key: cord-1022375-iy2yutq6 authors: Djidrovski, Ivo; Georgiou, Maria; Hughes, Grant L.; Patterson, Edward I.; Casas‐Sanchez, Aitor; Pennington, Shaun H.; Biagini, Giancarlo A.; Moya‐Molina, Marina; van den Bor, Jelle; Smit, Martine J.; Chung, Git; Lako, Majlinda; Armstrong, Lyle title: SARS‐CoV‐2 infects an upper airway model derived from induced pluripotent stem cells date: 2021-06-21 journal: Stem Cells DOI: 10.1002/stem.3422 sha: 98cbf0830bdf7d10fa88a3c38c6a065df5eac9ff doc_id: 1022375 cord_uid: iy2yutq6 As one of the primary points of entry of xenobiotic substances and infectious agents into the body, the lungs are subject to a range of dysfunctions and diseases that together account for a significant number of patient deaths. In view of this, there is an outstanding need for in vitro systems in which to assess the impact of both infectious agents and xenobiotic substances of the lungs. To address this issue, we have developed a protocol to generate airway epithelial basal‐like cells from induced pluripotent stem cells, which simplifies the manufacture of cellular models of the human upper airways. Basal‐like cells generated in this study were cultured on transwell inserts to allow formation of a confluent monolayer and then exposed to an air‐liquid interface to induce differentiation into a pseudostratified epithelial construct with a marked similarity to the upper airway epithelium in vivo. These constructs contain the component cell types required of an epithelial model system, produce mucus and functional cilia, and can support SARS‐CoV‐2 infection/replication and the secretion of cytokines in a manner similar to that of in vivo airways. This method offers a readily accessible and highly scalable protocol for the manufacture of upper airway models that could find applications in development of therapies for respiratory viral infections and the assessment of drug toxicity on the human lungs. both infectious agents and xenobiotic substances of the lungs. To address this issue, we have developed a protocol to generate airway epithelial basal-like cells from induced pluripotent stem cells, which simplifies the manufacture of cellular models of the human upper airways. Basal-like cells generated in this study were cultured on transwell inserts to allow formation of a confluent monolayer and then exposed to an air-liquid interface to induce differentiation into a pseudostratified epithelial construct with a marked similarity to the upper airway epithelium in vivo. These constructs contain the component cell types required of an epithelial model system, produce mucus and functional cilia, and can support SARS-CoV-2 infection/replication and the secretion of cytokines in a manner similar to that of in vivo airways. This method offers a readily accessible and highly scalable protocol for the manufacture of upper airway models that could find applications in development of therapies for respiratory viral infections and the assessment of drug toxicity on the human lungs. Demonstration of the ability of SARS-CoV-2 to infect an airway construct generated from induced pluripotent stem cells is significant since it paves the way for broader studies of viral airway infection using a system that can be manufactured reproducibly. [Correction added on 10 July 2021, after first online publication: Author name Shaun H. Penington has been changed to Shaun H. Pennington and Ivo Djidrovksi has been changed to Ivo The pandemic has resulted in major challenges to global healthcare systems and has severe consequences for the global economy if the spread of the virus is not effectively controlled. The causative agent of COVID-19, SARS-CoV-2, has been shown to infect the respiratory system resulting in viral pneumonia, but it may also affect the gastrointestinal system, heart, kidney, liver, and central nervous system leading to multiple organ failure. 2, 3 Previous studies have shown that SARS-CoV predominantly infects airway and alveolar epithelial cells, and macrophages 4 using the angiotensinconverting enzyme 2 (ACE2) receptor for entry. 5, 6 Rapid viral replication in these cells can lead to epithelial cell apoptosis causing the release of pro-inflammatory cytokines, 7 which can potentially cause airway damage and diminished patient survival. This is exemplified by the observation that in SARS-CoV-2-infected individuals, interleukin (IL)-6, IL-10, and tumor necrosis factor α (TNFα) surge during illness and decline during recovery. 8 Severely affected patients who require intensive care treatment can be distinguished by significantly higher levels of IL-6, IL-10, and TNFα and fewer CD4+ and CD8+ T cells. 9 Although it is likely that the ingress of large numbers of cytokinesecreting inflammatory macrophages into the lung tissue accounts for a significant proportion of the cytokines detected in such cases, the initial damage to the airway epithelial cells probably contributes not only to the overall concentration of cytokines but may also be responsible for recruitment of inflammatory macrophages. Other mechanisms besides apoptosis can lead to activation of the inflammasome. The binding of SARS-CoV-2 to the Toll-like receptor causes the release of pro-IL-1β, which is cleaved by caspase-1, followed by inflammasome activation and production of active mature IL-1β that mediate lung inflammation, fever, and fibrosis. 10 To underline this, suppression of pro-inflammatory IL-6 has been shown to have a therapeutic effect in COVID-19. 11 IL-1β can also enhance the constitutive detachment (or shedding) of an enzymatically active ectodomain fragment of ACE2 from the airway epithelial cells, an event associated with acute lung injury. 12, 13 Interestingly, SARS-CoV-2 infection is associated with ACE2 downregulation and ectodomain shedding thought to be induced by the SARS-CoV-2 Spike (S) protein. 14, 15 How the released form of ACE2 (the so-called soluble or sACE2) causes lung damage is not completely clear but it seems to be tightly coupled to TNFα production, so it may be involved in inflammatory response to SARS-CoV-2 infection. 16 Despite this understanding of the mechanisms by which SARS-CoV-type viruses damage the airway epithelia, there are no effective treatments for the resulting COVID-19 disease. Current management of COVID-19 is supportive, and respiratory failure from acute respiratory distress syndrome is the leading cause of mortality. In view of this, there is an urgent and currently unmet need for model systems that can function as high-throughput preclinical tools for the development of novel, effective therapies for COVID-19. The use of in vitro models mimicking the human airways generated from primary pulmonary epithelial cells grown at an air-liquid interface (ALI) has increased in popularity over the recent years. The cells can form a pseudostratified airway epithelium composed of all in vivo relevant cell types found in the airway epithelium including rare types such as pulmonary neuroendocrine cells and ionocytes. 17 This type of model has proven to be particularly useful for toxicological assessment of aerosol particles, 18, 19 drug discovery, 20 and more recently with the COVID-19 outbreak for viral infection studies. 21 Although these models offer significant advantages, their availability is limited to primary samples, which can significantly differ depending on the donor's genetic background and thus affecting the generated airway. Induced pluripotent stem cells (iPSCs) offer the potential to complement the limitations of primary cells by generating a large supply of cells with the genetic background of the donor. The pathways involved in differentiation into proximal and distal airway lineages have already been established by previous groups, who successfully differentiated functional ciliated cells, mucus-producing cells, and alveolar cells. [22] [23] [24] [25] [26] The current methods of lung differentiation use 3D self-forming spheroids or 2D cultures of mixed population of cells, which are more challenging when performing experiments similar to the primary airway ALI models. [27] [28] [29] In this study, we isolated a population of basal-like cells from differentiating iPSCs and used these to Mature ALI cultures prepared in this manner were fixed directly on the membrane with 4% paraformaldehyde (PFA) for 10 minutes at 3 C and then washed with PBS (3 Â 1.0 mL). The tissues were then removed together with the membrane, placed into molds, and embedded in optical coherence tomography (OCT) matrix (Cell Path, Newtown, UK, KMA-0100-00A). The molds were placed at À20 C to solidify. Once solid, they were sectioned into 5-to 10-μm slices on slides using a cryostat. The sectioned membrane was removed with PBS washes, and the slides were then stained using the same procedure as the basal cells using the same antibodies with the addition of antibodies (Tables 1 and 2 ). Once the staining was finalized, a few drops of Vectashield medium containing Hoechst were added to the slides then they were covered with coverslips, sealed with nail polish, and left to dry at 4 C. Basal airway-like cells generated in this manner were characterized by a combination of flow cytometry (Fortessa flow cytometer and FloJo analysis) and immunofluorescence (IF) (see Table 1 settes. Subsequently, paraffination was performed using the Excelsior AS Tissue processor (Thermo Fisher Scientific, A82300001), and the paraffinized tissues were placed into molds. Once the paraffin had solidified, 3-μm sections were made using a microtome. The slides were rehydrated using xylene and an ethanol series (100%, 96%, and 70%). Subsequently, the slides were stained using Mayer's hematoxylin (Sigma-Aldrich, MHS32) and alcoholic eosin Y (Sigma-Aldrich, HT110116) followed by dehydration using an ethanol series (80%, 96%, and 100%). Xylene-washed slides were mechanically covered using coverslips and dried at room temperature. Histology was assessed using an AxioVert 25 inverted microscope (Zeiss). Prior to imaging, the apical surface of the ALI cultures was washed using medium from the basal chamber. Subsequently, high-speed videos were captured using the Nikon Eclipse Ti2 LIPSI high content imaging microscope equipped with a phase 1 phase contrast ring and a CFI S Plan Fluor LWD Â20 objective. The middle of the Prime BSI sCMOS camera was used to capture 550 images (512 Â 512 pixels) over 5.5 seconds at a rate of 100 frames per second. During imaging, the atmosphere was constantly kept at 37 C, 5% CO 2 , and 95% humidity. At least three fields containing cilia for three inserts were imaged. For analysis MATLAB was used to calculate cilia beat frequency. Here, the intensity-time trace of each pixel was filtered for frequencies between 5 and 25 Hz using a band-pass filter. Subsequently, the power spectrum density was calculated. The frequency of each pixel is defined by the highest peak of the power spectrum density. The ciliated cells were visualized by plotting the frequency of each pixel into a heat map. The frequency distribution of the ciliated cells was visualized by plotting the amount of nonzero pixels into a histogram. The average frequency of all nonzero pixels was compared using a student's t test. Organoids were embedded in 1% low-melting agarose at 30 C containing Slowfade diamond mounting oil and imaged using a Zeiss LSM880 confocal laser scanning microscope. At Â400, sections were 3D reconstructed from a series of z-stacks (20-40-mm slices) with automatic optimal thickness and 1 Airyscan unit pinhole. Orthogonal views were generated using Zeiss Zen 3.3 software. Quantification of cytokines was performed using the V-PLEX Plus Figure 2D ) and the expression of mucin-1 (goblet cells, Figure 2D ). The presence of putative pulmonary neuroendocrine cells is indicated by the expression of synaptophysin ( Figure 2D ). The expression of all the four markers was similar between iPSC-derived and primary lung cell-derived ALI constructs ( Figure 2D ), although for MUC1 and CC10, the expression in the primary lung cell-derived ALI constructs was stronger and local- In conclusion, collectively our data show that airway epithelial constructs generated from iPSCs-derived basal cells have promise as models of the response of the airway epithelia to viral infections. F I G U R E 5 Lung airway constructs secrete inflammatory cytokines in response to SARS-CoV-2 infection. Data shown as mean ± SEM, n = 3 In this study, we established an efficient method of isolation of iPSCsderived basal-like cells from a mixed population of lung progenitors. Comparably with primary cells, when grown in BEGM and 3T3-J2 feeders, the iPSC-derived basal cells can self-renew and form colonies. They express genes associated with a basal cell phenotype (KRT14, deltaNp63, NGFR, Integrin alpha 6) while maintaining their multipotent capacity. One other group has reported the isolation of a basal cell-like population from lung spheroids 44 The authors declared no potential conflicts of interest. The data that support the findings of this study are available from the corresponding author upon reasonable request. Additional supporting information may be found online in the Supporting Information section at the end of this article. Epidemiology, genetic recombination, and pathogenesis of coronaviruses Novel coronavirus from patients with pneumonia in China Understanding SARS-CoV-2-mediated inflammatory responses: from mechanisms to potential therapeutic tools Single-cell RNA expression profiling of ACE2, the putative receptor of Wuhan A pneumonia outbreak associated with a new coronavirus of probable bat origin COVID-19, cytokines and immunosuppression: what can we learn from severe acute respiratory syndrome? Immune characteristics of severe and critical COVID-19 patients Reduction and functional exhaustion of T cells in patients Induction of pro-inflammatory cytokines (IL-1 and IL-6) and lung inflammation by coronavirus-19 (COVI-19 or SARS-CoV-2): anti-inflammatory strategies IL-6 inhibitors in the treatment of serious COVID-19: a promising therapy Angiotensin-converting enzyme 2 protects from severe acute lung failure A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury Differential downregulation of ACE2 by the spike proteins of severe acute respiratory syndrome coronavirus and human coronavirus NL63 Endocytosis of the receptor-binding domain of SARS-CoV spike protein together with virus receptor ACE2 Modulation of TNFalpha-converting enzyme by the spike protein of SARS-CoV and ACE2 induces TNF-alpha production and facilitates viral entry A revised airway epithelial hierarchy includes CFTR-expressing ionocytes Potential of in vitro reconstituted 3D human airway epithelia (MucilAir™) to assess respiratory sensitizers Realistic exposure methods for investigating the interaction of nanoparticles with the lung at the airliquid interface in vitro Air-liquid interface (ALI) culture of human bronchial epithelial cell monolayers as an in vitro model for airway drug transport studies Characterization and treatment of SARS-CoV-2 in nasal and bronchial human airway epithelia Efficient differentiation of human embryonic stem cells to definitive endoderm Generation of anterior foregut endoderm from human embryonic and induced pluripotent stem cells Efficient generation of functional CFTR-expressing airway epithelial cells from human pluripotent stem cells Directed induction of functional multi-ciliated cells in proximal airway epithelial spheroids from human pluripotent stem cells Generation of alveolar epithelial spheroids via isolated progenitor cells from human pluripotent stem cells Efficient generation of lung and airway epithelial cells from human pluripotent stem cells A bioengineered niche promotes in vivo engraftment and maturation of pluripotent stem cell derived human lung organoids Generation of multiciliated cells in functional airway epithelia from human induced pluripotent stem cells Disrupted alternative splicing for genes implicated in splicing and ciliogenesis causes PRPF31 retinitis pigmentosa iPSC modeling of severe aplastic anemia reveals impaired differentiation and telomere shortening in blood progenitors Keratin-14 expression in pneumocytes as a marker of lung regeneration/repair during diffuse alveolar damage Basal cells as stem cells of the mouse trachea and human airway epithelium Lineage-negative progenitors mobilize to regenerate lung epithelium after major injury The spike protein of SARS-CoV-a target for vaccine and therapeutic development Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor Interleukin-6 in Covid-19: a systematic review and meta-analysis SARS-CoV-2 triggers inflammatory responses and cell death through caspase-8 activation Innate immunity in the lung: how epithelial cells fight against respiratory pathogens Host defense function of the airway epithelium in health and disease: clinical background The airway epithelium: soldier in the fight against respiratory viruses Interferon gamma, TGF-β1 and RANTES expression in upper airway samples from SARS-CoV-2 infected patients Glycogen synthase kinase 3 induces multilineage maturation of human pluripotent stem cell-derived lung progenitors in 3D culture