Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Nature ; 629(8013): 869-877, 2024 May.
Article in English | MEDLINE | ID: mdl-38693267

ABSTRACT

Airway hillocks are stratified epithelial structures of unknown function1. Hillocks persist for months and have a unique population of basal stem cells that express genes associated with barrier function and cell adhesion. Hillock basal stem cells continually replenish overlying squamous barrier cells. They exhibit dramatically higher turnover than the abundant, largely quiescent classic pseudostratified airway epithelium. Hillocks resist a remarkably broad spectrum of injuries, including toxins, infection, acid and physical injury because hillock squamous cells shield underlying hillock basal stem cells from injury. Hillock basal stem cells are capable of massive clonal expansion that is sufficient to resurface denuded airway, and eventually regenerate normal airway epithelium with each of its six component cell types. Hillock basal stem cells preferentially stratify and keratinize in the setting of retinoic acid signalling inhibition, a known cause of squamous metaplasia2,3. Here we show that mouse hillock expansion is the cause of vitamin A deficiency-induced squamous metaplasia. Finally, we identify human hillocks whose basal stem cells generate functional squamous barrier structures in culture. The existence of hillocks reframes our understanding of airway epithelial regeneration. Furthermore, we show that hillocks are one origin of 'squamous metaplasia', which is long thought to be a precursor of lung cancer.


Subject(s)
Cell Plasticity , Epithelial Cells , Regeneration , Respiratory Mucosa , Stem Cells , Animals , Female , Humans , Male , Mice , Epithelial Cells/cytology , Epithelial Cells/pathology , Metaplasia/etiology , Metaplasia/pathology , Respiratory Mucosa/cytology , Respiratory Mucosa/injuries , Respiratory Mucosa/pathology , Stem Cells/cytology , Tretinoin/metabolism , Tretinoin/pharmacology , Vitamin A/metabolism , Vitamin A/pharmacology , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Mice, Inbred C57BL
2.
Nature ; 560(7718): 319-324, 2018 08.
Article in English | MEDLINE | ID: mdl-30069044

ABSTRACT

The airways of the lung are the primary sites of disease in asthma and cystic fibrosis. Here we study the cellular composition and hierarchy of the mouse tracheal epithelium by single-cell RNA-sequencing (scRNA-seq) and in vivo lineage tracing. We identify a rare cell type, the Foxi1+ pulmonary ionocyte; functional variations in club cells based on their location; a distinct cell type in high turnover squamous epithelial structures that we term 'hillocks'; and disease-relevant subsets of tuft and goblet cells. We developed 'pulse-seq', combining scRNA-seq and lineage tracing, to show that tuft, neuroendocrine and ionocyte cells are continually and directly replenished by basal progenitor cells. Ionocytes are the major source of transcripts of the cystic fibrosis transmembrane conductance regulator in both mouse (Cftr) and human (CFTR). Knockout of Foxi1 in mouse ionocytes causes loss of Cftr expression and disrupts airway fluid and mucus physiology, phenotypes that are characteristic of cystic fibrosis. By associating cell-type-specific expression programs with key disease genes, we establish a new cellular narrative for airways disease.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/genetics , Epithelial Cells/metabolism , Animals , Asthma/genetics , Epithelial Cells/cytology , Female , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Gene Expression Regulation , Goblet Cells/cytology , Goblet Cells/metabolism , Humans , Lung/cytology , Male , Mice , Sequence Analysis, RNA , Single-Cell Analysis , Trachea/cytology
3.
Nature ; 523(7562): 597-601, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26147083

ABSTRACT

Stem cells integrate inputs from multiple sources. Stem cell niches provide signals that promote stem cell maintenance, while differentiated daughter cells are known to provide feedback signals to regulate stem cell replication and differentiation. Recently, stem cells have been shown to regulate themselves using an autocrine mechanism. The existence of a 'stem cell niche' was first postulated by Schofield in 1978 to define local environments necessary for the maintenance of haematopoietic stem cells. Since then, an increasing body of work has focused on defining stem cell niches. Yet little is known about how progenitor cell and differentiated cell numbers and proportions are maintained. In the airway epithelium, basal cells function as stem/progenitor cells that can both self-renew and produce differentiated secretory cells and ciliated cells. Secretory cells also act as transit-amplifying cells that eventually differentiate into post-mitotic ciliated cells . Here we describe a mode of cell regulation in which adult mammalian stem/progenitor cells relay a forward signal to their own progeny. Surprisingly, this forward signal is shown to be necessary for daughter cell maintenance. Using a combination of cell ablation, lineage tracing and signalling pathway modulation, we show that airway basal stem/progenitor cells continuously supply a Notch ligand to their daughter secretory cells. Without these forward signals, the secretory progenitor cell pool fails to be maintained and secretory cells execute a terminal differentiation program and convert into ciliated cells. Thus, a parent stem/progenitor cell can serve as a functional daughter cell niche.


Subject(s)
Stem Cell Niche/physiology , Stem Cells/cytology , Animals , Cell Communication , Cell Differentiation , Cell Division , Cilia/metabolism , Female , Jagged-2 Protein , Male , Membrane Proteins/metabolism , Mice , Receptor, Notch2/metabolism , Signal Transduction , Stem Cells/metabolism , Trachea/cytology
4.
Genes Dev ; 27(2): 197-210, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23322301

ABSTRACT

The NKX2-1 transcription factor, a regulator of normal lung development, is the most significantly amplified gene in human lung adenocarcinoma. To study the transcriptional impact of NKX2-1 amplification, we generated an expression signature associated with NKX2-1 amplification in human lung adenocarcinoma and analyzed DNA-binding sites of NKX2-1 by genome-wide chromatin immunoprecipitation. Integration of these expression and cistromic analyses identified LMO3, itself encoding a transcription regulator, as a candidate direct transcriptional target of NKX2-1. Further cistromic and overexpression analyses indicated that NKX2-1 can cooperate with the forkhead box transcription factor FOXA1 to regulate LMO3 gene expression. RNAi analysis of NKX2-1-amplified cells compared with nonamplified cells demonstrated that LMO3 mediates cell survival downstream from NKX2-1. Our findings provide new insight into the transcriptional regulatory network of NKX2-1 and suggest that LMO3 is a transcriptional signal transducer in NKX2-1-amplified lung adenocarcinomas.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/physiopathology , Gene Expression Regulation, Neoplastic , LIM Domain Proteins/metabolism , Lung Neoplasms/physiopathology , Nuclear Proteins/genetics , Transcription Factors/genetics , Adenocarcinoma of Lung , Cell Line, Tumor , Chromatin/metabolism , Gene Expression Profiling , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Nuclear Proteins/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Thyroid Nuclear Factor 1 , Transcription Factors/metabolism
5.
Development ; 144(5): 755-766, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28246210

ABSTRACT

In contrast to a prior emphasis on the finality of cell fate decisions in developmental systems, cellular plasticity is now emerging as a general theme in the biology of multiple adult organ systems. In the lung, lineage tracing has been used to identify distinct epithelial stem and progenitor cell populations. These cells, together with their differentiated progeny, maintain a stable identity during steady state conditions, but can display remarkable lineage plasticity following injury. This Review summarizes our current understanding of the different cell lineages of the adult mammalian lung and their responses to injury. In the lung, which is constantly exposed to infection and aerosolized toxins, epithelial plasticity might be more of a rule than an exception, and it is likely that different injuries elicit different facultative responses.


Subject(s)
Cell Lineage , Lung/growth & development , Lung/physiology , Regeneration/physiology , Stem Cells/physiology , Animals , Cell Differentiation , Epithelial Cells/physiology , Homeostasis , Humans , Immune System , Lung Diseases/pathology , Mice , Stem Cells/cytology
6.
Am J Respir Cell Mol Biol ; 61(1): 31-41, 2019 07.
Article in English | MEDLINE | ID: mdl-30995076

ABSTRACT

Lung disease accounts for every sixth death globally. Profiling the molecular state of all lung cell types in health and disease is currently revolutionizing the identification of disease mechanisms and will aid the design of novel diagnostic and personalized therapeutic regimens. Recent progress in high-throughput techniques for single-cell genomic and transcriptomic analyses has opened up new possibilities to study individual cells within a tissue, classify these into cell types, and characterize variations in their molecular profiles as a function of genetics, environment, cell-cell interactions, developmental processes, aging, or disease. Integration of these cell state definitions with spatial information allows the in-depth molecular description of cellular neighborhoods and tissue microenvironments, including the tissue resident structural and immune cells, the tissue matrix, and the microbiome. The Human Cell Atlas consortium aims to characterize all cells in the healthy human body and has prioritized lung tissue as one of the flagship projects. Here, we present the rationale, the approach, and the expected impact of a Human Lung Cell Atlas.


Subject(s)
Lung Diseases/pathology , Lung/pathology , Humans , Lung/metabolism , Transcriptome/genetics
7.
Am J Pathol ; 188(5): 1149-1160, 2018 05.
Article in English | MEDLINE | ID: mdl-29476724

ABSTRACT

Improved tools have led to a burgeoning understanding of lung regeneration in mice, but it is not yet known how these insights may be relevant to acute lung injury in humans. We report in detail two cases of fulminant idiopathic acute lung injury requiring extracorporeal membrane oxygenation in previously healthy young adults with acute respiratory distress syndrome, one of whom required lung transplantation. Biopsy specimens showed diffuse alveolar injury with a striking paucity of alveolar epithelial regeneration, rare hyaline membranes, and diffuse contiguous airspace lining by macrophages. This novel constellation was termed diffuse alveolar injury with delayed epithelization. In addition, mirroring data from murine models of lung injury/regeneration, peribronchiolar basaloid pods (previously described as squamous metaplasia) and ciliated bronchiolarization were identified in these patients and in 39% of 57 historical cases with diffuse alveolar damage. These findings demonstrate a common and clinically relevant human disease correlate for murine models of severe acute lung injury. Evidence suggests that peribronchiolar basaloid pods and bronchiolarization are related spatially and temporally and likely represent overlapping sequential stages of the response to severe distal airway injury.


Subject(s)
Acute Lung Injury/pathology , Extracorporeal Membrane Oxygenation , Lung Transplantation , Pulmonary Fibrosis/pathology , Regeneration/physiology , Acute Lung Injury/surgery , Acute Lung Injury/therapy , Adult , Female , Humans , Male , Treatment Outcome
8.
J Immunol ; 199(8): 2873-2884, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28887431

ABSTRACT

Eicosanoids are a group of bioactive lipids that are shown to be important mediators of neutrophilic inflammation; selective targeting of their function confers therapeutic benefit in a number of diseases. Neutrophilic airway diseases, including cystic fibrosis, are characterized by excessive neutrophil infiltration into the airspace. Understanding the role of eicosanoids in this process may reveal novel therapeutic targets. The eicosanoid hepoxilin A3 is a pathogen-elicited epithelial-produced neutrophil chemoattractant that directs transepithelial migration in response to infection. Following hepoxilin A3-driven transepithelial migration, neutrophil chemotaxis is amplified through neutrophil production of a second eicosanoid, leukotriene B4 (LTB4). The rate-limiting step of eicosanoid generation is the liberation of arachidonic acid by phospholipase A2, and the cytosolic phospholipase A2 (cPLA2)α isoform has been specifically shown to direct LTB4 synthesis in certain contexts. Whether cPLA2α is directly responsible for neutrophil synthesis of LTB4 in the context of Pseudomonas aeruginosa-induced neutrophil transepithelial migration has not been explored. Human and mouse neutrophil-epithelial cocultures were used to evaluate the role of neutrophil-derived cPLA2α in infection-induced transepithelial signaling by pharmacological and genetic approaches. Primary human airway basal stem cell-derived epithelial cultures and micro-optical coherence tomography, a new imaging modality that captures two- and three-dimensional real-time dynamics of neutrophil transepithelial migration, were applied. Evidence from these studies suggests that cPLA2α expressed by neutrophils, but not epithelial cells, plays a significant role in infection-induced neutrophil transepithelial migration by mediating LTB4 synthesis during migration, which serves to amplify the magnitude of neutrophil recruitment in response to epithelial infection.


Subject(s)
Antigens, Human Platelet/metabolism , Cystic Fibrosis/immunology , Neutrophils/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Respiratory Mucosa/immunology , Transendothelial and Transepithelial Migration , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/metabolism , Animals , Cell Communication , Cell Line , Chemotaxis , Coculture Techniques , Cytosol/metabolism , Humans , Leukotriene B4/metabolism , Mice , Neutrophils/microbiology , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , Tomography, Optical Coherence
9.
Nature ; 503(7475): 218-23, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24196716

ABSTRACT

Cellular plasticity contributes to the regenerative capacity of plants, invertebrates, teleost fishes and amphibians. In vertebrates, differentiated cells are known to revert into replicating progenitors, but these cells do not persist as stable stem cells. Here we present evidence that differentiated airway epithelial cells can revert into stable and functional stem cells in vivo. After the ablation of airway stem cells, we observed a surprising increase in the proliferation of committed secretory cells. Subsequent lineage tracing demonstrated that the luminal secretory cells had dedifferentiated into basal stem cells. Dedifferentiated cells were morphologically indistinguishable from stem cells and they functioned as well as their endogenous counterparts in repairing epithelial injury. Single secretory cells clonally dedifferentiated into multipotent stem cells when they were cultured ex vivo without basal stem cells. By contrast, direct contact with a single basal stem cell was sufficient to prevent secretory cell dedifferentiation. In analogy to classical descriptions of amphibian nuclear reprogramming, the propensity of committed cells to dedifferentiate is inversely correlated to their state of maturity. This capacity of committed cells to dedifferentiate into stem cells may have a more general role in the regeneration of many tissues and in multiple disease states, notably cancer.


Subject(s)
Cell Dedifferentiation , Epithelial Cells/cytology , Stem Cells/cytology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Proliferation/drug effects , Cell Survival , Cells, Cultured , Doxycycline/pharmacology , Epithelial Cells/drug effects , Female , Male , Mice, Transgenic , Stem Cells/drug effects , Tamoxifen/pharmacology
10.
J Infect Dis ; 218(suppl_1): S44-S48, 2018 08 14.
Article in English | MEDLINE | ID: mdl-29878132

ABSTRACT

Residency training is a profound experience that greatly influences the career trajectory of every trainee. Currently, residency programs focus heavily (or almost exclusively) on the acquisition of medical knowledge and fail to foster intellectual curiosity and introduce residents to careers in investigation. We share 3 programs embedded in residency training where this focus is shifted with an emphasis on prompting intellectual curiosity and exciting residents about careers in investigation to revitalize the physician-scientist workforce.


Subject(s)
Internship and Residency , Physicians , Research Personnel , Career Choice , Health Workforce , Humans
12.
Am J Respir Cell Mol Biol ; 59(6): 684-694, 2018 12.
Article in English | MEDLINE | ID: mdl-29958012

ABSTRACT

The airway epithelial cell (AEC) response to allergens helps initiate and propagate allergic inflammation in asthma. CARMA3 is a scaffold protein that mediates G protein-coupled receptor-induced NF-κB activation in airway epithelium. In this study, we demonstrate that mice with CARMA3-deficient AECs have reduced airway inflammation, as well as reduced type 2 cytokine levels in response to Alternaria alternata. These mice also have reduced production of IL-33 and IL-25, and reduced numbers of innate lymphoid cells in the lung. We also show that CARMA3-deficient human AECs have decreased production of proasthmatic mediators in response to A. alternata. Finally, we show that CARMA3 interacts with inositol 1,4,5-trisphosphate receptors in AECs, and that inhibition of CARMA3 signaling reduces A. alternata-induced intracellular calcium release. In conclusion, we show that CARMA3 signaling in AECs helps mediate A. alternata-induced allergic airway inflammation, and that CARMA3 is an important signaling molecule for type 2 immune responses in the lung.


Subject(s)
Allergens/immunology , Alternaria/physiology , Alternariosis/immunology , Asthma/immunology , CARD Signaling Adaptor Proteins/metabolism , Pneumonia/immunology , Allergens/metabolism , Alternariosis/metabolism , Alternariosis/microbiology , Animals , Asthma/metabolism , Asthma/microbiology , Cells, Cultured , Disease Models, Animal , Humans , Mice , Pneumonia/metabolism , Pneumonia/microbiology
14.
J Immunol ; 195(2): 683-94, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26041536

ABSTRACT

Innate immune responses to allergens by airway epithelial cells (AECs) help initiate and propagate the adaptive immune response associated with allergic airway inflammation in asthma. Activation of the transcription factor NF-κB in AECs by allergens or secondary mediators via G protein-coupled receptors (GPCRs) is an important component of this multifaceted inflammatory cascade. Members of the caspase recruitment domain family of proteins display tissue-specific expression and help mediate NF-κB activity in response to numerous stimuli. We have previously shown that caspase recruitment domain-containing membrane-associated guanylate kinase protein (CARMA)3 is specifically expressed in AECs and mediates NF-κB activation in these cells in response to stimulation with the GPCR agonist lysophosphatidic acid. In this study, we demonstrate that reduced levels of CARMA3 in normal human bronchial epithelial cells decreases the production of proasthmatic mediators in response to a panel of asthma-relevant GPCR ligands such as lysophosphatidic acid, adenosine triphosphate, and allergens that activate GPCRs such as Alternaria alternata and house dust mite. We then show that genetically modified mice with CARMA3-deficient AECs have reduced airway eosinophilia and proinflammatory cytokine production in a murine model of allergic airway inflammation. Additionally, we demonstrate that these mice have impaired dendritic cell maturation in the lung and that dendritic cells from mice with CARMA3-deficient AECs have impaired Ag processing. In conclusion, we show that AEC CARMA3 helps mediate allergic airway inflammation, and that CARMA3 is a critical signaling molecule bridging the innate and adaptive immune responses in the lung.


Subject(s)
Asthma/immunology , CARD Signaling Adaptor Proteins/immunology , Dendritic Cells/immunology , Epithelial Cells/immunology , Lung/immunology , Adaptive Immunity , Adenosine Triphosphate/pharmacology , Allergens/immunology , Alternaria/immunology , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , CARD Signaling Adaptor Proteins/deficiency , CARD Signaling Adaptor Proteins/genetics , Cells, Cultured , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Gene Expression Regulation , Immunity, Innate , Lung/drug effects , Lung/pathology , Lysophospholipids/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/immunology , Ovalbumin/immunology , Pyroglyphidae/immunology , Signal Transduction
15.
Nature ; 455(7213): 627-32, 2008 Oct 02.
Article in English | MEDLINE | ID: mdl-18754011

ABSTRACT

One goal of regenerative medicine is to instructively convert adult cells into other cell types for tissue repair and regeneration. Although isolated examples of adult cell reprogramming are known, there is no general understanding of how to turn one cell type into another in a controlled manner. Here, using a strategy of re-expressing key developmental regulators in vivo, we identify a specific combination of three transcription factors (Ngn3 (also known as Neurog3) Pdx1 and Mafa) that reprograms differentiated pancreatic exocrine cells in adult mice into cells that closely resemble beta-cells. The induced beta-cells are indistinguishable from endogenous islet beta-cells in size, shape and ultrastructure. They express genes essential for beta-cell function and can ameliorate hyperglycaemia by remodelling local vasculature and secreting insulin. This study provides an example of cellular reprogramming using defined factors in an adult organ and suggests a general paradigm for directing cell reprogramming without reversion to a pluripotent stem cell state.


Subject(s)
Cell Transdifferentiation , Insulin-Secreting Cells/cytology , Pancreas, Exocrine/cytology , Transcription Factors/metabolism , Aging/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/analysis , Cell Shape , Cell Size , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hyperglycemia/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/ultrastructure , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Neovascularization, Physiologic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreas, Exocrine/embryology , Pancreas, Exocrine/metabolism , Regenerative Medicine/methods , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics
17.
Am J Respir Cell Mol Biol ; 48(3): 364-73, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23239495

ABSTRACT

Mucous cell metaplasia is a hallmark of airway diseases, such as asthma and chronic obstructive pulmonary disease. The majority of human airway epithelium is pseudostratified, but the cell of origin of mucous cells has not been definitively established in this type of airway epithelium. There is evidence that ciliated, club cell (Clara), and basal cells can all give rise to mucus-producing cells in different contexts. Because pseudostratified airway epithelium contains distinct progenitor cells from simple columnar airway epithelium, the lineage relationships of progenitor cells to mucous cells may be different in these two epithelial types. We therefore performed lineage tracing of the ciliated cells of the murine basal cell-containing airway epithelium in conjunction with the ovalbumin (OVA)-induced murine model of allergic lung disease. We genetically labeled ciliated cells with enhanced Yellow Fluorescent Protein (eYFP) before the allergen challenge, and followed the fate of these cells to determine whether they gave rise to newly formed mucous cells. Although ciliated cells increased in number after the OVA challenge, the newly formed mucous cells were not labeled with the eYFP lineage tag. Even small numbers of labeled mucous cells could not be detected, implying that ciliated cells make virtually no contribution to the new goblet cell pool. This demonstrates that, after OVA challenge, new mucous cells do not originate from ciliated cells in a pseudostratified basal cell-containing airway epithelium.


Subject(s)
Epithelial Cells/cytology , Ovalbumin/pharmacology , Respiratory Mucosa/cytology , Stem Cells/cytology , Allergens/immunology , Animals , Asthma/pathology , Cell Proliferation/drug effects , Cells, Cultured , Epithelial Cells/drug effects , Goblet Cells/cytology , Goblet Cells/drug effects , Hyperplasia/pathology , Male , Metaplasia/pathology , Mice , Mice, Inbred C57BL , Respiratory Mucosa/drug effects , Stem Cells/drug effects
18.
Am J Respir Cell Mol Biol ; 49(6): 1048-56, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23848320

ABSTRACT

Tissue-specific transgene expression using tetracycline (tet)-regulated promoter/operator elements has been used to revolutionize our understanding of cellular and molecular processes. However, because most tet-regulated mouse strains use promoters of genes expressed in multiple tissues, to achieve exclusive expression in an organ of interest is often impossible. Indeed, in the extreme case, unwanted transgene expression in other organ systems causes lethality and precludes the study of the transgene in the actual organ of interest. Here, we describe a novel approach to activating tet-inducible transgene expression solely in the airway by administering aerosolized doxycycline. By optimizing the dose and duration of aerosolized doxycycline exposure in mice possessing a ubiquitously expressed Rosa26 promoter-driven reverse tet-controlled transcriptional activator (rtTA) element, we induce transgene expression exclusively in the airways. We detect no changes in the cellular composition or proliferative behavior of airway cells. We used this newly developed method to achieve airway basal stem cell-specific transgene expression using a cytokeratin 5 (also known as keratin 5)-driven rtTA driver line to induce Notch pathway activation. We observed a more robust mucous metaplasia phenotype than in mice receiving doxycycline systemically. In addition, unwanted phenotypes outside of the lung that were evident when doxycycline was received systemically were now absent. Thus, our approach allows for rapid and efficient airway-specific transgene expression. After the careful strain by strain titration of the dose and timing of doxycycline inhalation, a suite of preexisting transgenic mice can now be used to study airway biology specifically in cases where transient transgene expression is sufficient to induce a phenotype.


Subject(s)
Doxycycline/administration & dosage , Respiratory System/drug effects , Respiratory System/metabolism , Transgenes/drug effects , Aerosols , Animals , Gene Expression/drug effects , Keratin-5/genetics , Metaplasia , Mice , Mice, Transgenic , Organ Specificity , Phenotype , Promoter Regions, Genetic , RNA, Untranslated/genetics , Receptors, Notch/metabolism , Respiratory System/pathology , Signal Transduction/drug effects , Tetracycline/pharmacology , Trans-Activators/genetics
19.
bioRxiv ; 2023 Aug 07.
Article in English | MEDLINE | ID: mdl-37609222

ABSTRACT

The airway epithelium is frequently exposed to pathogens and allergens, but the cells that are responsible for sampling these inhaled environmental agents have not been fully defined. Thus, there is a critical void in our understanding of how luminal antigens are delivered to the immune cells that drive the appropriate immune defenses against environmental assaults. In this study, we report the first single cell transcriptomes of airway Microfold (M) cells, whose gut counterparts have long been known for their antigen sampling abilities. Given their very recent discovery in the lower respiratory airways, the mechanisms governing the differentiation and functions of airway M cells are largely unknown. Here, we shed light on the pathways of airway M cell differentiation, establish their lineage, and identify a functional M cell-specific endocytic receptor, the complement receptor 2 (CR2). Lastly, we demonstrate that airway M cells can endocytose Aspergillus fumigatus conidia in a CR2-dependent manner. Collectively, this work lays a foundation for deepening our understanding of lung mucosal immunology and the mechanisms that drive lung immunity and tolerance.

20.
Elife ; 122023 03 30.
Article in English | MEDLINE | ID: mdl-36994985

ABSTRACT

The specific functional properties of a tissue are distributed amongst its component cell types. The various cells act coherently, as an ensemble, in order to execute a physiologic response. Modern approaches for identifying and dissecting novel physiologic mechanisms would benefit from an ability to identify specific cell types in live tissues that could then be imaged in real time. Current techniques require the use of fluorescent genetic reporters that are not only cumbersome, but which only allow the study of three or four cell types at a time. We report a non-invasive imaging modality that capitalizes on the endogenous autofluorescence signatures of the metabolic cofactors NAD(P)H and FAD. By marrying morphological characteristics with autofluorescence signatures, all seven of the airway epithelial cell types can be distinguished simultaneously in mouse tracheal explants in real time. Furthermore, we find that this methodology for direct cell type-specific identification avoids pitfalls associated with the use of ostensibly cell type-specific markers that are, in fact, altered by clinically relevant physiologic stimuli. Finally, we utilize this methodology to interrogate real-time physiology and identify dynamic secretory cell associated antigen passages (SAPs) that form in response to cholinergic stimulus. The identical process has been well documented in the intestine where the dynamic formation of SAPs and goblet cell associated antigen passages (GAPs) enable luminal antigen sampling. Airway secretory cells with SAPs are frequently juxtaposed to antigen presenting cells, suggesting that airway SAPs, like their intestinal counterparts, not only sample antigen but convey their cargo for immune cell processing.


Imaging several cell types, at the same time, within a living tissue is no small endeavor. To do so, scientists usually have to perform genetic manipulations that make certain proteins in each cell type fluorescent and therefore easy to track. However, these approaches are cumbersome, limited, and often not applicable to intact human tissues. A possible alternative would be to make use of autofluorescence ­ the fact that certain molecules in living cells naturally fluoresce when exposed to a particular wavelength of light. For example, this is the case for NAD(P)H and FAD, two small molecules necessary for life's biochemical processes, and whose intracellular levels and locations vary depending on cell type. In response, Shah, Hou et al. developed a new imaging technique that takes advantage of the unique autofluorescence signatures of NAD(P)H and FAD to distinguish between the seven different types of cells that line the surface of the airways of mice. Using their autofluorescence approach, Shah, Hou et al. were also able to discover a new role for secretory cells, which normally produce fluids, mucus and various proteins necessary for the lungs to work properly. The imaging experiments show that these cells could also sample material from the surface of the airway in a manner similar to how cells in the intestine take up material from the gut and pass their cargo to immune cells that mediate infection control or tolerance. Further studies should uncover more details about this new function of secretory lung cells. Other exciting discoveries may also emerge from researchers adopting the method developed by Shah, Hou et al. to examine a range of organs (both healthy and diseased), and attempting to apply it to human tissues.


Subject(s)
Antigens , Goblet Cells , Mice , Animals , Antigen-Presenting Cells , Phagocytosis , Optical Imaging
SELECTION OF CITATIONS
SEARCH DETAIL