51
|
Major J, Crotta S, Llorian M, McCabe TM, Gad HH, Priestnall SL, Hartmann R, Wack A. Type I and III interferons disrupt lung epithelial repair during recovery from viral infection. Science 2020; 369:712-717. [PMID: 32527928 PMCID: PMC7292500 DOI: 10.1126/science.abc2061] [Citation(s) in RCA: 321] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/08/2020] [Indexed: 12/27/2022]
Abstract
Interferons (IFNs) are central to antiviral immunity. Viral recognition elicits IFN production, which in turn triggers the transcription of IFN-stimulated genes (ISGs), which engage in various antiviral functions. Type I IFNs (IFN-α and IFN-β) are widely expressed and can result in immunopathology during viral infections. By contrast, type III IFN (IFN-λ) responses are primarily restricted to mucosal surfaces and are thought to confer antiviral protection without driving damaging proinflammatory responses. Accordingly, IFN-λ has been proposed as a therapeutic in coronavirus disease 2019 (COVID-19) and other such viral respiratory diseases (see the Perspective by Grajales-Reyes and Colonna). Broggi et al. report that COVID-19 patient morbidity correlates with the high expression of type I and III IFNs in the lung. Furthermore, IFN-λ secreted by dendritic cells in the lungs of mice exposed to synthetic viral RNA causes damage to the lung epithelium, which increases susceptibility to lethal bacterial superinfections. Similarly, using a mouse model of influenza infection, Major et al. found that IFN signaling (especially IFN-λ) hampers lung repair by inducing p53 and inhibiting epithelial proliferation and differentiation. Complicating this picture, Hadjadj et al. observed that peripheral blood immune cells from severe and critical COVID-19 patients have diminished type I IFN and enhanced proinflammatory interleukin-6– and tumor necrosis factor-α–fueled responses. This suggests that in contrast to local production, systemic production of IFNs may be beneficial. The results of this trio of studies suggest that the location, timing, and duration of IFN exposure are critical parameters underlying the success or failure of therapeutics for viral respiratory infections. Science, this issue p. 706, p. 712, p. 718; see also p. 626 Excessive cytokine signaling frequently exacerbates lung tissue damage during respiratory viral infection. Type I (IFN-α and IFN-β) and III (IFN-λ) interferons are host-produced antiviral cytokines. Prolonged IFN-α and IFN-β responses can lead to harmful proinflammatory effects, whereas IFN-λ mainly signals in epithelia, thereby inducing localized antiviral immunity. In this work, we show that IFN signaling interferes with lung repair during influenza recovery in mice, with IFN-λ driving these effects most potently. IFN-induced protein p53 directly reduces epithelial proliferation and differentiation, which increases disease severity and susceptibility to bacterial superinfections. Thus, excessive or prolonged IFN production aggravates viral infection by impairing lung epithelial regeneration. Timing and duration are therefore critical parameters of endogenous IFN action and should be considered carefully for IFN therapeutic strategies against viral infections such as influenza and coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Jack Major
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | - Stefania Crotta
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | - Miriam Llorian
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | - Teresa M McCabe
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | - Hans Henrik Gad
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Simon L Priestnall
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, UK.,Experimental Histopathology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Rune Hartmann
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
52
|
Chen X, Zhao C, Zhang C, Li Q, Chen J, Cheng L, Zhou J, Su X, Song Y. Vagal-α7nAChR signaling promotes lung stem cells regeneration via fibroblast growth factor 10 during lung injury repair. Stem Cell Res Ther 2020; 11:230. [PMID: 32522255 PMCID: PMC7288553 DOI: 10.1186/s13287-020-01757-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/19/2020] [Accepted: 06/01/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Proliferation and transdifferentiation of lung stem cells (LSCs) could promote lung injury repair. The distal airways of the lung are innervated by the vagus nerve. Vagal-alpha7 nicotinic acetylcholine receptor (α7nAChR) signaling plays a key role in regulating lung infection and inflammation; however, whether this pathway could regulate LSCs remains unknown. METHODS LSCs (Sca1+CD45-CD31- cells) were isolated and characterized according to a previously published protocol. α7nAChR knockout mice and wild-type littermates were intratracheally challenged with lipopolysaccharide (LPS) to induce lung injury. A cervical vagotomy was performed to study the regulatory effect of the vagus nerve on LSCs-mediated lung repair. α7nAChR agonist or fibroblast growth factor 10 (FGF10) was intratracheally delivered to mice. A single-cell suspension of lung cells was analyzed by flow cytometry. Lung tissues were collected for histology, quantitative real-time polymerase chain reaction (RT-PCR), and immunohistochemistry. RESULTS We found that LSCs maintained multilineage differentiation ability and transdifferentiated into alveolar epithelial type II cells (AEC2) following FGF10 stimulation in vitro. Vagotomy or α7nAChR deficiency reduced lung Ki67+ LSCs expansion and hampered the resolution of LPS-induced lung injury. Vagotomy or α7nAChR deficiency decreased lung FGF10 expression and the number of AEC2. The α7nAChR agonist-GTS-21 reversed the reduction of FGF10 expression in the lungs, as well as the number of Ki67+ cells, LSCs, Ki67+ LSCs, and AEC2 in LPS-challenged vagotomized mice. Supplementation with FGF10 counteracted the loss of Ki67+ LSCs and AEC2 in LPS-challenged α7nAChR knockout mice. CONCLUSIONS The vagus nerve deploys α7nAChR to enhance LSCs proliferation and transdifferentiation and promote lung repair in an FGF10-dependent manner during LPS-induced lung injury.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University and Shanghai Respiratory Research Institute, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Caiqi Zhao
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, People's Republic of China
| | - Cuiping Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University and Shanghai Respiratory Research Institute, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Qingmei Li
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, People's Republic of China
| | - Jie Chen
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, People's Republic of China
| | - Lianping Cheng
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, People's Republic of China
| | - Jian Zhou
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University and Shanghai Respiratory Research Institute, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Xiao Su
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, People's Republic of China.
| | - Yuanlin Song
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University and Shanghai Respiratory Research Institute, 180 Fenglin Road, Shanghai, 200032, People's Republic of China. .,Department of Pulmonary Medicine, Zhongshan Hospital, Qingpu Branch, Fudan University, Shanghai, People's Republic of China. .,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
53
|
Yu F, Jia R, Tang Y, Liu J, Wei B. SARS-CoV-2 infection and stem cells: Interaction and intervention. Stem Cell Res 2020; 46:101859. [PMID: 32570174 PMCID: PMC7263221 DOI: 10.1016/j.scr.2020.101859] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 02/08/2023] Open
Abstract
The emergence of the novel severe acute respiratory coronavirus 2 (SARS-CoV-2) in China and its rapid national and international spread have created a global health emergency. The resemblance with SARS-CoV in spike protein suggests that SARS-CoV-2 employs spike-driven entry into angiotensin-converting enzyme 2 (ACE2)-expressing cells. From a stem cell perspective, this review focuses on the possible involvement of ACE2+ stem/progenitor cells from both the upper and lower respiratory tracts in coronavirus infection. Viral infection-associated acute respiratory distress syndrome and acute lung injury occur because of dysregulation of the immune response. Mesenchymal stem cells appear to be a promising cell therapy given that they favorably modulate the immune response to reduce lung injury. The use of exogenous stem cells may lead to lung repair. Therefore, intervention by transplantation of exogenous stem cells may be required to replace, repair, remodel, and regenerate lung tissue in survivors infected with coronavirus. Ultimately, vaccines, natural killer cells and induced-pluripotent stem cell-derived virus-specific cytotoxic T lymphocytes may offer off-the-shelf therapeutics for preventing coronavirus reemergence.
Collapse
Affiliation(s)
- Fenggang Yu
- Institute of Life Science, Yingfeng Bilogical Group, Jinan, Shandong Province, China; Institute for Advanced Interdisciplinary Researc (iAIR), University of Jinan, Jinan 250022, China.
| | - Rufu Jia
- Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Yongyong Tang
- Yinfeng Dingcheng Bioengineering and Technology Ltd, Beijing, China
| | - Jin Liu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Benjie Wei
- Institute of Life Science, Yingfeng Bilogical Group, Jinan, Shandong Province, China
| |
Collapse
|
54
|
Sun F, Cheng L, Guo H, Sun Y, Ma Y, Wang Y, Feng W, Yuan Q, Dai X. Application of autologous SOX9 + airway basal cells in patients with bronchiectasis. CLINICAL RESPIRATORY JOURNAL 2020; 14:839-848. [PMID: 32436281 DOI: 10.1111/crj.13216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/30/2020] [Accepted: 05/12/2020] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Bronchiectasis is a common condition and a leading cause of respiratory morbidity and mortality. The treatment method for bronchiectasis is mainly symptomatic treatment or surgery; however, this condition is extremely prone to recurrence. OBJECTIVES To preliminarily evaluate the safety and efficacy of applying SOX9+ autologous airway basal cells (BCs) in patients with bronchiectasis. METHODS SOX9+ BCs were isolated from microscale tissue of a grade 3-5 bronchus by bronchoscopic brushing and expanded in vitro for approximately 4 weeks. Subsequently, the autologous SOX9+ BCs were transplanted into the diseased bronchus to treat patients with bronchiectasis. RESULTS The forced expiratory volume in1 second (FEV1)%, forced vital capacity (FVC)%, total lung capacity (TLC)%, residual volume (RV)% and RV/TLC ratio of predicted value in patients with bronchiectasis were improved at 4, 12, 24 and 48 weeks after cell transplantation, although the differences were not statistically significant (P > .05). Chest CT scans showed that the lesions in the pulmonary segment had not progressed at 4, 12 and 24 weeks after transplantation. No patients died during the follow-up. At 4, 12 and 24 weeks after transplantation, routine blood tests, liver function tests, renal function tests and myocardial enzymatic indexes were normal (P > .05). CONCLUSION Transplantation of autologous SOX9+ BCs has positive effects and is safe for patients with bronchiectasis.
Collapse
Affiliation(s)
- Fengjun Sun
- Department of Pharmacy, the First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Lin Cheng
- Department of Pharmacy, the First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Haiqing Guo
- Department of Respiratory Disease, the First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Yufen Sun
- Regend Therapeutics Co. Ltd, Zhejiang, China
| | - Yu Ma
- Regend Therapeutics Co. Ltd, Zhejiang, China
| | - Yu Wang
- Department of Pharmacy, the First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Wei Feng
- Department of Pharmacy, the First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Qian Yuan
- Department of Pharmacy, the First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaotian Dai
- Department of Respiratory Disease, the First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
55
|
LeMessurier KS, Tiwary M, Morin NP, Samarasinghe AE. Respiratory Barrier as a Safeguard and Regulator of Defense Against Influenza A Virus and Streptococcus pneumoniae. Front Immunol 2020; 11:3. [PMID: 32117216 PMCID: PMC7011736 DOI: 10.3389/fimmu.2020.00003] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/03/2020] [Indexed: 12/27/2022] Open
Abstract
The primary function of the respiratory system of gas exchange renders it vulnerable to environmental pathogens that circulate in the air. Physical and cellular barriers of the respiratory tract mucosal surface utilize a variety of strategies to obstruct microbe entry. Physical barrier defenses including the surface fluid replete with antimicrobials, neutralizing immunoglobulins, mucus, and the epithelial cell layer with rapidly beating cilia form a near impenetrable wall that separates the external environment from the internal soft tissue of the host. Resident leukocytes, primarily of the innate immune branch, also maintain airway integrity by constant surveillance and the maintenance of homeostasis through the release of cytokines and growth factors. Unfortunately, pathogens such as influenza virus and Streptococcus pneumoniae require hosts for their replication and dissemination, and prey on the respiratory tract as an ideal environment causing severe damage to the host during their invasion. In this review, we outline the host-pathogen interactions during influenza and post-influenza bacterial pneumonia with a focus on inter- and intra-cellular crosstalk important in pulmonary immune responses.
Collapse
Affiliation(s)
- Kim S LeMessurier
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| | - Meenakshi Tiwary
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| | - Nicholas P Morin
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Critical Care Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Amali E Samarasinghe
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| |
Collapse
|
56
|
Selman M, Pardo A. The leading role of epithelial cells in the pathogenesis of idiopathic pulmonary fibrosis. Cell Signal 2020; 66:109482. [DOI: 10.1016/j.cellsig.2019.109482] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022]
|
57
|
Kathiriya JJ, Brumwell AN, Jackson JR, Tang X, Chapman HA. Distinct Airway Epithelial Stem Cells Hide among Club Cells but Mobilize to Promote Alveolar Regeneration. Cell Stem Cell 2020; 26:346-358.e4. [PMID: 31978363 DOI: 10.1016/j.stem.2019.12.014] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/23/2019] [Accepted: 12/23/2019] [Indexed: 01/01/2023]
Abstract
Lung injury activates specialized adult epithelial progenitors to regenerate the epithelium. Depending on the extent of injury, both remaining alveolar type II cells (AEC2s) and distal airway stem/progenitors mobilize to cover denuded alveoli and restore normal barriers. The major source of airway stem/progenitors other than basal-like cells remains uncertain. Here, we define a distinct subpopulation (∼5%) of club-like lineage-negative epithelial progenitors (LNEPs) marked by high H2-K1 expression critical for alveolar repair. Quiescent H2-K1high cells account for virtually all in vitro regenerative activity of airway lineages. After bleomycin injury, H2-K1 cells expand and differentiate in vivo to alveolar lineages. However, injured H2-K1 cells eventually develop impaired self-renewal with features of senescence, limiting complete repair. Normal H2-K1high cells transplanted into injured lungs differentiate into alveolar cells and rescue lung function. These findings indicate that small subpopulations of specialized stem/progenitors are required for effective lung regeneration and are a potential therapeutic adjunct after major lung injury.
Collapse
Affiliation(s)
- Jaymin J Kathiriya
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, San Francisco, CA 94143, USA
| | - Alexis N Brumwell
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, San Francisco, CA 94143, USA
| | - Julia R Jackson
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, San Francisco, CA 94143, USA
| | - Xiaodan Tang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, San Francisco, CA 94143, USA; Department of Pulmonary Disease, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Harold A Chapman
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, San Francisco, CA 94143, USA.
| |
Collapse
|
58
|
Chao TL, Gu SY, Lin PH, Chou YT, Ling TY, Chang SY. Characterization of Influenza A Virus Infection in Mouse Pulmonary Stem/Progenitor Cells. Front Microbiol 2020; 10:2942. [PMID: 32038512 PMCID: PMC6985155 DOI: 10.3389/fmicb.2019.02942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/06/2019] [Indexed: 12/26/2022] Open
Abstract
The pulmonary stem/progenitor cells, which could be differentiated into downstream cells to repair tissue damage caused by influenza A virus, have also been shown to be the target cells of influenza virus infection. In this study, mouse pulmonary stem/progenitor cells (mPSCs) with capability to differentiate into type I or type II alveolar cells were used as an in vitro cell model to characterize replication and pathogenic effects of influenza viruses in PSCs. First, mPSCs and its immortalized cell line mPSCsOct4+ were shown to be susceptible to PR8, seasonal H1N1, 2009 pandemic H1N1, and H7N9 influenza viruses and can generate infectious virus particles, although with a lower virus titer, which could be attributed by the reduced vRNA replication and nucleoprotein (NP) aggregation in the cytoplasm. Nevertheless, a significant increase of interleukin (IL)-6 and interferon (IFN)-γ at 12 h and IFN-β at 24 h post infection in mPSCs implicates that mPSCs might function as a sensor to modulate immune responses to influenza virus infection. In summary, our results demonstrated mPSCs, as one of the target cells for influenza A viruses, could modulate early proinflammatory responses to influenza virus infection.
Collapse
Affiliation(s)
- Tai-Ling Chao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sing-Yi Gu
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pi-Han Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Tien Chou
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Thai-Yen Ling
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
59
|
Liu L, Song C, Li J, Wang Q, Zhu M, Hu Y, Chen J, Chen C, Zhang JS, Dong N, Chen C. Fibroblast growth factor 10 alleviates particulate matter-induced lung injury by inhibiting the HMGB1-TLR4 pathway. Aging (Albany NY) 2020; 12:1186-1200. [PMID: 31958320 PMCID: PMC7053597 DOI: 10.18632/aging.102676] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/25/2019] [Indexed: 12/12/2022]
Abstract
Exposure to particulate matter (PM) is associated with increased incidence of respiratory diseases. The present study aimed to investigate the roles of fibroblast growth factor 10 (FGF10) in PM-induced lung injury. Mice were intratracheally instilled with FGF10 or phosphate-buffered saline at one hour before instillation of PM for two consecutive days. In addition, the anti-inflammatory impact of FGF10 in vitro and its effect on the high-mobility group box 1 (HMGB1)-toll-like receptor 4 (TLR4) pathway was investigated. It was found that PM exposure is associated with increased inflammatory cell infiltration into the lung and increased vascular protein leakage, while FGF10 pretreatment attenuated both of these effects. FGF10 also decreased the PM-induced expression of interleukin (IL)-6, IL-8, tumor necrosis factor-α and HMGB1 in murine bronchoalveolar lavage fluid and in the supernatants of human bronchial epithelial cells exposed to PM. FGF10 exerted anti-inflammatory and cytoprotective effects by inhibiting the HMGB1-TLR4 pathway. These results indicate that FGF10 may have therapeutic values for PM-induced lung injury.
Collapse
Affiliation(s)
- Lingjing Liu
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Chenjian Song
- Department of Pulmonary Medicine, Yiwu Central Hospital, Yiwu 322000, China
| | - Jingli Li
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Qiang Wang
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Mingyang Zhu
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Yiran Hu
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Junjie Chen
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Chaolei Chen
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Jin-San Zhang
- Department of Pharmacy, Wenzhou Medical University Pharmacy School, Wenzhou 325006, China
| | - Nian Dong
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Chengshui Chen
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| |
Collapse
|
60
|
Li Y, Wu Q, Sun X, Shen J, Chen H. Organoids as a Powerful Model for Respiratory Diseases. Stem Cells Int 2020; 2020:5847876. [PMID: 32256609 PMCID: PMC7086445 DOI: 10.1155/2020/5847876] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/12/2020] [Accepted: 02/27/2020] [Indexed: 02/05/2023] Open
Abstract
Insults to the alveoli usually lead to inefficient gas exchange or even respiratory failure, which is difficult to model in animal studies. Over the past decade, stem cell-derived self-organizing three-dimensional organoids have emerged as a new avenue to recapitulate respiratory diseases in a dish. Alveolar organoids have improved our understanding of the mechanisms underlying tissue homeostasis and pathological alterations in alveoli. From this perspective, we review the state-of-the-art technology on establishing alveolar organoids from endogenous lung epithelial stem/progenitor cells or pluripotent stem cells, as well as the use of alveolar organoids for the study of respiratory diseases, including idiopathic pulmonary fibrosis, tuberculosis infection, and respiratory virus infection. We also discuss challenges that need to be overcome for future application of alveolar organoids in individualized medicine.
Collapse
Affiliation(s)
- Yu Li
- 1Department of Basic Medicine, Tianjin University Haihe Hospital, Tianjin, China
| | - Qi Wu
- 2Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
| | - Xin Sun
- 2Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
| | - Jun Shen
- 1Department of Basic Medicine, Tianjin University Haihe Hospital, Tianjin, China
- 2Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- 3Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Huaiyong Chen
- 1Department of Basic Medicine, Tianjin University Haihe Hospital, Tianjin, China
- 2Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- 3Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| |
Collapse
|
61
|
Chu X, Chen C, Chen C, Zhang JS, Bellusci S, Li X. Evidence for lung repair and regeneration in humans: key stem cells and therapeutic functions of fibroblast growth factors. Front Med 2019; 14:262-272. [PMID: 31741137 PMCID: PMC7095240 DOI: 10.1007/s11684-019-0717-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/05/2019] [Indexed: 01/19/2023]
Abstract
Regeneration carries the idea of regrowing partially or completely a missing organ. Repair, on the other hand, allows restoring the function of an existing but failing organ. The recognition that human lungs can both repair and regenerate is quite novel, the concept has not been widely used to treat patients. We present evidence that the human adult lung does repair and regenerate and introduce different ways to harness this power. Various types of lung stem cells are capable of proliferating and differentiating upon injury driving the repair/regeneration process. Injury models, primarily in mice, combined with lineage tracing studies, have allowed the identification of these important cells. Some of these cells, such as basal cells, broncho-alveolar stem cells, and alveolar type 2 cells, rely on fibroblast growth factor (FGF) signaling for their survival, proliferation and/or differentiation. While preclinical studies have shown the therapeutic benefits of FGFs, a recent clinical trial for acute respiratory distress syndrome (ARDS) using intravenous injection of FGF7 did not report the expected beneficial effects. We discuss the potential reasons for these negative results and propose the rationale for new approaches for future clinical trials, such as delivery of FGFs to the damaged lungs through efficient inhalation systems, which may be more promising than systemic exposure to FGFs. While this change in the administration route presents a challenge, the therapeutic promises displayed by FGFs are worth the effort.
Collapse
Affiliation(s)
- Xuran Chu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Justus-Liebig-University Giessen, 35392, Giessen, Germany
| | - Chengshui Chen
- Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Chaolei Chen
- Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jin-San Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Saverio Bellusci
- Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, China.
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Justus-Liebig-University Giessen, 35392, Giessen, Germany.
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
62
|
Liu L, Xia Z, Li J, Hu Y, Wang Q, Chen J, Fan S, Wu J, Dong N, Chen C. Fibroblast growth factor 10 protects against particulate matter-induced airway inflammatory response through regulating inflammatory signaling and apoptosis. Am J Transl Res 2019; 11:6977-6988. [PMID: 31814901 PMCID: PMC6895507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/25/2019] [Indexed: 06/10/2023]
Abstract
Chronic respiratory disorders are some of the most frequent and severe chronic diseases in China. Epidemiological research has shown that particulate matter (PM) is a risk factor and is closely correlated to the progression of numerous respiratory diseases. Fibroblast growth factor 10 (FGF10) is a mesenchymal-epithelial signaling messenger essential for the development and environmental stability of several tissues. Nevertheless, its role in PM-induced airway inflammation remains unclear. The present study aimed to explore the mechanisms underlying the FGF10-related slowing of lung injury and inflammation in vivo and in vitro, as well as the therapeutic potential of these phenomena. Mice were intraperitoneally injected with a vehicle (PBS) or FGF10 (0.5 mg/kg) at one hour before intratracheal treatment with vehicle (PBS) or PM (4 mg/kg) for two consecutive days. Human airway epithelial BEAS-2B cells were exposed to a vehicle (PBS) or FGF10 (10 ng/ml) in vitro at one hour prior to incubation with a vehicle or PM (200 ug/ml) for 24 hours. Then, the impact on inflammatory molecules was investigated. In vivo, it was found that FGF10 diminished the inflammatory cell aggregation and reduced the apoptosis. Interestingly, in the PM group, the level of FGF10 increased in the bronchoalveolar lavage fluid (BALF). However, the pre-treatment with FGF10 markedly impaired the PM-induced increase in IL-6, IL-8, TNF-α and PGE2 levels in BALF and the cell supernatant. In conclusion, the present findings indicate that FGF10 attenuates PM-induced airway inflammation by inhibiting apoptosis and inflammation. This may be exploited for the prevention and management of PM-induced airway inflammation.
Collapse
Affiliation(s)
- Lingjing Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Ziqiang Xia
- Department of Gastroenterology, First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Jingli Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Yiran Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Qiang Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Junjie Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Shiqian Fan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Jinming Wu
- Department of Gastroenterology, First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Nian Dong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Chengshui Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| |
Collapse
|
63
|
Fiege JK, Stone IA, Dumm RE, Waring BM, Fife BT, Agudo J, Brown BD, Heaton NS, Langlois RA. Long-term surviving influenza infected cells evade CD8+ T cell mediated clearance. PLoS Pathog 2019; 15:e1008077. [PMID: 31557273 PMCID: PMC6782110 DOI: 10.1371/journal.ppat.1008077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 10/08/2019] [Accepted: 09/11/2019] [Indexed: 01/02/2023] Open
Abstract
Influenza A virus (IAV) is a seasonal pathogen with the potential to cause devastating pandemics. IAV infects multiple epithelial cell subsets in the respiratory tract, eliciting damage to the lungs. Clearance of IAV is primarily dependent on CD8+ T cells, which must balance control of the infection with immunopathology. Using a virus expressing Cre recombinase to permanently label infected cells in a Cre-inducible reporter mouse, we previously discovered infected club cells that survive both lytic virus replication and CD8+ T cell-mediated clearance. In this study, we demonstrate that ciliated epithelial cells, type I and type II alveolar cells can also become survivor cells. Survivor cells are stable in the lung long-term and demonstrate enhanced proliferation compared to uninfected cells. When we investigated how survivor cells evade CD8+ T cell killing we observed that survivor cells upregulated the inhibitory ligand PD-L1, but survivor cells did not use PD-L1 to evade CD8+ T cell killing. Instead our data suggest that survivor cells are not inherently resistant to CD8+ T cell killing, but instead no longer present IAV antigen and cannot be detected by CD8+ T cells. Finally, we evaluate the failure of CD8+ T cells to kill these previously infected cells. This work demonstrates that additional cell types can survive IAV infection and that these cells robustly proliferate and are stable long term. By sparing previously infected cells, the adaptive immune system may be minimizing pathology associated with IAV infection.
Collapse
Affiliation(s)
- Jessica K. Fiege
- University of Minnesota, Department of Microbiology and Immunology and the Center for Immunology, Minneapolis, Minnesota, United States of America
| | - Ian A. Stone
- University of Minnesota, Department of Microbiology and Immunology and the Center for Immunology, Minneapolis, Minnesota, United States of America
| | - Rebekah E. Dumm
- Duke University School of Medicine, Department of Molecular Genetics and Microbiology, Durham, North Carolina, United States of America
| | - Barbara M. Waring
- University of Minnesota, Department of Microbiology and Immunology and the Center for Immunology, Minneapolis, Minnesota, United States of America
| | - Brian T. Fife
- University of Minnesota, Department of Medicine and the Center for Immunology, Minneapolis, Minnesota, United States of America
| | - Judith Agudo
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York City, New York, United States of America
| | - Brian D. Brown
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York City, New York, United States of America
| | - Nicholas S. Heaton
- Duke University School of Medicine, Department of Molecular Genetics and Microbiology, Durham, North Carolina, United States of America
| | - Ryan A. Langlois
- University of Minnesota, Department of Microbiology and Immunology and the Center for Immunology, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
64
|
Tighe RM, Redente EF, Yu YR, Herold S, Sperling AI, Curtis JL, Duggan R, Swaminathan S, Nakano H, Zacharias WJ, Janssen WJ, Freeman CM, Brinkman RR, Singer BD, Jakubzick CV, Misharin AV. Improving the Quality and Reproducibility of Flow Cytometry in the Lung. An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2019; 61:150-161. [PMID: 31368812 PMCID: PMC6670040 DOI: 10.1165/rcmb.2019-0191st] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Defining responses of the structural and immune cells in biologic systems is critically important to understanding disease states and responses to injury. This requires accurate and sensitive methods to define cell types in organ systems. The principal method to delineate the cell populations involved in these processes is flow cytometry. Although researchers increasingly use flow cytometry, technical challenges can affect its accuracy and reproducibility, thus significantly limiting scientific advancements. This challenge is particularly critical to lung immunology, as the lung is readily accessible and therefore used in preclinical and clinical studies to define potential therapeutics. Given the importance of flow cytometry in pulmonary research, the American Thoracic Society convened a working group to highlight issues and technical challenges to the performance of high-quality pulmonary flow cytometry, with a goal of improving its quality and reproducibility.
Collapse
|
65
|
Kalil AC, Thomas PG. Influenza virus-related critical illness: pathophysiology and epidemiology. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:258. [PMID: 31324202 PMCID: PMC6642581 DOI: 10.1186/s13054-019-2539-x] [Citation(s) in RCA: 265] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/10/2019] [Indexed: 12/22/2022]
Abstract
Influenza virus affects the respiratory tract by direct viral infection or by damage from the immune system response. In humans, the respiratory epithelium is the only site where the hemagglutinin (HA) molecule is effectively cleaved, generating infectious virus particles. Virus transmission occurs through a susceptible individual's contact with aerosols or respiratory fomites from an infected individual. The inability of the lung to perform its primary function of gas exchange can result from multiple mechanisms, including obstruction of the airways, loss of alveolar structure, loss of lung epithelial integrity from direct epithelial cell killing, and degradation of the critical extracellular matrix.Approximately 30-40% of hospitalized patients with laboratory-confirmed influenza are diagnosed with acute pneumonia. These patients who develop pneumonia are more likely to be < 5 years old, > 65 years old, Caucasian, and nursing home residents; have chronic lung or heart disease and history of smoking, and are immunocompromised.Influenza can primarily cause severe pneumonia, but it can also present in conjunction with or be followed by a secondary bacterial infection, most commonly by Staphylococcus aureus and Streptococcus pneumoniae. Influenza is associated with a high predisposition to bacterial sepsis and ARDS. Viral infections presenting concurrently with bacterial pneumonia are now known to occur with a frequency of 30-50% in both adult and pediatric populations. The H3N2 subtype has been associated with unprecedented high levels of intensive care unit (ICU) admission.Influenza A is the predominant viral etiology of acute respiratory distress syndrome (ARDS) in adults. Risk factors independently associated with ARDS are age between 36 and 55 years old, pregnancy, and obesity, while protective factors are female sex, influenza vaccination, and infections with Influenza A (H3N2) or Influenza B viruses.In the ICU, particularly during the winter season, influenza should be suspected not only in patients with typical symptoms and epidemiology, but also in patients with severe pneumonia, ARDS, sepsis with or without bacterial co-infection, as well as in patients with encephalitis, myocarditis, and rhabdomyolysis.
Collapse
Affiliation(s)
- Andre C Kalil
- Department of Internal Medicine, Division of Infectious Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Paul G Thomas
- Immunology Department, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
66
|
Nikolić MZ, Garrido-Martin EM, Greiffo FR, Fabre A, Heijink IH, Boots A, Greene CM, Hiemstra PS, Bartel S. From the pathophysiology of the human lung alveolus to epigenetic editing: Congress 2018 highlights from ERS Assembly 3 "Basic and Translational Science.". ERJ Open Res 2019; 5:00194-2018. [PMID: 31111040 PMCID: PMC6513036 DOI: 10.1183/23120541.00194-2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/23/2019] [Indexed: 12/16/2022] Open
Abstract
The European Respiratory Society (ERS) International Congress is the largest respiratory congress and brings together leading experts in all fields of respiratory medicine and research. ERS Assembly 3 shapes the basic and translational science aspects of this congress, aiming to combine cutting-edge novel developments in basic research with novel clinical findings. In this article, we summarise a selection of the scientific highlights from the perspective of the three groups within Assembly 3. In particular, we discuss new insights into the pathophysiology of the human alveolus, novel tools in organoid development and (epi)genome editing, as well as insights from the presented abstracts on novel therapeutic targets being identified for idiopathic pulmonary fibrosis. The amount of basic and translational science presented at #ERSCongress is steadily increasing, showing novel cutting-edge technologies and models.http://bit.ly/2GgXIJi
Collapse
Affiliation(s)
- Marko Z Nikolić
- University College London, Division of Medicine, London, UK.,These contributed equally to this work
| | - Eva M Garrido-Martin
- H12O-CNIO Lung Cancer Clinical Research Unit, Research Institute Hospital 12 Octubre - Spanish National Cancer Research Centre (CNIO), and Biomedical Research Networking Centre Consortium of Cancer (CIBERONC), Madrid, Spain.,These contributed equally to this work
| | - Flavia R Greiffo
- Comprehensive Pneumology Center, Ludwig-Maximilians University (LMU), University Hospital Grosshadern, and Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany.,These contributed equally to this work
| | - Aurélie Fabre
- University College Dublin, St Vincent's University Hospital, Elm Park, Dublin, Ireland
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Depts of Pathology and Medical Biology and Pulmonology, GRIAC Research Institute, Groningen, The Netherlands
| | - Agnes Boots
- Dept of Pharmacology and Toxicology, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Catherine M Greene
- Lung Biology Group, Dept of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Pieter S Hiemstra
- Dept of Pulmonology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Sabine Bartel
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| |
Collapse
|
67
|
Salwig I, Spitznagel B, Vazquez-Armendariz AI, Khalooghi K, Guenther S, Herold S, Szibor M, Braun T. Bronchioalveolar stem cells are a main source for regeneration of distal lung epithelia in vivo. EMBO J 2019; 38:embj.2019102099. [PMID: 31028085 DOI: 10.15252/embj.2019102099] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 12/22/2022] Open
Abstract
Bronchioalveolar stem cells (BASCs) are a potential source for lung regeneration, but direct in vivo evidence for a multipotential lineage contribution during homeostasis and disease is critically missing, since specific genetic labeling of BASCs has not been possible. We developed a novel cell tracing approach based on intein-mediated assembly of newly engineered split-effectors, allowing selective targeting of dual-marker expressing BASCs in the mouse lung. RNA sequencing of isolated BASCs demonstrates that BASCs show a distinct transcriptional profile, characterized by co-expression of bronchiolar and alveolar epithelial genes. We found that BASCs generate the majority of distal lung airway cells after bronchiolar damage but only moderately contribute to cellular turnover under homeostatic conditions. Importantly, DTA-mediated ablation of BASCs compromised proper regeneration of distal airways. The study defines BASCs as crucial components of the lung repair machinery and provides a paradigmatic example for the detection and manipulation of stem cells that cannot be recognized by a single marker alone.
Collapse
Affiliation(s)
- Isabelle Salwig
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Birgit Spitznagel
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Ana Ivonne Vazquez-Armendariz
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Keynoosh Khalooghi
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Stefan Guenther
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Susanne Herold
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Marten Szibor
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| |
Collapse
|
68
|
Paolicelli G, Luca AD, Jose SS, Antonini M, Teloni I, Fric J, Zelante T. Using Lung Organoids to Investigate Epithelial Barrier Complexity and IL-17 Signaling During Respiratory Infection. Front Immunol 2019; 10:323. [PMID: 30873173 PMCID: PMC6403157 DOI: 10.3389/fimmu.2019.00323] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/07/2019] [Indexed: 01/23/2023] Open
Affiliation(s)
| | - Antonella De Luca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Shyam S Jose
- Center for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czechia
| | - Martina Antonini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Irene Teloni
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Jan Fric
- Center for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czechia
| | - Teresa Zelante
- Center for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czechia
| |
Collapse
|
69
|
Non-lytic clearance of influenza B virus from infected cells preserves epithelial barrier function. Nat Commun 2019; 10:779. [PMID: 30770807 PMCID: PMC6377627 DOI: 10.1038/s41467-019-08617-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/22/2019] [Indexed: 01/11/2023] Open
Abstract
Influenza B virus (IBV) is an acute, respiratory RNA virus that has been assumed to induce the eventual death of all infected cells. We and others have shown however, that infection with apparently cytopathic viruses does not necessarily lead to cell death; some cells can intrinsically clear the virus and persist in the host long-term. To determine if any cells can survive direct IBV infection, we here generate a recombinant IBV capable of activating a host-cell reporter to permanently label all infected cells. Using this system, we demonstrate that IBV infection leads to the formation of a survivor cell population in the proximal airways that are ciliated-like, but transcriptionally and phenotypically distinct from both actively infected and bystander ciliated cells. We also show that survivor cells are critical to maintain respiratory barrier function. These results highlight a host response pathway that preserves the epithelium to limit the severity of IBV disease. Infection of a cell with influenza B virus (IBV) often results in cell death and the role of surviving cells in pathogenesis is unclear. Here, Dumm et al. generate a recombinant IBV that activates a host-cell reporter to permanently label infected cells, and show that surviving cells are important to preserve epithelial barrier function.
Collapse
|
70
|
Abstract
The acute respiratory distress syndrome (ARDS) is a common cause of respiratory failure in critically ill patients and is defined by the acute onset of noncardiogenic pulmonary oedema, hypoxaemia and the need for mechanical ventilation. ARDS occurs most often in the setting of pneumonia, sepsis, aspiration of gastric contents or severe trauma and is present in ~10% of all patients in intensive care units worldwide. Despite some improvements, mortality remains high at 30-40% in most studies. Pathological specimens from patients with ARDS frequently reveal diffuse alveolar damage, and laboratory studies have demonstrated both alveolar epithelial and lung endothelial injury, resulting in accumulation of protein-rich inflammatory oedematous fluid in the alveolar space. Diagnosis is based on consensus syndromic criteria, with modifications for under-resourced settings and in paediatric patients. Treatment focuses on lung-protective ventilation; no specific pharmacotherapies have been identified. Long-term outcomes of patients with ARDS are increasingly recognized as important research targets, as many patients survive ARDS only to have ongoing functional and/or psychological sequelae. Future directions include efforts to facilitate earlier recognition of ARDS, identifying responsive subsets of patients and ongoing efforts to understand fundamental mechanisms of lung injury to design specific treatments.
Collapse
|
71
|
Modeling Host-Pathogen Interactions in the Context of the Microenvironment: Three-Dimensional Cell Culture Comes of Age. Infect Immun 2018; 86:IAI.00282-18. [PMID: 30181350 DOI: 10.1128/iai.00282-18] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tissues and organs provide the structural and biochemical landscapes upon which microbial pathogens and commensals function to regulate health and disease. While flat two-dimensional (2-D) monolayers composed of a single cell type have provided important insight into understanding host-pathogen interactions and infectious disease mechanisms, these reductionist models lack many essential features present in the native host microenvironment that are known to regulate infection, including three-dimensional (3-D) architecture, multicellular complexity, commensal microbiota, gas exchange and nutrient gradients, and physiologically relevant biomechanical forces (e.g., fluid shear, stretch, compression). A major challenge in tissue engineering for infectious disease research is recreating this dynamic 3-D microenvironment (biological, chemical, and physical/mechanical) to more accurately model the initiation and progression of host-pathogen interactions in the laboratory. Here we review selected 3-D models of human intestinal mucosa, which represent a major portal of entry for infectious pathogens and an important niche for commensal microbiota. We highlight seminal studies that have used these models to interrogate host-pathogen interactions and infectious disease mechanisms, and we present this literature in the appropriate historical context. Models discussed include 3-D organotypic cultures engineered in the rotating wall vessel (RWV) bioreactor, extracellular matrix (ECM)-embedded/organoid models, and organ-on-a-chip (OAC) models. Collectively, these technologies provide a more physiologically relevant and predictive framework for investigating infectious disease mechanisms and antimicrobial therapies at the intersection of the host, microbe, and their local microenvironments.
Collapse
|
72
|
Transcriptome Analysis of Infected and Bystander Type 2 Alveolar Epithelial Cells during Influenza A Virus Infection Reveals In Vivo Wnt Pathway Downregulation. J Virol 2018; 92:JVI.01325-18. [PMID: 30111569 DOI: 10.1128/jvi.01325-18] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 08/12/2018] [Indexed: 12/29/2022] Open
Abstract
Influenza virus outbreaks remain a serious threat to public health. A greater understanding of how cells targeted by the virus respond to the infection can provide insight into the pathogenesis of disease. Here we examined the transcriptional profile of in vivo-infected and uninfected type 2 alveolar epithelial cells (AEC) in the lungs of influenza virus-infected mice. We show for the first time the unique gene expression profiles induced by the in vivo infection of AEC as well as the transcriptional response of uninfected bystander cells. This work allows us to distinguish the direct and indirect effects of infection at the cellular level. Transcriptome analysis revealed that although directly infected and bystander AEC from infected animals shared many transcriptome changes compared to AEC from uninfected animals, directly infected cells produce more interferon and express lower levels of Wnt signaling-associated transcripts, while concurrently expressing more transcripts associated with cell death pathways, than bystander uninfected AEC. The Wnt signaling pathway was downregulated in both in vivo-infected AEC and in vitro-infected human lung epithelial A549 cells. Wnt signaling did not affect type I and III interferon production by infected A549 cells. Our results reveal unique transcriptional changes that occur within infected AEC and show that influenza virus downregulates Wnt signaling. In light of recent findings that Wnt signaling is essential for lung epithelial stem cells, our findings reveal a mechanism by which influenza virus may affect host lung repair.IMPORTANCE Influenza virus infection remains a major public health problem. Utilizing a recombinant green fluorescent protein-expressing influenza virus, we compared the in vivo transcriptomes of directly infected and uninfected bystander cells from infected mouse lungs and discovered many pathways uniquely regulated in each population. The Wnt signaling pathway was downregulated in directly infected cells and was shown to affect virus but not interferon production. Our study is the first to discern the in vivo transcriptome changes induced by direct viral infection compared to mere exposure to the lung inflammatory milieu and highlight the downregulation of Wnt signaling. This downregulation has important implications for understanding influenza virus pathogenesis, as Wnt signaling is critical for lung epithelial stem cells and lung epithelial cell differentiation. Our findings reveal a mechanism by which influenza virus may affect host lung repair and suggest interventions that prevent damage or accelerate recovery of the lung.
Collapse
|
73
|
Yuan T, Volckaert T, Chanda D, Thannickal VJ, De Langhe SP. Fgf10 Signaling in Lung Development, Homeostasis, Disease, and Repair After Injury. Front Genet 2018; 9:418. [PMID: 30319693 PMCID: PMC6167454 DOI: 10.3389/fgene.2018.00418] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/06/2018] [Indexed: 12/15/2022] Open
Abstract
The lung is morphologically structured into a complex tree-like network with branched airways ending distally in a large number of alveoli for efficient oxygen exchange. At the cellular level, the adult lung consists of at least 40–60 different cell types which can be broadly classified into epithelial, endothelial, mesenchymal, and immune cells. Fibroblast growth factor 10 (Fgf10) located in the lung mesenchyme is essential to regulate epithelial proliferation and lineage commitment during embryonic development and post-natal life, and to drive epithelial regeneration after injury. The cells that express Fgf10 in the mesenchyme are progenitors for mesenchymal cell lineages during embryonic development. During adult lung homeostasis, Fgf10 is expressed in mesenchymal stromal niches, between cartilage rings in the upper conducting airways where basal cells normally reside, and in the lipofibroblasts adjacent to alveolar type 2 cells. Fgf10 protects and promotes lung epithelial regeneration after different types of lung injuries. An Fgf10-Hippo epithelial-mesenchymal crosstalk ensures maintenance of stemness and quiescence during homeostasis and basal stem cell (BSC) recruitment to further promote regeneration in response to injury. Fgf10 signaling is dysregulated in different human lung diseases including bronchopulmonary dysplasia (BPD), idiopathic pulmonary fibrosis (IPF), and chronic obstructive pulmonary disease (COPD), suggesting that dysregulation of the FGF10 pathway is critical to the pathogenesis of several human lung diseases.
Collapse
Affiliation(s)
- Tingting Yuan
- Division of Pulmonary, Department of Medicine, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham AL, United States
| | - Thomas Volckaert
- Division of Pulmonary, Department of Medicine, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham AL, United States
| | - Diptiman Chanda
- Division of Pulmonary, Department of Medicine, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham AL, United States
| | - Victor J Thannickal
- Division of Pulmonary, Department of Medicine, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham AL, United States
| | - Stijn P De Langhe
- Division of Pulmonary, Department of Medicine, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham AL, United States
| |
Collapse
|
74
|
Hui KPY, Ching RHH, Chan SKH, Nicholls JM, Sachs N, Clevers H, Peiris JSM, Chan MCW. Tropism, replication competence, and innate immune responses of influenza virus: an analysis of human airway organoids and ex-vivo bronchus cultures. THE LANCET RESPIRATORY MEDICINE 2018; 6:846-854. [PMID: 30001996 DOI: 10.1016/s2213-2600(18)30236-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Human airway organoids are three-dimensional cultures derived from stem cells, which self-organise in ex-vivo conditions to form so-called mini-airways. The cellular morphology of these cultures is physiologically similar to the human airway, with cilia, goblet cells, and club cells facing the inner lumen and basal cells situated at the outer layer. The aim of this study was to compare replication competence, tissue tropism, and host responses elicited by human and avian strains of influenza A virus in ex-vivo human bronchus and human airway organoids. METHODS Between Sept 29, 2016, and Jan 4, 2017, we obtained ex-vivo cultures of the human bronchus and cultured human airway organoids from lung stem cells obtained from human lung tissues removed as part of the routine clinical care of patients undergoing surgical resection at the Department of Cardiothoracic Surgery, University of Hong Kong, Queen Mary Hospital, Hong Kong. We compared viral replication competence, tissue tropism, and cytokine and chemokine induction of avian influenza A viruses isolated from humans (Sh2/H7N9, H5N1/483, H5N6/39715), and human H1N1pdm/415742 in airway organoids and ex-vivo bronchus explant cultures. FINDINGS Virus tropism and replication kinetics of human and avian influenza A viruses in human airway organoids mimicked those found in ex-vivo cultures of human bronchus explants. In both airway organoids and bronchus explants, influenza A H1N1 subtype (H1N1) and avian influenza A H7N9 viruses replicated to significantly higher titres than did the highly pathogenic avian influenza (HPAI) H5N1, whereas HPAI H5N6 replication was moderate. H1N1, H7N9, and H5N6 viruses infected ciliated cells and goblet cells, but not basal cells in both airway organoids and bronchus explants. The expression of cytokines, interleukin 6, and interferon β, and the chemokine regulated-on-activation, normal T-cell expressed and secreted, was significantly higher in human airway organoids infected with HPAI H5N1 virus than H1N1pdm/415742, Sh2/H7N9, and H5N6/39715 viruses, and the expression of monocyte chemoattractant protein-1 was significantly higher in human organoids infected with HPAI H5N1 virus than H1N1pdm/415742 and Sh2/H7N9 viruses. INTERPRETATION Human airway organoid cultures provided results that were comparable to those observed in human ex-vivo bronchus cultures, and thus provide an alternative physiologically relevant experimental model for investigating virus tropism and replication competence that could be used to assess the pandemic threat of animal influenza viruses. FUNDING US National Institute of Allergy and Infectious Diseases, Research Grants Council of the Hong Kong Special Administrative Region.
Collapse
Affiliation(s)
- Kenrie P Y Hui
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Rachel H H Ching
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Stan K H Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - John M Nicholls
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Norman Sachs
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Utrecht, Netherlands; Vertex Pharmaceuticals, San Diego, CA, USA
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Utrecht, Netherlands
| | - J S Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Michael C W Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
75
|
Sun X, Song L, Feng S, Li L, Yu H, Wang Q, Wang X, Hou Z, Li X, Li Y, Zhang Q, Li K, Cui C, Wu J, Qin Z, Wu Q, Chen H. Fatty Acid Metabolism is Associated With Disease Severity After H7N9 Infection. EBioMedicine 2018; 33:218-229. [PMID: 29941340 PMCID: PMC6085509 DOI: 10.1016/j.ebiom.2018.06.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/15/2018] [Accepted: 06/15/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Human infections with the H7N9 virus could lead to lung damage and even multiple organ failure, which is closely associated with a high mortality rate. However, the metabolic basis of such systemic alterations remains unknown. METHODS This study included hospitalized patients (n = 4) with laboratory-confirmed H7N9 infection, healthy controls (n = 9), and two disease control groups comprising patients with pneumonia (n = 9) and patients with pneumonia who received steroid treatment (n = 10). One H7N9-infected patient underwent lung biopsy for histopathological analysis and expression analysis of genes associated with lung homeostasis. H7N9-induced systemic alterations were investigated using metabolomic analysis of sera collected from the four patients by using ultra-performance liquid chromatography-mass spectrometry. Chest digital radiography and laboratory tests were also conducted. FINDINGS Two of the four patients did not survive the clinical treatments with antiviral medication, steroids, and oxygen therapy. Biopsy revealed disrupted expression of genes associated with lung epithelial integrity. Histopathological analysis demonstrated severe lung inflammation after H7N9 infection. Metabolomic analysis indicated that fatty acid metabolism may be inhibited during H7N9 infection. Serum levels of palmitic acid, erucic acid, and phytal may negatively correlate with the extent of lung inflammation after H7N9 infection. The changes in fatty acid levels may not be due to steroid treatment or pneumonia. INTERPRETATION Altered structural and secretory properties of the lung epithelium may be associated with the severity of H7N9-infection-induced lung disease. Moreover, fatty acid metabolism level may predict a fatal outcome after H7N9 virus infection.
Collapse
Affiliation(s)
- Xin Sun
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300070, China
| | - Lijia Song
- Department of Respiratory Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Shuang Feng
- Department of Clinical Laboratory, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Li Li
- Department of Respiratory Medicine, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Hongzhi Yu
- Department of Respiratory Medicine, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Qiaoxing Wang
- Department of Clinical Laboratory, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Xing Wang
- Department of Respiratory Medicine, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Zhili Hou
- Department of Tuberculosis, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Xue Li
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300070, China
| | - Yu Li
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300070, China
| | - Qiuyang Zhang
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300070, China
| | - Kuan Li
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300070, China
| | - Chao Cui
- Department of Thoracic Surgery, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Junping Wu
- Department of Respiratory Medicine, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Zhonghua Qin
- Department of Clinical Laboratory, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Qi Wu
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300070, China; Department of Respiratory Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin 300350, China.
| | - Huaiyong Chen
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300070, China; Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin 300350, China.
| |
Collapse
|
76
|
Crane MJ, Lee KM, FitzGerald ES, Jamieson AM. Surviving Deadly Lung Infections: Innate Host Tolerance Mechanisms in the Pulmonary System. Front Immunol 2018; 9:1421. [PMID: 29988424 PMCID: PMC6024012 DOI: 10.3389/fimmu.2018.01421] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/07/2018] [Indexed: 12/16/2022] Open
Abstract
Much research on infectious diseases focuses on clearing the pathogen through the use of antimicrobial drugs, the immune response, or a combination of both. Rapid clearance of pathogens allows for a quick return to a healthy state and increased survival. Pathogen-targeted approaches to combating infection have inherent limitations, including their pathogen-specific nature, the potential for antimicrobial resistance, and poor vaccine efficacy, among others. Another way to survive an infection is to tolerate the alterations to homeostasis that occur during a disease state through a process called host tolerance or resilience, which is independent from pathogen burden. Alterations in homeostasis during infection are numerous and include tissue damage, increased inflammation, metabolic changes, temperature changes, and changes in respiration. Given its importance and sensitivity, the lung is a good system for understanding host tolerance to infectious disease. Pneumonia is the leading cause of death for children under five worldwide. One reason for this is because when the pulmonary system is altered dramatically it greatly impacts the overall health and survival of a patient. Targeting host pathways involved in maintenance of pulmonary host tolerance during infection could provide an alternative therapeutic avenue that may be broadly applicable across a variety of pathologies. In this review, we will summarize recent findings on tolerance to host lung infection. We will focus on the involvement of innate immune responses in tolerance and how an initial viral lung infection may alter tolerance mechanisms in leukocytic, epithelial, and endothelial compartments to a subsequent bacterial infection. By understanding tolerance mechanisms in the lung we can better address treatment options for deadly pulmonary infections.
Collapse
Affiliation(s)
| | | | | | - Amanda M. Jamieson
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| |
Collapse
|
77
|
Zacharias WJ, Frank DB, Zepp JA, Morley MP, Alkhaleel FA, Kong J, Zhou S, Cantu E, Morrisey EE. Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor. Nature 2018; 555:251-255. [PMID: 29489752 PMCID: PMC6020060 DOI: 10.1038/nature25786] [Citation(s) in RCA: 449] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 01/24/2018] [Indexed: 12/11/2022]
Abstract
Functional tissue regeneration is required for the restoration of normal organ homeostasis after severe injury. Some organs, such as the intestine, harbour active stem cells throughout homeostasis and regeneration; more quiescent organs, such as the lung, often contain facultative progenitor cells that are recruited after injury to participate in regeneration. Here we show that a Wnt-responsive alveolar epithelial progenitor (AEP) lineage within the alveolar type 2 cell population acts as a major facultative progenitor cell in the distal lung. AEPs are a stable lineage during alveolar homeostasis but expand rapidly to regenerate a large proportion of the alveolar epithelium after acute lung injury. AEPs exhibit a distinct transcriptome, epigenome and functional phenotype and respond specifically to Wnt and Fgf signalling. In contrast to other proposed lung progenitor cells, human AEPs can be directly isolated by expression of the conserved cell surface marker TM4SF1, and act as functional human alveolar epithelial progenitor cells in 3D organoids. Our results identify the AEP lineage as an evolutionarily conserved alveolar progenitor that represents a new target for human lung regeneration strategies.
Collapse
Affiliation(s)
- William J Zacharias
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - David B Frank
- Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Jarod A Zepp
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Michael P Morley
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Farrah A Alkhaleel
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Jun Kong
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Su Zhou
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Edward Cantu
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Edward E Morrisey
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
78
|
Digital gene expression analysis in mice lung with coinfection of influenza and streptococcus pneumoniae. Oncotarget 2017; 8:112748-112760. [PMID: 29348862 PMCID: PMC5762547 DOI: 10.18632/oncotarget.23104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 11/26/2017] [Indexed: 01/02/2023] Open
Abstract
Influenza A virus (IAV) and Streptococcus pneumoniae (SP) are two major upper respiratory tract pathogens that can also cause infection in polarized bronchial epithelial cells to exacerbate disease in coinfected individuals which may result in significant morbidity. However, the underlying molecular mechanism is poorly understood. Here, we employed BALB/c ByJ mice inflected with SP, IAV, IAV followed by SP (IAV+SP) and PBS (Control) as models to survey the global gene expression using digital gene expression (DGE) profiling. We attempt to gain insights into the underlying genetic basis of this synergy at the expression level. Gene expression profiles were obtain using the Illimina/Hisseq sequencing technique, and further analyzed by enrichment analysis of Gene Ontology (GO) and Pathway function. The hematoxylin-eosin (HE) staining revealed different tissue changes in groups during which IAV+SP group showed the most severe cell apoptosis. Compared with Control, a total of 2731, 3221 and 3946 differentially expressed genes (DEGs) were detected in SP, IAV and IAV+SP respectively. Besides, sixty-two GO terms were identified by Gene Ontology functional enrichment analysis, such as cell killing, biological regulation, response to stimulus, signaling, biological adhesion, enzyme regulator activity, receptor regulator activity and translation regulator activity. Pathway significant enrichment analysis indicated the dysregulation of multiple pathways, including apoptosis pathway. Among these, five selected genes were further verified by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). This study shows that infection with SP, IAV or IAV+SP induces apoptosis with different degrees which might provide insights into the molecular mechanisms to facilitate further research.
Collapse
|
79
|
Li X, Yang L, Sun X, Wu J, Li Y, Zhang Q, Zhang Y, Li K, Wu Q, Chen H. The role of TGFβ‑HGF‑Smad4 axis in regulating the proliferation of mouse airway progenitor cells. Mol Med Rep 2017; 16:8155-8163. [PMID: 28983602 PMCID: PMC5779903 DOI: 10.3892/mmr.2017.7636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 08/07/2017] [Indexed: 02/05/2023] Open
Abstract
The interaction between airway epithelial progenitor cells and their microenvironment is critical for maintaining lung homeostasis. This microenvironment includes fibroblast cells, which support the growth of airway progenitor cells. However, the mechanism of this support is not fully understood. In the present study, the authors observed that inhibition of transforming growth factor (TGF)‑β signal with SB431542 promotes the expression of hepatocyte growth factor (HGF) in fibroblast cells. The HGF receptor, c‑Met, is expressed on airway progenitor cells; HGF promotes the colony‑forming ability of airway progenitor cells. The deletion of Smad4 in airway progenitor cells increases the colony‑forming ability, suggesting that Smad4 plays a negative role in the regulating the proliferation of airway progenitor cells. These data demonstrated that the regulation of airway progenitor cells by TGF‑β depends on TGF‑βR1/2 on stromal cells, rather than on epithelial progenitor cells. These data suggested a role for the TGF‑β‑TGF‑βR1/2‑HGF‑Smad4 axis in airway epithelial homeostasis and sheds new light on the interaction between airway progenitor cells and their microenvironment.
Collapse
Affiliation(s)
- Xue Li
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300350, P.R. China
| | - Li Yang
- Department of Respiratory Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Xin Sun
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin 300350, P.R. China
| | - Junping Wu
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin 300350, P.R. China
- Respiratory Department, Tianjin Haihe Hospital, Tianjin 300350, P.R. China
| | - Yu Li
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300350, P.R. China
| | - Qiuyang Zhang
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300350, P.R. China
| | - Yingchao Zhang
- Respiratory Department, Baodi Clinical College of Tianjin Medical University, Tianjin 301800, P.R. China
| | - Kuan Li
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300350, P.R. China
| | - Qi Wu
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300350, P.R. China
- Department of Respiratory Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin 300350, P.R. China
- Tianjin Institute of Respiratory Diseases, Tianjin 300350, P.R. China
- Correspondence to: Dr Huaiyong Chen or Dr Qi Wu, Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, 890 Jingu Road, Jinnan, Tianjin 300350, P.R. China, E-mail: , E-mail:
| | - Huaiyong Chen
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300350, P.R. China
- Tianjin Institute of Respiratory Diseases, Tianjin 300350, P.R. China
- Correspondence to: Dr Huaiyong Chen or Dr Qi Wu, Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, 890 Jingu Road, Jinnan, Tianjin 300350, P.R. China, E-mail: , E-mail:
| |
Collapse
|
80
|
Alvira CM, Morty RE. Can We Understand the Pathobiology of Bronchopulmonary Dysplasia? J Pediatr 2017; 190:27-37. [PMID: 29144252 PMCID: PMC5726414 DOI: 10.1016/j.jpeds.2017.08.041] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/28/2017] [Accepted: 08/16/2017] [Indexed: 01/17/2023]
Affiliation(s)
- Cristina M. Alvira
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California 94305
| | - Rory E. Morty
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center campus of the German Center for Lung Research, Giessen, Germany,Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
81
|
Surate Solaligue DE, Rodríguez-Castillo JA, Ahlbrecht K, Morty RE. Recent advances in our understanding of the mechanisms of late lung development and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2017; 313:L1101-L1153. [PMID: 28971976 DOI: 10.1152/ajplung.00343.2017] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/21/2017] [Accepted: 09/23/2017] [Indexed: 02/08/2023] Open
Abstract
The objective of lung development is to generate an organ of gas exchange that provides both a thin gas diffusion barrier and a large gas diffusion surface area, which concomitantly generates a steep gas diffusion concentration gradient. As such, the lung is perfectly structured to undertake the function of gas exchange: a large number of small alveoli provide extensive surface area within the limited volume of the lung, and a delicate alveolo-capillary barrier brings circulating blood into close proximity to the inspired air. Efficient movement of inspired air and circulating blood through the conducting airways and conducting vessels, respectively, generates steep oxygen and carbon dioxide concentration gradients across the alveolo-capillary barrier, providing ideal conditions for effective diffusion of both gases during breathing. The development of the gas exchange apparatus of the lung occurs during the second phase of lung development-namely, late lung development-which includes the canalicular, saccular, and alveolar stages of lung development. It is during these stages of lung development that preterm-born infants are delivered, when the lung is not yet competent for effective gas exchange. These infants may develop bronchopulmonary dysplasia (BPD), a syndrome complicated by disturbances to the development of the alveoli and the pulmonary vasculature. It is the objective of this review to update the reader about recent developments that further our understanding of the mechanisms of lung alveolarization and vascularization and the pathogenesis of BPD and other neonatal lung diseases that feature lung hypoplasia.
Collapse
Affiliation(s)
- David E Surate Solaligue
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - José Alberto Rodríguez-Castillo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Katrin Ahlbrecht
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and .,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| |
Collapse
|
82
|
Dial CF, Tune MK, Doerschuk CM, Mock JR. Foxp3 + Regulatory T Cell Expression of Keratinocyte Growth Factor Enhances Lung Epithelial Proliferation. Am J Respir Cell Mol Biol 2017; 57:162-173. [PMID: 28296468 DOI: 10.1165/rcmb.2017-0019oc] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Repair of the lung epithelium after injury is a critical component for resolution; however, the processes necessary to drive epithelial resolution are not clearly defined. Published data demonstrate that Foxp3+ regulatory T cells (Tregs) enhance alveolar epithelial proliferation after injury, and Tregs in vitro directly promote type II alveolar epithelial cell (AT2) proliferation, in part by a contact-independent mechanism. Therefore, we sought to determine the contribution of Treg-specific expression of a growth factor that is known to be important in lung repair, keratinocyte growth factor (kgf). The data demonstrate that Tregs express kgf and that Treg-specific expression of kgf regulates alveolar epithelial proliferation during the resolution phase of acute lung injury and in a model of regenerative alveologenesis in vivo. In vitro experiments demonstrate that AT2 cells cocultured with Tregs lacking kgf have decreased rates of proliferation compared with AT2 cells cocultured with wild-type Tregs. Moreover, Tregs isolated from lung tissue and grown in culture express higher levels of two growth factors that are important for lung repair (kgf and amphiregulin) compared with Tregs isolated from splenic tissue. Lastly, Tregs isolated from human lung tissue can be stimulated ex vivo to induce kgf expression. This study reveals mechanisms by which Tregs direct tissue-reparative effects during resolution after acute lung injury, further supporting the emerging role of Tregs in tissue repair.
Collapse
Affiliation(s)
- Catherine F Dial
- 1 Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine.,2 Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina
| | - Miriya K Tune
- 1 Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine.,2 Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina
| | - Claire M Doerschuk
- 1 Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine.,3 Center for Airways Disease, and.,2 Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina
| | - Jason R Mock
- 1 Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine.,2 Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
83
|
Abstract
Purpose of Review The lung research field has pioneered the use of organoids for the study of cell-cell interactions. Recent Findings The use of organoids for airway basal cells is routine. However, the development of organoids for the other regions of the lung is still in its infancy. Such cultures usually rely on cell-cell interactions between the stem cells and a putative niche cell for their growth and differentiation. Summary The use of co-culture organoid systems has facilitated the in vitro cultivation of previously inaccessible stem cell populations, providing a novel method for dissecting the molecular requirements of these cell-cell interactions. Future technology development will allow the growth of epithelial-only organoids in more defined media and also the introduction of specific non-epithelial cells for the study of cell interactions. These developments will require an improved understanding of the epithelial and non-epithelial cell types present in the lung and their lineage relationships.
Collapse
|
84
|
Deng M, Li J, Gan Y, Chen P. [Advances in Classification and Research Methods of Lung Epithelial Stem
and Progenitor Cells]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 20:130-137. [PMID: 28228225 PMCID: PMC5972970 DOI: 10.3779/j.issn.1009-3419.2017.02.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
分离和鉴定肺上皮干/祖细胞,深入了解他们在肺脏生理病理条件下的具体作用机理,对于防治包括肺癌在内的肺脏疾病有重要意义。本综述介绍了已鉴定的肺上皮干/祖细胞种类和肺上皮干/祖细胞研究方法的最新进展,前者具有区域特异性,主要包括位近端气道的基底细胞和导管细胞,位细支气管的Clara细胞、变异Clara细胞、细支气管肺泡干细胞和诱导出的krt5+细胞及位肺泡的Ⅱ型肺泡上皮细胞和Ⅱ型肺泡上皮祖细胞;后者主要包括肺损伤模型、谱系示踪技术、三维培养技术、移植、慢性标记细胞法及单细胞转录组学分析等。最后简述了肺上皮干/祖细胞与肺癌的关系以及肺癌干细胞靶向药物治疗进展。
Collapse
Affiliation(s)
- Minhua Deng
- Department of Respiratory Medicine, PLA Rocket Force General Hospital, Beijing 100088, China;Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Jinhua Li
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Ye Gan
- Department of Rehabilitation, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Ping Chen
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
85
|
Nardiello C, Mižíková I, Morty RE. Looking ahead: where to next for animal models of bronchopulmonary dysplasia? Cell Tissue Res 2016; 367:457-468. [PMID: 27917436 PMCID: PMC5320021 DOI: 10.1007/s00441-016-2534-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/01/2016] [Indexed: 11/16/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common complication of preterm birth, with appreciable morbidity and mortality in a neonatal intensive care setting. Much interest has been shown in the identification of pathogenic pathways that are amenable to pharmacological manipulation (1) to facilitate the development of novel therapeutic and medical management strategies and (2) to identify the basic mechanisms of late lung development, which remains poorly understood. A number of animal models have therefore been developed and continue to be refined with the aim of recapitulating pathological pulmonary hallmarks noted in lungs from neonates with BPD. These animal models rely on several injurious stimuli, such as mechanical ventilation or oxygen toxicity and infection and sterile inflammation, as applied in mice, rats, rabbits, pigs, lambs and nonhuman primates. This review addresses recent developments in modeling BPD in experimental animals and highlights important neglected areas that demand attention. Additionally, recent progress in the quantitative microscopic analysis of pathology tissue is described, together with new in vitro approaches of value for the study of normal and aberrant alveolarization. The need to examine long-term sequelae of damage to the developing neonatal lung is also considered, as is the need to move beyond the study of the lungs alone in experimental animal models of BPD.
Collapse
Affiliation(s)
- Claudio Nardiello
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Ivana Mižíková
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231, Bad Nauheim, Germany. .,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany.
| |
Collapse
|
86
|
Zhang YQ, Li P, Zhang FQ, Sun SJ, Cao YG. Lgr5 regulates the regeneration of lesioned nasal respiratory epithelium. Biochem Biophys Res Commun 2016; 481:195-200. [PMID: 27773817 DOI: 10.1016/j.bbrc.2016.10.075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 10/20/2016] [Indexed: 10/20/2022]
Abstract
Nasal respiratory epithelium is a ciliated pseudostratified columnar epithelium. The cellular components of nasal respiratory epithelium include ciliated cells, goblet cells, and basal cells. Until now, our knowledge in the development of nasal respiratory epithelium is still limited and the cellular mechanism of regeneration is still elusive. In this study, we found that adult stem cell marker leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5) is expressed in the mice nasal respiratory epithelium. Both immunostaining and lineage tracing analysis indicated Lgr5 positive cells in the nasal respiratory epithelium are proliferative stem/progenitor cells. Using the Rosa-Tdtomato and Rosa26-DTR mice, we elucidated that Lgr5+ cells participate in the regeneration of lesioned nasal respiratory epithelium, and this group of cells is necessary in the process of epithelium recovery. Using the in vitro culture system, we observed the formation of spheres from Lgr5+ cells and these spheres have the capacity to generate other types of cells. Above all, this study reported a group of previously unidentified progenitor/stem cells in nasal respiratory epithelium, unveiling the potential cellular mechanism in nasal respiratory epithelium regeneration.
Collapse
Affiliation(s)
- Yan-Qiang Zhang
- Department of Clinical Laboratory, Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University, Shandong Province, China
| | - Peng Li
- Department of Clinical Laboratory, Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University, Shandong Province, China
| | - Feng-Qin Zhang
- Department of Infusion and Injection Room, Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University, Shandong Province, China.
| | - Shao-Jun Sun
- Department of Clinical Laboratory, Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University, Shandong Province, China
| | - Yin-Guang Cao
- Department of Clinical Laboratory, Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University, Shandong Province, China
| |
Collapse
|
87
|
A Triple Co-Culture Model of the Human Respiratory Tract to Study Immune-Modulatory Effects of Liposomes and Virosomes. PLoS One 2016; 11:e0163539. [PMID: 27685460 PMCID: PMC5042471 DOI: 10.1371/journal.pone.0163539] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 09/09/2016] [Indexed: 12/19/2022] Open
Abstract
The respiratory tract with its ease of access, vast surface area and dense network of antigen-presenting cells (APCs) represents an ideal target for immune-modulation. Bio-mimetic nanocarriers such as virosomes may provide immunomodulatory properties to treat diseases such as allergic asthma. In our study we employed a triple co-culture model of epithelial cells, macrophages and dendritic cells to simulate the human airway barrier. The epithelial cell line 16HBE was grown on inserts and supplemented with human blood monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs) for exposure to influenza virosomes and liposomes. Additionally, primary human nasal epithelial cells (PHNEC) and EpCAM+ epithelial progenitor cell mono-cultures were utilized to simulate epithelium from large and smaller airways, respectively. To assess particle uptake and phenotype change, cell cultures were analyzed by flow cytometry and pro-inflammatory cytokine concentrations were measured by ELISA. All cell types internalized virosomes more efficiently than liposomes in both mono- and co-cultures. APCs like MDMs and MDDCs showed the highest uptake capacity. Virosome and liposome treatment caused a moderate degree of activation in MDDCs from mono-cultures and induced an increased cytokine production in co-cultures. In epithelial cells, virosome uptake was increased compared to liposomes in both mono- and co-cultures with EpCAM+ epithelial progenitor cells showing highest uptake capacity. In conclusion, all cell types successfully internalized both nanocarriers with virosomes being taken up by a higher proportion of cells and at a higher rate inducing limited activation of MDDCs. Thus virosomes may represent ideal carrier antigen systems to modulate mucosal immune responses in the respiratory tract without causing excessive inflammatory changes.
Collapse
|
88
|
Gilpin SE, Charest JM, Ren X, Tapias LF, Wu T, Evangelista-Leite D, Mathisen DJ, Ott HC. Regenerative potential of human airway stem cells in lung epithelial engineering. Biomaterials 2016; 108:111-9. [PMID: 27622532 DOI: 10.1016/j.biomaterials.2016.08.055] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/25/2016] [Accepted: 08/31/2016] [Indexed: 12/24/2022]
Abstract
Bio-engineered organs for transplantation may ultimately provide a personalized solution for end-stage organ failure, without the risk of rejection. Building upon the process of whole organ perfusion decellularization, we aimed to develop novel, translational methods for the recellularization and regeneration of transplantable lung constructs. We first isolated a proliferative KRT5(+)TP63(+) basal epithelial stem cell population from human lung tissue and demonstrated expansion capacity in conventional 2D culture. We then repopulated acellular rat scaffolds in ex vivo whole organ culture and observed continued cell proliferation, in combination with primary pulmonary endothelial cells. To show clinical scalability, and to test the regenerative capacity of the basal cell population in a human context, we then recellularized and cultured isolated human lung scaffolds under biomimetic conditions. Analysis of the regenerated tissue constructs confirmed cell viability and sustained metabolic activity over 7 days of culture. Tissue analysis revealed extensive recellularization with organized tissue architecture and morphology, and preserved basal epithelial cell phenotype. The recellularized lung constructs displayed dynamic compliance and rudimentary gas exchange capacity. Our results underline the regenerative potential of patient-derived human airway stem cells in lung tissue engineering. We anticipate these advances to have clinically relevant implications for whole lung bioengineering and ex vivo organ repair.
Collapse
Affiliation(s)
- Sarah E Gilpin
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Jonathan M Charest
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Xi Ren
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Luis F Tapias
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Tong Wu
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Daniele Evangelista-Leite
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Douglas J Mathisen
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States
| | - Harald C Ott
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| |
Collapse
|
89
|
Abstract
Seasonal and pandemic influenza are the two faces of respiratory infections caused by influenza viruses in humans. As seasonal influenza occurs on an annual basis, the circulating virus strains are closely monitored and a yearly updated vaccination is provided, especially to identified risk populations. Nonetheless, influenza virus infection may result in pneumonia and acute respiratory failure, frequently complicated by bacterial coinfection. Pandemics are, in contrary, unexpected rare events related to the emergence of a reassorted human-pathogenic influenza A virus (IAV) strains that often causes increased morbidity and spreads extremely rapidly in the immunologically naive human population, with huge clinical and economic impact. Accordingly, particular efforts are made to advance our knowledge on the disease biology and pathology and recent studies have brought new insights into IAV adaptation mechanisms to the human host, as well as into the key players in disease pathogenesis on the host side. Current antiviral strategies are only efficient at the early stages of the disease and are challenged by the genomic instability of the virus, highlighting the need for novel antiviral therapies targeting the pulmonary host response to improve viral clearance, reduce the risk of bacterial coinfection, and prevent or attenuate acute lung injury. This review article summarizes our current knowledge on the molecular basis of influenza infection and disease progression, the key players in pathogenesis driving severe disease and progression to lung failure, as well as available and envisioned prevention and treatment strategies against influenza virus infection.
Collapse
Affiliation(s)
- Christin Peteranderl
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Carole Schmoldt
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| |
Collapse
|
90
|
Abstract
Seasonal and pandemic influenza are the two faces of respiratory infections caused by influenza viruses in humans. As seasonal influenza occurs on an annual basis, the circulating virus strains are closely monitored and a yearly updated vaccination is provided, especially to identified risk populations. Nonetheless, influenza virus infection may result in pneumonia and acute respiratory failure, frequently complicated by bacterial coinfection. Pandemics are, in contrary, unexpected rare events related to the emergence of a reassorted human-pathogenic influenza A virus (IAV) strains that often causes increased morbidity and spreads extremely rapidly in the immunologically naive human population, with huge clinical and economic impact. Accordingly, particular efforts are made to advance our knowledge on the disease biology and pathology and recent studies have brought new insights into IAV adaptation mechanisms to the human host, as well as into the key players in disease pathogenesis on the host side. Current antiviral strategies are only efficient at the early stages of the disease and are challenged by the genomic instability of the virus, highlighting the need for novel antiviral therapies targeting the pulmonary host response to improve viral clearance, reduce the risk of bacterial coinfection, and prevent or attenuate acute lung injury. This review article summarizes our current knowledge on the molecular basis of influenza infection and disease progression, the key players in pathogenesis driving severe disease and progression to lung failure, as well as available and envisioned prevention and treatment strategies against influenza virus infection.
Collapse
Affiliation(s)
- Christin Peteranderl
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Carole Schmoldt
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| |
Collapse
|