Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Stroke ; 48(5): 1420-1423, 2017 05.
Article in English | MEDLINE | ID: mdl-28325846

ABSTRACT

BACKGROUND AND PURPOSE: Brain arteriovenous malformation (bAVM) is an important risk factor for intracranial hemorrhage. Current therapies are associated with high morbidities. Excessive vascular endothelial growth factor has been implicated in bAVM pathophysiology. Because soluble FLT1 binds to vascular endothelial growth factor with high affinity, we tested intravenous delivery of an adeno-associated viral vector serotype-9 expressing soluble FLT1 (AAV9-sFLT1) to alleviate the bAVM phenotype. METHODS: Two mouse models were used. In model 1, bAVM was induced in R26CreER;Eng2f/2f mice through global Eng gene deletion and brain focal angiogenic stimulation; AAV2-sFLT02 (an AAV expressing a shorter form of sFLT1) was injected into the brain at the time of model induction, and AAV9-sFLT1, intravenously injected 8 weeks after. In model 2, SM22αCre;Eng2f/2f mice had a 90% occurrence of spontaneous bAVM at 5 weeks of age and 50% mortality at 6 weeks; AAV9-sFLT1 was intravenously delivered into 4- to 5-week-old mice. Tissue samples were collected 4 weeks after AAV9-sFLT1 delivery. RESULTS: AAV2-sFLT02 inhibited bAVM formation, and AAV9-sFLT1 reduced abnormal vessels in model 1 (GFP versus sFLT1: 3.66±1.58/200 vessels versus 1.98±1.29, P<0.05). AAV9-sFLT1 reduced the occurrence of bAVM (GFP versus sFLT1: 100% versus 36%) and mortality (GFP versus sFLT1: 57% [12/22 mice] versus 24% [4/19 mice], P<0.05) in model 2. Kidney and liver function did not change significantly. Minor liver inflammation was found in 56% of AAV9-sFLT1-treated model 1 mice. CONCLUSIONS: By applying a regulated mechanism to restrict sFLT1 expression to bAVM, AAV9-sFLT1 can potentially be developed into a safer therapy to reduce the bAVM severity.


Subject(s)
Angiogenesis Inhibitors , Arteriovenous Fistula/therapy , Genetic Therapy/methods , Genetic Vectors , Intracranial Arteriovenous Malformations/therapy , Vascular Endothelial Growth Factor Receptor-1 , Animals , Dependovirus , Disease Models, Animal , Genetic Vectors/administration & dosage , Mice
2.
Am J Physiol Lung Cell Mol Physiol ; 310(11): L1098-110, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27036872

ABSTRACT

Impaired vascular endothelial growth factor (VEGF) signaling contributes to the pathogenesis of bronchopulmonary dysplasia (BPD). We hypothesized that the effects of VEGF on lung structure during development may be mediated through its downstream effects on both endothelial nitric oxide synthase (eNOS) and hepatocyte growth factor (HGF) activity, and that, in the absence of eNOS, trophic effects of VEGF would be mediated through HGF signaling. To test this hypothesis, we performed an integrative series of in vitro (fetal rat lung explants and isolated fetal alveolar and endothelial cells) and in vivo studies with normal rat pups and eNOS(-/-) mice. Compared with controls, fetal lung explants from eNOS(-/-) mice had decreased terminal lung bud formation, which was restored with recombinant human VEGF (rhVEGF) treatment. Neonatal eNOS(-/-) mice were more susceptible to hyperoxia-induced inhibition of lung growth than controls, which was prevented with rhVEGF treatment. Fetal alveolar type II (AT2) cell proliferation was increased with rhVEGF treatment only with mesenchymal cell (MC) coculture, and these effects were attenuated with anti-HGF antibody treatment. Unlike VEGF, HGF directly stimulated isolated AT2 cells even without MC coculture. HGF directly stimulates fetal pulmonary artery endothelial cell growth and tube formation, which is attenuated by treatment with JNJ-38877605, a c-Met inhibitor. rHGF treatment preserves alveolar and vascular growth after postnatal exposure to SU-5416, a VEGF receptor inhibitor. We conclude that the effects of VEGF on AT2 and endothelial cells during lung development are partly mediated through HGF-c-Met signaling and speculate that reciprocal VEGF-HGF signaling between epithelia and endothelia is disrupted in infants who develop BPD.


Subject(s)
Hepatocyte Growth Factor/physiology , Lung/growth & development , Vascular Endothelial Growth Factor A/physiology , Alveolar Epithelial Cells/physiology , Animals , Cell Adhesion , Cells, Cultured , Coculture Techniques , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Female , Lung/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/genetics , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Sheep
3.
Am J Physiol Lung Cell Mol Physiol ; 310(4): L287-98, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26566904

ABSTRACT

Prevention or treatment of lung diseases caused by the failure to form, or destruction of, existing alveoli, as observed in infants with bronchopulmonary dysplasia and adults with emphysema, requires understanding of the molecular mechanisms of alveolar development. In addition to its critical role in gas exchange, the pulmonary circulation also contributes to alveolar morphogenesis and maintenance by the production of paracrine factors, termed "angiocrines," that impact the development of surrounding tissue. To identify lung angiocrines that contribute to alveolar formation, we disrupted pulmonary vascular development by conditional inactivation of the Vegf-A gene during alveologenesis. This resulted in decreased pulmonary capillary and alveolar development and altered lung elastin and retinoic acid (RA) expression. We determined that RA is produced by pulmonary endothelial cells and regulates pulmonary angiogenesis and elastin synthesis by induction of VEGF-A and fibroblast growth factor (FGF)-18, respectively. Inhibition of RA synthesis in newborn mice decreased FGF-18 and elastin expression and impaired alveolarization. Treatment with RA and vitamin A partially reversed the impaired vascular and alveolar development induced by VEGF inhibition. Thus we identified RA as a lung angiocrine that regulates alveolarization through autocrine regulation of endothelial development and paracrine regulation of elastin synthesis via induction of FGF-18 in mesenchymal cells.


Subject(s)
Endothelial Cells/metabolism , Endothelium/metabolism , Fibroblast Growth Factors/metabolism , Lung/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Animals, Newborn , Capillaries/metabolism , Cells, Cultured , Mice, Transgenic , Neovascularization, Physiologic
4.
Cell Stem Cell ; 15(2): 123-38, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25105578

ABSTRACT

Respiratory disease is the third leading cause of death in the industrialized world. Consequently, the trachea, lungs, and cardiopulmonary vasculature have been the focus of extensive investigations. Recent studies have provided new information about the mechanisms driving lung development and differentiation. However, there is still much to learn about the ability of the adult respiratory system to undergo repair and to replace cells lost in response to injury and disease. This Review highlights the multiple stem/progenitor populations in different regions of the adult lung, the plasticity of their behavior in injury models, and molecular pathways that support homeostasis and repair.


Subject(s)
Lung/cytology , Stem Cells/cytology , Animals , Bronchioles/physiology , Cell Differentiation , Cell Lineage , Epithelium/physiology , Homeostasis , Humans , Lung/embryology , Mesoderm/physiology , Mice , Pulmonary Alveoli/physiology , Regeneration/physiology , Respiration , Respiratory System , Signal Transduction , Tissue Engineering/methods , Trachea/embryology , Trachea/physiology
5.
Anat Rec (Hoboken) ; 295(10): 1707-16, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22887719

ABSTRACT

Exposure to oxidant air pollutants in early childhood, with ozone as the key oxidant, has been linked to significant decrements in pulmonary function in young adults and exacerbation of airway remodeling in asthma. Development of lung parenchyma in rhesus monkeys is rapid during the first 2 years of life (comparable to the first 6 years in humans). Our hypothesis is that ozone inhalation during infancy alters alveolar morphogenesis. We exposed infant rhesus monkeys biweekly to 5, 8 hr/day, cycles of 0.5 ppm ozone with or without house dust mite allergen from 1 to 3 or 1 to 6 months of age. Monkeys were necropsied at 3 and 6 months of age. A morphometric approach was used to quantify changes in alveolar volume and number, the distribution of alveolar size, and capillary surface density per alveolar septa. Quantitative real time PCR was used to measure the relative difference in gene expression over time. Monkeys exposed to ozone alone or ozone combined with allergen had statistically larger alveoli that were less in number at 3 months of age. Alveolar capillary surface density was also decreased in the ozone exposed groups at 3 months of age. At 6 months of age, the alveolar number was similar between treatment groups and was associated with a significant rise in alveolar number from 3 to 6 months of age in the ozone exposed groups. This increase in alveolar number was not associated with any significant increase in microvascular growth as measured by morphometry or changes in angiogenic gene expression. Inhalation of ozone during infancy alters the appearance and timing of alveolar growth and maturation. Understanding the mechanism involved with this altered alveolar growth may provide insight into the parenchymal injury and repair process that is involved with chronic lung diseases such as severe asthma and COPD.


Subject(s)
Air Pollutants/toxicity , Ozone/toxicity , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/growth & development , Administration, Inhalation , Age Factors , Animals , Animals, Newborn , Atmosphere Exposure Chambers/adverse effects , Cell Count/methods , Environmental Exposure/adverse effects , Macaca mulatta , Male , Ozone/administration & dosage , Primates , Pulmonary Alveoli/cytology , Time Factors
6.
Anat Rec (Hoboken) ; 295(1): 167-76, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21956870

ABSTRACT

Disrupted lung alveolar myofibroblast and bronchial smooth muscle (BSM) cell development may lead to pulmonary disorders such as bronchopulmonary dysplasia. The molecular mechanisms that regulate BSM and alveolar myofibroblast development are not fully understood. Here we show that mSmile (murine Smile), a novel transmembrane protein with tetratricopeptide repeats, functions in lung alveolar myofibroblast and BSM cell development. mSmile mutant mice exhibit early neonatal lethality with few mice surviving up to 3 weeks. Mutant lungs display both airway branching morphogenesis defect during fetal lung development and alveolarization defect after birth. These defects are associated with reduced numbers of BSM cells in the peribronchial subepithelial region and clefts and myofibroblasts in alveolar septae. Expression of fibroblast growth factor-10 and its down stream target Bmp-4, which are important for BSM formation, is decreased. In vitro, mSmile mutant embryonic fibroblasts show reduced receptor activation and induction of myofibroblast formation in response to Transforming growth factor-ß (Tgf-ß), indicating that mSmile may mediate myofibroblast development through modulation of Tgf-ß signaling. These studies identify mSmile as a novel gene specifying both the BSM and lung alveolar myofibroblast lineages, contributing to our understanding of the biological control of the development of these cells, and may provide insights into the aberrant smooth muscle and alveolar myofibroblast development that occur in pathological conditions.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , Bronchi/embryology , Gene Expression Regulation, Developmental/genetics , Myocytes, Smooth Muscle/metabolism , Myofibroblasts/metabolism , Pulmonary Alveoli/embryology , Animals , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Bronchi/cytology , Cell Lineage/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocytes, Smooth Muscle/cytology , Myofibroblasts/cytology , Organogenesis/genetics , Organogenesis/physiology , Primary Cell Culture , Pulmonary Alveoli/cytology , Radiation Chimera
7.
J Clin Invest ; 121(7): 2855-62, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21701069

ABSTRACT

Laminins and their integrin receptors are implicated in epithelial cell differentiation and progenitor cell maintenance. We report here that a previously unrecognized subpopulation of mouse alveolar epithelial cells (AECs) expressing the laminin receptor α6ß4, but little or no pro-surfactant C (pro-SPC), is endowed with regenerative potential. Ex vivo, this subpopulation expanded clonally as progenitors but also differentiated toward mature cell types. Integrin ß4 itself was not required for AEC proliferation or differentiation. An in vivo embryonic lung organoid assay, which we believe to be novel, was used to show that purified ß4+ adult AECs admixed with E14.5 lung single-cell suspensions and implanted under kidney capsules self-organized into distinct Clara cell 10-kDa secretory protein (CC10+) airway-like and SPC+ saccular structures within 6 days. Using a bleomycin model of lung injury and an SPC-driven inducible cre to fate-map AECs, we found the majority of type II AECs in fibrotic areas were not derived from preexisting type II AECs, demonstrating that SPC- progenitor cells replenished type II AECs during repair. Our findings support the idea that there is a stable AEC progenitor population in the adult lung, provide in vivo evidence of AEC progenitor cell differentiation after parenchymal injury, and identify a strong candidate progenitor cell for maintenance of type II AECs during lung repair.


Subject(s)
Epithelial Cells/physiology , Integrin alpha6beta4/metabolism , Lung/anatomy & histology , Lung/physiology , Regeneration/physiology , Respiratory Mucosa/cytology , Animals , Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Cell Differentiation/physiology , Cells, Cultured , Epithelial Cells/cytology , Female , Lung/drug effects , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Alveoli/cytology , Pulmonary Surfactants/metabolism , Stem Cells/cytology , Stem Cells/physiology , Uteroglobin/metabolism
8.
Dis Model Mech ; 3(3-4): 224-35, 2010.
Article in English | MEDLINE | ID: mdl-20142327

ABSTRACT

Long bone development depends on endochondral bone formation, a complex process requiring exquisite balance between hypertrophic cartilage (HC) formation and its ossification. Dysregulation of this process may result in skeletal dysplasias and heterotopic ossification. Endochondral ossification requires the precise orchestration of HC vascularization, extracellular matrix remodeling, and the recruitment of osteoclasts and osteoblasts. Matrix metalloproteinase-9 (MMP-9), vascular endothelial growth factor (VEGF) and osteoclasts have all been shown to regulate endochondral ossification, but how their function interrelates is not known. We have investigated the functional relationship among these regulators of endochondral ossification, demonstrating that they have complementary but non-overlapping functions. MMP-9, VEGF and osteoclast deficiency all cause impaired growth plate ossification resulting in the accumulation of HC. VEGF mRNA and protein expression are increased at the MMP-9-/- growth plate, and VEGF activity contributes to endochondral ossification since sequestration of VEGF by soluble receptors results in further inhibition of growth plate vascularization and ossification. However, VEGF bioavailability is still limited in MMP-9 deficiency, as exogenous VEGF is able to rescue the MMP-9-/- phenotype, demonstrating that MMP-9 may partially, but not fully, regulate VEGF bioavailability. The organization of the HC extracellular matrix at the MMP-9-/- growth plate is altered, supporting a role for MMP-9 in HC remodeling. Inhibition of VEGF impairs osteoclast recruitment, whereas MMP-9 deficiency leads to an accumulation of osteoclasts at the chondro-osseous junction. Growth plate ossification in osteoclast-deficient mice is impaired in the presence of normal MMP-9 expression, indicating that other osteoclastic functions are also necessary. Our data delineate the complementary interplay between MMP-9, VEGF and osteoclast function that is necessary for normal endochondral bone formation and provide a molecular framework for investigating the molecular defects contributing to disorders of endochondral bone formation.


Subject(s)
Chondrocytes/pathology , Matrix Metalloproteinase 9/metabolism , Osteoclasts/enzymology , Osteoclasts/pathology , Osteogenesis , Vascular Endothelial Growth Factor A/metabolism , Acid Phosphatase/metabolism , Animals , Animals, Newborn , Bone and Bones/drug effects , Bone and Bones/enzymology , Bone and Bones/pathology , Bone and Bones/ultrastructure , Cartilage/pathology , Cartilage/ultrastructure , Chondrocytes/drug effects , Chondrocytes/enzymology , Chondrocytes/ultrastructure , Collagen Type II/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Growth Plate/drug effects , Growth Plate/enzymology , Growth Plate/pathology , Growth Plate/ultrastructure , Humans , Hypertrophy , Isoenzymes/metabolism , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase Inhibitors , Mice , Models, Biological , Osteoclasts/drug effects , Osteoclasts/ultrastructure , Osteogenesis/drug effects , Phenotype , Protease Inhibitors/pharmacology , Tartrate-Resistant Acid Phosphatase , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/pharmacology
9.
Anat Rec (Hoboken) ; 292(1): 123-30, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19115389

ABSTRACT

Epithelial-mesenchymal interactions are essential for tissue patterning during organogenesis. Distal lung epithelium and its adjacent mesenchyme comprise the epithelial-mesenchymal signaling unit that regulates lung branching morphogenesis. Tissue recombination experiments have demonstrated the importance of mesenchymal signals in inducing lung epithelial differentiation and branching, but the role of the epithelium in regulating mesenchymal signals has not been well characterized. Using transgenic mice, we ablated distal lung epithelial cells during lung development by inducing the expression of a constitutively active proapoptotic Bax protein under the surfactant protein C (SP-C) promoter. We found that epithelial cell ablation results in impaired lung branching morphogenesis, which progresses to emphysematous airspaces in the adults. Mesenchymal expression of fibroblast growth factor 10 (Fgf-10), whose strict spatial and temporal expression is critical for proper lung branching morphogenesis, is disrupted and loses its localized pattern. Interestingly, the expression of sonic hedgehog (Shh), an epithelial gene known to modulate Fgf-10 expression, is unchanged, indicating the existence of other distal epithelial signals that regulate mesenchymal Fgf-10expression. We propose that distal SP-C expressing lung epithelial cells provide essential signals for the downregulation of Fgf-10 expression in the distal mesenchyme during lung development. 292:123-130, 2009. (c) 2008 Wiley-Liss, Inc.


Subject(s)
Fibroblast Growth Factor 10/biosynthesis , Lung/cytology , Lung/metabolism , Morphogenesis/physiology , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Ablation Techniques/methods , Animals , Female , Humans , Lung/growth & development , Mice , Mice, Transgenic , Pregnancy , Pulmonary Emphysema/etiology , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Respiratory Mucosa/growth & development
10.
J Cell Sci ; 121(Pt 16): 2751-8, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18664493

ABSTRACT

Lipid droplets are complex and dynamic intracellular organelles that have an essential role in cholesterol and lipid homeostasis, and profoundly affect cellular structure and function. Variations in lipid-droplet composition exist between different cell types, but whether there are differences in the mechanisms of lipid-droplet accumulation remains to be elucidated. Here, we report that P311, previously identified to have a function in neuronal regeneration and a potential role in distal lung generation, regulates lipid droplet accumulation. P311 upregulates several classes of genes associated with lipid synthesis, significantly increases intracellular cholesterol and triglyceride levels, and increases intracellular lipid droplets. Interestingly, P311 expression is not necessary for lipogenesis in the well-established NIH3T3-L1 cell model of adipogenic differentiation. Instead, we demonstrate a novel role for P311 in an alternative pathway of lipid-droplet accumulation that is induced by the regeneration-inducing molecule retinoic acid.


Subject(s)
Lipid Metabolism/drug effects , Nerve Tissue Proteins/physiology , Tretinoin/pharmacology , 3T3-L1 Cells , Animals , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Silencing , Lipid Metabolism/genetics , Lung/metabolism , Lung/physiology , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Nerve Tissue Proteins/metabolism , Regeneration/drug effects , Regeneration/genetics , Signal Transduction/physiology , Up-Regulation/drug effects , Up-Regulation/genetics
11.
Dev Biol ; 308(1): 44-53, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17583691

ABSTRACT

There is increasing evidence that epithelial-vascular interactions are essential for tissue patterning. Here we identified components of the molecular cross talk between respiratory epithelial cells and pulmonary capillaries necessary for the formation of the gas exchange surface of the lung. Selective inactivation of the Vegf-A gene in respiratory epithelium results in an almost complete absence of pulmonary capillaries, demonstrating the dependence of pulmonary capillary development on epithelium-derived Vegf-A. Deficient capillary formation in Vegf-A deficient lungs is associated with a defect in primary septae formation, a morphogenetic process critical for distal lung morphogenesis, coupled with suppression of epithelial cell proliferation and decreased hepatocyte growth factor (Hgf) expression. Lung endothelial cells express Hgf, and selective deletion of the Hgf receptor gene in respiratory epithelium phenocopies the malformation of septae, confirming the requirement for epithelial Hgf signaling in normal septae formation and suggesting that Hgf serves as an endothelium-derived factor that signals to the epithelium. Our findings support a mechanism for primary septae formation dependent on reciprocal interactions between respiratory epithelium and the underlying vasculature, establishing the dependence of pulmonary capillary development on epithelium-derived Vegf-A, and identify Hgf as a putative endothelium-derived factor that mediates the reciprocal signaling from the vasculature to the respiratory epithelium.


Subject(s)
Hepatocyte Growth Factor/metabolism , Lung/embryology , Lung/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Base Sequence , Capillaries/embryology , Capillaries/metabolism , Cell Differentiation , Cell Proliferation , Cell Survival , DNA Primers/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/embryology , Epithelium/metabolism , Female , Fibroblast Growth Factors/genetics , Gene Dosage , Gene Expression Regulation, Developmental , In Situ Hybridization , Lung/blood supply , Male , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Neovascularization, Physiologic , Pregnancy , Signal Transduction , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics
12.
Birth Defects Res C Embryo Today ; 78(1): 80-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16622850

ABSTRACT

Epithelial-mesenchymal interactions and extracellular matrix remodeling are key processes of embryonic lung development. Lung smooth muscle cells, which are derived from the mesenchyme, form a sheath around bronchi and blood vessels. During lung organogenesis, smooth muscle differentiation coincides with epithelial branching morphogenesis and closely follows developing airways spatially and temporally. The precise function of parabronchial smooth muscle (PBSM) cells in healthy adult lung remains unclear. However, PBSM may regulate epithelial branching morphogenesis during lung development by the induction of mechanical stress or through regulation of paracrine signaling pathways. Alveolar myofibroblasts are interstitial contractile cells that share features and may share an origin with smooth muscle cells. Alveolar myofibroblasts are essential for secondary septation, a process critical for the development of the gas-exchange region of the lung. Dysregulation of PBSM or alveolar myofibroblast development is thought to underlie the pathogenesis of many lung diseases, including bronchopulmonary dysplasia, asthma, and interstitial fibrosis. We review the current understanding of the regulation of PBSM and alveolar myofibroblast development, and discuss the role of PBSM in lung development. We specifically focus on the role of these cells in the context of fibroblast growth factor-10, sonic hedgehog, bone morphogenetic protein-4, retinoic acid, and Wnt signaling pathways in the regulation of lung branching morphogenesis.


Subject(s)
Bronchi/cytology , Fibroblasts/cytology , Myocytes, Smooth Muscle/cytology , Organogenesis , Pulmonary Alveoli/cytology , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/metabolism , Bronchi/embryology , Extracellular Matrix/physiology , Fibroblast Growth Factor 10/metabolism , Hedgehog Proteins , Humans , Lung Diseases/metabolism , Lung Diseases/physiopathology , Pulmonary Alveoli/embryology , Trans-Activators/metabolism , Tretinoin/metabolism , Wnt1 Protein/metabolism
13.
Am J Respir Cell Mol Biol ; 35(1): 48-54, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16484684

ABSTRACT

Smoking-related destructive lung diseases such as chronic obstructive pulmonary disease (COPD) and emphysema are a major cause of morbidity and mortality worldwide. The immediate cause of emphysema is the obliteration of alveoli that are key functional units of the lungs where gas exchange takes place. Alveolar generation/regeneration under normal and pathologic conditions is a poorly understood process, but may hold the key to treatment of human emphysema. We used suppression subtractive hybridization to identify genes that may control alveolar generation during periods of pre- and postnatal active alveolar development. P311, a putative neuronal protein originally identified for its high expression in late-stage embryonic brain, was highly differentially expressed during periods of active distal lung morphogenesis. Quantitative real-time RT-PCR showed that the expression of P311 is developmentally regulated, with peak levels occurring during saccular and alveolar formation. Intriguingly, P311 gene expression was significantly decreased in lungs of individuals with emphysema compared with control subjects. Consistent with a role for this gene in alveolar formation, inhibition of alveolization by dexamethasone treatment in vivo resulted in decreased expression of P311. Together our data suggest that P311 expression is tightly regulated during the critical periods of alveolar formation, and that under pathologic conditions, its relative absence may contribute to failure of alveolar regeneration and lead to the development of human emphysema.


Subject(s)
Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Pulmonary Alveoli/metabolism , Aged , Animals , Animals, Newborn , Dexamethasone/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Fetus/cytology , Gene Expression Profiling , Humans , Male , Mice , Middle Aged , Pulmonary Alveoli/cytology , Pulmonary Alveoli/embryology , Pulmonary Alveoli/pathology , Pulmonary Emphysema/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
J Invest Dermatol ; 126(2): 486-96, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16374453

ABSTRACT

Degradation of the extracellular matrix, which is an indispensable step in tissue remodelling processes such as embryonic development and wound healing of the skin, has been attributed to collagenolytic activity of members of the matrix metalloproteinase family (MMPs). Here, we employed mmp13 knockout mice to elucidate the function of MMP13 in embryonic skin development, skin homeostasis, and cutaneous wound healing. Overall epidermal architecture and dermal composition of non-injured skin were indistinguishable from wild-type mice. Despite robust expression of MMP13 in the early phase of wound healing, wild-type and mmp13 knockout animals did not differ in their efficiency of re-epithelialization, inflammatory response, granulation tissue formation, angiogenesis, and restoration of basement membrane. Yet, among other MMPs also expressed during wound healing, MMP8 was found to be enhanced in wounds of MMP13-deficient mice. In summary, skin homeostasis and also tissue remodelling processes like embryonic skin development and cutaneous wound healing are independent of MMP13 either owing to MMP13 dispensability or owing to functional substitution by other collagenolytic proteinases such as MMP8.


Subject(s)
Collagenases/physiology , Epidermis/embryology , Granulation Tissue/growth & development , Skin/embryology , Wound Healing , Animals , Collagenases/deficiency , Collagenases/genetics , Epidermal Cells , Epidermis/enzymology , Matrix Metalloproteinase 13 , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 8/metabolism , Mice , Mice, Knockout , Neovascularization, Physiologic , Phenotype , Skin/cytology , Skin/enzymology , Wound Healing/genetics
15.
Mech Dev ; 122(7-8): 877-86, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15927453

ABSTRACT

The vasculature forms an intrinsic functional component of the lung and its development must be tightly regulated and coordinated with lung epithelial morphogenesis. Vascular endothelial growth factor (VEGF) and its receptors are highly expressed in a complementary pattern in the lungs during embryonic development. VEGF is expressed by epithelium and the receptors in the surrounding mesenchyme. To determine the function of VEGF in lung formation, we inhibited its activity using a soluble receptor in lung renal capsule grafts. Inhibition of VEGF results in inhibition of vascular development and significant alteration in epithelial development. Epithelial proliferation is inhibited, sacculation is impaired, and the epithelium undergoes apoptosis. Interestingly, when VEGF is attenuated, epithelial differentiation still proceeds, as shown by acquisition of both proximal and distal markers. These data show that VEGF co-ordinates epithelial and vascular development. It is required for the development of the lung vasculature and the vasculature is necessary for epithelial proliferation and morphogenesis, but not for cell differentiation.


Subject(s)
Epithelium/embryology , Lung/blood supply , Lung/embryology , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Differentiation , Cell Proliferation , Epithelium/metabolism , Gene Expression Regulation, Developmental , Lung/cytology , Lung/metabolism , Mice , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Transplantation, Homologous , Vascular Endothelial Growth Factor A/antagonists & inhibitors
17.
Shi Yan Sheng Wu Xue Bao ; 37(4): 303-9, 2004 Aug.
Article in Chinese | MEDLINE | ID: mdl-15511073

ABSTRACT

Lung is a critical respiratory organ of living body. The regulation of lung morphogenesis, especially the epithelial development in the late fetal stages, is not completely understood due to lack of efficiency strategies. In this study we showed that the growth of dissociated embryonic lung cells implanted underneath the kidney capsules of syngeneic hosts followed closely lung development in utero. The epithelium developed extensively and appeared to go through pseudoglandular stage, canallicular stage, and saccular stage at a pace similar to normal lung development. At the same time, the capillary-like vascular developed also. The greatest advantage of this model was that the dissociated single cells uptake antisence oligo at a high level, and introduced phenotype after grafting. Embryonic lung cell renal capsule graft is thus an excellent model for the study of lung morphogenesis.


Subject(s)
Lung/blood supply , Lung/embryology , Animals , Epithelium/embryology , Epithelium/physiology , Female , Immunohistochemistry , In Situ Hybridization , Kidney/blood supply , Kidney/cytology , Lung/physiology , Mice , Neovascularization, Physiologic/physiology , Pregnancy , Transplantation, Homologous
18.
J Biol Chem ; 279(51): 53848-56, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15456762

ABSTRACT

The epidermal growth factor receptor (EGFR) and its ligands function in diverse cellular functions including cell proliferation, differentiation, motility, and survival. EGFR signaling is important for the development of many tissues, including skin, lungs, intestines, and the craniofacial skeleton. We have now determined the role of EGFR signaling in endochondral ossification. We analyzed long bone development in EGFR-deficient mice. EGFR deficiency caused delayed primary ossification of the cartilage anlage and delayed osteoclast and osteoblast recruitment. Ossification of the growth plates was also abnormal resulting in an expanded area of growth plate hypertrophic cartilage and few bony trabeculae. The delayed osteoclast recruitment was not because of inadequate expression of matrix metalloproteinases, including matrix metalloproteinase-9, which have previously been shown to be important for osteoclast recruitment. EGFR was expressed by osteoclasts, suggesting that EGFR ligands may act directly to affect the formation and/or function of these cells. EGFR signaling regulated osteoclast formation. Inhibition of EGFR tyrosine kinase activity decreased the generation of osteoclasts from cultured bone marrow cells.


Subject(s)
Bone Development , Bone and Bones/metabolism , ErbB Receptors/genetics , ErbB Receptors/physiology , Osteoclasts/metabolism , Osteogenesis , Animals , Bone Marrow Cells/cytology , Bone and Bones/embryology , Cell Movement , Cell Proliferation , Cell Survival , Collagen Type I/biosynthesis , Gelatin/chemistry , In Situ Hybridization , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Osteocalcin/biosynthesis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Tissue Distribution
19.
Mech Dev ; 120(3): 305-13, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12591600

ABSTRACT

The study of distal lung morphogenesis and vascular development would be greatly facilitated by an in vitro or ex vivo experimental model. In this study we show that the growth of mouse embryonic day 12.5 lung rudiments implanted underneath the kidney capsules of syngeneic or immunodeficient hosts follows closely lung development in utero. The epithelium develops extensively with both proximal and distal differentiation to the saccular stage. The vasculature also develops extensively. Large blood vessels accompany large airways and capillaries develop within the saccular walls. Interestingly, vessels in the lung grafts develop from endothelial progenitor cells endogenous to the explants and host vessels do not vascularize the grafts independently. This suggests that embryonic lungs possess mechanisms to prevent the inappropriate ingrowth of surrounding vessels. However, vessels in the lung grafts do connect to host vessels, showing that embryonic lungs have the ability to stimulate host angiogenesis and recruit host vessel connections. These data support the hypothesis that the lung vasculature develops by both vasculogenic and angiogenic processes: a vascular network develops in situ in lung mesenchyme, which is then connected to angiogenic processes from central vessels. The lung renal capsule allograft is thus an excellent model to study the development of the pulmonary vasculature and of late fetal lung development that requires a functional blood supply.


Subject(s)
Blood Vessels/embryology , Kidney , Lung Transplantation/methods , Lung/embryology , Neovascularization, Physiologic/physiology , Animals , Epithelial Cells/physiology , Epithelium/embryology , Female , Lung/blood supply , Mice , Mice, Inbred Strains , Pregnancy , Stem Cells/physiology , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...