Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Am J Respir Cell Mol Biol ; 52(3): 295-303, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25054647

RESUMO

Bronchopulmonary dysplasia is a chronic lung disease of preterm infants characterized by arrested microvascularization and alveolarization. Studies show the importance of proangiogenic factors for alveolarization, but the importance of antiangiogenic factors is unknown. We proposed that hyperoxia increases the potent angiostatin, pigment epithelium-derived factor (PEDF), in neonatal lungs, inhibiting alveolarization and microvascularization. Wild-type (WT) and PEDF(-/-) mice were exposed to room air (RA) or 0.9 fraction of inspired oxygen from Postnatal Day 5 to 13. PEDF protein was increased in hyperoxic lungs compared with RA-exposed lungs (P < 0.05). In situ hybridization and immunofluorescence identified PEDF production primarily in alveolar epithelium. Hyperoxia reduced alveolarization in WT mice (P < 0.05) but not in PEDF(-/-) mice. WT hyperoxic mice had fewer platelet endothelial cell adhesion molecule (PECAM)-positive cells per alveolus (1.4 ± 0.4) than RA-exposed mice (4.3 ± 0.3; P < 0.05); this reduction was absent in hyperoxic PEDF(-/-) mice. The interactive regulation of lung microvascularization by vascular endothelial growth factor and PEDF was studied in vitro using MFLM-91U cells, a fetal mouse lung endothelial cell line. Vascular endothelial growth factor stimulation of proliferation, migration, and capillary tube formation was inhibited by PEDF. MFLM-91U cells exposed to conditioned medium (CM) from E17 fetal mouse lung type II (T2) cells cultured in 0.9 fraction of inspired oxygen formed fewer capillary tubes than CM from T2 cells cultured in RA (hyperoxia CM, 51 ± 10% of RA CM, P < 0.05), an effect abolished by PEDF antibody. We conclude that PEDF mediates reduced vasculogenesis and alveolarization in neonatal hyperoxia. Bronchopulmonary dysplasia likely results from an altered balance between pro- and antiangiogenic factors.


Assuntos
Animais Recém-Nascidos/metabolismo , Endotélio Vascular/metabolismo , Proteínas do Olho/metabolismo , Hiperóxia/metabolismo , Pulmão/metabolismo , Fatores de Crescimento Neural/metabolismo , Serpinas/metabolismo , Angiostatinas/metabolismo , Animais , Displasia Broncopulmonar/metabolismo , Linhagem Celular , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo
2.
BMC Dev Biol ; 13: 13, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23617334

RESUMO

BACKGROUND: MicroRNAs play important roles in regulating biological processes, including organ morphogenesis and maturation. However, little is known about specific pathways regulated by miRNA during lung development. Between the canalicular and saccular stages of the developing lung several important cellular events occur, including the onset of surfactant synthesis, microvascular remodeling and structural preparation for subsequent alveolarization. The miRNAs that are actively regulated, and the identity of their targets during this important developmental interval in the lung remain elusive. RESULTS: Using TLDA low density real-time PCR arrays, the expression of 376 miRNAs in male and female fetal mouse lungs of gestational days E15 - E18 were profiled. Statistical analyses identified 25 and 37 miRNAs that changed significantly between sexes and with gestation, respectively. In silico analysis using Ingenuity Pathway Analysis (IPA) identified specific pathways and networks known to be targets of these miRNAs which are important to lung development. Pathways that are targeted by sex regulated miRNAs include retinoin, IGFR1, Tp53 and Akt. Pathways targeted by gestation-regulated miRNAs include VEGFA and mediators of glucose metabolism. CONCLUSION: MiRNAs are differentially regulated across time and between sexes during the canalicular and saccular stages of lung development. Sex-associated differential miRNA expression may regulate the differences in structural and functional male and female lung development, as shown by networks generated using in silico analysis. These data provide a valuable resource to further enhance the understanding of miRNA control of lung development and maturation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Pulmão/embriologia , MicroRNAs/metabolismo , Animais , Feminino , Masculino , Camundongos , Organogênese , Gravidez , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Caracteres Sexuais
3.
Biochim Biophys Acta ; 1813(3): 480-91, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21195117

RESUMO

Maturation of pulmonary fetal type II cells to initiate adequate surfactant production is crucial for postnatal respiratory function. Little is known about specific mechanisms of signal transduction controlling type II cell maturation. The ErbB4 receptor and its ligand neuregulin (NRG) are critical for lung development. ErbB4 is cleaved at the cell membrane by the γ-secretase enzyme complex whose active component is either presenilin-1 (PSEN-1) or presenilin-2. ErbB4 cleavage releases the 80kDa intracellular domain (4ICD), which associates with chaperone proteins such as YAP (Yes-associated protein) and translocates to the nucleus to regulate gene expression. We hypothesized that PSEN-1 and YAP have a development-specific expression in fetal type II cells and are important for ErbB4 signaling in surfactant production. In primary fetal mouse E16, E17, and E18 type II cells, PSEN-1 and YAP expression increased at E17 and E18 over E16. Subcellular fractionation showed a strong cytosolic and a weaker membrane location of both PSEN-1 and YAP. This was enhanced by NRG stimulation. Co-immunoprecipitations showed ErbB4 associated separately with PSEN-1 and with YAP. Their association, phosphorylation, and co-localization were induced by NRG. Confocal immunofluorescence and nuclear fractionation confirmed these associations in a time-dependent manner after NRG stimulation. Primary ErbB4-deleted E17 type II cells were transfected with a mutant ErbB4 lacking the γ-secretase binding site. When compared to transfection with wild-type ErbB4, the stimulatory effect of NRG on surfactant protein mRNA expression was lost. We conclude that PSEN-1 and YAP have crucial roles in ErbB4 signal transduction during type II cell maturation.


Assuntos
Receptores ErbB/metabolismo , Feto/metabolismo , Pulmão/citologia , Pulmão/embriologia , Presenilina-1/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Proteínas de Ciclo Celular , Células Cultivadas , Receptores ErbB/análise , Feto/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Neurregulinas/metabolismo , Peptídeos/genética , Fosfoproteínas/análise , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Presenilina-1/análise , Presenilina-1/genética , Proteína C Associada a Surfactante Pulmonar , RNA Mensageiro/genética , Receptor ErbB-4 , Proteínas de Sinalização YAP
4.
Pediatr Pulmonol ; 57(10): 2327-2334, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35673713

RESUMO

OBJECTIVE: Noninvasive respiratory support has reduced the need for mechanical ventilation and surfactant administration in very premature neonates. We sought to determine how the increased use of noninvasive ventilation and less surfactant instillation has impacted the development of bronchopulmonary dysplasia (BPD) using four currently used BPD definitions. STUDY DESIGN: This is a retrospective, single-center cohort study of neonates born at less than 28 weeks gestation between 2010 and 2018. A respiratory practice change (less surfactant and more noninvasive ventilation) occurred in 2014 following participation in the Surfactant Positive Airway Pressure and Pulse Oximetry trial. Therefore, patients were divided into two epochs to compare postnatal respiratory and clinical course and BPD outcomes across four currently relevant definitions (Vermont Oxford Network, National Institute of Child Health and Human Development, Canadian, and Neonatal Research Network). RESULTS: Clinical and demographic variables were similar between epochs. Despite significant differences in maternal and infant characteristics and clinical course, the incidence of BPD was not significantly different between the two epochs regardless of the BPD definition utilized. There was a wide range in the incidence of BPD depending on the definition used. CONCLUSIONS: Despite decreased use of surfactant administration and invasive mechanical ventilation between the two epochs, the incidence of BPD did not change and there was wide variation depending on the definition used. A better understanding of the risk factors associated with BPD and a consensus definition is urgently needed to: 1) more accurately compare various studies, 2) help facilitate the conduct of clinical trials, and 3) enhance the development of novel therapeutic interventions to improve outcome.


Assuntos
Displasia Broncopulmonar , Surfactantes Pulmonares , Síndrome do Desconforto Respiratório do Recém-Nascido , Displasia Broncopulmonar/tratamento farmacológico , Displasia Broncopulmonar/terapia , Canadá/epidemiologia , Criança , Estudos de Coortes , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Surfactantes Pulmonares/uso terapêutico , Síndrome do Desconforto Respiratório do Recém-Nascido/terapia , Estudos Retrospectivos , Tensoativos
5.
Front Pediatr ; 9: 652038, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35096699

RESUMO

Background: Expressed breast milk (EBM) protein content is highly variable between mothers and often below published values that are still used for EBM protein fortification strategies. This approach may result in significant protein deficit and suboptimal protein energy (P/E) ratio. The study aim was to determine whether individualized EBM protein analysis and fortification will reduce preterm infant protein deficits and improve growth and neurodevelopmental outcome. Study Methods: In a single-center randomized, blinded study of infants born at 24 0/7-29 6/7 weeks, mother-specific protein values measured by a milk analyzer were used to individualize infant-specific protein intake (interventional group, IG), and compared this to a standardized protein fortification scheme based on published values of EBM protein content of 1.4 g/dL (control group, CG). For IG, milk analyzer protein values of mother's EBM were used to adjust protein content of the EBM. The CG EBM protein content was adjusted using the standard published value of 1.4 g/dL and not based on milk analyzer values. EBM protein content, protein intake, protein/energy (P/E) ratio, weight (WT), head circumference (HC), length (L), growth velocity (GV) from 2 to 6 weeks of age, WT, HC and L Z-Scores at 32- and 35-weeks PMA, and lean body mass (35 weeks PMA skin fold thickness) were measured. Neurodevelopment was assessed by Bayley III at average 24 months corrected gestational age (CGA). Results: EBM protein content before fortification was significantly below published values of 1.4 g/dL at all time points in both CG and IG. CG protein deficit was significantly decreased and progressively worsened throughout the study. Individualized protein fortification in IG avoided protein deficit and optimized P/E ratio. Although no significant change in short-term GV (at 6 weeks of age) was seen between groups, IG infants born at <27 weeks had significant improvements in WT and L z-scores, and leaner body mass at 32 and 35 weeks PMA. IG exhibited significantly improved cognitive scores at 24 months CGA. Conclusions: Infant-specific protein supplementation of mother's EBM optimized P/E ratio by eliminating protein deficit and improved growth z scores at 32- and 35-weeks PMA and neurocognitive testing at 24 months.

6.
Pediatr Res ; 68(1): 23-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20386491

RESUMO

Mice lacking aortic carboxypeptidase-like protein (ACLP) exhibit a gastroschisis (GS) like abdominal wall defect. The objectives of this study were to evaluate the pathophysiological features of GS in ACLP mice and to characterize the neuromuscular development of the eviscerated intestine (EI). ACLP mice were created by heterozygous mating from previously generated mice with targeted disruption of ACLP. Specimens were processed for H&E, and immunohistochemistry for smooth muscle cells [SMC, alpha-smooth muscle actin (alpha-SMA) antibody], interstitial cells of Cajal (ICC, c-kit-antibody), neural crest cells (NCC, Hox-b5-antibody), and enteric neurons (EN, PGP9.5-, alpha-internexin, and synaptophysin antibody). From 47 fetuses genotyped, 13 (27.7%) were wild type, 20 (42.5%) were heterozygous, and 14 (29.8%) were ACLP homozygous. In GS mice, expression of c-kit, Hox-b5, PGP-9.5, alpha-internexin, and synaptophysin were almost completely absent and only faint alpha-SMA expression was seen in the EI. In contrast, c-kit, Hox-b5, PGP9.5, alpha-internexin, synaptophysin, and alpha-SMA expression in intra-abdominal intestine in GS fetuses was the same as control intestine. The defect observed in ACLP mice closely resembles GS. Absence of ICC, NCC, EN, and immature differentiation of SMC supports an associated defect in neuromuscular development that is restricted to the EI.


Assuntos
Carboxipeptidases/metabolismo , Gastrosquise/patologia , Gastrosquise/fisiopatologia , Intestinos , Músculo Liso , Proteínas Repressoras/metabolismo , Animais , Biomarcadores/metabolismo , Carboxipeptidases/genética , Feminino , Feto/anatomia & histologia , Feto/patologia , Humanos , Células Intersticiais de Cajal/citologia , Células Intersticiais de Cajal/metabolismo , Intestinos/anormalidades , Intestinos/anatomia & histologia , Intestinos/embriologia , Camundongos , Camundongos Knockout , Músculo Liso/anormalidades , Músculo Liso/embriologia , Músculo Liso/inervação , Gravidez , Proteínas Repressoras/genética
7.
Am J Physiol Lung Cell Mol Physiol ; 297(1): L143-52, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19411307

RESUMO

In many organs, integrins and cadherins are partly regulated by Hox genes, but their interactions in airway morphogenesis and congenital lung diseases are unknown. We previously showed that the Hox protein HoxB5 is abnormally increased in bronchopulmonary sequestration (BPS) and congenital cystic adenomatoid malformation (CCAM), congenital lung lesions with abnormal airway branching. We now report on alpha(2)-, alpha(3)-, and beta(1)-integrin and E-cadherin expression in normal human lung and in BPS and CCAM tissue previously shown to have abnormal HoxB5 expression and on the relationship of cell adhesion molecule expression to Hoxb5 regulation. alpha(2)-, alpha(3)-, and beta(1)-integrins and E-cadherin expression in normal human lung and BPS and CCAM were evaluated using Western blot and immunohistochemistry. Fetal mouse lung fibroblasts with Hoxb5-specific siRNA downregulation were evaluated for alpha(2)-integrin protein levels by Western blot. Compared with normal human lung, a previously undetected alpha(2)-integrin isoform potentially lacking essential cytoplasmic sequences was significantly increased in BPS and CCAM, and alpha(2)-integrin spatial and cellular expression was more intense. E-cadherin protein levels were also significantly increased, whereas alpha(3) increased in CCAM compared with canalicular, but not with alveolar, stage lung. beta(1)-integrin levels were unchanged. We conclude that in BPS and CCAM, altered alpha(2)-integrin cytoplasmic signaling contributes to abnormal cellular behavior in these lung lesions. Aberrant cell adhesion molecule and Hox protein regulation are likely part of the mechanism involved in the development of BPS and CCAM.


Assuntos
Sequestro Broncopulmonar/metabolismo , Caderinas/metabolismo , Malformação Adenomatoide Cística Congênita do Pulmão/metabolismo , Integrinas/metabolismo , Animais , Western Blotting , Sequestro Broncopulmonar/patologia , Pré-Escolar , Malformação Adenomatoide Cística Congênita do Pulmão/patologia , Regulação para Baixo , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Proteínas de Homeodomínio/metabolismo , Humanos , Lactente , Recém-Nascido , Pulmão/citologia , Camundongos , Gravidez , Isoformas de Proteínas/metabolismo
8.
Neurosci Lett ; 430(2): 157-62, 2008 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-18063303

RESUMO

To investigate whether myelomeningocele (MMC) is associated with a global neuromuscular maldevelopment of the lower gastrointestinal (GI) tract and anorectum, the distribution and staining intensity of non-neuronal (alpha-smooth-muscle-actin), neural crest cell (NCC, [Hoxb5]), and neuronal markers (PGP-9.5, synaptophysin, neurotubulin-beta-III) within the distal colon, rectum, and anal sphincters were analyzed by immunohistochemistry and Western blot in rat fetuses with retinoic acid (RA) induced MMC. At term (E22), no gross-morphological differences of the anorectal unit of OIL (n=21) MMC (n=31), and RA-exposed-non MMC (RA, n=19) fetuses were found. Smooth muscle cells were evenly distributed within the muscle layers of the rectum and the internal anal sphincter in OIL, MMC, and RA fetuses. Density and staining intensity of NCC and mature enteric neurons within the myenteric plexus of the distal colon and rectum and innervation pattern within anal sphincters in MMC fetuses were analogous to RA and OIL controls. Normal smooth muscle and myenteric plexus development of the rectum and normal innervation of the anal sphincters and pelvic floor suggests that MMC is not associated with a global neuromuscular maldevelopment of lower GI structures in this short-gestational model.


Assuntos
Meningomielocele/patologia , Junção Neuromuscular/embriologia , Junção Neuromuscular/patologia , Tretinoína , Animais , Embrião de Mamíferos , Feminino , Proteínas de Homeodomínio/metabolismo , Intestino Grosso , Meningomielocele/induzido quimicamente , Meningomielocele/fisiopatologia , Músculo Liso/inervação , Músculo Liso/patologia , Gravidez , Ratos , Sinaptofisina/metabolismo , Tubulina (Proteína)/metabolismo , Ubiquitina Tiolesterase/metabolismo
9.
Birth Defects Res A Clin Mol Teratol ; 82(8): 571-84, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18553509

RESUMO

BACKGROUND: Hox transcription factors modulate signaling pathways controlling organ morphogenesis and maintain cell fate and differentiation in adults. Retinoid signaling, key in regulating Hox expression, is altered in pulmonary hypoplasia. Information on pattern-specific expression of Hox proteins in normal lung development and in pulmonary hypoplasia is minimal. Our objective was to determine how pulmonary hypoplasia alters temporal, spatial, and cellular expression of Hoxa5, Hoxb4, and Hoxb6 proteins compared to normal lung development. METHODS: Temporal, spatial, and cellular Hoxa5, Hoxb4, and Hoxb6 expression was studied in normal (untreated) and nitrofen-induced hypoplastic (NT-PH) lungs from gestational day 13.5, 16, and 19 fetuses and neonates using Western blot and immunohistochemistry. RESULTS: Modification of protein levels and spatial and cellular Hox expression patterns in NT-PH lungs was consistent with delayed lung development. Distinct protein isoforms were detected for each Hox protein. Expression levels of the Hoxa5 and Hoxb6 protein isoforms changed with development and were altered further in NT-PH lungs. Compared to normal lungs, GD19 and neonatal NT-PH lungs had decreased Hoxb6 and increased Hoxa5 and Hoxb4. Hoxa5 cellular localization changed from mesenchyme to epithelia earlier in normal lungs. Hoxb4 was expressed in mesenchyme and epithelial cells throughout development. Hoxb6 remained mainly in mesenchymal cells around distal airways. CONCLUSIONS: Unique spatial and cellular expression of Hoxa5, Hoxb4, and Hoxb6 participates in branching morphogenesis and terminal sac formation. Altered Hox protein temporal and cellular balance of expression either contributes to pulmonary hypoplasia or functions as a compensatory mechanism attempting to correct abnormal lung development and maturation in this condition.


Assuntos
Proteínas de Homeodomínio/metabolismo , Pulmão/anormalidades , Pulmão/crescimento & desenvolvimento , Fosfoproteínas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Feminino , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pneumopatias/induzido quimicamente , Pneumopatias/congênito , Pneumopatias/metabolismo , Camundongos , Éteres Fenílicos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Distribuição Tecidual/efeitos dos fármacos
10.
Front Pediatr ; 6: 125, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29951473

RESUMO

Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are common and significant morbidities of prematurely born infants. These diseases have in common altered and pathologic vascular formation in the face of incomplete organ development. Therefore, it is reasonable to question whether factors affecting angiogenesis could have a joint pathogenic role for both diseases. Inhibition or induced expression of a single angiogenic factor is unlikely to be 100% causative or protective of either of BPD or ROP. It is more likely that interactions of multiple factors leading to disordered angiogenesis are present, increasing the likelihood of common pathways in both diseases. This review explores this possibility by assessing the evidence showing involvement of specific angiogenic factors in the vascular development and maldevelopment in each disease. Theoretical interactions of specific factors mutually contributing to BPD and ROP are proposed and, where possible, a timeline of the proposed relationships between BPD and ROP is developed. It is hoped that future research will be inspired by the theories put forth in this review to enhance the understanding of the pathogenesis in both diseases.

11.
Front Biosci ; 12: 860-73, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17127343

RESUMO

Organ-specific patterning is partly determined by Hox gene regulatory interactions with the extracellular matrix (ECM), cell adhesion and fibroblast growth factor (FGFs) signaling pathways but coordination of these mechanisms in lung development is unknown. We have previously shown that Hoxb-5 affects airway patterning during lung morphogenesis. Hoxb-5 regulation in fetal lung affects ECM expression of tenascin-C and alters FGF10 spatial and cellular expression. To test this hypothesis, gestational day 13.5 (Gd13.5) fetal mouse lung fibroblasts and whole lungs were cultured with Hoxb-5-specific small interfering RNA (siRNA). Western blots showed that siRNA-down regulation of Hoxb-5 led to decreased tenascin-C and FGF10 and was associated with increased Hoxb-4 and decreased Hoxb-6 protein levels. Hoxa-5 protein levels were not affected. Hoxb-5 siRNA-treated whole lung cultures had a significant decrease in total lung and peripheral branching region surface area. Immunostaining showed negligible levels of Hoxb-5 protein and tenascin-C, and loss of FGF10 spatial restriction. We conclude that Hoxb-5 helps regulate lung airway development through modulation of ECM expression of tenascin-C. ECM changes induced by Hoxb-5 may affect mesenchymal-epithelial cell signaling to alter spatial and cellular restriction of FGF10. Hoxb-5 may also affect lung airway branching indirectly by cross regulation of other Hoxb genes.


Assuntos
Fator 10 de Crescimento de Fibroblastos/metabolismo , Proteínas de Homeodomínio/metabolismo , Pulmão/embriologia , Tenascina/metabolismo , Animais , Células Cultivadas , Regulação para Baixo , Feto , Fator 10 de Crescimento de Fibroblastos/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Pulmão/citologia , Pulmão/metabolismo , Camundongos , Técnicas de Cultura de Órgãos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Tenascina/genética
13.
J Cell Commun Signal ; 8(3): 231-44, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25073509

RESUMO

Hoxb5 and Hoxa5 transcription factor proteins uniquely impact lung morphogenesis at the developmental time point when extremely preterm infants are born. The effect of O2 exposure (0.4 FiO2) used in preterm infant care on these Hox proteins is unknown. We used ex vivo fetal mouse lung organ cultures to explore the effects of 0.4 FiO2 on lung airway and vascular formation in the context of Hoxb5 and Hoxa5 expression and regulation. Compared to room air, 48 h (h) 0.4 FiO2 adversely attenuated airway and microvasculature formation while reducing lung growth and epithelial cell volume, and increasing mesenchymal volume. 0.4 FiO2 decreased pro-angiogenic Hoxb5 and VEGFR2 while not altering protein levels of angiostatic Hoxa5. Lungs returned to RA after 24 h 0.4FiO2 had partial structural recovery but remained smaller and less developed. Mesenchymal cell apoptosis increased and proliferation decreased with time in O2 while epithelial cell proliferation significantly increased. Hoxb5 overexpression led to prominent peri-airway VEGFR2 expression and promoted lung vascular and airway patterning. Hoxa5 overexpression had the opposite effects. We conclude that 0.4 FiO2 exposure causes a profound loss of airway and lung microvascular development that occurs partially via reduction in pro-angiogenic Hoxb5 while angiostatic Hoxa5 expression is maintained.

14.
PLoS One ; 8(2): e55911, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23409087

RESUMO

Epithelial-mesenchymal interactions play a crucial role in branching morphogenesis, but very little is known about how endothelial cells contribute to this process. Here, we examined how anti-angiogenic miR-221 and pro-angiogenic miR-130a affect airway and vascular development in the fetal lungs. Lung-specific effects of miR-130a and miR-221 were studied in mouse E14 whole lungs cultured for 48 hours with anti-miRs or mimics to miR-130a and miR-221. Anti-miR 221 treated lungs had more distal branch generations with increased Hoxb5 and VEGFR2 around airways. Conversely, mimic 221 treated lungs had reduced airway branching, dilated airway tips and decreased Hoxb5 and VEGFR2 in mesenchyme. Anti-miR 130a treatment led to reduced airway branching with increased Hoxa5 and decreased VEGFR2 in the mesenchyme. Conversely, mimic 130a treated lungs had numerous finely arborized branches extending into central lung regions with diffusely localized Hoxa5 and increased VEGFR2 in the mesenchyme. Vascular morphology was analyzed by GSL-B4 (endothelial cell-specific lectin) immunofluorescence. Observed changes in airway morphology following miR-221 inhibition and miR-130a enhancement were mirrored by changes in vascular plexus formation around the terminal airways. Mouse fetal lung endothelial cells (MFLM-91U) were used to study microvascular cell behavior. Mimic 221 treatment resulted in reduced tube formation and cell migration, where as the reverse was observed with mimic 130a treatment. From these data, we conclude that miR-221 and miR-130a have opposing effects on airway and vascular morphogenesis of the developing lung.


Assuntos
Pulmão/embriologia , Pulmão/metabolismo , MicroRNAs/genética , Animais , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox , Pulmão/irrigação sanguínea , Camundongos , MicroRNAs/metabolismo , Morfogênese/genética , Neovascularização Fisiológica/genética
15.
Biomed Res Int ; 2013: 320249, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24078914

RESUMO

Androgens enhance airway branching but delay alveolar maturation contributing to increased respiratory morbidity in prematurely born male infants. Hoxb5 protein positively regulates airway branching in developing lung. In other organs, androgen regulation intersects with Hox proteins and TGF ß -SMAD signaling, but these interactions have not been studied in the lung. We hypothesized that androgen alteration of airway branching early in lung development requires Hoxb5 expression and that these androgen-Hoxb5 interactions occur partially through regional changes in TGF ß signaling. To evaluate acute effects of androgen and TGF ß on Hoxb5, E11 whole fetal mouse lungs were cultured with dihydrotestosterone (DHT) with/without Hoxb5 siRNA or TGF ß inhibitory antibody. Chronic in utero DHT exposure was accomplished by exposing pregnant mice to DHT (subcutaneous pellet) from E11 to E18. DHT's ability to enhance airway branching and alter phosphorylated SMAD2 cellular localization was partially dependent on Hoxb5. Hoxb5 inhibition also changed the cellular distribution of SMAD7 protein. Chronic in utero DHT increased Hoxb5 and altered SMAD7 mesenchymal localization. TGF ß inhibition enhanced airway branching, and Hoxb5 protein cellular localization was more diffuse. We conclude that DHT controls lung airway development partially through modulation of Hoxb5 protein expression and that this level of regulation involves interactions with TGF ß signaling.


Assuntos
Proteínas de Homeodomínio/metabolismo , Pulmão/embriologia , Pulmão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Androgênios , Animais , Di-Hidrotestosterona/farmacologia , Feminino , Feto/efeitos dos fármacos , Feto/metabolismo , Técnicas de Silenciamento de Genes , Pulmão/citologia , Pulmão/efeitos dos fármacos , Masculino , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Camundongos , Morfogênese/efeitos dos fármacos , Gravidez , Proteínas Smad/metabolismo
16.
Am J Physiol Lung Cell Mol Physiol ; 293(2): L429-35, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17545485

RESUMO

Insufficient fetal surfactant production leads to respiratory distress syndrome among preterm infants. Neuregulin signals the onset of fetal surfactant phospholipid synthesis through formation of erbB receptor dimers. We hypothesized that erbB4 downregulation in fetal type II epithelial cells will downregulate not only fetal surfactant phospholipid synthesis, but also affect proliferation and erbB receptor localization. We tested these hypotheses using small interfering RNA (siRNA) directed against the erbB4 gene to silence erbB4 receptor function in cultures of primary day 19 fetal rat lung type II cells. ErbB4 siRNA treatment inhibited erbB4 receptor protein expression, fibroblast-conditioned medium induced erbB4 phosphorylation, and fetal surfactant phospholipid synthesis. Cell proliferation, measured as thymidine incorporation, was also inhibited by erbB4 siRNA treatment. Downregulation of erbB4 receptor protein changed erbB1 localization at baseline and after stimulation, as determined by confocal microscopy and subcellular fractionation. We conclude that erbB4 is an important receptor in the control of fetal lung type II cell maturation.


Assuntos
Receptores ErbB/metabolismo , Fosfolipídeos/metabolismo , Surfactantes Pulmonares/metabolismo , Mucosa Respiratória/embriologia , Animais , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Colina/farmacocinética , Dimerização , Regulação para Baixo/fisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Receptores ErbB/química , Receptores ErbB/genética , Feminino , Pulmão/citologia , Pulmão/embriologia , Gravidez , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Receptor ErbB-4 , Mucosa Respiratória/citologia , Timidina/farmacocinética
17.
J Pediatr Surg ; 41(4): 774-80, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16567192

RESUMO

BACKGROUND/PURPOSE: Mesenchymal nuclear transcription factors (MNTF) are involved in lung development and maturation and regulate surfactant protein (SP) expression. Prolonged (>2 weeks) fetal tracheal occlusion (TO) has been shown to accelerate lung growth and inhibit pulmonary surfactant synthesis. The effects of TO on SP expression and MNTF, however, have not been formally assessed. The objectives of this study were to evaluate the effects of short-term (3 days) TO on normal lung growth and protein expression of pulmonary MNTF involved in SP synthesis. METHODS: At E19 (term, 22 days), 2 fetuses per time-dated Sprague-Dawley rats underwent either TO (n = 23) or a sham (n = 22) operation. Lungs were harvested 72 hours post surgery. Pulmonary SP-A; SP-B; SP-C messenger RNA (mRNA) expression; and SP-A and SP-B, Hoxb5, thyroid transcription factor 1, and retinoic X receptor-alpha protein expression were analyzed. RESULTS: Lung weight was significantly increased by TO (TO 0.32 +/- 0.02g vs SHAM 0.14 +/- 0.01 g; P < .001), resulting in 123% increase of the lung-to-body-weight ratio. No difference of SP-A-mRNA (177 +/- 4.3 TO vs 169 +/- 4.4 SHAM; P = .25), SP-B-mRNA (87.7 +/- 0.2 TO vs 87.4 +/- 0.02 SHAM; P = .33), and SP-C-mRNA (186.5 +/- 3.2 TO vs 183.2 +/- 2.7 SHAM; P = .45) expression was found. Surfactant protein A (175.6 +/- 25.3 TO vs 192.5 +/- 19.8 SHAM; P = .59) and SP-B (163.4 +/- 5.2 TO vs 166.8 +/- 9.3 SHAM; P = .75) protein expression were similar in both groups; however, Hoxb5 (70.3 +/- 18.9 TO vs 130.6 +/- 5.1 SHAM; P = .02) and thyroid transcription factor 1 (102.6 +/- 19 TO vs 181.1 +/- 6.3 SHAM; P = .007) expression were significantly decreased. Retinoic X receptor-alpha expression tended to be increased by TO (171.9 +/- 6.0 TO vs 155.4 +/- 6.7 SHAM; P = .06). CONCLUSIONS: Short-term TO late in gestation induces rapid lung growth. Surfactant protein-mRNA and protein expression are not significantly altered. Thyroid transcription factor 1 and Hoxb5 are down-regulated by TO, suggesting that duration and timing of occlusion are important in balancing the effects of TO on lung growth vs lung maturation.


Assuntos
Proteínas de Homeodomínio/biossíntese , Proteínas Nucleares/biossíntese , Proteínas Associadas a Surfactantes Pulmonares/biossíntese , Receptor X Retinoide alfa/biossíntese , Fatores de Transcrição/biossíntese , Animais , Ratos , Ratos Sprague-Dawley , Fator Nuclear 1 de Tireoide , Traqueia
18.
J Surg Res ; 108(2): 203-11, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12505043

RESUMO

BACKGROUND: Nitrofen-induced pulmonary hypoplasia (PH) with or without coexistent congenital diaphragmatic hernia (CDH) in animals mimics the human condition. We have demonstrated reduced steroid-thyroid-retinoid receptors in hypoplastic lungs. Therefore, we hypothesize that expression of two additional mesenchymally derived nuclear transcription factors, retinoid X receptor alpha (RXR-alpha) and homeobox b-5 (Hoxb-5), would also be altered in hypoplastic lungs. MATERIALS AND METHODS: We used timed-pregnant CD-1 mice either untreated or gavaged with nitrofen on gestational day (Gd) 8. Normal lungs were compared with hypoplastic lungs with severe left PH and CDH at Gd 14, 16, and 19 and from neonates. We performed immunoblotting for RXR-alpha and Hoxb-5 proteins and immunohistochemistry for Hoxb-5 protein. RESULTS: Almost 70-80% of nitrofen-exposed mice had no apparent external phenotypic abnormalities, such as craniofacial defects. Fetal body and lung weights were reduced. RXR-alpha and Hoxb-5 proteins were highest at Gd 14 and decreased as development progressed. Densitometric analysis of RXR-alpha or Hoxb-5 proteins in normal and hypoplastic lungs showed no significant difference; however, the immunolocalization pattern of Hoxb-5 protein differed. Hoxb-5 protein was primarily in mesenchymal cells of normal lungs on Gd 14; however, by Gd 19, it appeared to be mainly in the epithelial cells of prealveolar structures and in adjacent subepithelial mesenchymal cells. In hypoplastic lungs no significant changes were observed in Hoxb-5 staining in mesenchymal cells at Gd 14 nor at Gd 16; however, Hoxb-5 expression persisted in mesenchyme and epithelium at Gd 19 and in neonatal hypoplastic lungs, unlike normal lungs. CONCLUSIONS: (1) Unaltered RXR-alpha protein implies that despite altered retinoic acid receptors (RARs) in hypoplastic lungs, the mechanisms of nitrofen-induced PH may be independent of RXR-alpha pathways. (2) In hypoplastic lungs, the persistent mesenchymal expression of Hoxb-5, in later stages of development and at birth, suggests delayed development and maturation compared to normal lungs. We speculate that nitrofen induces PH via RAR-dependent but RXR-independent interactions, which may be downstream of the Hoxb-5 gene or may involve other more anteriorly expressed Hox genes.


Assuntos
Núcleo Celular/metabolismo , Pulmão/anormalidades , Mesoderma/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos/metabolismo , Anormalidades Congênitas/embriologia , Desenvolvimento Embrionário e Fetal/fisiologia , Idade Gestacional , Proteínas de Homeodomínio/metabolismo , Immunoblotting , Técnicas Imunológicas , Pulmão/efeitos dos fármacos , Pulmão/embriologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos , Éteres Fenílicos/farmacologia , Receptores do Ácido Retinoico/metabolismo , Valores de Referência , Receptores X de Retinoides , Coloração e Rotulagem
19.
Mol Genet Metab ; 80(1-2): 242-54, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14567974

RESUMO

We recently showed that T3 treatment of cultured gestational day 11.5 early pseudoglandular period mouse lungs, accelerated terminal airway development at the expense of decreased branching morphogenesis. As the ability of T3 to influence epithelial cell differentiation increases with advancing development, we hypothesized that in the late pseudoglandular period, T3 would cause further premature changes in the morphology of the distal airways leading to abnormal saccular development. Gestational day 13.5 embryonic mouse lungs were cultured for 3 and 7 days without or with added T3. Increasing T3 dose and time in culture resulted in progressive development of thin walled, abnormal saccules, an increase in cuboidal and flattened epithelia and airway space with a concomitant decrease in mesenchymal cell volume. Consistent with increased cuboidal and flattened epithelial cell volume identified by morphometry, immunostaining suggested increased cell proliferation detected by localization of proliferating cell nuclear antigen (PCNA) in epithelial cells of T3 treated lungs. T3 decreased mesenchymal expression of Hoxb-5 protein and caused progressive localization of Nkx2.1 and SP-C proteins to distal cuboidal epithelia of early abnormal saccules, evidence that T3 prematurely and abnormally advanced mesenchymal and epithelial cell differentiation. Western blot showed a T3-dependent decrease in Hoxb-5 and a trend towards decreased Nkx2.1 and SP-C, after 3 and 7 days of culture, respectively. We conclude that exogenous T3 treatment during the late pseudoglandular period prematurely and abnormally accelerates terminal saccular development. This may lead to abnormal mesenchymal and epithelial cell fate.


Assuntos
Diferenciação Celular/fisiologia , Células Epiteliais/citologia , Pulmão/embriologia , Morfogênese/fisiologia , Tri-Iodotironina/farmacologia , Animais , Divisão Celular/fisiologia , Embrião de Mamíferos , Feminino , Pulmão/citologia , Camundongos , Técnicas de Cultura de Órgãos , Gravidez , Antígeno Nuclear de Célula em Proliferação/metabolismo
20.
Am J Physiol Lung Cell Mol Physiol ; 282(3): L359-69, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11839528

RESUMO

Although thyroid hormone (T(3)) influences epithelial cell differentiation during late fetal lung development, its effects on early lung morphogenesis are unknown. We hypothesized that T(3) would alter embryonic lung airway branching and temporal-spatial differentiation of the lung epithelium and mesenchyme. Gestational day 11.5 embryonic mouse lungs were cultured for 72 h in BGJb serum-free medium without or with added T(3) (0.2, 2.0, 10.0, or 100 nM). Evaluation of terminal bud counts showed a dose- and time-dependent decrease in branching morphogenesis. Cell proliferation was also significantly decreased with higher doses of T(3). Morphometric analysis of lung histology showed that T(3) caused a dose-dependent decrease in mesenchyme and increase in cuboidal epithelia and airway space. Immunocytochemistry showed that with T(3) treatment, Nkx2.1 and surfactant protein SP-C proteins became progressively localized to cuboidal epithelial cells and mesenchymal expression of Hoxb5 was reduced, a pattern resembling late fetal lung development. We conclude that exogenous T(3) treatment during early lung development accelerated epithelial and mesenchymal cell differentiation at the expense of premature reduction in new branch formation and lung growth.


Assuntos
Pulmão/embriologia , Tri-Iodotironina/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Técnicas de Cultura , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Desenvolvimento Embrionário e Fetal/fisiologia , Camundongos , Tri-Iodotironina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA