Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters










Publication year range
1.
Anat Rec (Hoboken) ; 2023 Feb 19.
Article in English | MEDLINE | ID: mdl-36808710

ABSTRACT

Exciting new imaging and molecular tools, combined with state-of-the-art genetically modified mouse models, have recently boosted interest in pulmonary (vagal) sensory pathway investigations. In addition to the identification of diverse sensory neuronal subtypes, visualization of intrapulmonary projection patterns attracted renewed attention on morphologically identified sensory receptor end-organs, such as the pulmonary neuroepithelial bodies (NEBs) that have been our area of expertise for the past four decades. The current review aims at providing an overview of the cellular and neuronal components of the pulmonary NEB microenvironment (NEB ME) in mice, underpinning the role of these complexly organized structures in the mechano- and chemosensory potential of airways and lungs. Interestingly, the pulmonary NEB ME additionally harbors different types of stem cells, and emerging evidence suggests that the signal transduction pathways that are active in the NEB ME during lung development and repair also determine the origin of small cell lung carcinoma. Although documented for many years that NEBs appear to be affected in several pulmonary diseases, the current intriguing knowledge on the NEB ME seems to encourage researchers that are new to the field to explore the possibility that these versatile sensor-effector units may be involved in lung pathogenesis or pathobiology.

8.
Respir Res ; 19(1): 207, 2018 Oct 26.
Article in English | MEDLINE | ID: mdl-30367659

ABSTRACT

BACKGROUND: The microenvironment (ME) of neuroepithelial bodies (NEBs) harbors densely innervated groups of pulmonary neuroendocrine cells that are covered by Clara-like cells (CLCs) and is believed to be important during development and for adult airway epithelial repair after severe injury. Yet, little is known about its potential stem cell characteristics in healthy postnatal lungs. METHODS: Transient mild lung inflammation was induced in mice via a single low-dose intratracheal instillation of lipopolysaccharide (LPS). Bronchoalveolar lavage fluid (BALF), collected 16 h after LPS instillation, was used to challenge the NEB ME in ex vivo lung slices of control mice. Proliferating cells in the NEB ME were identified and quantified following simultaneous LPS instillation and BrdU injection. RESULTS: The applied LPS protocol induced very mild and transient lung injury. Challenge of lung slices with BALF of LPS-treated mice resulted in selective Ca2+-mediated activation of CLCs in the NEB ME of control mice. Forty-eight hours after LPS challenge, a remarkably selective and significant increase in the number of divided (BrdU-labeled) cells surrounding NEBs was observed in lung sections of LPS-challenged mice. Proliferating cells were identified as CLCs. CONCLUSIONS: A highly reproducible and minimally invasive lung inflammation model was validated for inducing selective activation of a quiescent stem cell population in the NEB ME. The model creates new opportunities for unraveling the cellular mechanisms/pathways regulating silencing, activation, proliferation and differentiation of this unique postnatal airway epithelial stem cell population.


Subject(s)
Cell Proliferation/physiology , Neuroepithelial Cells/metabolism , Respiratory Mucosa/metabolism , Stem Cell Niche/physiology , Stem Cells/metabolism , Animals , Female , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Respiratory Mucosa/cytology
9.
Curr Biol ; 28(5): 722-732.e6, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29478854

ABSTRACT

Root hairs facilitate a plant's ability to acquire soil anchorage and nutrients. Root hair growth is regulated by the plant hormone auxin and dependent on localized synthesis, secretion, and modification of the root hair tip cell wall. However, the exact cell wall regulators in root hairs controlled by auxin have yet to be determined. In this study, we describe the characterization of ERULUS (ERU), an auxin-induced Arabidopsis receptor-like kinase, whose expression is directly regulated by ARF7 and ARF19 transcription factors. ERU belongs to the Catharanthus roseus RECEPTOR-LIKE KINASE 1-LIKE (CrRLK1L) subfamily of putative cell wall sensor proteins. Imaging of a fluorescent fusion protein revealed that ERU is localized to the apical root hair plasma membrane. ERU regulates cell wall composition in root hairs and modulates pectin dynamics through negative control of pectin methylesterase (PME) activity. Mutant eru (-/-) root hairs accumulate de-esterified homogalacturonan and exhibit aberrant pectin Ca2+-binding site oscillations and increased PME activity. Up to 80% of the eru root hair phenotype is rescued by pharmacological supplementation with a PME-inhibiting catechin extract. ERU transcription is altered in specific cell wall-related root hair mutants, suggesting that it is a target for feedback regulation. Loss of ERU alters the phosphorylation status of FERONIA and H+-ATPases 1/2, regulators of apoplastic pH. Furthermore, H+-ATPases 1/2 and ERU are differentially phosphorylated in response to auxin. We conclude that ERULUS is a key auxin-controlled regulator of cell wall composition and pectin dynamics during root hair tip growth.


Subject(s)
Arabidopsis/genetics , Catharanthus/genetics , Gene Expression Regulation, Plant , Plant Proteins/genetics , Plant Roots/growth & development , Arabidopsis/growth & development , Catharanthus/metabolism , Cell Differentiation , Cell Wall/chemistry , Cell Wall/genetics , Indoleacetic Acids/metabolism , Organogenesis, Plant/genetics , Plant Growth Regulators/metabolism , Plant Proteins/metabolism , Plant Roots/genetics , Plants, Genetically Modified/genetics , Plants, Genetically Modified/growth & development
10.
Ann Transl Med ; 5(6): 129, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28462209

ABSTRACT

Dipeptidyl peptidase 4 (DPP4) is a cell surface protease that has been reported to play a role in glucose homeostasis, cancer, HIV, autoimmunity, immunology and inflammation. A role for DPP4 in ischemia-reperfusion injury (IRI) in the heart has been established. Dipeptidyl peptidase 4 inhibition (DPP4i) appeared to decrease infarct size, improves cardiac function and promotes myocardial regeneration. Lung ischemia reperfusion injury is caused by a complex mechanism in which macrophages and neutrophils play an important role. Generation of reactive oxygen species (ROS), uncoupling of nitric oxide synthase (NOS), activation of nuclear factor-κB (NF-κB), activation of nicotinamide adenine dinucleotide phosphate metabolism, and generation of pro-inflammatory cytokines lead to acute lung injury (ALI). In this review we present the current knowledge on DPP4 as a target to treat IRI in the lung. We also provide evidence of the roles of the DPP4 substrates glucagon-like peptide 1 (GLP-1), vasoactive intestinal peptide (VIP) and stromal cell-derived factor-1α (SDF-1α) in protection against oxidative stress through activation of the mitogen-activated protein kinase (MAPK) 1/2 and phosphatidylinositol 3'-kinase (PI3K)/Akt signal transduction pathways.

11.
Ann Transl Med ; 5(6): 130, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28462210

ABSTRACT

The pathophysiology of lung diseases is very complex and proteolytic enzymes may play a role or could be used as biomarkers. In this review, the literature was searched to make an overview of what is known on the expression of the proline-specific peptidases dipeptidyl peptidase (DPP) 4, 8, 9, prolyl oligopeptidase (PREP) and fibroblast activation protein α (FAP) in the healthy and diseased lung. Search terms included asthma, chronic obstructive pulmonary disease (COPD), lung cancer, fibrosis, ischemia reperfusion injury and pneumonia. Knowledge on the loss or gain of protein expression and activity during disease might tie these enzymes to certain cell types, substrates or interaction partners that are involved in the pathophysiology of the disease, ultimately leading to the elucidation of their functional roles and a potential therapeutic target. Most data could be found on DPP4, while the other enzymes are less explored. Published data however often appear to be conflicting, the applied methods divers and the specificity of the assays used questionable. In conclusion, information on the expression of the proline-specific peptidases in the healthy and diseased lung is lacking, begging for further well-designed research.

12.
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
13.
Cell Tissue Res ; 368(3): 459-467, 2017 06.
Article in English | MEDLINE | ID: mdl-28190088

ABSTRACT

Being continuously exposed to a plethora of antigens ranging from food antigens to potential pathogenic organisms, the gastrointestinal (GI) tract harbors the largest collection of immune cells in the mammalian body. This immune system has to maintain a delicate balance between mounting an active immune response and maintaining tolerance. The GI tract is also home to an elaborate intrinsic nervous system, the enteric nervous system (ENS). Various in vitro studies of neuro-immune communication have suggested that vasoactive intestinal peptide (VIP), an important GI neurotransmitter, modulates mononuclear phagocytes (MNPs), i.e., dendritic cells and macrophages. Using a combined approach of reverse transcription plus the polymerase chain reaction, immunofluorescence, three-dimensional maximum intensity projections and immunoelectron microscopy, we investigate the interaction between the enteric innervation and MNPs in the ileal lamina propria (LP). We demonstrate that VIP-ergic fibers of the ENS lie adjacent to CX3CR1+ MNPs and that VPAC1 is constitutively expressed on ileal CX3CR1+ cells in the LP of the mouse. We also identify, for the first time, CX3CR1+ immune cells in the LP at the ultrastructural level. Our data thus reveal the in situ presence of the molecular components that are necessary for a VIP-mediated neuro-immune interaction between the ENS and CX3CR1-expressing immune cells in the LP of the ileum.


Subject(s)
Chemokine CX3CL1/metabolism , Ileum/immunology , Ileum/innervation , Nerve Fibers/metabolism , Neuroimmunomodulation , Vasoactive Intestinal Peptide/metabolism , Animals , Ileum/metabolism , Ileum/ultrastructure , Mice , Mice, Inbred C57BL , Mononuclear Phagocyte System/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Signal Transduction
14.
Auton Neurosci ; 191: 39-47, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25953244

ABSTRACT

Specific subpopulations of lung-related primary afferent neurons in dorsal root and vagal sensory ganglia have been reported to express P2X2 and P2X3 receptors both in the neuronal cell bodies and in their peripheral terminals. The afferent innervation of airways and lungs is organised as sensory receptor structures, of which at least seven types with a vagal origin and two with a spinal origin have been reported. In view of the recently suggested therapeutic promise of ATP antagonism - specifically at P2X3 receptor expressing nociceptive fibres - in respiratory disorders, the present work focusses on four distinct populations of pulmonary sensory receptors that have so far been reported to express P2X2/3 receptors. Three of them originate from myelinated nerve fibres that display similar mechanosensor-like morphological and neurochemical characteristics. Two of the latter concern vagal nodose sensory fibres, either related to pulmonary neuroepithelial bodies (NEBs), or giving rise to smooth muscle-associated airway receptors (SMARs); the third gives rise to visceral pleura receptors (VPRs) and most likely arises from dorsal root ganglia. The fourth population concerns C-fibre receptors (CFRs) that also derive from neuronal cell bodies located in vagal nodose ganglia. Although the majority of the airway- and lung-related sensory receptors that express P2X2/3 receptors apparently do not belong to accepted nociceptive populations, these data definitely point out that ATP may be an important player in the physiological transduction of different lung-related afferent signals from the periphery to the CNS. The observed variety within the populations of pulmonary sensory receptors that express P2X2/3 receptors argues for a critical and careful interpretation of the functional data.


Subject(s)
Central Nervous System/metabolism , Receptors, Purinergic P2X2/metabolism , Receptors, Purinergic P2X3/metabolism , Respiratory System/metabolism , Animals , Humans , Respiratory Muscles/metabolism
15.
Anat Rec (Hoboken) ; 298(5): 903-16, 2015 May.
Article in English | MEDLINE | ID: mdl-25644190

ABSTRACT

The gastrointestinal (GI) tract, just like the skin and the airways, is constantly exposed to both harmless and pathogenic organisms and hence requires a tightly regulated immune homeostasis to function properly. A central role in the regulation of this balance is played by the dendritic cells (DCs), a heterogeneous population of antigen-presenting cells that can be further divided into distinct subsets with different functions depending on the tissue they reside in. In recent years, the DC population in the lamina propria (LP) of the intestine has emerged as a key player in immune surveillance. Given the extensive innervation of the GI mucosa, these DC subsets possibly are also regulated by interactions with neuronal components. Current knowledge, be it still fragmentary, indicates that dysregulation of this neuroimmune communication leads to the onset of pathological disorders. The present review article deals with the identification and interaction of distinct subtypes of mouse intestinal LP DCs with elements of the enteric nervous system (ENS) in normal and inflammatory conditions. Furthermore, the question is addressed whether any parallels can be drawn between intestinal LP DCs and DCs residing in the skin and lung in order to gain a better insight into common or clearly distinct mechanistic pathways and the possible impact of the mucosal components in the microenvironment. The exact way in which the ENS is serving its immunomodulatory roles in the GI tract is still largely unknown, although there are significant indications for a crosstalk between LP DCs and components of the ENS. This review clearly shows that in the three different organ systems the same neurotransmitters (i.e., SP, CGRP, and VIP) reoccur, serving similar functions. Mechanistic lessons learned from other organ systems, such as the skin and lung, may be of substantial help in further exploring the nature of the neuroimmune communication between GI innervation and LP DCs.


Subject(s)
Cell Communication/physiology , Dendritic Cells/cytology , Intestines/cytology , Neuroimmunomodulation/physiology , Animals , Dendritic Cells/immunology , Enteric Nervous System/immunology , Humans , Intestines/immunology
16.
Cell Tissue Res ; 359(2): 453-463, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25358399

ABSTRACT

Corticotropin-releasing factor (CRF) and urocortins (UCNs) are important ligands in the CRF signaling pathways, which are most known for their role in the hypothalamic-pituitary-adrenal stress axis. However, peripheral CRF signaling also has profound effects on gastrointestinal functions. Although the murine animal model is highly relevant for the exploration of this complexly balanced pathway via genetic manipulation, little is known about the expression of CRF and UCNs in the mouse intestine. This study aims to investigate the cellular localization of CRF and UCNs in the ileum and to explore whether and how this cellular expression is altered in conditions of intestinal Schistosoma mansoni-induced inflammation. The results show a distinct expression pattern for the different CRF receptor ligands in the ileum. CRF was located in nerve fibers and stromal cells. All UCNs were expressed in polymorphonuclear leukocytes. Furthermore, UCN2 and UCN3 were found in the musculature. During acute schistosomiasis, UCN1 showed an increased immunoreactivity in blood vessels and UCN3 was de novo expressed mainly in submucous neurons. Typical features of S. mansoni-inflamed ileum, such as nerve fiber sprouting, muscle layer thickening and granuloma formation thus all have an impact on the CRF signaling pathways. In conclusion, we outline for the first time the expression of CRF signaling ligands in the mouse ileum; our results point to important changes of this signaling system in S. mansoni-induced intestinal inflammation, which warrants further functional investigation with specific focus on CRF2, given the exclusive binding of UCN2 and UCN3 to this receptor.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Ileum/parasitology , Schistosoma mansoni/physiology , Schistosomiasis mansoni/metabolism , Schistosomiasis mansoni/parasitology , Urocortins/metabolism , Animals , Corticotropin-Releasing Hormone/genetics , Ileum/metabolism , Ileum/pathology , Immunohistochemistry , Inflammation/pathology , Leukocytes/metabolism , Ligands , Male , Mice, Inbred C57BL , Muscles/metabolism , Nerve Fibers/metabolism , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/pathology , Stromal Cells/metabolism , Up-Regulation , Urocortins/genetics
17.
J Exp Bot ; 65(18): 5485-95, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25147272

ABSTRACT

The synthesis and composition of cell walls is dynamically adapted in response to many developmental and environmental signals. In this respect, cell wall proteins involved in controlling cell elongation are critical for cell development. Transcriptome analysis identified a gene in Arabidopsis thaliana, which was named proline-rich protein-like, AtPRPL1, based on sequence similarities from a phylogenetic analysis. The most resemblance was found to AtPRP1 and AtPRP3 from Arabidopsis, which are known to be involved in root hair growth and development. In A. thaliana four proline-rich cell wall protein genes, playing a role in building up the cross-connections between cell wall components, can be distinguished. AtPRPL1 is a small gene that in promoter::GUS (ß-glucuronidase) analysis has high expression in trichoblast cells and in the collet. Chemical or mutational interference with root hair formation inhibited this expression. Altered expression levels in knock-out or overexpression lines interfered with normal root hair growth and etiolated hypocotyl development, but Fourier transform-infrared (FT-IR) analysis did not identify consistent changes in cell wall composition of root hairs and hypocotyl. Co-localization analysis of the AtPRPL1-green fluorescent protein (GFP) fusion protein and different red fluorescent protein (RFP)-labelled markers confirmed the presence of AtPRPL1-GFP in small vesicles moving over the endoplasmic reticulum. Together, these data indicate that the AtPRPL1 protein is involved in the cell's elongation process. How exactly this is achieved remains unclear at present.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Plant Roots/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Gene Expression Regulation, Plant/genetics , Gene Expression Regulation, Plant/physiology , Hypocotyl/genetics , Hypocotyl/metabolism , Plant Roots/genetics
18.
Contrast Media Mol Imaging ; 9(6): 400-8, 2014.
Article in English | MEDLINE | ID: mdl-24753446

ABSTRACT

In this study, the interaction between cells and micron-sized paramagnetic iron oxide (MPIO) particles was investigated by characterizing MPIO in their original state, and after cellular uptake in vitro as well as in vivo. Moreover, MPIO in the olfactory bulb were studied 9 months after injection. Using various imaging techniques, cell-MPIO interactions were investigated with increasing spatial resolution. Live cell confocal microscopy demonstrated that MPIO co-localize with lysosomes after in vitro cellular uptake. In more detail, a membrane surrounding the MPIO was observed by high-angle annular dark-field scanning transmission electron microscopy (HAADF-STEM). Following MPIO uptake in vivo, the same cell-MPIO interaction was observed by HAADF-STEM in the subventricular zone at 1 week and in the olfactory bulb at 9 months after MPIO injection. These findings provide proof for the current hypothesis that MPIO are internalized by the cell through endocytosis. The results also show MPIO are not biodegradable, even after 9 months in the brain. Moreover, they show the possibility of HAADF-STEM generating information on the labeled cell as well as on the MPIO. In summary, the methodology presented here provides a systematic route to investigate the interaction between cells and nanoparticles from the micrometer level down to the nanometer level and beyond.


Subject(s)
Ferric Compounds , Mesenchymal Stem Cells/ultrastructure , Multimodal Imaging/methods , Olfactory Bulb/ultrastructure , Animals , Cell Line , Cell Tracking/methods , Endocytosis/drug effects , Ferric Compounds/chemistry , Humans , Lysosomes/chemistry , Metal Nanoparticles/chemistry , Mice , Microscopy, Electron, Scanning Transmission , Particle Size , Staining and Labeling
19.
PLoS One ; 8(11): e80294, 2013.
Article in English | MEDLINE | ID: mdl-24282533

ABSTRACT

Airway branching morphogenesis in utero is essential for optimal postnatal lung function. In the fetus, branching morphogenesis occurs during the pseudoglandular stage (weeks 9-17 of human gestation, embryonic days (E)11.5-16.5 in mouse) in a hypercalcaemic environment (~1.7 in the fetus vs. ~1.1-1.3 mM for an adult). Previously we have shown that fetal hypercalcemia exerts an inhibitory brake on branching morphogenesis via the calcium-sensing receptor. In addition, earlier studies have shown that nifedipine, a selective blocker of L-type voltage-gated Ca(2+) channels (VGCC), inhibits fetal lung growth, suggesting a role for VGCC in lung development. The aim of this work was to investigate the expression of VGCC in the pseudoglandular human and mouse lung, and their role in branching morphogenesis. Expression of L-type (CaV1.2 and CaV1.3), P/Q type (CaV2.1), N-type (CaV2.2), R-type (CaV2.3), and T-type (CaV3.2 and CaV3.3) VGCC was investigated in paraffin sections from week 9 human fetal lungs and E12.5 mouse embryos. Here we show, for the first time, that Cav1.2 and Cav1.3 are expressed in both the smooth muscle and epithelium of the developing human and mouse lung. Additionally, Cav2.3 was expressed in the lung epithelium of both species. Incubating E12.5 mouse lung rudiments in the presence of nifedipine doubled the amount of branching, an effect which was partly mimicked by the Cav2.3 inhibitor, SNX-482. Direct measurements of changes in epithelial cell membrane potential, using the voltage-sensitive fluorescent dye DiSBAC2(3), demonstrated that cyclic depolarisations occur within the developing epithelium and coincide with rhythmic occlusions of the lumen, driven by the naturally occurring airway peristalsis. We conclude that VGCC are expressed and functional in the fetal human and mouse lung, where they play a role in branching morphogenesis. Furthermore, rhythmic epithelial depolarisations evoked by airway peristalsis would allow for branching to match growth and distension within the developing lung.


Subject(s)
Calcium Channels/physiology , Calcium/blood , Lung/embryology , Morphogenesis , Animals , Epithelium/metabolism , Humans , Mice , Muscle, Smooth/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...