Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 4956, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37653024

ABSTRACT

The molecular etiology of idiopathic pulmonary fibrosis (IPF) has been extensively investigated to identify new therapeutic targets. Although anti-inflammatory treatments are not effective for patients with IPF, damaged alveolar epithelial cells play a critical role in lung fibrogenesis. Here, we establish an organoid-based lung fibrosis model using mouse and human lung tissues to assess the direct communication between damaged alveolar type II (AT2)-lineage cells and lung fibroblasts by excluding immune cells. Using this in vitro model and mouse genetics, we demonstrate that bleomycin causes DNA damage and activates p53 signaling in AT2-lineage cells, leading to AT2-to-AT1 transition-like state with a senescence-associated secretory phenotype (SASP). Among SASP-related factors, TGF-ß plays an exclusive role in promoting lung fibroblast-to-myofibroblast differentiation. Moreover, the autocrine TGF-ß-positive feedback loop in AT2-lineage cells is a critical cellular system in non-inflammatory lung fibrogenesis. These findings provide insights into the mechanism of IPF and potential therapeutic targets.


Subject(s)
Idiopathic Pulmonary Fibrosis , Transforming Growth Factor beta , Humans , Animals , Mice , Feedback , Alveolar Epithelial Cells , Idiopathic Pulmonary Fibrosis/genetics , Cell Differentiation
2.
Stem Cells ; 41(8): 809-820, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37468433

ABSTRACT

Single-cell RNA sequencing is a valuable tool for dissecting cellular heterogeneity in complex systems. However, it is still challenging to estimate the proliferation and differentiation potentials of subpopulations within dormant tissue stem cells. Here, we established a new single-cell analysis method for profiling the organoid-forming capacity and differentiation potential of tissue stem cells to disclose stem cell subpopulations by integrating single-cell morphometrics, organoid-forming assay, and RNA sequencing, a method named scMORN. To explore lung epithelial stem cells, we initially developed feeder-free culture system, which could expand all major lung stem cells, including basal, club, and alveolar type 2 (AT2) cells, and found that club cells contained a subpopulation, which showed better survival rate and high proliferation capacity and could differentiate into alveolar cells. Using the scMORN method, we discovered a club cell subpopulation named Muc5b+ and large club (ML-club) cells that efficiently formed organoids than other club or AT2 cells in our feeder-free organoid culture and differentiated into alveolar cells in vitro. Single-cell transcriptome profiling and immunohistochemical analysis revealed that ML-club cells localized at the intrapulmonary proximal airway and distinct from known subpopulations of club cells such as BASCs. Furthermore, we identified CD14 as a cell surface antigen of ML-club cells and showed that purified CD14+ club cells engrafted into injured mouse lungs had better engraftment rate and expansion than other major lung stem cells, reflecting the observations in organoid culture systems. The scMORN method could be adapted to different stem cell tissues to discover useful stem-cell subpopulations.


Subject(s)
Lung , Transcriptome , Animals , Mice , Transcriptome/genetics , Stem Cells/metabolism , Organoids/metabolism , Gene Expression Profiling , Cell Differentiation
3.
Nat Protoc ; 17(11): 2699-2719, 2022 11.
Article in English | MEDLINE | ID: mdl-35978039

ABSTRACT

Development of visceral organs such as the esophagus, lung, liver and stomach are coordinated by reciprocal signaling interactions between the endoderm and adjacent mesoderm cells in the fetal foregut. Although the recent successes in recapitulating developmental signaling in vitro has enabled the differentiation of human pluripotent stem cells (hPSCs) into various types of organ-specific endodermal epithelium, the generation of organ-specific mesenchyme has received much less attention. This is a major limitation in ongoing efforts to engineer complex human tissue. Here, we describe a protocol to differentiate hPSCs into different types of organ-specific mesoderm, leveraging signaling networks and molecular markers elucidated from single-cell transcriptomics of mouse foregut organogenesis. Building on established methods, hPSC-derived lateral plate mesoderm treated with either retinoic acid (RA) or RA together with a Hedgehog (HH) agonist generates posterior or anterior foregut splanchnic mesoderm, respectively, after 4-d cultures. These are directed into organ-specific mesenchyme lineages by the combinatorial activation or inhibition of WNT, BMP, RA or HH pathways from days 4 to 7 in cultures. By day 7, the cultures are enriched for different types of mesoderm with distinct molecular signatures: 60-90% pure liver septum transversum/mesothelium-like, 70-80% pure liver-like fibroblasts and populations of ~35% respiratory-like mesoderm, gastric-like mesoderm or esophageal-like mesoderm. This protocol can be performed by anyone with moderate experience differentiating hPSCs, provides a novel platform to study human mesoderm development and can be used to engineer more complex foregut tissue for disease modeling and regenerative medicine.


Subject(s)
Hedgehog Proteins , Pluripotent Stem Cells , Humans , Mice , Animals , Hedgehog Proteins/metabolism , Mesoderm , Endoderm , Cell Differentiation , Tretinoin/pharmacology , Lung
4.
Kobe J Med Sci ; 67(2): E61-E70, 2021 Nov 02.
Article in English | MEDLINE | ID: mdl-34795157

ABSTRACT

Initially, endothelin (ET)-2 was described as an endothelium-derived vasoconstrictor. However, accumulating evidence suggests the involvement of ET-2 in non-cardiovascular physiology and disease pathophysiology. The deficiency of ET-2 in mice can be lethal, and such mice exhibit a distinct developmental abnormality in the lungs. Nonetheless, the definite role of ET-2 in the lungs remains unclear. The ET-2 isoform, ET-1, promotes pulmonary fibrosis in mice. Although endothelin receptor antagonists (ERAs) show improvements in bleomycin-induced pulmonary fibrosis in mouse models, clinical trials examining ERAs for pulmonary fibrosis treatment have been unsuccessful, even showing harmful effects in patients. We hypothesized that ET-2, which activates the same receptor as ET-1, plays a distinct role in pulmonary fibrosis. In this study, we showed that ET-2 is expressed in the lung epithelium, and ET-2 deletion in epithelial cells of mice results in the exacerbation of bleomycin-induced pulmonary fibrosis. ET-2 knockdown in lung epithelial cell lines resulted in increased apoptosis mediated via oxidative stress induction. In contrast to the effects of ET-1, which induced fibroblast activation, ET-2 hampered fibroblast activation in primary mouse lung fibroblast cells by inhibiting the TGF-ß-SMAD2/3 pathway. Our results demonstrated the divergent roles of ET-1 and ET-2 in pulmonary fibrosis pathophysiology and suggested that ET-2, expressed in epithelial cells, exerts protective effects against the development of pulmonary fibrosis in mice.


Subject(s)
Bleomycin/toxicity , Endothelin-2/metabolism , Lung/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Animals , Bleomycin/administration & dosage , Epithelial Cells , Epithelium/metabolism , Epithelium/pathology , Lung/pathology , Mice , Transforming Growth Factor beta/metabolism
5.
Commun Biol ; 4(1): 1204, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34671097

ABSTRACT

Congenital malformations cause life-threatening diseases in pediatrics, yet the molecular mechanism of organogenesis is poorly understood. Here we show that Dyrk2-deficient mice display congenital malformations in multiple organs. Transcriptome analysis reveals molecular pathology of Dyrk2-deficient mice, particularly with respect to Foxf1 reduction. Mutant pups exhibit sudden death soon after birth due to respiratory failure. Detailed analyses of primordial lungs at the early developmental stage demonstrate that Dyrk2 deficiency leads to altered airway branching and insufficient alveolar development. Furthermore, the Foxf1 expression gradient in mutant lung mesenchyme is disrupted, reducing Foxf1 target genes, which are necessary for proper airway and alveolar development. In ex vivo lung culture system, we rescue the expression of Foxf1 and its target genes in Dyrk2-deficient lung by restoring Shh signaling activity. Taken together, we demonstrate that Dyrk2 is essential for embryogenesis and its disruption results in congenital malformation.


Subject(s)
Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Gene Expression , Lung Diseases/genetics , Protein Serine-Threonine Kinases/deficiency , Protein-Tyrosine Kinases/deficiency , Animals , Forkhead Transcription Factors/metabolism , Lung Diseases/congenital , Mice , Dyrk Kinases
6.
Development ; 148(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34228796

ABSTRACT

The trachea delivers inhaled air into the lungs for gas exchange. Anomalies in tracheal development can result in life-threatening malformations, such as tracheoesophageal fistula and tracheomalacia. Given the limitations of current therapeutic approaches, development of technologies for the reconstitution of a three-dimensional trachea from stem cells is urgently required. Recently, single-cell sequencing technologies and quantitative analyses from cell to tissue scale have been employed to decipher the cellular basis of tracheal morphogenesis. In this Review, recent advances in mammalian tracheal development and the generation of tracheal tissues from pluripotent stem cells are summarized.


Subject(s)
Lung/growth & development , Morphogenesis/physiology , Trachea/growth & development , Tracheoesophageal Fistula/pathology , Animals , Cartilage/growth & development , Cell Differentiation , Epithelium , Humans , Mesoderm/growth & development , Mice , Morphogenesis/genetics , Respiratory System , Trachea/abnormalities , Tracheomalacia , Transcriptome
7.
Dev Cell ; 56(13): 1917-1929.e9, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34129836

ABSTRACT

During development, quiescent airway basal stem cells are derived from proliferative primordial progenitors through the cell-cycle slowdown. In contrast, basal cells contribute to adult tissue regeneration by shifting from slow cycling to proliferating and subsequently back to slow cycling. Although sustained proliferation results in tumorigenesis, the molecular mechanisms regulating these transitions remain unknown. Using temporal single-cell transcriptomics of developing murine airway progenitors and genetic validation experiments, we found that TGF-ß signaling decelerated cell cycle by inhibiting Id2 and contributed to slow-cycling basal cell specification during development. In adult tissue regeneration, reduced TGF-ß signaling restored Id2 expression and initiated regeneration. Id2 overexpression and Tgfbr2 knockout enhanced epithelial proliferation; however, persistent Id2 expression drove basal cell hyperplasia that resembled a precancerous state. Together, the TGF-ß-Id2 axis commonly regulates the proliferation transitions in basal cells during development and regeneration, and its fine-tuning is critical for normal regeneration while avoiding basal cell hyperplasia.


Subject(s)
Cell Proliferation/genetics , Inhibitor of Differentiation Protein 2/genetics , Regeneration/genetics , Transforming Growth Factor beta/genetics , Animals , Cell Differentiation/genetics , Epithelial Cells/cytology , Humans , Lung/growth & development , Mice , Respiratory System/growth & development , Stem Cells/cytology
8.
Dev Dyn ; 250(11): 1552-1567, 2021 11.
Article in English | MEDLINE | ID: mdl-33840142

ABSTRACT

The trachea is a rigid air duct with some mobility, which comprises the upper region of the respiratory tract and delivers inhaled air to alveoli for gas exchange. During development, the tracheal primordium is first established at the ventral anterior foregut by interactions between the epithelium and mesenchyme through various signaling pathways, such as Wnt, Bmp, retinoic acid, Shh, and Fgf, and then segregates from digestive organs. Abnormalities in this crosstalk result in lethal congenital diseases, such as tracheal agenesis. Interestingly, these molecular mechanisms also play roles in tissue regeneration in adulthood, although it remains less understood compared with their roles in embryonic development. In this review, we discuss cellular and molecular mechanisms of trachea development that regulate the morphogenesis of this simple tubular structure and identities of individual differentiated cells. We also discuss how the facultative regeneration capacity of the epithelium is established during development and maintained in adulthood.


Subject(s)
Gene Expression Regulation, Developmental , Organogenesis , Endoderm/metabolism , Female , Humans , Mesoderm/metabolism , Organogenesis/physiology , Pregnancy , Trachea/abnormalities
9.
Stem Cell Res Ther ; 12(1): 54, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436065

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is an incurable and debilitating chronic disease characterized by progressive airflow limitation associated with abnormal levels of tissue inflammation. Therefore, stem cell-based approaches to tackle the condition are currently a focus of regenerative therapies for COPD. Extracellular vesicles (EVs) released by all cell types are crucially involved in paracrine, extracellular communication. Recent advances in the field suggest that stem cell-derived EVs possess a therapeutic potential which is comparable to the cells of their origin. METHODS: In this study, we assessed the potential anti-inflammatory effects of human umbilical cord mesenchymal stem cell (hUC-MSC)-derived EVs in a rat model of COPD. EVs were isolated from hUC-MSCs and characterized by the transmission electron microscope, western blotting, and nanoparticle tracking analysis. As a model of COPD, male Sprague-Dawley rats were exposed to cigarette smoke for up to 12 weeks, followed by transplantation of hUC-MSCs or application of hUC-MSC-derived EVs. Lung tissue was subjected to histological analysis using haematoxylin and eosin staining, Alcian blue-periodic acid-Schiff (AB-PAS) staining, and immunofluorescence staining. Gene expression in the lung tissue was assessed using microarray analysis. Statistical analyses were performed using GraphPad Prism 7 version 7.0 (GraphPad Software, USA). Student's t test was used to compare between 2 groups. Comparison among more than 2 groups was done using one-way analysis of variance (ANOVA). Data presented as median ± standard deviation (SD). RESULTS: Both transplantation of hUC-MSCs and application of EVs resulted in a reduction of peribronchial and perivascular inflammation, alveolar septal thickening associated with mononuclear inflammation, and a decreased number of goblet cells. Moreover, hUC-MSCs and EVs ameliorated the loss of alveolar septa in the emphysematous lung of COPD rats and reduced the levels of NF-κB subunit p65 in the tissue. Subsequent microarray analysis revealed that both hUC-MSCs and EVs significantly regulate multiple pathways known to be associated with COPD. CONCLUSIONS: In conclusion, we show that hUC-MSC-derived EVs effectively ameliorate by COPD-induced inflammation. Thus, EVs could serve as a new cell-free-based therapy for the treatment of COPD.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Pulmonary Disease, Chronic Obstructive , Animals , Humans , Inflammation/therapy , Male , Pulmonary Disease, Chronic Obstructive/therapy , Rats , Rats, Sprague-Dawley , Umbilical Cord
10.
Neurosci Res ; 170: 122-132, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33309869

ABSTRACT

During mammalian corticogenesis, Notch signaling is essential to maintain neural stem cells called radial glial cells (RGCs) and the cortical architecture. Because the conventional knockout of either Notch1 or Notch2 causes a neuroepithelial loss prior to neurogenesis, their functional relationship in RGCs remain elusive. Here, we investigated the impacts of single knockout of Notch1 and Notch2 genes, and their conditional double knockout (DKO) on mouse corticogenesis. We demonstrated that Notch1 single knockout affected RGC maintenance in early to mid-neurogenesis whereas Notch2 knockout caused no apparent defect. In contrast, Notch2 plays a role in the RGC maintenance as Notch1 does at the late stage. Notch1 and Notch2 DKO resulted in the complete loss of RGCs, suggesting their cooperative function. We found that Notch activity in RGCs depends on the Notch gene dosage irrespective of Notch1 or Notch2 at late neurogenic stage, and that Notch1 and Notch2 have a similar activity, most likely due to a drastic increase in Notch2 transcription. Our results revealed that Notch1 has an essential role in establishing the RGC pool during the early stage, whereas Notch1 and Notch2 subsequently exhibit a comparable function for RGC maintenance and neurogenesis in the late neurogenic period in the mouse telencephalon.


Subject(s)
Neural Stem Cells , Receptor, Notch1 , Animals , Ependymoglial Cells , Mice , Neurogenesis , Receptor, Notch1/genetics , Signal Transduction
11.
Dis Model Mech ; 13(12)2020 12 21.
Article in English | MEDLINE | ID: mdl-33355253

ABSTRACT

Mammalian lungs have the ability to recognize external environments by sensing different compounds in inhaled air. Pulmonary neuroendocrine cells (PNECs) are rare, multi-functional epithelial cells currently garnering attention as intrapulmonary sensors; PNECs can detect hypoxic conditions through chemoreception. Because PNEC overactivation has been reported in patients suffering from respiratory diseases - such as asthma, chronic obstructive pulmonary disease, bronchopulmonary dysplasia and other congenital diseases - an improved understanding of the fundamental characteristics of PNECs is becoming crucial in pulmonary biology and pathology. During the past decade, murine genetics and disease models revealed the involvement of PNECs in lung ventilation dynamics, mechanosensing and the type 2 immune responses. Single-cell RNA sequencing further unveiled heterogeneous gene expression profiles in the PNEC population and revealed that a small number of PNECs undergo reprogramming during regeneration. Aberrant large clusters of PNECs have been observed in neuroendocrine tumors, including small-cell lung cancer (SCLC). Modern innovation of imaging analyses has enabled the discovery of dynamic migratory behaviors of PNECs during airway development, perhaps relating to SCLC malignancy. This Review summarizes the findings from research on PNECs, along with novel knowledge about their function. In addition, it thoroughly addresses the relevant questions concerning the molecular pathology of pulmonary diseases and related therapeutic approaches.


Subject(s)
Homeostasis , Lung Diseases/pathology , Lung Diseases/physiopathology , Lung/pathology , Lung/physiopathology , Neuroendocrine Cells/pathology , Animals , Humans , Stem Cell Niche , Stem Cells/metabolism
12.
Nat Commun ; 11(1): 4159, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32855415

ABSTRACT

The periodic cartilage and smooth muscle structures in mammalian trachea are derived from tracheal mesoderm, and tracheal malformations result in serious respiratory defects in neonates. Here we show that canonical Wnt signaling in mesoderm is critical to confer trachea mesenchymal identity in human and mouse. At the initiation of tracheal development, endoderm begins to express Nkx2.1, and then mesoderm expresses the Tbx4 gene. Loss of ß-catenin in fetal mouse mesoderm causes loss of Tbx4+ tracheal mesoderm and tracheal cartilage agenesis. The mesenchymal Tbx4 expression relies on endodermal Wnt activation and Wnt ligand secretion but is independent of known Nkx2.1-mediated respiratory development, suggesting that bidirectional Wnt signaling between endoderm and mesoderm promotes trachea development. Activating Wnt, Bmp signaling in mouse embryonic stem cell (ESC)-derived lateral plate mesoderm (LPM) generates tracheal mesoderm containing chondrocytes and smooth muscle cells. For human ESC-derived LPM, SHH activation is required along with WNT to generate proper tracheal mesoderm. Together, these findings may contribute to developing applications for human tracheal tissue repair.


Subject(s)
Endoderm/metabolism , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Trachea/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Endoderm/cytology , Endoderm/embryology , Human Embryonic Stem Cells/metabolism , Humans , Mesoderm/cytology , Mesoderm/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mouse Embryonic Stem Cells/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/metabolism , Trachea/cytology , Trachea/embryology , beta Catenin/metabolism
13.
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
14.
Dev Growth Differ ; 62(1): 67-79, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31613406

ABSTRACT

The respiratory system has ideal tissue structure and cell types for efficient gas exchange to intake oxygen and release carbon dioxide. This complex system develops through orchestrated intercellular signaling among various cell types, such as club, ciliated, basal, neuroendocrine, AT1, AT2, endothelial, and smooth muscle cells. Notch signaling is a highly conserved cell-cell signaling pathway ideally suited for very short-range cellular communication because Notch signals are transmitted by direct contact with an adjacent cell. Enthusiastic efforts by Notch researchers over the last two decades have led to the identification of critical roles of this signaling pathway during development, homeostasis, and regeneration of the respiratory system. The dysregulation of Notch signaling results in a wide range of respiratory diseases such as pulmonary artery hypertension (PAH), chronic obstructive pulmonary disease (COPD), interstitial pulmonary fibrosis (IPF), and lung cancer. Thus, a deep understanding of the biological functions of Notch signaling will help identify novel treatment targets in various respiratory diseases.


Subject(s)
Homeostasis , Lung Diseases , Lung/physiology , Receptors, Notch , Regeneration , Signal Transduction , Trachea/physiology , Tracheal Diseases , Animals , Humans , Lung Diseases/genetics , Lung Diseases/metabolism , Lung Diseases/pathology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Tracheal Diseases/genetics , Tracheal Diseases/metabolism , Tracheal Diseases/pathology
15.
Nat Commun ; 9(1): 2816, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30026494

ABSTRACT

Tube morphogenesis is essential for internal-organ development, yet the mechanisms regulating tube shape remain unknown. Here, we show that different mechanisms regulate the length and diameter of the murine trachea. First, we found that trachea development progresses via sequential elongation and expansion processes. This starts with a synchronized radial polarization of smooth muscle (SM) progenitor cells with inward Golgi-apparatus displacement regulates tube elongation, controlled by mesenchymal Wnt5a-Ror2 signaling. This radial polarization directs SM progenitor cell migration toward the epithelium, and the resulting subepithelial morphogenesis supports tube elongation to the anteroposterior axis. This radial polarization also regulates esophageal elongation. Subsequently, cartilage development helps expand the tube diameter, which drives epithelial-cell reshaping to determine the optimal lumen shape for efficient respiration. These findings suggest a strategy in which straight-organ tubulogenesis is driven by subepithelial cell polarization and ring cartilage development.


Subject(s)
Cartilage/metabolism , Esophagus/metabolism , Morphogenesis/genetics , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Trachea/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Cartilage/cytology , Cartilage/growth & development , Cell Differentiation , Cell Polarity , Embryo, Mammalian , Esophagus/cytology , Esophagus/growth & development , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Muscle, Smooth/cytology , Myocytes, Smooth Muscle/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction , Trachea/cytology , Trachea/growth & development , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism
16.
Nat Commun ; 9(1): 2815, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30022023

ABSTRACT

Tubulogenesis is essential for the formation and function of internal organs. One such organ is the trachea, which allows gas exchange between the external environment and the lungs. However, the cellular and molecular mechanisms underlying tracheal tube development remain poorly understood. Here, we show that the potassium channel KCNJ13 is a critical modulator of tracheal tubulogenesis. We identify Kcnj13 in an ethylnitrosourea forward genetic screen for regulators of mouse respiratory organ development. Kcnj13 mutants exhibit a shorter trachea as well as defective smooth muscle (SM) cell alignment and polarity. KCNJ13 is essential to maintain ion homeostasis in tracheal SM cells, which is required for actin polymerization. This process appears to be mediated, at least in part, through activation of the actin regulator AKT, as pharmacological increase of AKT phosphorylation ameliorates the Kcnj13-mutant trachea phenotypes. These results provide insight into the role of ion homeostasis in cytoskeletal organization during tubulogenesis.


Subject(s)
Morphogenesis/genetics , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Proto-Oncogene Proteins c-akt/genetics , Trachea/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Cell Polarity , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Ion Transport , Mice, Knockout , Muscle, Smooth/cytology , Myocytes, Smooth Muscle/cytology , Phosphorylation , Polymerization , Potassium Channels, Inwardly Rectifying/deficiency , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Trachea/cytology , Trachea/growth & development
17.
Blood ; 132(11): 1167-1179, 2018 09 13.
Article in English | MEDLINE | ID: mdl-29853539

ABSTRACT

Platelets participate in not only thrombosis and hemostasis but also other pathophysiological processes, including tumor metastasis and inflammation. However, the putative role of platelets in the development of solid organs has not yet been described. Here, we report that platelets regulate lung development through the interaction between the platelet-activation receptor, C-type lectin-like receptor-2 (Clec-2; encoded by Clec1b), and its ligand, podoplanin, a membrane protein. Clec-2 deletion in mouse platelets led to lung malformation, which caused respiratory failure and neonatal lethality. In these embryos, α-smooth muscle actin-positive alveolar duct myofibroblasts (adMYFs) were almost absent in the primary alveolar septa, which resulted in loss of alveolar elastic fibers and lung malformation. Our data suggest that the lack of adMYFs is caused by abnormal differentiation of lung mesothelial cells (luMCs), the major progenitor of adMYFs. In the developing lung, podoplanin expression is detected in alveolar epithelial cells (AECs), luMCs, and lymphatic endothelial cells (LECs). LEC-specific podoplanin knockout mice showed neonatal lethality and Clec1b-/--like lung developmental abnormalities. Notably, these Clec1b-/--like lung abnormalities were also observed after thrombocytopenia or transforming growth factor-ß depletion in fetuses. We propose that the interaction between Clec-2 on platelets and podoplanin on LECs stimulates adMYF differentiation of luMCs through transforming growth factor-ß signaling, thus regulating normal lung development.


Subject(s)
Blood Platelets/metabolism , Cell Differentiation/physiology , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Pulmonary Alveoli/embryology , Signal Transduction/physiology , Animals , Blood Platelets/cytology , Endothelial Cells , Epithelial Cells/cytology , Epithelial Cells/metabolism , Lectins, C-Type/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Myofibroblasts/cytology , Myofibroblasts/metabolism , Pulmonary Alveoli/cytology , Respiratory Mucosa/cytology , Respiratory Mucosa/embryology
18.
Dev Growth Differ ; 59(1): 3, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28122132
19.
Proc Natl Acad Sci U S A ; 113(29): 8242-7, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27364009

ABSTRACT

Abnormal enlargement of the alveolar spaces is a hallmark of conditions such as chronic obstructive pulmonary disease and bronchopulmonary dysplasia. Notch signaling is crucial for differentiation and regeneration and repair of the airway epithelium. However, how Notch influences the alveolar compartment and integrates this process with airway development remains little understood. Here we report a prominent role of Notch signaling in the epithelial-mesenchymal interactions that lead to alveolar formation in the developing lung. We found that alveolar type II cells are major sites of Notch2 activation and show by Notch2-specific epithelial deletion (Notch2(cNull)) a unique contribution of this receptor to alveologenesis. Epithelial Notch2 was required for type II cell induction of the PDGF-A ligand and subsequent paracrine activation of PDGF receptor-α signaling in alveolar myofibroblast progenitors. Moreover, Notch2 was crucial in maintaining the integrity of the epithelial and smooth muscle layers of the distal conducting airways. Our data suggest that epithelial Notch signaling regulates multiple aspects of postnatal development in the distal lung and may represent a potential target for intervention in pulmonary diseases.


Subject(s)
Lung/metabolism , Receptor, Notch2/metabolism , Respiratory Mucosa/metabolism , Animals , Cell Line , Cell Proliferation , Epithelial Cells/metabolism , Fucosyltransferases/genetics , Lung/anatomy & histology , Mice, Transgenic , Muscle, Smooth/anatomy & histology , Muscle, Smooth/metabolism , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Respiratory Mucosa/anatomy & histology , Signal Transduction
20.
Cell Rep ; 13(12): 2679-86, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26711336

ABSTRACT

The airway epithelium consists of diverse cell types, including neuroendocrine (NE) cells. These cells are thought to function as chemoreceptors and as a component of the stem cell niche as well as the cells of origin in small-cell lung cancer. NE cells often localize at bifurcation points of airway tubes, forming small clusters called neuroepithelial bodies (NEBs). To investigate NEB development, we established methods for 3D mapping and ex vivo 4D imaging of developing lungs. We found that NEBs localize at stereotypic positions in the bifurcation area irrespective of variations in size. Notch-Hes1 signaling contributes to the differentiation of solitary NE cells, regulating their number but not localization. Live imaging revealed that individual NE cells migrate distally to and cluster at bifurcation points, driving NEB formation. We propose that NEB development is a multistep process involving differentiation of individual NE cells and their directional migration to organize NEBs.


Subject(s)
Cell Movement/physiology , Lung/cytology , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Neuroepithelial Bodies/cytology , Animals , Immunohistochemistry , Lung/metabolism , Mice , Neuroepithelial Bodies/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...