Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
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.
Development ; 149(18)2022 09 15.
Article in English | MEDLINE | ID: mdl-36134690

ABSTRACT

Heart regeneration requires multiple cell types to enable cardiomyocyte (CM) proliferation. How these cells interact to create growth niches is unclear. Here, we profile proliferation kinetics of cardiac endothelial cells (CECs) and CMs in the neonatal mouse heart and find that they are spatiotemporally coupled. We show that coupled myovascular expansion during cardiac growth or regeneration is dependent upon VEGF-VEGFR2 signaling, as genetic deletion of Vegfr2 from CECs or inhibition of VEGFA abrogates both CEC and CM proliferation. Repair of cryoinjury displays poor spatial coupling of CEC and CM proliferation. Boosting CEC density after cryoinjury with virus encoding Vegfa enhances regeneration. Using Mendelian randomization, we demonstrate that circulating VEGFA levels are positively linked with human myocardial mass, suggesting that Vegfa can stimulate human cardiac growth. Our work demonstrates the importance of coupled CEC and CM expansion and reveals a myovascular niche that may be therapeutically targeted for heart regeneration.


Subject(s)
Endothelial Cells , Vascular Endothelial Growth Factor A , Animals , Cell Proliferation , Endothelial Cells/physiology , Heart/physiology , Humans , Infant, Newborn , Mice , Myocytes, Cardiac/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
3.
EMBO J ; 39(21): e106697, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33001445

ABSTRACT

Stem cell-derived organoid models have emerged as a valuable tool for studying organogenesis, cell-to-cell stromal communication and disease. In this issue, Vazquez-Armendariz et al (2020) report a murine lung stem cell-based bronchioalveolar organoid system and provide insights into the effect of co-culturing with immune and mesenchymal cells.


Subject(s)
Lung , Organoids , Animals , Cell Communication , Mice , Organogenesis , Stem Cells
4.
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
5.
Am J Respir Cell Mol Biol ; 69(3): 255-265, 2023 09.
Article in English | MEDLINE | ID: mdl-37315312

ABSTRACT

Targeted delivery of transgenes to tissue-resident stem cells and related niches offers avenues for interrogating pathways and editing endogenous alleles for therapeutic interventions. Here, we survey multiple adeno-associated virus (AAV) serotypes, administered via intranasal and retroorbital routes in mice, to target lung alveolar stem cell niches. We found that AAV5, AAV4, and AAV8 efficiently and preferentially transduce alveolar type-2 stem cells (AT2s), endothelial cells, and PDGFRA+ fibroblasts, respectively. Notably, some AAVs show different cell tropisms depending on the route of administration. Proof-of-concept experiments reveal the versatility of AAV5-mediated transgenesis for AT2-lineage labeling, clonal cell tracing after cell ablation, and conditional gene inactivation in both postnatal and adult mouse lungs in vivo. AAV6, but not AAV5, efficiently transduces both mouse and human AT2s in alveolar organoid cultures. Furthermore, AAV5 and AAV6 can be used to deliver guide RNAs and transgene cassettes for homologous recombination in vivo and ex vivo, respectively. Using this system coupled with clonal derivation of AT2 organoids, we demonstrate efficient and simultaneous editing of multiple loci, including targeted insertion of a payload cassette in AT2s. Taken together, our studies highlight the powerful utility of AAVs for interrogating alveolar stem cells and other specific cell types both in vivo and ex vivo.


Subject(s)
Dependovirus , Endothelial Cells , Mice , Animals , Humans , Dependovirus/genetics , Transduction, Genetic , Genetic Vectors , Gene Transfer Techniques , Stem Cells
6.
Am J Respir Cell Mol Biol ; 69(6): 623-637, 2023 12.
Article in English | MEDLINE | ID: mdl-37523502

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) of BAL cells has provided insights into coronavirus disease (COVID-19). However, reports have been limited by small patient cohorts. We performed a meta-analysis of BAL scRNA-seq data from healthy control subjects (n = 13) and patients with COVID-19 (n = 20), sourced from six independent studies (167,280 high-quality cells in total). Consistent with the source reports, increases in infiltrating leukocyte subtypes were noted, several with type I IFN signatures and unique gene expression signatures associated with transcellular chemokine signaling. Noting dramatic reductions of inferred NKX2-1 and NR4A1 activity in alveolar epithelial type II (AT-II) cells, we modeled pseudotemporal AT-II-to-AT-I progression. This revealed changes in inferred AT-II cell metabolic activity, increased transitional cells, and a previously undescribed AT-I state. This cell state was conspicuously marked by the induction of genes of the epidermal differentiation complex, including the cornified envelope protein SPRR3 (small proline-rich protein 3), upregulation of multiple KRT (keratin) genes, inferred mitochondrial dysfunction, and cell death signatures including apoptosis and ferroptosis. Immunohistochemistry of lungs from patients with COVID-19 confirmed upregulation and colocalization of KRT13 and SPRR3 in the distal airspaces. Forced overexpression of SPRR3 in human alveolar epithelial cells ex vivo did not activate caspase-3 or upregulate KRT13, suggesting that SPRR3 marks an AT-I cornification program in COVID-19 but is not sufficient for phenotypic changes.


Subject(s)
Alveolar Epithelial Cells , COVID-19 , Humans , COVID-19/genetics , COVID-19/metabolism , Lung , Epithelial Cells/metabolism , Sequence Analysis, RNA
7.
Am J Physiol Lung Cell Mol Physiol ; 325(3): L288-L298, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37366541

ABSTRACT

Pompe disease is an autosomal recessive glycogen storage disease caused by mutations in the gene that encodes acid alpha-glucosidase (GAA)-an enzyme responsible for hydrolyzing lysosomal glycogen. GAA deficiency results in systemic lysosomal glycogen accumulation and cellular disruption. Glycogen accumulation in skeletal muscles, motor neurons, and airway smooth muscle cells is known to contribute to respiratory insufficiency in Pompe disease. However, the impact of GAA deficiency on the distal alveolar type 1 and type 2 cells (AT1 and AT2) has not been evaluated. AT1 cells rely on lysosomes for cellular homeostasis so that they can maintain a thin barrier for gas exchange, whereas AT2 cells depend on lysosome-like structures (lamellar bodies) for surfactant production. Using a mouse model of Pompe disease, the Gaa-/- mouse, we investigated the consequences of GAA deficiency on AT1 and AT2 cells using histology, pulmonary function and mechanics, and transcriptional analysis. Histological analysis revealed increased accumulation of lysosomal-associated membrane protein 1 (LAMP1) in the Gaa-/- mice lungs. Furthermore, ultrastructural examination showed extensive intracytoplasmic vacuoles enlargement and lamellar body engorgement. Respiratory dysfunction was confirmed using whole body plethysmography and forced oscillometry. Finally, transcriptomic analysis demonstrated dysregulation of surfactant proteins in AT2 cells, specifically reduced levels of surfactant protein D in the Gaa-/- mice. We conclude that GAA enzyme deficiency leads to glycogen accumulation in the distal airway cells that disrupts surfactant homeostasis and contributes to respiratory impairments in Pompe disease.NEW & NOTEWORTHY This research highlights the impact of Pompe disease on distal airway cells. Prior to this work, respiratory insufficiency in Pompe disease was classically attributed to pathology in respiratory muscles and motor neurons. Using the Pompe mouse model, we note significant pathology in alveolar type 1 and 2 cells with reductions in surfactant protein D and disrupted surfactant homeostasis. These novel findings highlight the potential contributions of alveolar pathology to respiratory insufficiency in Pompe disease.


Subject(s)
Glycogen Storage Disease Type II , Respiratory Insufficiency , Humans , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , Pulmonary Surfactant-Associated Protein D/metabolism , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism , Muscle, Skeletal/metabolism , Glycogen/metabolism
8.
EMBO Rep ; 22(7): e51921, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34096150

ABSTRACT

Epithelial tissues respond to a wide variety of environmental and genotoxic stresses. As an adaptive mechanism, cells can deviate from their natural paths to acquire new identities, both within and across lineages. Under extreme conditions, epithelial tissues can utilize "shape-shifting" mechanisms whereby they alter their form and function at a tissue-wide scale. Mounting evidence suggests that in order to acquire these alternate tissue identities, cells follow a core set of "tissue logic" principles based on developmental paradigms. Here, we review the terminology and the concepts that have been put forward to describe cell plasticity. We also provide insights into various cell intrinsic and extrinsic factors, including genetic mutations, inflammation, microbiota, and therapeutic agents that contribute to cell plasticity. Additionally, we discuss recent studies that have sought to decode the "syntax" of plasticity-i.e., the cellular and molecular principles through which cells acquire new identities in both homeostatic and malignant epithelial tissues-and how these processes can be manipulated for developing novel cancer therapeutics.


Subject(s)
Cell Plasticity , Neoplasms , Epithelial Cells , Homeostasis , Humans , Inflammation , Neoplasms/genetics
9.
Am J Respir Crit Care Med ; 203(10): 1275-1289, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33321047

ABSTRACT

Rationale: Identification of the specific cell types expressing CFTR (cystic fibrosis [CF] transmembrane conductance regulator) is required for precision medicine therapies for CF. However, a full characterization of CFTR expression in normal human airway epithelia is missing. Objectives: To identify the cell types that contribute to CFTR expression and function within the proximal-distal axis of the normal human lung. Methods: Single-cell RNA (scRNA) sequencing (scRNA-seq) was performed on freshly isolated human large and small airway epithelial cells. scRNA in situ hybridization (ISH) and single-cell qRT-PCR were performed for validation. In vitro culture systems correlated CFTR function with cell types. Lentiviruses were used for cell type-specific transduction of wild-type CFTR in CF cells. Measurements and Main Results: scRNA-seq identified secretory cells as dominating CFTR expression in normal human large and, particularly, small airway superficial epithelia, followed by basal cells. Ionocytes expressed the highest CFTR levels but were rare, whereas the expression in ciliated cells was infrequent and low. scRNA ISH and single-cell qRT-PCR confirmed the scRNA-seq findings. CF lungs exhibited distributions of CFTR and ionocytes similar to those of normal control subjects. CFTR mediated Cl- secretion in cultures tracked secretory cell, but not ionocyte, densities. Furthermore, the nucleotide-purinergic regulatory system that controls CFTR-mediated hydration was associated with secretory cells and not with ionocytes. Lentiviral transduction of wild-type CFTR produced CFTR-mediated Cl- secretion in CF airway secretory cells but not in ciliated cells. Conclusions: Secretory cells dominate CFTR expression and function in human airway superficial epithelia. CFTR therapies may need to restore CFTR function to multiple cell types, with a focus on secretory cells.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , Epithelial Cells/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Case-Control Studies , Cell Culture Techniques , Humans
10.
Proc Natl Acad Sci U S A ; 116(5): 1603-1612, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30655340

ABSTRACT

Current therapeutic interventions for the treatment of respiratory infections are hampered by the evolution of multidrug resistance in pathogens as well as the lack of effective cellular targets. Despite the identification of multiple region-specific lung progenitor cells, the identity of molecules that might be therapeutically targeted in response to infections to promote activation of progenitor cell types remains elusive. Here, we report that loss of Abl1 specifically in SCGB1A1-expressing cells leads to a significant increase in the proliferation and differentiation of bronchiolar epithelial cells, resulting in dramatic expansion of an SCGB1A1+ airway cell population that coexpresses SPC, a marker for type II alveolar cells that promotes alveolar regeneration following bacterial pneumonia. Furthermore, treatment with an Abl-specific allosteric inhibitor enhanced regeneration of the alveolar epithelium and promoted accelerated recovery of mice following pneumonia. These data reveal a potential actionable target that may be exploited for efficient recovery after pathogen-induced infections.


Subject(s)
Lung/metabolism , Lung/physiopathology , Pneumonia, Bacterial/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Regeneration/physiology , Stem Cells/metabolism , Uteroglobin/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/physiology , Animals , Bronchioles/metabolism , Bronchioles/physiopathology , Cell Differentiation/physiology , Cell Line , Female , Humans , Male , Mice , Mice, Inbred C57BL , Pneumonia, Bacterial/physiopathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/physiopathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/physiopathology , Stem Cells/physiology
11.
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
12.
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
13.
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
14.
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
15.
bioRxiv ; 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38585928

ABSTRACT

Proteins undergo reversible S -acylation via a thioester linkage in vivo. S -palmitoylation, modification by C16:0 fatty acid, is a common S -acylation that mediates critical protein-membrane and protein-protein interactions. The most widely used S -acylation assays, including acyl-biotin exchange and acyl resin-assisted capture, utilize blocking of free Cys thiols, hydroxylamine-dependent cleavage of the thioester and subsequent labeling of nascent thiol. These assays generally require >500 micrograms of protein input material per sample and numerous reagent removal and washing steps, making them laborious and ill-suited for high throughput and low input applications. To overcome these limitations, we devised Acyl-Trap, a suspension trap-based assay that utilizes a thiol-reactive quartz to enable buffer exchange and hydroxylamine-mediated S -acyl enrichment. We show that the method is compatible with protein-level detection of S -acylated proteins (e.g. H-Ras) as well as S -acyl site identification and quantification using on-trap isobaric labeling and LC-MS/MS from as little as 20 micrograms of protein input. In mouse brain, Acyl-Trap identified 279 reported sites of S -acylation and 1298 previously unreported putative sites. Also described are conditions for long-term hydroxylamine storage, which streamlines the assay. More generally, Acyl-Trap serves as a proof-of-concept for PTM-tailored suspension traps suitable for both traditional protein detection and chemoproteomic workflows.

16.
Dev Cell ; 59(7): 830-840.e4, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38377991

ABSTRACT

Tissue repair requires a highly coordinated cellular response to injury. In the lung, alveolar type 2 cells (AT2s) act as stem cells to replenish both themselves and alveolar type 1 cells (AT1s); however, the complex orchestration of stem cell activity after injury is poorly understood. Here, we establish longitudinal imaging of AT2s in murine intact tissues ex vivo and in vivo in order to track their dynamic behavior over time. We discover that a large fraction of AT2s become motile following injury and provide direct evidence for their migration between alveolar units. High-resolution morphokinetic mapping of AT2s further uncovers the emergence of distinct motile phenotypes. Inhibition of AT2 migration via genetic depletion of ArpC3 leads to impaired regeneration of AT2s and AT1s in vivo. Together, our results establish a requirement for stem cell migration between alveolar units and identify properties of stem cell motility at high cellular resolution.


Subject(s)
Alveolar Epithelial Cells , Lung , Mice , Animals , Lung/physiology , Alveolar Epithelial Cells/metabolism , Stem Cells/metabolism , Cell Movement , Cell Differentiation/physiology
17.
Cell Rep ; 43(4): 114076, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38607917

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 pandemic is characterized by the emergence of novel variants of concern (VOCs) that replace ancestral strains. Here, we dissect the complex selective pressures by evaluating variant fitness and adaptation in human respiratory tissues. We evaluate viral properties and host responses to reconstruct forces behind D614G through Omicron (BA.1) emergence. We observe differential replication in airway epithelia, differences in cellular tropism, and virus-induced cytotoxicity. D614G accumulates the most mutations after infection, supporting zoonosis and adaptation to the human airway. We perform head-to-head competitions and observe the highest fitness for Gamma and Delta. Under these conditions, RNA recombination favors variants encoding the B.1.617.1 lineage 3' end. Based on viral growth kinetics, Alpha, Gamma, and Delta exhibit increased fitness compared to D614G. In contrast, the global success of Omicron likely derives from increased transmission and antigenic variation. Our data provide molecular evidence to support epidemiological observations of VOC emergence.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/physiology , SARS-CoV-2/genetics , COVID-19/virology , COVID-19/transmission , Virus Replication , Mutation/genetics , Respiratory Mucosa/virology , Genetic Fitness , Animals , Epithelial Cells/virology , Chlorocebus aethiops , Adaptation, Physiological/genetics , Vero Cells
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.
J Clin Invest ; 133(22)2023 11 15.
Article in English | MEDLINE | ID: mdl-37966116

ABSTRACT

Over the last decade, several organoid models have evolved to acquire increasing cellular, structural, and functional complexity. Advanced lung organoid platforms derived from various sources, including adult, fetal, and induced pluripotent stem cells, have now been generated, which more closely mimic the cellular architecture found within the airways and alveoli. In this regard, the establishment of novel protocols with optimized stem cell isolation and culture conditions has given rise to an array of models able to study key cellular and molecular players involved in lung injury and repair. In addition, introduction of other nonepithelial cellular components, such as immune, mesenchymal, and endothelial cells, and employment of novel precision gene editing tools have further broadened the range of applications for these systems by providing a microenvironment and/or phenotype closer to the desired in vivo scenario. Thus, these developments in organoid technology have enhanced our ability to model various aspects of lung biology, including pathogenesis of diseases such as chronic obstructive pulmonary disease, pulmonary fibrosis, cystic fibrosis, and infectious disease and host-microbe interactions, in ways that are often difficult to undertake using only in vivo models. In this Review, we summarize the latest developments in lung organoid technology and their applicability for disease modeling and outline their strengths, drawbacks, and potential avenues for future development.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Endothelial Cells , Lung , Organoids/pathology
20.
Cell Stem Cell ; 30(11): 1486-1502.e9, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37922879

ABSTRACT

Organ regeneration requires dynamic cell interactions to reestablish cell numbers and tissue architecture. While we know the identity of progenitor cells that replace lost tissue, the transient states they give rise to and their role in repair remain elusive. Here, using multiple injury models, we find that alveolar fibroblasts acquire distinct states marked by Sfrp1 and Runx1 that influence tissue remodeling and reorganization. Unexpectedly, ablation of alveolar epithelial type-1 (AT1) cells alone is sufficient to induce tissue remodeling and transitional states. Integrated scRNA-seq followed by genetic interrogation reveals RUNX1 is a key driver of fibroblast states. Importantly, the ectopic induction or accumulation of epithelial transitional states induce rapid formation of transient alveolar fibroblasts, leading to organ-wide fibrosis. Conversely, the elimination of epithelial or fibroblast transitional states or RUNX1 loss, leads to tissue simplification resembling emphysema. This work uncovered a key role for transitional states in orchestrating tissue topologies during regeneration.


Subject(s)
Core Binding Factor Alpha 2 Subunit , Lung , Epithelial Cells , Stem Cells , Cell Communication
SELECTION OF CITATIONS
SEARCH DETAIL