveolar remodeling was accompanied by type II cell hyperplasia, obliteration of pulmonary capillaries, and widespread expression of alpha-smooth muscle actin, indicating myofibroblast transformation in the lung parenchyma. Dysplastic epithelial cells lining conducting airways stained intensely for the mucin, MUC5A/C. Tissue concentrations of proinflammatory cytokines were not substantially altered in the SP-C (-/-) mice. Production of matrix metalloproteinases (MMP-2 and MMP-9) was increased in alveolar macrophages from SP-C (-/-) mice. Absence of SP-C caused a severe progressive pulmonary disorder with histologic features consistent with interstitial pneumonitis.


Subject(s)
Emphysema/genetics , Peptides/genetics , Peptides/physiology , Pneumonia/genetics , Animals , Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins , Lung/metabolism , Lung/pathology , Lung/ultrastructure , Macrophages/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 3/metabolism , Mice , Microscopy, Electron , Phospholipids/metabolism , Pressure , Protein Precursors/metabolism , Proteolipids/metabolism , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein C , Pulmonary Surfactant-Associated Protein D/metabolism , Pulmonary Surfactants
19.
J Histochem Cytochem ; 50(5): 651-60, 2002 May.
Article in English | MEDLINE | ID: mdl-11967276

ABSTRACT

Immunoreactive surfactant protein-D (SP-D) was assessed in human fetal, newborn, and adult tissues. In the fetal lung, SP-D was detected on airway surfaces by 10 weeks' gestation, staining increasing in the distal airways, decreasing in the proximal conducting airways with advancing gestation. In lungs from near-term infants and adults, SP-D was detected in Type II cells, serous cells of tracheobronchial glands, and subsets of cells lining peripheral airways. Immunostaining was decreased or absent in areas of lungs of neonates after injury to Type II cells, infection, or hemorrhage and was decreased in collapsed or unseptated airways from older infants with bronchopulmonary dysplasia. SP-D was also detected in many organs at all ages. SP-D was readily detected in epithelial cells and luminal material in lacrimal glands, salivary glands, pancreas, bile ducts, renal tubules, esophageal muscle and glands, parietal cells of the stomach, crypts of Lieberkuhn, sebaceous and eccrine sweat glands, Von Ebner's glands, endocervical glands, seminal vesicles, adrenal cortex, myocardium, and anterior pituitary gland. SP-D is a widely distributed member of the "collectin" family of polypeptides secreted onto luminal surfaces by epithelial cells lining ducts of many organs, where it likely plays a role in innate host defense.


Subject(s)
Aging/metabolism , Glycoproteins/metabolism , Pulmonary Surfactants/metabolism , Adult , Animals , Fetus/metabolism , Glycoproteins/immunology , Humans , Immune Sera , Immunohistochemistry , Infant, Newborn , Lung/embryology , Lung/growth & development , Lung/metabolism , Mice , Organ Specificity , Pulmonary Surfactant-Associated Protein D , Pulmonary Surfactants/immunology , Retrospective Studies
20.
J Biol Chem ; 277(25): 22743-9, 2002 Jun 21.
Article in English | MEDLINE | ID: mdl-11927601

ABSTRACT

The structure and functions of the airways of the lung change dramatically along their lengths. Large-diameter conducting airways are supported by cartilaginous rings and smooth muscle tissue and are lined by ciliated and secretory epithelial cells that are involved in mucociliary clearance. Smaller peripheral airways formed during branching morphogenesis are lined by cuboidal and squamous cells that facilitate gas exchange to a network of fine capillaries. The factors that mediate formation of these changing cell types and structures along the length of the airways are unknown. We report here that conditional expression of fibroblast growth factor (FGF)-18 in epithelial cells of the developing lung caused the airway to adopt structural features of proximal airways. Peripheral lung tubules were markedly diminished in numbers, whereas the size and extent of conducting airways were increased. Abnormal smooth muscle and cartilage were found in the walls of expanded distal airways, which were accompanied by atypically large pulmonary blood vessels. Expression of proteins normally expressed in peripheral lung tubules, including SP-B and pro-SP-C, was inhibited. FGF-18 mRNA was detected in normal mouse lung in stromal cells surrounding proximal airway cartilage and in peripheral lung mesenchyme. Effects were unique to FGF-18 because expression of other members of the FGF family had different consequences. These data show that FGF-18 is capable of enhancing proximal and inhibiting peripheral programs during lung morphogenesis.


Subject(s)
Fibroblast Growth Factors/metabolism , Lung/embryology , Lung/pathology , Animals , Cartilage/metabolism , Crosses, Genetic , DNA, Complementary/metabolism , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/physiology , Immunohistochemistry , In Situ Hybridization , Lung/ultrastructure , Mice , Mice, Transgenic , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Protein Binding , Protein Isoforms , Proteolipids/metabolism , Pulmonary Surfactants/metabolism , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...