Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Nature ; 604(7904): 111-119, 2022 04.
Article in English | MEDLINE | ID: mdl-35355018

ABSTRACT

Mapping the spatial distribution and molecular identity of constituent cells is essential for understanding tissue dynamics in health and disease. We lack a comprehensive map of human distal airways, including the terminal and respiratory bronchioles (TRBs), which are implicated in respiratory diseases1-4. Here, using spatial transcriptomics and single-cell profiling of microdissected distal airways, we identify molecularly distinct TRB cell types that have not-to our knowledge-been previously characterized. These include airway-associated LGR5+ fibroblasts and TRB-specific alveolar type-0 (AT0) cells and TRB secretory cells (TRB-SCs). Connectome maps and organoid-based co-cultures reveal that LGR5+ fibroblasts form a signalling hub in the airway niche. AT0 cells and TRB-SCs are conserved in primates and emerge dynamically during human lung development. Using a non-human primate model of lung injury, together with human organoids and tissue specimens, we show that alveolar type-2 cells in regenerating lungs transiently acquire an AT0 state from which they can differentiate into either alveolar type-1 cells or TRB-SCs. This differentiation programme is distinct from that identified in the mouse lung5-7. Our study also reveals mechanisms that drive the differentiation of the bipotent AT0 cell state into normal or pathological states. In sum, our findings revise human lung cell maps and lineage trajectories, and implicate an epithelial transitional state in primate lung regeneration and disease.


Subject(s)
Cell Lineage , Lung , Stem Cells , Alveolar Epithelial Cells , Animals , Cell Differentiation , Connectome , Fibroblasts , Gene Expression Profiling , Humans , Lung/cytology , Lung Diseases , Mice , Organoids , Primates , Regeneration , Single-Cell Analysis , Stem Cells/cytology
2.
Annu Rev Cell Dev Biol ; 27: 493-512, 2011.
Article in English | MEDLINE | ID: mdl-21639799

ABSTRACT

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


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/physiology , Lung Diseases/physiopathology , Lung/cytology , Lung/growth & development , Stem Cells/cytology , Stem Cells/physiology , Airway Remodeling , Animals , Cell Differentiation , Cell Lineage , Homeostasis , Humans , Lung/pathology , Lung/physiology , Lung Diseases/pathology , Organogenesis/physiology , Respiratory System , Wound Healing
3.
Nature ; 517(7536): 621-5, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25533958

ABSTRACT

Broadly, tissue regeneration is achieved in two ways: by proliferation of common differentiated cells and/or by deployment of specialized stem/progenitor cells. Which of these pathways applies is both organ- and injury-specific. Current models in the lung posit that epithelial repair can be attributed to cells expressing mature lineage markers. By contrast, here we define the regenerative role of previously uncharacterized, rare lineage-negative epithelial stem/progenitor (LNEP) cells present within normal distal lung. Quiescent LNEPs activate a ΔNp63 (a p63 splice variant) and cytokeratin 5 remodelling program after influenza or bleomycin injury in mice. Activated cells proliferate and migrate widely to occupy heavily injured areas depleted of mature lineages, at which point they differentiate towards mature epithelium. Lineage tracing revealed scant contribution of pre-existing mature epithelial cells in such repair, whereas orthotopic transplantation of LNEPs, isolated by a definitive surface profile identified through single-cell sequencing, directly demonstrated the proliferative capacity and multipotency of this population. LNEPs require Notch signalling to activate the ΔNp63 and cytokeratin 5 program, and subsequent Notch blockade promotes an alveolar cell fate. Persistent Notch signalling after injury led to parenchymal 'micro-honeycombing' (alveolar cysts), indicative of failed regeneration. Lungs from patients with fibrosis show analogous honeycomb cysts with evidence of hyperactive Notch signalling. Our findings indicate that distinct stem/progenitor cell pools repopulate injured tissue depending on the extent of the injury, and the outcomes of regeneration or fibrosis may depend in part on the dynamics of LNEP Notch signalling.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/pathology , Lung Injury/pathology , Lung/cytology , Lung/pathology , Re-Epithelialization , Stem Cells/cytology , Animals , Bleomycin , Cell Lineage , Cell Proliferation , Cell Separation , Cysts/metabolism , Cysts/pathology , Epithelial Cells/metabolism , Female , Humans , Keratin-5/metabolism , Lung/physiology , Lung Injury/chemically induced , Lung Injury/virology , Male , Mice , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Stem Cell Transplantation , Stem Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
4.
J Physiol ; 596(9): 1549-1574, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29430647

ABSTRACT

KEY POINTS: Enteric neurotransmission is essential for gastrointestinal (GI) motility, although the cells and conductances responsible for post-junctional responses are controversial. The calcium-activated chloride conductance (CaCC), anoctamin-1 (Ano1), was expressed by intramuscular interstitial cells of Cajal (ICC-IM) in proximal stomach and not resolved in smooth muscle cells (SMCs). Cholinergic nerve fibres were closely apposed to ICC-IM. Conductances activated by cholinergic stimulation in isolated ICC-IM and SMCs were determined. A CaCC was activated by carbachol in ICC-IM and a non-selective cation conductance in SMCs. Responses to cholinergic nerve stimulation were studied. Excitatory junction potentials (EJPs) and mechanical responses were evoked in wild-type mice but absent or greatly reduced with knockout/down of Ano1. Drugs that block Ano1 inhibited the conductance activated by carbachol in ICC-IM and EJPs and mechanical responses in tissues. The data of the present study suggest that electrical and mechanical responses to cholinergic nerve stimulation are mediated by Ano1 expressed in ICC-IM and not SMCs. ABSTRACT: Enteric motor neurotransmission is essential for normal gastrointestinal (GI) motility. Controversy exists regarding the cells and ionic conductance(s) that mediate post-junctional neuroeffector responses to motor neurotransmitters. Isolated intramuscular ICC (ICC-IM) and smooth muscle cells (SMCs) from murine fundus muscles were used to determine the conductances activated by carbachol (CCh) in each cell type. The calcium-activated chloride conductance (CaCC), anoctamin-1 (Ano1) is expressed by ICC-IM but not resolved in SMCs, and CCh activated a Cl- conductance in ICC-IM and a non-selective cation conductance in SMCs. We also studied responses to nerve stimulation using electrical-field stimulation (EFS) of intact fundus muscles from wild-type and Ano1 knockout mice. EFS activated excitatory junction potentials (EJPs) in wild-type mice, although EJPs were absent in mice with congenital deactivation of Ano1 and greatly reduced in animals in which the CaCC-Ano1 was knocked down using Cre/loxP technology. Contractions to cholinergic nerve stimulation were also greatly reduced in Ano1 knockouts. SMCs cells also have receptors and ion channels activated by muscarinic agonists. Blocking acetylcholine esterase with neostigmine revealed a slow depolarization that developed after EJPs in wild-type mice. This depolarization was still apparent in mice with genetic deactivation of Ano1. Pharmacological blockers of Ano1 also inhibited EJPs and contractile responses to muscarinic stimulation in fundus muscles. The data of the present study are consistent with the hypothesis that ACh released from motor nerves binds muscarinic receptors on ICC-IM with preference and activates Ano1. If metabolism of acetylcholine is inhibited, ACh overflows and binds to extrajunctional receptors on SMCs, eliciting a slower depolarization response.


Subject(s)
Acetylcholine/metabolism , Interstitial Cells of Cajal/physiology , Myocytes, Smooth Muscle/physiology , Stomach/physiology , Synaptic Transmission , Animals , Anoctamin-1/physiology , Chloride Channels/physiology , Electric Stimulation , Gastric Fundus/cytology , Gastric Fundus/physiology , Interstitial Cells of Cajal/cytology , Mice , Mice, Knockout , Muscle Contraction , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Stomach/cytology
5.
Proc Natl Acad Sci U S A ; 112(8): 2575-80, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25675481

ABSTRACT

The developmental rehearsal for the debut of hearing is marked by massive changes in the membrane properties of hair cells (HCs) and spiral ganglion neurons (SGNs). Whereas the underlying mechanisms for the developing HC transition to mature stage are understood in detail, the maturation of SGNs from hyperexcitable prehearing to quiescent posthearing neurons with broad dynamic range is unknown. Here, we demonstrated using pharmacological approaches, caged-Ca(2+) photolysis, and gramicidin patch recordings that the prehearing SGN uses Ca(2+)-activated Cl(-) conductance to depolarize the resting membrane potential and to prime the neurons in a hyperexcitable state. Immunostaining of the cochlea preparation revealed the identity and expression of the Ca(2+)-activated Cl(-) channel transmembrane member 16A (TMEM16A) in SGNs. Moreover, null deletion of TMEM16A reduced the Ca(2+)-activated Cl(-) currents and action potential firing in SGNs. To determine whether Cl(-) ions and TMEM16A are involved in the transition between pre- and posthearing features of SGNs we measured the intracellular Cl(-) concentration [Cl(-)]i in SGNs. Surprisingly, [Cl(-)]i in SGNs from prehearing mice was ∼90 mM, which was significantly higher than posthearing neurons, ∼20 mM, demonstrating discernible altered roles of Cl(-) channels in the developing neuron. The switch in [Cl(-)]i stems from delayed expression of the development of intracellular Cl(-) regulating mechanisms. Because the Cl(-) channel is the only active ion-selective conductance with a reversal potential that lies within the dynamic range of SGN action potentials, developmental alteration of [Cl(-)]i, and hence the equilibrium potential for Cl(-) (ECl), transforms pre- to posthearing phenotype.


Subject(s)
Chloride Channels/metabolism , Membrane Potentials , Neurons/physiology , Spiral Ganglion/physiology , Action Potentials/drug effects , Animals , Anoctamin-1 , Anoctamins , Calcium/pharmacology , Cell Membrane/drug effects , Cell Membrane/physiology , Chloride Channels/antagonists & inhibitors , Chlorides/metabolism , Female , Hearing/physiology , Male , Membrane Potentials/drug effects , Mice, Knockout , Neurons/drug effects , Phenotype , Solute Carrier Family 12, Member 2/metabolism , Spiral Ganglion/drug effects , Symporters/metabolism , K Cl- Cotransporters
6.
J Urol ; 196(4): 1295-302, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27173853

ABSTRACT

PURPOSE: Lower urinary tract malformations are among the most common congenital anomalies in humans. Molecular genetic studies of mouse external genital development have begun to identify mechanisms that pattern the genital tubercle and orchestrate urethral tubulogenesis. The urethral plate epithelium is an endodermal signaling region that has an essential role in external genital development. However, little is known about the molecular identity of this cell population or the genes that regulate its activity. MATERIALS AND METHODS: We used microarray analysis to characterize differences in gene expression between urethral plate epithelium and surrounding tissue in mouse genital tubercles. In situ hybridizations were performed to map gene expression patterns and ToppCluster (https://toppcluster.cchmc.org/) was used to analyze gene associations. RESULTS: A total of 84 genes were enriched at least 20-fold in urethral plate epithelium relative to surrounding tissue. The majority of these genes were expressed throughout the urethral plate in males and females at embryonic day 12.5 when the urethral plate is known to signal. Functional analysis using ToppCluster revealed genetic pathways with known functions in other organ systems but unknown roles in external genital development. Additionally, a 3-dimensional molecular atlas of genes enriched in urethral plate epithelium was generated and deposited at the GUDMAP (GenitoUrinary Development Molecular Anatomy Project) website (http://gudmap.org/). CONCLUSIONS: We identified dozens of genes previously unknown to be expressed in urethral plate epithelium at a crucial developmental period. It provides a novel panel of genes for analysis in animal models and in humans with external genital anomalies.


Subject(s)
Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , RNA/genetics , Urethra/embryology , Urothelium/embryology , Animals , Female , Hedgehog Proteins/biosynthesis , In Situ Hybridization , Male , Mice , Models, Animal , Protein Array Analysis , Signal Transduction , Urethra/metabolism , Urothelium/metabolism
7.
BMC Dev Biol ; 15: 32, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26376663

ABSTRACT

Recent studies have identified epithelial stem and progenitor cell populations of the lung. We are just beginning to understand the mechanisms that regulate their homeostatic, regenerative and maladaptive behaviors. Here, we discuss evidence of regulatory niches for epithelial stem cells of the lung.


Subject(s)
Airway Remodeling , Lung/cytology , Stem Cell Niche , Animals , Bronchi/cytology , Disease Models, Animal , Fibrosis/pathology , Humans , Lung/pathology , Mice , Pulmonary Alveoli/cytology
8.
Pflugers Arch ; 467(6): 1203-13, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24974903

ABSTRACT

Intestinal epithelial electrolyte secretion is activated by increase in intracellular cAMP or Ca(2+) and opening of apical Cl(-) channels. In infants and young animals, but not in adults, Ca(2+)-activated chloride channels may cause secretory diarrhea during rotavirus infection. While detailed knowledge exists concerning the contribution of cAMP-activated cystic fibrosis transmembrane conductance regulator (CFTR) channels, analysis of the role of Ca(2+)-dependent Cl(-) channels became possible through identification of the anoctamin (TMEM16) family of proteins. We demonstrate expression of several anoctamin paralogues in mouse small and large intestines. Using intestinal-specific mouse knockout models for anoctamin 1 (Ano1) and anoctamin 10 (Ano10) and a conventional knockout model for anoctamin 6 (Ano6), we demonstrate the role of anoctamins for Ca(2+)-dependent Cl(-) secretion induced by the muscarinic agonist carbachol (CCH). Ano1 is preferentially expressed in the ileum and large intestine, where it supports Ca(2+)-activated Cl(-) secretion. In contrast, Ano10 is essential for Ca(2+)-dependent Cl(-) secretion in jejunum, where expression of Ano1 was not detected. Although broadly expressed, Ano6 has no role in intestinal cholinergic Cl(-) secretion. Ano1 is located in a basolateral compartment/membrane rather than in the apical membrane, where it supports CCH-induced Ca(2+) increase, while the essential and possibly only apical Cl(-) channel is CFTR. These results define a new role of Ano1 for intestinal Ca(2+)-dependent Cl(-) secretion and demonstrate for the first time a contribution of Ano10 to intestinal transport.


Subject(s)
Calcium Signaling , Chloride Channels/metabolism , Chlorides/metabolism , Intestinal Mucosa/metabolism , Phospholipid Transfer Proteins/metabolism , Animals , Anoctamin-1 , Anoctamins , Calcium/metabolism , Chloride Channels/genetics , Mice , Organ Specificity , Phospholipid Transfer Proteins/genetics
9.
Proc Natl Acad Sci U S A ; 109(13): 4910-5, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22411819

ABSTRACT

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


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Animals , Bronchioles/metabolism , Bronchioles/pathology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Disease Progression , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/metabolism , Hyperplasia , Intercellular Signaling Peptides and Proteins , Lung Neoplasms/genetics , Mice , Models, Biological , Mutant Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Peptides/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Pulmonary Surfactant-Associated Protein C , SOXB1 Transcription Factors/metabolism , Time Factors , Transcriptome/genetics , Uteroglobin/metabolism
10.
Proc Natl Acad Sci U S A ; 109(40): 16354-9, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22988107

ABSTRACT

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


Subject(s)
Chloride Channels/metabolism , Mucins/metabolism , Muscle Contraction/physiology , Muscle, Smooth/physiology , Respiratory System/cytology , Respiratory System/metabolism , Animals , Anoctamin-1 , Cells, Cultured , Epithelial Cells/metabolism , Humans , Immunohistochemistry , Mice , Microscopy, Fluorescence
11.
J Physiol ; 592(18): 4051-68, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25063822

ABSTRACT

Interstitial cells of Cajal (ICC) are pacemaker cells that generate electrical activity to drive contractility in the gastrointestinal tract via ion channels. Ano1 (Tmem16a), a Ca(2+)-activated Cl(-) channel, is an ion channel expressed in ICC. Genetic deletion of Ano1 in mice resulted in loss of slow waves in smooth muscle of small intestine. In this study, we show that Ano1 is required to maintain coordinated Ca(2+) transients between myenteric ICC (ICC-MY) of small intestine. First, we found spontaneous Ca(2+) transients in ICC-MY in both Ano1 WT and knockout (KO) mice. However, Ca(2+) transients within the ICC-MY network in Ano1 KO mice were uncoordinated, while ICC-MY Ca(2+) transients in Ano1 WT mice were rhythmic and coordinated. To confirm the role of Ano1 in the loss of Ca(2+) transient coordination, we used pharmacological inhibitors of Ano1 activity and shRNA-mediated knock down of Ano1 expression in organotypic cultures of Ano1 WT small intestine. Coordinated Ca(2+) transients became uncoordinated using both these approaches, supporting the conclusion that Ano1 is required to maintain coordination/rhythmicity of Ca(2+) transients. We next determined the effect on smooth muscle contractility using spatiotemporal maps of contractile activity in Ano1 KO and WT tissues. Significantly decreased contractility that appeared to be non-rhythmic and uncoordinated was observed in Ano1 KO jejunum. In conclusion, Ano1 has a previously unidentified role in the regulation of coordinated gastrointestinal smooth muscle function through coordination of Ca(2+) transients in ICC-MY.


Subject(s)
Calcium Signaling , Chloride Channels/metabolism , Interstitial Cells of Cajal/metabolism , Jejunum/metabolism , Muscle Contraction , Animals , Anoctamin-1 , Calcium/metabolism , Chloride Channels/genetics , Interstitial Cells of Cajal/physiology , Jejunum/physiology , Mice
12.
Kidney Int ; 85(6): 1369-81, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24476694

ABSTRACT

The role of calcium-activated chloride channels for renal function is unknown. By immunohistochemistry we demonstrate dominant expression of the recently identified calcium-activated chloride channels, Anoctamin 1 (Ano1, TMEM16A) in human and mouse proximal tubular epithelial (PTE) cells, with some expression in podocytes and other tubular segments. Ano1-null mice had proteinuria and numerous large reabsorption vesicles in PTE cells. Selective knockout of Ano1 in podocytes (Ano1-/-/Nphs2-Cre) did not impair renal function, whereas tubular knockout in Ano1-/-/Ksp-Cre mice increased urine protein excretion and decreased urine electrolyte concentrations. Purinergic stimulation activated calcium-dependent chloride currents in isolated proximal tubule epithelial cells from wild-type but not from Ano1-/-/Ksp-Cre mice. Ano1 currents were activated by acidic pH, suggesting parallel stimulation of Ano1 chloride secretion with activation of the proton-ATPase. Lack of calcium-dependent chloride secretion in cells from Ano1-/-/Ksp-Cre mice was paralleled by attenuated proton secretion and reduced endosomal acidification, which compromised proximal tubular albumin uptake. Tubular knockout of Ano1 enhanced serum renin and aldosterone concentrations, probably leading to enhanced compensatory distal tubular reabsorption, thus maintaining normal blood pressure levels. Thus, Ano1 has a role in proximal tubular proton secretion and protein reabsorption. The results correspond to regulation of the proton-ATPase by the Ano1-homolog Ist2 in yeast.


Subject(s)
Chloride Channels/metabolism , Kidney Tubules, Proximal/metabolism , Podocytes/metabolism , Renal Reabsorption , Adenosine Triphosphate/pharmacology , Aldosterone/blood , Animals , Anoctamin-1 , Cells, Cultured , Chloride Channels/deficiency , Chloride Channels/drug effects , Chloride Channels/genetics , Female , Genotype , Humans , Hydrogen-Ion Concentration , Ion Channel Gating , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/physiopathology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phenotype , Podocytes/drug effects , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/physiopathology , Renal Reabsorption/drug effects , Renin/blood , Time Factors , Vacuolar Proton-Translocating ATPases/metabolism
13.
Proc Natl Acad Sci U S A ; 108(52): E1475-83, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22123957

ABSTRACT

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


Subject(s)
Cell Differentiation/physiology , Pulmonary Alveoli/cytology , Pulmonary Fibrosis/pathology , Stromal Cells/cytology , Actins/metabolism , Animals , Biomarkers/metabolism , Bleomycin/toxicity , Bromodeoxyuridine , Cell Proliferation , Epithelial-Mesenchymal Transition/physiology , Extracellular Matrix Proteins/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Mice , Myofibroblasts/cytology , Pericytes/metabolism , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/chemically induced , Real-Time Polymerase Chain Reaction , Stromal Cells/metabolism
14.
Biol Reprod ; 86(1): 1-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21976594

ABSTRACT

Myosalpinx contractions are critical for oocyte transport along the oviduct. A specialized population of pacemaker cells-oviduct interstitial cells of Cajal-generate slow waves, the electrical events underlying myosalpinx contractions. The ionic basis of oviduct pacemaker activity is unknown. We examined the role of a new class of Ca(2+)-activated Cl(-) channels (CaCCs)-anoctamin 1, encoded by Tmem16a-in oviduct slow wave generation. RT-PCR revealed the transcriptional expression of Tmem16a-encoded CaCCs in the myosalpinx. Intracellular microelectrode recordings were performed in the presence of two pharmacologically distinct Cl(-) channel antagonists, anthracene-9-carboxylic acid and niflumic acid. Both of these inhibitors caused membrane hyperpolarization, reduced the duration of slow waves, and ultimately inhibited pacemaker activity. Niflumic acid also inhibited propagating calcium waves within the myosalpinx. Slow waves were present at birth in wild-type and heterozygous oviducts but failed to develop by birth in mice homozygous for a null allele of Tmem16a (Tmem16a(tm1Bdh/tm1Bdh)). These data suggest that Tmem16a-encoded CaCCs contribute to membrane potential and are responsible for the upstroke and plateau phases of oviduct slow waves.


Subject(s)
Chloride Channels/metabolism , Electrophysiological Phenomena/physiology , Fallopian Tubes/physiology , Gene Expression Regulation/physiology , Animals , Anoctamin-1 , Anthracenes/pharmacology , Calcium/metabolism , Chloride Channels/antagonists & inhibitors , Chloride Channels/genetics , Female , Genotype , Mice , Mice, Inbred BALB C , Mice, Knockout , Muscle, Smooth/metabolism , Mutation , Reverse Transcriptase Polymerase Chain Reaction
15.
Proc Natl Acad Sci U S A ; 106(31): 12771-5, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19625615

ABSTRACT

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


Subject(s)
Lung/cytology , Stem Cells/cytology , Trachea/cytology , Animals , Cell Proliferation , Cells, Cultured , Female , Gene Expression Profiling , Humans , Male , Mice , Oligonucleotide Array Sequence Analysis , Receptor, Nerve Growth Factor/analysis , Trachea/metabolism
16.
Proc Natl Acad Sci U S A ; 106(50): 21413-8, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19965375

ABSTRACT

Calcium-activated chloride channels (CaCC) with similar hallmark features are present in many cell types and mediate important physiological functions including epithelial secretion, sensory signal transduction, and smooth muscle contraction. Having identified TMEM16A of the transmembrane proteins with unknown function (TMEM) 16 family as a CaCC subunit, we have developed antibodies specific for mouse TMEM16A, as evidenced by the absence of immunoreactivity in TMEM16A knockout mice. Here, we show that TMEM16A is located in the apical membranes of epithelial cells in exocrine glands and trachea. In addition, TMEM16A is expressed in airway smooth muscle cells and the smooth muscle cells of reproductive tracts, the oviduct and ductus epididymis. In the gastrointestinal (GI) tract, TMEM16A is absent from smooth muscle cells, but present in the interstitial cells of Cajal (ICC), the pacemaker cells that control smooth muscle contraction. The physiological importance of TMEM16A is underscored by the diminished rhythmic contraction of gastric smooth muscle from TMEM16A knockout mice. The TMEM16A expression pattern established in this study thus provides a roadmap for the analyses of physiological functions of calcium-activated chloride channels that contain TMEM16A subunits.


Subject(s)
Chloride Channels/analysis , Animals , Anoctamin-1 , Antibodies, Monoclonal , Exocrine Glands/chemistry , Exocrine Glands/cytology , Gastrointestinal Tract/chemistry , Gastrointestinal Tract/cytology , Immunohistochemistry , Mice , Tissue Distribution , Trachea/chemistry , Trachea/cytology , Urogenital System/chemistry , Urogenital System/cytology
17.
J Clin Invest ; 132(9)2022 05 02.
Article in English | MEDLINE | ID: mdl-35316222

ABSTRACT

Pericyte-mediated capillary constriction decreases cerebral blood flow in stroke after an occluded artery is unblocked. The determinants of pericyte tone are poorly understood. We show that a small rise in cytoplasmic Ca2+ concentration ([Ca2+]i) in pericytes activated chloride efflux through the Ca2+-gated anion channel TMEM16A, thus depolarizing the cell and opening voltage-gated calcium channels. This mechanism strongly amplified the pericyte [Ca2+]i rise and capillary constriction evoked by contractile agonists and ischemia. In a rodent stroke model, TMEM16A inhibition slowed the ischemia-evoked pericyte [Ca2+]i rise, capillary constriction, and pericyte death; reduced neutrophil stalling; and improved cerebrovascular reperfusion. Genetic analysis implicated altered TMEM16A expression in poor patient recovery from ischemic stroke. Thus, pericyte TMEM16A is a crucial regulator of cerebral capillary function and a potential therapeutic target for stroke and possibly other disorders of impaired microvascular flow, such as Alzheimer's disease and vascular dementia.


Subject(s)
Pericytes , Stroke , Calcium/metabolism , Cerebrovascular Circulation/genetics , Humans , Ischemia/metabolism , Pericytes/metabolism , Stroke/metabolism
19.
J Biol Chem ; 285(17): 12990-3001, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20177062

ABSTRACT

Activation of an apical Ca(2+)-dependent Cl(-) channel (CaCC) is the rate-limiting step for fluid secretion in many exocrine tissues. Here, we compared the properties of native CaCC in mouse submandibular salivary gland acinar cells to the Ca(2+)-gated Cl(-) currents generated by Tmem16A and Best2, members from two distinct families of Ca(2+)-activated Cl(-) channels found in salivary glands. Heterologous expression of Tmem16A and Best2 transcripts in HEK293 cells produced Ca(2+)-activated Cl(-) currents with time and voltage dependence and inhibitor sensitivity that resembled the Ca(2+)-activated Cl(-) current found in native salivary acinar cells. Best2(-/-) and Tmem16A(-/-) mice were used to further characterize the role of these channels in the exocrine salivary gland. The amplitude and the biophysical footprint of the Ca(2+)-activated Cl(-) current in submandibular gland acinar cells from Best2-deficient mice were the same as in wild type cells. Consistent with this observation, the fluid secretion rate in Best2 null mice was comparable with that in wild type mice. In contrast, submandibular gland acinar cells from Tmem16A(-/-) mice lacked a Ca(2+)-activated Cl(-) current and a Ca(2+)-mobilizing agonist failed to stimulate Cl(-) efflux, requirements for fluid secretion. Furthermore, saliva secretion was abolished by the CaCC inhibitor niflumic acid in wild type and Best2(-/-) mice. Our results demonstrate that both Tmem16A and Best2 generate Ca(2+)-activated Cl(-) current in vitro with similar properties to those expressed in native cells, yet only Tmem16A appears to be a critical component of the acinar Ca(2+)-activated Cl(-) channel complex that is essential for saliva production by the submandibular gland.


Subject(s)
Calcium/metabolism , Chloride Channels/metabolism , Chlorides/metabolism , Submandibular Gland/metabolism , Animals , Anoctamin-1 , Chloride Channels/genetics , Gene Expression Regulation/physiology , Ion Transport/physiology , Mice , Mice, Knockout , Organ Specificity/physiology , Submandibular Gland/cytology
20.
Am J Physiol Gastrointest Liver Physiol ; 301(6): G1044-51, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21940901

ABSTRACT

Ano1 is a recently discovered Ca(2+)-activated Cl(-) channel expressed on interstitial cells of Cajal (ICC) that has been implicated in slow-wave activity in the gut. However, Ano1 is expressed on all classes of ICC, even those that do not contribute to generation of the slow wave, suggesting that Ano1 may have an alternate function in these cells. Ano1 is also highly expressed in gastrointestinal stromal tumors. Mice lacking Ano1 had fewer proliferating ICC in whole mount preparations and in culture, raising the possibility that Ano1 is involved in proliferation. Cl(-) channel blockers decreased proliferation in cells expressing Ano1, including primary cultures of ICC and in the pancreatic cancer-derived cell line, CFPAC-1. Cl(-) channel blockers had a reduced effect on Ano1(-/-) cultures, confirming that the blockers are acting on Ano1. Ki67 immunoreactivity, 5-ethynyl-2'-deoxyuridine incorporation, and cell-cycle analysis of cells grown in low-Cl(-) media showed fewer proliferating cells than in cultures grown in regular medium. We confirmed that mice lacking Ano1 had less phosphorylated retinoblastoma protein compared with controls. These data led us to conclude that Ano1 regulates proliferation at the G(1)/S transition of the cell cycle and may play a role in tumorigenesis.


Subject(s)
Chloride Channels/physiology , Interstitial Cells of Cajal/cytology , Interstitial Cells of Cajal/physiology , Neoplasm Proteins/physiology , Pancreatic Neoplasms/pathology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Animals, Newborn , Anoctamin-1 , Antineoplastic Agents, Hormonal/pharmacology , Cell Division/physiology , Cell Line, Tumor , Chloride Channels/antagonists & inhibitors , Chlorides/metabolism , Cyclooxygenase Inhibitors/pharmacology , Female , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/physiopathology , Gastrointestinal Stromal Tumors/pathology , Gastrointestinal Stromal Tumors/physiopathology , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred BALB C , Neoplasm Proteins/antagonists & inhibitors , Niflumic Acid/pharmacology , Pancreatic Neoplasms/physiopathology , Primary Cell Culture , Retinoblastoma Protein/metabolism , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL