Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Respir Res ; 18(1): 87, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28482837

ABSTRACT

BACKGROUND: The pulmonary neuroepithelial body (NEB) microenvironment (ME) consists of innervated cell clusters that occur sparsely distributed in the airway epithelium, an organization that has so far hampered reliable selective gene expression analysis. Although the NEB ME has been suggested to be important for airway epithelial repair after ablation, little is known about their potential stem cell characteristics in healthy postnatal lungs. Here we report on a large-scale selective gene expression analysis of the NEB ME. METHODS: A GAD67-GFP mouse model was used that harbors GFP-fluorescent NEBs, allowing quick selection and pooling by laser microdissection (LMD) without further treatment. A panel of stem cell-related PCR arrays was used to selectively compare mRNA expression in the NEB ME to control airway epithelium (CAE). For genes that showed a higher expression in the NEB ME, a ranking was made based on the relative expression level. Single qPCR and immunohistochemistry were used to validate and quantify the PCR array data. RESULTS: Careful optimization of all protocols appeared to be essential to finally obtain high-quality RNA from pooled LMD samples of NEB ME. About 30% of the more than 600 analyzed genes showed an at least two-fold higher expression compared to CAE. The gene that showed the highest relative expression in the NEB ME, Delta-like ligand 3 (Dll3), was investigated in more detail. Selective Dll3 gene expression in the NEB ME could be quantified via single qPCR experiments, and Dll3 protein expression could be localized specifically to NEB cell surface membranes. CONCLUSIONS: This study emphasized the importance of good protocols and RNA quality controls because of the, often neglected, fast RNA degradation in postnatal lung samples. It was shown that sufficient amounts of high-quality RNA for reliable complex gene expression analysis can be obtained from pooled LMD-collected NEB ME samples of postnatal lungs. Dll3 expression, which has also been reported to be important in high-grade pulmonary tumor-initiating cells, was used as a proof-of-concept to confirm that the described methodology represents a promising tool for further unraveling the molecular basis of NEB ME physiology in general, and its postnatal stem cell capacities in particular.


Subject(s)
Gene Expression Profiling/methods , Intracellular Signaling Peptides and Proteins/metabolism , Lung/metabolism , Membrane Proteins/metabolism , Neuroepithelial Bodies/cytology , Neuroepithelial Bodies/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Animals, Newborn , Cells, Cultured , Lung/cytology , Mice , Mice, Transgenic
2.
Cell Rep ; 19(2): 246-254, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28402849

ABSTRACT

There is evidence that certain club cells (CCs) in the murine airways associated with neuroepithelial bodies (NEBs) and terminal bronchioles are resistant to the xenobiotic naphthalene (Nap) and repopulate the airways after Nap injury. The identity and significance of these progenitors (variant CCs, v-CCs) have remained elusive. A recent screen for CC markers identified rare Uroplakin3a (Upk3a)-expressing cells (U-CCs) with a v-CC-like distribution. Here, we employ lineage analysis in the uninjured and chemically injured lungs to investigate the role of U-CCs as epithelial progenitors. U-CCs proliferate and generate CCs and ciliated cells in uninjured airways long-term and, like v-CCs, after Nap. U-CCs have a higher propensity to generate ciliated cells than non-U-CCs. Although U-CCs do not contribute to alveolar maintenance long-term, they generate alveolar type I and type II cells after Bleomycin (Bleo)-induced alveolar injury. Finally, we report that Upk3a+ cells exist in the NEB microenvironment of the human lung and are aberrantly expanded in conditions associated with neuroendocrine hyperplasias.


Subject(s)
Bronchioles/metabolism , Cellular Microenvironment/genetics , Stem Cells/metabolism , Uroplakin III/biosynthesis , Animals , Bleomycin/toxicity , Bronchioles/drug effects , Bronchioles/injuries , Cell Lineage/drug effects , Cellular Microenvironment/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Mice , Naphthalenes/toxicity , Neuroepithelial Bodies/metabolism , Neuroepithelial Bodies/pathology , Pulmonary Alveoli/injuries , Stem Cells/drug effects , Uroplakin III/genetics , Wound Healing
3.
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
4.
Cell ; 163(2): 394-405, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26435104

ABSTRACT

Epithelial cells are normally stably anchored, maintaining their relative positions and association with the basement membrane. Developmental rearrangements occur through cell intercalation, and cells can delaminate during epithelial-mesenchymal transitions and metastasis. We mapped the formation of lung neuroepithelial bodies (NEBs), innervated clusters of neuroendocrine/neurosensory cells within the bronchial epithelium, revealing a targeted mode of cell migration that we named "slithering," in which cells transiently lose epithelial character but remain associated with the membrane while traversing neighboring epithelial cells to reach cluster sites. Immunostaining, lineage tracing, clonal analysis, and live imaging showed that NEB progenitors, initially distributed randomly, downregulate adhesion and polarity proteins, crawling over and between neighboring cells to converge at diametrically opposed positions at bronchial branchpoints, where they reestablish epithelial structure and express neuroendocrine genes. There is little accompanying progenitor proliferation or apoptosis. Activation of the slithering program may explain why lung cancers arising from neuroendocrine cells are highly metastatic.


Subject(s)
Cell Movement , Lung/cytology , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Neuroepithelial Bodies/cytology , Animals , Cell Lineage , Down-Regulation , Epithelial-Mesenchymal Transition , Lung/embryology , Lung/metabolism , Mice , Neuroepithelial Bodies/metabolism , Stem Cells/cytology , Stem Cells/metabolism
5.
Adv Exp Med Biol ; 860: 309-13, 2015.
Article in English | MEDLINE | ID: mdl-26303495

ABSTRACT

Sustained exposure to low oxygen concentration leads to profound changes in gene expression to restore oxygen homeostasis. Hypoxia-inducible factors (HIFs) comprise a group of transcription factors which accumulate under hypoxia and contribute to the complex changes in gene expression. Under normoxic conditions HIFs are degraded by prolyl-hydroxylases (PHD), however during hypoxia this degradation is inhibited causing HIF accumulation and subsequent changes in gene expression. Pulmonary neuroepithelial bodies (NEB) are innervated serotonin (5-HT)-producing cells distributed throughout the airway epithelium. These putative O(2) sensors are hypothesized to contribute to the ventilatory response to hypoxia. NEB dysfunction has been implicated in several paediatric lung diseases including neuroendocrine cell hyperplasia of infancy and sudden infant death syndrome, both characterized by a marked NEB hyperplasia with unknown functional significance. We have previously reported striking NEB hyperplasia in PHD1(-/-) mice making these mice a potential model to study the role of NEBs in paediatric lung diseases. Here we report in vitro studies on 5-HT release from NEB using this model.


Subject(s)
Lung Diseases/etiology , Neuroepithelial Bodies/metabolism , Procollagen-Proline Dioxygenase/physiology , Serotonin/metabolism , Animals , Cells, Cultured , Mice , Mice, Knockout
6.
PLoS One ; 9(11): e112623, 2014.
Article in English | MEDLINE | ID: mdl-25409164

ABSTRACT

The pulmonary neuroepithelial bodies (NEBs) constitute polymodal airway chemosensors for monitoring and signaling ambient gas concentrations (pO2, pCO2/H+) via complex innervation to the brain stem controlling breathing. NEBs produce the bioactive amine, serotonin (5-HT), and a variety of peptides with multiple effects on lung physiology and other organ systems. NEBs in mammals appear prominent and numerous during fetal and neonatal periods, and decline in the post-natal period suggesting an important role during perinatal adaptation. The naked mole-rat (NMR), Heterocephalus glaber, has adapted to the extreme environmental conditions of living in subterranean burrows in large colonies (up to 300 colony mates). The crowded, unventilated burrows are environments of severe hypoxia and hypercapnia. However, NMRs adjust readily to above ground conditions. The chemosensory NEBs of this species were characterized and compared to those of the conventional Wistar rat (WR) to identify similarities and differences that could explain the NMR's adaptability to environments. A multilabel immunohistochemical analysis combined with confocal microscopy revealed that the expression patterns of amine, peptide, neuroendocrine, innervation markers and chemosensor component proteins in NEBs of NMR were similar to that of WR. However, we found the following differences: 1) NEBs in both neonatal and adult NMR lungs were significantly larger and more numerous as compared to WR; 2) NEBs in NMR had a more variable compact cell organization and exhibited significant differences in the expression of adhesion proteins; 3) NMR NEBs showed a significantly greater ratio of 5-HT positive cells with an abundance of 5-HT; 4) NEBs in NMR expressed the proliferating cell nuclear antigen (PCNA) and the neurogenic gene (MASH1) indicating active proliferation and a state of persistent differentiation. Taken together our findings suggest that NEBs in lungs of NMR are in a hyperactive, functional and developmental state, reminiscent of a persistent fetal state that extends postnatally.


Subject(s)
Adaptation, Physiological , Lung/cytology , Lung/physiology , Neuroepithelial Bodies/cytology , Phenotype , Animals , Biomarkers/metabolism , Carbon Dioxide/metabolism , Cell Adhesion , Cell Proliferation , Gene Expression Regulation , Immunohistochemistry , Lung/innervation , Mole Rats , Neuroepithelial Bodies/metabolism , Neurogenesis , Neurosecretory Systems/metabolism , Neurosecretory Systems/physiology , Oxygen/metabolism
7.
Morfologiia ; 145(1): 60-2, 2014.
Article in Russian | MEDLINE | ID: mdl-25051803

ABSTRACT

This paper describes the study of the neuroepithelial bodies (NEB) in the lungs of adult healthy Wistar rats (n = 12). Using the immunocytochemical reaction demonstrating synaptophysin, NEB and immunopositive nerve terminals approaching them, were visualized. It was found that NEB were the structures constantly presented in the rat lung. In contrast to the diffuse neuroendocrine elements, NEB are characterized by grouped distribution of cells. It was found that some part of NEB had no efferent innervation.


Subject(s)
Lung/cytology , Neuroepithelial Bodies/cytology , Animals , Lung/innervation , Lung/metabolism , Neuroepithelial Bodies/metabolism , Peripheral Nerves/cytology , Rats , Rats, Wistar , Synaptophysin/genetics , Synaptophysin/metabolism
8.
Respir Physiol Neurobiol ; 193: 52-4, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24418354

ABSTRACT

Neuroepithelial bodies (NEBs) serve a niche for lung stem cells and proliferate in a variety of pulmonary diseases. We hypothesize that NEBs play an important role in lung injury repair processes, such as during pulmonary fibrosis. To test this hypothesis, we examined NEBs in a bleomycin-induced lung fibrosis mouse model. We divided FVB/NJ mice into bleomycin-treated (BL) and normal saline-treated (NS) groups. Two weeks after intravenous treatment, we immune-stained NEBs with anti-calcitonin gene-related peptide (CGRP) in whole mount preparations and found that the number of NEBs per unit area of airway almost tripled in the BL group (1.11±0.28 number/mm(2); n=5) compared with the NS group (0.32±0.14 number/mm(2); n=4, p=0.001). The size of NEBs increased significantly in the BL group. Our findings support that NEBs play an important role in the pathogenesis of pulmonary fibrosis.


Subject(s)
Bronchioles/pathology , Lung/pathology , Neuroepithelial Bodies/pathology , Pulmonary Fibrosis/pathology , Trachea/pathology , Animals , Bleomycin , Bronchioles/metabolism , Calcitonin Gene-Related Peptide/metabolism , Fluorescent Antibody Technique , Lung/metabolism , Male , Mice , Neuroepithelial Bodies/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Trachea/metabolism
9.
Histol Histopathol ; 29(5): 659-68, 2014 May.
Article in English | MEDLINE | ID: mdl-24301684

ABSTRACT

Adult male mice were continuously treated with bromodeoxyuridine (BrdU) for 1, 2, or 4 weeks by an osmotic pump. To detect BrdU-label-retaining cells (LRCs), putative progenitor/stem cells, other animals were continuously treated with BrdU for 2 weeks, and were then kept without any treatments for 2, 6, or 18 months. The lungs were fixed with 4% paraformaldehyde, and were paraffin-embedded. We observed terminal bronchioles with BrdU immunostaining alone or with BrdU immunostaining accompanying immunostaining for Clara cell secretory protein (CCSP), forkhead box protein J1 (FoxJ1), or calcitonin gene-related peptide (CGRP). The average incidences of BrdU-incorporated cells in the terminal bronchioles after 1, 2, and 4 weeks of continuous BrdU infusion were 6.2%, 11.9%, and 23.1%, respectively. Most BrdU-incorporated cells in these periods were CCSP-immunoreactive (91.7%, 91.3%, and 88.2%, respectively), which means progenitor function of Clara cells. FoxJ1-immunoreactive BrdU-incorporated cells were fewer (5.4%, 3.0%, 2.7%, respectively). The average incidences of BrdU-LRCs in the terminal bronchioles after 2, 6, and 18 months were 7.2%, 4.3, and 2.7%, respectively. Most BrdU-LRCs were CCSP-immunoreactive (91.0%, 92.7%, and 89.6%, respectively), and FoxJ1-immunoreactive BrdU-LRCs were fewer (6.0%, 5.7%, and 2.1%, respectively). CGRP-positive BrdU-incorporated cells were occasional. CGRP-positive BrdU-LRCs were detected in 17.6% of neuroepithelial bodies (NEBs) at 2 months, but disappeared at 6 months. BrdU-positive stem cell candidates, which locate at the brochiolo-alveolar duct junction or cover NEB, were few throughout this study. In conclusion, in the lungs treated only with BrdU, CCSP-immunoreactive cells are important to maintain homeostasis in the terminal bronchiolar epithelium.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Bromodeoxyuridine/pharmacokinetics , Bronchioles/cytology , Bronchioles/metabolism , Animals , Bromodeoxyuridine/administration & dosage , Calcitonin Gene-Related Peptide/metabolism , Forkhead Transcription Factors/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred ICR , Neuroepithelial Bodies/cytology , Neuroepithelial Bodies/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Time Factors , Uteroglobin/metabolism
10.
J Cell Sci ; 126(Pt 19): 4490-501, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23886943

ABSTRACT

The Ca(2+)-sensing receptor (CaSR) is the master regulator of whole-body extracellular free ionized [Ca(2+)]o. In addition to sensing [Ca(2+)]o, CaSR integrates inputs from a variety of different physiological stimuli. The CaSR is also expressed in many regions outside the [Ca(2+)]o homeostatic system, including the fetal lung where it plays a crucial role in lung development. Here, we show that neuroepithelial bodies (NEBs) of the postnatal mouse lung express a functional CaSR. NEBs are densely innervated groups of neuroendocrine epithelial cells in the lung representing complex sensory receptors in the airways and exhibiting stem cell characteristics. qRT-PCR performed on laser microdissected samples from GAD67-GFP mouse lung cryosections revealed exclusive expression of the CaSR in the NEB microenvironment. CaSR immunoreactivity was present at NEB cells from postnatal day 14 onwards. Confocal imaging of lung slices revealed that NEB cells responded to an increase of [Ca(2+)]o with a rise in intracellular Ca(2+) ([Ca(2+)]i); an effect mimicked by several membrane-impermeant CaSR agonists (e.g. the calcimimetic R-568) and that was blocked by the calcilytic Calhex-231. Block of TRPC channels attenuated the CaSR-dependent increases in [Ca(2+)]i, suggesting that Ca(2+) influx through TRPC channels contributes to the total [Ca(2+)]i signal evoked by the CaSR in NEBs. CaSR also regulated baseline [Ca(2+)]i in NEBs and, through paracrine signaling from Clara-like cells, coordinated intercellular communication in the NEB microenvironment. These data suggest that the NEB CaSR integrates multiple signals converging on this complex chemosensory unit, and is a key regulator of this intrapulmonary airway stem cell niche.


Subject(s)
Lung/metabolism , Neuroendocrine Cells/cytology , Neuroepithelial Bodies/metabolism , Receptors, Calcium-Sensing/biosynthesis , Animals , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroendocrine Cells/metabolism , Receptors, Calcium-Sensing/metabolism
11.
Histochem Cell Biol ; 140(5): 549-66, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23568330

ABSTRACT

Gamma-aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the central nervous system (CNS) of vertebrates, but has also been reported in multiple cell types outside the CNS. A GABAergic system has been proposed in neuroepithelial bodies (NEBs) in monkey lungs. Pulmonary NEBs are known as complex intraepithelial sensory airway receptors and are part of the NEB microenvironment. Aim of the present study was to unravel a GABAergic signaling system in the NEB microenvironment in mouse lungs, enabling the use of genetically modified animals for future functional studies. Immunostaining of mouse lungs revealed that glutamic acid decarboxylase 65/67 (GAD65/67), a rate-limiting enzyme in the biosynthesis of GABA, and the vesicular GABA transporter (VGAT) were exclusively expressed in NEB cells. In GAD67-green fluorescent protein (GFP) knock-in mice, all pulmonary NEBs appeared to express GFP. For confocal live cell imaging, ex vivo vibratome lung slices of GAD67-GFP mice can be directly loaded with fluorescent functional probes, e.g. a red-fluorescent calcium dye, without the necessity of time-consuming prior live visualization of NEBs. RT-PCR of the NEB microenvironment obtained by laser microdissection revealed the presence of both GABAA and GABAB (R1 and R2) receptors, which was confirmed by immunostaining. In conclusion, the present study not only revealed the presence of a GABAergic signaling pathway, but also the very selective expression of GFP in pulmonary NEBs in a GAD67-GFP mouse model. Different proof of concept experiments have clearly shown that adoption of the GAD67-GFP mouse model will certainly boost future functional imaging and gene expression analysis of the mouse NEB microenvironment.


Subject(s)
Cellular Microenvironment , GABAergic Neurons/metabolism , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/metabolism , Lung/metabolism , Neuroepithelial Bodies/metabolism , Signal Transduction , Animals , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , Immunohistochemistry , Lung/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Imaging , Neuroepithelial Bodies/cytology , Reverse Transcriptase Polymerase Chain Reaction , gamma-Aminobutyric Acid/metabolism
12.
Semin Cell Dev Biol ; 24(1): 40-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23022441

ABSTRACT

Pulmonary neuroepithelial bodies are polymodal sensors widely distributed within the airway mucosa of mammals and other species. Neuroepithelial body cells store and most likely release serotonin and peptides as transmitters. Neuroepithelial bodies have a complex innervation that includes vagal sensory afferent fibers and dorsal root ganglion fibers. Neuroepithelial body cells respond to a number of intraluminal airway stimuli, including hypoxia, hypercarbia, and mechanical stretch. This article reviews recent findings in the cellular and molecular biology of neuroepithelial body cells and their potential role as airway sensors involved in the control of respiration, particularly during the perinatal period. Alternate hypotheses and areas of controversy regarding potential function as mechanosensory receptors involved in pulmonary reflexes are discussed.


Subject(s)
Lung/metabolism , Neuroepithelial Bodies/metabolism , Animals , Humans , Lung/innervation , Oxygen/metabolism , Oxygen Consumption
13.
Development ; 139(23): 4365-73, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23132245

ABSTRACT

In the developing lung, it is thought that the terminal buds of elongating airways contain a population of multipotent epithelial progenitors. As the bronchial tree extends, descendants of these cells give rise to lineage-restricted progenitors in the conducting airways via Notch signaling, which is involved in the establishment of epithelial Clara, ciliated and pulmonary neuroendocrine (NE) cell populations. However, the precise molecular details of this selection process are still emerging. Our stepwise removal of the three Notch receptors from the developing lung epithelium reveals that, whereas Notch2 mediates the Clara/ciliated cell fate decision with negligible contributions from Notch1 and Notch3, all three Notch receptors contribute in an additive manner to regulate the abundance of NE cells and the size of the presumptive pulmonary neuroepithelial body (pNEB) as a result of mutual interactions between NE cells and the Notch-dependent, SSEA-1(+), CC10(-) cell population surrounding the pNEB (SPNC cells). Ectopic expression of the Notch1 or Notch2 intracellular domain was sufficient to induce SSEA-1(+) cells and to suppress pNEB formation without expending Clara cells. We provide evidence that the additive functions of Notch receptors, together with other signaling pathways, maintains the expression of Hes1, a key regulator of NE cell fate, and that maintenance of Hes1 expression in epithelial cells is key to the regulation of pNEB size. These results suggest that two different assemblies of Notch receptors coordinate the numbers and distribution of the major epithelial cell types in the conducting airway during lung organogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Homeodomain Proteins/biosynthesis , Lung/embryology , Neuroendocrine Cells/physiology , Neuroepithelial Bodies/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Respiratory Mucosa/cytology , Animals , Cell Differentiation , Epithelial Cells/physiology , Gene Expression Regulation, Developmental , Lewis X Antigen/biosynthesis , Lewis X Antigen/genetics , Lewis X Antigen/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/biosynthesis , Receptor, Notch1/biosynthesis , Receptor, Notch2/biosynthesis , Respiratory Mucosa/embryology , Respiratory Mucosa/metabolism , Respiratory System/metabolism , Signal Transduction , Transcription Factor HES-1 , Uteroglobin/biosynthesis
14.
Proc Natl Acad Sci U S A ; 109(31): 12592-7, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22797898

ABSTRACT

Clara cells of mammalian airways have multiple functions and are morphologically heterogeneous. Although Notch signaling is essential for the development of these cells, it is unclear how Notch influences Clara cell specification and if diversity is established among Clara cell precursors. Here we identify expression of the secretoglobin Scgb3a2 and Notch activation as early events in a program of secretory cell fate determination in developing murine airways. We show that Scgb3a2 expression in vivo is Notch-dependent at early stages and ectopically induced by constitutive Notch1 activation, and also that in vitro Notch signaling together with the pan-airway transcription factor Ttf1 (Nkx2.1) synergistically regulate secretoglobin gene transcription. Furthermore, we identified a subpopulation of secretory precursors juxtaposed to presumptive neuroepithelial bodies (NEBs), distinguished by their strong Scgb3a2 and uroplakin 3a (Upk3a) signals and reduced Ccsp (Scgb1a1) expression. Genetic ablation of Ascl1 prevented NEB formation and selectively interfered with the formation of this subpopulation of cells. Lineage labeling of Upk3a-expressing cells during development showed that these cells remain largely uncommitted during embryonic development and contribute to Clara and ciliated cells in the adult lung. Together, our findings suggest a role for Notch in the induction of a Clara cell-specific program of gene expression, and reveals that the NEB microenvironment in the developing airways is a niche for a distinct subset of Clara-like precursors.


Subject(s)
Neuroepithelial Bodies/metabolism , Respiratory System/embryology , Stem Cell Niche/physiology , Stem Cells/metabolism , Animals , Female , Gene Expression Regulation, Developmental/physiology , Mice , Mice, Knockout , Neuroepithelial Bodies/cytology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Respiratory System/cytology , Secretoglobins/biosynthesis , Secretoglobins/genetics , Stem Cells/cytology , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Acta Histochem ; 114(4): 370-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21831413

ABSTRACT

Anatomical and functional studies on the autonomic innervation as well as the location of airway receptors in the air-bladder of lepisosteids are very fragmentary. These water-breathing fishes share in common with the bichirs the presence of a glottis (not a ductus pneumaticus) opening into the esophagus. In contrast to a high concentration of neuroepithelial cells (NECs) contained in the furrowed epithelium in the lung of Polypterus, these cells are scattered as solitary cells in the glottal epithelium, and grouped to form neuroepithelial bodies (NEBs) in the mucociliated epithelium investing the main trabeculae in the air-bladder of Lepisosteus osseus and L. oculatus. The present immunohistochemical studies also demonstrated the presence of nerve fibers in the trabecular striated musculature and a possible relation to NEBs in these species, and identified immunoreactive elements of this innervation. Tyrosine hydroxylase (TH), choline acetyltransferase (ChAT), 5-HT and neuropeptide immunoreactivities were detected in the intramural nerve fibers. 5-HT and VIP immunopositive nerve fibers are apparently associated with NEBs. TH, VIP and SP immunoreactivities are also present in nerve fibers coursing in the radially arranged striated muscle surrounding the glottis and its submucosa. 5-HT positive neurons are also found in submucosal and the muscle layers of the glottis. The physiological function of the adrenergic and inhibitory innervation of the striated muscle as well as the neurochemical coding and morphology of the innervation of the NEBs are not known. Future studies are needed to provide evidence for these receptors with the capacity of chemoreceptors and/or mechanoreceptors.


Subject(s)
Fishes/anatomy & histology , Muscle, Striated , Neuroepithelial Bodies/ultrastructure , Neuroepithelial Cells/ultrastructure , Respiratory System , Animals , Choline O-Acetyltransferase/biosynthesis , Immunohistochemistry , Mucous Membrane/metabolism , Mucous Membrane/ultrastructure , Muscle, Striated/anatomy & histology , Muscle, Striated/innervation , Nerve Fibers/metabolism , Nerve Fibers/ultrastructure , Neuroepithelial Bodies/metabolism , Neuroepithelial Cells/metabolism , Neuropeptides/biosynthesis , Respiratory System/anatomy & histology , Respiratory System/innervation , Serotonin/biosynthesis , Tyrosine 3-Monooxygenase/biosynthesis
16.
Histochem Cell Biol ; 136(4): 371-85, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21822716

ABSTRACT

Afferent activities arising from sensory nerve terminals located in lungs and airways are carried almost exclusively by fibres travelling through the vagus nerve. Based on electrophysiological investigations, intrapulmonary airway-related vagal afferent receptors have been classified into three main subtypes, two of which are myelinated and mechanosensitive, i.e., rapidly and slowly adapting receptors. To allow for a full functional identification of the distinct populations of airway receptors, morphological and neurochemical characteristics still need to be determined. Nerve terminals visualised using markers for myelinated vagal afferents seem to be almost uniquely associated with two morphologically well-formed airway receptor end organs, smooth muscle-associated airway receptors (SMARs) and neuroepithelial bodies (NEBs), localised in airway smooth muscle and epithelium, respectively. Due to the lack of a selective marker for SMARs in mice, no further neurochemical coding is available today. NEBs are extensively innervated diffusely spread groups of neuroendocrine cells in the airway epithelium, and are known to receive at least two separate populations of myelinated vagal afferent nerve terminals. So far, however, no evidence has been reported for the expression of channels that may underlie direct sensing and transduction of mechanical stimuli by the receptor terminals in NEBs and SMARs. This study focused on the expression of mechanogated two-pore domain K(+) (K(2P)) channels, TREK-1 and TRAAK, in mouse airways and more particular in the NEB micro-environment and in SMARs by multiple immunostaining. TREK-1 could be detected on smooth muscle cells surrounding intrapulmonary airways and blood vessels, while TRAAK was expressed on myelinated vagal afferents terminating both in SMARs and in the NEB micro-environment. Co-stainings with known markers for subpopulations of myelinated vagal afferents and general neuronal markers revealed that all identified SMARs exhibit TRAAK immunoreactivity, and that at least three subpopulations exist in mouse airways. Also, the intraepithelial terminals of both subpopulations of NEB-associated myelinated vagal sensory nerve fibres were shown to express TRAAK. In conclusion, the present study finally characterised an intrinsically mechanosensitive ion channel, the K(2P) channel TRAAK, on the terminals of identified myelinated vagal nodose airway afferents, organised as SMARs and as components of the innervation of NEBs. These data support the hypothesis that both SMARs and NEBs harbour the morphological counterparts of electrophysiologically identified myelinated vagal airway mechanoreceptors. TRAAK appears to be strongly involved in regulating airway mechanosensing since it was found to be expressed on the terminals of all subpopulations of potential vagal mechanosensors.


Subject(s)
Lung/metabolism , Muscle, Smooth/metabolism , Neuroepithelial Bodies/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Potassium Channels/metabolism , Sensory Receptor Cells/metabolism , Animals , Female , Immunohistochemistry , Lung/cytology , Male , Mice , Mice, Inbred C57BL , Staining and Labeling
17.
Histol Histopathol ; 25(8): 975-84, 2010 08.
Article in English | MEDLINE | ID: mdl-20552548

ABSTRACT

Important physiological functions of neurotrophins (NTs) in airways and lungs are the early development, differentiation and maintenance of peripheral sensory neurons. The main pulmonary sensory innervation is of vagal origin, with several nerve fibre populations that selectively contact complex morphologically well-characterized receptor end-organs, called neuroepithelial bodies (NEBs). NEBs in mouse lungs are innervated by at least two separate myelinated vagal sensory nerve fibre populations, of which the neurochemical coding is suggestive of a mechanosensory function. Since neurotrophin-4 (NT-4) has been especially described to be important for the maintenance of mechanosensory nerve terminals, the present study aimed at investigating the NT-4 dependency of the two myelinated vagal sensory nerve fibre populations innervating mouse pulmonary NEBs. Multiple immunostaining in 21-day-old and adult mouse lungs revealed the expression of the NT-4 receptor TrkB on the two different myelinated vagal sensory nerve fibre populations, i.e., the vesicular glutamate transporter/calbindin-positive and the P2X2/3-positive fibres, which selectively contact pulmonary NEBs. Examination of the effect of the lack of NT-4 on these NEB-related nerve fibre populations, by comparing adult NT-4-/- and wild-type mice, revealed that in NT-4-/- mice the percentage of NEBs contacted by P2X2/3+ is reduced by 75%, while the VGLUT+/CB+ population seemed to be unaffected. This study demonstrated that although mouse pulmonary NEBs are contacted by two distinct TrkB expressing populations of vagal myelinated afferents, only one is distinctly reduced in NT-4 deficient mice, suggesting the involvement of NTs. In view of the growing evidence for the involvement of NTs in neuronal plasticity associated with airway diseases, pulmonary NEBs innervated by NT-sensitive vagal afferents may play a significant role.


Subject(s)
Lung/innervation , Lung/physiology , Nerve Endings/physiology , Neuroepithelial Bodies/metabolism , Animals , Calbindins , Lung/metabolism , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Nerve Endings/metabolism , Nerve Fibers/metabolism , Nerve Growth Factors/metabolism , Neurons, Afferent/metabolism , Neurons, Afferent/physiology , Respiratory System/metabolism , S100 Calcium Binding Protein G , Sensory Receptor Cells
18.
Adv Exp Med Biol ; 648: 427-38, 2009.
Article in English | MEDLINE | ID: mdl-19536508

ABSTRACT

In pulmonary neuroepithelial bodies (NEB), presumed airway chemoreceptors, classical NADPH oxidase (gp91 phox, NOX2) is co-expressed with O(2) sensitive K(+) channels (K(+)O(2)) and functions as an O(2) sensor. Here we examined related NADPH oxidase homologues "novel oxidases "(NOX 1, 3&4) and their possible involvement in O(2) sensing. For immunolocalization we used specific antibodies against various NADPH components and K(+) (O(2)) subunits to label NEB in rat /rabbit lung and NEB related H146 tumor cell line. For gene expression profiling of NEB cells microdissected from human lung, and H146 cells, we used custom MultiGene-12TM RT-PCR array that included NADPH oxidase components and homologues /accessory proteins (NOX1-4, phox-p22, p40, p47, p67, Rac1, NOXO1 and NOXA1) and K(+)O(2) channels (Kv -1.2, 1.5, 2.1, 3.1, 3.3, 3.4, 4.2, 4.3;TASK1-3). In rat lung, NOX2, NOX4, p22phox, Kv3.3 (and Kv3.4 in rabbit) and TASK1 localized to the apical plasma membrane of NEB cells, and membrane or sub-membrane regions in H146 cells. NEB and H146 cells expressed all NOX proteins except NOX3, as well as all K(+)O(2) channels, except Kv1.5 and Kv4.3. Co-immunoprecipitation using Western blot multicolor Quantum dot labeling showed NOX2 molecular complexes with Kv but not with TASK, while NOX4 associated with TASK1 but not with Kv channel proteins. Hypoxia -induced serotonin release was inhibited in H 146 cells by siRNA to NOX2, while siRNA to NOX4 had only a partial effect, implicating NOX 2 as the predominant NEB cell O(2) sensor. Present findings support NEB cell specific plasma membrane model of O(2) sensing, and suggest unique NOX/K(+)O(2) channel combinations for diverse physiological NEB functions.


Subject(s)
Lung/cytology , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Neuroepithelial Bodies/metabolism , Oxygen/metabolism , Animals , Base Sequence , Cell Line , Epitopes/metabolism , Gene Expression Profiling , Humans , Hypoxia/metabolism , Immunohistochemistry , Infant , Male , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , NADPH Oxidase 2 , NADPH Oxidases/chemistry , NADPH Oxidases/genetics , Potassium Channels/chemistry , Potassium Channels/metabolism , Protein Transport , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Serotonin/metabolism
19.
Adv Exp Med Biol ; 648: 439-45, 2009.
Article in English | MEDLINE | ID: mdl-19536509

ABSTRACT

Pulmonary neuroepithelial bodies (NEB) act as airway oxygen sensors and produce serotonin, a variety of neuropeptides and are involved in autonomic nervous system control of breathing, especially during the neonatal period. We now report that NEB cells also express a GABAergic signaling loop that is increased by prenatal nicotine exposure. In this study, cultured monkey NEB cells show hypoxia-evoked action potentials and hypoxia-sensitive K(+) current. As shown by both immunofluorescence and RT-PCR, monkey NEB cells synthesize and contain serotonin. The monkey NEB cells express the beta2 and beta3 GABA_A receptor subunits, GAD and also express alpha7, alpha4 and beta4 nicotinic receptor (nAChR) subunits. The alpha7 nAChR is co-expressed with GAD in NEB. The numbers of NEB and beta3 GABA_A receptor subunits expressed in NEB cells in lungs from control newborn monkeys were compared to lungs from animals that received nicotine during gestation. Prenatal nicotine exposure increased the numbers of NEB by 46% in lung and the numbers of NEB cells expressing GAD and GABA_A beta3 receptors increased by 67% and 66%, respectively. This study suggests that prenatal nicotine exposure can modulate NEB function by increasing the numbers of NEB cells and by increasing both GAD expression and beta3 GABA_A receptor subunit expression. The interaction of the intrinsic GABAergic system in the lung with nicotinic receptors in PNEC/NEB may provide a mechanism to explain the link between smoking during pregnancy and SIDS.


Subject(s)
Lung/cytology , Maternal Exposure , Neuroepithelial Bodies/drug effects , Neuroepithelial Bodies/metabolism , Nicotine/pharmacology , Receptors, GABA-A/metabolism , Action Potentials/drug effects , Animals , Cells, Cultured , Electric Conductivity , Female , Gene Expression Regulation/drug effects , Hypoxia/metabolism , Lung/drug effects , Macaca mulatta , Neuroepithelial Bodies/cytology , Potassium/metabolism , Signal Transduction/drug effects
20.
Histochem Cell Biol ; 131(1): 55-74, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18762965

ABSTRACT

As best characterized for rats, it is clear that pulmonary neuroepithelial bodies (NEBs) are contacted by a plethora of nerve fiber populations, suggesting that they represent an extensive group of multifunctional intraepithelial airway receptors. Because of the importance of genetically modified mice for functional studies, and the current lack of data, the main aim of the present study was to achieve a detailed analysis of the origin and neurochemical properties of nerve terminals associated with NEBs in mouse lungs. Antibodies against known selective markers for sensory and motor nerve terminals in rat lungs were used on lungs from control and vagotomized mice of two different strains, i.e., Swiss and C57-Bl6. NEB cells were visualized by antibodies against either the general neuroendocrine marker protein gene-product 9.5 (PGP9.5) or calcitonin gene-related peptide (CGRP). Thorough immunohistochemical examination of NEB cells showed that some of these NEB cells also exhibit calbindin D-28 k (CB) and vesicular acetylcholine transporter (VAChT) immunoreactivity (IR). Mouse pulmonary NEBs were found to receive intraepithelial nerve terminals of at least two different populations of myelinated vagal afferents: (1) Immunoreactive (ir) for vesicular glutamate transporters (VGLUTs) and CB; (2) expressing P2X(2) and P2X(3) ATP receptors. CGRP IR was seen in varicose vagal nerve fibers and in delicate non-vagal fibers, both in close proximity to NEBs. VAChT immunostaining showed very weak IR in the NEB-related intraepithelial vagal sensory nerve terminals. nNOS- or VIP-ir nerve terminals could be observed at the base of pulmonary NEBs. While a single NEB can be contacted by multiple nerve fiber populations, it was clear that none of the so far characterized nerve fiber populations contacts all pulmonary NEBs. The present study revealed that mouse lungs harbor several populations of nerve terminals that may selectively contact NEBs. Although at present the physiological significance of the innervation pattern of NEBs remains enigmatic, it is likely that NEBs are receptor-effector end-organs that may host complex and/or multiple functional properties in normal airways. The neurochemical information on the innervation of NEBs in mouse lungs gathered in the present study will be essential for the interpretation of upcoming functional data and for the study of transgenic mice.


Subject(s)
Lung/innervation , Neuroepithelial Bodies/chemistry , Animals , Immunohistochemistry , Lung/cytology , Lung/metabolism , Mice , Mice, Inbred C57BL , Neuroepithelial Bodies/cytology , Neuroepithelial Bodies/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...