Abstract
The intertwined relationship between structure and function has been key to understanding human organ physiology and disease pathogenesis. An organ-on-a-chip (organ chip) is a bioengineered microfluidic cell culture device lined by living cells and tissues that recapitulates organ-level functions in vitro. This is accomplished by recreating organ-specific tissue-tissue interfaces and microenvironmental biochemical and mechanical cues while providing dynamic perfusion through endothelium-lined vascular channels. In this review, we discuss how this emerging technology has contributed to the understanding of human lung structure-function relationships at the cell, tissue, and organ levels.
Keywords: lung physiology, mechanobiology, microphysiological system, organ chip, 3-D tissue models
Introduction
Effective characterization of biological structure-function relations requires availability of experimental models that incorporate a sufficient amount of structural information while still allowing functional data to be interpreted with reasonable simplicity (1). This represents a major challenge during physiological investigation of the human lung, which is an organ that consists of >50 heterogeneous types of cells (2) that are arranged into specific architectural units with the primary function of gas exchange between the air and blood. The complexity is heightened further by the fact that the lung is a highly dynamic organ such that its constituent tissues and cells experience a variety of mechanical forces, including cyclic deformation (strain), hydrostatic pressure, and shear stress due to flow of both air and blood (3). Moreover, the influence of these mechanical cues can be significantly impacted by the relative stiffness or compliance of the extracellular matrix (ECM) that underlies the lung epithelium and endothelium and surrounds cells within the intervening connective tissue (4).
The rise of molecular biology and pursuit of therapeutic interventions for human diseases have elevated cellular physiology and the investigation of molecular mechanisms to the forefront of physiology research. This trend is underpinned by the development of human two-dimensional (2-D) culture models that enable high-throughput cell biological analyses and have resulted in some drug discovery success. For example, the use of patient-derived human bronchial epithelial cells contributed to the development of cystic fibrosis (CF) treatments (5). However, the limitations of conventional in vitro culture technology in modeling complex responses, disease phenotypes, and pharmacological behaviors necessitate additional testing in animal models (6), especially genetically engineered mice models that preserve in vivo structural and functional complexity while enabling investigation of the physiological impact of particular gene defects. This research paradigm has been a core pillar of basic biomedical research and preclinical drug discovery for the last 40 years (6). However, the extremely low success rate of preclinical-to-clinical translation has highlighted the genetic, structural, and functional differences among species. For example, results from mouse models fail to predict human responses to inflammatory diseases (7), viral vector-mediated gene therapy (8), and side effects of RNA vaccines against COVID-19 (9). The human lung also differs substantially from its mouse counterpart in terms of structure, constituent cell types, and patterns of gene expression (2, 10, 11). Thus, it is imperative to develop models of the human lung that balance simplicity with the need for relevant organ-level structures and functions, which are required to generate adequate functional information that is directly relevant to lung physiology.
Human organoids and organ chips are two technologies that have emerged over the last decade to address this need. Although they employ distinct design principles, both aim to replicate critical structural and functional features of an organ in vitro with three-dimensional (3-D) cell culture. Organoids are generated by culturing stem cell-derived or tissue progenitor cells within an ECM gel or synthetic hydrogels, where they differentiate and compartmentalize into closed spheroids that exhibit tissue structures with forms that often resemble those seen in vivo. For example, lung alveolar organoids and airway organoids lined by a differentiated lung epithelium have been generated and applied to study lung regeneration, cell fate decisions, and disease processes in vitro (12, 13). Organ chips, on the other hand, build upon the idea that development of differentiated features at the cell and tissue levels alone is not sufficient to fully replicate organ-level responses and that reconstitution of organ-level structures in a physiologically relevant microenvironment containing organ-specific mechanical cues is required as well, as recent evidence suggests that extrinsic microenvironmental controllers complement cell self-organization to facilitate spatial and temporal organoid patterning (14). By using an engineering approach to reproduce cellular scaffolds, multicellular structures, tissue-tissue interfaces, physicochemical microenvironments, and circulatory perfusion, organ chips can achieve tissue- and organ-level complexity, maturity, and functionality that are not attainable with standard 2-D or 3-D culture techniques, including organoids (15, 16).
Organ chips have been utilized to model numerous human tissues, organs, and organ systems, including lung, heart, gastrointestinal (GI) tract, liver, kidney, and brain (15, 17). More general discussions on the use of organ chips in basic biological research, disease modeling, drug development, and personalized medicine can be found in recent reviews (15–17). Here, we focus on the lung organ chip, the first described multitissue organ chip (18) and most well-characterized system. We begin by summarizing lung chip designs with varying degrees of structural complexity for mimicking the lung alveolus or the conducting airway. Then, we describe new functional insights into human lung pathophysiology at the cell, tissue, organ, and organism levels that these models have enabled. Finally, we discuss challenges and future opportunities so that the full potential of organ chips in advancing lung biology and pulmonary medicine can be achieved.
Human Lung Chips
Although the concepts of tissue engineering and “human-body-on-a-chip” have been around since the late 1990s (16), the first microfluidic culture device that recapitulated organ-level structures (FIGURE 1A) and functions of a major functional unit of a human organ—the distal lung gas exchange unit or lung alveolus—was described in Science in 2010 (18). This lung alveolus chip is composed of optically clear, elastomeric polydimethylsiloxane (PDMS) material that contains two parallel hollow channels separated by a porous ECM-coated membrane lined with human lung alveolar epithelial cells on one side and with human vascular endothelium on the other, recreating the native gas-exchanging alveolar-capillary interface (FIGURE 1B). Culture medium is flowed through the endothelium-lined channel to mimic vascular perfusion, and air is introduced into the epithelial channel to mimic the air-liquid interface (ALI). In addition, cyclic suction is applied to hollow side chambers to induce the flexible tissue-tissue interfaces that extend and retract to the same degree and at the same rate as the alveolus experiences because of lung breathing motions. All these elements combined result in a microfluidic microphysiological system that enables unprecedented structural and functional recapitulation of the human distal lung. This reductionist approach to reconstructing the structure and function of human organs created a conceptual framework for the organ chip field. Since then, various organ chip models of the lung and other organs have been designed, fabricated, and used for preclinical disease modeling and drug testing (15). Some have even greater cellular and structural complexity to achieve a higher level of in vivo mimicry, whereas others sacrifice structural complexity for throughput (21).
Human Lung Alveolus Chips
Initial versions of the two-channel alveolus chip model used an established human alveolar epithelial cell line (A549 or NCI-H441) and human umbilical vein endothelial cells (18, 22). The model has since been improved by replacing cells with more relevant primary cells from commercial vendors (23–27), human cadaver lungs (24), or organoid cultures (28). These alveolar chip models have been used to study pulmonary nanotoxicology (18, 29), bacterial infection (18), drug toxicity-induced pulmonary edema (22), orthotopic lung cancer growth (25), intravascular thrombosis (23), and infection by influenza (24, 30) and SARS-CoV-2 (26, 27, 30, 31) viruses.
Single-channel microfluidic devices that only contain alveolar epithelial cells have been developed as well. One model used an elastic membrane consisting of poly(ε-)caprolactone and gelatin that has similar stiffness (Young’s modulus) as the basement membrane of the lung alveolus, to mimic the microenvironment of human alveolar epithelium (32). Despite lacking endothelium, this microfluidic model can be useful for studying stretch-induced functional changes of alveolar epithelium. Another single-channel alveolus chip enables real-time monitoring of cellular mechanics during breathing movements by integrating photonic nanoparticles in the underlying membrane on which the alveolar epithelial cells were seeded (19) (FIGURE 1C). This model allows the investigation of dynamic relationships between cellular deformations and disease phenotypes, such as idiopathic pulmonary fibrosis (IPF) (19).
More complicated models have been created by incorporating additional cell types of the distal lung, such as stromal cells. To accomplish this, the most frequently used design incorporates a middle layer seeded with ECM gel alone or with lung fibroblasts (33) (FIGURE 1D). An open-top chip also has been engineered that allows analysis of stromal-epithelial interactions and cellular cross talk between the three chip compartments. Similar 3-D alveolar barrier models have also been fabricated by inkjet printing alveolar epithelial cells, lung fibroblasts, ECM, and lung microvascular endothelial cells (34), although vascular perfusion and cyclic mechanical strain are absent in this static model. Another strategy for increasing complexity focused on modeling not only the tissue interface but also the 3-D architectures of the alveolar air sac. A biological, stretchable, and biodegradable membrane made of collagen and elastin was cast on a thin gold mesh to emulate an array of tiny alveoli with in vivo-like dimensions (35). Similarly, a 3-D porous hydrogel made of low-stiffness gelatin methacryloyl (GelMA) has been used to construct an inverse opal structure, which is bonded to a compartmentalized PDMS chip device that provides the medium supply, ALI, and the cyclic mechanical movements (20) (FIGURE 1E). Although these more complex chips provide more accurate 3-D structural representations of the in vivo lung alveolus than the two-channel models, it remains to be demonstrated whether they can provide additional novel biological insights.
Human Lung Airway Chips
Human lung airways are divided into two compartments, the larger conducting airways (bronchi) and the smaller respiratory airways (bronchioles), and organ chips have been designed to model both compartments (FIGURE 2A). The two-channel microfluidic design used in the alveolus chip was adapted to create a human lung small airway chip that supports full differentiation of a columnar, pseudostratified, mucociliary bronchiolar epithelium (39). This model was successfully used to model disease processes associated with the small airway, including asthma (39, 40), chronic obstructive pulmonary disease (COPD) (39), and the effects of exposure to cigarette smoke (41). More recently, the chip was modified to contain a membrane with different chemistry (PDMS vs. polyethylene terephthalate) and material properties (flexible vs. rigid) and larger pores (7 mm vs. 0.4 mm) to culture human bronchial airway basal cells and to model the human large bronchial airway (30) (FIGURE 2B). This model has proven to be powerful for studying viral-host interactions (24, 30), modeling disease processes related to CF (42), identifying candidate antiviral therapeutics (30), testing drug efficacy of novel biologics including RNA therapeutics (43), and assessing influenza viral evolution in responses to antiviral drug treatment in vitro (30, 37). Various human diseased airway chips also have been generated by populating chips with primary bronchial or bronchiolar epithelial cells obtained from patients with lung diseases such as COPD (39) or CF (42). These chips have been shown to recapitulate disease-specific phenotypes, for example, selective cytokine hypersecretion, increased neutrophil recruitment, and clinical exacerbation by exposure to viral and bacterial infections (39, 42).
A simplified one-channel microfluidic device composed of proximal airway epithelial progenitor cells derived from human induced pluripotent stem cells (iPSCs) enabled the investigation of fluid shear stress during ciliated cell development and disease modeling for primary ciliary dyskinesia (44). The use of gravity-assisted media exchange instead of perfusion pumps also allowed for an increase in throughput. For example, a human small airway chip employing this method allowed high-throughput studies of direct gas/aerosol exposures to inhaled toxicants (45). A streamlined plastic 384-well format organ chip design also was described recently and used to create a human bronchial airway cultured under an ALI (46) (FIGURE 2C). Although this chip lacked endothelial cells, the model enabled high-throughput infection and drug screening against respiratory viral infections (46).
More complicated models have been created by incorporating other relevant cell types in the conducting airways, such as fibroblasts and smooth muscle cells (SMCs). A common design approach for multilayer microfluidic airway chips is to use a three-channel configuration: a top channel for airway epithelial cells, a middle channel for interstitial fibroblasts, and a bottom channel for microvascular endothelial cells (47). A similar three-compartment design was used to incorporate SMCs by replacing the middle channel with a thin hydrogel layer for ECM and a bottom medium reservoir for SMC culture (48). Another model comprises three hollow channels within a 3-D collagen matrix containing pulmonary fibroblasts; the central channel is lined with primary human bronchial epithelial cells cultured under ALI, whereas the two flanking channels are lined with primary human lung microvascular cells to form the vascular compartment (38) (FIGURE 2D).
A more complicated airway chip disease model was created to study idiopathic pulmonary fibrosis (IPF), in which healthy or IPF primary fibroblasts were cultured in a fibrin-collagen hydrogel filling one channel in parallel to an airway epithelial-endothelial barrier. This model revealed communication between the interstitium and the airway epithelium as well as fibrotic changes in club cells and ciliated cells in the airway epithelium (49). In addition to a single endothelial channel, 3-D printing using a decellularized ECM bioink has been used to encapsulate and print endothelial cells and fibroblasts to form a vascular network in an airway chip (50). Physical forces such as bronchial compressive stress can be introduced to the airway chip as well. Another bronchial airway chip model reconstitutes 3-D cocultures of primary human airway SMCs and bronchial epithelial cells that are fully differentiated under an ALI (51). In addition, physiologically relevant compressive stress was applied to the pseudostratified epithelium by regulating air pressure, and the resulting changes in mechanical properties of SMCs were quantified in real time with optical magnetic twisting cytometry (51). This model can be used to study the interplay of mechanical and biochemical signals in bronchospasm. Finally, by linking a nasal, a bronchus, and an alveolar chip through the air channel, a multicompartment human airway-on-chip platform was fabricated to emulate the airborne route of respiratory pathogens and to investigate regional lung cross talk for viral infection pathways (52).
Human Organ Chips Inform Lung Physiology at the Cell Level
Among the >50 cell types found in the human lung, only a small subset has been well studied (2), and most of these are epithelial cells. Located at the interface between the environment and the organism, epithelial cells serve many important functions including barrier protection, fluid balance, clearance of particulate, initiation of immune responses, production of mucus, and regeneration after injury. Although much of our understanding of these cells has come from mouse studies, recent work using human organ chips has revealed some unique insights.
The two major epithelial lineages within the alveoli are alveolar type 1 (AT1) and AT2 epithelial cells. The thin flat AT1 cells that cover >95% of the lung surface area are responsible for gas exchange, whereas the less frequent cuboidal AT2 cells produce pulmonary surfactant to preserve alveolar surface tension as well as acting as progenitors for AT1 cells. Several studies from human alveolus chips have found that cyclic mechanical strain (which mimics physiological breathing motions) increases surfactant production, decreases cell proliferation, and enhances ECM deposition (18, 19, 24, 53). Cyclic strain also increases nanoparticle absorption across the alveolar-capillary barrier (18), enhances interleukin-2 (IL-2)-induced pulmonary edema via a signaling pathway involving the transient receptor potential vanilloid-type 4 (TRPV4) mechanosensitive ion channel (22), and suppresses lung cancer cell growth and invasion (25).
More recently, physical forces associated with cyclic breathing motions also were found to stimulate lung innate immune responses to viral infection in alveolar epithelial cells and microvascular endothelial cells (24). RNA sequencing (RNA-seq) analysis revealed that TRPV4 and the receptor for advanced glycation end products (RAGE), an inflammatory mediator and marker for ATI cells, play central roles in this response (24). Modulating these pathways may offer great therapeutic potential against viral pneumonia, ventilator-induced lung injury, and other inflammatory respiratory diseases. Moreover, although physiological strain was protective, application of hyperphysiological strains on-chip revealed that they can induce cell apoptosis, activate inflammatory responses, and impair barrier integrity (24, 32) as experienced by patients on mechanical ventilators or with abnormal lung compliance.
The trachea and proximal airways are lined by a pseudostratified epithelium that contains multiple cell lineages, including multiciliated cells, secretory cells, goblet cells, and basal stem cells, as well as many other less abundant cell types, such as neuroendocrine cells (54). The two-channel human small airway chip enables the generation of cilia with the same structure, length, beating frequency, and transport velocity as their in vivo counterparts on the apical surface of the polarized epithelium (39). In addition, fluid shear stress applied through the vascular channel promotes synchronized cilium beating and coordinated mucociliary movement (39). A recent study using a monoculture of iPSC-derived airway epithelial cells exhibited similar behavior even though cells were cultured under a submerged condition, which was previously regarded as not suitable for inducing cilium differentiation (20). It is possible that fluid shear stress can compensate for ALI through an unidentified mechanotransduction mechanism. Investigating how different types of physical stresses alter differentiation, gene expression, and responses to external stimuli in lung epithelial and endothelial cells on-chip will provide critical insights into lung mechanobiology.
Human Organ Chips Inform Lung Physiology at the Tissue Level
The respiratory system in its totality is composed of multiple tissue systems, including the epithelial, endothelial, mesenchymal, and immune components. So far, the greatest understanding from studies of lung organ chips has been obtained for the epithelial tissues. Lung epithelium serves as the first-line defense against environmental pathogens by creating a relatively tight permeability barrier. Human lung organ chips are well suited to study the formation, damage, and repair of this critical tissue interface. Barrier integrity can be measured by quantifying permeability of fluorescent tracers of different sizes or using integrated electrical sensors to measure transepithelial electrical resistance (TEER) on-chip (before these assays, medium needs to be introduced to the air channel in chips under ALI culture conditions) (55, 56). Tissue barrier permeabilities measured within two-channel lung chips are similar to those measured in vivo (18, 41). Compromised barrier function also can be detected on-chip when exposed to a number of insults that are known to contribute to acute lung injury, such as increased levels of IL-2 (22) and TNF-α (23), as well as exposure to bacterial lipopolysaccharide endotoxin (LPS) (23) or infection by influenza virus (24, 30). With easy access to both the air and blood sides of the lung barrier on-chip, it is possible to determine whether any of these insults lead to direct (pulmonary) or indirect (extrapulmonary) lung injury in vitro. For example, LPS causes significant barrier disruption only when dosed in the air channel of the alveolus chip, supporting the hypothesis of direct epithelial injury (23). Interestingly, LPS also induced vascular thrombosis in this model via its effect on the epithelium, as it had no effect on coagulation when infused directly within the endothelium-lined vascular channel.
Both gas-exchanging alveoli and conducting airways are capable of regeneration after injury. However, our understanding of this process remains limited, with much of our knowledge derived from studies of the mouse trachea, which most closely resembles the structure of human proximal airway (57). Human lung organ chips are beginning to produce valuable insights into this process. Similar to in vivo (58, 59), alveolar epithelial cells and endothelial cells are relatively quiescent on-chip but proliferate rapidly upon influenza-induced lung injury (24). Future work is needed to demonstrate whether the airway has similar regenerative capacity on-chip after viral infection.
Human organ chip experiments also have demonstrated airway cell plasticity. Goblet cells constitute ∼10–20% of the total cell population in the human small airway epithelium, but the percentage increased significantly after exposure of the epithelium to interleukin-13 (IL-13) (39). This goblet cell hyperplasia mimics the clinical presentation in the airway mucosa of individuals with asthma (39). The application of recently developed lineage tracing methodologies, such as cellular barcoding (60), to the organ chip field is likely to shed fresh light on cell fate determination during lung development and regeneration.
Human Organ Chips Inform Lung Physiology at the Organ Level
The ability to coculture multiple tissues and cells in an organ-relevant structural context represents the greatest advantage of human organ chips. This allows exploration of cellular cross talk that is vital for the development, injury response, and regeneration of the respiratory system. The process involved in forming the tissues in a lung chip in vitro are also reminiscent of lung developmental processes that occur in vivo. For example, time course RNA-seq analysis suggests significant epithelial-endothelial signaling cross talk that may contribute to distal lung epithelial maturation and vascularization under these conditions (24). Future work is required to examine whether any of the signaling axes that were previously reported in mice (e.g., BMP4-NFATc1-thrombospondin-1, MMP14-EGFR; Ref. 61) are also utilized during tissue differentiation in human lung chips. Another microphysiological model of the airway has revealed cellular cross talk between airway epithelial cells and smooth muscle cells (51). A mechanical stimulus mimicking a bronchospastic challenge was shown to trigger a marked contraction and delayed relaxation of airway smooth muscle. This was mediated by the discordant expression of cyclooxygenase genes in epithelial cells and regulated by the mechanosensor and transcriptional coactivator Yes-associated protein.
Perhaps the use of human organ chips in elucidating tissue cross talk is best exemplified by recent work that used lung chips to study respiratory viral infections. When respiratory viruses, such as influenza virus or SARS-CoV-2, infect airway or alveolar epithelial cells in lung chips, significant inflammation and injury also can be observed in endothelial cells, even though these viruses do not infect endothelial cells in monoculture (24, 26, 27, 31). At least in the case of influenza virus infection, these effects on endothelium are likely due to paracrine signals from the infected epithelial cells, as viral RNA was not detected in the endothelium (24). Alternatively, in the case of SARS-CoV-2 infection, it may be due to mitochondrial dysfunction and activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway (27). The activation of endothelial cells then initiates a series of responses, including the adherence of neutrophils, monocytes, B cells, and T cells to the endothelium and their subsequent transmigration into the air space, which can further amplify release of inflammatory cytokines (24, 30, 31). This vicious cycle can lead to hyperinflammation that causes irreversible tissue damage and loss of lung function (62). A number of immunomodulatory drugs have been tested and showed efficacy in disrupting this process on-chip, including IL-6 antibody tocilizumab (26) and the RAGE inhibitor azeliragon (24). The use of organ chip technology to identify targets that disrupt cellular communication should result in the identification of more effective therapies against human infectious diseases.
Human Organ Chips Inform Lung Physiology at the Organism Level
Compared with other 3-D tissue culture models, one of the greatest advantages of organ chips is their ability to study interorgan communications because they contain vascular channels that experience dynamic flow. This is achieved by fluidically linking the vascular channels of different organ chips to form the so-called human “body-on-chips” (15, 63, 64). The most frequently used application of such multiorgan chip systems is to mimic drug absorption, distribution, and clearance via metabolism or excretion. When combined with mass spectrometry and computational simulation, these body chip systems may offer a replacement to animal models to measure drug pharmacokinetics (PK) and pharmacodynamics (PD) (64–69). By linking the lung with other organs such as the liver, it is possible to evaluate drug delivery and PK/PD of aerosolized small molecular drugs or other therapeutic modalities (70, 71). For example, a multiorgan system composed of the lung, liver, and breast cancer that was created to mimic inhalation therapy of curcumin revealed that the air route is more effective and allows more frequent dosing than intravenous delivery in the same system (72).
Human Organ Chips Inform Lung Microbe-Host Interactions
The lung is susceptible to infection by a wide variety of microorganisms, including viruses, bacteria, and fungi. Human lung chips have been used to model pathogen-host interaction associated with these pathogens. Besides studying host responses and tissue cross talk during infection as discussed above, lung chips can also be used to study how the host shapes the viral evolution landscape. For example, serial passaging of influenza virus between multiple human airway chips (mimicking human-to-human transmission) revealed mutations that have been observed in human transmission of influenza viruses, as well as novel ones that have not been previously reported in human clinical isolates (37). Recent research also suggests that ferrets or mink (which are often used to model viral infections) are poor models for human lung, particularly when investigating spike RBD mutants, as mutations that adapt SARS-CoV-2 to mink or ferret do not increase fitness in the human airway (73). Because the human airway chip enables more faithful mimicry of host innate immune response and host selection pressure compared with traditional cell line models or animal models, it provides a better approach to examine and predict antigenic shift and antigenic drifts, which could inform future vaccine designs (37).
In addition to modeling viral infection, lung chips have also been used to model various bacterial infections, such as those caused by Pseudomonas aeruginosa (42) or Mycobacterium tuberculosis (Mtb) (74); a similar approach has been used to study effects of aerosolized LPS droplets that mimic bacterial infection (75). These studies have provided many insights into host defense. For example, a two-channel lung chip revealed that pulmonary surfactant plays an important role in host innate immunity during early Mtb infection (74). Recently, a human alveolus chip was used to model Aspergillus fumigatus infection, and it was shown that human macrophages partially inhibited the growth of the fungus and contributed to the release of proinflammatory cytokines, which was associated with an increased number of invasive hyphae (76). Organ chips can even be used to study multikingdom (bacterial-fungal-human) human microbiome and their interaction with the host, as demonstrated by a human bronchiole model containing airway, vascular, and ECM compartments (38).
Challenges and Future Directions
During the first decade of the organ chip field, much attention has been paid to microfluidic device design and engineering and demonstration of their capacity to emulate key tissue and organ functions. The advancements discussed above illustrate that human organ chips can be utilized to address a wide range of questions relevant to human lung physiology and pathophysiology at scales from single cells to body-level functions (FIGURE 3) that could not be addressed with conventional culture systems, static microphysiological systems, or even animal models. Moving forward, a few obstacles must be addressed before the full potential of organ chips in lung biology research can be realized. Apart from typical issues related to all organ chip models, such as procurement of high-quality human cells, long-term culture, alternate flexible elastomeric materials, online readouts, and benchmarking (15, 16), there are a few questions that are particularly pertinent to the lung.
The first question is whether we can leverage organ chips to better define intercellular cross talk between the cells that comprise the multiple different tissues of human lung. Although many of the existing lung chip models still largely focus on the epithelium, other tissue types are equally important for maintaining lung structure and function. For example, endothelial cells are involved in the differentiation of alveolar epithelial cells, as well as in alveolar regeneration after acute injury (77–80). A growing body of evidence also indicates that stromal cells and the ECM play a critical role in the development and pathogenesis of interstitial lung diseases, such as pulmonary fibrosis (36, 81–83). However, none of the animal models that are commonly used in current preclinical research provides a faithful representation of pathophysiology of this fibrotic disease (84). The three-channel chip design that incorporates an ECM gel seeded with fibroblasts may represent a promising tool to study this process (33). Another component that is yet to be included on lung chip models is immune cells located in the lung parenchyma, such as alveolar macrophages and tissue-resident memory immune cells, both of which are critical in the host response to respiratory pathogens (85, 86). Finally, the potential to mimic anatomical regions unique to humans may provide a significant benefit in studying the physiology and disorders linked with those regions, such as the submucosal gland and the bronchiole.
Is it possible to model the heterogeneity of the lung with an organ chip? The lung is extremely heterogeneous in terms of its cell composition, topography, and mechanical forces acting on the cells. The variability of lung cell types was only recently mapped out with advances in single-cell analysis techniques. These studies have uncovered previously unidentified populations of cells, such as general capillary endothelial cells and aerocytes that are specialized for gas exchange and leukocyte trafficking inside the capillary endothelium (87). Although it is unrealistic and perhaps also unnecessary for organ chips to represent the full cellular variability of intact organs, characterizing cell heterogeneity on existing chip models or incorporating rare cell types, such as neuroendocrine cells (54) and ionocytes (88), in future models would enable more in-depth analysis of relevant biological questions (89). Similarly, engineering chip models that recapitulate 3-D tissue architecture may enhance their ability to mimic in vivo structural and mechanical anisotropy of lung tissues that are important for its function (20, 90).
Conclusions
The rapid growth of the organ chip field over the last decade has demonstrated that this technology is capable of not only recapitulating what we already know about human lung pathophysiology but also shedding light on previously unknown mechanisms. Many human organ chips have been designed and fabricated to model key functional units of the human respiratory system. However, to fully exploit the potential of lung chips in physiology research, it is critical that these tools be used to address biological questions on a wider scale. There are a few hurdles to this endeavor. Besides common considerations for the organ chip field, such as standardization, democratization, and regulatory challenges (15), model-builders must improve existing designs and develop new ones that can meet the ever-increasing needs of traditional lung biologists. On the other hand, lung biologists who are accustomed to using traditional models must be educated about advances in the organ chip field and be kept current on the rapidly expanding capabilities of these new tools in addressing research questions in lung biology and pulmonary medicine. As with organ chips, it is only through intimate interactions and cross talk within research communities that new knowledge about lung physiology can be obtained.
Acknowledgments
This research was supported by the Wyss Institute for Biologically Inspired Engineering at Harvard University, the National Heart, Lung, and Blood Institute (UH3-HL-141797-02), and the Defense Advanced Research Projects Agency (DARPA) (HR0011-22-20017).
D.E.I. is a founder, board member, and SAB chair and holds equity in Emulate Inc. H.B. has no conflicts of interest, financial or otherwise, to disclose.
H.B. prepared figures; H.B. drafted manuscript; H.B. and D.E.I. edited and revised manuscript; H.B. and D.E.I. approved final version of manuscript.
References
- 1.Weibel ER. The Pathway for Oxygen: Structure and Function in the Mammalian Respiratory System. Cambridge, MA: Harvard University Press, 1984. [Google Scholar]
- 2.Travaglini KJ, Nabhan AN, Penland L, Sinha R, Gillich A, Sit RV, Chang S, Conley SD, Mori Y, Seita J, Berry GJ, Shrager JB, Metzger RJ, Kuo CS, Neff N, Weissman IL, Quake SR, Krasnow MA. A molecular cell atlas of the human lung from single-cell RNA sequencing. Nature 587: 619–625, 2020. doi: 10.1038/s41586-020-2922-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Fredberg JJ, Kamm RD. Stress transmission in the lung: pathways from organ to molecule. Annu Rev Physiol 68: 507–541, 2006. doi: 10.1146/annurev.physiol.68.072304.114110. [DOI] [PubMed] [Google Scholar]
- 4.Ingber DE. Cellular mechanotransduction: putting all the pieces together again. FASEB J 20: 811–827, 2006. doi: 10.1096/fj.05-5424rev. [DOI] [PubMed] [Google Scholar]
- 5.Neuberger T, Burton B, Clark H, Van Goor F. Use of primary cultures of human bronchial epithelial cells isolated from cystic fibrosis patients for the pre-clinical testing of CFTR modulators. Methods Mol Biol 741: 39–54, 2011. doi: 10.1007/978-1-61779-117-8_4. [DOI] [PubMed] [Google Scholar]
- 6.Ingber DE. Is it time for Reviewer 3 to request human organ chip experiments instead of animal validation studies? Adv Sci (Weinh) 7: 2002030, 2020. doi: 10.1002/advs.202002030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, , et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci USA 110: 3507–3512, 2013. doi: 10.1073/pnas.1222878110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Keiser MS, Ranum PT, Yrigollen CM, Carrell EM, Smith GR, Muehlmatt AL, Chen YH, Stein JM, Wolf RL, Radaelli E, Lucas TJ 2nd, Gonzalez-Alegre P, Davidson BL. Toxicity after AAV delivery of RNAi expression constructs into nonhuman primate brain. Nat Med 27: 1982–1989, 2021. doi: 10.1038/s41591-021-01522-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Tahtinen S, Tong AJ, Himmels P, Oh J, Paler-Martinez A, Kim L, Wichner S, Oei Y, McCarron MJ, Freund EC, Amir ZA, de la Cruz CC, Haley B, Blanchette C, Schartner JM, Ye W, Yadav M, Sahin U, Delamarre L, Mellman I. IL-1 and IL-1ra are key regulators of the inflammatory response to RNA vaccines. Nat Immunol 23: 532–542, 2022. doi: 10.1038/s41590-022-01160-y. [DOI] [PubMed] [Google Scholar]
- 10.Basil MC, Morrisey EE. Lung regeneration: a tale of mice and men. Semin Cell Dev Biol 100: 88–100, 2020. doi: 10.1016/j.semcdb.2019.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Basil MC, Cardenas-Diaz FL, Kathiriya JJ, Morley MP, Carl J, Brumwell AN, Katzen J, Slovik KJ, Babu A, Zhou S, Kremp MM, McCauley KB, Li S, Planer JD, Hussain SS, Liu X, Windmueller R, Ying Y, Stewart KM, Oyster M, Christie JD, Diamond JM, Engelhardt JF, Cantu E, Rowe SM, Kotton DN, Chapman HA, Morrisey EE. Human distal airways contain a multipotent secretory cell that can regenerate alveoli. Nature 604: 120–126, 2022. doi: 10.1038/s41586-022-04552-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Liberti DC, Morrisey EE. Organoid models: assessing lung cell fate decisions and disease responses. Trends Mol Med 27: 1159–1174, 2021. doi: 10.1016/j.molmed.2021.09.008. [DOI] [PubMed] [Google Scholar]
- 13.Wang Y, Wang P, Qin J. Human organoids and organs-on-chips for addressing COVID-19 challenges. Adv Sci (Weinh), 9: e2105187, 2022. doi: 10.1002/advs.202105187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Gjorevski N, Nikolaev M, Brown TE, Mitrofanova O, Brandenberg N, DelRio FW, Yavitt FM, Liberali P, Anseth KS, Lutolf MP. Tissue geometry drives deterministic organoid patterning. Science 375: eaaw9021, 2022. doi: 10.1126/science.aaw9021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ingber DE. Human organs-on-chips for disease modelling, drug development and personalized medicine. Nat Rev Genet 2022: 1–25, 2022. doi: 10.1038/s41576-022-00466-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Vunjak-Novakovic G, Ronaldson-Bouchard K, Radisic M. Organs-on-a-chip models for biological research. Cell 184: 4597–4611, 2021. doi: 10.1016/j.cell.2021.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Low LA, Mummery C, Berridge BR, Austin CP, Tagle DA. Organs-on-chips: into the next decade. Nat Rev Drug Discov 20: 345–361, 2021. doi: 10.1038/s41573-020-0079-3. [DOI] [PubMed] [Google Scholar]
- 18.Huh D, Matthews BD, Mammoto A, Montoya-Zavala M, Hsin HY, Ingber DE. Reconstituting organ-level lung functions on a chip. Science 328: 1662–1668, 2010. doi: 10.1126/science.1188302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zhu Y, Sun L, Wang Y, Cai L, Zhang Z, Shang Y, Zhao Y. A biomimetic human lung-on-a-chip with colorful display of microphysiological breath. Adv Mater 34: e2108972, 2022. doi: 10.1002/adma.202108972. [DOI] [PubMed] [Google Scholar]
- 20.Huang D, Liu T, Liao J, Maharjan S, Xie X, Pérez M, Anaya I, Wang S, Tirado Mayer A, Kang Z, Kong W, Mainardi VL, Garciamendez-Mijares CE, Garcia Martínez G, Moretti M, Zhang W, Gu Z, Ghaemmaghami AM, Zhang YS. Reversed-engineered human alveolar lung-on-a-chip model. Proc Natl Acad Sci USA 118: e2016146118, 2021. doi: 10.1073/pnas.2016146118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lagowala DA, Kwon S, Sidhaye VK, Kim DH. Human microphysiological models of airway and alveolar epithelia. Am J Physiol Lung Cell Mol Physiol 321: L1072–L1088, 2021. doi: 10.1152/ajplung.00103.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Huh D, Leslie DC, Matthews BD, Fraser JP, Jurek S, Hamilton GA, Thorneloe KS, McAlexander MA, Ingber DE. A human disease model of drug toxicity-induced pulmonary edema in a lung-on-a-chip microdevice. Sci Transl Med 4: 159ra147, 2012. doi: 10.1126/scitranslmed.3004249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Jain A, Barrile R, van der Meer AD, Mammoto A, Mammoto T, De Ceunynck K, Aisiku O, Otieno MA, Louden CS, Hamilton GA, Flaumenhaft R, Ingber DE. Primary human lung alveolus-on-a-chip model of intravascular thrombosis for assessment of therapeutics. Clin Pharmacol Ther 103: 332–340, 2018. doi: 10.1002/cpt.742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bai H, Si L, Jiang A, Belgur C, Zhai Y, Plebani R, Oh CY, Rodas M, Patil A, Nurani A, Gilpin SE, Powers RK, Goyal G, Prantil-Baun R, Ingber DE. Mechanical control of innate immune responses against viral infection revealed in a human lung alveolus chip. Nat Commun 13: 1928, 2022. doi: 10.1038/s41467-022-29562-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hassell BA, Goyal G, Lee E, Sontheimer-Phelps A, Levy O, Chen CS, Ingber DE. Human organ chip models recapitulate orthotopic lung cancer growth, therapeutic responses, and tumor dormancy in vitro. Cell Rep 23: 3698, 2018. doi: 10.1016/j.celrep.2018.06.028. [DOI] [PubMed] [Google Scholar]
- 26.Thacker VV, Sharma K, Dhar N, Mancini GF, Sordet-Dessimoz J, McKinney JD. Rapid endotheliitis and vascular damage characterize SARS-CoV-2 infection in a human lung-on-chip model. EMBO Rep 22: e52744, 2021. doi: 10.15252/embr.202152744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Domizio JD, Gulen MF, Saidoune F, Thacker VV, Yatim A, Sharma K, Nass T, Guenova E, Schaller M, Conrad C, Goepfert C, De Leval L, Garnier CV, Berezowska S, Dubois A, Gilliet M, Ablasser A. The cGAS-STING pathway drives type I IFN immunopathology in COVID-19. Nature 603: 145–151, 2022. doi: 10.1038/s41586-022-04421-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.van Riet S, van Schadewijk A, Khedoe P, Limpens R, Bárcena M, Stolk J, Hiemstra PS, van der Does AM. Organoid-based expansion of patient-derived primary alveolar type 2 cells for establishment of alveolus epithelial lung-chip cultures. Am J Physiol Lung Cell Mol Physiol 322: L526–L538, 2022. doi: 10.1152/ajplung.00153.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zheng L, Dong H, Zhao W, Zhang X, Duan X, Zhang H, Liu S, Sui G. An air-liquid interface organ-level lung microfluidics platform for analysis on molecular mechanisms of cytotoxicity induced by cancer-causing fine particles. ACS Sens 4: 907–917, 2019. doi: 10.1021/acssensors.8b01672. [DOI] [PubMed] [Google Scholar]
- 30.Si L, Bai H, Rodas M, Cao W, Oh CY, Jiang A, , et al. A human-airway-on-a-chip for the rapid identification of candidate antiviral therapeutics and prophylactics. Nat Biomed Eng 5: 815–829, 2021. doi: 10.1038/s41551-021-00718-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhang M, Wang P, Luo R, Wang Y, Li Z, Guo Y, Yao Y, Li M, Tao T, Chen W, Han J, Liu H, Cui K, Zhang X, Zheng Y, Qin J. Biomimetic human disease model of SARS-CoV-2 induced lung injury and immune responses on organ chip system. Adv Sci (Weinh), 8: 2002928, 2021. doi: 10.1002/advs.202002928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Doryab A, Taskin MB, Stahlhut P, Schröppel A, Wagner DE, Groll J, Schmid O. A biomimetic, copolymeric membrane for cell-stretch experiments with pulmonary epithelial cells at the air-liquid interface. Adv Funct Mater 31: 2004707, 2021. doi: 10.1002/adfm.202004707. [DOI] [Google Scholar]
- 33.Varone A, Nguyen JK, Leng L, Barrile R, Sliz J, Lucchesi C, Wen N, Gravanis A, Hamilton GA, Karalis K, Hinojosa CD. A novel organ-chip system emulates three-dimensional architecture of the human epithelia and the mechanical forces acting on it. Biomaterials 275: 120957, 2021. doi: 10.1016/j.biomaterials.2021.120957. [DOI] [PubMed] [Google Scholar]
- 34.Kang D, Park JA, Kim W, Kim S, Lee HR, Kim WJ, Yoo JY, Jung S. All-inkjet-printed 3D alveolar barrier model with physiologically relevant microarchitecture. Adv Sci (Weinh) 8: 2004990, 2021. doi: 10.1002/advs.202004990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zamprogno P, Wüthrich S, Achenbach S, Thoma G, Stucki JD, Hobi N, Schneider-Daum N, Lehr CM, Huwer H, Geiser T, Schmid RA, Guenat OT. Second-generation lung-on-a-chip with an array of stretchable alveoli made with a biological membrane. Commun Biol 4: 168, 2021. doi: 10.1038/s42003-021-01695-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zepp JA, Morrisey EE. Cellular crosstalk in the development and regeneration of the respiratory system. Nat Rev Mol Cell Biol 20: 551–566, 2019. doi: 10.1038/s41580-019-0141-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Si L, Bai H, Oh CY, Jin L, Prantil-Baun R, Ingber DE. Clinically relevant influenza virus evolution reconstituted in a human lung airway-on-a-chip. Microbiol Spectr 9: e0025721, 2021. doi: 10.1128/Spectrum.00257-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Barkal LJ, Procknow CL, Álvarez-García YR, Niu M, Jiménez-Torres JA, Brockman-Schneider RA, Gern JE, Denlinger LC, Theberge AB, Keller NP, Berthier E, Beebe DJ. Microbial volatile communication in human organotypic lung models. Nat Commun 8: 1770, 2017. doi: 10.1038/s41467-017-01985-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Benam KH, Villenave R, Lucchesi C, Varone A, Hubeau C, Lee HH, Alves SE, Salmon M, Ferrante TC, Weaver JC, Bahinski A, Hamilton GA, Ingber DE. Small airway-on-a-chip enables analysis of human lung inflammation and drug responses in vitro. Nat Methods 13: 151–157, 2016. doi: 10.1038/nmeth.3697. [DOI] [PubMed] [Google Scholar]
- 40.Nawroth JC, Lucchesi C, Cheng D, Shukla A, Ngyuen J, Shroff T, Varone A, Karalis K, Lee HH, Alves S, Hamilton GA, Salmon M, Villenave R. A microengineered airway lung chip models key features of viral-induced exacerbation of asthma. Am J Respir Cell Mol Biol 63: 591–600, 2020. doi: 10.1165/rcmb.2020-0010MA. [DOI] [PubMed] [Google Scholar]
- 41.Benam KH, Novak R, Nawroth J, Hirano-Kobayashi M, Ferrante TC, Choe Y, Prantil-Baun R, Weaver JC, Bahinski A, Parker KK, Ingber DE. Matched-comparative modeling of normal and diseased human airway responses using a microengineered breathing lung chip. Cell Syst 3: 456–466.e4, 2016. PMC]doi: 10.1016/j.cels.2016.10.003. [DOI] [PubMed] [Google Scholar]
- 42.Plebani R, Potla R, Soong M, Bai H, Izadifar Z, Jiang A, Travis RN, Belgur C, Dinis A, Cartwright MJ, Prantil-Baun R, Jolly P, Gilpin SE, Romano M, Ingber DE. Modeling pulmonary cystic fibrosis in a human lung airway-on-a-chip: cystic fibrosis airway chip. J Cyst Fibros 2021: S1569-1993(21)02106-8, 2021. doi: 10.1016/j.jcf.2021.10.004. [DOI] [PubMed] [Google Scholar]
- 43.Si L, Bai H, Oh CY, Zhang T, Hong F, Jiang A, Ye Y, Jordan Tx Logue J, McGrath M, Belgur C, Nurani A, Cao W, Prantil-Baun R, Gygi SP, Powers RK, Frieman M, tenOever BR, Ingber DE. Self-assembling short immunostimulatory duplex RNAs with broad spectrum antiviral activity (Preprint). bioRxiv 2021.2011.2019.469183, 2021. doi: 10.1101/2021.11.19.469183. [DOI] [PMC free article] [PubMed]
- 44.Sone N, Konishi S, Igura K, Tamai K, Ikeo S, Korogi Y, Kanagaki S, Namba T, Yamamoto Y, Xu Y, Takeuchi K, Adachi Y, Chen-Yoshikawa TF, Date H, Hagiwara M, Tsukita S, Hirai T, Torisawa YS, Gotoh S. Multicellular modeling of ciliopathy by combining iPS cells and microfluidic airway-on-a-chip technology. Sci Transl Med 13: eabb1298, 2021. doi: 10.1126/scitranslmed.abb1298. [DOI] [PubMed] [Google Scholar]
- 45.Sakolish C, Georgescu A, Huh DD, Rusyn I. A model of human small airway on a chip for studies of sub-acute effects of inhalation toxicants. Toxicol Sci 187: 267–278, 2022. doi: 10.1093/toxsci/kfac036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gard AL, Luu RJ, Miller CR, Maloney R, Cain BP, Marr EE, Burns DM, Gaibler R, Mulhern TJ, Wong CA, Alladina J, Coppeta JR, Liu P, Wang JP, Azizgolshani H, Fezzie RF, Balestrini JL, Isenberg BC, Medoff BD, Finberg RW, Borenstein JT. High-throughput human primary cell-based airway model for evaluating influenza, coronavirus, or other respiratory viruses in vitro. Sci Rep 11: 14961, 2021. doi: 10.1038/s41598-021-94095-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Sellgren KL, Butala EJ, Gilmour BP, Randell SH, Grego S. A biomimetic multicellular model of the airways using primary human cells. Lab Chip 14: 3349–3358, 2014. doi: 10.1039/c4lc00552j. [DOI] [PubMed] [Google Scholar]
- 48.Humayun M, Chow CW, Young EW. Microfluidic lung airway-on-a-chip with arrayable suspended gels for studying epithelial and smooth muscle cell interactions. Lab Chip 18: 1298–1309, 2018. doi: 10.1039/c7lc01357d. [DOI] [PubMed] [Google Scholar]
- 49.Mejías JC, Nelson MR, Liseth O, Roy K. A 96-well format microvascularized human lung-on-a-chip platform for microphysiological modeling of fibrotic diseases. Lab Chip 20: 3601–3611, 2020. doi: 10.1039/d0lc00644k. [DOI] [PubMed] [Google Scholar]
- 50.Park JY, Ryu H, Lee B, Ha DH, Ahn M, Kim S, Kim JY, Jeon NL, Cho DW. Development of a functional airway-on-a-chip by 3D cell printing. Biofabrication 11: 015002, 2018. doi: 10.1088/1758-5090/aae545. [DOI] [PubMed] [Google Scholar]
- 51.Kilic O, Yoon A, Shah SR, Yong HM, Ruiz-Valls A, Chang H, Panettieri RA Jr, Liggett SB, Quinones-Hinojosa A, An SS, Levchenko A. A microphysiological model of the bronchial airways reveals the interplay of mechanical and biochemical signals in bronchospasm. Nat Biomed Eng 3: 532–544, 2019. doi: 10.1038/s41551-019-0366-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Nof E, Zidan H, Artzy-Schnirman A, Mouhadeb O, Beckerman M, Bhardwaj S, Elias-Kirma S, Gur D, Beth-Din A, Levenberg S, Korin N, Ordentlich A, Sznitman J. Human multi-compartment airways-on-chip platform for emulating respiratory airborne transmission: from nose to pulmonary acini. Front Physiol 13: 853317, 2022. doi: 10.3389/fphys.2022.853317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kumar V, Madhurakkat Perikamana SK, Tata A, Hoque J, Gilpin A, Tata PR, Varghese S. An in vitro microfluidic alveolus model to study lung biomechanics. Front Bioeng Biotechnol 10: 848699, 2022. doi: 10.3389/fbioe.2022.848699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Branchfield K, Nantie L, Verheyden JM, Sui P, Wienhold MD, Sun X. Pulmonary neuroendocrine cells function as airway sensors to control lung immune response. Science 351: 707–710, 2016. doi: 10.1126/science.aad7969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Maoz BM, Herland A, Henry OY, Leineweber WD, Yadid M, Doyle J, Mannix R, Kujala VJ, FitzGerald EA, Parker KK, Ingber DE. Organs-on-chips with combined multi-electrode array and transepithelial electrical resistance measurement capabilities. Lab Chip 17: 2294–2302, 2017. doi: 10.1039/c7lc00412e. [DOI] [PubMed] [Google Scholar]
- 56.Henry OY, Villenave R, Cronce MJ, Leineweber WD, Benz MA, Ingber DE. Organs-on-chips with integrated electrodes for trans-epithelial electrical resistance (TEER) measurements of human epithelial barrier function. Lab Chip 17: 2264–2271, 2017. doi: 10.1039/c7lc00155j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Basil MC, Katzen J, Engler AE, Guo M, Herriges MJ, Kathiriya JJ, Windmueller R, Ysasi AB, Zacharias WJ, Chapman HA, Kotton DN, Rock JR, Snoeck HW, Vunjak-Novakovic G, Whitsett JA, Morrisey EE. The cellular and physiological basis for lung repair and regeneration: past, present, and future. Cell Stem Cell 26: 482–502, 2020. doi: 10.1016/j.stem.2020.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Major J, Crotta S, Llorian M, McCabe TM, Gad HH, Priestnall SL, Hartmann R, Wack A. Type I and III interferons disrupt lung epithelial repair during recovery from viral infection. Science 369: 712–717, 2020. doi: 10.1126/science.abc2061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zhao G, Weiner AI, Neupauer KM, de Mello Costa MF, Palashikar G, Adams-Tzivelekidis S, Mangalmurti NS, Vaughan AE. Regeneration of the pulmonary vascular endothelium after viral pneumonia requires COUP-TF2. Sci Adv 6: eabc4493, 2020. doi: 10.1126/sciadv.abc4493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kebschull JM, Zador AM. Cellular barcoding: lineage tracing, screening and beyond. Nat Methods 15: 871–879, 2018. doi: 10.1038/s41592-018-0185-x. [DOI] [PubMed] [Google Scholar]
- 61.Ding BS, Nolan DJ, Guo P, Babazadeh AO, Cao Z, Rosenwaks Z, Crystal RG, Simons M, Sato TN, Worgall S, Shido K, Rabbany SY, Rafii S. Endothelial-derived angiocrine signals induce and sustain regenerative lung alveolarization. Cell 147: 539–553, 2011. doi: 10.1016/j.cell.2011.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Lamers MM, Haagmans BL. SARS-CoV-2 pathogenesis. Nat Rev Microbiol 20: 270–284, 2022. doi: 10.1038/s41579-022-00713-0. [DOI] [PubMed] [Google Scholar]
- 63.Novak R, Ingram M, Marquez S, Das D, Delahanty A, Herland A, , et al. Robotic fluidic coupling and interrogation of multiple vascularized organ chips. Nat Biomed Eng 4: 407–420, 2020. doi: 10.1038/s41551-019-0497-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wang X, Cirit M, Wishnok JS, Griffith LG, Tannenbaum SR. Analysis of an integrated human multiorgan microphysiological system for combined tolcapone metabolism and brain metabolomics. Anal Chem 91: 8667–8675, 2019. doi: 10.1021/acs.analchem.9b02224. [DOI] [PubMed] [Google Scholar]
- 65.Herland A, Maoz BM, Das D, Somayaji MR, Prantil-Baun R, Novak R, Cronce M, Huffstater T, Jeanty SS, Ingram M, Chalkiadaki A, Benson Chou D, Marquez S, Delahanty A, Jalili-Firoozinezhad S, Milton Y, Sontheimer-Phelps A, Swenor B, Levy O, Parker KK, Przekwas A, Ingber DE. Quantitative prediction of human pharmacokinetic responses to drugs via fluidically coupled vascularized organ chips. Nat Biomed Eng 4: 421–436, 2020. doi: 10.1038/s41551-019-0498-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Prantil-Baun R, Novak R, Das D, Somayaji MR, Przekwas A, Ingber DE. Physiologically based pharmacokinetic and pharmacodynamic analysis enabled by microfluidically linked organs-on-chips. Annu Rev Pharmacol Toxicol 58: 37–64, 2018. doi: 10.1146/annurev-pharmtox-010716-104748. [DOI] [PubMed] [Google Scholar]
- 67.Grant J, Özkan A, Oh C, Mahajan G, Prantil-Baun R, Ingber DE. Simulating drug concentrations in PDMS microfluidic organ chips. Lab Chip 21: 3509–3519, 2021. doi: 10.1039/d1lc00348h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ronaldson-Bouchard K, Teles D, Yeager K, Tavakol DN, Zhao Y, Chramiec A, Tagore S, Summers M, Stylianos S, Tamargo M, Lee BM, Halligan SP, Abaci EH, Guo Z, Jacków J, Pappalardo A, Shih J, Soni RK, Sonar S, German C, Christiano AM, Califano A, Hirschi KK, Chen CS, Przekwas A, Vunjak-Novakovic G. A multi-organ chip with matured tissue niches linked by vascular flow. Nat Biomed Eng 6: 351–371, 2022. doi: 10.1038/s41551-022-00882-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Picollet-D’hahan N, Zuchowska A, Lemeunier I, Le Gac S. Multiorgan-on-a-chip: a systemic approach to model and decipher inter-organ communication. Trends Biotechnol 39: 788–810, 2021. doi: 10.1016/j.tibtech.2020.11.014. [DOI] [PubMed] [Google Scholar]
- 70.Bovard D, Sandoz A, Luettich K, Frentzel S, Iskandar A, Marescotti D, Trivedi K, Guedj E, Dutertre Q, Peitsch MC, Hoeng J. A lung/liver-on-a-chip platform for acute and chronic toxicity studies. Lab Chip 18: 3814–3829, 2018. doi: 10.1039/c8lc01029c. [DOI] [PubMed] [Google Scholar]
- 71.Skardal A, Aleman J, Forsythe S, Rajan S, Murphy S, Devarasetty M, Pourhabibi Zarandi N, Nzou G, Wicks R, Sadri-Ardekani H, Bishop C, Soker S, Hall A, Shupe T, Atala A. Drug compound screening in single and integrated multi-organoid body-on-a-chip systems. Biofabrication 12: 025017, 2020. doi: 10.1088/1758-5090/ab6d36. [DOI] [PubMed] [Google Scholar]
- 72.Miller PG, Chen CY, Wang YI, Gao E, Shuler ML. Multiorgan microfluidic platform with breathable lung chamber for inhalation or intravenous drug screening and development. Biotechnol Bioeng 117: 486–497, 2020. doi: 10.1002/bit.27188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zhou J, Peacock TP, Brown JC, Goldhill DH, Elrefaey AM, Penrice-Randal R, Cowton VM, De Lorenzo G, Furnon W, Harvey WT, Kugathasan R, Frise R, Baillon L, Lassaunière R, Thakur N, Gallo G, Goldswain H, Donovan-Banfield I, Dong X, Randle NP, Sweeney F, Glynn MC, Quantrill JL, McKay PF, Patel AH, Palmarini M, Hiscox JA, Bailey D, Barclay WS. Mutations that adapt SARS-CoV-2 to mink or ferret do not increase fitness in the human airway. Cell Rep 38: 110344, 2022. doi: 10.1016/j.celrep.2022.110344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Thacker VV, Dhar N, Sharma K, Barrile R, Karalis K, McKinney JD. A lung-on-chip model of early Mycobacterium tuberculosis infection reveals an essential role for alveolar epithelial cells in controlling bacterial growth. Elife 9: e59961, 2020. doi: 10.7554/eLife.59961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Artzy-Schnirman A, Zidan H, Elias-Kirma S, Ben-Porat L, Tenenbaum-Katan J, Carius P, Fishler R, Schneider-Daum N, Lehr CM, Sznitman J. Capturing the onset of bacterial pulmonary infection in acini-on-chips. Adv Biosyst 3: e1900026, 2019. doi: 10.1002/adbi.201900026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Hoang TN, Cseresnyés Z, Hartung S, Blickensdorf M, Saffer C, Rennert K, Mosig AS, von Lilienfeld-Toal M, Figge MT. Invasive aspergillosis-on-chip: A quantitative treatment study of human Aspergillus fumigatus infection. Biomaterials 283: 121420, 2022. doi: 10.1016/j.biomaterials.2022.121420. [DOI] [PubMed] [Google Scholar]
- 77.Mammoto A, Mammoto T. Vascular niche in lung alveolar development, homeostasis, and regeneration. Front Bioeng Biotechnol 7: 318, 2019. doi: 10.3389/fbioe.2019.00318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Yao J, Guihard PJ, Wu X, Blazquez-Medela AM, Spencer MJ, Jumabay M, Tontonoz P, Fogelman AM, Boström KI, Yao Y. Vascular endothelium plays a key role in directing pulmonary epithelial cell differentiation. J Cell Biol 216: 3369–3385, 2017. doi: 10.1083/jcb.201612122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Rafii S, Cao Z, Lis R, Siempos II, Chavez D, Shido K, Rabbany SY, Ding BS. Platelet-derived SDF-1 primes the pulmonary capillary vascular niche to drive lung alveolar regeneration. Nat Cell Biol 17: 123–136, 2015. doi: 10.1038/ncb3096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Lee JH, Bhang DH, Beede A, Huang TL, Stripp BR, Bloch KD, Wagers AJ, Tseng YH, Ryeom S, Kim CF. Lung stem cell differentiation in mice directed by endothelial cells via a BMP4-NFATc1-thrombospondin-1 axis. Cell 156: 440–455, 2014. doi: 10.1016/j.cell.2013.12.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zepp JA, Morley MP, Loebel C, Kremp MM, Chaudhry FN, Basil MC, Leach JP, Liberti DC, Niethamer TK, Ying Y, Jayachandran S, Babu A, Zhou S, Frank DB, Burdick JA, Morrisey EE. Genomic, epigenomic, and biophysical cues controlling the emergence of the lung alveolus. Science 371: eabc3172, 2021. doi: 10.1126/science.abc3172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Nabhan AN, Brownfield DG, Harbury PB, Krasnow MA, Desai TJ. Single-cell Wnt signaling niches maintain stemness of alveolar type 2 cells. Science 359: 1118–1123, 2018. doi: 10.1126/science.aam6603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Zhou Y, Horowitz JC, Naba A, Ambalavanan N, Atabai K, Balestrini J, Bitterman PB, Corley RA, Ding BS, Engler AJ, Hansen KC, Hagood JS, Kheradmand F, Lin QS, Neptune E, Niklason L, Ortiz LA, Parks WC, Tschumperlin DJ, White ES, Chapman HA, Thannickal VJ. Extracellular matrix in lung development, homeostasis and disease. Matrix Biol 73: 77–104, 2018. doi: 10.1016/j.matbio.2018.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Moore BB, Lawson WE, Oury TD, Sisson TH, Raghavendran K, Hogaboam CM. Animal models of fibrotic lung disease. Am J Respir Cell Mol Biol 49: 167–179, 2013. doi: 10.1165/rcmb.2013-0094TR. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Shenoy AT, Lyon De Ana C, Arafa EI, Salwig I, Barker KA, Korkmaz FT, Ramanujan A, Etesami NS, Soucy AM, Martin IM, Tilton BR, Hinds A, Goltry WN, Kathuria H, Braun T, Jones MR, Quinton LJ, Belkina AC, Mizgerd JP. Antigen presentation by lung epithelial cells directs CD4(+) TRM cell function and regulates barrier immunity. Nat Commun 12: 5834, 2021. doi: 10.1038/s41467-021-26045-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Peteranderl C, Morales-Nebreda L, Selvakumar B, Lecuona E, Vadász I, Morty RE, Schmoldt C, Bespalowa J, Wolff T, Pleschka S, Mayer K, Gattenloehner S, Fink L, Lohmeyer J, Seeger W, Sznajder JI, Mutlu GM, Budinger GR, Herold S. Macrophage-epithelial paracrine crosstalk inhibits lung edema clearance during influenza infection. J Clin Invest 126: 1566–1580, 2016. doi: 10.1172/JCI83931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Gillich A, Zhang F, Farmer CG, Travaglini KJ, Tan SY, Gu M, Zhou B, Feinstein JA, Krasnow MA, Metzger RJ. Capillary cell-type specialization in the alveolus. Nature 586: 785–789, 2020. doi: 10.1038/s41586-020-2822-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Montoro DT, Haber AL, Biton M, Vinarsky V, Lin B, Birket SE, Yuan F, Chen S, Leung HM, Villoria J, Rogel N, Burgin G, Tsankov AM, Waghray A, Slyper M, Waldman J, Nguyen L, Dionne D, Rozenblatt-Rosen O, Tata PR, Mou H, Shivaraju M, Bihler H, Mense M, Tearney GJ, Rowe SM, Engelhardt JF, Regev A, Rajagopal J. A revised airway epithelial hierarchy includes CFTR-expressing ionocytes. Nature 560: 319–324, 2018. doi: 10.1038/s41586-018-0393-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Mertz DR, Ahmed T, Takayama S. Engineering cell heterogeneity into organs-on-a-chip. Lab Chip 18: 2378–2395, 2018. doi: 10.1039/c8lc00413g. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Mondrinos MJ, Alisafaei F, Yi AY, Ahmadzadeh H, Lee I, Blundell C, Seo J, Osborn M, Jeon TJ, Kim SM, Shenoy VB, Huh D. Surface-directed engineering of tissue anisotropy in microphysiological models of musculoskeletal tissue. Sci Adv 7: eabe9446, 2021. doi: 10.1126/sciadv.abe9446. [DOI] [PMC free article] [PubMed] [Google Scholar]