Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Physiol Genomics ; 43(21): 1226-40, 2011 Nov 07.
Article in English | MEDLINE | ID: mdl-21878612

ABSTRACT

Better understanding alveolarization mechanisms could help improve prevention and treatment of diseases characterized by reduced alveolar number. Although signaling through fibroblast growth factor (FGF) receptors is essential for alveolarization, involved ligands are unidentified. FGF18, the expression of which peaks coincidentally with alveolar septation, is likely to be involved. Herein, a mouse model with inducible, lung-targeted FGF18 transgene was used to advance the onset of FGF18 expression peak, and genome-wide expression changes were determined by comparison with littermate controls. Quantitative RT-PCR was used to confirm expression changes of selected up- and downregulated genes and to determine their expression profiles in the course of lung postnatal development. This allowed identifying so-far unknown target genes of the factor, among which a number are known to be involved in alveolarization. The major target was adrenomedullin, a promoter of lung angiogenesis and alveolar development, whose transcript was increased 6.9-fold. Other genes involved in angiogenesis presented marked expression increases, including Wnt2 and cullin2. Although it appeared to favor cell migration notably through enhanced expression of Snai1/2, FGF18 also induced various changes consistent with prevention of epithelial-mesenchymal transition. Together with antifibrotic effects driven by induction of E prostanoid receptor 2 and repression of numerous myofibroblast markers, this could prevent alveolar septation-driving mechanisms from becoming excessive and deleterious. Last, FGF18 up- or downregulated genes of extracellular matrix components and epithelial cell markers previously shown to be up- or downregulated during alveolarization. These findings therefore argue for an involvement of FGF18 in the control of various developmental events during the alveolar stage.


Subject(s)
Fibroblast Growth Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Pulmonary Alveoli/growth & development , Animals , Animals, Newborn , Down-Regulation/drug effects , Down-Regulation/genetics , Doxycycline/pharmacology , Gene Expression Regulation, Developmental/drug effects , Mice , Oligonucleotide Array Sequence Analysis , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transgenes/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
2.
Am J Physiol Lung Cell Mol Physiol ; 298(6): L849-56, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20348277

ABSTRACT

Lung hypoplasia (LH) is a life-threatening congenital abnormality with various causes. It involves vascular bed underdevelopment with abnormal arterial muscularization leading to pulmonary hypertension. Because underlying molecular changes are imperfectly known and sometimes controversial, we determined key factors of angiogenesis along intrauterine development, focusing at the angiopoietin (ANG)/Tie-2 system. Lung specimens from medical terminations of pregnancy (9-37 wk) were used, including LH due to congenital diaphragmatic hernia (CDH) or other causes, and nonpulmonary disease samples were used as controls. ELISA determination indicated little ANG-1 change during pregnancy and no effect of LH, whereas Tie-2 declined similarly between 9 and 37 wk in LH and controls. By contrast, ANG-2 markedly increased in LH from 24 wk, whereas it remained stable in controls. Because VEGF increased also, this was interpreted as an attempt to overcome vascular underdevelopment. Hypothesizing that its inefficiency might be due to impaired downstream mechanism, endothelial nitric oxide synthase (eNOS) was determined by semiquantitative Western blot and found to be reduced by approximately 75%, mostly in the instance of CDH. In conclusion, angiogenesis remains defective in hypoplastic lungs despite reactive enhancement of VEGF and ANG-2 production, which could be due, at least in part, to insufficient eNOS expression.


Subject(s)
Angiopoietin-2/metabolism , Lung/blood supply , Nitric Oxide Synthase Type III/deficiency , Vascular Endothelial Growth Factor A/metabolism , Angiopoietin-1/metabolism , Female , Humans , Hypertension, Pulmonary , Lung/abnormalities , Lung/embryology , Neovascularization, Pathologic/physiopathology , Pregnancy , Receptor, TIE-2/metabolism
3.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L965-76, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19700645

ABSTRACT

Acute lung injury and compromised alveolar development characterize bronchopulmonary dysplasia (BPD) of the premature neonate. High levels of keratinocyte growth factor (KGF), a cell-cell mediator with pleiotrophic lung effects, are associated with low BPD risk. KGF decreases mortality in hyperoxia-exposed newborn rodents, a classic model of injury-induced impaired alveolarization, although the pulmonary mechanisms of this protection are poorly defined. These were explored through in vitro and in vivo approaches in the rat. Hyperoxia decreased by 30% the rate of wound closure of a monolayer of fetal alveolar epithelial cells, due to cell death, which was overcome by recombinant human KGF (100 ng/ml). In rat pups exposed to >95% O2 from birth, increased viability induced by intraperitoneal injection of KGF (2 microg/g body wt) every other day was associated with prevention of neutrophil influx in bronchoalveolar lavage (BAL), prevention of decreases in whole lung DNA content and cell proliferation rate, partial prevention of apoptosis increase, and a markedly increased proportion of surfactant protein B-immunoreactive cells in lung parenchyma. Increased lung antioxidant capacity is likely to be due in part to enhanced CAAT/enhancer binding protein alpha expression. By contrast, KGF neither corrected changes induced by hyperoxia in parameters of lung morphometry that clearly indicated impaired alveolarization nor had any significant effect on tissue or BAL surfactant phospholipids. These findings evidence KGF alveolar epithelial cell protection, enhancing effects on alveolar repair capacity, and anti-inflammatory effects in the injured neonatal lung that may account, at least in part, for its ability to reduce mortality. They argue in favor of a therapeutic potential of KGF in the injured neonatal lung.


Subject(s)
Fibroblast Growth Factor 7/pharmacology , Hyperoxia/pathology , Lung/drug effects , Lung/pathology , Animals , Animals, Newborn , Apoptosis/drug effects , Bronchoalveolar Lavage Fluid/cytology , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation/drug effects , DNA/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Phospholipids/metabolism , Pregnancy , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Pulmonary Surfactant-Associated Protein B/metabolism , Rats , Rats, Sprague-Dawley , Survival Analysis , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism
4.
Trends Mol Med ; 15(4): 169-79, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19303361

ABSTRACT

Bronchopulmonary dysplasia of the premature neonate and emphysema of the adult lung are common diseases that are characterized by increased airspace size and respiratory insufficiency and that presently lack efficient treatment. Although the former leads to impaired alveolar development and the latter to alveolar destruction, they have striking similarities in their pathophysiology, including the precipitating effect of oxidative stress, sustained inflammation, enhanced apoptosis, protease-antiprotease imbalance, elastic fiber deterioration and altered microvascularization. This review aims to comparatively analyze their molecular mechanisms to try identify common therapeutic targets. The recent discovery that alveolar developmental and maintenance programs share the same signal molecules and pathways, together with considerable increase in their understanding, have facilitated the development of common innovative strategies that have started to be tested in experimental models and pilot clinical studies.


Subject(s)
Bronchopulmonary Dysplasia/drug therapy , Bronchopulmonary Dysplasia/pathology , Pulmonary Emphysema/drug therapy , Pulmonary Emphysema/pathology , Animals , Apoptosis/physiology , Bronchopulmonary Dysplasia/metabolism , Cellular Senescence/physiology , Humans , Infant, Newborn , Inflammation/physiopathology , Oxidative Stress/physiology , Pulmonary Emphysema/metabolism
5.
Physiol Genomics ; 32(1): 128-41, 2007 Dec 19.
Article in English | MEDLINE | ID: mdl-17911382

ABSTRACT

Little is known about the molecular basis of lung alveolarization. We used a microarray profiling strategy to identify novel genes that may regulate the secondary septation process. Rat lung fibroblasts were extemporaneously isolated on postnatal days 2, 7, and 21, i.e., before, during, and after septation, respectively. Total RNA was extracted, and cRNAs were hybridized to Affymetrix rat genome 230 2.0 microarrays. Expression levels of a selection of genes were confirmed by real-time PCR. In addition to genes already known to be upregulated during alveolarization including drebrin, midkine, Fgfr3, and Fgfr4, the study allowed us to identify two remarkable groups of genes with opposite profiles, i.e., gathering genes either transiently up- or downregulated on day 7. The former group includes the transcription factors retinoic acid receptor (RXR)-gamma and homeobox (Hox) a2, a4, and a5 and genes involved in Wnt signaling (Wnt5a, Fzd1, and Ndp); the latter group includes the extracellular matrix components Comp and Opn and the signal molecule Slfn4. Profiling in whole lung from fetal life to adulthood confirmed that changes were specific for alveolarization. Two treatments that arrest septation, hyperoxia and dexamethasone, inhibited the expression of genes that are upregulated during alveolarization and conversely enhanced that of genes weakly expressed during alveolarization and upregulated thereafter. The possible roles of these genes in secondary septation are discussed. Gene expression profiling analysis on freshly isolated cells represents a powerful approach to provide new information about differential regulation of genes during alveolarization and pathways potentially involved in the pathogenesis of bronchopulmonary dysplasia.


Subject(s)
Gene Expression Profiling , Lung/physiology , Pulmonary Alveoli/physiology , Animals , Animals, Newborn , Dexamethasone/pharmacology , Female , Gene Expression Regulation/drug effects , Homeodomain Proteins/genetics , Lung/cytology , Lung/drug effects , Oligonucleotide Array Sequence Analysis , Pregnancy , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Rats
6.
PLoS Med ; 4(7): e237, 2007 Jul 31.
Article in English | MEDLINE | ID: mdl-17676984

ABSTRACT

BACKGROUND: Pulmonary hypoplasia and persistent pulmonary hypertension account for significant mortality and morbidity in neonates with congenital diaphragmatic hernia (CDH). Global lung immaturity and studies in animal models suggest the presence of surfactant deficiency that may further complicate the pathophysiology of CDH. However, data about surfactant status in human fetuses with CDH at birth are contradictory. The lack of a chronological study of surfactant content in late pregnancy has been a significant limitation. The appropriateness of administering surfactant supplements to neonates with CDH is therefore a debated question. METHODS AND FINDINGS: We investigated surfactant content in human fetuses with CDH compared to age-matched fetuses with nonpulmonary diseases used as controls. Concentrations of disaturated phosphatidylcholine and surfactant proteins were found to be similar at a given stage of pregnancy, with both components showing a similar pattern of increase with progressing pregnancy in fetuses with CDH and in control fetuses. Thyroid transcription factor 1, a critical regulator of surfactant protein transcription, similarly displayed no difference in abundance. Finally, we examined the expression of three glucocorticoid-regulated diffusible mediators involved in lung epithelial maturation, namely: keratinocyte growth factor (KGF), leptin, and neuregulin 1 beta 1 (NRG1-beta1). KGF expression decreased slightly with time in control fetuses, but remained unchanged in fetuses with CDH. Leptin and NRG1-beta1 similarly increased in late pregnancy in control and CDH lungs. These maturation factors were also determined in the sheep fetus with surgical diaphragmatic hernia, in which surfactant deficiency has been reported previously. In contrast to the findings in humans, surgical diaphragmatic hernia in the sheep fetus was associated with decreased KGF and neuregulin expression. Fetoscopic endoluminal tracheal occlusion performed in the sheep model to correct lung hypoplasia increased leptin expression, partially restored KGF expression, and fully restored neuregulin expression. CONCLUSIONS: Our results indicate that CDH does not impair surfactant storage in human fetuses. CDH lungs exhibited no trend toward a decrease in contents, or a delay in developmental changes for any of the studied surfactant components and surfactant maturation factors. Surfactant amounts are likely to be appropriate to lung size. These findings therefore do not support the use of surfactant therapy for infants with CDH. Moreover, they raise the question of the relevance of CDH animal models to explore lung biochemical maturity.


Subject(s)
Fetus/metabolism , Hernia, Diaphragmatic/metabolism , Pulmonary Surfactants/metabolism , Animals , Blotting, Western , Female , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Gene Expression Regulation, Developmental , Hernia, Diaphragmatic/genetics , Hernias, Diaphragmatic, Congenital , Humans , Infant, Newborn , Leptin/genetics , Leptin/metabolism , Lung/embryology , Lung/metabolism , Male , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuregulin-1 , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Sheep
7.
Pediatr Res ; 62(1): 20-5, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17515830

ABSTRACT

Lung development requires extracellular matrix remodeling. This involves matrix metalloproteinases (MMPs) and their endogenous inhibitors [tissue inhibitors of metalloproteinases (TIMPs)]. Because these have been generally studied only in whole lung, we focused specifically on mesenchymal and epithelial cells freshly isolated at various developmental stages. In fibroblasts, the most striking developmental change was a peak (fourfold the prenatal level) of membrane type 1 (MT1)-MMP transcript during alveolarization, consistent with the known crucial role of MT1-MMP in this process. TIMP-1 and -2 mRNAs transiently increased on postnatal d (pn) 3. In alveolar epithelial cells (AECs), MMP-2 expression was maximal on fetal d (f) 19 when alveolar type II cells (ATII) differentiate and on pn5; by contrast, MT1-MMP expression changed little and TIMP-1 expression decreased with advancing gestation. In cells expressing in vitro the ATI phenotype, TIMP-1 and -2 activities were nine- and fivefold those in cells expressing ATII features, respectively, whereas ATII presented higher MMP-2 activity and were the only cell type to express MMP-9. This indicates higher remodeling potential for ATII. Pulmonary mesenchymal and epithelial cells have therefore quite distinct MMP/TIMP expression patterns. Changes in cell compartments should be specifically documented in developing lung diseases such as bronchopulmonary dysplasia in which changes in MMP activities have been reported.


Subject(s)
Epithelial Cells/metabolism , Isoenzymes/metabolism , Lung , Matrix Metalloproteinases/metabolism , Mesoderm/cytology , Tissue Inhibitor of Metalloproteinases/metabolism , Animals , Cells, Cultured , Epithelial Cells/cytology , Extracellular Matrix/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Lung/cytology , Lung/embryology , Lung/metabolism , Matrix Metalloproteinases/genetics , Mesoderm/metabolism , Phenotype , Pregnancy , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinases/genetics
8.
Am J Respir Crit Care Med ; 175(10): 1066-77, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17303798

ABSTRACT

RATIONALE: Lung hypoplasia in congenital diaphragmatic hernia (CDH) seems to involve impaired alveolar septation. We hypothesized that disturbed deposition of elastin and expression of fibroblast growth factor 18 (FGF18), an elastogenesis stimulus, occurs in CDH. OBJECTIVES: To document FGF18 and elastin in human CDH and ovine surgical and rat nitrofen models and to use models to evaluate the benefit of treatments. METHODS: Human CDH and control lungs were collected post mortem. Diaphragmatic hernia was created in sheep at 85 days; fetal lungs were collected at 139 days (term = 145 days). Pregnant rats received nitrofen at 12 days; fetal lungs were collected at 21 days (term = 22 days). Some of the sheep fetuses with hernia underwent tracheal occlusion (TO); some of the nitrofen-treated pregnant rats received vitamin A. Both treatments are known to promote lung growth. MEASUREMENTS AND MAIN RESULTS: Coincidental with the onset of secondary septation, FGF18 protein increased threefold in control human lungs, which failed to occur in CDH. FGF18 labeling was found in interstitial cells of septa. Elastin staining demonstrated poor septation and markedly decreased elastin density in CDH lungs. Consistently, lung FGF18 transcripts were diminished 60 and 83% by CDH in sheep and rats, respectively, and elastin density and expression were diminished. TO and vitamin A restored FGF18 and elastin expression in sheep and rats, respectively. TO restored elastin density. CONCLUSIONS: Impaired septation in CDH is associated with decreased FGF18 expression and elastic fiber deposition. Simultaneous correction of FGF18 and elastin defects by TO and vitamin A suggests that defective elastogenesis may result, at least partly, from FGF18 deficiency.


Subject(s)
Elastin/deficiency , Fibroblast Growth Factors/deficiency , Hernia, Diaphragmatic/metabolism , Lung/abnormalities , Amino Acid Sequence , Animals , Base Sequence , Disease Models, Animal , Elastin/analysis , Elastin/genetics , Female , Fibroblast Growth Factors/analysis , Fibroblast Growth Factors/genetics , Hernia, Diaphragmatic/drug therapy , Hernia, Diaphragmatic/surgery , Humans , Infant , Infant, Newborn , Lung/chemistry , Lung/metabolism , Male , Molecular Sequence Data , Pulmonary Alveoli/abnormalities , Pulmonary Alveoli/chemistry , Pulmonary Alveoli/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sheep , Trachea/surgery , Vitamin A/therapeutic use
10.
Am J Respir Cell Mol Biol ; 34(6): 738-45, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16484688

ABSTRACT

Exposure of newborn rats to hyperoxia impairs alveolarization. Nitric oxide (NO) may prevent this evolution. Angiogenesis and factors involved in this process, but also other growth factors (GFs) involved in alveolar development, are likely potential therapeutic targets for NO. We studied the effects of the NO donor, [Z]-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)aminio]diazen-1-ium-1, 2-diolate, also termed DETANONOate (D-NO), on hyperoxia-induced changes in key regulatory factors of alveolar development in neonatal rats, and its possible preventive effect on the physiologic consequences of hyperoxia. Newborn rat pups were randomized at birth to hyperoxia (> 95% O2) or room air exposure for 6 or 10 d, while receiving D-NO or its diluent. On Day 6, several GFs and their receptors were studied at pre- and/or post-translational levels. Elastin transcript determination on Day 6, and elastin deposition in tissue and morphometric analysis of the lungs on Day 10, were also performed. Hyperoxia decreased the expression of vascular endothelial growth factor (VEGF) receptor (VEGFR) 2, fibroblast growth factor (FGF)-18, and FGF receptors (FGFRs) FGFR3 and FGFR4, increased mortality, and impaired alveolarization and capillary growth. D-NO treatment of hyperoxia-exposed pups restored the expression level of FGF18 and FGFR4, induced an increase of both VEGF mRNA and protein, enhanced elastin expression, and partially restored elastin deposition in alveolar walls. Although, under the present conditions, D-NO failed to prevent the physiologic consequences of hyperoxia in terms of survival and lung alveolarization, our findings demonstrate molecular effects of NO on GFs involved in alveolar development that may have contributed to the protective effects previously reported for NO.


Subject(s)
Gene Expression Regulation, Developmental , Hyperoxia/metabolism , Lung/metabolism , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Animals , Animals, Newborn , Elastin/genetics , Elastin/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hyperoxia/genetics , Hyperoxia/mortality , Lung/drug effects , Lung/growth & development , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
Am J Physiol Lung Cell Mol Physiol ; 288(1): L43-51, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15447937

ABSTRACT

The fibroblast growth factors (FGFs) are key players in fetal lung development, but little is known about their status in postnatal lung. Here, we investigated the expression pattern of FGF-18 transcripts through the perinatal period and evidenced a sevenfold increase after birth that paralleled changes in elastin expression. In vitro, recombinant human (rh)FGF-18 had a mitogenic activity on day 21 fetal rat lung fibroblasts and stimulated its own expression in the latter, whereas FGF-2 inhibited it. At 50 or 100 ng/ml, rhFGF-18 increased the expression of alpha-smooth muscle actin (alpha-SMA; 2.5-fold), a characteristic marker of myofibroblasts, of tropoelastin (6.5-fold), of lysyl oxidase (2-fold), and of fibulins 1 and 5 (8- and 2.2-fold) in confluent fibroblasts isolated from fetal day 21 lung; similar results were obtained with fibroblasts from day 3 postnatal lungs. Elastin protein expression was also slightly increased in fetal fibroblasts. Lung analysis on day 4 in rat pups that had received rhFGF-18 (3 microg) on days 0 and 1 showed a 1.7-fold increase of tropoelastin transcripts, whereas alpha-SMA transcripts were unchanged. In contrast, rhFGF-2 markedly decreased expression of elastin in vitro and in vivo and of fibulin 5 in vitro. In addition, vitamin A, which is known to enhance alveolar development, elevated FGF-18 and elastin expressions in day 2 lungs, thus advancing the biological increase. We postulate that FGF-18 is involved in postnatal lung development through stimulating myofibroblast proliferation and differentiation.


Subject(s)
Animals, Newborn , Elastin/biosynthesis , Fibroblast Growth Factors/metabolism , Fibroblasts/metabolism , Lung/metabolism , Myocytes, Smooth Muscle/metabolism , Actins/genetics , Actins/metabolism , Aging/metabolism , Animals , Animals, Newborn/growth & development , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Division/drug effects , Cells, Cultured , Elastin/genetics , Fetus/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacology , Fibroblasts/cytology , Lung/embryology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/cytology , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tropoelastin/genetics , Up-Regulation , Vitamin A/pharmacology
12.
Am J Physiol Lung Cell Mol Physiol ; 288(3): L562-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15557086

ABSTRACT

To determine whether prenatal surfactant storage was altered in a model of systemic arteriovenous fistula (SAVF) with pulmonary hypertension, a fistula was created between the internal jugular vein and the carotid artery in 120-day fetal lambs, and surfactant material was explored at 134 days. Total phospholipids (TPL) and disaturated phosphatidylcholine (DSPC) were increased in whole lung tissue. Phospholipid analysis of isolated lamellar body fraction evidenced a specific increase of surfactant pool size: TPL and DSPC in this fraction were enhanced 1.9 and 2.9 times, respectively, when referred to DNA. Although the steady-state level of transcripts of surfactant protein (SP)-A and SP-B was not found to be changed at the time of death, semiquantitative Western blot analysis revealed elevated SP-A and SP-B protein contents three- and twofold, respectively. These findings indicate markedly enhanced accumulation of surfactant material in the presence of surgically induced prenatal pulmonary hypertension. Although total lung cell number was increased by 26%, SP-B immunolabeling indicated that increased surfactant amount did not result from an increased alveolar type II cell proportion, but rather from an increased rate of storage. Whether similar changes in surfactant are encountered in human neonates with persistent pulmonary hypertension is worthy of investigation.


Subject(s)
Arteriovenous Fistula/complications , Fetal Diseases/metabolism , Hypertension, Pulmonary/metabolism , Phospholipids/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism , Animals , Arteriovenous Fistula/embryology , Blotting, Western , Carotid Arteries , DNA/metabolism , Disease Models, Animal , Fetal Diseases/pathology , Gestational Age , Hypertension, Pulmonary/embryology , Hypertension, Pulmonary/etiology , Jugular Veins , Lung/metabolism , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein B/metabolism , Sheep
13.
Am J Physiol Lung Cell Mol Physiol ; 286(6): L1293-301, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14742307

ABSTRACT

Previous investigations gained from in vivo or lung explant studies suggested that VEGF is an autocrine proliferation and maturation factor for developing alveolar type II cells. The objective of this work was to determine whether VEGF exerted its growth and maturation effects directly on isolated type II cells. These were isolated from 19-day fetal rat lung and cultured in defined medium. The presence of VEGF receptor-2 was assessed in cultured cells at the pre- and posttranslational levels. Recombinant VEGF(165), formerly found to be active on lung explants, failed to enhance type II cell proliferation estimated by thymidine and 5-bromo-2'-deoxy-uridine incorporation. It increased choline incorporation in saturated phosphatidylcholine by 27% but did not increase phospholipid surfactant pool size. VEGF (100 ng/ml) left unchanged the transcript level of surfactant proteins (SP)-A, SP-C, and SP-D but increased SP-B transcripts to four times the control steady-state level. VEGF slightly retarded, but did not prevent, the in vitro transdifferentiation of type II into type I cells, as assessed by immunolabeling of the type I cell marker T1alpha. We conclude that, with the exception of SP-B expression, which appears to be controlled directly, the previously observed effects of this VEGF isoform on type II cells are likely to be exerted indirectly through reciprocal paracrine interactions involving other lung cell types.


Subject(s)
Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Bromodeoxyuridine , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Fractionation , Cells, Cultured , Choline/pharmacokinetics , Endothelial Cells , Female , In Vitro Techniques , Phosphatidylcholines/metabolism , Pregnancy , Pulmonary Alveoli/embryology , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein C/genetics , Pulmonary Surfactant-Associated Protein D/genetics , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Respiratory Mucosa/embryology , Thymidine/pharmacokinetics , Tritium , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
Am J Respir Cell Mol Biol ; 29(5): 620-6, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14568882

ABSTRACT

Previous investigations have evidenced the importance of CCAAT/enhancer-binding proteins (C/EBPs) and peroxisome proliferator-activated receptor (PPAR)gamma for lung development, especially for alveolar type II cells (ATII). This prompted us to explore whether ATII maturation-promoting mediators controlled their expression in isolated ATII. In whole rat lung, C/EBPalpha, beta, delta, and PPARgamma mRNAs increased 3-5 times between gestational day 18 and term (Day 22), dropped around birth, then reincreased. C/EBPbeta and delta, but not PPARgamma, displayed similar profile in isolated ATII; C/EBPalpha transcript disappeared and the protein became hardly detectable in isolated cells. In cultured ATII, dexamethasone increased C/EBPbeta and PPARgamma mRNAs 2-4 times, and cyclic AMP increased C/EBPbeta and delta mRNAs approximately 1.5 times. Whereas retinoic acid increased C/EBPbeta and PPARgamma mRNAs 1.5 times in ATII in vitro, vitamin-A deficiency strongly decreased fetal lung C/EBPalpha, beta, and PPARgamma transcripts in vivo. C/EBPbeta, delta, and PPARgamma mRNAs were also increased in vitro by epidermal growth factor and keratinocyte growth factor, whereas they were unchanged by the maturation inhibitor transforming growth factor-beta. C/EBPalpha expression was not reinduced by any mediator. Changes in transcripts were reflected in protein levels analyzed through Western blotting. These results argue for a role of these factors in ATII functional maturation, and indicate a multifactorial control of their ontogeny.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Lung/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Epithelium/metabolism , Female , Gene Expression Profiling , Male , Rats , Vitamin A Deficiency/metabolism
15.
Pediatr Res ; 53(2): 231-9, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538780

ABSTRACT

Keratinocyte growth factor (KGF, or fibroblast growth factor 7) was previously reported to enhance the synthesis of surfactant in alveolar type II cells. We investigated the possible interactions between KGF and a glucocorticoid, dexamethasone (Dex), on surfactant protein (SP) gene expression. In cultured fetal rat type II cells, KGF and Dex induced greater-than-additive stimulating effects on SP-A and SP-B expressions that were enhanced three-fold and 30-fold, respectively, but had only additive effects on SP-C expression. Using murine lung epithelial (MLE) cells, KGF increased SP-A, SP-B (up to two-fold), and SP-C (up to three-fold) mRNA levels in a dose-dependent way. Dex 10(-9) to 10(-7) M increased SP-A and SP-B mRNA 1.5-fold and SP-C mRNA two-fold. Consistent with type II cell findings, simultaneous treatment by KGF and Dex induced a synergistic increase of SP-A and SP-B transcripts (three-fold and 4.5-fold, respectively), but not of SP-C transcripts. SP-A protein was present in MLE-15 and was increased about three-fold by KGF plus Dex. Expression study of a reporter gene placed under either the SP-A or the SP-B gene regulatory sequences and transfected in MLE-15 cells indicated that the Dex-KGF synergy was achieved mainly through a transcriptional effect for SP-A, and both transcriptional and nontranscriptional effects for SP-B. For the latter, increased mRNA stability was evidenced with the aid of actinomycin D. The Dex-KGF synergy may have potential interest for diseases associated with surfactant deficiency.


Subject(s)
Dexamethasone/pharmacology , Fibroblast Growth Factors/metabolism , Gene Expression Regulation/drug effects , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein B/genetics , Animals , Blotting, Western/methods , Cell Line , Cell Line, Transformed , Cells, Cultured , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Drug Synergism , Epithelial Cells , Fetus , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/physiology , Gene Expression Regulation/physiology , Genes, Reporter/drug effects , Genes, Reporter/genetics , Mice , Molecular Weight , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/embryology , Pulmonary Surfactant-Associated Protein A/biosynthesis , Pulmonary Surfactant-Associated Protein A/immunology , Pulmonary Surfactant-Associated Protein C/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/biosynthesis , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...