Skip to main content

Building a human lung from pluripotent stem cells to model respiratory viral infections

Abstract

To protect against the constant threat of inhaled pathogens, the lung is equipped with cellular defenders. In coordination with resident and recruited immune cells, this defence is initiated by the airway and alveolar epithelium following their infection with respiratory viruses. Further support for viral clearance and infection resolution is provided by adjacent endothelial and stromal cells. However, even with these defence mechanisms, respiratory viral infections are a significant global health concern, causing substantial morbidity, socioeconomic losses, and mortality, underlining the need to develop effective vaccines and antiviral medications. In turn, the identification of new treatment options for respiratory infections is critically dependent on the availability of tractable in vitro experimental models that faithfully recapitulate key aspects of lung physiology. For such models to be informative, it is important these models incorporate human-derived, physiologically relevant versions of all cell types that normally form part of the lungs anti-viral response. This review proposes a guideline using human induced pluripotent stem cells (iPSCs) to create all the disease-relevant cell types. iPSCs can be differentiated into lung epithelium, innate immune cells, endothelial cells, and fibroblasts at a large scale, recapitulating in vivo functions and providing genetic tractability. We advocate for building comprehensive iPSC-derived in vitro models of both proximal and distal lung regions to better understand and model respiratory infections, including interactions with chronic lung diseases.

Introduction

Respiratory viral infections are the most common cause of human illness, contributing substantially to morbidity, socioeconomic losses, and mortality worldwide [1]. Infections with respiratory syncytial virus (RSV) or influenza virus are leading causes of mortality in children and the elderly [2, 3]. Severe respiratory viral infections in infancy can predispose to later development of chronic lung diseases, including asthma [4, 5], and infections are a common trigger of chronic lung disease exacerbations [6,7,8]. Respiratory viruses infect individuals of all age groups; however, some respiratory viruses have seasonal oscillation of their outbreaks, with cases of RSV, influenza virus, and coronavirus (CoV) infections peaking during the winter, while rhinovirus, human metapneumovirus, and adenovirus are detected year round [9]. Furthermore, the emergence of novel viral strains, such as severe acute respiratory syndrome (SARS)-CoV-1 and SARS-CoV-2, is a reminder of the potential for new viruses to significantly impact public health.

The primary function of the lungs, to provide oxygen to the blood and expel carbon dioxide, renders the lungs constantly susceptible to inhaled pathogens. Air enters the lungs via the proximal conducting airway composed of the trachea, bronchi, and bronchioles, where it is delivered to the distal gas exchanging zones of the respiratory bronchioles and alveoli. To protect the lung, epithelial cells, resident and recruited immune cells, endothelial and mesenchymal cells coordinate defence against invading pathogens (summarised in Table 1).

Table 1 Summary of lung cell type functions at homeostasis and during respiratory viral infections, with reference to production of each cell type from iPSCs

The airway epithelium is a pseudostratified cell layer lining the air-exposed surfaces of the conducting airways, providing a physical barrier between the sterile internal areas of the body and the environment. Specialised multiciliated cells, goblet cells and club cells provide additional protection by trapping dust, allergens, pollutants, and pathogens in mucous and removing them from the airway by mucociliary clearance. Basal cells are the multipotent stem cell population residing at the base of the epithelium and differentiate into multiciliated cells, club, and goblet cells (reviewed [10]). There are additional rare cell types in the airway including pulmonary neuroendocrine cells, which relay environmental information to the brain [11], tuft cells which have specialised innate immune functions [12], and pulmonary ionocytes [13], which have specialised ion transport functions [14]. Additionally, airway epithelial cells operate as sentinels for respiratory infections as they are the primary target for respiratory viral infection [15, 16] where they function to secrete antimicrobial peptides and pro-inflammatory cytokines, to orchestrate appropriate immune responses [17]. In the alveoli, type 2 alveolar epithelial cells (AT2s) provide similar protection to the type 1 alveolar epithelial cells (AT1s) [18,19,20]. AT1 cells reside adjacent to endothelial cells of the alveolar capillary plexus to facilitate gas exchange with the blood. AT2 cells produce pulmonary surfactant which reduces surface tension preventing alveolar collapse and are facultative progenitors of the alveolus, akin to basal cells of the airway, which actively divide and differentiate into AT1 cells during development and after infection or injury [10, 19].

Supporting the epithelia of the proximal and distal lung is a stromal layer of extracellular matrix (ECM), secreted, maintained, and remodelled by fibroblasts in response to damage or inflammation. The stroma contributes to the maintenance of structural integrity and provides support to tissue resident cells while also remaining navigable for migratory cells, including resident and infiltrating immune cells. A dense network of blood vessels, comprised primarily of endothelial cells, extends throughout the lung. Blood vessels and capillaries are supported by the stroma in the proximal airways and are in direct contact with the basolateral surface of the alveolar epithelium. This facilitates gas exchange and enables the rapid recruitment of innate and adaptive immune cells following infections.

There is a dearth of vaccines or effective antiviral medications for most respiratory viruses. This scarcity of treatments is partly attributable to the absence of tractable human-relevant models for studying viral replication or for identifying host response pathways that could be targeted with pharmaceuticals. Moreover, the need for development of appropriate models is heightened by the human-specific nature of many viruses, diminishing the utility of animal models [21,22,23]. Likewise, while transformed human cell lines are easy to culture and manipulate, they lack the complexity and diversity needed to recapitulate the physiological reactions of the broad range of human cell types involved in viral pathogenesis [24,25,26].

Pathogenesis and tissue repair mechanisms following respiratory viral infection are species and cell type specific. To dissect these mechanisms, in vitro models need to recapitulate the cell type diversity, cellular interactions, and the extracellular milieu of both the airway and alveolus in vivo. Primary airway epithelial cells (AECs) have been the predominant in vitro model used to elucidate respiratory viral infection pathogenesis, and have been vital in reliably predicting drug responsiveness, such as cystic fibrosis transmembrane conductance regulator (CFTR) modulators prior to clinical trials [27]. However, challenges in obtaining tissue, limited expansion capability ex vivo and poor gene editing potential have restricted their wide-spread application. Furthermore, epithelial-only models do not recapitulate the intricate communication between the epithelium, immune cells, endothelial cells, and stromal cells that ensue following a viral infection and which are central to effective pathogen clearance (Fig. 1). Indeed, the utility of more complex cellular models for understanding respiratory infections has recently been demonstrated using co-cultures of primary airway epithelial cells, pulmonary fibroblasts, endothelial cells and monocytes [28]. Models that mimic the cellular and spatial organisation of the lung will be crucial for discovering and testing new therapeutics, including preventative measures, such as mucosal vaccines.

The discovery of induced pluripotent stem cells (iPSCs) [29] and application of directed differentiation approaches to derive disease-relevant cell types can overcome these limitations. Generated directly from adult somatic cells, iPSCs undergo reprogramming by introducing pluripotency factors [30]. Their self-renewal capacity, ease of access, and potential to differentiate into any desired cell type has enabled the study of lung diseases caused by genetic disorders and/or environmental triggers [31,32,33,34,35,36,37,38,39,40,41,42,43,44]. Of note, iPSCs can be differentiated into lung cell types of interest, including lung airway and alveolar epithelial cells, immune cells, endothelial cells, and fibroblasts, at near limitless scale. Importantly, most of these in vitro derived cell types recapitulate functions of their in vivo counterparts, but with the added benefit of being amenable to genetic manipulation, greatly expanding the accessibility and utility of such models for diverse applications. In addition, iPSC-based platforms would enable all cell types to share a common and consistent genetic background, eliminating variability that is inherent within any system that requires primary material.

The ultimate goal of in vitro lung models is to move beyond traditional observational studies and to gain insights into disease mechanisms by utilising the full suite of molecular and genetic tools available. For example, CRISPR machinery could be deployed in an inducible and cell-type specific manner for high throughput genetic screens to uncover host mechanisms underlying respiratory viral infections [41]. Further, the application of diverse reporters and biosensors offer an enormous diversity of tools to deconstruct mechanisms across space, time, and cell type at unprecedented scale in a physiologically relevant context. In our opinion, combining cell types of interest derived by directed differentiation of iPSCs [45] offers the best path forward for achieving these goals.

In this review, we discuss the development of iPSC-derived lung epithelial cell models and their application to study respiratory viral infections. We also summarise the roles of non-epithelial cell types in respiratory infections as well as their ontogeny in the context of generating iPSC-derived progeny. We draw on this information to propose a roadmap for building a complete iPSC-derived in vitro model of the proximal and distal lung to model existing and emerging respiratory infections, and their interaction with chronic lung diseases.

Fig. 1
figure 1

Summary of the cell types and functions of ideal iPSC-derived in vitro airway and alveolar lung models. The airways and alveoli are distinct compartments with unique epithelial cell types and therefore should be modelled separately. We envisage a modular system whereby the airways or alveoli can be modelled at a homeostatic steady state, as well as an inflamed, infected state for comparison. Our ideal system incorporates stromal and endothelial cells, as well as resident (macrophages and dendritic cell [DC]) and infiltrating (neutrophils and natural kill [NK] cell) immune cells. Coupled with the genetic tractability and scalability afforded by iPSC-derived cell types, the intercellular crosstalk, as well as cell intrinsic processes in physiological contexts can be deconstructed with the complete suite of available molecular and genetic tools

Epithelium

Primary airway epithelial cells (AECs) in vitro have been used extensively to interrogate human respiratory infection mechanisms of pathogenesis in a physiologically relevant context [46, 47]. These protocols expand basal cells from brushings, which can be cryopreserved before undergoing terminal differentiation [48]. To recapitulate the pseudostratified airway epithelium, basal cells are matured in air-liquid interface (ALI) cultures or 3D spherical organoids [49, 50]. Airway epithelial ALI cultures have become a mainstay of respiratory research and applied in diverse contexts from drug testing, particle and vapour exposure, toxicity testing and host pathogen interactions (reviewed [49, 51,52,53,54,55,56])). ALI differentiated cultures are fundamentally distinct from undifferentiated basal cells, illustrated by differential viral growth kinetics and cytokine responses to respiratory virus infection [57, 58]. During infection, respiratory viruses first encounter the apical side of the epithelium, often gaining cellular entry through apical cell surface receptors. This aspect of the viral replication cycle can be recapitulated using ALI cultures, with studies showing that a suite of respiratory viruses preferentially infect via the apical cell surface [59,60,61,62,63,64]. Notably, AEC ALI cultures confirmed the ciliated cell tropism for parainfluenza virus 3 [60], human metapneumovirus [65], human rhinovirus (HRV) [66, 67], SARS-CoV-1 [62], SARS-CoV-2 [68, 69] and RSV [64, 70]. The cell type specific tropism of respiratory viruses also highlights the essential need to validate therapeutic interventions in appropriate models. For example, hydroxychloroquine showed potent inhibition of SARS-CoV-2 infection of Vero E6 cells [71,72,73] but limited efficacy in clinical trials of COVID19 patients [74], likely through TMPRSS2 [75, 76], which is highly expressed in the airway epithelium [77, 78], but absent from Vero E6 cells.

Airway epithelial spheroid cultures are a commonly used alternative to the ALI platform. In this system, basal cells are suspended in matrix scaffold where they divide, subsequently forming spheres containing differentiated airway epithelial cells [79]. As for AEC ALIs, airway organoids have also been used to model respiratory infections (reviewed [80, 81]). Matrix-embedded airway organoids develop an epithelium with the apical surface facing the lumen, which complicates infection models since respiratory viruses infect from the apical side. Therefore, organoids must be removed from the matrix, dissociated for infection, and then re-embedded to reassemble their native topology while infected. Direct comparison of 3D airway organoids versus ALI systems confirmed robust infection at ALI and minimal infection in 3D [82], with the ALI system performing equally or better for evaluation of cellular tropism and fitness [83, 84].

Models using primary alveolar epithelial cells are scarce for two major reasons. Firstly, alveolar epithelial cells are not easily accessible [85]. Secondly, AT1 cells are extremely fragile, and most are destroyed during tissue dissociation and processing. Despite these hurdles, a limited number of models have been developed. Isolated primary AT2s differentiate to AT1 cells in culture with the addition of serum [86,87,88,89] and recently developed defined conditions [90]. Using this system, tropism, replication and inflammatory responses to SARS-CoV-1 and influenza virus in the alveolar epithelium has been explored [91,92,93,94]. Alternatively, AT2s can be maintained in spheroid cultures either in the presence of fibroblast feeder cells [19, 95], or in feeder-free, chemically defined culture conditions [88, 96,97,98,99] to study cellular responses to SARS-CoV-2 [88, 96, 98, 99], although it should be noted that these systems also have the same limitations of spherical airway organoid cultures discussed above.

Primary epithelial cells have inherent restraints, chiefly stemming from difficulties related to their acquisition and to their restricted potential for prolonged growth in vitro [100]. This limitation makes it challenging to obtain cells with a consistent genetic background for ongoing studies or to compare results across different laboratories. Additionally, difficulties in generating clonal populations hinder reliable gene editing of primary airway or alveolar epithelial cells in vitro. Moreover, accessing tissue, especially from patients with rare or debilitating conditions, presents an almost insurmountable barrier to mechanistic research. In contrast, lung epithelial cells derived from induced pluripotent stem cells (iPSCs) or human embryonic stem cells overcome these limitations while offering further advantages.

iPSCs have the capacity to differentiate into any cell type of the body. Directed differentiation of iPSCs in vitro to lung epithelium recapitulates aspects of lung ontogeny, leveraging decades of foundational research on lung development (reviewed [101]). iPSCs are initially specified to definitive endoderm through activin signalling [102, 103] (Fig. 2). Anterior foregut endoderm [104] and primordial lung progenitors expressing NKX2.1 are subsequently derived [42, 105,106,107,108,109]. Enrichment of lung progenitors can be achieved by FACS purification of cells expressing an NKX2.1-fluorescent reporter gene or specific cell surface marker combinations [33, 42]. At this juncture, bipotent lung progenitors can differentiate to airway or alveolar epithelium, depending on the level and duration of WNT signalling (Fig. 2) [34, 109,110,111,112]. Airway basal cells can be further purified through cell sorting, serially passaged, and plated as required at ALI, where they differentiate to a pseudostratified airway epithelium containing multiciliated cells, club, and goblet cells [34, 100, 113,114,115]. Alternatively, WNT activation promotes AT2 cell specification [38, 42, 106, 116,117,118,119,120]. ALI culture of iAT2s induces further maturation by reducing cell cycling and promoting the formation of tight junctions [31, 121]. Recently, iAT1 cells have been derived from iAT2s by manipulating the level of nuclear YAP signalling. iAT1 cells derived by this pathway had a canonical flattened morphology and exhibited increased expression and secretion of alveolar signalling ligands important for mesenchymal and endothelial homeostasis [90].

Fig. 2
figure 2

Schematic of directed differentiation of iPSCs to airway and alveolar epithelium. Differentiation of lung epithelial cells from induced pluripotent stem cell (iPSCs) begins with the induction of definitive endoderm and its subsequent specification to anterior foregut endoderm. Expression of the transcription factor NKX2-1 marks the appearance of bipotent lung progenitor cells, capable of forming both airway epithelial and alveolar epithelial cells. To generate airway epithelial cells, lung progenitors are first differentiated to basal cells that, following enrichment by flow cytometry, can be maintained as spheroids for up to ten passages. Plating these basal cells in air-liquid interface (ALI) cultures with specific differentiation media promotes the formation of a mature pseudostratified epithelium. To generate alveolar epithelial cells, lung progenitors are differentiated to type 2 alveolar epithelial (iAT2) cells using specific growth factors and small molecules. Once formed, iAT2s can also be expanded as 3D spheroids for up to 9 months. When required, iAT2s can be seeded at ALI to induce further maturation. In addition, iAT1 cells can also be generated from iAT2s at ALI using specific media and seeding conditions

iPSC-derived airway and alveolar epithelial models have been deployed to study respiratory viruses, including SARS-CoV-2. In ALI cultures, iPSC-derived AECs infected with SARS-CoV-2 mount a robust interferon response. Within these cultures, multiciliated cells are the primary target of infection, mirroring findings from ALI cultures of primary AECs [122]. Similarly, SARS-CoV-2 infection of iAT2 cells in ALI cultures elicits an epithelial intrinsic inflammatory response and loss of the mature alveolar program, demonstrated by downregulation of surfactant protein genes [123]. In these experiments, virions were observed in lamellar bodies, illustrating the potential importance of this AT2-specific organelle in COVID19 pathogenesis. Moreover, analysis of SARS-CoV-2 infected iAT2 cultures revealed dysregulation of cellular organisation and lipid metabolism. Significantly, these perturbations were not detected in commonly used immortal cell lines, A549, Caco-2 and Vero E6 infected with SARS-CoV-2 [35, 124], highlighting the potentially greater physiological relevance of the iPSC-based model systems.

The scalable nature of iPSC-derived lung epithelial cells makes them an ideal platform for drug screening, demonstrated by recent studies that have used these systems to uncover novel inhibitors of SARS-CoV-2 entry or replication [35, 125, 126]. Furthermore, the genetic tractability of iPSCs has underpinned recent investigations into SARS-CoV-2 entry mechanisms in airway or alveolar epithelial cells [127, 128]. Together these studies highlight that iPSC-derived lung epithelial cells not only provide insights into the biology of their in vivo counterparts, but also offer additional benefits such as scalability and ease of genetic manipulation to interrogate respiratory virus pathogenesis.

Innate immune cells

Viral clearance and infection resolution require a coordinated cellular response. This response is initiated by the infected epithelium and the innate immune cells that are both resident in the lung and rapidly recruited to the site of injury. Achieving a balance between an effective antiviral response versus an exaggerated, damaging immune response is paramount. Below we summarise the importance of various innate immune cells for viral clearance, highlighting any studies focused on the interplay between the human epithelium and immune cells. We also briefly survey iPSC directed differentiation protocols for creating innate immune cells of interest.

Macrophages

Macrophages are responsible for maintaining lung homeostasis, balancing inflammation, promoting regeneration, and countering infections. They are professional phagocytes and antigen presenting cells, absorbing and digesting pathogens and debris, while also initiating adaptive immune responses by presenting antigens to T lymphocytes in tissues [129, 130]. Macrophages are versatile; capable of both producing proinflammatory and chemotactic signals at sites of respiratory infection, as well as producing anti-inflammatory mediators, and fostering tissue repair in the aftermath of injury [131, 132]. Commonly, macrophages are categorised as having two distinct phenotypes; proinflammatory (M1) and anti-inflammatory (M2) [133], although the full spectrum of phenotypes is not captured by these binary classifications [134]. Nevertheless, irrespective of their designation as M1, M2 or something in between, the diverse range of macrophage effector functions can be traced back to microenvironmental stimuli [135].

Studies in mice demonstrate that at birth, tissue resident macrophages in the lung trace their origins back to progenitor cells in the embryonic yolk sac [136,137,138]. While extensive studies have demonstrated the self-renewing characteristics of these embryonically-derived macrophages [139,140,141], it is noteworthy that monocytes originating in the bone marrow play a pivotal role in replenishing the macrophage population in the lung, particularly during periods of infection and inflammation [142,143,144]. Indeed, in healthy aging, the alveolar macrophages in the human lung are replenished entirely from circulating monocytes [145].

Until recently, models employed for human macrophage studies were mostly confined to immortalized cell lines, and peripheral blood monocyte derived macrophages (MDMs) (reviewed in [135]). While MDMs can be readily obtained in large numbers from human donors, they cannot be maintained in culture for extended periods, and inconsistency is introduced by donor variability, owing to different genetics and infection history [137, 138, 141, 146]. In order to overcome the barriers existing in human macrophage studies, increasing efforts have focused on deriving macrophages from iPSCs [135, 147].

Given the complex nature of macrophage ontogeny, it is worth considering how distinct developmental pathways contribute to the various macrophage populations that must confront respiratory viral infections in the adult human (Fig. 3). The first wave of human haematopoiesis occurs in the yolk sac [138], predominately yielding primitive erythroid cells and macrophages [148,149,150], including the resident macrophages in the brain, microglia [137]. The yolk sac is also the site of the second wave of blood cell production, which generates erythromyeloid progenitors (EMPs). EMP derived macrophages seed a number of tissues, including the liver, where they undergo maturation and contribute to the generation of tissue resident macrophages [138, 151]. Definitive haematopoiesis, the third wave, primarily originates in the aorta-gonad-mesonephros region and generates multipotent progenitors, including haematopoietic stem cells (HSCs), capable of giving rise to all the erythroid, myeloid, and lymphoid cell types associated with the adult haematopoietic system. After transiting through the foetal liver [138, 152,153,154], HSCs migrate to the bone marrow, and at birth they become the source of adult haematopoiesis. During adult life, HSCs underpin the continuous production of cells representing all lineages, including monocytes that can further differentiate into MDMs [155,156,157]. In summary, macrophages are ultimately produced in all waves of haematopoiesis, with embryonic haematopoiesis giving rise to the first lung-resident macrophages, while definitive haematopoiesis through derivation of HSCs then monocytes will produce macrophages in later stages of life.

Numerous protocols for generating macrophages from iPSC (iMacs) have been described [135]. In general, protocols involve the stepwise differentiation of iPSCs to mesoderm, haemogenic endothelium, hematopoietic progenitors and then, in the presence of Macrophage colony stimulating factor (MCSF or CSF-1), into iMacs. Current iMac differentiation protocols can be broadly viewed as emulating either primitive or definitive haematopoiesis. Frequently, differentiation protocols produce iMacs that closely resemble those formed from primitive haematopoiesis, characterized by MYB-independent myeloid differentiation [158, 159]. These protocols are independent of WNT signalling, yielding haematopoietic progenitors that lack the expression of HOXA genes [160]. In addition to MCSF, these protocols often include interleukin (IL)-3 [161,162,163,164,165,166], a factor which acts to drive proliferation of progenitor populations. Alternatively, haematopoietic progenitors generated through WNT-dependent pathways more closely resemble those that arise from intra-embryonic definitive haematopoiesis [167, 168]. Interestingly, iMacs generated through primitive versus definitive routes in vitro display functional differences, including intrinsic inflammatory responses and phagocytosis [168], suggesting they may respond differently to specific immunological challenges, including respiratory viral infections.

Efforts to establish co-cultures of human iPSC-derived lung epithelial cells with macrophages has increased in recent years. iPSC-derived alveolar-like organoids can be combined with macrophages through microinjection [169] or forced aggregation [170], with the iMacs partially augmenting responses to inflammatory stimuli in vitro [169]. Prior polarisation of iMacs to M1 or M2 phenotypes influences iPSC-derived lung epithelial responses to SARS-CoV-2 infection [171]. Similar co-culture approaches have been trialled by combining primary airway epithelial cells and MDMs in 3D [172] or ALI [173]. Although the application of these models to significantly advance the study of respiratory viruses is currently in the early stages, these studies highlight the potential to apply co-cultured iPSC-derived lung epithelial cells and macrophages at large scales.

Neutrophils

Neutrophils are the predominant cell type recovered from the airways at the peak of respiratory viral infection [174]. Typically, neutrophils migrate to the lungs via the bloodstream in response to an infection, and attack invading pathogens through production of reactive oxygen species, phagocytosis, degranulation and/or neutrophil extracellular traps [175,176,177,178,179,180,181,182,183,184,185]. While neutrophils dominate the early immune response to respiratory pathogens, these defence mechanisms are often maladaptive, leading to collateral damage to the lungs [176, 181, 182, 186,187,188]. Moreover, recent evidence shows that neutrophilia in the lungs prior to infection actually predisposes to a more severe infection [189]. Given the clear involvement of neutrophils in the early response to respiratory infections, the incorporation of these cells into iPSC-based infectious disease models is vital.

Neutrophils are derived in vivo from HSCs residing in the bone marrow, through differentiation of common myeloid progenitors, into granulocyte-monocyte progenitors then granulocyte progenitors [190, 191] (Fig. 3). Granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor then commits these progenitors to a neutrophil fate [192, 193]. Thus, protocols to differentiate iPSCs into neutrophils attempt to recapitulate this sequence of events. This first involves the derivation of CD34 + haematopoietic progenitors from haemogenic endothelium [194,195,196,197,198]. The subsequent differentiation and expansion of progenitors differs across protocols, but generally relies on culture with stem cell factor, FMS-like tyrosine kinase 3 ligand and/or IL-3 or IL-6 [194,195,196,197,198]. Nevertheless, all protocols conclude with high concentrations of granulocyte colony-stimulating factor to induce neutrophil commitment and expansion [194,195,196,197,198]. iPSC-derived neutrophils display typical characteristics of neutrophils, including morphology and behaviour (e.g., production of reactive oxygen species, formation of neutrophil extracellular traps, phagocytosis, and chemotaxis) [194, 197]. While these iPSC-derived neutrophils have been applied for genetic disease modelling in vitro [196] or as a cellular therapy in mouse models in vivo [195, 198], to date they have not been applied to the study of respiratory infections.

Fig. 3
figure 3

Summary of the haematopoiesis of innate immune cells of interest. Primitive haematopoiesis commences in the yolk sac, giving rise to erythro-myeloid progenitors (EMPs) which can mature into progenitors in the periphery, or through the foetal liver. Definitive haematopoiesis, which includes the production of haematopoietic stem cells (HSCs), can give rise to neutrophils, monocytes (including monocyte-derived dendritic cells [moDCs] and monocyte-derived macrophages), conventional and plasmacytoid dendritic cells (cDC1, cDC2 and pDCs) and natural killer (NK) cells

Dendritic cells

Dendritic cells (DCs) reside both within the epithelium and dispersed through the lung interstitium and play a central role in integrating innate and adaptive immunity. In the mouse lung, the roles of various subsets of DCs during respiratory infections have been extensively described, including CD11b + conventional DCs (cDC), CD103 + DCs, monocyte-derived DCs (moDC) and plasmacytoid DCs (pDCs) [199,200,201]. Similar classification has occurred with respect to human DC populations (CD141 + cDC1, CD1c + cDC2, moDCs and pDCs) [202,203,204,205]; however, studies describing the role of these subsets during infections with human-specific respiratory pathogens are limited.

The primary purpose of DCs is to capture and process foreign antigens [206]. Following this, DCs migrate to local mediastinal lymph nodes to present antigens to T cells which in turn initiate antigen-specific cellular and humoral immune responses. Beyond this role, subsets of conventional DCs potentiate inflammation by releasing cytokines and chemokines [199, 207]. In vitro models have demonstrated that peripheral blood mononuclear cell-derived DCs augment cytokine production and epithelial repair [208, 209]. Moreover, pDCs are essential for antiviral responses, producing vast quantities of type I interferons [210,211,212], and the absence of this cell type impairs responses to respiratory viral infections in mice [213,214,215,216].

In vivo, DC development initiates from HSCs in the bone marrow. HSCs differentiate to common myeloid progenitors, macrophage-DC progenitors, then common DC progenitors, prior to commitment to either pDCs or pre-cDCs (reviewed in [217] (Fig. 3). Pre-cDCs migrate into the periphery where they differentiate to either cDC1 or cDC2s. pDCs can also arise from common lymphoid progenitors [218] (Fig. 3). Finally, moDCs arise in the periphery from monocytes, which can be derived from monocyte-DC progenitor lineages or granulocyte-macrophage progenitor lineages (see Fig. 3).

Several directed differentiation approaches to generate cDCs and moDCs from iPSCs have been developed [219,220,221,222]. To recapitulate development, protocols differentiate iPSCs to haematopoietic progenitors, then use IL-4 and granulocyte-macrophage colony-stimulating factor to specify to cDCs [219]. In some cases, Notch signalling is important for differentiation of DCs [221, 223]. iPSC-derived cDCs display phagocytic, T cell stimulatory and cytokine-producing properties [219, 221, 224]. The generation of pDCs has been less well described; however, the exclusion of IL-4 during the differentiation time course appears to be important [222]. Finally, some applications have successfully derived iPSCs from peripheral blood DCs, in an effort to then increase efficiency of iPSC-derived DC differentiation potential and maturation [224, 225]. The intricate ontogeny of DCs is poorly captured by these directed-differentiation approaches, and further work will be necessary to create precise subsets of DCs to build a lung model that is useful to interrogate respiratory viral infections.

Natural killer cells

Natural Killer (NK) cells are innate immune cells that play a key role in antitumour and antiviral defence [226]. While NK cells in the human lung parenchyma are mainly CD56dim CD16+, indicative of a mature phenotype, few express the tissue residency marker CD69 [227], suggesting the lung is populated by circulating NK cells from the blood, rather than a stable pool of tissue-resident NK cells. During respiratory infections, NK cells constrain viral infection by killing infected cells, either through their direct lysis, and/or through the activation of cell death receptors on target cells (reviewed in [228]). NK cells also shape the immune response through production of cytokines, in particular IFN-γ. Genetic deficiencies that influence NK cell numbers or function are associated with the severity of viral infections in the lung [229]. Furthermore, severe lower respiratory viral infections are associated with low NK cell numbers [230, 231], indicating the important role these cells play in the lung’s response to infection.

In the bone marrow, HSCs give rise to common lymphoid progenitors that can differentiate into NK cells, which undergo further maturation in the bone marrow and/or secondary lymphoid tissues [232,233,234,235] (Fig. 3). Thus, to recapitulate this development in vitro, iPSC-derived NK (iNK) cell differentiation protocols first derive haematopoietic progenitors [236,237,238,239,240]. Several approaches have been employed to generate iNK cells from iPSCs, including methods that include the use of stromal cell lines [240, 241] or protocols that rely on the formation of embryoid bodies in defined media conditions [236, 239, 242]. IL-15 is an essential cytokine for NK cell differentiation [233] and is employed in all directed-differentiation protocols reported to date. iNK cells express canonical markers including CD56, NKG2D, NKp44, NKp46, CD16, and Killer Ig-like receptors, and display cytotoxic activity against tumour cells [240, 243]. At present, we are unaware of any studies using iNK cells to enhance understanding of respiratory viral infections. Given the capacity of NK cells to detect and kill virally infected cells, examining the interaction between iNK cells and infected epithelial cells promises to be an exciting area of future research.

Future iPSC-derived immune cells to consider

As highlighted so far, very few studies have utilised models that have incorporated iPSC-derived immune cells with epithelial cells to interrogate disease mechanisms underlying respiratory infections. Where these models have been tested, work has focused on innate immune cell types which are relatively simple to generate in vitro, such as macrophages. However, for some innate immune lineages, robust iPSC differentiation protocols have not been developed (e.g., basophils). Moreover, it is well recognised that adaptive immune cell types also play important roles in the response to respiratory viral infections. The incorporation of these cell types into an in vitro model of lung infections faces both technical and logistical hurdles. For cells of the adaptive immune system, generation of cells representing these lineages only makes sense when the specificity of their antigen receptors can be pre-determined. Although there has been some success in creating iPSC derived CD8 + Cytotoxic T cells with T cell receptors of known specificity [244], the generation of antigen specific CD4 + T cells or antibody producing B cells is still a major challenge. Ideally, the availability of T cells with T cell receptors that recognise specific viral antigens would enable the construction of complex in vitro cultures that could be used to model respiratory viral infections. The continued improvement of protocols generating various innate and adaptive immune cells from iPSCs will allow future research questions surrounding the involvement of these cells in respiratory viral infections to be answered.

Endothelium

Endothelial dysfunction occurs during respiratory infections

The pulmonary endothelium has a critical role in gas exchange, transport of solutes and fluid in the lung, vascular tone, coagulation, and immune cell trafficking [245]. Following a respiratory infection, the endothelium is an active player in coordinating immune responses. For instance, the endothelium is crucial in regulating immune cell entry from the bloodstream into the airspace [246, 247]. Cytokines activate endothelial cells to adopt a leaky, proinflammatory phenotype [248], and endothelial cells are themselves a source of cytokines [249]. Moreover, the activated pulmonary endothelium releases reactive oxygen species and reactive nitrogen species to augment leukocyte chemotaxis (e.g., neutrophils) [250]. Following infections, endothelial cells also adopt a pro-coagulation phenotype to limit damage and restrict infection to the lung [251] (see Table 1).

Endothelial cells play a predominant role in the pathobiology of disease during severe respiratory infections, such as acute respiratory distress syndrome. As these infections progress, the endothelial barrier is disrupted due to cytokines, reactive oxygen species, histamine, or mechanical stretch. Moreover, respiratory pathogens themselves can directly damage endothelial cells [252, 253]. This loss of the barrier and increased permeability causes pulmonary oedema, due to movement of fluid into the interstitial space [245], causing dyspnoea. Given the importance of the pulmonary endothelium in response and repair following respiratory infection, the incorporation of endothelial cells into human lung models of infection is likely to be very important.

iPSC-derived endothelial cells

Pulmonary endothelial cells exhibit extensive heterogeneity based on anatomical location and functional properties, including barrier properties, proliferation, adhesion receptor expression and leukocyte trafficking [254,255,256,257,258,259]. Endothelial cells in the lung arise through angiogenesis (growth of existing vessels) and vasculogenesis (new formation of vessels) [260,261,262]. During embryogenesis, the pulmonary endothelium arises from splanchnic mesoderm [263], while lymphatic endothelial cells arise from paraxial mesoderm [264]. Lineage tracing studies in mice have uncovered further heterogeneity in pulmonary endothelial cell specification, with cardiopulmonary progenitors giving rise to smooth muscle and proximal endothelial cells, but not distal capillary endothelial cells which arise from earlier endothelium [265]. The developmental complexity of endothelial cell specification in the lung is important when considering iPSC-derived endothelial cells to study respiratory viral infections.

There are many approaches for generating endothelial cells from iPSCs, including directed differentiation protocols that allow derivation of arterial or venous endothelial cells [266,267,268,269]. In general, a limitation is that iPSC-derived endothelial cells remain limited in their ability to form microvasculature [270], although advances in 3D culture may start to address this [271]. While there are no protocols for creating pulmonary patterned endothelial cells, iPSC-derived endothelial cells seeded into decellularized whole lung scaffolds adopt markers of the endogenous endothelium [272]. These studies suggest the adoption of a pulmonary endothelial fate may require the co-culture of iPSC-derived endothelial cells in a “lung promoting” environment, which may also include airway or alveolar epithelial cells. While iPSC-derived airway epithelium matures in the presence of primary pulmonary microvascular cells [273], the effect of the epithelium on iPSC-derived endothelial cells has not been explored. Notwithstanding these challenges, “generic” iPSC-derived endothelial cells have been successfully applied to understand the pathogenesis of lung diseases, such as pulmonary arterial hypertension in vitro [274,275,276,277,278].

Stroma

Fibrotic scarring follows severe respiratory infections

Fibroblasts reside in the lung interstitium at low frequency. In response to injury, fibroblasts are activated, leading to an increase in their numbers. Fibroblast activation, which is thought to promote tissue repair, is typically driven by signals from adjacent injured alveolar epithelial cells. Recent studies in mice have shown that during acute respiratory infection, subsets of fibroblasts adopt interferon-responsive, proinflammatory or pro-repair phenotypes [279]. Similarly, fibroblast-derived ECM proteases remodel the lung microenvironment following infection [279, 280]. While this new role for fibroblasts during acute infection is emerging, considerably more is known about their role during wound repair.

Pulmonary fibrosis can develop following severe or persistent damage to the lung induced by respiratory infections, such as acute respiratory distress syndrome and pneumonia [281, 282]. Studies of patients with severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome, both immediately following infection and 15 years later, noted impaired lung function and radiographic findings consistent with pulmonary fibrosis [283, 284]. A similar scenario is unfolding in the wake of the COVID19 pandemic, with pulmonary fibrosis a common feature of Post-Acute Sequelae of COVID-19, also termed “long COVID” [285,286,287]. Post-mortem examinations of post-COVID19 lungs have revealed hallmark abnormalities such as alveolar epithelial cell dysfunction, fibroblastic foci, alterations in the extracellular matrix, and immune cell infiltration [287]. While these studies demonstrate the association between pulmonary fibrosis and prior severe respiratory infections, the molecular mechanisms linking these two events has not been established, highlighting the need for further mechanistic studies.

iPSC-derived stromal cells

Lung fibroblasts can be classified based on spatial localization, or function. For instance, fibroblasts can be broadly characterized as adventitial fibroblasts, residing proximally, and alveolar fibroblasts, residing in the distal region of the lung. These spatially distinct populations display distinct characteristics, such as differential expression of ECM proteins [279, 288, 289]. Alternatively, functional heterogeneity enables lung fibroblasts to be classified as either myofibroblasts, lipofibroblasts or matrix fibroblasts. Myofibroblasts have contractile properties, are important in wound repair, and play pathological roles in pulmonary fibrosis [290]. Lipofibroblasts reside in close proximity to, and provide surfactant lipids to AT2s [291]. Matrix fibroblasts generate the lung ECM [292]. During injury, there is evidence that fibroblasts can convert between lipofibroblast and myofibroblast states, demonstrating the plasticity of fibroblast identities [293].

Methods for the derivation of tissue-specific mesenchyme from iPSCs have lagged behind other lung lineages. A recent study by Alber et al. described the generation of the first mouse iPSC-derived lung mesenchyme that express key transcriptional and functional features of in vivo mesenchyme [294]. Co-culture of this lung mesenchyme with iPSC-derived epithelial cells upregulated gene signatures associated with alveolar and adventitial fibroblasts. Moreover, the authors observed this cross talk was bidirectional with epithelial fate influenced too [294]. Similarly, iPSC-derived (non-tissue specific) mesenchyme supports the development of iPSC-derived alveolar epithelial cells [295]. These early forays into generating iPSC derived lung stromal cells represent an important step to recreating the diversity of lung stromal populations. In this context, further work will be required to understand the nature of human iPSC-derived lung mesenchymal cells, particularly how these relate to specific fibroblast subtypes.

Applications for iPSC-derived models

In establishing iPSC-derived models of the lung, several practical and technical points need to be considered. Practically, working with iPSCs is time-consuming, expensive and requires tacit knowledge through experience. This reality is likely to place such models beyond the reach of research groups whose work is primarily focused on the biology of lung cells, rather than how these cells are made. Technically, differentiation protocols for many of the cell types required to construct a complex lung model are still a work in progress. Directed differentiation protocols for proximal and distal lung epithelium have matured considerably in recent years; however, the occasional emergence of non-lung endoderm cells can occur [33, 296, 297]. Furthermore, rare cell types such as tuft cells, pulmonary neuroendocrine cells and pulmonary ionocytes have limited representation in iPSC-derived AEC models, although recent successes have occurred in this area [298,299,300]. For the alveolar compartment, co-culture of iAT1 and iAT2 cells, as well as the introduction of mechanical forces [301], which occur during breathing, represents an opportunity to establish more physiologically relevant models. Finally, directed differentiation protocols for some cell types that might be required to complete a proposed model (e.g., stromal cells), are a headwind in the near term; however, this will improve over time as protocols evolve.

In constructing a “mini lung” model (Fig. 4), the optimal combination of immune, stromal, endothelial, and epithelial cells will depend on the research question under consideration, whether that involves specific pathogens or genetic diseases. Likewise, the optimisation of medium composition, extracellular matrix, cell seeding sequence, and cell density will likely be required to enable “mini lung” systems to adequately model different human pathologies. Ideally, advances in cryopreservation methods for each iPSC-derived cell type will allow an “off-the-shelf” approach, freeing researchers from the challenge of coordinating multiple directed differentiation outcomes. We foresee that the technical challenges for establishing iPSC-based platforms will be far outweighed by the applications.

Fig. 4
figure 4

Idealised workflow to create iPSC-derived “mini lungs”. Proposed models will build on existing induced pluripotent stem cell (iPSC) derived airway epithelial cell (iAEC) and type 2 alveolar epithelial cell (iAT2) air-liquid interface (ALI) cultures. Distinct directed differentiation of mesodermal lineages, including endothelial cells, haematopoietic cells, and fibroblasts would be performed, prior to incorporation with epithelial cells to mimic the homeostatic or infected airway or alveolus

The role of host factors during viral infection are cell type specific, precluding simplistic or one size fits all approaches to characterising virus-host interactions. High-throughput pooled genetic screens are an attractive methodology [302] to probe these complex interactions, enabling cellular genes crucial to the viral replication cycle to be identified in an unbiased fashion. Cognisance of relevant cell types is also important, as it was shown that key genes enabling SARS-CoV-2 infection could vary greatly between cell types [303]. In this context, determining the optimal frequency of different cell types within a complex mini lung model will be a necessary prelude to using these models for drug discovery. Notwithstanding these challenges, iPSC-derived respiratory models will provide an ideal system for physiologically relevant, high-throughput screens for lung diseases (reviewed [304]).

Another application where iPSC-derived lung models show great promise is in modelling genetic diseases, including cystic fibrosis, primary ciliary dyskinesia, alpha-1 antitrypsin deficiency, and interstitial lung disease. Donor-derived iPSCs can be created, and disease-causing mutations corrected in vitro to generate isogenic control lines which are critical for assessing the impact of genetic variants. For instance, pairs of iPSC lines differing only by the presence of a specific disease causing mutation have been generated for cystic fibrosis [34, 305, 306], primary ciliary dyskinesia [34, 307], alpha-1 antitrypsin deficiency [308, 309] and childhood interstitial lung disease [32, 43, 310]. The availability of these paired iPSC lines is critical for mechanistic studies and drug screening protocols, which prior to the advent of iPSC technologies, were challenging because of limitations in obtaining and manipulating primary cells. iPSC-derived models can circumvent these limitations as they enable complex genetic modifications, including the generation of isogenic controls, and provide the scalability required for mechanistic studies and high-throughput screens.

Conclusions

Respiratory infections inflict an enormous toll in terms of mortality, morbidity, and economic loss, yet comparatively few antiviral drugs, vaccines or therapies exist for these common infections and their associated diseases. The relative dearth of effective therapeutics is due, in part, to the lack of tractable in vitro systems that enable incorporation of relevant human cell types that accurately model the behaviour of their in vivo counterparts. Building models of the lung with iPSC-derived progeny overcomes the limitations of immortalised cells and primary cell systems while retaining the advantages of both, including scalability, ease of gene editing and physiological complexity. Additionally, iPSC-derived models provide an accessible platform for the study of genetic and chronic diseases, and how specific human cell types interact with drugs, toxins, and importantly, respiratory infections. Robust protocols now exist to create iPSC-derived lung epithelial cells which faithfully recapitulate the biology of both the airway and alveolar epithelium. The next step will be to combine these with non-epithelial cell types, such as stromal, endothelial, and immune cells, to create complex “mini lung” models that better reflect the true cellular diversity of the lung. The generation of such complex “mini lung” models has the potential to expand our understanding of how cellular interactions shape the response to viral infections and usher in a new era for respiratory research and therapeutic development.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

AEC:

Airway epithelial cell

ALI:

Air–liquid interface

AT1:

Type 1 alveolar epithelial cell

AT2:

Type 2 alveolar epithelial cell

cDC:

Conventional dendritic cell

CFTR:

Cystic fibrosis transmembrane conductance regulator

CoV:

Coronavirus

DC:

Dendritic cell

ECM:

Extracellular matrix

EMP:

Erythromyeloid progenitor

hESC:

Human embryonic stem cell

HRV:

Human rhinovirus

HSC:

Haematopoietic stem cell

iAT1:

iPSC–derived type 1 alveolar epithelial cell

iAT2:

iPSC–derived type 2 alveolar epithelial cell

iBC:

iPSC–derived basal cell

IL:

Interleukin

iMac:

iPSC–derived macrophage

iPSC:

Induced pluripotent stem cell

M-CSF:

Macrophage colony–stimulating factor

MDM:

Monocyte–derived macrophage

moDC:

Monocyte–derived dendritic cell

NK:

Natural killer

pDC:

Plasmacytoid dendritic cell

RSV:

Respiratory syncytial virus

SARS:

Severe acute respiratory syndrome

References

  1. World Health Organisation. WHO methods and data sources for global burden of disease estimates 2000–2019. Department of Data and Analytics Division of Data AaDfIW, Geneva; 2020. Contract No.: WHO/ DDI/DNA/GHE/2020.3.

  2. Thompson WW, Shay DK, Weintraub E, Brammer L, Cox N, Anderson LJ, et al. Mortality associated with influenza and respiratory syncytial virus in the United States. JAMA. 2003;289(2):179–86.

    Article  Google Scholar 

  3. Li Y, Wang X, Blau DM, Caballero MT, Feikin DR, Gill CJ, et al. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in children younger than 5 years in 2019: a systematic analysis. Lancet. 2022;399(10340):2047–64.

    Article  Google Scholar 

  4. Sigurs N, Aljassim F, Kjellman B, Robinson PD, Sigurbergsson F, Bjarnason R, et al. Asthma and allergy patterns over 18 years after severe RSV bronchiolitis in the first year of life. Thorax. 2010;65(12):1045–52.

    Article  Google Scholar 

  5. Holt PG, Sly PD. Viral infections and atopy in asthma pathogenesis: new rationales for asthma prevention and treatment. Nat Med. 2012;18(5):726–35.

    Article  CAS  Google Scholar 

  6. Johnston SL, Pattemore PK, Sanderson G, Smith S, Lampe F, Josephs L, et al. Community study of role of viral infections in exacerbations of asthma in 9–11 year old children. BMJ. 1995;310(6989):1225–9.

    Article  CAS  Google Scholar 

  7. Papadopoulos NG, Christodoulou I, Rohde G, Agache I, Almqvist C, Bruno A, et al. Viruses and bacteria in acute asthma exacerbations–a GA(2) LEN-DARE systematic review. Allergy. 2011;66(4):458–68.

    Article  CAS  Google Scholar 

  8. Papi A, Bellettato CM, Braccioni F, Romagnoli M, Casolari P, Caramori G, et al. Infections and airway inflammation in chronic obstructive pulmonary disease severe exacerbations. Am J Respir Crit Care Med. 2006;173(10):1114–21.

    Article  Google Scholar 

  9. Moriyama M, Hugentobler WJ, Iwasaki A. Seasonality of respiratory viral infections. Annual Rev Virol. 2020;7(1):83–101.

    Article  CAS  Google Scholar 

  10. Zepp JA, Morrisey EE. Cellular crosstalk in the development and regeneration of the respiratory system. Nat Rev Mol Cell Biol. 2019;20(9):551–66.

    Article  CAS  Google Scholar 

  11. Noguchi M, Furukawa KT, Morimoto M. Pulmonary neuroendocrine cells: physiology, tissue homeostasis and disease. Dis Model Mech. 2020;13(12).

  12. Strine MS, Wilen CB. Tuft cells are key mediators of interkingdom interactions at mucosal barrier surfaces. PLoS Pathog. 2022;18(3):e1010318.

    Article  CAS  Google Scholar 

  13. Plasschaert LW, Žilionis R, Choo-Wing R, Savova V, Knehr J, Roma G, et al. A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature. 2018;560(7718):377–81.

    Article  CAS  Google Scholar 

  14. Shah VS, Chivukula RR, Lin B, Waghray A, Rajagopal J. Cystic fibrosis and the cells of the airway epithelium: what are ionocytes and what do they do? Annu Rev Pathol. 2022;17(1):23–46.

    Article  CAS  Google Scholar 

  15. Ahn JH, Kim J, Hong SP, Choi SY, Yang MJ, Ju YS et al. Nasal ciliated cells are primary targets for SARS-CoV-2 replication in the early stage of COVID-19. J Clin Invest. 2021;131(13).

  16. Sungnak W, Huang N, Bécavin C, Berg M, Queen R, Litvinukova M, et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med. 2020;26(5):681–7.

    Article  CAS  Google Scholar 

  17. Pieter SH, Paul B, McCray B Jr. The innate immune function of airway epithelial cells in inflammatory lung disease. Eur Respir J. 2015;45(4):1150.

    Article  Google Scholar 

  18. Mason RJ, Williams MC. Type II alveolar cell. Defender of the alveolus. Am Rev Respir Dis. 1977;115(6 Pt 2):81–91.

    CAS  Google Scholar 

  19. Barkauskas CE, Cronce MJ, Rackley CR, Bowie EJ, Keene DR, Stripp BR, et al. Type 2 alveolar cells are stem cells in adult lung. J Clin Invest. 2013;123(7):3025–36.

    Article  CAS  Google Scholar 

  20. Toulmin SA, Bhadiadra C, Paris AJ, Lin JH, Katzen J, Basil MC, et al. Type II alveolar cell MHCII improves respiratory viral disease outcomes while exhibiting limited antigen presentation. Nat Commun. 2021;12(1):3993.

    Article  CAS  Google Scholar 

  21. Ibricevic A, Pekosz A, Walter MJ, Newby C, Battaile JT, Brown EG, et al. Influenza virus receptor specificity and cell tropism in mouse and human airway epithelial cells. J Virol. 2006;80(15):7469–80.

    Article  CAS  Google Scholar 

  22. Rock JR, Randell SH, Hogan BL. Airway basal stem cells: a perspective on their roles in epithelial homeostasis and remodeling. Dis Models Mech. 2010;3(9–10):545–56.

    Article  CAS  Google Scholar 

  23. Takaki H, Oshiumi H, Shingai M, Matsumoto M, Seya T. Development of mouse models for analysis of human virus infections. Microbiol Immunol. 2017;61(3–4):107–13.

    Article  CAS  Google Scholar 

  24. Kanagaki S, Suezawa T, Moriguchi K, Nakao K, Toyomoto M, Yamamoto Y, et al. Hydroxypropyl cyclodextrin improves amiodarone-induced aberrant lipid homeostasis of alveolar cells. Am J Respir Cell Mol Biol. 2021;64(4):504–14.

    Article  CAS  Google Scholar 

  25. Corbière V, Dirix V, Norrenberg S, Cappello M, Remmelink M, Mascart F. Phenotypic characteristics of human type II alveolar epithelial cells suitable for antigen presentation to T lymphocytes. Respir Res. 2011;12(1):15.

    Article  Google Scholar 

  26. Martens K, Hellings PW, Steelant B. Calu-3 epithelial cells exhibit different immune and epithelial barrier responses from freshly isolated primary nasal epithelial cells in vitro. Clin Transl Allergy. 2018;8:40.

    Article  Google Scholar 

  27. McCarthy C, Brewington JJ, Harkness B, Clancy JP, Trapnell BC. Personalised CFTR pharmacotherapeutic response testing and therapy of cystic fibrosis. Eur Respir J. 2018;51(6):1702457.

    Article  Google Scholar 

  28. Roe MM, Do T, Turner S, Jevitt AM, Chlebicz M, White K, et al. Blood myeloid cells differentiate to lung resident cells and respond to pathogen stimuli in a 3D human tissue-engineered lung model. Front Bioeng Biotechnol. 2023;11:1212230.

    Article  Google Scholar 

  29. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–72.

    Article  CAS  Google Scholar 

  30. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76.

    Article  CAS  Google Scholar 

  31. Abo KM, Sainz de Aja J, Lindstrom-Vautrin J, Alysandratos KD, Richards A, Garcia-de-Alba C et al. Air-liquid interface culture promotes maturation and allows environmental exposure of pluripotent stem cell-derived alveolar epithelium. JCI Insight. 2022;7(6).

  32. Alysandratos KD, Russo SJ, Petcherski A, Taddeo EP, Acin-Perez R, Villacorta-Martin C, et al. Patient-specific iPSCs carrying an SFTPC mutation reveal the intrinsic alveolar epithelial dysfunction at the inception of interstitial lung disease. Cell Rep. 2021;36(9):109636.

    Article  CAS  Google Scholar 

  33. Hawkins F, Kramer P, Jacob A, Driver I, Thomas DC, McCauley KB, et al. Prospective isolation of NKX2-1-expressing human lung progenitors derived from pluripotent stem cells. J Clin Invest. 2017;127(6):2277–94.

    Article  Google Scholar 

  34. Hawkins FJ, Suzuki S, Beermann ML, Barillà C, Wang R, Villacorta-Martin C, et al. Derivation of airway basal stem cells from human pluripotent stem cells. Cell Stem Cell. 2021;28(1):79–e958.

    Article  CAS  Google Scholar 

  35. Hekman RM, Hume AJ, Goel RK, Abo KM, Huang J, Blum BC, et al. Actionable cytopathogenic host responses of human alveolar type 2 cells to SARS-CoV-2. Mol Cell. 2020;80(6):1104–e229.

    Article  CAS  Google Scholar 

  36. Huang J, Hume AJ, Abo KM, Werder RB, Villacorta-Martin C, Alysandratos KD, et al. SARS-CoV-2 infection of pluripotent stem cell-derived human lung alveolar type 2 cells elicits a rapid epithelial-intrinsic inflammatory response. Cell Stem Cell. 2020;27(6):962–73. e7.

    Article  CAS  Google Scholar 

  37. Jacob A, Morley M, Hawkins F, McCauley KB, Jean JC, Heins H, et al. Differentiation of human pluripotent stem cells into functional lung alveolar epithelial cells. Cell Stem Cell. 2017;21(4):472–88. e10.

    Article  CAS  Google Scholar 

  38. Jacob A, Vedaie M, Roberts DA, Thomas DC, Villacorta-Martin C, Alysandratos KD, et al. Derivation of self-renewing lung alveolar epithelial type II cells from human pluripotent stem cells. Nat Protoc. 2019;14(12):3303–32.

    Article  CAS  Google Scholar 

  39. McCauley KB, Hawkins F, Serra M, Thomas DC, Jacob A, Kotton DN. Efficient derivation of functional human airway epithelium from pluripotent stem cells via temporal regulation of wnt signaling. Cell Stem Cell. 2017;20(6):844–57. e6.

    Article  CAS  Google Scholar 

  40. Werder RB, Berthiaume KA, Merritt C, Gallagher M, Villacorta-Martin C, Wang F, et al. The COPD GWAS gene ADGRG6 instructs function and injury response in human iPSC-derived type II alveolar epithelial cells. Am J Hum Genet. 2023.

  41. Werder RB, Liu T, Abo KM, Lindstrom-Vautrin J, Villacorta-Martin C, Huang J, et al. CRISPR interference interrogation of COPD GWAS genes reveals the functional significance of desmoplakin in iPSC-derived alveolar epithelial cells. Sci Adv. 2022;8(28):eabo6566.

    Article  CAS  Google Scholar 

  42. Gotoh S, Ito I, Nagasaki T, Yamamoto Y, Konishi S, Korogi Y, et al. Generation of alveolar epithelial spheroids via isolated progenitor cells from human pluripotent stem cells. Stem Cell Rep. 2014;3(3):394–403.

    Article  CAS  Google Scholar 

  43. Korogi Y, Gotoh S, Ikeo S, Yamamoto Y, Sone N, Tamai K, et al. In vitro disease modeling of Hermansky-Pudlak syndrome type 2 using human induced pluripotent stem cell-derived alveolar organoids. Stem Cell Rep. 2019.

  44. Suezawa T, Kanagaki S, Korogi Y, Nakao K, Hirai T, Murakami K, et al. Modeling of lung phenotype of Hermansky-Pudlak syndrome type I using patient-specific iPSCs. Respir Res. 2021;22(1):284.

    Article  CAS  Google Scholar 

  45. Eicher AK, Kechele DO, Sundaram N, Berns HM, Poling HM, Haines LE, et al. Functional human gastrointestinal organoids can be engineered from three primary germ layers derived separately from pluripotent stem cells. Cell Stem Cell. 2022;29(1):36–e516.

    Article  CAS  Google Scholar 

  46. Lechner JF, Haugen A, McClendon IA, Pettis EW. Clonal growth of normal adult human bronchial epithelial cells in a serum-free medium. Vitro. 1982;18(7):633–42.

    Article  CAS  Google Scholar 

  47. Barrett LA, McDowell EM, Frank AL, Harris CC, Trump BF. Long-term organ culture of human bronchial epithelium. Cancer Res. 1976;36(3):1003–10.

    CAS  Google Scholar 

  48. Fulcher ML, Randell SH. Human nasal and tracheo-bronchial respiratory epithelial cell culture. Methods Mol Biol. 2013;945:109–21.

    Article  Google Scholar 

  49. Cao X, Coyle JP, Xiong R, Wang Y, Heflich RH, Ren B, et al. Invited review: human air-liquid-interface organotypic airway tissue models derived from primary tracheobronchial epithelial cells-overview and perspectives. Vitro Cell Dev Biol Anim. 2021;57(2):104–32.

    Article  Google Scholar 

  50. Müller L, Brighton LE, Carson JL, Fischer WA 2nd, Jaspers I. Culturing of human nasal epithelial cells at the air liquid interface. J Vis Exp. 2013(80).

  51. Bérubé K, Prytherch Z, Job C, Hughes T. Human primary bronchial lung cell constructs: the new respiratory models. Toxicology. 2010;278(3):311–8.

    Article  Google Scholar 

  52. Lacroix G, Koch W, Ritter D, Gutleb AC, Larsen ST, Loret T, et al. Air–liquid interface in vitro models for respiratory toxicology research: consensus workshop and recommendations. Appl Vitro Toxicol. 2018;4(2):91–106.

    Article  Google Scholar 

  53. Rijsbergen LC, van Dijk LLA, Engel MFM, de Vries RD, de Swart RL. In vitro modelling of respiratory virus infections in human airway epithelial cells - a systematic review. Front Immunol. 2021;12:683002.

    Article  CAS  Google Scholar 

  54. Denney L, Ho L-P. The role of respiratory epithelium in host defence against influenza virus infection. Biomedical J. 2018;41(4):218–33.

    Article  Google Scholar 

  55. Ryu G, Shin HW. SARS-CoV-2 infection of airway epithelial cells. Immune Netw. 2021;21(1):e3.

    Article  Google Scholar 

  56. Bridges JP, Vladar EK, Huang H, Mason RJ. Respiratory epithelial cell responses to SARS-CoV-2 in COVID-19. Thorax. 2022;77(2):203–9.

    Article  Google Scholar 

  57. Chan RW, Yuen KM, Yu WC, Ho CC, Nicholls JM, Peiris JS, et al. Influenza H5N1 and H1N1 virus replication and innate immune responses in bronchial epithelial cells are influenced by the state of differentiation. PLoS ONE. 2010;5(1):e8713.

    Article  Google Scholar 

  58. Jia HP, Look DC, Shi L, Hickey M, Pewe L, Netland J, et al. ACE2 receptor expression and severe acute respiratory syndrome coronavirus infection depend on differentiation of human airway epithelia. J Virol. 2005;79(23):14614–21.

    Article  CAS  Google Scholar 

  59. Slepushkin VA, Staber PD, Wang G, McCray PB Jr., Davidson BL. Infection of human airway epithelia with h1N1, H2N2, and H3N2 influenza a virus strains. Mol Ther. 2001;3(3):395–402.

    Article  CAS  Google Scholar 

  60. Zhang L, Bukreyev A, Thompson CI, Watson B, Peeples ME, Collins PL, et al. Infection of ciliated cells by human parainfluenza virus type 3 in an in vitro model of human airway epithelium. J Virol. 2005;79(2):1113–24.

    Article  CAS  Google Scholar 

  61. Palermo LM, Porotto M, Yokoyama CC, Palmer SG, Mungall BA, Greengard O, et al. Human parainfluenza virus infection of the airway epithelium: viral hemagglutinin-neuraminidase regulates fusion protein activation and modulates infectivity. J Virol. 2009;83(13):6900–8.

    Article  CAS  Google Scholar 

  62. Sims AC, Baric RS, Yount B, Burkett SE, Collins PL, Pickles RJ. Severe acute respiratory syndrome coronavirus infection of human ciliated airway epithelia: role of ciliated cells in viral spread in the conducting airways of the lungs. J Virol. 2005;79(24):15511–24.

    Article  CAS  Google Scholar 

  63. Klimyte EM, Smith SE, Oreste P, Lembo D, Dutch RE. Inhibition of human metapneumovirus binding to heparan sulfate blocks infection in human lung cells and airway tissues. J Virol. 2016;90(20):9237–50.

    Article  CAS  Google Scholar 

  64. Zhang L, Peeples ME, Boucher RC, Collins PL, Pickles RJ. Respiratory syncytial virus infection of human airway epithelial cells is polarized, specific to ciliated cells, and without obvious cytopathology. J Virol. 2002;76(11):5654–66.

    Article  CAS  Google Scholar 

  65. de Graaf M, Herfst S, Aarbiou J, Burgers PC, Zaaraoui-Boutahar F, Bijl M, et al. Small hydrophobic protein of human metapneumovirus does not affect virus replication and host gene expression in vitro. PLoS ONE. 2013;8(3):e58572.

    Article  Google Scholar 

  66. Griggs TF, Bochkov YA, Basnet S, Pasic TR, Brockman-Schneider RA, Palmenberg AC, et al. Rhinovirus C targets ciliated airway epithelial cells. Respir Res. 2017;18(1):84.

    Article  Google Scholar 

  67. Jakiela B, Gielicz A, Plutecka H, Hubalewska-Mazgaj M, Mastalerz L, Bochenek G, et al. Th2-type cytokine–induced mucus metaplasia decreases susceptibility of human bronchial epithelium to Rhinovirus infection. Am J Respir Cell Mol Biol. 2014;51(2):229–41.

    Article  Google Scholar 

  68. Hou YJ, Okuda K, Edwards CE, Martinez DR, Asakura T, Dinnon KH 3, et al. SARS-CoV-2 reverse genetics reveals a variable infection gradient in the respiratory tract. Cell. 2020;182(2):429–e4614.

    Article  CAS  Google Scholar 

  69. Fiege JK, Thiede JM, Nanda HA, Matchett WE, Moore PJ, Montanari NR, et al. Single cell resolution of SARS-CoV-2 tropism, antiviral responses, and susceptibility to therapies in primary human airway epithelium. PLoS Pathog. 2021;17(1):e1009292.

    Article  CAS  Google Scholar 

  70. Guo-Parke H, Canning P, Douglas I, Villenave R, Heaney LG, Coyle PV, et al. Relative respiratory syncytial virus cytopathogenesis in upper and lower respiratory tract epithelium. Am J Respir Crit Care Med. 2013;188(7):842–51.

    Article  Google Scholar 

  71. Wang M, Cao R, Zhang L, Yang X, Liu J, Xu M, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30(3):269–71.

    Article  CAS  Google Scholar 

  72. Yao X, Ye F, Zhang M, Cui C, Huang B, Niu P, et al. In vitro antiviral activity and projection of optimized dosing design of hydroxychloroquine for the treatment of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Clin Infect Dis. 2020;71(15):732–9.

    Article  CAS  Google Scholar 

  73. Liu J, Cao R, Xu M, Wang X, Zhang H, Hu H, et al. Hydroxychloroquine, a less toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro. Cell Discov. 2020;6(1):16.

    Article  CAS  Google Scholar 

  74. Axfors C, Schmitt AM, Janiaud P, van’t Hooft J, Abd-Elsalam S, Abdo EF, et al. Mortality outcomes with hydroxychloroquine and chloroquine in COVID-19 from an international collaborative meta-analysis of randomized trials. Nat Commun. 2021;12(1):2349.

    Article  CAS  Google Scholar 

  75. Ou T, Mou H, Zhang L, Ojha A, Choe H, Farzan M. Hydroxychloroquine-mediated inhibition of SARS-CoV-2 entry is attenuated by TMPRSS2. PLoS Pathog. 2021;17(1):e1009212.

    Article  CAS  Google Scholar 

  76. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181(2):271–e808.

    Article  CAS  Google Scholar 

  77. Sajuthi SP, DeFord P, Li Y, Jackson ND, Montgomery MT, Everman JL, et al. Type 2 and interferon inflammation regulate SARS-CoV-2 entry factor expression in the airway epithelium. Nat Commun. 2020;11(1):5139.

    Article  CAS  Google Scholar 

  78. Lukassen S, Chua RL, Trefzer T, Kahn NC, Schneider MA, Muley T, et al. SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells. Embo j. 2020;39(10):e105114.

    Article  CAS  Google Scholar 

  79. Rock JR, Onaitis MW, Rawlins EL, Lu Y, Clark CP, Xue Y, et al. Basal cells as stem cells of the mouse trachea and human airway epithelium. Proc Natl Acad Sci U S A. 2009;106(31):12771–5.

    Article  CAS  Google Scholar 

  80. Kim J, Koo B-K, Clevers H. Organoid studies in COVID-19 research. Int J Stem Cells. 2022;15(1):3–13.

    Article  CAS  Google Scholar 

  81. van der Vaart J, Clevers H. Airway organoids as models of human disease. J Intern Med. 2021;289(5):604–13.

    Article  Google Scholar 

  82. Sano E, Suzuki T, Hashimoto R, Itoh Y, Sakamoto A, Sakai Y, et al. Cell response analysis in SARS-CoV-2 infected bronchial organoids. Commun Biol. 2022;5(1):516.

    Article  CAS  Google Scholar 

  83. Zhou J, Li C, Sachs N, Chiu MC, Wong BH, Chu H, et al. Differentiated human airway organoids to assess infectivity of emerging influenza virus. Proc Natl Acad Sci U S A. 2018;115(26):6822–7.

    Article  CAS  Google Scholar 

  84. Chiu MC, Li C, Liu X, Yu Y, Huang J, Wan Z, et al. A bipotential organoid model of respiratory epithelium recapitulates high infectivity of SARS-CoV-2 Omicron variant. Cell Discov. 2022;8(1):57.

    Article  CAS  Google Scholar 

  85. Gonzalez RF, Allen L, Gonzales L, Ballard PL, Dobbs LG. HTII-280, a biomarker specific to the apical plasma membrane of human lung alveolar type II cells. J Histochem Cytochem. 2010;58(10):891–901.

    Article  CAS  Google Scholar 

  86. Beers MF, Moodley Y. When is an alveolar type 2 cell an alveolar type 2 cell? A Conundrum for Lung Stem Cell Biology and Regenerative Medicine. Am J Respir Cell Mol Biol. 2017;57(1):18–27.

    Article  CAS  Google Scholar 

  87. Wang J, Edeen K, Manzer R, Chang Y, Wang S, Chen X, et al. Differentiated human alveolar epithelial cells and reversibility of their phenotype in vitro. Am J Respir Cell Mol Biol. 2007;36(6):661–8.

    Article  CAS  Google Scholar 

  88. Katsura H, Sontake V, Tata A, Kobayashi Y, Edwards CE, Heaton BE, et al. Human lung stem cell-based alveolospheres provide insights into SARS-CoV-2-mediated interferon responses and pneumocyte dysfunction. Cell Stem Cell. 2020;27(6):890–e9048.

    Article  CAS  Google Scholar 

  89. Ghaedi M, Calle EA, Mendez JJ, Gard AL, Balestrini J, Booth A, et al. Human iPS cell-derived alveolar epithelium repopulates lung extracellular matrix. J Clin Invest. 2013;123(11):4950–62.

    Article  CAS  Google Scholar 

  90. Burgess CL, Huang J, Bawa PS, Alysandratos K-D, Minakin K, Ayers LJ et al. Generation of human alveolar epithelial type I cells from pluripotent stem cells. Cell Stem Cell. 2024.

  91. Mossel EC, Wang J, Jeffers S, Edeen KE, Wang S, Cosgrove GP, et al. SARS-CoV replicates in primary human alveolar type II cell cultures but not in type I-like cells. Virology. 2008;372(1):127–35.

    Article  CAS  Google Scholar 

  92. Qian Z, Travanty EA, Oko L, Edeen K, Berglund A, Wang J, et al. Innate immune response of human alveolar type II cells infected with severe acute respiratory syndrome-coronavirus. Am J Respir Cell Mol Biol. 2013;48(6):742–8.

    Article  CAS  Google Scholar 

  93. Wang J, Oberley-Deegan R, Wang S, Nikrad M, Funk CJ, Hartshorn KL, et al. Differentiated human alveolar type II cells secrete antiviral IL-29 (IFN-lambda 1) in response to influenza A infection. J Immunol. 2009;182(3):1296–304.

    Article  CAS  Google Scholar 

  94. Wang J, Nikrad MP, Phang T, Gao B, Alford T, Ito Y, et al. Innate immune response to influenza a virus in differentiated human alveolar type II cells. Am J Respir Cell Mol Biol. 2011;45(3):582–91.

    Article  CAS  Google Scholar 

  95. Sucre JMS, Jetter CS, Loomans H, Williams J, Plosa EJ, Benjamin JT, et al. Successful establishment of primary type II alveolar epithelium with 3D organotypic coculture. Am J Respir Cell Mol Biol. 2018;59(2):158–66.

    Article  CAS  Google Scholar 

  96. Youk J, Kim T, Evans KV, Jeong YI, Hur Y, Hong SP, et al. Three-dimensional human alveolar stem cell culture models reveal infection response to SARS-CoV-2. Cell Stem Cell. 2020;27(6):905–e1910.

    Article  CAS  Google Scholar 

  97. Salahudeen AA, Choi SS, Rustagi A, Zhu J, van Unen V. Progenitor identification and SARS-CoV-2 infection in human distal lung organoids. Nature. 2020;588(7839):670–5.

    Article  CAS  Google Scholar 

  98. Ebisudani T, Sugimoto S, Haga K, Mitsuishi A, Takai-Todaka R, Fujii M, et al. Direct derivation of human alveolospheres for SARS-CoV-2 infection modeling and drug screening. Cell Rep. 2021;35(10):109218.

    Article  CAS  Google Scholar 

  99. Mulay A, Konda B, Garcia G Jr., Yao C, Beil S, Villalba JM, et al. SARS-CoV-2 infection of primary human lung epithelium for COVID-19 modeling and drug discovery. Cell Rep. 2021;35(5):109055.

    Article  CAS  Google Scholar 

  100. Mou H, Vinarsky V, Tata PR, Brazauskas K, Choi SH, Crooke AK, et al. Dual SMAD signaling inhibition enables long-term expansion of diverse epithelial basal cells. Cell Stem Cell. 2016;19(2):217–31.

    Article  CAS  Google Scholar 

  101. Herriges M, Morrisey EE. Lung development: orchestrating the generation and regeneration of a complex organ. Development. 2014;141(3):502–13.

    Article  CAS  Google Scholar 

  102. Loh KM, Ang LT, Zhang J, Kumar V, Ang J, Auyeong JQ, et al. Efficient endoderm induction from human pluripotent stem cells by logically directing signals controlling lineage bifurcations. Cell Stem Cell. 2014;14(2):237–52.

    Article  CAS  Google Scholar 

  103. Kubo A, Shinozaki K, Shannon JM, Kouskoff V, Kennedy M, Woo S, et al. Development of definitive endoderm from embryonic stem cells in culture. Development. 2004;131(7):1651–62.

    Article  CAS  Google Scholar 

  104. Green MD, Chen A, Nostro MC, d’Souza SL, Schaniel C, Lemischka IR, et al. Generation of anterior foregut endoderm from human embryonic and induced pluripotent stem cells. Nat Biotechnol. 2011;29(3):267–72.

    Article  CAS  Google Scholar 

  105. Rankin SA, Han L, McCracken KW, Kenny AP, Anglin CT, Grigg EA, et al. A retinoic acid-hedgehog cascade coordinates mesoderm-inducing signals and endoderm competence during lung specification. Cell Rep. 2016;16(1):66–78.

    Article  CAS  Google Scholar 

  106. Huang SX, Islam MN, O’Neill J, Hu Z, Yang YG, Chen YW, et al. Efficient generation of lung and airway epithelial cells from human pluripotent stem cells. Nat Biotechnol. 2014;32(1):84–91.

    Article  CAS  Google Scholar 

  107. Serra M, Alysandratos KD, Hawkins F, McCauley KB, Jacob A, Choi J, et al. Pluripotent stem cell differentiation reveals distinct developmental pathways regulating lung- versus thyroid-lineage specification. Development. 2017;144(21):3879–93.

    CAS  Google Scholar 

  108. Ikonomou L, Herriges MJ, Lewandowski SL, Marsland R 3rd, Villacorta-Martin C, Caballero IS, et al. The in vivo genetic program of murine primordial lung epithelial progenitors. Nat Commun. 2020;11(1):635.

    Article  CAS  Google Scholar 

  109. Yamamoto Y, Korogi Y, Hirai T, Gotoh S. A method of generating alveolar organoids using human pluripotent stem cells. Methods Cell Biol. 2020;159:115–41.

    Article  CAS  Google Scholar 

  110. McCauley KB, Hawkins F, Serra M, Thomas DC, Jacob A, Kotton DN. Efficient derivation of functional human airway epithelium from pluripotent stem cells via temporal regulation of wnt signaling. Cell Stem Cell. 2017;20(6):844–e576.

    Article  CAS  Google Scholar 

  111. McCauley KB, Hawkins F, Kotton DN. Derivation of epithelial-only airway organoids from human pluripotent stem cells. Curr Protoc Stem Cell Biol. 2018;45(1):e51.

    Article  Google Scholar 

  112. Konishi S, Gotoh S, Tateishi K, Yamamoto Y, Korogi Y, Nagasaki T, et al. Directed induction of functional multi-ciliated cells in proximal airway epithelial spheroids from human pluripotent stem cells. Stem Cell Rep. 2016;6(1):18–25.

    Article  CAS  Google Scholar 

  113. Tadokoro T, Gao X, Hong CC, Hotten D, Hogan BL. BMP signaling and cellular dynamics during regeneration of airway epithelium from basal progenitors. Development. 2016;143(5):764–73.

    CAS  Google Scholar 

  114. Miller AJ, Yu Q, Czerwinski M, Tsai YH, Conway RF, Wu A, et al. In vitro and in vivo development of the human airway at single-cell resolution. Dev Cell. 2020;53(1):117–e286.

    Article  CAS  Google Scholar 

  115. Suzuki S, Hawkins FJ, Barillà C, Beermann ML, Kotton DN, Davis BR. Differentiation of human pluripotent stem cells into functional airway basal stem cells. STAR Protocols. 2021;2(3):100683.

    Article  Google Scholar 

  116. Gonzales LW, Guttentag SH, Wade KC, Postle AD, Ballard PL. Differentiation of human pulmonary type II cells in vitro by glucocorticoid plus cAMP. Am J Physiology-Lung Cell Mol Physiol. 2002;283(5):L940–51.

    Article  CAS  Google Scholar 

  117. Jacob A, Morley M, Hawkins F, McCauley KB, Jean JC, Heins H, et al. Differentiation of human pluripotent stem cells into functional lung alveolar epithelial cells. Cell Stem Cell. 2017;21(4):472–e8810.

    Article  CAS  Google Scholar 

  118. Yamamoto Y, Gotoh S, Korogi Y, Seki M, Konishi S, Ikeo S, et al. Long-term expansion of alveolar stem cells derived from human iPS cells in organoids. Nat Methods. 2017;14(11):1097–106.

    Article  CAS  Google Scholar 

  119. Hein RFC, Conchola AS, Fine AS, Xiao Z, Frum T, Brastrom LK et al. Stable iPSC-derived NKX2-1 + lung bud tip progenitor organoids give rise to airway and alveolar cell types. Development. 2022;149(20).

  120. Huang SX, Green MD, de Carvalho AT, Mumau M, Chen YW, D’Souza SL, et al. The in vitro generation of lung and airway progenitor cells from human pluripotent stem cells. Nat Protoc. 2015;10(3):413–25.

    Article  CAS  Google Scholar 

  121. Schruf E, Schroeder V, Le HQ, Schönberger T, Raedel D, Stewart EL, et al. Recapitulating idiopathic pulmonary fibrosis related alveolar epithelial dysfunction in a human iPSC-derived air-liquid interface model. FASEB J. 2020;34(6):7825–46.

    Article  CAS  Google Scholar 

  122. Wang R, Hume AJ, Beermann ML, Simone-Roach C, Lindstrom-Vautrin J, Le Suer J, et al. Human airway lineages derived from pluripotent stem cells reveal the epithelial responses to SARS-CoV-2 infection. Am J Physiol Lung Cell Mol Physiol. 2022;322(3):L462–78.

    Article  CAS  Google Scholar 

  123. Huang J, Hume AJ, Abo KM, Werder RB, Villacorta-Martin C, Alysandratos KD, et al. SARS-CoV-2 infection of pluripotent stem cell-derived human lung alveolar type 2 cells elicits a rapid epithelial-intrinsic inflammatory response. Cell Stem Cell. 2020;27(6):962–e737.

    Article  CAS  Google Scholar 

  124. Pei R, Feng J, Zhang Y, Sun H, Li L, Yang X, et al. Host metabolism dysregulation and cell tropism identification in human airway and alveolar organoids upon SARS-CoV-2 infection. Protein Cell. 2021;12(9):717–33.

    Article  CAS  Google Scholar 

  125. Han Y, Duan X, Yang L, Nilsson-Payant BE, Wang P, Duan F, et al. Identification of SARS-CoV-2 inhibitors using lung and colonic organoids. Nature. 2021;589(7841):270–5.

    Article  CAS  Google Scholar 

  126. Samuel RM, Majd H, Richter MN, Ghazizadeh Z, Zekavat SM, Navickas A, et al. Androgen signaling regulates SARS-CoV-2 receptor levels and is associated with severe COVID-19 symptoms in men. Cell Stem Cell. 2020;27(6):876–e8912.

    Article  CAS  Google Scholar 

  127. Niwa R, Sakai K, Lung MSY, Matsumoto T, Mikawa R, Maehana S, et al. ACE2 knockout hinders SARS-CoV-2 propagation in iPS cell-derived airway and alveolar epithelial cells. Front Cell Dev Biol. 2023;11:1290876.

    Article  Google Scholar 

  128. Dobrindt K, Hoagland DA, Seah C, Kassim B, O’Shea CP, Murphy A, et al. Common genetic variation in humans impacts in vitro susceptibility to SARS-CoV-2 infection. Stem Cell Rep. 2021;16(3):505–18.

    Article  CAS  Google Scholar 

  129. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496(7446):445–55.

    Article  CAS  Google Scholar 

  130. Weiss G, Schaible UE. Macrophage defense mechanisms against intracellular bacteria. Immunol Rev. 2015;264(1):182–203.

    Article  CAS  Google Scholar 

  131. Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M. Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol. 2013;229(2):176–85.

    Article  CAS  Google Scholar 

  132. Watanabe S, Alexander M, Misharin AV, Budinger GS. The role of macrophages in the resolution of inflammation. J Clin Investig. 2019;129(7):2619–28.

    Article  Google Scholar 

  133. Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM. M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol. 2000;164(12):6166–73.

    Article  CAS  Google Scholar 

  134. Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity. 2014;40(2):274–88.

    Article  CAS  Google Scholar 

  135. Lyadova I, Gerasimova T, Nenasheva T. Macrophages derived from human induced pluripotent stem cells: the diversity of protocols, future prospects, and outstanding questions. Front Cell Dev Biology. 2021;9:640703.

    Article  Google Scholar 

  136. Schulz C, Perdiguero EG, Chorro L, Szabo-Rogers H, Cagnard N, Kierdorf K, et al. A lineage of myeloid cells independent of myb and hematopoietic stem cells. Science. 2012;336(6077):86–90.

    Article  CAS  Google Scholar 

  137. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010;330(6005):841–5.

    Article  CAS  Google Scholar 

  138. Hoeffel G, Chen J, Lavin Y, Low D, Almeida FF, See P, et al. C-Myb + erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity. 2015;42(4):665–78.

    Article  CAS  Google Scholar 

  139. Guilliams M, De Kleer I, Henri S, Post S, Vanhoutte L, De Prijck S, et al. Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J Exp Med. 2013;210(10):1977–92.

    Article  CAS  Google Scholar 

  140. Yona S, Kim K-W, Wolf Y, Mildner A, Varol D, Breker M, et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity. 2013;38(1):79–91.

    Article  CAS  Google Scholar 

  141. Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf M, et al. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity. 2013;38(4):792–804.

    Article  CAS  Google Scholar 

  142. Coillard A, Segura E. Vivo differentiation of human monocytes. Front Immunol. 2019;10:1907.

    Article  CAS  Google Scholar 

  143. Hume DA, Irvine KM, Pridans C. The mononuclear phagocyte system: the relationship between monocytes and macrophages. Trends Immunol. 2019;40(2):98–112.

    Article  CAS  Google Scholar 

  144. Epelman S, Lavine KJ, Beaudin AE, Sojka DK, Carrero JA, Calderon B, et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity. 2014;40(1):91–104.

    Article  CAS  Google Scholar 

  145. Byrne AJ, Powell JE, O’Sullivan BJ, Ogger PP, Hoffland A, Cook J et al. Dynamics of human monocytes and airway macrophages during healthy aging and after transplant. J Exp Med. 2020;217(3).

  146. Klichinsky M, Ruella M, Shestova O, Lu XM, Best A, Zeeman M, et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol. 2020;38(8):947–53.

    Article  CAS  Google Scholar 

  147. Li S, Song L, Zhang Y, Zhan Z, Yang Y, Yu L et al. Optimizing the method for differentiation of macrophages from human induced pluripotent stem cells. Stem Cells Int. 2022;2022.

  148. Palis J, Yoder MC. Yolk-sac hematopoiesis: the first blood cells of mouse and man. Exp Hematol. 2001;29(8):927–36.

    Article  CAS  Google Scholar 

  149. Lacaud G, Kouskoff V. Hemangioblast, hemogenic endothelium, and primitive versus definitive hematopoiesis. Exp Hematol. 2017;49:19–24.

    Article  Google Scholar 

  150. Ivanovs A, Rybtsov S, Ng ES, Stanley EG, Elefanty AG, Medvinsky A. Human haematopoietic stem cell development: from the embryo to the dish. Development. 2017;144(13):2323–37.

    Article  CAS  Google Scholar 

  151. Frame JM, McGrath KE, Palis J. Erythro-myeloid progenitors:definitive hematopoiesis in the conceptus prior to the emergence of hematopoietic stem cells. Blood Cells Mol Dis. 2013;51(4):220–5.

    Article  Google Scholar 

  152. Kumaravelu P, Hook L, Morrison AM, Ure J, Zhao S, Zuyev S et al. Quantitative developmental anatomy of definitive haematopoietic stem cells/long-term repopulating units (HSC/RUs): role of the aorta-gonad-mesonephros (AGM) region and the yolk sac in colonisation of the mouse embryonic liver. Development. 2002;129(21):4891-9.

  153. Boisset J-C, van Cappellen W, Andrieu-Soler C, Galjart N, Dzierzak E, Robin C. In vivo imaging of haematopoietic cells emerging from the mouse aortic endothelium. Nature. 2010;464(7285):116–20.

    Article  CAS  Google Scholar 

  154. Imanirad P. Hematopoietic progenitor and stem cell regulation during development: hypoxia and niches. Blood Cells Mol Dis. 2013;51(4):256-63.

  155. Cummings M, Peters AD, Whitehead GF, Menon BR, Micklefield J, Webb SJ, et al. Assembling a plug-and-play production line for combinatorial biosynthesis of aromatic polyketides in Escherichia coli. PLoS Biol. 2019;17(7):e3000347.

    Article  CAS  Google Scholar 

  156. Palis J, Robertson S, Kennedy M, Wall C, Keller G. Development of erythroid and myeloid progenitors in the yolk sac and embryo proper of the mouse. Development. 1999;126(22):5073–84.

    Article  CAS  Google Scholar 

  157. Laurenti E, Göttgens B. From haematopoietic stem cells to complex differentiation landscapes. Nature. 2018;553(7689):418–26.

    Article  CAS  Google Scholar 

  158. Cao X, Yakala GK, van den Hil FE, Cochrane A, Mummery CL, Orlova VV. Differentiation and functional comparison of monocytes and macrophages from hiPSCs with peripheral blood derivatives. Stem cell Rep. 2019;12(6):1282–97.

    Article  CAS  Google Scholar 

  159. Lee CZ, Kozaki T, Ginhoux F. Studying tissue macrophages in vitro: are iPSC-derived cells the answer? Nat Rev Immunol. 2018;18(11):716–25.

    Article  CAS  Google Scholar 

  160. Sturgeon CM, Ditadi A, Awong G, Kennedy M, Keller G. Wnt signaling controls the specification of definitive and primitive hematopoiesis from human pluripotent stem cells. Nat Biotechnol. 2014;32(6):554–61.

    Article  CAS  Google Scholar 

  161. Karlsson KR, Cowley S, Martinez FO, Shaw M, Minger SL, James W. Homogeneous monocytes and macrophages from human embryonic stem cells following coculture-free differentiation in M-CSF and IL-3. Exp Hematol. 2008;36(9):1167–75.

    Article  CAS  Google Scholar 

  162. Wilgenburg Bv, Browne C, Vowles J, Cowley SA. Efficient, long term production of monocyte-derived macrophages from human pluripotent stem cells under partly-defined and fully-defined conditions. PLoS ONE. 2013;8(8):e71098.

    Article  Google Scholar 

  163. Vanhee S, De Mulder K, Van Caeneghem Y, Verstichel G, Van Roy N, Menten B, et al. In vitro human embryonic stem cell hematopoiesis mimics MYB-independent yolk sac hematopoiesis. Haematologica. 2015;100(2):157–66.

    Article  CAS  Google Scholar 

  164. Klimchenko O, Di Stefano A, Geoerger B, Hamidi S, Opolon P, Robert T, et al. Monocytic cells derived from human embryonic stem cells and fetal liver share common differentiation pathways and homeostatic functions. Blood. 2011;117(11):3065–75.

    Article  CAS  Google Scholar 

  165. Lopez-Yrigoyen M, May A, Ventura T, Taylor H, Fidanza A, Cassetta L, et al. Production and characterization of human macrophages from pluripotent stem cells. J Vis Exp. 2020;158:e61038.

    Google Scholar 

  166. Takata K, Kozaki T, Lee CZW, Thion MS, Otsuka M, Lim S, et al. Induced-pluripotent-stem-cell-derived primitive macrophages provide a platform for modeling tissue-resident macrophage differentiation and function. Immunity. 2017;47(1):183–98. e6.

    Article  CAS  Google Scholar 

  167. Ng ES, Azzola L, Bruveris FF, Calvanese V, Phipson B, Vlahos K, et al. Differentiation of human embryonic stem cells to HOXA + hemogenic vasculature that resembles the aorta-gonad-mesonephros. Nat Biotechnol. 2016;34(11):1168–79.

    Article  CAS  Google Scholar 

  168. Bredemeyer AL, Amrute JM, Koenig AL, Idol RA, He L, Luff SA, et al. Derivation of extra-embryonic and intra-embryonic macrophage lineages from human pluripotent stem cells. Development. 2022;149(8):dev200016.

    Article  CAS  Google Scholar 

  169. Seo HR, Han HJ, Lee Y, Noh YW, Cho SJ, Kim JH. Human pluripotent stem cell-derived alveolar organoid with macrophages. Int J Mol Sci. 2022;23(16).

  170. Heo H-R, Hong S-H. Generation of macrophage containing alveolar organoids derived from human pluripotent stem cells for pulmonary fibrosis modeling and drug efficacy testing. Cell Biosci. 2021;11(1):216.

    Article  CAS  Google Scholar 

  171. Lian Q, Zhang K, Zhang Z, Duan F, Guo L, Luo W, et al. Differential effects of macrophage subtypes on SARS-CoV-2 infection in a human pluripotent stem cell-derived model. Nat Commun. 2022;13(1):2028.

    Article  CAS  Google Scholar 

  172. Roe MM, Do T, Turner S, Jevitt AM, Chlebicz M, White K et al. Blood myeloid cells differentiate to lung resident cells and respond to pathogen stimuli in a 3D human tissue-engineered lung model. Front Bioeng Biotechnol. 2023;11.

  173. Barreto-Duran E, Szczepański A, Gałuszka-Bulaga A, Surmiak M, Siedlar M, Sanak M, et al. The interplay between the airway epithelium and tissue macrophages during the SARS-CoV-2 infection. Front Immunol. 2022;13:991991.

    Article  CAS  Google Scholar 

  174. McNamara PS, Ritson P, Selby A, Hart CA, Smyth RL. Bronchoalveolar lavage cellularity in infants with severe respiratory syncytial virus bronchiolitis. Arch Dis Child. 2003;88(10):922–6.

    Article  CAS  Google Scholar 

  175. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303(5663):1532–5.

    Article  CAS  Google Scholar 

  176. Narasaraju T, Yang E, Samy RP, Ng HH, Poh WP, Liew A-A, et al. Excessive neutrophils and neutrophil extracellular traps contribute to acute lung injury of influenza pneumonitis. Am J Pathol. 2011;179(1):199–210.

    Article  CAS  Google Scholar 

  177. Toussaint M, Jackson DJ, Swieboda D, Guedan A, Tsourouktsoglou T-D, Ching YM et al. Host DNA released by NETosis promotes rhinovirus-induced type-2 allergic asthma exacerbation. Nat Med. 2017;advance online publication.

  178. Moorthy AN, Tan KB, Wang S, Narasaraju T, Chow VT. Effect of high-fat diet on the formation of pulmonary neutrophil extracellular traps during influenza pneumonia in BALB/c mice. Front Immunol. 2016;7:289.

    Article  Google Scholar 

  179. Eum S-Y, Kong J-H, Hong M-S, Lee Y-J, Kim J-H, Hwang S-H, et al. Neutrophils are the predominant infected phagocytic cells in the airways of patients with active pulmonary TB. Chest. 2010;137(1):122–8.

    Article  Google Scholar 

  180. Hashimoto Y, Moki T, Takizawa T, Shiratsuchi A, Nakanishi Y. Evidence for phagocytosis of influenza virus-infected, apoptotic cells by neutrophils and macrophages in mice. J Immunol. 2007;178(4):2448–57.

    Article  CAS  Google Scholar 

  181. Soehnlein O, Oehmcke S, Rothfuchs AG, Frithiof R, Rooijen Nv, Mörgelin M, et al. Neutrophil degranulation mediates severe lung damage triggered by streptococcal M1 protein. Eur Respir J. 2008;32(2):405–12.

    Article  CAS  Google Scholar 

  182. Hoenderdos K, Lodge KM, Hirst RA, Chen C, Palazzo SG, Emerenciana A, et al. Hypoxia upregulates neutrophil degranulation and potential for tissue injury. Thorax. 2016;71(11):1030–8.

    Article  Google Scholar 

  183. Effah CY, Drokow EK, Agboyibor C, Ding L, He S, Liu S, et al. Neutrophil-dependent immunity during pulmonary infections and inflammations. Front Immunol. 2021;12:689866.

    Article  CAS  Google Scholar 

  184. Mizgerd JP. Molecular mechanisms of neutrophil recruitment elicited by bacteria in the lungs. Semin Immunol. 2002;14(2):123–32.

    Article  CAS  Google Scholar 

  185. Radermecker C, Detrembleur N, Guiot J, Cavalier E, Henket M, d’Emal C et al. Neutrophil extracellular traps infiltrate the lung airway, interstitial, and vascular compartments in severe COVID-19. J Exp Med. 2020;217(12).

  186. Lefrançais E, Mallavia B, Zhuo H, Calfee CS, Looney MR. Maladaptive role of neutrophil extracellular traps in pathogen-induced lung injury. JCI Insight. 2018;3(3).

  187. Zhu L, Liu L, Zhang Y, Pu L, Liu J, Li X, et al. High level of Neutrophil Extracellular traps correlates with poor prognosis of severe influenza a infection. J Infect Dis. 2018;217(3):428–37.

    Article  CAS  Google Scholar 

  188. Curren B, Ahmed T, Howard DR, Ashik Ullah M, Sebina I, Rashid RB, et al. IL-33-induced neutrophilic inflammation and NETosis underlie rhinovirus-triggered exacerbations of asthma. Mucosal Immunol. 2023;16(5):671–84.

    Article  Google Scholar 

  189. Habibi MS, Thwaites RS, Chang M, Jozwik A, Paras A, Kirsebom F et al. Neutrophilic inflammation in the respiratory mucosa predisposes to RSV infection. Science. 2020;370(6513).

  190. Görgens A, Radtke S, Möllmann M, Cross M, Dürig J, Horn PA, et al. Revision of the human hematopoietic tree: granulocyte subtypes derive from distinct hematopoietic lineages. Cell Rep. 2013;3(5):1539–52.

    Article  Google Scholar 

  191. von Vietinghoff S, Ley K. Homeostatic regulation of blood neutrophil counts. J Immunol. 2008;181(8):5183–8.

    Article  Google Scholar 

  192. Liu F, Wu HY, Wesselschmidt R, Kornaga T, Link DC. Impaired production and increased apoptosis of neutrophils in granulocyte colony-stimulating factor receptor-deficient mice. Immunity. 1996;5(5):491–501.

    Article  CAS  Google Scholar 

  193. Lieschke GJ, Grail D, Hodgson G, Metcalf D, Stanley E, Cheers C, et al. Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization. Blood. 1994;84(6):1737–46.

    Article  CAS  Google Scholar 

  194. Sweeney CL, Merling RK, Choi U, Priel DL, Kuhns DB, Wang H, et al. Generation of functionally mature neutrophils from induced pluripotent stem cells. Methods Mol Biol. 2014;1124:189–206.

    Article  Google Scholar 

  195. Miyauchi M, Ito Y, Nakahara F, Hino T, Nakamura F, Iwasaki Y, et al. Efficient production of human neutrophils from iPSCs that prevent murine lethal infection with immune cell recruitment. Blood. 2021;138(24):2555–69.

    Article  CAS  Google Scholar 

  196. Tsui M, Min W, Ng S, Dobbs K, Notarangelo LD, Dror Y, et al. The use of induced pluripotent stem cells to study the effects of adenosine deaminase deficiency on human neutrophil development. Front Immunol. 2021;12:748519.

    Article  CAS  Google Scholar 

  197. Brok-Volchanskaya VS, Bennin DA, Suknuntha K, Klemm LC, Huttenlocher A, Slukvin I. Effective and rapid generation of functional neutrophils from induced pluripotent stem cells using ETV2-modified mRNA. Stem cell Rep. 2019;13(6):1099–110.

    Article  CAS  Google Scholar 

  198. Trump LR, Nayak RC, Singh AK, Emberesh S, Wellendorf AM, Lutzko CM, et al. Neutrophils derived from genetically modified human induced pluripotent stem cells circulate and phagocytose bacteria in vivo. Stem Cells Transl Med. 2019;8(6):557–67.

    Article  CAS  Google Scholar 

  199. Neyt K, Lambrecht BN. The role of lung dendritic cell subsets in immunity to respiratory viruses. Immunol Rev. 2013;255(1):57–67.

    Article  Google Scholar 

  200. Guilliams M, Lambrecht BN, Hammad H. Division of labor between lung dendritic cells and macrophages in the defense against pulmonary infections. Mucosal Immunol. 2013.

  201. Plantinga M, Guilliams M, Vanheerswynghels M, Deswarte K, Branco-Madeira F, Toussaint W, et al. Conventional and monocyte-derived CD11b + dendritic cells initiate and maintain T helper 2 cell-mediated immunity to house dust mite allergen. Immunity. 2013;38(2):322–35.

    Article  CAS  Google Scholar 

  202. Collin M, Bigley V. Human dendritic cell subsets: an update. Immunology. 2018;154(1):3–20.

    Article  CAS  Google Scholar 

  203. Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, Miltenyi S, et al. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. J Immunol. 2000;165(11):6037–46.

    Article  CAS  Google Scholar 

  204. MacDonald KP, Munster DJ, Clark GJ, Dzionek A, Schmitz J, Hart DN. Characterization of human blood dendritic cell subsets. Blood. 2002;100(13):4512–20.

    Article  CAS  Google Scholar 

  205. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, et al. Nomenclature of monocytes and dendritic cells in blood. Blood. 2010;116(16):e74–80.

    Article  CAS  Google Scholar 

  206. Steinman RM. The dendritic cell system and its role in immunogenicity. Annu Rev Immunol. 1991;9:271–96.

    Article  CAS  Google Scholar 

  207. Luber CA, Cox J, Lauterbach H, Fancke B, Selbach M, Tschopp J, et al. Quantitative proteomics reveals subset-specific viral recognition in dendritic cells. Immunity. 2010;32(2):279–89.

    Article  CAS  Google Scholar 

  208. Nguyen Hoang AT, Chen P, Juarez J, Sachamitr P, Billing B, Bosnjak L, et al. Dendritic cell functional properties in a three-dimensional tissue model of human lung mucosa. Am J Physiol Lung Cell Mol Physiol. 2012;302(2):L226–37.

    Article  Google Scholar 

  209. Harrington H, Cato P, Salazar F, Wilkinson M, Knox A, Haycock JW, et al. Immunocompetent 3D model of human upper airway for disease modeling and in vitro drug evaluation. Mol Pharm. 2014;11(7):2082–91.

    Article  CAS  Google Scholar 

  210. Colonna M, Trinchieri G, Liu Y-J. Plasmacytoid dendritic cells in immunity. Nat Immunol. 2004;5(12):1219–26.

    Article  CAS  Google Scholar 

  211. Chehimi J, Starr SE, Kawashima H, Miller DS, Trinchieri G, Perussia B, et al. Dendritic cells and IFN-alpha-producing cells are two functionally distinct non-B, non-monocytic HLA-DR + cell subsets in human peripheral blood. Immunology. 1989;68(4):486–90.

    CAS  Google Scholar 

  212. Cella M, Jarrossay D, Facchetti F, Alebardi O, Nakajima H, Lanzavecchia A, et al. Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon. Nat Med. 1999;5(8):919–23.

    Article  CAS  Google Scholar 

  213. Lynch JP, Werder RB, Loh Z, Sikder MAA, Curren B, Zhang V, et al. Plasmacytoid dendritic cells protect from viral bronchiolitis and asthma through semaphorin 4a-mediated T reg expansion. J Exp Med. 2018;215(2):537–57.

    Article  CAS  Google Scholar 

  214. Swiecki M, Gilfillan S, Vermi W, Wang Y, Colonna M. Plasmacytoid dendritic cell ablation impacts early interferon responses and antiviral NK and CD8(+) T cell accrual. Immunity. 2010;33(6):955–66.

    Article  CAS  Google Scholar 

  215. Davidson S, Kaiko G, Loh Z, Lalwani A, Zhang V, Spann K, et al. Plasmacytoid dendritic cells promote host defense against acute pneumovirus infection via the TLR7-MyD88-dependent signaling pathway. J Immunol. 2011;186(10):5938–48.

    Article  CAS  Google Scholar 

  216. Wang H, Peters N, Schwarze J. Plasmacytoid dendritic cells limit viral replication, pulmonary inflammation, and airway hyperresponsiveness in respiratory syncytial virus infection. J Immunol. 2006;177(9):6263–70.

    Article  CAS  Google Scholar 

  217. Merad M, Sathe P, Helft J, Miller J, Mortha A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol. 2013;31:563-604.

  218. Dress RJ, Dutertre CA, Giladi A, Schlitzer A, Low I, Shadan NB, et al. Plasmacytoid dendritic cells develop from Ly6D(+) lymphoid progenitors distinct from the myeloid lineage. Nat Immunol. 2019;20(7):852–64.

    Article  CAS  Google Scholar 

  219. Sachamitr P, Leishman AJ, Davies TJ, Fairchild PJ. Directed differentiation of human induced pluripotent stem cells into dendritic cells displaying tolerogenic properties and resembling the CD141(+) subset. Front Immunol. 2017;8:1935.

    Article  Google Scholar 

  220. Monkley S, Krishnaswamy JK, Göransson M, Clausen M, Meuller J, Thörn K, et al. Optimised generation of iPSC-derived macrophages and dendritic cells that are functionally and transcriptionally similar to their primary counterparts. PLoS ONE. 2020;15(12):e0243807.

    Article  CAS  Google Scholar 

  221. Makino K, Long MD, Kajihara R, Matsueda S, Oba T, Kanehira K, et al. Generation of cDC-like cells from human induced pluripotent stem cells via notch signaling. J Immunother Cancer. 2022;10(1):e003827.

    Article  Google Scholar 

  222. Sontag S, Förster M, Qin J, Wanek P, Mitzka S, Schüler HM, et al. Modelling IRF8 deficient human hematopoiesis and dendritic cell development with engineered iPS cells. Stem Cells. 2017;35(4):898–908.

    CAS  Google Scholar 

  223. Oba T, Makino K, Kajihara R, Yokoi T, Araki R, Abe M, et al. In situ delivery of iPSC-derived dendritic cells with local radiotherapy generates systemic antitumor immunity and potentiates PD-L1 blockade in preclinical poorly immunogenic tumor models. J Immunother Cancer. 2021;9(5):e002432.

    Article  Google Scholar 

  224. Horton C, Davies TJ, Lahiri P, Sachamitr P, Fairchild PJ. Induced pluripotent stem cells reprogrammed from primary dendritic cells provide an abundant source of immunostimulatory dendritic cells for use in immunotherapy. Stem Cells. 2020;38(1):67–79.

    Article  CAS  Google Scholar 

  225. Chhabra A, Chen I-P, Batra D. Human dendritic cell–derived induced pluripotent stem cell lines are not immunogenic. J Immunol. 2017;198(5):1875–86.

    Article  CAS  Google Scholar 

  226. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol. 2008;9(5):503–10.

    Article  CAS  Google Scholar 

  227. Marquardt N, Kekäläinen E, Chen P, Kvedaraite E, Wilson JN, Ivarsson MA, et al. Human lung natural killer cells are predominantly comprised of highly differentiated hypofunctional CD69(-)CD56(dim) cells. J Allergy Clin Immunol. 2017;139(4):1321–e304.

    Article  CAS  Google Scholar 

  228. Ramírez-Labrada A, Pesini C, Santiago L, Hidalgo S, Calvo-Pérez A, Oñate C, et al. All about (NK cell-mediated) death in two acts and an unexpected encore: initiation, execution and activation of adaptive immunity. Front Immunol. 2022;13:896228.

    Article  Google Scholar 

  229. Noyola DE, Juárez-Vega G, Monjarás-Ávila C, Escalante-Padrón F, Rangel-Ramírez V, Cadena-Mota S, et al. NK cell immunophenotypic and genotypic analysis of infants with severe respiratory syncytial virus infection. Microbiol Immunol. 2015;59(7):389–97.

    Article  CAS  Google Scholar 

  230. Larrañaga CL, Ampuero SL, Luchsinger VF, Carrión FA, Aguilar NV, Morales PR, et al. Impaired immune response in severe human lower tract respiratory infection by respiratory syncytial virus. Pediatr Infect Dis J. 2009;28(10):867–73.

    Article  Google Scholar 

  231. Welliver TP, Garofalo RP, Hosakote Y, Hintz KH, Avendano L, Sanchez K, et al. Severe human lower respiratory tract illness caused by respiratory syncytial virus and influenza virus is characterized by the absence of pulmonary cytotoxic lymphocyte responses. J Infect Dis. 2007;195(8):1126–36.

    Article  CAS  Google Scholar 

  232. Miller JS, Alley KA, McGlave P. Differentiation of natural killer (NK) cells from human primitive marrow progenitors in a stroma-based long-term culture system: identification of a CD34 + 7 + NK progenitor. Blood. 1994;83(9):2594–601.

    Article  CAS  Google Scholar 

  233. Mrózek E, Anderson P, Caligiuri MA. Role of interleukin-15 in the development of human CD56 + natural killer cells from CD34 + hematopoietic progenitor cells. Blood. 1996;87(7):2632–40.

    Article  Google Scholar 

  234. Freud AG, Becknell B, Roychowdhury S, Mao HC, Ferketich AK, Nuovo GJ, et al. A human CD34(+) subset resides in lymph nodes and differentiates into CD56bright natural killer cells. Immunity. 2005;22(3):295–304.

    Article  CAS  Google Scholar 

  235. Freud AG, Yokohama A, Becknell B, Lee MT, Mao HC, Ferketich AK, et al. Evidence for discrete stages of human natural killer cell differentiation in vivo. J Exp Med. 2006;203(4):1033–43.

    Article  CAS  Google Scholar 

  236. Motazedian A, Bruveris FF, Kumar SV, Schiesser JV, Chen T, Ng ES, et al. Multipotent RAG1 + progenitors emerge directly from haemogenic endothelium in human pluripotent stem cell-derived haematopoietic organoids. Nat Cell Biol. 2020;22(1):60–73.

    Article  CAS  Google Scholar 

  237. Ng ES, Davis R, Stanley EG, Elefanty AG. A protocol describing the use of a recombinant protein-based, animal product-free medium (APEL) for human embryonic stem cell differentiation as spin embryoid bodies. Nat Protoc. 2008;3(5):768–76.

    Article  CAS  Google Scholar 

  238. Ng ES, Davis RP, Azzola L, Stanley EG, Elefanty AG. Forced aggregation of defined numbers of human embryonic stem cells into embryoid bodies fosters robust, reproducible hematopoietic differentiation. Blood. 2005;106(5):1601–3.

    Article  CAS  Google Scholar 

  239. Knorr DA, Ni Z, Hermanson D, Hexum MK, Bendzick L, Cooper LJ, et al. Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Transl Med. 2013;2(4):274–83.

    Article  CAS  Google Scholar 

  240. Woll PS, Martin CH, Miller JS, Kaufman DS. Human embryonic stem cell-derived NK cells acquire functional receptors and cytolytic activity. J Immunol. 2005;175(8):5095–103.

    Article  CAS  Google Scholar 

  241. Woll PS, Grzywacz B, Tian X, Marcus RK, Knorr DA, Verneris MR, et al. Human embryonic stem cells differentiate into a homogeneous population of natural killer cells with potent in vivo antitumor activity. Blood. 2009;113(24):6094–101.

    Article  CAS  Google Scholar 

  242. Zhu H, Kaufman DS. An improved method to produce clinical-scale natural killer cells from human pluripotent stem cells. Methods Mol Biol. 2019;2048:107–19.

    Article  CAS  Google Scholar 

  243. Goldenson BH, Zhu H, Wang YM, Heragu N, Bernareggi D, Ruiz-Cisneros A, et al. Umbilical cord blood and iPSC-derived natural killer cells demonstrate key differences in cytotoxic activity and KIR profiles. Front Immunol. 2020;11:561553.

    Article  CAS  Google Scholar 

  244. Kaneko S. In vitro generation of antigen-specific T cells from induced pluripotent stem cells of antigen-specific T cell origin. Methods Mol Biol. 2016;1393:67–73.

    Article  Google Scholar 

  245. Maniatis NA, Kotanidou A, Catravas JD, Orfanos SE. Endothelial pathomechanisms in acute lung injury. Vascul Pharmacol. 2008;49(4–6):119–33.

    Article  CAS  Google Scholar 

  246. Sans M, Panés J, Ardite E, Elizalde JI, Arce Y, Elena M, et al. VCAM-1 and ICAM-1 mediate leukocyte-endothelial cell adhesion in rat experimental colitis. Gastroenterology. 1999;116(4):874–83.

    Article  CAS  Google Scholar 

  247. Laudes IJ, Guo RF, Riedemann NC, Speyer C, Craig R, Sarma JV, et al. Disturbed homeostasis of lung intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 during sepsis. Am J Pathol. 2004;164(4):1435–45.

    Article  CAS  Google Scholar 

  248. Sedgwick JB, Menon I, Gern JE, Busse WW. Effects of inflammatory cytokines on the permeability of human lung microvascular endothelial cell monolayers and differential eosinophil transmigration. J Allergy Clin Immunol. 2002;110(5):752–6.

    Article  CAS  Google Scholar 

  249. Teijaro JR, Walsh KB, Cahalan S, Fremgen DM, Roberts E, Scott F, et al. Endothelial cells are central orchestrators of cytokine amplification during influenza virus infection. Cell. 2011;146(6):980–91.

    Article  CAS  Google Scholar 

  250. Kratzer E, Tian Y, Sarich N, Wu T, Meliton A, Leff A, et al. Oxidative stress contributes to lung injury and barrier dysfunction via microtubule destabilization. Am J Respir Cell Mol Biol. 2012;47(5):688–97.

    Article  CAS  Google Scholar 

  251. Levi M, ten Cate H, van der Poll T. Endothelium: interface between coagulation and inflammation. Crit Care Med. 2002;30(5 Suppl):S220–4.

    Article  Google Scholar 

  252. Zhao B, Bowden RA, Stavchansky SA, Bowman PD. Human endothelial cell response to gram-negative lipopolysaccharide assessed with cDNA microarrays. Am J Physiol Cell Physiol. 2001;281(5):C1587–95.

    Article  CAS  Google Scholar 

  253. Bannerman DD, Goldblum SE. Mechanisms of bacterial lipopolysaccharide-induced endothelial apoptosis. Am J Physiology-Lung Cell Mol Physiol. 2003;284(6):L899–914.

    Article  CAS  Google Scholar 

  254. Niethamer TK, Stabler CT, Leach JP, Zepp JA, Morley MP, Babu A, et al. Defining the role of pulmonary endothelial cell heterogeneity in the response to acute lung injury. eLife. 2020;9:e53072.

    Article  CAS  Google Scholar 

  255. Schupp JC, Adams TS, Cosme C, Raredon MSB, Yuan Y, Omote N, et al. Integrated single-cell atlas of endothelial cells of the human lung. Circulation. 2021;144(4):286–302.

    Article  CAS  Google Scholar 

  256. Parker JC, Stevens T, Randall J, Weber DS, King JA. Hydraulic conductance of pulmonary microvascular and macrovascular endothelial cell monolayers. Am J Physiol Lung Cell Mol Physiol. 2006;291(1):L30–7.

    Article  CAS  Google Scholar 

  257. Stevens T. Molecular and cellular determinants of lung endothelial cell heterogeneity. Chest. 2005;128(6 Suppl):s558–64.

    Article  Google Scholar 

  258. Aird WC. Phenotypic heterogeneity of the endothelium: II. Representative vascular beds. Circul Res. 2007;100(2):174–90.

    Article  CAS  Google Scholar 

  259. Aird WC. Phenotypic heterogeneity of the endothelium: I. structure, function, and mechanisms. Circul Res. 2007;100(2):158–73.

    Article  CAS  Google Scholar 

  260. deMello DE, Sawyer D, Galvin N, Reid LM. Early fetal development of lung vasculature. Am J Respir Cell Mol Biol. 1997;16(5):568–81.

    Article  CAS  Google Scholar 

  261. Anderson-Berry A, O’Brien EA, Bleyl SB, Lawson A, Gundersen N, Ryssman D, et al. Vasculogenesis drives pulmonary vascular growth in the developing chick embryo. Dev Dyn. 2005;233(1):145–53.

    Article  CAS  Google Scholar 

  262. Schachtner SK, Wang Y, Scott Baldwin H. Qualitative and quantitative analysis of embryonic pulmonary vessel formation. Am J Respir Cell Mol Biol. 2000;22(2):157–65.

    Article  CAS  Google Scholar 

  263. Hall SM, Hislop AA, Pierce CM, Haworth SG. Prenatal origins of human intrapulmonary arteries: formation and smooth muscle maturation. Am J Respir Cell Mol Biol. 2000;23(2):194–203.

    Article  CAS  Google Scholar 

  264. Stone OA, Stainier DYR. Paraxial mesoderm is the major source of lymphatic endothelium. Dev Cell. 2019;50(2):247–e553.

    Article  CAS  Google Scholar 

  265. Peng T, Tian Y, Boogerd CJ, Lu MM, Kadzik RS, Stewart KM, et al. Coordination of heart and lung co-development by a multipotent cardiopulmonary progenitor. Nature. 2013;500(7464):589–92.

    Article  CAS  Google Scholar 

  266. Ang LT, Nguyen AT, Liu KJ, Chen A, Xiong X, Curtis M et al. Generating human artery and vein cells from pluripotent stem cells highlights the arterial tropism of Nipah and Hendra viruses. Cell. 2022.

  267. Ditadi A, Sturgeon CM, Tober J, Awong G, Kennedy M, Yzaguirre AD, et al. Human definitive haemogenic endothelium and arterial vascular endothelium represent distinct lineages. Nat Cell Biol. 2015;17(5):580–91.

    Article  CAS  Google Scholar 

  268. Rosa S, Praça C, Pitrez PR, Gouveia PJ, Aranguren XL, Ricotti L, et al. Functional characterization of iPSC-derived arterial- and venous-like endothelial cells. Sci Rep. 2019;9(1):3826.

    Article  CAS  Google Scholar 

  269. Sriram G, Tan JY, Islam I, Rufaihah AJ, Cao T. Efficient differentiation of human embryonic stem cells to arterial and venous endothelial cells under feeder- and serum-free conditions. Stem Cell Res Ther. 2015;6:261.

    Article  Google Scholar 

  270. Bezenah JR, Kong YP, Putnam AJ. Evaluating the potential of endothelial cells derived from human induced pluripotent stem cells to form microvascular networks in 3D cultures. Sci Rep. 2018;8(1):2671.

    Article  Google Scholar 

  271. Schmidt S, Alt Y, Deoghare N, Krüger S, Kern A, Rockel AF, et al. A blood vessel organoid model recapitulating aspects of vasculogenesis, angiogenesis and vessel wall maturation. Organoids. 2022;1(1):41–53.

    Article  Google Scholar 

  272. Yuan Y, Leiby KL, Greaney AM, Raredon MSB, Qian H, Schupp JC, et al. A pulmonary vascular model from endothelialized whole organ scaffolds. Front Bioeng Biotechnol. 2021;9:760309.

    Article  Google Scholar 

  273. Burkhanova U, Harris A, Leir SH. Enhancement of airway epithelial cell differentiation by pulmonary endothelial cell co-culture. Stem Cell Res. 2022;65:102967.

    Article  CAS  Google Scholar 

  274. Gu M, Shao NY, Sa S, Li D, Termglinchan V, Ameen M, et al. Patient-specific iPSC-derived endothelial cells uncover pathways that protect against pulmonary hypertension in BMPR2 mutation carriers. Cell Stem Cell. 2017;20(4):490–e5045.

    Article  CAS  Google Scholar 

  275. Sa S, Gu M, Chappell J, Shao NY, Ameen M, Elliott KA, et al. Induced pluripotent stem cell model of pulmonary arterial hypertension reveals novel gene expression and patient specificity. Am J Respir Crit Care Med. 2017;195(7):930–41.

    Article  CAS  Google Scholar 

  276. West JD, Austin ED, Gaskill C, Marriott S, Baskir R, Bilousova G, et al. Identification of a common wnt-associated genetic signature across multiple cell types in pulmonary arterial hypertension. Am J Physiol Cell Physiol. 2014;307(5):C415–30.

    Article  CAS  Google Scholar 

  277. Kiskin FN, Chang CH, Huang CJZ, Kwieder B, Cheung C, Dunmore BJ, et al. Contributions of BMPR2 mutations and extrinsic factors to cellular phenotypes of pulmonary arterial hypertension revealed by induced pluripotent stem cell modeling. Am J Respir Crit Care Med. 2018;198(2):271–5.

    Article  CAS  Google Scholar 

  278. Gu M, editor. Using patient-specific induced pluripotent stem cells to understand and treat pulmonary arterial hypertension. Molecular mechanism of congenital heart disease and pulmonary hypertension; 2020 2020//; Singapore: Springer Singapore.

  279. Boyd DF, Allen EK, Randolph AG, Guo XJ, Weng Y, Sanders CJ, et al. Exuberant fibroblast activity compromises lung function via ADAMTS4. Nature. 2020;587(7834):466–71.

    Article  CAS  Google Scholar 

  280. Rojas-Quintero J, Wang X, Tipper J, Burkett PR, Zuñiga J, Ashtekar AR et al. Matrix metalloproteinase-9 deficiency protects mice from severe influenza a viral infection. JCI Insight. 2018;3(24).

  281. Burnham EL, Hyzy RC, Paine R 3rd, Kelly AM, Quint LE, Lynch D, et al. Detection of fibroproliferation by chest high-resolution CT scan in resolving ARDS. Chest. 2014;146(5):1196–204.

    Article  Google Scholar 

  282. Molyneaux PL, Maher TM. The role of infection in the pathogenesis of idiopathic pulmonary fibrosis. Eur Respir Rev. 2013;22(129):376–81.

    Article  Google Scholar 

  283. Hui DS, Joynt GM, Wong KT, Gomersall CD, Li TS, Antonio G, et al. Impact of severe acute respiratory syndrome (SARS) on pulmonary function, functional capacity and quality of life in a cohort of survivors. Thorax. 2005;60(5):401–9.

    Article  CAS  Google Scholar 

  284. Zhang P, Li J, Liu H, Han N, Ju J, Kou Y, et al. Long-term bone and lung consequences associated with hospital-acquired severe acute respiratory syndrome: a 15-year follow-up from a prospective cohort study. Bone Res. 2020;8:8.

    Article  CAS  Google Scholar 

  285. Duong-Quy S, Vo-Pham-Minh T, Tran-Xuan Q, Huynh-Anh T, Vo-Van T, Vu-Tran-Thien Q, et al. Post-COVID-19 pulmonary fibrosis: facts-challenges and futures: a narrative review. Pulm Ther. 2023;9(3):295–307.

    Article  Google Scholar 

  286. Spagnolo P, Balestro E, Aliberti S, Cocconcelli E, Biondini D, Casa GD, et al. Pulmonary fibrosis secondary to COVID-19: a call to arms? Lancet Respir Med. 2020;8(8):750–2.

    Article  CAS  Google Scholar 

  287. Rendeiro AF, Ravichandran H, Bram Y, Chandar V, Kim J, Meydan C, et al. The spatial landscape of lung pathology during COVID-19 progression. Nature. 2021;593(7860):564–9.

    Article  CAS  Google Scholar 

  288. Redente EF, Chakraborty S, Sajuthi S, Black BP, Edelman BL, Seibold MA et al. Loss of Fas signaling in fibroblasts impairs homeostatic fibrosis resolution and promotes persistent pulmonary fibrosis. JCI Insight. 2020;6(1).

  289. Xie T, Wang Y, Deng N, Huang G, Taghavifar F, Geng Y, et al. Single-cell deconvolution of fibroblast heterogeneity in mouse pulmonary fibrosis. Cell Rep. 2018;22(13):3625–40.

    Article  CAS  Google Scholar 

  290. Hinz B. The role of myofibroblasts in wound healing. Curr Res Transl Med. 2016;64(4):171–7.

    CAS  Google Scholar 

  291. Rehan VK, Sugano S, Wang Y, Santos J, Romero S, Dasgupta C, et al. Evidence for the presence of lipofibroblasts in human lung. Exp Lung Res. 2006;32(8):379–93.

    Article  CAS  Google Scholar 

  292. Ushakumary MG, Riccetti M, Perl A-KT. Resident interstitial lung fibroblasts and their role in alveolar stem cell niche development, homeostasis, injury, and regeneration. Stem Cells Transl Med. 2021;10(7):1021–32.

    Article  CAS  Google Scholar 

  293. El Agha E, Moiseenko A, Kheirollahi V, De Langhe S, Crnkovic S, Kwapiszewska G, et al. Two-way conversion between lipogenic and myogenic fibroblastic phenotypes marks the progression and resolution of lung fibrosis. Cell Stem Cell. 2017;20(2):261–e733.

    Article  Google Scholar 

  294. Alber AB, Marquez HA, Ma L, Kwong G, Thapa BR, Villacorta-Martin C, et al. Directed differentiation of mouse pluripotent stem cells into functional lung-specific mesenchyme. Nat Commun. 2023;14(1):3488.

    Article  CAS  Google Scholar 

  295. Tamai K, Sakai K, Yamaki H, Moriguchi K, Igura K, Maehana S, et al. iPSC-derived mesenchymal cells that support alveolar organoid development. Cell Rep Methods. 2022;2(10):100314.

    Article  CAS  Google Scholar 

  296. Hurley K, Ding J, Villacorta-Martin C, Herriges MJ, Jacob A, Vedaie M, et al. Reconstructed single-cell fate trajectories define lineage plasticity windows during differentiation of human PSC-derived distal lung progenitors. Cell Stem Cell. 2020;26(4):593–e6088.

    Article  CAS  Google Scholar 

  297. McCauley KB, Alysandratos K-D, Jacob A, Hawkins F, Caballero IS, Vedaie M, et al. Single-cell transcriptomic profiling of pluripotent stem cell-derived SCGB3A2 + airway epithelium. Stem Cell Rep. 2018;10(5):1579–95.

    Article  CAS  Google Scholar 

  298. Chen HJ, Poran A, Unni AM, Huang SX, Elemento O, Snoeck HW, et al. Generation of pulmonary neuroendocrine cells and SCLC-like tumors from human embryonic stem cells. J Exp Med. 2019;216(3):674–87.

    Article  CAS  Google Scholar 

  299. Hor P, Punj V, Calvert BA, Castaldi A, Miller AJ, Carraro G, et al. Efficient generation and transcriptomic profiling of human iPSC-derived pulmonary neuroendocrine cells. iScience. 2020;23(5):101083.

    Article  CAS  Google Scholar 

  300. Wang R, Simone-Roach C, Lindstrom-Vautrin J, Wang F, Rollins S, Bawa PS, et al. De novo generation of pulmonary ionocytes from normal and cystic fibrosis human induced pluripotent stem cells. Am J Respir Crit Care Med. 2023;207(9):1249–53.

    Article  CAS  Google Scholar 

  301. Huh D, Matthews BD, Mammoto A, Montoya-Zavala M, Hsin HY, Ingber DE. Reconstituting organ-level lung functions on a chip. Science. 2010;328(5986):1662–8.

    Article  CAS  Google Scholar 

  302. Srivastava K, Pandit B. Genome-wide CRISPR screens and their applications in infectious disease. Front Genome Ed. 2023;5:1243731.

    Article  Google Scholar 

  303. Biering SB, Sarnik SA, Wang E, Zengel JR, Leist SR, Schäfer A, et al. Genome-wide bidirectional CRISPR screens identify mucins as host factors modulating SARS-CoV-2 infection. Nat Genet. 2022;54(8):1078–89.

    Article  CAS  Google Scholar 

  304. Li K, Ouyang M, Zhan J, Tian R. CRISPR-based functional genomics screening in human-pluripotent-stem-cell-derived cell types. Cell Genom. 2023;3(5):100300.

    Article  CAS  Google Scholar 

  305. Firth AL, Menon T, Parker GS, Qualls SJ, Lewis BM, Ke E, et al. Functional gene correction for cystic fibrosis in lung epithelial cells generated from patient iPSCs. Cell Rep. 2015;12(9):1385–90.

    Article  CAS  Google Scholar 

  306. Crane AM, Kramer P, Bui JH, Chung WJ, Li XS, Gonzalez-Garay ML, et al. Targeted correction and restored function of the CFTR gene in cystic fibrosis induced pluripotent stem cells. Stem cell Rep. 2015;4(4):569–77.

    Article  CAS  Google Scholar 

  307. Sone N, Konishi S, Igura K, Tamai K, Ikeo S, Korogi Y, et al. Multicellular modeling of ciliopathy by combining iPS cells and microfluidic airway-on-a-chip technology. Sci Transl Med. 2021;13(601):eabb1298.

    Article  CAS  Google Scholar 

  308. Werder RB, Kaserman JE, Packer MS, Lindstrom-Vautrin J, Villacorta-Martin C, Young LE, et al. Adenine base editing reduces misfolded protein accumulation and toxicity in alpha-1 antitrypsin deficient patient iPSC-hepatocytes. Mol Ther. 2021;29(11):3219–29.

    Article  CAS  Google Scholar 

  309. Kaserman JE, Hurley K, Dodge M, Villacorta-Martin C, Vedaie M, Jean JC, et al. A highly phenotyped open access repository of alpha-1 antitrypsin deficiency pluripotent stem cells. Stem Cell Rep. 2020;15(1):242–55.

    Article  CAS  Google Scholar 

  310. Sun YL, Hennessey EE, Heins H, Yang P, Villacorta-Martin C, Kwan J et al. Human pluripotent stem cell modeling of alveolar type 2 cell dysfunction caused by ABCA3 mutations. J Clin Invest. 2024;134(2).

  311. Lee S-J, Park C, Lee JY, Kim S, Kwon PJ, Kim W, et al. Generation of pure lymphatic endothelial cells from human pluripotent stem cells and their therapeutic effects on wound repair. Sci Rep. 2015;5(1):11019.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to members of the Immune Development and Blood Development laboratories at Murdoch Children’s Research Institute (MCRI) for helpful discussions. Figures were created using BioRender.com.

Funding

This work was supported by the Australian National Health and Medical Research Council, the Stafford Fox Medical Research Foundation, and the Novo Nordisk Foundation Center for Stem Cell Medicine (grant NNF21CC0073729).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, R.B.W.; literature review, D.L.T., S.A., H.B., and R.B.W.; manuscript preparation, D.L.T., S.A. and R.B.W.; manuscript edits and proofing, H.B., E.S. and R.B.W. All authors critically reviewed and approved the final version of the manuscript.

Corresponding author

Correspondence to Rhiannon B. Werder.

Ethics declarations

Ethical approval

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Turner, D.L., Amoozadeh, S., Baric, H. et al. Building a human lung from pluripotent stem cells to model respiratory viral infections. Respir Res 25, 277 (2024). https://doi.org/10.1186/s12931-024-02912-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12931-024-02912-0

Keywords