Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
iScience ; 25(2): 103797, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35198866

ABSTRACT

Metabolism is vital to cellular function and tissue homeostasis during human lung development. In utero, embryonic pluripotent stem cells undergo endodermal differentiation toward a lung progenitor cell fate that can be mimicked in vitro using induced human pluripotent stem cells (hiPSCs) to study genetic mutations. To identify differences between wild-type and surfactant protein B (SFTPB)-deficient cell lines during endoderm specification toward lung, we used an untargeted metabolomics approach to evaluate the developmental changes in metabolites. We found that the metabolites most enriched during the differentiation from pluripotent stem cell to lung progenitor cell, regardless of cell line, were sphingomyelins and phosphatidylcholines, two important lipid classes in lung development. The SFTPB mutation had no metabolic impact on early endodermal lung development. The identified metabolite signatures during lung progenitor cell differentiation may be utilized as biomarkers for normal embryonic lung development.

2.
Sci Rep ; 9(1): 13450, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31530844

ABSTRACT

Surfactant protein B (SFTPB) deficiency is a fatal disease affecting newborn infants. Surfactant is produced by alveolar type II cells which can be differentiated in vitro from patient specific induced pluripotent stem cell (iPSC)-derived lung organoids. Here we show the differentiation of patient specific iPSCs derived from a patient with SFTPB deficiency into lung organoids with mesenchymal and epithelial cell populations from both the proximal and distal portions of the human lung. We alter the deficiency by infecting the SFTPB deficient iPSCs with a lentivirus carrying the wild type SFTPB gene. After differentiating the mutant and corrected cells into lung organoids, we show expression of SFTPB mRNA during endodermal and organoid differentiation but the protein product only after organoid differentiation. We also show the presence of normal lamellar bodies and the secretion of surfactant into the cell culture medium in the organoids of lentiviral infected cells. These findings suggest that a lethal lung disease can be targeted and corrected in a human lung organoid model in vitro.


Subject(s)
Genetic Therapy/methods , Induced Pluripotent Stem Cells/cytology , Lung/cytology , Pulmonary Alveolar Proteinosis/congenital , Pulmonary Surfactant-Associated Protein B/deficiency , Cell Differentiation , Epithelial Cells/physiology , Fibroblasts/cytology , Genetic Markers , Green Fluorescent Proteins/genetics , Humans , Induced Pluripotent Stem Cells/transplantation , Lentivirus/genetics , Organoids , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/therapy , Pulmonary Alveoli/cytology , Pulmonary Surfactant-Associated Protein B/genetics
3.
Stem Cells Dev ; 24(1): 21-35, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25079436

ABSTRACT

Reciprocal signaling between the lung mesenchyme and epithelium is crucial for differentiation and branching morphogenesis. We hypothesized that the combination of signaling pathways comprising early epithelial-mesenchymal interactions and a 3D spatial environment are necessary for an efficient induction of embryonic and induced pluripotent stem cells (ESCs and iPSCs) into a lung cell phenotype with hallmarks of the distal niche. Aggregating early, but not late, embryonic lung mesenchyme with endoderm-induced mouse ESCs and iPSCs for 6 days resulted in organization into tubular structures and differentiation of the tubular lining cells to an NKX2-1(+)/SOX2(-)/SOX9(+)/proSFTPC(+) lineage. Over 80% of the endoderm-induced cells committed to an NKX2-1(+) lineage. Electron microscopy analysis demonstrated numerous multivesicular bodies and glycogen deposits in the tubular lining cells, characteristic features of type II epithelial cell progenitors. Using soluble FGFR2 receptor antagonists, we demonstrate that reciprocal fibroblast growth factor (FGF) 2, 7, and 10 signaling is essential for differentiation of endoderm-induced cells to an NKX2-1(+)/proSFTPC(+) phenotype within 3D aggregates. Only FGF2 was able to commit endoderm-induced cells in monolayer cultures to an NKX2-1(+) lineage, however with a significant lower efficiency (∼16%) than seen with mesenchyme. Thus, while FGF2 signaling alone can induce a primed population of ESCs and iPSCs, the cells do not differentiate to distal lung epithelial progenitors with the same efficiency and level of maturity that is achieved when the complex tissue and 3D environment of the developing lung is more accurately recapitulated.


Subject(s)
Cell Culture Techniques/methods , Epithelial Cells/metabolism , Fibroblast Growth Factor 2/metabolism , Induced Pluripotent Stem Cells/metabolism , Lung/metabolism , Signal Transduction , Animals , Antigens, Differentiation/biosynthesis , Cell Line , Epithelial Cells/cytology , Induced Pluripotent Stem Cells/cytology , Lung/cytology , Mice
4.
Am J Respir Cell Mol Biol ; 52(4): 448-58, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25180700

ABSTRACT

This study investigated whether hypoxia-inducible factor (HIF)-1 influences postnatal vascularization and alveologenesis in mice and whether stable (constitutive-active) HIF could prevent hyperoxia-induced lung injury. We assessed postnatal vessel and alveolar formation in transgenic mice, expressing a stable, constitutive-active, HIF1α-subunit (HIF-1αΔODD) in the distal lung epithelium. In addition, we compared lung function, histology, and morphometry of neonatal transgenic and wild-type mice subjected to hyperoxia. We found that postnatal lungs of HIF-1αΔODD mice had a greater peripheral vessel density and displayed advanced alveolarization compared with control lungs. Stable HIF-1α expression was associated with increased postnatal expression of angiogenic factors, including vascular endothelial growth factor, angiopoietins 1 and 2, Tie2, and Ephrin B2 and B4. Hyperoxia-exposed neonatal HIF-1αΔODD mice exhibited worse lung function but had similar histological and surfactant abnormalities compared with hyperoxia-exposed wild-type controls. In conclusion, expression of constitutive-active HIF-1α in the lung epithelium was associated with increased postnatal vessel growth via up-regulation of angiogenic factors. The increase in postnatal vasculature was accompanied by enhanced alveolar formation. However, stable HIF-1α expression in the distal lung did not prevent hyperoxia-induced lung injury in neonates but instead worsened lung function.


Subject(s)
Hyperoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Lung/metabolism , Pulmonary Alveoli/pathology , Animals , HEK293 Cells , Humans , Hyperoxia/pathology , Lung/pathology , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Alveoli/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
Am J Physiol Lung Cell Mol Physiol ; 305(11): L795-804, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23934924

ABSTRACT

Mechanical ventilation induces pulmonary apoptosis and inhibits alveolar development in preterm infants, but the molecular basis for the apoptotic injury is unknown. The objective was to determine the signaling mechanism(s) of ventilation (stretch)-induced apoptosis in newborn rat lung. Seven-day-old rats were ventilated with room air for 24 h using moderate tidal volumes (8.5 ml/kg). Isolated fetal rat lung epithelial and fibroblast cells were subjected to continuous cyclic stretch (5, 10, or 17% elongation) for up to 12 h. Prolonged ventilation significantly increased the number of apoptotic alveolar type II cells (i.e., terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling and anti-cleaved caspase-3 immunochemistry) and was associated with increased expression of the apoptotic mediator Fas ligand (FasL). Fetal lung epithelial cells, but not fibroblasts, subjected to maximal (i.e., 17%, but not lesser elongation) cyclic stretch exhibited increased apoptosis (i.e., nuclear fragmentation and DNA laddering), which appeared to be mediated via the extrinsic pathway (increased expression of FasL and cleaved caspase-3, -7, and -8). The intrinsic pathway appeared not to be involved [minimal mitochondrial membrane depolarization (JC-1 flow analysis) and no activation of caspase-9]. Universal caspases inhibition and neutralization of FasL abrogated the stretch-induced apoptosis. Prolonged mechanical ventilation induces apoptosis of alveolar type II cells in newborn rats and the mechanism appears to involve activation of the extrinsic death pathway via the FasL/Fas system.


Subject(s)
Alveolar Epithelial Cells/physiology , Apoptosis , Fas Ligand Protein/metabolism , Ventilator-Induced Lung Injury/metabolism , fas Receptor/metabolism , Animals , Biomechanical Phenomena , Caspases/metabolism , Cells, Cultured , Fibroblasts/physiology , Rats , Respiration, Artificial/adverse effects , Signal Transduction , Stress, Physiological , Ventilator-Induced Lung Injury/etiology , Ventilator-Induced Lung Injury/pathology
6.
Am J Respir Cell Mol Biol ; 49(4): 627-36, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23713442

ABSTRACT

The disruption of angiogenic pathways, whether through genetic predisposition or as a consequence of life-saving interventions, may underlie many pulmonary diseases of infancy, including bronchopulmonary dysplasia. Neuropilin-1 (Nrp1) is a transmembrane receptor that plays essential roles in normal and pathological vascular development and binds two distinct ligand families: vascular endothelial growth factor (Vegf) and class 3 semaphorins (Sema3). Although Nrp1 is critical for systemic vascular development, the importance of Nrp1 in pulmonary vascular morphogenesis is uncertain. We hypothesized that Sema3-Nrp1 and Vegf-Nrp1 interactions are important pathways in the orchestration of pulmonary vascular development during alveolarization. Complete ablation of Nrp1 signaling would therefore lead to interruption of normal angiogenic and vascular maturation processes that are relevant to the pathogenesis of bronchopulmonary dysplasia. We have previously shown that congenital loss of Sema3-Nrp1 signaling in transgenic Nrp1(Sema-) mice resulted in disrupted alveolar-capillary interface formation and high neonatal mortality. Here, pathohistological examination of Nrp1(Sema-) survivors in the alveolar period revealed moderate to severe respiratory distress, alveolar hemorrhaging, abnormally dilated capillaries, and disintegrating alveolar septa, demonstrating continued instability of the alveolar-capillary interface. Moreover, consistent with a reduced capillary density and consequent increases in vascular resistance, hypertensive remodeling was observed. In contrast, conditional Nrp1 deletion beginning at postnatal day 5 had only a transient effect upon alveolar and vascular development or pneumocyte differentiation despite an increase in mortality. Our results demonstrate that although Sema3-Nrp1 signaling is critical during fetal pulmonary development, Nrp1 signaling does not appear to be essential for alveolar development or vascular function in the postnatal period.


Subject(s)
Fetus/embryology , Neuropilin-1/genetics , Pulmonary Alveoli/embryology , Pulmonary Alveoli/metabolism , Semaphorins/genetics , Animals , Capillaries/metabolism , Endothelium, Vascular/metabolism , Female , Fetus/metabolism , Male , Mice , Neuropilin-1/metabolism , Pregnancy , Semaphorins/metabolism , Sequence Deletion/genetics , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Am J Pathol ; 180(1): 48-60, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22063298

ABSTRACT

Despite modern treatments, congenital diaphragmatic hernia (CDH) remains associated with variable survival and significant morbidity. The associated pulmonary hypoplasia is a major determinant of outcome. To develop better treatments, improved comprehension of the pathogenesis of lung hypoplasia is warranted. We developed an in vitro cell recombinant model to mimic pulmonary hypoplasia and specifically to investigate epithelial-mesenchymal interactions and to decipher which tissue layer is primarily defective in nitrofen-induced CDH-associated lung hypoplasia. Epithelial cells (E) and fibroblasts (F) were isolated from E19 control ((C)) and nitrofen-induced hypoplastic rat lungs ((N)). Cells were recombined and cultured as either homotypic [(F(C))(E(C)) and (F(N))(E(N))] or heterotypic [(F(C))(E(N)) and (F(N))(E(C))] recombinants. Recombinants containing F(N) fibroblasts had a thickened fibroblast tissue layer and there were fewer organized alveolar-like epithelial structures compared with those in control (F(C))(E(C)) recombinants. These F(N) recombinants exhibited a decrease in terminal deoxynucleotidyl transferase dUTP nick end labeling and cleaved caspase-3 positive cells. Cell proliferation was arrested in recombinants containing F(N) fibroblasts, which also exhibited increased p27(Kip1) and p57(Kip2) expression. In conclusion, fibroblasts, and not epithelial cells, appear to be the defective cell type in nitrofen-induced hypoplastic lungs due to a decreased ability to undergo apoptosis and maintain overall proliferation. This may explain the characteristic pulmonary interstitial thickening and hypoplasia observed in both nitrofen-induced hypoplastic lungs as well as human hypoplastic CDH lungs.


Subject(s)
Epithelial Cells/pathology , Fibroblasts/pathology , Hernias, Diaphragmatic, Congenital , Lung/abnormalities , Animals , Apoptosis/physiology , Cell Differentiation , Cell Proliferation , Epithelial-Mesenchymal Transition/physiology , Female , Hernia, Diaphragmatic/pathology , Immunohistochemistry , Mesoderm/pathology , Phenyl Ethers/toxicity , Pregnancy , Rats , Rats, Sprague-Dawley , Teratogens/toxicity
8.
Am J Physiol Lung Cell Mol Physiol ; 298(5): L670-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20118299

ABSTRACT

Prenatal exposures to immunogenic stimuli, such as bacterial LPS, have shown to influence the neonatal immune system and lung function. However, no detailed analysis of the immunomodulatory effects of LPS on postnatal T helper cell differentiation has been performed. Using a rat model, we investigated the effect of prenatal LPS exposure on postnatal T cell differentiation and experimental allergic airway disease. Pregnant rats were injected with LPS on day 20 and 21 (term = 22 days). Some of the offspring were sensitized and challenged with ovalbumin. Positive control animals were placebo exposed to saline instead of LPS, whereas negative controls were sensitized with saline. Expression of T cell-related transcription factors and cytokines was quantified in the lung, and airway hyperresponsiveness was measured. Prenatal LPS exposure induced a T helper 1 (T(H)1) immune milieu in the offspring of rats [i.e., increased T-bet and T(H)1 cytokine expression while expression of T(H)2-associated transcription factors (GATA3 and STAT6) and cytokines was decreased]. Prenatal LPS exposure did not trigger T(H)17 cell differentiation in the offspring. Furthermore, prenatal LPS exposure reduced ovalbumin-induced (T(H)2-mediated) airway inflammation, eosinophilia, and airway responsiveness. Thus, in utero exposure to endotoxin promotes a T(H)1 immune environment, which suppresses the development of allergic airway disease later in life.


Subject(s)
Asthma/prevention & control , Lipopolysaccharides/administration & dosage , Maternal-Fetal Exchange/immunology , Animals , Antigens/administration & dosage , Asthma/genetics , Asthma/immunology , Asthma/pathology , Cytokines/genetics , Disease Models, Animal , Female , Gene Expression , Lung/immunology , Lung/pathology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Pregnancy , Rats , Rats, Wistar , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/prevention & control , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology
9.
Am J Respir Cell Mol Biol ; 43(1): 74-87, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19684306

ABSTRACT

CTP:phosphocholine cytidylyltransferase (CCTalpha) plays a key role in the biosynthesis of surfactant phosphatidylcholine. In this study, we investigated the role of its membrane-binding (M) domain in modulating its structure, function, and cellular distribution. Multiple enhanced green fluorescent protein-CCTalpha constructs were generated to evaluate the subcellular distribution in A549 cells. The M domain targeted CCTalpha to the perinuclear (membrane-rich) region. Microinjections with glutathione-S-transferase fusion protein containing the M domain corroborated the perinuclear targeting. Deletion of the M domain or substitutions of the hydrophobic residues with arginine/serine in the VEEKS(267-277) motif of the M domain resulted in a nuclear appearance and indented nuclei. Membrane binding of CCTalpha decreased gradually as the number of positively charged arginine residues increased in the VEEKS motif. To identify whether membrane-protein interactions cause structural alterations in CCTalpha, we visualized the protein in the absence and presence of lipids by transmission electron microscopy. These studies revealed that CCTalpha forms a dimer-like complex that condenses upon binding to lipid vesicles, but not lipid monolayers. The influence of the M domain on CCTalpha activity was assessed in transgenic mice overexpressing the N-terminal catalytic domain (CCTalpha(1-239)), N-terminal catalytic plus M domain (CCTalpha(1-290)), or full-length CCTalpha(1-367) in fetal type II cells by using the surfactant protein C promoter. Only overexpression of CCTalpha(1-367) increased surfactant phosphatidylcholine synthesis. Thus, the M domain influences membrane binding, cellular distribution, and topology of CCTalpha, but the domain alone is not sufficient to confer CCT activity in alveolar type II cells in vivo.


Subject(s)
Choline-Phosphate Cytidylyltransferase/genetics , Choline-Phosphate Cytidylyltransferase/physiology , Lung/cytology , Pulmonary Alveoli/metabolism , Animals , Catalytic Domain , Cell Line, Tumor , Humans , Lipids/chemistry , Mice , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Mutation , Phosphatidylcholines/chemistry , Protein Structure, Tertiary , Rats , Structure-Activity Relationship
10.
Am J Physiol Lung Cell Mol Physiol ; 296(5): L726-37, 2009 May.
Article in English | MEDLINE | ID: mdl-19218354

ABSTRACT

Maternal bacterial infections adversely affect lung development by crossing the placental barrier and infecting the developing fetus. The underlying mechanism negatively affecting pulmonary development remains unknown. Herein, we investigated whether a systemic maternal infection affects postnatal inflammation and alveolar development. Pregnant rats were injected with 2.5 mg/kg LPS on day 20 and 21 (term = 22 days). Postnatal (PN0-21) mRNA and protein expression of cytokines (IL-1beta, IL-6, IL-10, CXCL1/2, TNFalpha) and genes implicated in alveologenesis [tropoelastin, lysyl oxidase (LOX), lysyl oxidase-like (LOXL)1, tenascin-C (TNC), fibulin 5, vascular endothelial growth factor (VEGF-A), VEGF receptor (VEGFR)2, VEGFR1, platelet-derived growth factor (PDGF)A, PDGFB, and PDGFRalpha] were quantified by real-time PCR and beadlyte technology. Lung transcript and protein levels of IL-1beta, IL-6, and CXCL1/2 were significantly greater in LPS-exposed pups than those of control pups at PN0, 2, 6, 10, and 14. Bronchoalveolar lavage fluid (BALF) of LPS-exposed animals contained significantly more macrophages at PN2 and 14 than BALF of control pups. Morphometric analysis revealed that LPS-exposed animals had fewer and larger alveoli, fewer secondary septa, and decreased peripheral vessel density when compared with control pups. This morphological delay in alveolar development disappeared after PN14. Tropoelastin, LOXL1, VEGF, VEGFR2, and PDGFRalpha mRNA expression of LPS-exposed animals was significantly greater than those of control animals in PN2-14 lungs. TNC, LOX, fibulin 5, VEGFR1, PDGFA, and PDGFB expression was not affected by maternal LPS exposure. Together, the data demonstrate that maternal exposure to endotoxin results in a prolonged pulmonary inflammation postnatally, altered gene expression of molecules implicated in alveologenesis, and delayed morphological maturation of the lung.


Subject(s)
Endotoxins/administration & dosage , Endotoxins/pharmacology , Organogenesis/drug effects , Prenatal Exposure Delayed Effects/pathology , Pulmonary Alveoli/embryology , Animals , Animals, Newborn , Body Weight/drug effects , Bronchoalveolar Lavage Fluid/cytology , Chemokines/genetics , Chemokines/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Developmental/drug effects , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Organ Size/drug effects , Placenta/drug effects , Placenta/metabolism , Placentation , Pregnancy , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Pulmonary Alveoli/growth & development , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Tropoelastin/genetics , Tropoelastin/metabolism
11.
Pediatr Res ; 61(2): 191-6, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17237721

ABSTRACT

Infection/inflammation and mechanical ventilation have both independently been shown to increase cytokine/chemokine levels in lung tissue and blood samples of premature patients. Little is known about the combined effect of systemic inflammation and mechanical ventilation on cytokine expression in the lung. We tested whether pre-existing inflammation induced by lipopolysaccharide (LPS) exposure would modify cytokine/chemokine response in newborn rat lungs to high tidal volume ventilation (HTVV). Newborn rats were randomly assigned to four groups: groups I and II (saline); groups III and IV: 3 mg/kg LPS. Groups II and IV were 24h later subjected to 3h of ventilation with a tidal volume of 25 mL/kg. HTVV alone increased IL-1beta, IL-6 and the chemokine (C-X-C motif) ligand 2 (CXCL2) mRNA expression. Although the cytokine response to LPS alone had disappeared after 24 h, the combination of LPS pretreatment and HTVV significantly increased the expression of IL-6 and IL-1beta mRNA when compared with HTVV alone. TNF-alpha expression was increased neither by HTVV alone nor in combination with LPS. IL-6 protein content in bronchoalveolar lavage increased due to the combined treatment. Thus, a subtle pre-existing inflammation combined with HTVV amplifies the proinflammatory cytokine/chemokine expression in the newborn rat lung compared with HTVV alone.


Subject(s)
Cytokines/metabolism , Lipopolysaccharides/pharmacology , Lung/growth & development , Lung/immunology , Respiration, Artificial , Animals , Animals, Newborn , Bronchoalveolar Lavage Fluid/chemistry , Cytokines/genetics , Lung/drug effects , Pneumonia/etiology , Pneumonia/immunology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Tidal Volume , Time Factors
12.
Am J Physiol Lung Cell Mol Physiol ; 292(3): L725-41, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17114281

ABSTRACT

Classical tissue recombination experiments have reported that at early gestation both tracheal and distal lung epithelium have the plasticity to respond to mesenchymal signals. Herein we examined the role of epithelial-mesenchymal interactions in maintaining epithelial differentiation at late (E19-E21, term = 22 days) fetal gestation in the rat. Isolated distal lung epithelial cells were recombined with mesenchymal cells from lung, skin, and intestine, and the homotypic or heterotypic recombinant cell aggregates were cultured for up to 5 days. Recombining lung epithelial cells with mesenchyme from various sources induced a morphological pattern that was specific to the type of inducing mesenchyme. In situ analysis of surfactant protein (SP)-C, SP-B, and Clara cell secretory protein (CCSP) expression, as well as SP-C and CCSP promoter transactivation experiments, revealed that distal lung epithelium requires lung mesenchyme to maintain the alveolar, but not bronchiolar, phenotype. Incubation of lung recombinants with an anti-FGF7 antibody resulted in a partial inhibition of mesenchyme-induced SP-C promoter transactivation. Immunoreactivity for Delta and Lunatic fringe, components of the Notch pathway that regulates cell differentiation, was downregulated in the heterotypic recombinants. In contrast, Hes1 mRNA expression was increased in these recombinants. Cumulatively, these results suggest that at late fetal gestation, distal lung epithelial cells are not fully committed to a specific phenotype and still have the plasticity to respond to various signals. Their alveolar phenotype is likely maintained by Notch/Notch ligand interactions and mesenchymal factors, including FGF7.


Subject(s)
Epithelial Cells/metabolism , Fetal Development , Lung/embryology , Mesoderm/metabolism , Animals , Cells, Cultured , Epithelial Cells/ultrastructure , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Immunoenzyme Techniques , In Situ Hybridization , Lung/cytology , Lung/ultrastructure , Male , Mesoderm/cytology , Mesoderm/ultrastructure , Phenotype , Pulmonary Surfactants/metabolism , RNA Probes , Rats , Rats, Wistar , Uteroglobin/genetics , Uteroglobin/metabolism
13.
J Biol Chem ; 279(53): 55946-57, 2004 Dec 31.
Article in English | MEDLINE | ID: mdl-15498769

ABSTRACT

CTP:phosphocholine cytidylyltransferase (CCT) is a rate-determining enzyme in the de novo synthesis of phosphatidylcholine (PtdCho). Alveolar type II cells synthesize large quantities of disaturated PtdCho, the surface-active agent of pulmonary surfactant, particularly at late gestation when the lung prepares itself for postnatal air breathing. To clarify the role of CCTalpha in lung surfactant maturation, we overexpressed CCTalpha(1-367) using the surfactant protein-C promoter. Lungs of transgenic mice were analyzed at day 18 of gestation (term = 19 days). Overexpression of CCTalpha(1-367) increased the synthesis and content of PtdCho in fetal type II cells isolated from the transgenic mice. Also, PtdCho content of fetal lung fluid was increased. No changes in surfactant protein content were detected. Interestingly, fetal type II cells of transgenic mice contained more glycogen than control cells. Incorporation studies with [U-(14)C]glucose demonstrated that overexpression of CCTalpha(1-367) in fetal type II cells increased glycogen synthesis without affecting glycogen breakdown. To determine which domain contributes to this glycogen phenotype, two additional transgenes were created overexpressing either CCTalpha(1-239) or CCTalpha(239-367). Glycogen synthesis and content were increased in fetal type II cells expressing CCTalpha(239-367) but not CCTalpha(1-239)(.) We conclude that overexpression of CCTalpha increases surfactant PtdCho synthesis without affecting surfactant protein levels but that it disrupts glycogen metabolism in differentiating type II cells via its regulatory domain.


Subject(s)
Choline-Phosphate Cytidylyltransferase/genetics , Choline-Phosphate Cytidylyltransferase/physiology , Glycogen/metabolism , Phosphatidylcholines/chemistry , Animals , Blotting, Western , Choline/metabolism , Genotype , Glucose/metabolism , Immunoblotting , Lasers , Lung/metabolism , Lung/pathology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Microscopy, Fluorescence , Models, Genetic , Phenotype , Phosphatidylcholines/metabolism , Promoter Regions, Genetic , Protein Conformation , Protein Structure, Tertiary , Pulmonary Surfactants/metabolism , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transgenes
14.
Am J Respir Cell Mol Biol ; 26(4): 506-15, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11919088

ABSTRACT

Pulmonary surfactant phosphatidylcholine (PC) formation increases as alveolar type II cells mature and arrest in G0/G1 state of the cell cycle at late fetal gestation. To determine whether this G0/G1 arrest is responsible for the increase in PC synthesis, we investigated the rates of PC synthesis and the activity, phosphorylation, intracellular distribution, synthesis, and degradation of a key enzyme of PC synthesis, cytidine triphosphate (CTP):phosphocholine cytidylyltransferase (CCTalpha). In synchronized mouse lung epithelial (MLE)-15 cells, PC production and CCTalpha activity peaked at G0/G1, declined during transition to G1/S, and remained low during S and G2/M. The changes in CCTalpha activity were not due to alterations in CCTalpha gene and protein expression. CCTalpha protein degradation also did not change during the cell cycle. Indirect immunofluorescence and immunogold electron microscopy revealed that CCTalpha localized to the cytoplasmic compartment and that its cytosolic localization did not change with the cell cycle. Although immunoblotting suggested no major redistribution of CCTalpha mass from cytosol to endoplasmic reticulum, activity measurements revealed that the ratio of particulate/soluble CCTalpha activity was cell cycle-dependent. The particulate/soluble ratio peaked at G0/G1 and declined with cell-cycle progression. Furthermore, the decrease in CCTalpha activity during exit from G0/G1 was associated with an increase in CCTalpha phosphorylation. These data suggest that the cell-cycle changes in PC synthesis are likely not due to alterations in CCTalpha expression and degradation but are primarily a consequence of changes in CCTalpha activity, phosphorylation, and membrane affinity.


Subject(s)
Cell Cycle/physiology , Choline-Phosphate Cytidylyltransferase/metabolism , Lung/cytology , Lung/metabolism , Phosphatidylcholines/biosynthesis , Animals , Cells, Cultured , Epithelial Cells/metabolism , G1 Phase/physiology , Mice , Phosphorylation , S Phase/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...