Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Annu Rev Cell Dev Biol ; 27: 493-512, 2011.
Article in English | MEDLINE | ID: mdl-21639799

ABSTRACT

The vertebrate lung is elegantly patterned to carry out gas exchange and host defense. Similar to other organ systems, endogenous stem and progenitor cells fuel the organogenesis of the lung and maintain homeostasis in the face of normal wear and tear. In the context of acute injury, these progenitor populations are capable of effecting efficient repair. However, chronic injury, inflammation, and immune rejection frequently result in pathological airway remodeling and serious impairment of lung function. Here, we review the development, maintenance, and repair of the vertebrate respiratory system with an emphasis on the roles of epithelial stem and progenitor cells. We discuss what is currently known about their identities, lineage relationships, and the mechanisms that regulate their differentiation along various lineages. A deeper understanding of these progenitor populations will undoubtedly accelerate the discovery of improved cellular, genetic, molecular, and bioengineered therapies for lung disease.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/physiology , Lung Diseases/physiopathology , Lung/cytology , Lung/growth & development , Stem Cells/cytology , Stem Cells/physiology , Airway Remodeling , Animals , Cell Differentiation , Cell Lineage , Homeostasis , Humans , Lung/pathology , Lung/physiology , Lung Diseases/pathology , Organogenesis/physiology , Respiratory System , Wound Healing
2.
Genes Dev ; 28(17): 1929-39, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25184679

ABSTRACT

Cell type-specific conditional activation of oncogenic K-Ras is a powerful tool for investigating the cell of origin of adenocarcinomas in the mouse lung. Our previous studies showed that K-Ras activation with a CC10(Scgb1a1)-CreER driver leads to adenocarcinoma in a subset of alveolar type II cells and hyperplasia in the bronchioalveolar duct region. However, no tumors develop in the bronchioles, although recombination occurs throughout this region. To explore underlying mechanisms, we simultaneously modulated either Notch signaling or Sox2 levels in the CC10+ cells along with activation of K-Ras. Inhibition of Notch strongly inhibits adenocarcinoma formation but promotes squamous hyperplasia in the alveoli. In contrast, activation of Notch leads to widespread Sox2+, Sox9+, and CC10+ papillary adenocarcinomas throughout the bronchioles. Chromatin immunoprecipitation demonstrates Sox2 binding to NOTCH1 and NOTCH2 regulatory regions. In transgenic mouse models, overexpression of Sox2 leads to a significant reduction of Notch1 and Notch2 transcripts, while a 50% reduction in Sox2 leads to widespread papillary adenocarcinoma in the bronchioles. Taken together, our data demonstrate that the cell of origin of K-Ras-induced tumors in the lung depends on levels of Sox2 expression affecting Notch signaling. In addition, the subtype of tumors arising from type II cells is determined in part by Notch activation or suppression.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Genes, ras/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Receptors, Notch/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Gene Expression Regulation, Neoplastic , Mice , Mice, Transgenic , Protein Binding , Pulmonary Alveoli/pathology , Receptors, Notch/genetics , Signal Transduction , Transcriptional Activation/genetics
3.
Development ; 145(17)2018 08 16.
Article in English | MEDLINE | ID: mdl-30115640

ABSTRACT

The historic town of Taos, New Mexico, with its rich multicultural history of art and craft, was the site of the second Keystone Symposium on 'Endoderm Development and Disease', which was held in February 2018. The theme of the meeting was 'Cross-Organ Comparison and Interplay', emphasizing an integrative and multisystem approach to the broad topics of organ physiology, homeostasis, repair, regeneration and disease. As we review here, participants shared their recent discoveries and discussed how new technologies developed in one organ system might be applied to answer crucial questions in another. Other integrative themes were how agents such as parasites, microbes, immune cells, physical forces and innervation can affect tissue organization and progenitor cell dynamics, and how defects in the development of an organ can impact its adult function. Participants came away with a broader vision of their field and a renewed sense of collective energy empowered by novel tools and fresh ideas.


Subject(s)
Endoderm , Animals , Congresses as Topic , Humans , New Mexico
4.
Development ; 145(9)2018 05 11.
Article in English | MEDLINE | ID: mdl-29752282

ABSTRACT

The bone morphogenetic protein (BMP) signaling pathway, including antagonists, functions in lung development and regeneration of tracheal epithelium from basal stem cells. Here, we explore its role in the alveolar region, where type 2 epithelial cells (AT2s) and Pdgfrα+ type 2-associated stromal cells (TASCs) are components of the stem cell niche. We use organoids and in vivo alveolar regrowth after pneumonectomy (PNX) - a process that requires proliferation of AT2s and differentiation into type 1 cells (AT1s). BMP signaling is active in AT2s and TASCs, transiently declines post-PNX in association with upregulation of antagonists, and is restored during differentiation of AT2s to AT1s. In organoids, BMP4 inhibits AT2 proliferation, whereas antagonists (follistatin, noggin) promote AT2 self-renewal at the expense of differentiation. Gain- and loss-of-function genetic manipulation reveals that reduced BMP signaling in AT2s after PNX allows self-renewal but reduces differentiation; conversely, increased BMP signaling promotes AT1 formation. Constitutive BMP signaling in Pdgfrα+ cells reduces their AT2 support function, both after PNX and in organoid culture. Our data reveal multiple cell-type-specific roles for BMP signaling during alveolar regeneration.


Subject(s)
Alveolar Epithelial Cells/metabolism , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation/physiology , Cell Proliferation/physiology , Signal Transduction/physiology , Smad Proteins/metabolism , Stem Cells/metabolism , Alveolar Epithelial Cells/cytology , Animals , Bone Morphogenetic Protein 4/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Mice , Mice, Transgenic , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Smad Proteins/genetics , Stem Cells/cytology
5.
Development ; 144(6): 986-997, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28292845

ABSTRACT

Lungs are composed of a system of highly branched tubes that bring air into the alveoli, where gas exchange takes place. The proximal and distal regions of the lung contain epithelial cells specialized for different functions: basal, secretory and ciliated cells in the conducting airways and type II and type I cells lining the alveoli. Basal, secretory and type II cells can be grown in three-dimensional culture, with or without supporting stromal cells, and under these conditions they give rise to self-organizing structures known as organoids. This Review summarizes the different methods for generating organoids from cells isolated from human and mouse lungs, and compares their final structure and cellular composition with that of the airways or alveoli of the adult lung. We also discuss the potential and limitations of organoids for addressing outstanding questions in lung biology and for developing new drugs for disorders such as cystic fibrosis and asthma.


Subject(s)
Lung/cytology , Organoids/cytology , Alveolar Epithelial Cells/cytology , Animals , Humans , Stem Cells/cytology
6.
Development ; 143(5): 764-73, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26811382

ABSTRACT

The pseudostratified epithelium of the lung contains ciliated and secretory luminal cells and basal stem/progenitor cells. To identify signals controlling basal cell behavior we screened factors that alter their self-renewal and differentiation in a clonal organoid (tracheosphere) assay. This revealed that inhibitors of the canonical BMP signaling pathway promote proliferation but do not affect lineage choice, whereas exogenous Bmp4 inhibits proliferation and differentiation. We therefore followed changes in BMP pathway components in vivo in the mouse trachea during epithelial regeneration from basal cells after injury. The findings suggest that BMP signaling normally constrains proliferation at steady state and this brake is released transiently during repair by the upregulation of endogenous BMP antagonists. Early in repair, the packing of epithelial cells along the basal lamina increases, but density is later restored by active extrusion of apoptotic cells. Systemic administration of the BMP antagonist LDN-193189 during repair initially increases epithelial cell number but, following the shedding phase, normal density is restored. Taken together, these results reveal crucial roles for both BMP signaling and cell shedding in homeostasis of the respiratory epithelium.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Respiratory Mucosa/metabolism , Stem Cells/metabolism , Animals , Apoptosis , Basement Membrane/metabolism , Cell Differentiation , Cell Proliferation , Epithelial Cells/metabolism , Green Fluorescent Proteins/metabolism , Ligands , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Pyrazoles/chemistry , Pyrimidines/chemistry , Regeneration , Respiratory Mucosa/cytology , Signal Transduction , Trachea/metabolism , Trachea/pathology
7.
Am J Respir Cell Mol Biol ; 59(6): 706-712, 2018 12.
Article in English | MEDLINE | ID: mdl-30011373

ABSTRACT

The alveolar region of the lung is composed of two major epithelial cell types: cuboidal alveolar type 2 cells (AT2 cells), which produce surfactant proteins, and large, thin, alveolar type 1 cells (AT1 cells), specialized for efficient gas exchange. AT1 cells cover more than 95% of the alveolar surface and constitute a major barrier to the entry of pathogenic agents. Relatively few genetic tools are available for studying the development of AT1 cells, the function of genes expressed in them, and the effect of specifically killing them in vivo in the adult lung. One distinguishing feature of AT1 cells is the high level of expression of the gene Ager, encoding the advanced glycation endproduct-specific receptor, a member of the immunoglobulin superfamily of cell surface receptors. In this paper, we report the generation of a novel Ager-CreERT2 allele in which Cre recombinase is inserted into the first coding exon of the endogenous gene. After treatment with tamoxifen the allele enables Ager+ progenitor cells to be efficiently lineage labeled during late embryonic development and AT1 cells to be killed in the adult lung using a Rosa26-diphtheria toxin A allele. Significantly, adult mice in which approximately 50% of the AT1 cells are killed survive the loss; repair is associated with increased proliferation of SFTPC+ (surfactant protein C-positive) AT2 cells and the upregulation of Ager expression. The Ager-CreERT2 allele thus expands the repertoire of genetic tools for studying AT1 turnover, physiology, and repair.


Subject(s)
Homeostasis , Integrases/metabolism , Organogenesis , Pulmonary Alveoli/cytology , Pulmonary Alveoli/physiology , Receptor for Advanced Glycation End Products/metabolism , Receptors, Estrogen/metabolism , Animals , Cell Differentiation , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Receptor for Advanced Glycation End Products/genetics , Receptors, Estrogen/genetics
8.
Proc Natl Acad Sci U S A ; 112(16): 5099-104, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25840590

ABSTRACT

Telomere syndromes have their most common manifestation in lung disease that is recognized as idiopathic pulmonary fibrosis and emphysema. In both conditions, there is loss of alveolar integrity, but the underlying mechanisms are not known. We tested the capacity of alveolar epithelial and stromal cells from mice with short telomeres to support alveolar organoid colony formation and found that type 2 alveolar epithelial cells (AEC2s), the stem cell-containing population, were limiting. When telomere dysfunction was induced in adult AEC2s by conditional deletion of the shelterin component telomeric repeat-binding factor 2, cells survived but remained dormant and showed all the hallmarks of cellular senescence. Telomere dysfunction in AEC2s triggered an immune response, and this was associated with AEC2-derived up-regulation of cytokine signaling pathways that are known to provoke inflammation in the lung. Mice uniformly died after challenge with bleomycin, underscoring an essential role for telomere function in AEC2s for alveolar repair. Our data show that alveoloar progenitor senescence is sufficient to recapitulate the regenerative defects, inflammatory responses, and susceptibility to injury that are characteristic of telomere-mediated lung disease. They suggest alveolar stem cell failure is a driver of telomere-mediated lung disease and that efforts to reverse it may be clinically beneficial.


Subject(s)
Pulmonary Alveoli/pathology , Stem Cells/pathology , Telomere Shortening , Telomere/pathology , Aging/pathology , Animals , Cell Differentiation , Cell Proliferation , Epithelial Cells/metabolism , Gene Deletion , Immunity , Inflammation/pathology , Intercellular Signaling Peptides and Proteins , Mesoderm/pathology , Mice , Paracrine Communication , Peptides/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Surfactant-Associated Protein C , Signal Transduction/immunology , Spheroids, Cellular/pathology , Stromal Cells/pathology , Telomeric Repeat Binding Protein 2/metabolism , Tumor Suppressor Protein p53/metabolism
9.
Proc Natl Acad Sci U S A ; 111(35): E3641-9, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136113

ABSTRACT

The pseudostratified airway epithelium of the lung contains a balanced proportion of multiciliated and secretory luminal cells that are maintained and regenerated by a population of basal stem cells. However, little is known about how these processes are modulated in vivo, and about the potential role of cytokine signaling between stem and progenitor cells and their niche. Using a clonal 3D organoid assay, we found that IL-6 stimulated, and Stat3 inhibitors reduced, the generation of ciliated vs. secretory cells from basal cells. Gain-of-function and loss-of-function studies with cultured mouse and human basal cells suggest that IL-6/Stat3 signaling promotes ciliogenesis at multiple levels, including increases in multicilin gene and forkhead box protein J1 expression and inhibition of the Notch pathway. To test the role of IL-6 in vivo genetically, we followed the regeneration of mouse tracheal epithelium after ablation of luminal cells by inhaled SO2. Stat3 is activated in basal cells and their daughters early in the repair process, correlating with an increase in Il-6 expression in platelet-derived growth factor receptor alpha(+) mesenchymal cells in the stroma. Conditional deletion in basal cells of suppressor of cytokine signaling 3, encoding a negative regulator of the Stat3 pathway, results in an increase in multiciliated cells at the expense of secretory and basal cells. By contrast, Il-6 null mice regenerate fewer ciliated cells and an increased number of secretory cells after injury. The results support a model in which IL-6, produced in the reparative niche, functions to enhance the differentiation of basal cells, and thereby acts as a "friend" to promote airway repair rather than a "foe."


Subject(s)
Interleukin-6/metabolism , Respiratory Mucosa/cytology , STAT3 Transcription Factor/metabolism , Animals , Bronchi/cytology , Cell Differentiation/physiology , Cilia/physiology , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/physiology , Green Fluorescent Proteins/genetics , Humans , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation/physiology , Primary Cell Culture , Regeneration/physiology , Respiratory Mucosa/physiology , STAT3 Transcription Factor/genetics , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/physiology , Trachea/cytology
10.
Proc Natl Acad Sci U S A ; 110(23): 9356-61, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23690579

ABSTRACT

Most of the airways of the human lung are lined by an epithelium made up of ciliated and secretory luminal cells and undifferentiated basal progenitor cells. The integrity of this epithelium and its ability to act as a selective barrier are critical for normal lung function. In other epithelia, there is evidence that transcription factors of the evolutionarily conserved grainyheadlike (GRHL) family play key roles in coordinating multiple cellular processes required for epithelial morphogenesis, differentiation, remodeling, and repair. However, only a few target genes have been identified, and little is known about GRHL function in the adult lung. Here we focus on the role of GRHL2 in primary human bronchial epithelial cells, both as undifferentiated progenitors and as they differentiate in air-liquid interface culture into an organized mucociliary epithelium with transepithelial resistance. Using a dominant-negative protein or shRNA to inhibit GRHL2, we follow changes in epithelial phenotype and gene transcription using RNA sequencing or microarray analysis. We identify several hundreds of genes that are directly or indirectly regulated by GRHL2 in both undifferentiated cells and air-liquid interface cultures. Using ChIP sequencing to map sites of GRHL2 binding in the basal cells, we identify 7,687 potential primary targets and confirm that GRHL2 binding is strongly enriched near GRHL2-regulated genes. Taken together, the results support the hypothesis that GRHL2 plays a key role in regulating many physiological functions of human airway epithelium, including those involving cell morphogenesis, adhesion, and motility.


Subject(s)
DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Respiratory Mucosa/physiology , Transcription Factors/metabolism , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Chromatin Immunoprecipitation , DNA-Binding Proteins/antagonists & inhibitors , Epithelial Cells/metabolism , Gene Expression Regulation/physiology , Genetic Vectors , Humans , Immunohistochemistry , Lentivirus , Microarray Analysis , Morphogenesis/genetics , Morphogenesis/physiology , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/metabolism , Sequence Analysis, RNA , Transcription Factors/antagonists & inhibitors
11.
Proc Natl Acad Sci U S A ; 109(13): 4910-5, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22411819

ABSTRACT

Identifying the cells of origin of lung cancer may lead to new therapeutic strategies. Previous work has focused upon the putative bronchoalveolar stem cell at the bronchioalveolar duct junction as a cancer cell of origin when a codon 12 K-Ras mutant is induced via adenoviral Cre inhalation. In the present study, we use two "knock-in" Cre-estrogen receptor alleles to inducibly express K-RasG12D in CC10(+) epithelial cells and Sftpc(+) type II alveolar cells of the adult mouse lung. Analysis of these mice identifies type II cells, Clara cells in the terminal bronchioles, and putative bronchoalveolar stem cells as cells of origin for K-Ras-induced lung hyperplasia. However, only type II cells appear to progress to adenocarcinoma.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Animals , Bronchioles/metabolism , Bronchioles/pathology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Disease Progression , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/metabolism , Hyperplasia , Intercellular Signaling Peptides and Proteins , Lung Neoplasms/genetics , Mice , Models, Biological , Mutant Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Peptides/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Pulmonary Surfactant-Associated Protein C , SOXB1 Transcription Factors/metabolism , Time Factors , Transcriptome/genetics , Uteroglobin/metabolism
12.
Proc Natl Acad Sci U S A ; 109(40): 16354-9, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22988107

ABSTRACT

Mucous cell hyperplasia and airway smooth muscle (ASM) hyperresponsiveness are hallmark features of inflammatory airway diseases, including asthma. Here, we show that the recently identified calcium-activated chloride channel (CaCC) TMEM16A is expressed in the adult airway surface epithelium and ASM. The epithelial expression is increased in asthmatics, particularly in secretory cells. Based on this and the proposed functions of CaCC, we hypothesized that TMEM16A inhibitors would negatively regulate both epithelial mucin secretion and ASM contraction. We used a high-throughput screen to identify small-molecule blockers of TMEM16A-CaCC channels. We show that inhibition of TMEM16A-CaCC significantly impairs mucus secretion in primary human airway surface epithelial cells. Furthermore, inhibition of TMEM16A-CaCC significantly reduces mouse and human ASM contraction in response to cholinergic agonists. TMEM16A-CaCC blockers, including those identified here, may positively impact multiple causes of asthma symptoms.


Subject(s)
Chloride Channels/metabolism , Mucins/metabolism , Muscle Contraction/physiology , Muscle, Smooth/physiology , Respiratory System/cytology , Respiratory System/metabolism , Animals , Anoctamin-1 , Cells, Cultured , Epithelial Cells/metabolism , Humans , Immunohistochemistry , Mice , Microscopy, Fluorescence
13.
Proc Natl Acad Sci U S A ; 108(52): E1475-83, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22123957

ABSTRACT

There are currently few treatment options for pulmonary fibrosis. Innovations may come from a better understanding of the cellular origin of the characteristic fibrotic lesions. We have analyzed normal and fibrotic mouse and human lungs by confocal microscopy to define stromal cell populations with respect to several commonly used markers. In both species, we observed unexpected heterogeneity of stromal cells. These include numerous cells with molecular and morphological characteristics of pericytes, implicated as a source of myofibroblasts in other fibrotic tissues. We used mouse genetic tools to follow the fates of specific cell types in the bleomcyin-induced model of pulmonary fibrosis. Using inducible transgenic alleles to lineage trace pericyte-like cells in the alveolar interstitium, we show that this population proliferates in fibrotic regions. However, neither these cells nor their descendants express high levels of the myofibroblast marker alpha smooth muscle actin (Acta2, aSMA). We then used a Surfactant protein C-CreER(T2) knock-in allele to follow the fate of Type II alveolar cells (AEC2) in vivo. We find no evidence at the cellular or molecular level for epithelial to mesenchymal transition of labeled cells into myofibroblasts. Rather, bleomycin accelerates the previously reported conversion of AEC2 into AEC1 cells. Similarly, epithelial cells labeled with our Scgb1a1-CreER allele do not give rise to fibroblasts but generate both AEC2 and AEC1 cells in response to bleomycin-induced lung injury. Taken together, our results show a previously unappreciated heterogeneity of cell types proliferating in fibrotic lesions and exclude pericytes and two epithelial cell populations as the origin of myofibroblasts.


Subject(s)
Cell Differentiation/physiology , Pulmonary Alveoli/cytology , Pulmonary Fibrosis/pathology , Stromal Cells/cytology , Actins/metabolism , Animals , Biomarkers/metabolism , Bleomycin/toxicity , Bromodeoxyuridine , Cell Proliferation , Epithelial-Mesenchymal Transition/physiology , Extracellular Matrix Proteins/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Mice , Myofibroblasts/cytology , Pericytes/metabolism , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/chemically induced , Real-Time Polymerase Chain Reaction , Stromal Cells/metabolism
14.
Am J Respir Cell Mol Biol ; 49(5): 788-97, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23742075

ABSTRACT

Bronchiolitis obliterans (BO) is a major cause of chronic airway dysfunction after toxic chemical inhalation. The pathophysiology of BO is not well understood, but epithelial cell injury has been closely associated with the development of fibrotic lesions in human studies and in animal models of both toxin-induced and transplant-induced BO. However, whereas almost all cases and models of BO include epithelial injury, not all instances of epithelial injury result in BO, suggesting that epithelial damage per se is not the critical event leading to the development of BO. Here, we describe a model of chlorine-induced BO in which mice develop tracheal and large airway obliterative lesions within 10 days of exposure to high (350 parts per million [ppm]), but not low (200 ppm), concentrations of chlorine gas. Importantly, these lesions arise only under conditions and in areas in which basal cells, the resident progenitor cells for large airway epithelium, are eliminated by chlorine exposure. In areas of basal cell loss, epithelial regeneration does not occur, resulting in persistent regions of epithelial denudation. Obliterative airway lesions arise specifically from regions of epithelial denudation in a process that includes inflammatory cell infiltration by Day 2 after exposure, fibroblast infiltration and collagen deposition by Day 5, and the ingrowth of blood vessels by Day 7, ultimately leading to lethal airway obstruction by Days 9-12. We conclude that the loss of epithelial progenitor cells constitutes a critical factor leading to the development of obliterative airway lesions after chemical inhalation.


Subject(s)
Bronchi/pathology , Bronchiolitis Obliterans/pathology , Chlorine , Epithelial Cells/pathology , Respiratory Mucosa/pathology , Stem Cells/pathology , Trachea/pathology , Animals , Bronchi/metabolism , Bronchiolitis Obliterans/chemically induced , Bronchiolitis Obliterans/metabolism , Cell Death , Collagen/metabolism , Disease Models, Animal , Disease Progression , Epithelial Cells/metabolism , Female , Fibrosis , Gases , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Inhalation Exposure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology , Re-Epithelialization , Respiratory Mucosa/metabolism , Stem Cells/metabolism , Time Factors , Trachea/metabolism
15.
Development ; 137(24): 4171-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21068065

ABSTRACT

The stratification and differentiation of the epidermis are known to involve the precise control of multiple signaling pathways. By contrast, little is known about the development of the mouse esophagus and forestomach, which are composed of a stratified squamous epithelium. Based on prior work in the skin, we hypothesized that bone morphogenetic protein (BMP) signaling is a central player. To test this hypothesis, we first used a BMP reporter mouse line harboring a BRE-lacZ allele, along with in situ hybridization to localize transcripts for BMP signaling components, including various antagonists. We then exploited a Shh-Cre allele that drives recombination in the embryonic foregut epithelium to generate gain- or loss-of-function models for the Bmpr1a (Alk3) receptor. In gain-of-function (Shh-Cre;Rosa26(CAG-loxpstoploxp-caBmprIa)) embryos, high levels of ectopic BMP signaling stall the transition from simple columnar to multilayered undifferentiated epithelium in the esophagus and forestomach. In loss-of-function experiments, conditional deletion of the BMP receptor in Shh-Cre;Bmpr1a(flox/flox) embryos allows the formation of a multilayered squamous epithelium but this fails to differentiate, as shown by the absence of expression of the suprabasal markers loricrin and involucrin. Together, these findings suggest multiple roles for BMP signaling in the developing esophagus and forestomach.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Esophagus/cytology , Esophagus/metabolism , Gastric Mucosa/metabolism , Signal Transduction/physiology , Stomach/cytology , Animals , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Epithelium/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Signal Transduction/genetics
16.
Development ; 136(22): 3741-5, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19855016

ABSTRACT

The conducting airways (bronchi and bronchioles) and peripheral gas exchange (alveolar) regions of the mammalian lung are generated by a process of branching morphogenesis. Evidence suggests that during embryonic development, the undifferentiated epithelial progenitors are located at the distal tips of the branching epithelium. To test this hypothesis, we used an Id2-CreER(T2) knock-in mouse strain to lineage trace the distal epithelial tip cells during either the pseudoglandular or canalicular phases of development. During the pseudoglandular stage, the tip cells both self-renew and contribute descendents to all epithelial cell lineages, including neuroendocrine cells. In addition, individual Id2(+) tip cells can self-renew and contribute descendents to both the bronchiolar and alveolar compartments. By contrast, during the later canalicular stage, the distal epithelial tip cells only contribute descendents to the alveoli. Taken together, this evidence supports a model in which the distal tip of the developing lung contains a multipotent epithelial population, the fate of which changes during development.


Subject(s)
Embryonic Stem Cells/cytology , Inhibitor of Differentiation Protein 2/metabolism , Lung/cytology , Multipotent Stem Cells/cytology , Animals , Epithelium/metabolism , Gene Knock-In Techniques , Inhibitor of Differentiation Protein 2/genetics , Mice , Pulmonary Alveoli/cytology
17.
Proc Natl Acad Sci U S A ; 106(31): 12771-5, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19625615

ABSTRACT

The pseudostratified epithelium of the mouse trachea and human airways contains a population of basal cells expressing Trp-63 (p63) and cytokeratins 5 (Krt5) and Krt14. Using a KRT5-CreER(T2) transgenic mouse line for lineage tracing, we show that basal cells generate differentiated cells during postnatal growth and in the adult during both steady state and epithelial repair. We have fractionated mouse basal cells by FACS and identified 627 genes preferentially expressed in a basal subpopulation vs. non-BCs. Analysis reveals potential mechanisms regulating basal cells and allows comparison with other epithelial stem cells. To study basal cell behaviors, we describe a simple in vitro clonal sphere-forming assay in which mouse basal cells self-renew and generate luminal cells, including differentiated ciliated cells, in the absence of stroma. The transcriptional profile identified 2 cell-surface markers, ITGA6 and NGFR, which can be used in combination to purify human lung basal cells by FACS. Like those from the mouse trachea, human airway basal cells both self-renew and generate luminal daughters in the sphere-forming assay.


Subject(s)
Lung/cytology , Stem Cells/cytology , Trachea/cytology , Animals , Cell Proliferation , Cells, Cultured , Female , Gene Expression Profiling , Humans , Male , Mice , Oligonucleotide Array Sequence Analysis , Receptor, Nerve Growth Factor/analysis , Trachea/metabolism
18.
Am J Gastroenterol ; 106(6): 1039-47, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21448147

ABSTRACT

OBJECTIVES: An early event in the pathogenesis of gastroesophageal reflux disease (GERD) is an acid-induced increase in junctional (paracellular) permeability in esophageal epithelium (EE). The molecular events that account for this change are unknown. E-cadherin is a junctional protein important in barrier function in EE. Therefore, defects in barrier function in EE were sought in GERD as well as whether their presence correlated with abnormalities in e-cadherin. METHODS: Endoscopic biopsies of EE from GERD (n=20; male 10; female 10; mean age 50 ± 10 years) and subjects with a healthy esophagus (controls; n=23; male 11; female 12; mean age 51 ± 11 years) were evaluated in mini-Ussing chambers and by western blot and immunochemistry; and serum analyzed by enzyme-linked immunosorbent assay (ELISA). A role for e-cadherin was also assessed using a unique conditional knockout of e-cadherin in adult mouse esophagus. RESULTS: EE from GERD patients had lower electrical resistance and higher fluorescein flux than EE from controls; and the findings in GERD associated with cleavage of e-cadherin. Cleavage of e-cadherin in GERD was documented in EE by the presence of a 35-kDa, C-terminal fragment of the molecule on western blot and by an increase in soluble N-terminal fragments of the molecule in serum. Activation of the membrane metalloproteinase, A Disintegrin And Metalloproteinase (ADAM-10), was identified as a likely cause for cleavage of e-cadherin by western blot and immunostaining and a role for e-cadherin in the increased junctional permeability in EE from GERD supported by showing increased permeability after deletion of e-cadherin in mouse EE. CONCLUSIONS: The EE in GERD has increased junctional permeability and this is in association with proteolytic cleavage of e-cadherin. As loss of e-cadherin can, alone, account for the increase in junctional permeability, cleavage of e-cadherin likely represents a critical molecular event in the pathogenesis of GERD, and identification of cleaved fragments may, if confirmed in larger studies, be valuable as a biomarker of GERD.


Subject(s)
Cadherins/metabolism , Esophagus/pathology , Gastroesophageal Reflux/metabolism , Gastroesophageal Reflux/pathology , Adolescent , Adult , Aged , Animals , Biomarkers/analysis , Biomarkers/metabolism , Biopsy, Needle , Blotting, Western , Cadherins/analysis , Case-Control Studies , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Esophagogastric Junction/pathology , Esophagogastric Junction/physiopathology , Esophagoscopy/methods , Female , Fluorescent Antibody Technique , Gastroesophageal Reflux/physiopathology , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout , Middle Aged , Prognosis , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Severity of Illness Index
19.
J Cell Biol ; 172(7): 1045-56, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16567502

ABSTRACT

Charged MVB protein 5 (CHMP5) is a coiled coil protein homologous to the yeast Vps60/Mos10 gene and other ESCRT-III complex members, although its precise function in either yeast or mammalian cells is unknown. We deleted the CHMP5 gene in mice, resulting in a phenotype of early embryonic lethality, reflecting defective late endosome function and dysregulation of signal transduction. Chmp5-/- cells exhibit enlarged late endosomal compartments that contain abundant internal vesicles expressing proteins that are characteristic of late endosomes and lysosomes. This is in contrast to ESCRT-III mutants in yeast, which are defective in multivesicular body (MVB) formation. The degradative capacity of Chmp5-/- cells was reduced, and undigested proteins from multiple pathways accumulated in enlarged MVBs that failed to traffic their cargo to lysosomes. Therefore, CHMP5 regulates late endosome function downstream of MVB formation, and the loss of CHMP5 enhances signal transduction by inhibiting lysosomal degradation of activated receptors.


Subject(s)
Carrier Proteins/physiology , Embryonic Development/physiology , Endosomes/physiology , Signal Transduction/physiology , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Cell Line , Cells, Cultured , Down-Regulation , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Endocytosis/genetics , Endocytosis/physiology , Endosomal Sorting Complexes Required for Transport , Gene Expression Regulation, Developmental/genetics , Histocompatibility Antigens Class II/metabolism , Horseradish Peroxidase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , NIH 3T3 Cells , Phenotype , Phosphorylation , Protein Serine-Threonine Kinases , RNA, Small Interfering/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Sequence Homology, Amino Acid , Signal Transduction/genetics , Stem Cells/metabolism , Transfection
20.
Proc Natl Acad Sci U S A ; 105(43): 16626-30, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18922767

ABSTRACT

During mouse development, the sophisticated vascular network of the lung is established from embryonic day (E) approximately 10.5 and continues to develop postnatally. This network is composed of endothelial cells enclosed by vascular smooth muscle, pericytes, and other mesenchymal cells. Recent in vivo lineage labeling studies in the developing heart and intestine suggest that some of the vascular smooth muscle cells arise from the surface mesothelium. In the developing lung, the Wilm's tumor 1 gene (Wt1) is expressed only in the mesothelial cells. Therefore, we lineage-labeled the mesothelium in vivo by using a Wt1-Cre transgene in combination with either Rosa26R(lacZ), Rosa26R(CAG-hPLAP), or Rosa26R(EYFP) reporter alleles. In all three cases, cells derived from lineage-labeled mesothelium are found inside the lung and as smooth muscle actin (SMA) and PDGF receptor-beta positive cells in the walls of pulmonary blood vessels. To corroborate this finding, we used 5-(and-6)-carboxy-2',7'-dichlorofluorescein diacetate, succinimidyl ester "mixed isomers" (CCFSE) dye to label mesothelial cells on the surface of the embryonic lung. Over the course of 72-h culture, dye-labeled cells also appear within the lung mesenchyme. Together, our data provide evidence that mesothelial cells serve as a source of vascular smooth muscle cells in the developing lung and suggest that a conserved mechanism applies to the development of blood vessels in all coelomic organs.


Subject(s)
Epithelium/physiology , Lung/blood supply , Lung/growth & development , Animals , Blood Vessels/embryology , Blood Vessels/growth & development , Cell Lineage , Embryo, Mammalian , Fluorescent Dyes , Genes, Reporter , Lung/embryology , Mesoderm , Mice , Muscle, Smooth, Vascular , Organogenesis , Receptor, Platelet-Derived Growth Factor beta/analysis
SELECTION OF CITATIONS
SEARCH DETAIL