Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Nature ; 541(7636): 182-187, 2017 01 12.
Article in English | MEDLINE | ID: mdl-28052057

ABSTRACT

Despite the global prevalence of gastric disease, there are few adequate models in which to study the fundus epithelium of the human stomach. We differentiated human pluripotent stem cells (hPSCs) into gastric organoids containing fundic epithelium by first identifying and then recapitulating key events in embryonic fundus development. We found that disruption of Wnt/ß-catenin signalling in mouse embryos led to conversion of fundic to antral epithelium, and that ß-catenin activation in hPSC-derived foregut progenitors promoted the development of human fundic-type gastric organoids (hFGOs). We then used hFGOs to identify temporally distinct roles for multiple signalling pathways in epithelial morphogenesis and differentiation of fundic cell types, including chief cells and functional parietal cells. hFGOs are a powerful model for studying the development of the human fundus and the molecular bases of human gastric physiology and pathophysiology, and also represent a new platform for drug discovery.


Subject(s)
Gastric Fundus/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Body Patterning , Cell Differentiation , Cell Lineage , Drug Discovery/methods , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/embryology , Epithelium/metabolism , Female , Gastric Fundus/cytology , Gastric Fundus/embryology , Homeodomain Proteins/metabolism , Humans , Male , Mice , Organoids/cytology , Organoids/embryology , Organoids/metabolism , Parietal Cells, Gastric/cytology , Parietal Cells, Gastric/metabolism , Pluripotent Stem Cells/cytology , SOXB1 Transcription Factors/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Trans-Activators/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/agonists
2.
Development ; 144(21): 3879-3893, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28947536

ABSTRACT

The in vitro-directed differentiation of pluripotent stem cells (PSCs) through stimulation of developmental signaling pathways can generate mature somatic cell types for basic laboratory studies or regenerative therapies. However, there has been significant uncertainty regarding a method to separately derive lung versus thyroid epithelial lineages, as these two cell types each originate from Nkx2-1+ foregut progenitors and the minimal pathways claimed to regulate their distinct lineage specification in vivo or in vitro have varied in previous reports. Here, we employ PSCs to identify the key minimal signaling pathways (Wnt+BMP versus BMP+FGF) that regulate distinct lung- versus thyroid-lineage specification, respectively, from foregut endoderm. In contrast to most previous reports, these minimal pathways appear to be evolutionarily conserved between mice and humans, and FGF signaling, although required for thyroid specification, unexpectedly appears to be dispensable for lung specification. Once specified, distinct Nkx2-1+ lung or thyroid progenitor pools can now be independently derived for functional 3D culture maturation, basic developmental studies or future regenerative therapies.


Subject(s)
Body Patterning , Cell Differentiation , Lung/cytology , Lung/embryology , Pluripotent Stem Cells/cytology , Signal Transduction , Thyroid Gland/cytology , Animals , Biomarkers/metabolism , Body Patterning/genetics , Bone Morphogenetic Proteins/metabolism , Cell Lineage , Embryo, Mammalian/cytology , Embryonic Development , Endoderm/cytology , Endoderm/metabolism , Epithelial Cells/cytology , Fibroblast Growth Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Reproducibility of Results , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Thyroid Gland/embryology , Transcriptome/genetics , Wnt Proteins/metabolism
3.
Dev Biol ; 434(1): 121-132, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29217200

ABSTRACT

A small number of signaling pathways are used repeatedly during organogenesis, and they can have drastically different effects on the same population of cells depending on the embryonic stage. How cellular competence changes over developmental time is not well understood. Here we used Xenopus, mouse, and human pluripotent stem cells to investigate how the temporal sequence of Wnt, BMP, and retinoic acid (RA) signals regulates endoderm developmental competence and organ induction, focusing on respiratory fate. While Nkx2-1+ lung fate is not induced until late somitogenesis stages, here we show that lung competence is restricted by the gastrula stage as a result of Wnt and BMP-dependent anterior-posterior (A-P) patterning. These early Wnt and BMP signals make posterior endoderm refractory to subsequent RA/Wnt/BMP-dependent lung induction. We further mapped how RA modulates the response to Wnt and BMP in a temporal specific manner. In the gastrula RA promotes posterior identity, however in early somite stages of development RA regulates respiratory versus pharyngeal potential in anterior endoderm and midgut versus hindgut potential in posterior endoderm. Together our data suggest a dynamic and conserved response of vertebrate endoderm during organogenesis, wherein early Wnt/BMP/RA impacts how cells respond to later Wnt/BMP/RA signals, illustrating how reiterative combinatorial signaling can regulate both developmental competence and subsequent fate specification.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Endoderm/embryology , Organogenesis/drug effects , Tretinoin/pharmacology , Wnt Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Endoderm/cytology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Organogenesis/physiology , Somites/cytology , Somites/embryology , Species Specificity , Xenopus laevis
4.
Dev Biol ; 443(1): 50-63, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30153454

ABSTRACT

Organogenesis is regulated by mesenchymal-epithelial signaling events that induce expression of cell-type specific transcription factors critical for cellular proliferation, differentiation and appropriate tissue patterning. While mesenchymal transcription factors play a key role in mesenchymal-epithelial interactions, transcriptional networks in septum transversum and splanchnic mesenchyme remain poorly characterized. Forkhead Box F1 (FOXF1) transcription factor is expressed in mesenchymal cell lineages; however, its role in organogenesis remains uncharacterized due to early embryonic lethality of Foxf1-/- mice. In the present study, we generated mesenchyme-specific Foxf1 knockout mice (Dermo1-Cre Foxf1-/-) and demonstrated that FOXF1 is required for development of respiratory, cardiovascular and gastrointestinal organ systems. Deletion of Foxf1 from mesenchyme caused embryonic lethality in the middle of gestation due to multiple developmental defects in the heart, lung, liver and esophagus. Deletion of Foxf1 inhibited mesenchyme proliferation and delayed branching lung morphogenesis. Gene expression profiling of micro-dissected distal lung mesenchyme and ChIP sequencing of fetal lung tissue identified multiple target genes activated by FOXF1, including Wnt2, Wnt11, Wnt5A and Hoxb7. FOXF1 decreased expression of the Wnt inhibitor Wif1 through direct transcriptional repression. Furthermore, using a global Foxf1 knockout mouse line (Foxf1-/-) we demonstrated that FOXF1-deficiency disrupts the formation of the lung bud in foregut tissue explants. Finally, deletion of Foxf1 from smooth muscle cell lineage (smMHC-Cre Foxf1-/-) caused hyper-extension of esophagus and trachea, loss of tracheal and esophageal muscle, mispatterning of esophageal epithelium and decreased proliferation of smooth muscle cells. Altogether, FOXF1 promotes lung morphogenesis by regulating mesenchymal-epithelial signaling and stimulating cellular proliferation in fetal lung mesenchyme.


Subject(s)
Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Lung/embryology , Animals , Cell Proliferation , Forkhead Transcription Factors/physiology , Gene Expression Regulation, Developmental/genetics , Lung/cytology , Lung/metabolism , Mesoderm/metabolism , Mice/embryology , Mice, Inbred C57BL , Mice, Knockout , Organogenesis/physiology , Transcription Factors/metabolism , Transcriptome/genetics
5.
Dev Biol ; 427(1): 93-105, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28501476

ABSTRACT

Crosstalk between mesenchymal and epithelial cells influences organogenesis in multiple tissues, such as lung, pancreas, liver, and the nervous system. Lung mesenchyme comprises multiple cell types, however, and precise identification of the mesenchymal cell type(s) that drives early events in lung development remains unknown. Endothelial cells have been shown to be required for some aspects of lung epithelial patterning, lung stem cell differentiation, and regeneration after injury. Furthermore, endothelial cells are involved in early liver and pancreas development. From these observations we hypothesized that endothelial cells might also be required for early specification of the respiratory field and subsequent lung bud initiation. We first blocked VEGF signaling in E8.5 cultured foreguts with small molecule VEGFR inhibitors and found that lung specification and bud formation were unaltered. However, when we examined E9.5 mouse embryos carrying a mutation in the VEGFR Flk-1, which do not develop endothelial cells, we found that respiratory progenitor specification was impeded. Because the E9.5 embryos were substantially smaller than control littermates, suggesting the possibility of developmental delay, we isolated and cultured foreguts from mutant and control embryos on E8.5, when no size differences were apparent. We found that both specification of the respiratory field and lung bud formation occurred in mutant and control explants. These observations were unaffected by the presence or absence of serum. We also observed that hepatic specification and initiation occurred in the absence of endothelial cells, and that expansion of the liver epithelium in culture did not differ between mutant and control explants. Consistent with previously published results, we also found that pancreatic buds were not maintained in cultured foreguts when endothelial cells were absent. Our observations support the conclusion that endothelial cells are not required for early specification of lung progenitors and bud initiation, and that the diminished lung specification seen in E9.5 Flk-/- embryos is likely due to developmental delay resulting from the insufficient delivery of oxygen, nutrients, and other factors in the absence of a vasculature.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Lung/metabolism , Organogenesis/genetics , Animals , Cell Movement/genetics , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Endothelial Cells/cytology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Immunohistochemistry , In Situ Hybridization , Lung/cytology , Lung/embryology , Mice , Mice, Knockout , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Nuclear Factor 1 , Time Factors , Transcription Factors/genetics , Transcription Factors/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
7.
J Biol Chem ; 290(30): 18559-74, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26048993

ABSTRACT

Pulmonary surfactant, a mixture of proteins and phospholipids, plays an important role in facilitating gas exchange by maintaining alveolar stability. Saturated phosphatidylcholine (SatPC), the major component of surfactant, is synthesized both de novo and by the remodeling of unsaturated phosphatidylcholine (PC) by lyso-PC acyltransferase 1 (LPCAT1). After synthesis in the endoplasmic reticulum, SatPC is routed to lamellar bodies (LBs) for storage prior to secretion. The mechanism by which SatPC is transported to LB is not understood. The specificity of LPCAT1 for lyso-PC as an acyl acceptor suggests that formation of SatPC via LPCAT1 reacylation is a final step in SatPC synthesis prior to transport. We hypothesized that LPCAT1 forms a transient complex with SatPC and specific phospholipid transport protein(s) to initiate trafficking of SatPC from the endoplasmic reticulum to the LB. Herein we have assessed the ability of different StarD proteins to interact with LPCAT1. We found that LPCAT1 interacts with StarD10, that this interaction is direct, and that amino acids 79-271 of LPCAT1 and the steroidogenic acute regulatory protein-related lipid transfer (START) domain of START domain-containing protein 10 (StarD10) are sufficient for this interaction. The role of StarD10 in trafficking of phospholipid to LB was confirmed by the observation that knockdown of StarD10 significantly reduced transport of phospholipid to LB. LPCAT1 also interacted with one isoform of StarD7 but showed no interaction with StarD2/PC transfer protein.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Lipid Metabolism , Phospholipids/biosynthesis , Phosphoproteins/metabolism , Pulmonary Alveoli/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Humans , Mice , Phosphatidylcholines/biosynthesis , Phosphatidylcholines/metabolism , Phospholipids/metabolism , Phosphoproteins/genetics , Protein Interaction Maps/genetics , Pulmonary Alveoli/cytology , Pulmonary Surfactants/metabolism
8.
Circ Res ; 115(8): 709-20, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25091710

ABSTRACT

RATIONALE: Inactivating mutations in the Forkhead Box transcription factor F1 (FOXF1) gene locus are frequently found in patients with alveolar capillary dysplasia with misalignment of pulmonary veins, a lethal congenital disorder, which is characterized by severe abnormalities in the respiratory, cardiovascular, and gastrointestinal systems. In mice, haploinsufficiency of the Foxf1 gene causes alveolar capillary dysplasia and developmental defects in lung, intestinal, and gall bladder morphogenesis. OBJECTIVE: Although FOXF1 is expressed in multiple mesenchyme-derived cell types, cellular origins and molecular mechanisms of developmental abnormalities in FOXF1-deficient mice and patients with alveolar capillary dysplasia with misalignment of pulmonary veins remain uncharacterized because of lack of mouse models with cell-restricted inactivation of the Foxf1 gene. In the present study, the role of FOXF1 in endothelial cells was examined using a conditional knockout approach. METHODS AND RESULTS: A novel mouse line harboring Foxf1-floxed alleles was generated by homologous recombination. Tie2-Cre and Pdgfb-CreER transgenes were used to delete Foxf1 from endothelial cells. FOXF1-deficient embryos exhibited embryonic lethality, growth retardation, polyhydramnios, cardiac ventricular hypoplasia, and vascular abnormalities in the lung, placenta, yolk sac, and retina. Deletion of FOXF1 from endothelial cells reduced endothelial proliferation, increased apoptosis, inhibited vascular endothelial growth factor signaling, and decreased expression of endothelial genes critical for vascular development, including vascular endothelial growth factor receptors Flt1 and Flk1, Pdgfb, Pecam1, CD34, integrin ß3, ephrin B2, Tie2, and the noncoding RNA Fendrr. Chromatin immunoprecipitation assay demonstrated that Flt1, Flk1, Pdgfb, Pecam1, and Tie2 genes are direct transcriptional targets of FOXF1. CONCLUSIONS: FOXF1 is required for the formation of embryonic vasculature by regulating endothelial genes critical for vascular development and vascular endothelial growth factor signaling.


Subject(s)
Blood Vessels/metabolism , Endothelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis/genetics , Base Sequence , Blood Vessels/embryology , Blotting, Western , Cell Line , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Lung/blood supply , Lung/embryology , Lung/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
9.
J Pathol ; 236(1): 41-52, 2015 May.
Article in English | MEDLINE | ID: mdl-25500911

ABSTRACT

Inherited syndromes provide unique opportunities to identify key regulatory mechanisms governing human disease. We previously identified germline loss-of-function DICER1 mutations in a human syndrome defined by the childhood lung neoplasm pleuropulmonary blastoma (PPB), which arises during lung development. DICER1 regulates many biological processes critical in development and disease pathogenesis. Significant challenges in defining the role of DICER1 in human disease are identifying cause-effect relationships and generating manipulatable systems that model the complexity of organ development and disease pathogenesis. Here we report the generation of a murine model for PPB and demonstrate that precise temporal and cell type-specific Dicer1 ablation is necessary and sufficient for the development of cystic lungs that histologically and phenotypically model PPB. Dicer1 ablation in the distal airway epithelium during early stages of lung development resulted in a cystic lung phenotype indistinguishable from PPB, whereas DICER1 function was not required for development of the proximal airway epithelium or during later stages of organogenesis. Mechanistic studies demonstrate that Dicer1 loss results in epithelial cell death, followed by cystic airway dilatation accompanied by epithelial and mesenchymal proliferation. These studies define precise temporal and epithelial cell type-specific DICER1 functions in the developing lung and demonstrate that loss of these DICER1 functions is sufficient for the development of cystic PPB. These results also provide evidence that PPB arise through a novel mechanism of non-cell-autonomous tumour initiation, in which the genetic abnormality initiating the neoplasm does not occur in the cells that ultimately transform, but rather occurs in a benign-appearing epithelial cell component that predisposes underlying mesenchymal cells to malignant transformation.


Subject(s)
DEAD-box RNA Helicases/metabolism , Germ-Line Mutation/genetics , Lung Neoplasms/metabolism , Pulmonary Blastoma/metabolism , Ribonuclease III/metabolism , Animals , DEAD-box RNA Helicases/genetics , Disease Models, Animal , Epithelium/metabolism , Epithelium/pathology , Humans , Lung Neoplasms/pathology , Mice , Pulmonary Blastoma/pathology , Ribonuclease III/genetics
10.
Dev Dyn ; 244(4): 553-63, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25581492

ABSTRACT

BACKGROUND: Early lung morphogenesis is driven by tissue interactions. Signals from the lung mesenchyme drive epithelial morphogenesis, but which individual mesenchymal cell types are influencing early epithelial branching and differentiation remains unclear. It has been shown that endothelial cells are involved in epithelial repair and regeneration in the adult lung, and they may also play a role in driving early lung epithelial branching. These data, in combination with evidence that endothelial cells influence early morphogenetic events in the liver and pancreas, led us to hypothesize that endothelial cells are necessary for early lung epithelial branching. RESULTS: We blocked vascular endothelial growth factor (VEGF) signaling in embryonic day (E) 12.5 lung explants with three different VEGF receptor inhibitors (SU5416, Ki8751, and KRN633) and found that in all cases the epithelium was able to branch despite the loss of endothelial cells. Furthermore, we found that distal lung mesenchyme depleted of endothelial cells retained its ability to induce terminal branching when recombined with isolated distal lung epithelium (LgE). Additionally, isolated E12.5 primary mouse lung endothelial cells, or human lung microvascular endothelial cells (HMVEC-L), were not able to induce branching when recombined with LgE. CONCLUSIONS: Our observations support the conclusion that endothelial cells are not required for early lung branching.


Subject(s)
Endothelial Cells/cytology , Epithelial Cells/cytology , Epithelium/embryology , Lung/embryology , Lung/metabolism , Animals , Cell Differentiation , Humans , Lung/pathology , Mesoderm/metabolism , Mice , Morphogenesis , Mutation , Myocytes, Smooth Muscle/cytology , Pericytes/cytology , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
11.
Dev Biol ; 362(1): 24-41, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22094019

ABSTRACT

Hypoxia inducible factor (HIF) 1a, EPAS1 and NEPAS are expressed in the embryonic mouse lung and each isoform exhibits distinct spatiotemporal expression patterns throughout morphogenesis. To further assess the role of the HIF1a isoform in lung epithelial cell differentiation and homeostasis, we created transgenic mice that express a constitutively active isoform of human HIF-1a (HIF-1a three point mutant (TPM)), in a doxycycline-dependent manner. Expression of HIF1a TPM in the developing pulmonary epithelium resulted in lung hypoplasia characterized by defective branching morphogenesis, altered cellular energetics and impaired epithelial maturation, culminating in neonatal lethality at birth from severe respiratory distress. Histological and biochemical analyses revealed expanded glycogen pools in the pulmonary epithelial cells at E18.5, concomitant with decreased pulmonary surfactant, suggesting a delay or an arrest in maturation. Importantly, these defects occurred in the absence of apoptosis or necrosis. In addition, sub-pleural hemorrhaging was evident as early as E14.5 in HIF1a TPM lungs, despite normal patterning of the blood vasculature, consistent with defects in endothelial barrier function. Epithelial expression of HIF1a TPM also resulted in increased VEGFA and VEGFC production, an increase in the number of lymphatic vessels and indirect activation of the multiple Notch pathway components in endothelial precursor cells. Collectively, these data indicate that HIF-1a protein levels in the pulmonary epithelium must be tightly controlled for proper development of the epithelial and mesenchymal compartments.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/embryology , Lymphangiogenesis/physiology , Respiratory Mucosa/embryology , Analysis of Variance , Animals , DNA Primers/genetics , DNA, Mitochondrial/genetics , Doxycycline , Genetic Vectors/genetics , Glycogen/metabolism , Immune Sera/genetics , Immunoblotting , Immunohistochemistry , Lung/metabolism , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Phosphatidylcholines/metabolism , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/metabolism , Transgenes/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/metabolism
12.
Proc Natl Acad Sci U S A ; 106(26): 10684-9, 2009 Jun 30.
Article in English | MEDLINE | ID: mdl-19541651

ABSTRACT

Hypoxia, through the hypoxia-inducible transcription factors HIF-1alpha and HIF-2alpha (HIFs), induces angiogenesis by up-regulating a common set of angiogenic cytokines. Unlike HIF-1alpha, which regulates a unique set of genes, most genes regulated by HIF-2alpha overlap with those induced by HIF-1alpha. Thus, the unique contribution of HIF-2alpha remains largely obscure. By using adenoviral mutant HIF-1alpha and adenoviral mutant HIF-2alpha constructs, where the HIFs are transcriptionally active under normoxic conditions, we show that HIF-2alpha but not HIF-1alpha regulates adenosine A(2A) receptor in primary cultures of human lung endothelial cells. Further, siRNA knockdown of HIF-2alpha completely inhibits hypoxic induction of A(2A) receptor. Promoter studies show a 2.5-fold induction of luciferase activity with HIF-2alpha cotransfection. Analysis of the A(2A) receptor gene promoter revealed a hypoxia-responsive element in the region between -704 and -595 upstream of the transcription start site. By using a ChIP assay, we demonstrate that HIF-2alpha binding to this region is specific. In addition, we demonstrate that A(2A) receptor has angiogenic potential, as assessed by increases in cell proliferation, cell migration, and tube formation. Additional data show increased expression of A(2A) receptor in human lung tumor cancer samples relative to adjacent normal lung tissue. These data also demonstrate that A(2A) receptor is regulated by hypoxia and HIF-2alpha in human lung endothelial cells but not in mouse-derived endothelial cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Endothelial Cells/physiology , Neovascularization, Physiologic/physiology , Receptors, Adenosine A2/genetics , Amino Acids, Dicarboxylic/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia , Cell Line , Cell Movement , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression/drug effects , Humans , Luciferases/genetics , Luciferases/metabolism , Lung/cytology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Adenosine A2/metabolism , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
13.
Dev Biol ; 347(2): 301-14, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20816795

ABSTRACT

Foxm1 is a member of the Forkhead Box (Fox) family of transcription factors. Foxm1 (previously called Foxm1b, HFH-11B, Trident, Win, or MPP2) is expressed in multiple cell types and plays important roles in cellular proliferation, differentiation and tumorigenesis. Genetic deletion of Foxm1 from mouse respiratory epithelium during initial stages of lung development inhibits lung maturation and causes respiratory failure after birth. However, the role of Foxm1 during postnatal lung morphogenesis remains unknown. In the present study, Foxm1 expression was detected in epithelial cells of conducting and peripheral airways and changing dynamically with lung maturation. To discern the biological role of Foxm1 in the prenatal and postnatal lung, a novel transgenic mouse line that expresses a constitutively active form of FoxM1 (FoxM1 N-terminal deletion mutant or FoxM1-ΔN) under the control of lung epithelial-specific SPC promoter was produced. Expression of the FoxM1-ΔN transgene during embryogenesis caused epithelial hyperplasia, inhibited lung sacculation and expression of the type II epithelial marker, pro-SPC. Expression of FoxM1-ΔN mutant during the postnatal period did not influence alveologenesis but caused focal airway hyperplasia and increased proliferation of Clara cells. Likewise, expression of FoxM1-ΔN mutant in conducting airways with Scgb1a1 promoter was sufficient to induce Clara cell hyperplasia. Furthermore, FoxM1-ΔN cooperated with activated K-Ras to induce lung tumor growth in vivo. Increased activity of Foxm1 altered lung sacculation, induced proliferation in the respiratory epithelium and accelerated lung tumor growth, indicating that precise regulation of Foxm1 is critical for normal lung morphogenesis and development of lung cancer.


Subject(s)
Forkhead Transcription Factors/genetics , Lung/embryology , Lung/growth & development , Respiratory Mucosa/embryology , Respiratory Mucosa/growth & development , Animals , Cell Enlargement , Female , Forkhead Box Protein M1 , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, ras , Humans , Hyperplasia , Lung/cytology , Lung/metabolism , Lung Neoplasms/genetics , Male , Mice , Mice, Transgenic , Mutant Proteins/genetics , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism
14.
Dis Model Mech ; 2020 Dec 16.
Article in English | MEDLINE | ID: mdl-33328171

ABSTRACT

Congenital tracheomalacia, resulting from incomplete tracheal cartilage development, is a relatively common birth defect that severely impairs breathing in neonates. Mutations in the Hedgehog (HH) pathway and downstream Gli transcription factors are associated with tracheomalacia in patients and mouse models; however, the underlying molecular mechanisms are unclear. Using multiple HH/Gli mouse mutants including one that mimics Pallister-Hall Syndrome, we show that excessive Gli repressor activity prevents specification of tracheal chondrocytes. Lineage tracing experiments show that Sox9+ chondrocytes arise from HH-responsive splanchnic mesoderm in the fetal foregut that expresses the transcription factor Foxf1. Disrupted HH/Gli signaling results in 1) loss of Foxf1 which in turn is required to support Sox9+ chondrocyte progenitors and 2) a dramatic reduction in Rspo2, a secreted ligand that potentiates Wnt signaling known to be required for chondrogenesis. These results reveal a HH-Foxf1-Rspo2 signaling axis that governs tracheal cartilage development and informs the etiology of tracheomalacia.

15.
Nat Commun ; 11(1): 4158, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32855417

ABSTRACT

Visceral organs, such as the lungs, stomach and liver, are derived from the fetal foregut through a series of inductive interactions between the definitive endoderm (DE) and the surrounding splanchnic mesoderm (SM). While DE patterning is fairly well studied, the paracrine signaling controlling SM regionalization and how this is coordinated with epithelial identity is obscure. Here, we use single cell transcriptomics to generate a high-resolution cell state map of the embryonic mouse foregut. This identifies a diversity of SM cell types that develop in close register with the organ-specific epithelium. We infer a spatiotemporal signaling network of endoderm-mesoderm interactions that orchestrate foregut organogenesis. We validate key predictions with mouse genetics, showing the importance of endoderm-derived signals in mesoderm patterning. Finally, leveraging these signaling interactions, we generate different SM subtypes from human pluripotent stem cells (hPSCs), which previously have been elusive. The single cell data can be explored at: https://research.cchmc.org/ZornLab-singlecell .


Subject(s)
Digestive System/metabolism , Endoderm/metabolism , Gene Regulatory Networks , Mesoderm/metabolism , Organogenesis/genetics , Signal Transduction/genetics , Animals , Cell Lineage/genetics , Digestive System/cytology , Digestive System/embryology , Endoderm/cytology , Endoderm/embryology , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Humans , Internet , Mesoderm/cytology , Mesoderm/embryology , Mice, Inbred C57BL , Single-Cell Analysis/methods , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Nat Commun ; 11(1): 635, 2020 01 31.
Article in English | MEDLINE | ID: mdl-32005814

ABSTRACT

Multipotent Nkx2-1-positive lung epithelial primordial progenitors of the foregut endoderm are thought to be the developmental precursors to all adult lung epithelial lineages. However, little is known about the global transcriptomic programs or gene networks that regulate these gateway progenitors in vivo. Here we use bulk RNA-sequencing to describe the unique genetic program of in vivo murine lung primordial progenitors and computationally identify signaling pathways, such as Wnt and Tgf-ß superfamily pathways, that are involved in their cell-fate determination from pre-specified embryonic foregut. We integrate this information in computational models to generate in vitro engineered lung primordial progenitors from mouse pluripotent stem cells, improving the fidelity of the resulting cells through unbiased, easy-to-interpret similarity scores and modulation of cell culture conditions, including substratum elastic modulus and extracellular matrix composition. The methodology proposed here can have wide applicability to the in vitro derivation of bona fide tissue progenitors of all germ layers.


Subject(s)
Epithelial Cells/cytology , Lung/cytology , Mice/genetics , Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Epithelial Cells/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Germ Layers/embryology , Germ Layers/metabolism , Lung/embryology , Lung/metabolism , Male , Mice/embryology , Mice/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Pluripotent Stem Cells/metabolism , Signal Transduction , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/metabolism , Transcriptome , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
17.
Dev Biol ; 320(1): 149-60, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18544451

ABSTRACT

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


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Lung/cytology , Lung/embryology , Morphogenesis , Transcription Factors/metabolism , Animals , Cell Proliferation , DNA-Binding Proteins/genetics , Epithelium/embryology , Gene Expression Regulation, Developmental , Humans , Mice , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-ets/metabolism , Pulmonary Surfactant-Associated Protein C/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , Transcription Factors/genetics
18.
Dev Biol ; 316(2): 441-55, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18342301

ABSTRACT

Microarray experiments designed to identify genes differentially expressed in the E11.5 lung and trachea showed that melanoma inhibitory activity (Mia1) was expressed only in the lung. Mia1 was abundantly expressed during early lung development, but was virtually absent by the end of gestation. Distal embryonic lung epithelium showed high levels of Mia1 expression, which was suppressed by treatment with either retinoic acid or the FGF signaling antagonist SU5402. Late-gestation fetuses in which lung epithelial hyperplasia was induced by misexpression of FGF7 or FGF10 showed continued expression of Mia1 in areas of aberrant morphogenesis. Mia1 expression was also significantly increased in urethane-induced lung adenomas. Treatment of E18.5 lung explants with exogenous MIA caused significant reductions in the expression of the lung differentiation markers Sftpa, Sftpb, Sftpc, and Abca3. Bitransgenic mice expressing MIA under the control of the SFTPC promoter after E16.5, the age when Mia1 is normally silenced, died from respiratory failure at birth with morphologically immature lungs associated with reduced levels of saturated phosphatidylcholine and mature SP-B. Microarray analysis showed significant reductions in the expression of Sftpa, Sftpb, Abca3, Aqp5, Lzp-s, Scd2, and Aytl2 in lungs misexpressing MIA. These results suggest that the silencing of Mia1 that occurs in late gestation may be required for maturation of the surfactant system.


Subject(s)
Extracellular Matrix Proteins/genetics , Lung/embryology , Trachea/embryology , Animals , Cell Culture Techniques , Embryonic Development , Epithelial Cells/cytology , Gene Expression Regulation, Developmental , Genome , Mesoderm/cytology , Mice , Mice, Inbred Strains , Morphogenesis , Oligonucleotide Array Sequence Analysis , Organ Culture Techniques
19.
FASEB J ; 20(10): 1698-700, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16807366

ABSTRACT

Bronchopulmonary dysplasia (BPD), a chronic lung disease affecting preterm neonates, is associated with significant childhood and adult health problems. Histopathologic features of BPD include impaired vascular and distal airway development. We previously showed that activation of hypoxia-inducible factors (HIFs) by inhibition of prolyl hydroxylase domain-containing proteins (PHDs) is feasible and that it stimulates vascular endothelial growth factor (VEGF)-dependent angiogenesis in vitro. We tested the hypothesis that enhancement of angiogenesis by activation of HIFs improves lung growth and function in prematurely born neonates in vivo. Preterm baboons (125 day+14 day pro re nata O2 model, corresponding to 27 human gestational weeks) were treated for 14 days with intravenous (i.v.) FG-4095, a PHD inhibitor. Notably, 77% of diminished total alveolar surface area in untreated controls was recovered by FG-4095 treatment. Functional significance of the structural changes was indicated by improved oxygenation and lung compliance in FG-4095-treated newborns. Surfactant proteins B and C and saturated phosphatidylcholine were unchanged. Incidence of spontaneous ductus arteriosus closure was increased, likely contributing to lower ratio of pulmonary to systemic blood flow in FG-4095 group. These findings indicate that HIF stimulation by PHD inhibition ameliorates pathological and physiological consequences of BPD.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Lung Diseases/drug therapy , Lung/growth & development , Premature Birth , Animals , Animals, Newborn , Chronic Disease , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Female , Hypoxia-Inducible Factor 1/physiology , Lung/pathology , Lung/physiopathology , Lung Diseases/etiology , Male , Neovascularization, Physiologic/drug effects , Papio , Respiratory Function Tests , Treatment Outcome
20.
Nat Cell Biol ; 19(8): 904-914, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28737769

ABSTRACT

After influenza infection, lineage-negative epithelial progenitors (LNEPs) exhibit a binary response to reconstitute epithelial barriers: activating a Notch-dependent ΔNp63/cytokeratin 5 (Krt5) remodelling program or differentiating into alveolar type II cells (AEC2s). Here we show that local lung hypoxia, through hypoxia-inducible factor (HIF1α), drives Notch signalling and Krt5pos basal-like cell expansion. Single-cell transcriptional profiling of human AEC2s from fibrotic lungs revealed a hypoxic subpopulation with activated Notch, suppressed surfactant protein C (SPC), and transdifferentiation toward a Krt5pos basal-like state. Activated murine Krt5pos LNEPs and diseased human AEC2s upregulate strikingly similar core pathways underlying migration and squamous metaplasia. While robust, HIF1α-driven metaplasia is ultimately inferior to AEC2 reconstitution in restoring normal lung function. HIF1α deletion or enhanced Wnt/ß-catenin activity in Sox2pos LNEPs blocks Notch and Krt5 activation, instead promoting rapid AEC2 differentiation and migration and improving the quality of alveolar repair.


Subject(s)
Cell Lineage , Cell Proliferation , Cell Transdifferentiation , Epithelial Cells/metabolism , Hypoxia/metabolism , Influenza, Human/metabolism , Orthomyxoviridae Infections/metabolism , Oxygen/metabolism , Pulmonary Alveoli/metabolism , Regeneration , Animals , Cell Movement , Cells, Cultured , Disease Models, Animal , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Gene Expression Profiling , Genotype , Humans , Hypoxia/genetics , Hypoxia/pathology , Hypoxia/virology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/genetics , Influenza, Human/pathology , Influenza, Human/virology , Keratin-5/genetics , Keratin-5/metabolism , Male , Mice, Transgenic , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Phenotype , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pulmonary Alveoli/pathology , Pulmonary Alveoli/virology , Receptors, Notch/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Single-Cell Analysis , Time Factors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway
SELECTION OF CITATIONS
SEARCH DETAIL