Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Am J Respir Cell Mol Biol ; 67(6): 623-631, 2022 12.
Article in English | MEDLINE | ID: mdl-36036918

ABSTRACT

The epithelium lining airspaces of the human lung is maintained by regional stem cells, including basal cells of pseudostratified airways and alveolar type 2 (AT2) pneumocytes of the gas-exchange region. Despite effective techniques for long-term preservation of airway basal cells, procedures for efficient preservation of functional epithelial cell types of the distal gas-exchange region are lacking. Here we detail a method for cryobanking of epithelial cells from either mouse or human lung tissue for preservation of their phenotypic and functional characteristics. Flow cytometric profiling, epithelial organoid-forming efficiency, and single-cell transcriptomic analysis were used to compare cells recovered from cryobanked tissue with those of freshly dissociated tissue. AT2 cells within single-cell suspensions of enzymatically digested cryobanked distal lung tissue retained expression of the pan-epithelial marker CD326 and the AT2 cell surface antigen recognized by monoclonal antibody HT II-280, allowing antibody-mediated enrichment and downstream analysis. Isolated AT2 cells from cryobanked tissue were comparable with those of freshly dissociated tissue both in their single-cell transcriptome and their capacity for in vitro organoid formation in three-dimensional cultures. We conclude that the cryobanking method described herein allows long-term preservation of distal human lung tissue for downstream analysis of lung cell function and molecular phenotype and is ideally suited for the creation of an easily accessible tissue resource for the research community.


Subject(s)
Epithelial Cells , Lung , Humans , Mice , Animals , Cell Differentiation/physiology , Epithelial Cells/metabolism , Alveolar Epithelial Cells/metabolism , Phenotype
2.
Cell Rep ; 35(5): 109055, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33905739

ABSTRACT

Coronavirus disease 2019 (COVID-19) is the latest respiratory pandemic caused by severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2). Although infection initiates in the proximal airways, severe and sometimes fatal symptoms of the disease are caused by infection of the alveolar type 2 (AT2) cells of the distal lung and associated inflammation. In this study, we develop primary human lung epithelial infection models to understand initial responses of proximal and distal lung epithelium to SARS-CoV-2 infection. Differentiated air-liquid interface (ALI) cultures of proximal airway epithelium and alveosphere cultures of distal lung AT2 cells are readily infected by SARS-CoV-2, leading to an epithelial cell-autonomous proinflammatory response with increased expression of interferon signaling genes. Studies to validate the efficacy of selected candidate COVID-19 drugs confirm that remdesivir strongly suppresses viral infection/replication. We provide a relevant platform for study of COVID-19 pathobiology and for rapid drug screening against SARS-CoV-2 and emergent respiratory pathogens.


Subject(s)
Alveolar Epithelial Cells/virology , COVID-19 Drug Treatment , COVID-19/pathology , Lung/virology , SARS-CoV-2/drug effects , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adult , Aged , Alanine/analogs & derivatives , Alanine/pharmacology , Alveolar Epithelial Cells/metabolism , COVID-19/metabolism , COVID-19/virology , Child, Preschool , Drug Discovery/methods , Epithelial Cells/virology , Epithelium/metabolism , Epithelium/virology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Lung/pathology , Male , Middle Aged , Models, Biological , Primary Cell Culture , Respiratory Mucosa/virology , SARS-CoV-2/physiology , Virus Replication/drug effects
3.
Biol Open ; 10(4)2021 04 15.
Article in English | MEDLINE | ID: mdl-33913472

ABSTRACT

Otitis media (OM) is the most common paediatric disease and leads to significant morbidity. Although understanding of underlying disease mechanisms is hampered by complex pathophysiology, it is clear that epithelial abnormalities underpin the disease. The mechanisms underpinning epithelial remodelling in OM remain unclear. We recently described a novel in vitro model of mouse middle ear epithelial cells (mMEECs) that undergoes mucociliary differentiation into the varied epithelial cell populations seen in the middle ear cavity. We now describe genome wide gene expression profiles of mMEECs as they undergo differentiation. We compared the gene expression profiles of original (uncultured) middle ear cells, confluent cultures of undifferentiated cells and cells that had been differentiated for 7 days at an air liquid interface (ALI). >5000 genes were differentially expressed among the three groups of cells. Approximately 4000 genes were differentially expressed between the original cells and day 0 of ALI culture. The original cell population was shown to contain a mix of cell types, including contaminating inflammatory cells that were lost on culture. Approximately 500 genes were upregulated during ALI induced differentiation. These included some secretory genes and some enzymes but most were associated with the process of ciliogenesis. The data suggest that the in vitro model of differentiated murine middle ear epithelium exhibits a transcriptional profile consistent with the mucociliary epithelium seen within the middle ear. Knowledge of the transcriptional landscape of this epithelium will provide a basis for understanding the phenotypic changes seen in murine models of OM.


Subject(s)
Biomarkers , Ear, Middle/cytology , Ear, Middle/metabolism , Epithelium/metabolism , Gene Expression Profiling , Transcriptome , Animals , Cells, Cultured , Computational Biology/methods , Disease Susceptibility , Epithelial Cells , Genome-Wide Association Study , Mice , Molecular Sequence Annotation , Otitis Media/etiology , Otitis Media/metabolism , Otitis Media/pathology
4.
Am J Respir Crit Care Med ; 203(6): 707-717, 2021 03 15.
Article in English | MEDLINE | ID: mdl-32991815

ABSTRACT

Rationale: Idiopathic pulmonary fibrosis (IPF) is an insidious and fatal interstitial lung disease associated with declining pulmonary function. Accelerated aging, loss of epithelial progenitor cell function and/or numbers, and cellular senescence are implicated in the pathogenies of IPF.Objectives: We sought to investigate the role of alveolar type 2 (AT2) cellular senescence in initiation and/or progression of pulmonary fibrosis and therapeutic potential of targeting senescence-related pathways and senescent cells.Methods: Epithelial cells of 9 control donor proximal and distal lung tissues and 11 IPF fibrotic lung tissues were profiled by single-cell RNA sequencing to assesses the contribution of epithelial cells to the senescent cell fraction for IPF. A novel mouse model of conditional AT2 cell senescence was generated to study the role of cellular senescence in pulmonary fibrosis.Measurements and Main Results: We show that AT2 cells isolated from IPF lung tissue exhibit characteristic transcriptomic features of cellular senescence. We used conditional loss of Sin3a in adult mouse AT2 cells to initiate a program of p53-dependent cellular senescence, AT2 cell depletion, and spontaneous, progressive pulmonary fibrosis. We establish that senescence rather than loss of AT2 cells promotes progressive fibrosis and show that either genetic or pharmacologic interventions targeting p53 activation or senescence block fibrogenesis.Conclusions: Senescence of AT2 cells is sufficient to drive progressive pulmonary fibrosis. Early attenuation of senescence-related pathways and elimination of senescent cells are promising therapeutic approaches to prevent pulmonary fibrosis.


Subject(s)
Aging/pathology , Alveolar Epithelial Cells/pathology , Cellular Senescence , Idiopathic Pulmonary Fibrosis/pathology , Aged , Aged, 80 and over , Female , Humans , Male
5.
Cell Stem Cell ; 27(6): 869-875.e4, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33259798

ABSTRACT

Current smoking is associated with increased risk of severe COVID-19, but it is not clear how cigarette smoke (CS) exposure affects SARS-CoV-2 airway cell infection. We directly exposed air-liquid interface (ALI) cultures derived from primary human nonsmoker airway basal stem cells (ABSCs) to short term CS and then infected them with SARS-CoV-2. We found an increase in the number of infected airway cells after CS exposure with a lack of ABSC proliferation. Single-cell profiling of the cultures showed that the normal interferon response was reduced after CS exposure with infection. Treatment of CS-exposed ALI cultures with interferon ß-1 abrogated the viral infection, suggesting one potential mechanism for more severe viral infection. Our data show that acute CS exposure allows for more severe airway epithelial disease from SARS-CoV-2 by reducing the innate immune response and ABSC proliferation and has implications for disease spread and severity in people exposed to CS.


Subject(s)
COVID-19/physiopathology , Respiratory Mucosa/physiopathology , Smoking/adverse effects , Stem Cells/virology , COVID-19/genetics , COVID-19/immunology , COVID-19/therapy , Cells, Cultured , Down-Regulation , Humans , Immunity, Innate , Interferon-beta/therapeutic use , Patient Acuity , Respiratory Mucosa/virology
6.
J Vis Exp ; (161)2020 07 21.
Article in English | MEDLINE | ID: mdl-32773760

ABSTRACT

Epithelial organoid models serve as valuable tools to study the basic biology of an organ system and for disease modeling. When grown as organoids, epithelial progenitor cells can self-renew and generate differentiating progeny that exhibit cellular functions similar to those of their in vivo counterparts. Herein we describe a step-by-step protocol to isolate region-specific progenitors from human lung and generate 3D organoid cultures as an experimental and validation tool. We define proximal and distal regions of the lung with the goal of isolating region-specific progenitor cells. We utilized a combination of enzymatic and mechanical dissociation to isolate total cells from the lung and trachea. Specific progenitor cells were then fractionated from the proximal or distal origin cells using fluorescence associated cell sorting (FACS) based on cell type-specific surface markers, such as NGFR for sorting basal cells and HTII-280 for sorting alveolar type II cells. Isolated basal or alveolar type II progenitors were used to generate 3D organoid cultures. Both distal and proximal progenitors formed organoids with a colony forming efficiency of 9-13% in distal region and 7-10% in proximal region when plated 5000 cell/well on day 30. Distal organoids maintained HTII-280+ alveolar type II cells in culture whereas proximal organoids differentiated into ciliated and secretory cells by day 30. These 3D organoid cultures can be used as an experimental tool for studying the cell biology of lung epithelium and epithelial mesenchymal interactions, as well as for the development and validation of therapeutic strategies targeting epithelial dysfunction in a disease.


Subject(s)
Cell Culture Techniques , Cell Separation/methods , Epithelial Cells/cytology , Lung/cytology , Organoids/cytology , Stem Cells/cytology , Cell Differentiation , Cell Fractionation , Humans , Organoids/metabolism , Staining and Labeling
7.
Am J Respir Crit Care Med ; 202(11): 1540-1550, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32692579

ABSTRACT

Rationale: Declining lung function in patients with interstitial lung disease is accompanied by epithelial remodeling and progressive scarring of the gas-exchange region. There is a need to better understand the contribution of basal cell hyperplasia and associated mucosecretory dysfunction to the development of idiopathic pulmonary fibrosis (IPF).Objectives: We sought to decipher the transcriptome of freshly isolated epithelial cells from normal and IPF lungs to discern disease-dependent changes within basal stem cells.Methods: Single-cell RNA sequencing was used to map epithelial cell types of the normal and IPF human airways. Organoid and air-liquid interface cultures were used to investigate functional properties of basal cell subtypes.Measurements and Main Results: We found that basal cells included multipotent and secretory primed subsets in control adult lung tissue. Secretory primed basal cells include an overlapping molecular signature with basal cells obtained from the distal lung tissue of IPF lungs. We confirmed that NOTCH2 maintains undifferentiated basal cells and restricts basal-to-ciliated differentiation, and we present evidence that NOTCH3 functions to restrain secretory differentiation.Conclusions: Basal cells are dynamically regulated in disease and are specifically biased toward the expansion of the secretory primed basal cell subset in IPF. Modulation of basal cell plasticity may represent a relevant target for therapeutic intervention in IPF.


Subject(s)
Cell Plasticity , Cell Proliferation/genetics , Cell Self Renewal/genetics , Epithelial Cells/cytology , Idiopathic Pulmonary Fibrosis/genetics , Respiratory Mucosa/cytology , Aged , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , Basement Membrane , Case-Control Studies , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Male , Middle Aged , RNA-Seq , Respiratory Mucosa/metabolism , Single-Cell Analysis , Transcriptome , Young Adult
8.
Int J Pediatr Otorhinolaryngol ; 130 Suppl 1: 109833, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31901291

ABSTRACT

OBJECTIVE: To summarize recently published key articles on the topics of biomedical engineering, biotechnology and new models in relation to otitis media (OM). DATA SOURCES: Electronic databases: PubMed, Ovid Medline, Cochrane Library and Clinical Evidence (BMJ Publishing). REVIEW METHODS: Articles on biomedical engineering, biotechnology, material science, mechanical and animal models in OM published between May 2015 and May 2019 were identified and subjected to review. A total of 132 articles were ultimately included. RESULTS: New imaging technologies for the tympanic membrane (TM) and the middle ear cavity are being developed to assess TM thickness, identify biofilms and differentiate types of middle ear effusions. Artificial intelligence (AI) has been applied to train software programs to diagnose OM with a high degree of certainty. Genetically modified mice models for OM have further investigated what predisposes some individuals to OM and consequent hearing loss. New vaccine candidates protecting against major otopathogens are being explored and developed, especially combined vaccines, targeting more than one pathogen. Transcutaneous vaccination against non-typeable Haemophilus influenzae has been successfully tried in a chinchilla model. In terms of treatment, novel technologies for trans-tympanic drug delivery are entering the clinical domain. Various growth factors and grafting materials aimed at improving healing of TM perforations show promising results in animal models. CONCLUSION: New technologies and AI applications to improve the diagnosis of OM have shown promise in pre-clinical models and are gradually entering the clinical domain. So are novel vaccines and drug delivery approaches that may allow local treatment of OM. IMPLICATIONS FOR PRACTICE: New diagnostic methods, potential vaccine candidates and the novel trans-tympanic drug delivery show promising results, but are not yet adapted to clinical use.


Subject(s)
Haemophilus Infections/prevention & control , Otitis Media/diagnosis , Otitis Media/therapy , Tympanic Membrane/diagnostic imaging , Animals , Artificial Intelligence , Biofilms , Biomedical Engineering , Biotechnology , Disease Models, Animal , Ear, Middle/diagnostic imaging , Haemophilus Vaccines , Haemophilus influenzae , Humans , Otitis Media/prevention & control , Otitis Media with Effusion/diagnostic imaging , Tympanic Membrane/surgery
9.
Methods Mol Biol ; 1940: 157-168, 2019.
Article in English | MEDLINE | ID: mdl-30788824

ABSTRACT

Epithelial abnormalities underpin the development of the middle ear disease, otitis media (OM). Until now, a well-characterized in vitro model of the middle ear (ME) epithelium that replicates the complex cellular composition of the middle ear has not been available. This chapter describes the development of a novel in vitro model of mouse middle ear epithelial cells (mMECs), cultured at the air-liquid interface (ALI). This system enables recapitulation of the characteristics of the native murine ME epithelium. We demonstrate that mMECs undergo differentiation into the varied cell populations seen within the native middle ear. Overall, our mMEC culture system can help better understand the cell biology of the middle ear and improve our understanding of the pathophysiology of OM. The model also has the potential to serve as a platform for validation of treatments designed to reverse aspects of epithelial remodeling underpinning OM development.


Subject(s)
Cell Culture Techniques/methods , Ear, Middle/cytology , Epithelial Cells/cytology , Epithelium/growth & development , Otitis Media/pathology , Animals , Cells, Cultured , Culture Media/chemistry , Ear, Middle/surgery , Mice , Mice, Inbred C57BL
10.
Sci Rep ; 8(1): 3128, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449589

ABSTRACT

Otitis Media (OM) is characterized by epithelial abnormalities and defects in innate immunity in the middle ear (ME). Although, BPIFA1, a member of the BPI fold containing family of putative innate defence proteins is abundantly expressed by the ME epithelium and SNPs in Bpifa1 have been associated with OM susceptibility, its role in the ME is not well characterized. We investigated the role of BPIFA1 in protection of the ME and the development of OM using murine models. Loss of Bpifa1 did not lead to OM development. However, deletion of Bpifa1 in Evi1Jbo/+ mice, a model of chronic OM, caused significant exacerbation of OM severity, thickening of the ME mucosa and increased collagen deposition, without a significant increase in pro-inflammatory gene expression. Our data suggests that BPIFA1 is involved in maintaining homeostasis within the ME under steady state conditions and its loss in the presence of inflammation, exacerbates epithelial remodelling leading to more severe OM.


Subject(s)
Glycoproteins/metabolism , Otitis Media/metabolism , Phosphoproteins/metabolism , Animals , Disease Models, Animal , Ear, Middle/metabolism , Ear, Middle/physiology , Epithelium/metabolism , Female , Gene Expression , Genes, Regulator , Glycoproteins/genetics , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Otitis Media/genetics , Phosphoproteins/genetics , Proteostasis
11.
Dis Model Mech ; 9(11): 1405-1417, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27660200

ABSTRACT

Otitis media (OM), or middle ear inflammation, is the most common paediatric disease and leads to significant morbidity. Although understanding of underlying disease mechanisms is hampered by complex pathophysiology it is clear that epithelial abnormalities underpin the disease. There is currently a lack of a well-characterised in vitro model of the middle ear (ME) epithelium that replicates the complex cellular composition of the middle ear. Here, we report the development of a novel in vitro model of mouse middle ear epithelial cells (mMECs) at an air-liquid interface (ALI) that recapitulates the characteristics of the native murine ME epithelium. We demonstrate that mMECs undergo differentiation into the varied cell populations seen within the native middle ear. Proteomic analysis confirmed that the cultures secrete a multitude of innate defence proteins from their apical surface. We showed that the mMECs supported the growth of the otopathogen, nontypeable Haemophilus influenzae (NTHi), suggesting that the model can be successfully utilised to study host-pathogen interactions in the middle ear. Overall, our mMEC culture system can help to better understand the cell biology of the middle ear and improve our understanding of the pathophysiology of OM. The model also has the potential to serve as a platform for validation of treatments designed to reverse aspects of epithelial remodelling that underpin OM development.


Subject(s)
Ear, Middle/anatomy & histology , Epithelium/anatomy & histology , Animals , Biomarkers/metabolism , Cell Culture Techniques , Cell Shape , Cells, Cultured , Cilia/metabolism , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Epithelium/metabolism , Epithelium/ultrastructure , Haemophilus Infections/microbiology , Haemophilus Infections/pathology , Haemophilus influenzae/physiology , Mass Spectrometry , Mice, Inbred C57BL , Models, Animal , Proteome/metabolism
12.
Cell Tissue Res ; 350(3): 455-64, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22986921

ABSTRACT

Despite being initially identified in mice, little is known about the sites of production of members of the BPI fold (BPIF) containing (PLUNC) family of putative innate defence proteins in this species. These proteins have largely been considered to be specificaly expressed in the respiratory tract, and we have recently shown that they exhibit differential expression in the epithelium of the proximal airways. In this study, we have used species-specific antibodies to systematically localize two members of this protein family; BPIFA1 (PLUNC/SPLUNC1) and BPIFB1 (LPLUNC1) in adult mice. In general, these proteins exhibit distinct and only partially overlapping localization. BPIFA1 is highly expressed in the respiratory epithelium and Bowman's glands of the nasal passages, whereas BPIFB1 is present in small subset of goblet cells in the nasal passage and pharynx. BPIFB1 is also present in the serous glands in the proximal tongue where is co-localised with the salivary gland specific family member, BPIFA2E (parotid secretory protein) and also in glands of the soft palate. Both proteins exhibit limited expression outside of these regions. These results are consistent with the localization of the proteins seen in man. Knowledge of the complex expression patterns of BPIF proteins in these regions will allow the use of tractable mouse models of disease to dissect their function.


Subject(s)
Carrier Proteins/metabolism , Glycoproteins/metabolism , Mouth/cytology , Mouth/metabolism , Nasal Cavity/cytology , Nasal Cavity/metabolism , Phosphoproteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Cell Differentiation/physiology , Glycoproteins/genetics , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phosphoproteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...