Abstract
Intestinal organoids have become indispensable tools for many gastrointestinal researchers, advancing their studies of nontransformed intestinal epithelial cells, and their roles in an array of diseases, including inflammatory bowel disease and colon cancer. In many cases. these diseases, as well as many enteric infections, reflect pathogenic interactions between bacteria and the gut epithelium. The complexity of studying this microbe–epithelial interface in vivo has led to significant focus on modeling this cross-talk using organoid models. Considering how quickly the organoid field is advancing, it can be difficult to keep up to date with the latest techniques, as well as their respective strengths and weaknesses. This review addresses the advantages of using organoids derived from adult stem cells and the recently identified differences that biopsy location and patient age can have on organoids and their interactions with microbes. Several approaches to introducing bacteria in a relevant (apical) manner (ie, microinjecting 3-dimensional spheroids, polarity-reversed organoids, and 2-dimensional monolayers) also are addressed, as are the key readouts that can be obtained using these models. Lastly, the potential for new approaches, such as air–liquid interface, to facilitate studying bacterial interactions with important but understudied epithelial subsets such as goblet cells and their products, is evaluated.
Keywords: Organoid, Enteroid, Intestinal epithelium, Host-microbe interaction, Co-culture
Abbreviations used in this paper: 2D, 2-dimensional; 3D, 3-dimensional; ALI, air–liquid interface; ASC, adult stem cell; EAEC, enteroaggregative Escherichia coli; ECM, extracellular matrix; GI, gastrointestinal; IEC, intestinal epithelial cell; iPSC, inducible pluripotent stem cell; LGR5, leucine-rich repeat-containing G-protein–coupled receptor 5; LPS, lipopolysaccharide; NOD2, nucleotide-binding oligomerization domain-containing protein 2; SCFA, short-chain fatty acid; TLR, Toll-like receptor
Summary.
This review provides a step-by-step guide for investigating host–microbe interactions using organoids, covering the importance of organoid type selection, the advantages and disadvantages of different techniques to introduce microbes, and cutting-edge advances made to better mimic in vivo conditions.
Many aspects of gastrointestinal (GI) health, including intestinal development, nutrition, digestion, and healing, all are impacted positively by signals coming from the gut microbiome. Correspondingly, maladaptive interactions between the host and specific gut microbes can lead to an array of infectious and idiopathic diseases including inflammatory bowel disease and colon cancer.1, 2, 3 In most cases, these interactions reflect microbial contact with the intestinal epithelium. This single layer of cells, comprising an array of functionally distinct cellular subsets, acts as a protective barrier as well as a messenger, enabling cross-talk between luminal microbes and the underlying immune cells, nerves, and other cell types.4,5
Studying microbe–intestinal epithelial cell (IEC) interactions in vivo is highly complex, necessitating the use of reductionist co-culture systems that until recently focused on transformed cell lines. These cell models have expanded our understanding of microbe–IEC interactions, but their inability to reproduce the complex structure and functionality of the epithelium along the length of the GI tract has limited their relevance. Just over a decade ago, a revolution enabled the culturing and propagation of stem cells to form organ-specific cell types in 3-dimensional (3D) cultures, better known as organoids.6,7 Since its emergence, intestinal organoid research has been a rapidly evolving field. Novel technologies and methodologies are being published daily, and many of these advances have been incorporated into modeling microbe–IEC interactions.
For this review, we have compiled these advances into a convenient guide focused on bacteria–host interactions within the GI tract because virus–organoid interactions recently were reviewed.8 Criteria for choosing the proper sources of stem cells as well as the advantages and limitations of the different bacteria–organoid co-culture approaches are discussed. Moreover, options for better simulating the in vivo conditions of the GI tract, as well as modeling both pathogenic and commensal microbe interactions with organoids, are detailed. It is hoped that this guide (Figure 1) will benefit researchers seeking to enter the field as well as experienced organoid researchers looking to optimize their current model systems.
Figure 1.
Brief guide on designing bacteria–IEC interaction experiments using organoids. Organoids need to be generated from the most relevant (and obtainable) stem cell source, GI region, and appropriate host. Before introducing the microbe, organoids also need to model specific in vivo intestinal epithelium conditions such as undergoing differentiation to form specific IEC subtypes, a functional mucus layer, and a gradient oxygen level. These conditions facilitate the addition, adhesion, and replication of microbes in vitro, which can be modeled in various ways, each having its advantages and disadvantages. Staining of images in order of appearance: differentiation: blue, DAPI; red, MUC2; green, E-cadherin; mucus layer: blue, DAPI; red, E-cadherin; green, AAL; polarity reversal: blue, DAPI; red, villin; green, E-cadherin; 2D monolayer: blue, DAPI; red, actin; green, GFP S Typhimurium. DAPI, 4′,6-diamidino-2-phenylindole; EACC, Enteroid-anaerobe co-cuture system; GFP, Green fluorescent protein; IHAC, intestinal hemi-anaerobic co-culture system.
Choosing Your Stem Cells: Pluripotent or Adult?
The first and perhaps most important choice for researchers is whether they should use inducible pluripotent stem cells (iPSCs) or adult stem cells (ASCs) to generate intestinal organoids. Their creation and maintenance require distinct protocols, in concert with their structures and functions being fundamentally different.
iPSCs can be induced to differentiate and form organoids through the manipulation of complex developmental cues that include key interactions with extracellular matrix (ECM).9 The main advantage of working with iPSCs is the ability to generate organoids without the need for primary tissues, because obtaining human tissue biopsy specimens can be logistically challenging for many researchers. Despite their convenience, because iPSC-organoids are derived using developmental cues, they remain in a more fetal-like state and often are missing key features of a mature epithelium.10 Similarly, because the phenotype of these organoids is described as being less organ-specific, they may not exactly mimic the epithelium of a particular intestinal region.10 Most iPSC-derived organoids also contain mesenchymal cells as a by-product of their differentiation,9 although recent methodologic advancements can generate organoids free of mesenchymal cells.11 Notably, the presence of mesenchymal cells can be beneficial because they supply cues for IEC development and maintenance, yet their presence can hinder studies in which IEC–intrinsic responses are desired.
In contrast, ASCs are isolated from host tissues and, as a result, organoids derived from ASCs can provide the cellular diversity that is biologically representative of the originating tissue. In 2009, Hans Clevers and his group6 isolated leucine-rich repeat-containing G-protein–coupled receptor 5 (Lgr5)+ stem cells from intestinal crypts, culturing the cells in a complex ECM (Matrigel, Corning, #356255) with a precise supplementation of growth factors that enabled long-term propagation. ASC-derived small intestinal (enteroids) and large intestinal (colonoids) organoids were shown to mature into fully functional crypts and differentiated epithelium that provided a snapshot of the gut epithelium in vivo.12 Moreover, ASC organoids do not contain the mesenchymal cells found in iPSC organoids. Although obtaining the tissues necessary for human ASC organoids can be challenging, biobanks that serve as sources for tissues and organoids, while also providing donor information (tissue site, age, sex, disease), are growing in number and accessibility. Consequently, to study microbe interactions with the gut epithelium, ASC-derived organoids appear more relevant and convenient than iPSC-derived organoids and thus are the focus of this review.
Introducing Bacteria to the Organoid System: Choosing the Best Technique
Exploring the interactions that occur between billions of luminal bacteria and the intestinal epithelium is key to understanding gut function under homeostatic and disease states. As shown with gnotobiotic mice, in the absence of a microbiota, the gut epithelium undergoes abnormal development, increased metabolic stress and defects in barrier function, and mucin secretion.13,14 In contrast, enteric bacterial pathogens often directly infect IECs, causing physiological dysfunction as well as inflammatory responses that potentially contribute to many chronic diseases.15 Thus, it is fundamental to select an appropriate method to introduce the microbe of interest, largely keeping faithful to the in vivo interactions, while also considering cost and time.
Adding Bacterial Products to the Media
A healthy intestinal epithelium normally undergoes very limited direct interactions with the gut microbiota. Instead, bacterial products and metabolites diffuse out of the intestinal lumen to the mucosal surface where IECs sense bacterial-derived, microbe-associated molecular patterns or metabolites such as short-chain fatty acids (SCFAs). This can lead to differential gene expression and phenotypic changes in IEC stemness and their immune functions.16 With this in mind, ideally, organoids should undergo chronic, low-level stimulation with bacterial products to be more reflective of the in vivo intestinal epithelium.
Various strategies have been used to study the effects of commensal-derived products on IEC homeostasis such as the direct supplementation of SCFA to organoid media. Through transcriptomic analysis, this showed that butyrate, propionate, or SCFA mixtures enhanced IEC turnover and differentiation, in concert with more budding and reduced stem cell numbers by reducing histone deacetylase 3 activity.17,18 This effect was reversed by adding the bacterial metabolites phytate and inositol trisphosphate to the organoids, which stimulate histone deacetylase 3 activity.19 In both human and murine enteroids, butyrate promoted the production of retinoic acid, a critical signaling molecule for IEC development, homeostasis, and immunity.20,21 Microbe-associated molecular patterns also were found to have equally crucial roles in maintaining homeostasis, as was shown with SCFAs. For example, lipopolysaccharide (LPS) isolated from commensal bacteria activated Toll-like receptor 4 (TLR4) in colonoids, leading to inhibition of Lgr5+ stem cell–based IEC proliferation.22
In addition, products from bacterial pathogens can be used in place of live bacteria to study their involvement in IEC pathophysiology and innate immune responses, especially with secreted toxins and effectors that can act on IECs without the pathogen itself needing close contact. For example, Clostridium difficile, as an obligate anaerobe, can be difficult to culture. Researchers instead added the C difficile toxinA (TcdA) and toxin B (TcdB) to colonoids, finding they directly interact with the Wingless/Integrated (Wnt)-receptor Frizzled, as well as triggering caspase 3/7-induced apoptosis, while other forms of cell death were dispensable for protection against these toxins.23,24 Thus, bacterial products can partially replicate the effects of live bacteria, although adding these factors to intact 3D organoids (basolateral side of IECs) may not mimic their actions in vivo because bacteria and their products normally contact the apical surface of IECs.16 In other instances, it still is essential to work with live bacteria, especially in the case of those pathogens that cause disease through direct interactions with epithelial surfaces, such as by invading or adhering to IECs.
Fragmentation
Fragmentation involves recovering organoids from ECM, then mechanically disrupting them into fragments to be mixed and incubated with bacteria or bacterial products, before reseeding them back into the ECM. As a result, these fragments reassemble into 3D organoids with the introduced bacteria/products entrapped within their lumens,25 thereby permitting bacterial interactions with both the apical and basal sides of IECs. A study introducing muramyl dipeptide via organoid fragmentation showed the cytoprotective effects of muramyl dipeptide because it activated the nucleotide-binding oligomerization domain-containing protein (NOD) 2-receptor pathway as well as autophagy to protect Lgr5+ stem cells from oxidative stress, thereby protecting crypt homeostasis.26 Similarly, enteroid fragmentation and consequent proteomic analysis were used to show that infection with the pathogen Listeria monocytogenes altered innate host defenses, including the complement and coagulation cascades, Hypoxia-inducible factor-1 signaling, ferroptosis, and NOD-like receptor signaling.27 Although clearly this method can be used to study both homeostatic and pathogenic bacteria–organoid interactions, there are several limitations, including variation in the quantity of bacteria/products entrapped per organoid and the fact that this approach leads to bacterial interactions with both the apical and basolateral sides of IECs, hence the physiological relevance could be questioned.
Microinjection Into Organoid Lumens
Another way to introduce bacteria/products into enclosed 3D organoids is by microinjecting them into the organoid lumen. This method maintains the 3D organoid structure while ensuring contact between the bacteria and the physiologically relevant apical side of the epithelium.28 There are several studies outlining the standardization and optimization of this technique, including methods for calculating optimal injection volumes,29,30 the use of fluorescent bacteria to aid injection visualization and assessment,31,32 repeated microinjections,33 and even the construction of a high-throughput microinjection platform.34
Both commensal and pathogenic bacteria have been microinjected into organoids. Notably, when a mixture of commensal bacteria from stool filtrates was microinjected into the hypoxic lumens of colonoids, they maintained their species diversity for more than 96 hours.34 Moreover, the growth of the anaerobe Bifidobacterium adolescentis was sustained for at least 72 hours after microinjection, emphasizing the advantage of the hypoxic organoid lumen for maintaining commensal diversity and growth.34 For pathogens, Salmonella enterica serovar Typhimurium was observed to stimulate Paneth cell granule secretion (including α-defensins) in enteroids, but only when administered on the apical side, underscoring the importance of proper microbe orientation when studying pathogenesis.29 Another study investigated the effects of long-term, repeated microinjection of genotoxic Escherichia coli, and found that the injected enteroids acquired mutational changes comparable with the signatures of colorectal cancer, highlighting the carcinogenic effect of this E. coli.33 Taken together, the microinjection approach provides advantages over other methods by allowing researchers to study both commensal and pathogen interactions with IECs in an integral 3D structure. Other advantages include the ability to inject a precise dose of bacteria, as well as providing hypoxic conditions, and longer experimental time frames that enable the study of organoid proliferation and the emergence of IEC subtypes. However, microinjection can cause structural damage to organoids, while the high cost of the microinjection equipment and the technical challenges of this approach are not ideal for beginners, or for high-throughput applications.
Organoid Polarity Reversal
A recently described technique reverses the polarity of 3D organoids so that their apical side faces outward. Achieved by removing ECM proteins from the culture system, mature organoids are grown in suspension for 3 days to allow the organoids to turn inside out, with the final inverted organoids showing proper epithelial barrier integrity and nutrient uptake.
Once achieved, this approach bypasses the need for microinjection because microbes can be simply added to the culture media to directly interact with the organoid’s (now outward facing) apical surface. In the initial study describing this approach, researchers used 2 invasive pathogens, S. Typhimurium and L. monocytogenes to infect apical-out, basal-out, or mixed apical-/basal-out enteroids, and found that S. Typhimurium preferentially infected the apical surface whereas L. monocytogenes preferred the basal surface of the enteroids. These findings reinforced the concept that these 2 enteric pathogens use distinct strategies to invade a polarized epithelium.35 Similarly, enteroaggregative E. coli (EAEC) was found to preferentially attach to the apical side of mucin-secreting goblet cells on apical-out organoids.36 Thus, this technique models microbe interactions with the apical surface of IECs using a 3D structure and consequently facilitates the collection and assessment of chemokines and other secretions released from the apical side of IECs into the media during these interactions. A major limitation, however, is that organoids require approximately 3 days in suspension before they undergo reversal, thus reducing available experimental time frames. Other technical challenges include increased cell death and the fact that reversed organoids often adhere to each other owing to the lack of ECM. Moreover, because reversed organoids are grown in suspension, the difficulty of changing media can lead to unrestrained bacterial growth as well as the accumulation of toxins that could impact organoid viability negatively. These issues pose obstacles for researchers to design long-term, microbe–organoid experiments, making studies focused on homeostatic host–microbe interactions difficult to address using this approach.
2-Dimensional Monolayers and Air Liquid Interface
The most widely adapted method of introducing bacteria/products involves disrupting 3D organoids to allow IECs to form 2-dimensional (2D) monolayers. Sacrificing 3D structure, 2D organoid-derived monolayers provide a simplified yet relevant model that allows the application of many conventional cell culture techniques to evaluate IEC functionality, such as tight junctions, cell differentiation, and immune responses.37 Using Transwells to separate the apical and basal compartment furthers IEC differentiation12 and has the potential to incorporate additional host factors such as mesenchymal cells or immune cells to better mimic the in vivo environment. Upon apical infection with pathogenic L. monocytogenes, Shigella flexneri, or S. enterica Typhi, transepithelial electrical resistance and fluorescein isothiocyanate–dextran assays showed increased permeability in organoid monolayers. Infections also were accompanied by increased secretion (both basolateral and apical) of interleukin 8, along with other proinflammatory responses unique to each pathogen.38, 39, 40 Similarly, another study added flagellin to a colonoid-derived monolayer, and observed TLR5 activation promoted interleukin 8 secretion, which was amplified by an endoplasmic reticulum stress response.41 In contrast, the addition of other pathogenic bacterial products including lipoteichoic acid, as well as LPS, to enteroid-derived monolayers did not trigger IEC innate immune responses.42
A recent study showed an unfortunate limitation of the current 2D monolayer system: long-term culturing of organoids as monolayers generated low oxygen tension, leading to cellular stress, reduced differentiation, and a lack of secretory cells.43 Notably, these effects could be reversed by using air–liquid interface (ALI) culturing, where the apical side of the monolayer is exposed to air while the basal side remains submerged in media. ALI was found to reduce oxygen stress, thereby re-establishing healthy and differentiated monolayers with homeodomain-only protein homeobox (Hopx+) quiescent stem cells.43,44
Taken together, 2D organoid-derived monolayers provide the simplest approach to study microbe–IEC interactions and offer the potential to measure the apical release of host protective factors such as mucins. Nonetheless, this method requires a large number of IECs for the initial setup and may not accurately depict IEC proliferation, migration, and death. With the development of more technologies geared toward 3D applications, such as light-sheet and 2-photon microscopy,45 as well as microfluidic chip technology that permits the morphogenesis of tube-shaped intestinal structures with an intact lumen and the addition of 3D vascular structures,46, 47, 48 the field eventually could create a niche that integrates both 2D and 3D technologies. This would yield improved and more relevant in vitro models to further enhance host–microbe interaction studies.
Modeling Age and Site Specifications of the Intestinal Epithelium
Many microbes require specific structural and functional features to interact with their host epithelium, which could be impacted if the in vitro organoid model does not re-create relevant in vivo conditions. This can involve mimicking an appropriate developmental stage, a specific GI region or IEC subtype, the mucus layer, and proper oxygen levels.
Development and Organization
A large gap exists in our understanding of how the human intestine functions in early life, especially how its development is impacted by microbiota colonization. Early colonization with pathogenic microbes or impaired maturation could lead to profound developmental defects and disorders such as necrotizing enterocolitis.49 Organoids offer the potential to model some of these processes and improve our understanding of neonatal intestinal diseases.
The neonatal intestinal epithelium undergoes distinctive developmental changes when it is exposed to different microbial populations. A murine neonatal enteroid study suggests that IEC maturation to the adult state depends on intrinsic factors, largely independent of the microbiota, when the enteroids are maintained in culture over 4 weeks.50 In contrast, a separate study showed that commensal metabolites isolated from baby fecal filtrates up-regulated growth pathways in murine enteroids, facilitating stem cell proliferation.51 In comparison with human adult enteroids, human fetal enteroids show reduced markers of IEC maturation, as well as altered immune responses and barrier function. Only late-stage fetal enteroids are responsive to LPS and heat-killed bacteria, whereas early stage fetal enteroids remain unresponsive when compared with adult enteroids.52 Furthermore, fetal IEC immune responses are attenuated when in the presence of indole-3-lactic acid derived from Bifidobacterium longum infantis, a metabolite found in breastmilk.53 These studies highlight the differential responses between immature and mature IECs and emphasize the need for further studies on the role of microbes in IEC development (Figure 2).
Figure 2.
Maturation, function, location, and species specificity of intestinal organoids and bacterial interactions. (A) Schematic representation of human IEC maturation and function of intestinal organoids relative to developmental stages. (B) Schematic representation of organoid bacterial interactions relative to species and GI locations. Number of (+) signs indicate the degree of the response, (-) negative signs indicate no response, and N/A indicates no data available. FliC, flagellin; Pam3CSK4, Pam3CysSerLys4.
As the GI tract matures, it differentiates into segments with unique functions, cell populations, and microbiota composition. Consequently, organoids isolated from each GI region (enteroids, colonoids, and gasteroids) show corresponding differences in IEC composition, function, and architecture, which also can be species specific.12,54,55 For example, enteroids can contain Paneth cells, which are not found in colonoids, and Paneth cells, both in vivo and in vitro, secrete antimicrobial peptides that can curb S Typhimurium growth.32,56,57 Likewise, the enteric pathogen EAEC preferentially interacts with organoids derived from certain GI regions, with site-specific adherence patterns to duodenal and jejunal enteroids, and much less adherence to ileal enteroids or colonoids. The inflammatory responses from each segment also varied, although it seemed largely dependent on the level of EAEC infection.36,58 Moreover, human and murine organoids from different GI regions express varying levels of pattern recognition receptors, and thus their responses to bacterial stimulation differ. Although organoids from all sites express TLR4, only murine gasteroids and colonoids respond to LPS owing to their expression of the accessory protein CD14, whereas human organoids from other regions of the GI tract were completely unresponsive owing to their lack of CD14.55,59 These results highlight the importance of selecting organoids originating from the appropriate GI segment and species when studying specific bacteria–organoid interactions because fundamental differences exist between human and murine organoids, as well as between those collected from different GI segments. These differences likely will lead to varying degrees of bacterial adherence and growth, while also causing discrepancies in host responses (Figure 2).
IEC Subtypes
Modeling host–microbe interactions reflective of the in vivo situation demands the development of organoid systems containing mature and functional IECs, including the enrichment of IEC subtypes to investigate more specific interactions.60 Unfortunately, many widely used protocols generate organoids containing only semidifferentiated IECs that lack rarer IEC subtypes.54,56 The incorporation of differentiation factors such as DAPT, a Notch signaling inhibitor, and CHIR99021, a Wnt signaling potentiator in organoid media, promotes differentiation and enriches for Paneth cells within enteroids.61, 62, 63, 64 Supplementation with stem cell niche factors insulin growth factor-1 and fibroblast growth factor also can preserve the in vivo cellular diversity and self-renewing capacity of intestinal crypts.65
Specific cell-type–enriched organoids have been used when modeling enteric pathogens that target specific IEC subtypes. For instance, supplementing enteroids with tumor necrosis factor α and receptor activator of nuclear factor-κB ligand (NFκB) enriches for M cells, and such enteroids were used previously to confirm that S flexneri use M cells as portals for invasive entry.39 In addition to Paneth cells61,66,67 and M cells,39,68, 69, 70 researchers successfully have enriched organoids for goblet cells,62,66,67 tuft cells,71 and enteroendocrine cells.72,73 Together, these enriched organoids provide useful models when investigating cell-type–specific interactions with bacteria. Nonetheless, it is important to consider whether a phenotype observed in these enriched organoids may be an indirect effect of their specialized media because many of the growth factors and inhibitors used to manipulate IEC differentiation also may affect other signaling pathways.67 In addition, enriched organoids develop crypt-like structures leading to reduced lumen volume, consequently affecting the ability to introduce microbes, particularly through microinjection, because this procedure is largely dependent on the volume of bacterial culture delivered into the organoid lumen.
Modeling the Mucus Layer
The intestinal mucus layer is a dynamic and highly complex matrix that physically separates intestinal luminal contents from the underlying epithelium.74 It plays an active role in controlling microbial interactions with IECs because its heavily glycosylated mucin proteins are critical for bacterial adhesion and also act as a nutrient source for commensals and pathogens alike.13,75 Thus, as we develop more accurate host–bacterial organoid models that include different IEC subtypes, it is equally important to develop functional goblet cells able to generate a mucus layer.
Many studies remove Wnt ligands from the media to induce organoid-derived monolayers to produce a mucus layer. In a recent study, human colonoid monolayers formed a mucus layer 5 days after Wnt removal and, interestingly, the bacterial pathogen enterohemorrhagic E coli (EHEC) preferred infecting these monolayers over undifferentiated monolayers, suggesting a role for mucus as an anchor or nutrient source for EHEC.76 Similarly, mucus-producing, differentiated, colonoid monolayers were used to define the role of an EAEC-produced serine protease, showing that its ability to degrade organoid-produced mucus improved bacterial penetration and colonization of the underlying IECs.77 Mucus-producing, enteroid-derived monolayers also have been used to model C difficile–induced enteritis and confirmed a protective role for mucus (Muc2) because the addition of Muc2 delayed C difficile–derived toxins from reaching the monolayers.78 These results highlight that an intact mucus layer can alter bacterial pathogenesis in vitro and thus its addition may aid in understanding complex enteric pathogen–host interactions.
Additional techniques now are being used to enrich for mucus in organoid models. One method mimics commensal cues via the addition of SCFAs. SCFAs epigenetically modify the enteroids, leading to concentration-dependent increases in the expression of goblet cell markers, as well as common mucus constituents and antimicrobial peptides.18 Notably, it remains unclear if the mucus produced by these monolayers truly reflects the in vivo mucus layer. Mucus should be largely impenetrable to commensal bacteria, show a complex glycosylation pattern, as well as contain an array of mucus-associated proteins as shown in Table 1. Moreover, the mucus layer should sustain its own renewal by tightly regulating its production, secretion, and degradation. Thus, it is crucial to validate the composition (glycosylation, proteins) and functionality (permeability, secretion, production) of mucus layers produced in vitro. Currently, there are few studies in which organoids are characterized for the presence and function of goblet cells or a mucus layer, however, several methods used in other systems may be applicable and are summarized in Table 1.
Table 1.
Overview of Techniques Used to Evaluate Goblet Cell and Mucus Function
| Techniques | Used on | Compatible organoid method | Goblet cell properties/mucus functions | Reference |
|---|---|---|---|---|
| Immunofluorescent staining for mucus composition | In vitro ALI monolayer; in vivo mouse colon staining | 2D monolayer; 3D organoids | Expression of mucus protein components MUC2, MUC5AC, TFF3, AGR2, RELMβ, FCGBP, CLCA1, and ZG16 | 75,90 |
| Isotope label of mucin | In vivo and ex vivo mouse colon and mucus | 2D monolayer | Mucin synthesis and production after 3H-glucosamine injection | 91 |
| GalNAz label of mucin (Click-iT) | In vivo mouse small intestine | 2D monolayer; 3D organoids | Mucin synthesis and production by labeling mucin glycoproteins | 92, 93, 94 |
| Secretagogue stimulation | Ex vivo mouse colon | 2D monolayer; 3D organoids | Mucin secretion and goblet cell maturity stimulated by acetylcholine or analogs | 95,96 |
| Activated charcoal and micropipettor | Ex vivo mouse colon | 2D monolayer | Mucus thickness measured by adding charcoal particles on mucus apical surface | 75,95, 96, 97 |
| Fluorescent beads | In vitro monolayer; ex vivo mouse intestine | 2D monolayer | Mucus thickness measured by distance of beads to epithelium | 90,94,98 |
| Penetrable beads | In vitro monolayer; ex vivo mouse intestine | 2D monolayer | Mucus permeability to bacteria-sized fluorescent beads | 75,95,96 |
| LC-MSMS | In vivo mouse colon mucus | 2D monolayer | Proteomic analysis of the mucus layer | 75,94,98 |
AGR2, anterior gradient 2; CLCA1, chloride channel regulator, calcium-activated-1; FCGBP, IgGFc-binding protein; GalNAz, N-azidoacetylgalactosamine; LC-MSMS, liquid chromatography with tandem mass spectrometry; MUC2, mucin 2; MUC5AC, mucin5AC; RELMβ, resistin-like molecule β; TFF3, Trefoil Factor 3; ZG16, zymogen granule membrane protein 16.
Oxygen Levels and Anaerobic Bacteria
The majority of intestinal commensal bacteria are facultative or obligate anaerobes that can survive only in environments containing minimal oxygen.79,80 Correspondingly, the colonic lumen is hypoxic under healthy conditions.81 In contrast, most researchers perform their bacteria-organoid studies using ambient incubator conditions of 18%–21% O2.82 O2 levels are affected further by factors such as ALI and ECM density,44 resulting in oxygen gradients that are not representative of in vivo conditions.81 Thus, researchers need to develop systems that create a dynamic oxygen gradient from the apical to the basolateral sides of IEC monolayers, for example, growing cultures in modified anaerobic chambers. The enteroid–anaerobe co-culture system (EACC), as well as the intestinal hemi-anaerobic co-culture system (iHAC), recently were developed to establish a physiological hypoxia gradient for tissue culture.83,84 Both systems deliver oxygen to the basolateral side of the monolayer, with IECs consuming the O2, preventing it from reaching the apical side, rendering the apical region hypoxic. Notably, this O2 gradient promotes increased IEC barrier integrity and facilitates the survival of obligate anaerobes such as Bacteroides thetaiotaomicron and Blautia species for up to 24 hours. B adolescentis, Bacteroides fragilis, Clostridium butyricum, and Akkermansia muciniphila also can survive for 48 hours in this system, driving IECs toward a more mature and immunotolerant state.83,84 These 2 robust models re-create the oxygen gradient seen in vivo to address interactions between anaerobic microbes and gut epithelium, and define the extent to which disrupting this system can result in dysbiosis and intestinal inflammatory disorders.79,81,85
IEC Intrinsic Immune Responses to Bacteria Using Organoids
Aside from their other functions, IECs serve as a central hub in mucosal innate immunity, expressing an array of innate receptors that sense various microbial ligands from the gut lumen and respond through the release of antimicrobial peptides and cytokines. IEC responses often shape subsequent immune responses from underlying immune cells.55,59 Interestingly, many recent studies have shown that the innate immune responses of ASC-derived organoids to commensal and pathogenic bacteria are very different from those obtained using cancer cell lines.
Aside from the TLRs and NOD receptors mentioned earlier, IECs also use intrinsic inflammasomes to sense and respond to invading pathogens. In vivo activation of inflammasomes during S Typhimurium infection selectively expels infected IECs into the intestinal lumen.86 Recently, we confirmed these responses reflect IEC intrinsic inflammasomes, by infecting murine colonoid-derived monolayers with S Typhimurium. It was notable that the monolayers did not express the same levels of inflammatory caspases as in vivo until they were treated with interferon (IFN)-γ, which helped IECs reach a state of heightened immune activation. This led the treated IECs to express the same inflammatory caspases as their in vivo counterparts, and efficiently extrude infected cells.87 In a separate study, we also identified differences in the preferential activation of these inflammatory caspases between human and murine enteroids upon their infection with S Typhimurium.88 Because inflammasomes are expressed by many cell types such as macrophages and neutrophils, studying enteroids helps clarify the role of IEC intrinsic signaling and allows side-by-side comparison between human beings and mice. This further emphasizes the importance of choosing the appropriate species and stimulation when using organoids to study host–microbe interactions because findings made with cells from 1 species or microbe may not prove relevant under other conditions. Moreover, it highlights that to model the in vivo epithelium, baseline IEC responses should be assessed in the context of integrating commensal microbes,42,55,59 as well as immune cells to organoid models, a subject that was reviewed recently and is summarized in Table 2.89
Table 2.
List of Organoid–Immune Cell Co-culture Studies
| Co-culture | Organoid type | Immune cell used | Microbe | Technique | Findings | Reference |
|---|---|---|---|---|---|---|
| Organoids and immune cells | Human fetal enteroids, colonoids | Fetal CD4+ Tem cells | NA | Mix in Matrigel | Low numbers of CD4+ Tem cells support stem cell growth, while high numbers impair IEC development via TNF-α | 99 |
| Murine enteroids | Th1, Th2, Th17, iTreg cells | NA | In media; mix in Matrigel | MHCII on Lgr5+ ISCs interact with Th and Treg cells, each producing different cytokines to regulate ISC renewal and differentiation | 100 | |
| Murine enteroids | ILC3 | NA | Mix in Matrigel | ILC3 produces IL22 to promote ISC proliferation (via STAT3 signaling) after tissue damage | 101 | |
| Murine enteroids | ILC2-enriched MLN cells | NA | Transwell | IL33 induces ILC2 to produce IL13, a cytokine that enhances IEC differentiation to goblet cells | 102 | |
| Murine enteroids | IEL | NA | Mix in Matrigel | IELs propagate and could migrate along the basolateral surface of IEC into and out of the enteroids | 103 | |
| Murine enteroids | CD4+, CD8+ T cells | NA | Mix in Matrigel | T cells can be co-cultured long term with enteroids and acquire IEL-like migration and expression patterns | 104 | |
| Mouse and human organoids | T cells, DCs | NA | Mix in Matrigel | Dysregulated T-cell activation from bone marrow transplant leads to increased IFNγ production and ISC injury | 105 | |
| Organoids, immune cells, and microbes | Human enteroid monolayer | Human PBMC-derived macrophages | EPEC, ETEC | Transwell | Macrophages project through Transwell filter to interact and phagocytose ETEC and EPEC found on the apical side of the enteroid monolayers | 106 |
| Human enteroid monolayer | PBMC |
C difficile toxin A, E coli O6 |
Transwell (ALI) | Mucus layer prevented toxin-mediated junction disruptions and bacteria-induced cytokine secretions from PBMCs | 90 |
DC, dendritic cell; EPEC, Enteropathogenic E coli; ETEC, Enterotoxigenic E coli; IEL, intraepithelial lymphocyte; IFN, interferon; IL, interleukin; ILC, innate lymphoid cell; ISC, intestinal stem cell; iTreg, induced T regulatory cells; MHCII, Major histocompatibility complex II; MLN, mesenteric lymph node; PBMC, peripheral blood mononuclear cell; STAT3, Signal transducer and activator transcription 3; Tem, T-effector memory cells; Th, T helper cells; TNF, tumor necrosis factor.
Conclusions
Taken together, it is clear that intestinal organoids offer impressive potential to accurately model in vivo bacteria–IEC interactions. Many variables need to be taken into consideration when using organoids: the stem cell origin, the tissue location, the culturing method, not to mention all the complexities regarding the heterogeneity of human samples such as sex, age, ethnicity, and disease-related status. Although new organoid models have tackled several of these issues, including improving our ability to introduce microbes, stimulants, or immune cells, as well as produce mucus and control O2 levels, there are still several physiologically relevant factors that are missing, such as mechanical forces from luminal flow and peristalsis. Consequently, it is essential to continue exploring how to generate more relevant features of the native gut epithelium in organoids, to better define their interactions with gut microbes. Another pressing issue is to develop methods to generate organoids on a larger scale in a more reproducible, accessible, and inexpensive fashion. Acknowledging the impressive advances already made, organoids will be a key translational platform to discovering personalized treatments unique to individual patients.
Acknowledgments
Author contributions
Xiao Han reviewed the concept and design, performed the literature review, drafted the manuscript, and approved the final manuscript; Matthias Mslati, Emily Davies, and Yan Chen performed the literature review, drafted the manuscript, and approved the final manuscript; Joannie M. Allaire was responsible for study supervision, literature review, critical revision of the manuscript for important intellectual content, and approved the final manuscript; and Bruce A. Vallance was responsible for study supervision, critical revision of the manuscript for important intellectual content, and approved the final manuscript.
Footnotes
Conflicts of interest The authors disclose no conflicts.
Funding Supported by operating grants from the Canadian Institutes of Health Research (PJT-148846 and 159528) and Crohn’s and Colitis Canada (B.A.V.), and by the Michael Smith Foundation for Health Research and the Canadian Institutes of Health Research fellowships (J.M.A.).
Contributor Information
Joannie M. Allaire, Email: jallaire@bcchr.ca.
Bruce A. Vallance, Email: bvallance@cw.bc.ca.
References
- 1.Sekirov I., Russell S.L., Caetano M., Antunes L., Finlay B.B. Gut microbiota in health and disease. Physiol Rev. 2010;90:859–904. doi: 10.1152/physrev.00045.2009. [DOI] [PubMed] [Google Scholar]
- 2.Kinross J.M., Darzi A.W., Nicholson J.K. Gut microbiome-host interactions in health and disease. Genome Med. 2011;3:1–12. doi: 10.1186/gm228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Cianci R., Pagliari D., Piccirillo C.A., Fritz J.H., Gambassi G. The microbiota and immune system crosstalk in health and disease. Mediators Inflamm. 2018;2018:2912539. doi: 10.1155/2018/2912539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Peterson L.W., Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol. 2014;14:141–153. doi: 10.1038/nri3608. [DOI] [PubMed] [Google Scholar]
- 5.Allaire J.M., Morampudi V., Crowley S.M., Stahl M., Yu H., Bhullar K., Knodler L.A., Bressler B., Jacobson K., Vallance B.A. Frontline defenders: goblet cell mediators dictate host-microbe interactions in the intestinal tract during health and disease. Am J Physiol Gastrointest Liver Physiol. 2018;314:G360–G377. doi: 10.1152/ajpgi.00181.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Sato T., Vries R.G., Snippert H.J., Van De Wetering M., Barker N., Stange D.E., Van Es J.H., Abo A., Kujala P., Peters P.J., Clevers H. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262–265. doi: 10.1038/nature07935. [DOI] [PubMed] [Google Scholar]
- 7.Ootani A., Li X., Sangiorgi E., Ho Q.T., Ueno H., Toda S., Sugihara H., Fujimoto K., Weissman I.L., Capecchi M.R., Kuo C.J. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche. Nat Med. 2009;15:701–706. doi: 10.1038/nm.1951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ramani S., Crawford S.E., Blutt S.E., Estes M.K. Human organoid cultures: transformative new tools for human virus studies. Curr Opin Virol. 2018;29:79–86. doi: 10.1016/j.coviro.2018.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Spence J.R., Mayhew C.N., Rankin S.A., Kuhar M.F., Vallance J.E., Tolle K., Hoskins E.E., Kalinichenko V.V., Wells S.I., Zorn A.M., Shroyer N.F., Wells J.M. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 2011;470:105–110. doi: 10.1038/nature09691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Watanabe N., Santostefano K.E., Yachnis A.T., Terada N. A pathologist’s perspective on induced pluripotent stem cells. Lab Invest. 2017;97:1126–1132. doi: 10.1038/labinvest.2017.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mithal A., Capilla A., Heinze D., Berical A., Villacorta-Martin C., Vedaie M., Jacob A., Abo K., Szymaniak A., Peasley M., Stuffer A., Mahoney J., Kotton D.N., Hawkins F., Mostoslavsky G. Generation of mesenchyme free intestinal organoids from human induced pluripotent stem cells. Nat Commun. 2020;11:215. doi: 10.1038/s41467-019-13916-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kozuka K., He Y., Koo-McCoy S., Kumaraswamy P., Nie B., Shaw K., Chan P., Leadbetter M., He L., Lewis J.G., Zhong Z., Charmot D., Balaa M., King A.J., Caldwell J.S., Siegel M. Development and characterization of a human and mouse intestinal epithelial cell monolayer platform. Stem Cell Rep. 2017;9:1976–1990. doi: 10.1016/j.stemcr.2017.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Donohoe D.R., Garge N., Zhang X., Sun W., O’Connell T.M., Bunger M.K., Bultman S.J. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011;13:517–526. doi: 10.1016/j.cmet.2011.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Birchenough G.M.H., Johansson M.E.V., Gustafsson J.K., Bergström J.H., Hansson G.C. New developments in goblet cell mucus secretion and function. Mucosal Immunol. 2015;8:712–719. doi: 10.1038/mi.2015.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Frank D.N., St. Amand AL, Feldman R.A., Boedeker E.C., Harpaz N., Pace N.R. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A. 2007;104:13780–13785. doi: 10.1073/pnas.0706625104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Venegas D.P., De La Fuente M.K., Landskron G., González M.J., Quera R., Dijkstra G., Harmsen H.J.M., Faber K.N., Hermoso M.A. Short chain fatty acids (SCFAs) mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019;10:1486. doi: 10.3389/fimmu.2019.01486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kaiko G.E., Ryu S.H., Koues O.I., Collins P.L., Solnica-Krezel L., Pearce E.J., Pearce E.L., Oltz E.M., Stappenbeck T.S. The colonic crypt protects stem cells from microbiota-derived metabolites. Cell. 2016;165:1708–1720. doi: 10.1016/j.cell.2016.05.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Pearce S.C., Weber G.J., Van Sambeek D.M., Soares J.W., Racicot K., Breault D.T. Intestinal enteroids recapitulate the effects of short-chain fatty acids on the intestinal epithelium. PLoS One. 2020;15 doi: 10.1371/journal.pone.0230231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Wu S., Hashimoto-Hill S., Woo V., Eshleman E.M., Whitt J., Engleman L., Karns R., Denson L.A., Haslam D.B., Alenghat T. Microbiota-derived metabolite promotes HDAC3 activity in the gut. Nature. 2020;586:108–112. doi: 10.1038/s41586-020-2604-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Schilderink R., Verseijden C., Seppen J., Muncan V., van den Brink G.R., Lambers T.T., van Tol E.A., de Jonge W.J. The SCFA butyrate stimulates the epithelial production of retinoic acid via inhibition of epithelial HDAC. Am J Physiol Liver Physiol. 2016;310:G1138–G1146. doi: 10.1152/ajpgi.00411.2015. [DOI] [PubMed] [Google Scholar]
- 21.Lukonin I., Serra D., Challet Meylan L., Volkmann K., Baaten J., Zhao R., Meeusen S., Colman K., Maurer F., Stadler M.B., Jenkins J., Liberali P. Phenotypic landscape of intestinal organoid regeneration. Nature. 2020;586:275–280. doi: 10.1038/s41586-020-2776-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Naito T., Mulet C., Castro C De, Molinaro A., Saffarian A., Nigro G., Bérard M., Clerc M., Pedersen A.B., Sansonetti P.J., Thierry P. Lipopolysaccharide from crypt-specific core microbiota modulates the colonic epithelial proliferation-to-differentiation balance. mBio. 2017;8:1–16. doi: 10.1128/mBio.01680-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Tao L., Zhang J., Meraner P., Tovaglieri A., Wu X., Gerhard R., Zhang X., Stallcup W.B., Miao J., He X., Hurdle J.G., Breault D.T., Brass A.L., Dong M. Frizzled proteins are colonic epithelial receptors for C. difficile toxin B. Nature. 2016;538:350–355. doi: 10.1038/nature19799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Saavedra P.H.V., Huang L., Ghazavi F., Kourula S., Vanden Berghe T., Takahashi N., Vandenabeele P., Lamkanfi M. Apoptosis of intestinal epithelial cells restricts Clostridium difficile infection in a model of pseudomembranous colitis. Nat Commun. 2018;9:4846. doi: 10.1038/s41467-018-07386-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Dutta D., Clevers H. Organoid culture systems to study host–pathogen interactions. Curr Opin Immunol. 2017;48:15–22. doi: 10.1016/j.coi.2017.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Levy A., Stedman A., Deutsch E., Donnadieu F., Virgin H.W., Sansonetti P.J., Nigro G. Innate immune receptor NOD2 mediates LGR5+ intestinal stem cell protection against ROS cytotoxicity via mitophagy stimulation. Proc Natl Acad Sci U S A. 2020;117:1994–2003. doi: 10.1073/pnas.1902788117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Huang J, Zhou C, Zhou G, Ye K. TMT-based quantitative proteomic analysis of intestinal organoids infected by Listeria monocytogenes with different virulence, bioRxiv 2020;164061. [DOI] [PMC free article] [PubMed]
- 28.Bartfeld S., Clevers H. Organoids as model for infectious diseases: culture of human and murine stomach organoids and microinjection of helicobacter pylori. J Vis Exp. 2015;105:53359. doi: 10.3791/53359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Yokoi Y., Nakamura K., Yoneda T., Kikuchi M., Sugimoto R., Shimizu Y., Ayabe T. Paneth cell granule dynamics on secretory responses to bacterial stimuli in enteroids. Sci Rep. 2019;9:1–10. doi: 10.1038/s41598-019-39610-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Engevik M.A., Aihara E., Montrose M.H., Shull G.E., Hassett D.J., Worrell R.T. Loss of NHE3 alters gut microbiota composition and influences Bacteroides thetaiotaomicron growth. Am J Physiol Gastrointest Liver Physiol. 2013;305:G697–G711. doi: 10.1152/ajpgi.00184.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhang Y.G., Wu S., Xia Y., Sun J. Salmonella-infected crypt-derived intestinal organoid culture system for host–bacterial interactions. Physiol Rep. 2014;2:e12147. doi: 10.14814/phy2.12147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wilson S.S., Tocchi A., Holly M.K., Parks W.C., Smith J.G. A small intestinal organoid model of non-invasive enteric pathogen-epithelial cell interactions. Mucosal Immunol. 2015;8:352–361. doi: 10.1038/mi.2014.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Pleguezuelos-Manzano C., Puschhof J., Huber A.R., Van Hoeck A., Wood H.M., Nomburg J., Gurjao C., Manders F., Dalmasso G., Stege P.B., Paganelli F.L., Geurts M.H., Beumer J., Mizutani T., Van Der Linden R., Van Elst S., England G., Consortium R., Top J., Willems R.J.L., Giannakis M., Bonnet R., Quirke P., Meyerson M., Cuppen E., Van Boxtel R., Clevers H. Mutational signature in colorectal cancer caused by genotoxic pks + E. coli. Nature. 2020;580:269–273. doi: 10.1038/s41586-020-2080-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Williamson I.A., Arnold J.W., Samsa L.A., Gaynor L., DiSalvo M., Cocchiaro J.L., Carroll I., Azcarate-Peril M.A., Rawls J.F., Allbritton N.L., Magness S.T. A high-throughput organoid microinjection platform to study gastrointestinal microbiota and luminal physiology. Cell Mol Gastroenterol Hepatol. 2018;6:301–319. doi: 10.1016/j.jcmgh.2018.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Co J.Y., Margalef-Català M., Li X., Mah A.T., Kuo C.J., Monack D.M., Amieva M.R. Controlling epithelial polarity: a human enteroid model for host-pathogen interactions. Cell Rep. 2019;26:2509–2520.e4. doi: 10.1016/j.celrep.2019.01.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Rajan A., Robertson M.J., Carter H.E., Poole N.M., Clark J.R., Green S.I., Criss Z.K., Zhao B., Karandikar U., Xing Y., Margalef-Català M., Jain N., Wilson R.L., Bai F., Hyser J.M., Petrosino J., Shroyer N.F., Blutt S.E., Coarfa C., Song X., Prasad B.V., Amieva M.R., Grande-Allen J., Estes M.K., Okhuysen P.C., Maresso A.W., Enteroaggregative E. coli adherence to human heparan sulfate proteoglycans drives segment and host specific responses to infection. PLOS Pathog. 2020;16 doi: 10.1371/journal.ppat.1008851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Foulke-Abel J., In J., Kovbasnjuk O., Zachos N.C., Ettayebi K., Blutt S.E., Hyser J.M., Zeng X.L., Crawford S.E., Broughman J.R., Estes M.K., Donowitz M. Human enteroids as an ex-vivo model of host–pathogen interactions in the gastrointestinal tract. Exp Biol Med. 2014;239:1124–1134. doi: 10.1177/1535370214529398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Roodsant T., Navis M., Aknouch I., Renes I.B., van Elburg R.M., Pajkrt D., Wolthers K.C., Schultsz C., van der Ark K.C.H., Sridhar A., Muncan V. A human 2D primary organoid-derived epithelial monolayer model to study host-pathogen interaction in the small intestine. Front Cell Infect Microbiol. 2020;10:272. doi: 10.3389/fcimb.2020.00272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ranganathan S., Doucet M., Grassel C.L., Delaine-Elias B., Zachos N.C., Barry E.M. Evaluating Shigella flexneri pathogenesis in the human enteroid model. Infect Immun. 2019;87:e00740–e00818. doi: 10.1128/IAI.00740-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Nickerson K.P., Senger S., Zhang Y., Lima R., Patel S., Ingano L., Flavahan W.A., Kumar D.K.V., Fraser C.M., Faherty C.S., Sztein M.B., Fiorentino M., Fasano A. Salmonella typhi colonization provokes extensive transcriptional changes aimed at evading host mucosal immune defense during early infection of human intestinal tissue. EBioMedicine. 2018;31:92–109. doi: 10.1016/j.ebiom.2018.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Rees W.D., Stahl M., Jacobson K., Bressler B., Sly L.M., Vallance B.A., Steiner T.S. Enteroids derived from inflammatory bowel disease patients display dysregulated endoplasmic reticulum stress pathways, leading to differential inflammatory responses and dendritic cell maturation. J Crohns Colitis. 2020;14:948–961. doi: 10.1093/ecco-jcc/jjz194. [DOI] [PubMed] [Google Scholar]
- 42.Ruan W., Engevik M.A., Chang-Graham A.L., Danhof H.A., Goodwin A., Engevik K.A., Shi Z., Hall A., Rienzi S.C.D., Venable S., Britton R.A., Hyser J., Versalovic J. Enhancing responsiveness of human jejunal enteroids to host and microbial stimuli. J Physiol. 2020;598:3085–3105. doi: 10.1113/JP279423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wang Y., Chiang I.L., Ohara T.E., Fujii S., Cheng J., Muegge B.D., Ver Heul A., Han N.D., Lu Q., Xiong S., Chen F., Lai C.W., Janova H., Wu R., Whitehurst C.E., VanDussen K.L., Liu T.C., Gordon J.I., Sibley L.D., Stappenbeck T.S. Long-term culture captures injury-repair cycles of colonic stem cells. Cell. 2019;179:1144–1159.e15. doi: 10.1016/j.cell.2019.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.DiMarco R.L., Su J., Yan K.S., Dewi R., Kuo C.J., Heilshorn S.C. Engineering of three-dimensional microenvironments to promote contractile behavior in primary intestinal organoids. Integr Biol (Camb) 2014;6:127–142. doi: 10.1039/c3ib40188j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Dekkers J.F., Alieva M., Wellens L.M., Ariese H.C.R., Jamieson P.R., Vonk A.M., Amatngalim G.D., Hu H., Oost K.C., Snippert H.J.G., Beekman J.M., Wehrens E.J., Visvader J.E., Clevers H., Rios A.C. High-resolution 3D imaging of fixed and cleared organoids. Nat Protoc. 2019;14:1756–1771. doi: 10.1038/s41596-019-0160-8. [DOI] [PubMed] [Google Scholar]
- 46.Bein A., Shin W., Jalili-firoozinezhad S., Park M.H., Sontheimer-phelps A., Tovaglieri A., Chalkiadaki A., Kim H.J., Ingber D.E. Microfluidic organ-on-a-chip models of human intestine. Cell Mol Gastroenterol Hepatol. 2018;5:659–668. doi: 10.1016/j.jcmgh.2017.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Brassard J.A., Nikolaev M., Hübscher T., Hofer M., Lutolf M.P. Recapitulating macro-scale tissue self-organization through organoid bioprinting. Nat Mater. 2021;20:22–29. doi: 10.1038/s41563-020-00803-5. [DOI] [PubMed] [Google Scholar]
- 48.Nikolaev M., Mitrofanova O., Broguiere N., Geraldo S., Dutta D., Tabata Y., Elci B., Brandenberg N., Kolotuev I., Gjorevski N., Clevers H., Lutolf M.P. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature. 2020;585:574–578. doi: 10.1038/s41586-020-2724-8. [DOI] [PubMed] [Google Scholar]
- 49.Niño F Diego, Chhinder P., Sodhi H.J.D. Necrotizing enterocolitis: new insights into pathogenesis and mechanisms. Physiol Behav. 2016;176:139–148. doi: 10.1038/nrgastro.2016.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Navis M., Martins Garcia T., Renes I.B., Vermeulen J.L., Meisner S., Wildenberg M.E., van den Brink G.R., van Elburg R.M., Muncan V. Mouse fetal intestinal organoids: new model to study epithelial maturation from suckling to weaning. EMBO Rep. 2019;20:1–12. doi: 10.15252/embr.201846221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Dougherty M.W., Kudin O., Mühlbauer M., Neu J., Gharaibeh R.Z., Jobin C. Gut microbiota maturation during early human life induces enterocyte proliferation via microbial metabolites. BMC Microbiol. 2020;20:205. doi: 10.1186/s12866-020-01892-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Senger S., Ingano L., Freire R., Anselmo A., Zhu W., Sadreyev R., Walker W.A., Fasano A. Human fetal-derived enterospheres provide insights on intestinal development and a novel model to study necrotizing enterocolitis (NEC) Cell Mol Gastroenterol Hepatol. 2018;5:549–568. doi: 10.1016/j.jcmgh.2018.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Meng D., Sommella E., Salviati E., Campiglia P., Ganguli K., Djebali K., Zhu W., Walker W.A. Indole-3-lactic acid, a metabolite of tryptophan, secreted by Bifidobacterium longum subspecies infantis is anti-inflammatory in the immature intestine. Pediatr Res. 2020;88:209–217. doi: 10.1038/s41390-019-0740-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Middendorp S., Schneeberger K., Wiegerinck C.L., Mokry M., Akkerman R.D.L., Van Wijngaarden S., Clevers H., Nieuwenhuis E.E.S. Adult stem cells in the small intestine are intrinsically programmed with their location-specific function. Stem Cells. 2014;32:1083–1091. doi: 10.1002/stem.1655. [DOI] [PubMed] [Google Scholar]
- 55.Kayisoglu O., Weiss F., Niklas C., Pierotti I., Pompaiah M., Wallaschek N., Germer C.-T., Wiegering A., Bartfeld S. Location-specific cell identity rather than exposure to GI microbiota defines many innate immune signalling cascades in the gut epithelium. Gut. 2021;70:687–697. doi: 10.1136/gutjnl-2019-319919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Stelzner M., Helmrath M., Dunn J.C.Y., Henning S.J., Houchen C.W., Kuo C., Lynch J., Li L., Magness S.T., Martin M.G., Wong M.H., Yu J. A nomenclature for intestinal in vitro cultures. Am J Physiol Gastrointest Liver Physiol. 2012;302:G1359–G1363. doi: 10.1152/ajpgi.00493.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Rodriguez N.R.M., Eloi M.D., Huynh A., Dominguez T., Lam A.H.C., Dayana C.M., Naser Z., Desharnais R., Salzman N.H., Porter E. Expansion of Paneth cell population in response to enteric salmonella enterica serovar typhimurium infection. Infect Immun. 2012;80:266–275. doi: 10.1128/IAI.05638-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Rajan A., Vela L., Zeng X.L., Yu X., Shroyer N., Blutt S.E., Poole N.M., Carlin L.G., Nataro J.P., Estes M.K., Okhuysen P.C., Maresso A.W. Novel segment- and host-specific patterns of enteroaggregative Escherichia coli adherence to human intestinal enteroids. MBio. 2018;9: doi: 10.1128/mBio.02419-17. e02419–e02417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Price A.E., Shamardani K., Lugo K.A., Deguine J., Roberts A.W., Lee B.L., Barton G.M. A map of Toll-like receptor expression in the intestinal epithelium reveals distinct spatial, cell type-specific, and temporal patterns. Immunity. 2018;49:560–575.e6. doi: 10.1016/j.immuni.2018.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Sato T., Stange D.E., Ferrante M., Vries R.G.J., Van Es J.H., Van Den Brink S., Van Houdt W.J., Pronk A., Van Gorp J., Siersema P.D., Clevers H. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141:1762–1772. doi: 10.1053/j.gastro.2011.07.050. [DOI] [PubMed] [Google Scholar]
- 61.Mead B.E., Ordovas-Montanes J., Braun A.P., Levy L.E., Bhargava P., Szucs M.J., Ammendolia D.A., MacMullan M.A., Yin X., Hughes T.K., Wadsworth M.H., Ahmad R., Rakoff-Nahoum S., Carr S.A., Langer R., Collins J.J., Shalek A.K., Karp J.M. Harnessing single-cell genomics to improve the physiological fidelity of organoid-derived cell types. BMC Biol. 2018;16:62. doi: 10.1186/s12915-018-0527-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Yin X., Farin H.F., Van Es J.H., Clevers H., Langer R., Karp J.M. Niche-independent high-purity cultures of Lgr5 + intestinal stem cells and their progeny. Nat Methods. 2014;11:106–112. doi: 10.1038/nmeth.2737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.VanDussen K.L., Carulli A.J., Keeley T.M., Patel S.R., Puthoff B.J., Magness S.T., Tran I.T., Maillard I., Siebel C., Kolterud Å., Grosse A.S., Gumucio D.L., Ernst S.A., Tsai Y.-H., Dempsey P.J., Samuelson L.C. Notch signaling modulates proliferation and differentiation of intestinal crypt base columnar stem cells. Development. 2012;139:488LP–497LP. doi: 10.1242/dev.070763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Farin H.F., Karthaus W.R., Kujala P., Rakhshandehroo M., Schwank G., Vries R.G.J., Kalkhoven E., Nieuwenhuis E.E.S., Clevers H. Paneth cell extrusion and release of antimicrobial products is directly controlled by immune cell–derived IFN-γ. J Exp Med. 2014;211:1393–1405. doi: 10.1084/jem.20130753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Fujii M., Matano M., Toshimitsu K., Takano A., Mikami Y., Nishikori S., Sugimoto S., Sato T. Human intestinal organoids maintain self-renewal capacity and cellular diversity in niche-inspired culture condition. Cell Stem Cell. 2018;23:787–793.e6. doi: 10.1016/j.stem.2018.11.016. [DOI] [PubMed] [Google Scholar]
- 66.Luu L., Matthews Z.J., Armstrong S.D., Powell P.P., Wileman T., Wastling J.M., Coombes J.L. Proteomic profiling of enteroid cultures skewed toward development of specific epithelial lineages. Proteomics. 2018;18:1–5. doi: 10.1002/pmic.201800132. [DOI] [PubMed] [Google Scholar]
- 67.Treveil A., Sudhakar P., Matthews Z.J., Wrzesiński T., Jones E.J., Brooks J., Ölbei M., Hautefort I., Hall L.J., Carding S.R., Mayer U., Powell P.P., Wileman T., Di Palma F., Haerty W., Korcsmáros T. Regulatory network analysis of Paneth cell and goblet cell enriched gut organoids using transcriptomics approaches. Mol Omics. 2020;16:39–58. doi: 10.1039/c9mo00130a. [DOI] [PubMed] [Google Scholar]
- 68.de Lau W., Kujala P., Schneeberger K., Middendorp S., Li V.S.W., Barker N., Martens A., Hofhuis F., DeKoter R.P., Peters P.J., Nieuwenhuis E., Clevers H. Peyer’s patch M cells derived from Lgr5+ stem cells require SpiB and are induced by RankL in cultured “miniguts.”. Mol Cell Biol. 2012;32:3639–3647. doi: 10.1128/MCB.00434-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Rouch J.D., Scott A., Lei N.Y., Solorzano-Vargas R.S., Wang J., Hanson E.M., Kobayashi M., Lewis M., Stelzner M.G., Dunn J.C.Y., Eckmann L., Martín M.G. Development of functional microfold (M) cells from intestinal stem cells in primary human enteroids. PLoS One. 2016;11 doi: 10.1371/journal.pone.0148216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Ding S., Song Y., Brulois K.F., Pan J., Co J.Y., Ren L., Feng N., Yasukawa L.L., Sánchez-Tacuba L., Wosen J.E., Mellins E.D., Monack D.M., Amieva M.R., Kuo C.J., Butcher E.C., Greenberg H.B. Retinoic acid and lymphotoxin signaling promote differentiation of human intestinal M cells. Gastroenterology. 2020;159:214–226.e1. doi: 10.1053/j.gastro.2020.03.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Nadjsombati M.S., McGinty J.W., Lyons-Cohen M.R., Jaffe J.B., DiPeso L., Schneider C., Miller C.N., Pollack J.L., Nagana Gowda G.A., Fontana M.F., Erle D.J., Anderson M.S., Locksley R.M., Raftery D., von Moltke J. Detection of succinate by intestinal tuft cells triggers a type 2 innate immune circuit. Immunity. 2018;49:33–41.e7. doi: 10.1016/j.immuni.2018.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Basak O., Beumer J., Wiebrands K., Seno H., van Oudenaarden A., Clevers H. Induced quiescence of Lgr5+ stem cells in intestinal organoids enables differentiation of hormone-producing enteroendocrine cells. Cell Stem Cell. 2017;20:177–190.e4. doi: 10.1016/j.stem.2016.11.001. [DOI] [PubMed] [Google Scholar]
- 73.Zachos N.C. Using enteroendocrine cell–enriched human enteroids to evaluate responses to gut stimuli. Cell Mol Gastroenterol Hepatol. 2019;8:291–292. doi: 10.1016/j.jcmgh.2019.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Johansson M.E.V., Sjövall H., Hansson G.C. The gastrointestinal mucus system in health and disease. Nat Rev Gastroenterol Hepatol. 2013;10:352–361. doi: 10.1038/nrgastro.2013.35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Erickson N.A., Nystrom E.E.L., Mundhenk L., Arike L., Glauben R., Heimesaat M.M., Fischer A., Bereswill S., Birchenough G.M.H., Gruber A.D., Johansson M.E.V. The goblet cell protein Clca1 (Alias mClca3 or Gob-5) is not required for intestinal mucus synthesis, structure and barrier function in naive or DSS-challenged mice. PLoS One. 2015;10 doi: 10.1371/journal.pone.0131991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.In J., Foulke-Abel J., Zachos N.C., Hansen A.M., Kaper J.B., Bernstein H.D., Halushka M., Blutt S., Estes M.K., Donowitz M., Kovbasnjuk O. Enterohemorrhagic Escherichia coli reduces mucus and intermicrovillar bridges in human stem cell-derived colonoids. Cell Mol Gastroenterol Hepatol. 2016;2:48–62.e3. doi: 10.1016/j.jcmgh.2015.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Liu L., Saitz-Rojas W., Smith R., Gonyar L., In J.G., Kovbasnjuk O., Zachos N.C., Donowitz M., Nataro J.P., Ruiz-Perez F. Mucus layer modeling of human colonoids during infection with enteroaggragative E. coli. Sci Rep. 2020;10:1–14. doi: 10.1038/s41598-020-67104-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Engevik M.A., Danhof H.A., Chang-Graham A.L., Spinler J.K., Engevik K.A., Herrmann B., Endres B.T., Garey K.W., Hyser J.M., Britton R.A., Versalovic J. Human intestinal enteroids as a model of Clostridioides difficile-induced enteritis. Am J Physiol Liver Physiol. 2020;318:G870–G888. doi: 10.1152/ajpgi.00045.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Colgan S.P., Taylor C.T. Hypoxia: an alarm signal during intestinal inflammation. Nat Rev Gastroenterol Hepatol. 2010;7:281–287. doi: 10.1038/nrgastro.2010.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Friedman E.S., Bittinger K., Esipova T.V., Hou L., Chau L., Jiang J., Mesaros C., Lund P.J., Liang X., FitzGerald G.A., Goulian M., Lee D., Garcia B.A., Blair I.A., Vinogradov S.A., Wu G.D. Microbes vs. chemistry in the origin of the anaerobic gut lumen. Proc Natl Acad Sci U S A. 2018;115:4170–4175. doi: 10.1073/pnas.1718635115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zeitouni N.E., Chotikatum S., von Köckritz-Blickwede M., Naim H.Y. The impact of hypoxia on intestinal epithelial cell functions: consequences for invasion by bacterial pathogens. Mol Cell Pediatr. 2016;3:14. doi: 10.1186/s40348-016-0041-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.von Köckritz-Blickwede M., Zeitouni N., Fandrey J., Naim H.Y. Measuring oxygen levels in Caco-2 cultures. Hypoxia. 2015;3:53. doi: 10.2147/HP.S85625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Fofanova T., Stewart C., Auchtung J., Wilson R., Britton R., Grande-Allen K., Estes M., Petrosino J. A novel human enteroid-anaerobe co-culture system to study microbial-host interaction under physiological hypoxia. bioRxiv. 2019:555755. [Google Scholar]
- 84.Sasaki N., Miyamoto K., Maslowski K.M., Ohno H., Kanai T., Sato T. Development of a scalable coculture system for gut anaerobes and human colon epithelium. Gastroenterology. 2020;159:388–390.e5. doi: 10.1053/j.gastro.2020.03.021. [DOI] [PubMed] [Google Scholar]
- 85.Van Welden S., Selfridge A.C., Hindryckx P. Intestinal hypoxia and hypoxia-induced signalling as therapeutic targets for IBD. Nat Rev Gastroenterol Hepatol. 2017;14:596–611. doi: 10.1038/nrgastro.2017.101. [DOI] [PubMed] [Google Scholar]
- 86.Sellin M.E., Müller A.A., Felmy B., Dolowschiak T., Diard M., Tardivel A., Maslowski K.M., Hardt W.D. Epithelium-intrinsic NAIP/NLRC4 inflammasome drives infected enterocyte expulsion to restrict salmonella replication in the intestinal mucosa. Cell Host Microbe. 2014;16:237–248. doi: 10.1016/j.chom.2014.07.001. [DOI] [PubMed] [Google Scholar]
- 87.Crowley S.M., Han X., Allaire J.M., Stahl M., Rauch I., Knodler L.A., Vallance B.A. Intestinal restriction of Salmonella Typhimurium requires caspase-1 and caspase-11 epithelial intrinsic inflammasomes. PLOS Pathog. 2020;16 doi: 10.1371/journal.ppat.1008498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Holly M.K., Han X., Zhao E.J., Crowley S.M., Allaire J.M., Knodler A., Vallance B.A., Smith J.G., Columbia B. Vol. 88. 2020. Salmonella enterica infection of murine and human enteroid-derived monolayers elicits differential activation of epithelial-intrinsic inflammasomes; pp. e00017–e00020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Bar-Ephraim Y.E., Kretzschmar K., Clevers H. Organoids in immunological research. Nat Rev Immunol. 2020;20:279–293. doi: 10.1038/s41577-019-0248-y. [DOI] [PubMed] [Google Scholar]
- 90.Wang Y., Kim R., Sims C.E., Allbritton N.L. Building a thick mucus hydrogel layer to improve the physiological relevance of in vitro primary colonic epithelial models. Cell Mol Gastroenterol Hepatol. 2019;8:653–655.e5. doi: 10.1016/j.jcmgh.2019.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Leon-Coria A., Kumar M., Moreau F., Chadee K. Defining cooperative roles for colonic microbiota and Muc2 mucin in mediating innate host defense against Entamoeba histolytica. PLoS Pathog. 2018;14:1–22. doi: 10.1371/journal.ppat.1007466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Johansson M.E.V. Fast renewal of the distal colonic mucus layers by the surface goblet cells as measured by in vivo labeling of mucin glycoproteins. PLoS One. 2012;7 doi: 10.1371/journal.pone.0041009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Schneider H., Pelaseyed T., Svensson F., Johansson M.E.V. Study of mucin turnover in the small intestine by in vivo labeling. Sci Rep. 2018;8:1–11. doi: 10.1038/s41598-018-24148-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Bergstrom K., Shan X., Casero D., Batushansky A., Lagishetty V., Jacobs J.P., Hoover C., Kondo Y., Shao B., Gao L., Zandberg W., Noyovitz B., McDaniel J.M., Gibson D.L., Pakpour S., Kazemian N., McGee S., Houchen C.W., Rao C.V., Griffin T.M., Sonnenburg J.L., McEver R.P., Braun J., Xia L. Proximal colon-derived O-glycosylated mucus encapsulates and modulates the microbiota. Science. 2020;370:467–472. doi: 10.1126/science.aay7367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Gustafsson J.K., Ermund A., Johansson M.E.V., Schütte A., Hansson G.C., Sjövall H. An ex vivo method for studying mucus formation, properties, and thickness in human colonic biopsies and mouse small and large intestinal explants. Am J Physiol Liver Physiol. 2012;302:G430–G438. doi: 10.1152/ajpgi.00405.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Birchenough G.M.H., Nystrom E.E.L., Johansson M.E.V., Hansson G.C. A sentinel goblet cell guards the colonic crypt by triggering Nlrp6-dependent Muc2 secretion. Science. 2016;352:1535–1542. doi: 10.1126/science.aaf7419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Strugala V., Allen A., Dettmar P.W., Pearson J.P. Colonic mucin: methods of measuring mucus thickness. Proc Nutr Soc. 2003;62:237–243. doi: 10.1079/pns2002205. [DOI] [PubMed] [Google Scholar]
- 98.Schroeder B.O., Birchenough G.M.H., Ståhlman M., Arike L., Johansson M.E.V., Hansson G.C., Bäckhed F. Bifidobacteria or fiber protects against diet-induced microbiota-mediated colonic mucus deterioration. Cell Host Microbe. 2018;23:27–40.e7. doi: 10.1016/j.chom.2017.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Schreurs R.R.C.E., Baumdick M.E., Sagebiel A.F., Kaufmann M., Mokry M., Klarenbeek P.L., Schaltenberg N., Steinert F.L., van Rijn J.M., Drewniak A., The S.M.M.L., Bakx R., Derikx J.P.M., de Vries N., Corpeleijn W.E., Pals S.T., Gagliani N., Friese M.A., Middendorp S., Nieuwenhuis E.E.S., Reinshagen K., Geijtenbeek T.B.H., van Goudoever J.B., Bunders M.J. Human fetal TNF-α-cytokine-producing CD4 + effector memory T cells promote intestinal development and mediate inflammation early in life. Immunity. 2019;50:462–476.e8. doi: 10.1016/j.immuni.2018.12.010. [DOI] [PubMed] [Google Scholar]
- 100.Biton M., Haber A.L., Rogel N., Burgin G., Beyaz S., Schnell A., Ashenberg O., Su C.W., Smillie C., Shekhar K., Chen Z., Wu C., Ordovas-Montanes J., Alvarez D., Herbst R.H., Zhang M., Tirosh I., Dionne D., Nguyen L.T., Xifaras M.E., Shalek A.K., von Andrian U.H., Graham D.B., Rozenblatt-Rosen O., Shi H.N., Kuchroo V., Yilmaz O.H., Regev A., Xavier R.J. T helper cell cytokines modulate intestinal stem cell renewal and differentiation. Cell. 2018;175:1307–1320.e22. doi: 10.1016/j.cell.2018.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Lindemans C.A., Calafiore M., Mertelsmann A.M., O’Connor M.H., Dudakov J.A., Jenq R.R., Velardi E., Young L.F., Smith O.M., Lawrence G., Ivanov J.A., Fu Y.Y., Takashima S., Hua G., Martin M.L., O’Rourke K.P., Lo Y.H., Mokry M., Romera-Hernandez M., Cupedo T., Dow L.E., Nieuwenhuis E.E., Shroyer N.F., Liu C., Kolesnick R., Van Den Brink M.R.M., Hanash A.M. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature. 2015;528:560–564. doi: 10.1038/nature16460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Waddell A., Vallance J.E., Hummel A., Alenghat T., Rosen M.J. IL-33 induces murine intestinal goblet cell differentiation indirectly via innate lymphoid cell IL-13 secretion. J Immunol. 2019;202:598–607. doi: 10.4049/jimmunol.1800292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Nozaki K., Mochizuki W., Matsumoto Y., Matsumoto T., Fukuda M., Mizutani T., Watanabe M., Nakamura T. Co-culture with intestinal epithelial organoids allows efficient expansion and motility analysis of intraepithelial lymphocytes. J Gastroenterol. 2016;51:206–213. doi: 10.1007/s00535-016-1170-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Rogoz A., Reis B.S., Karssemeijer R.A., Mucida D. A 3-D enteroid-based model to study T-cell and epithelial cell interaction. J Immunol Methods. 2015;421:89–95. doi: 10.1016/j.jim.2015.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Takashima S., Martin M.L., Jansen S.A., Fu Y., Bos J., Chandra D., O’Connor M.H., Mertelsmann A.M., Vinci P., Kuttiyara J., Devlin S.M., Middendorp S., Calafiore M., Egorova A., Kleppe M., Lo Y., Shroyer N.F., Cheng E.H., Levine R.L., Liu C., Kolesnick R., Lindemans C.A., Hanash A.M. T cell-derived interferon-γ programs stem cell death in immune-mediated intestinal damage. Sci Immunol. 2019;4 doi: 10.1126/sciimmunol.aay8556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Noel G., Baetz N.W., Staab J.F., Donowitz M., Kovbasnjuk O., Pasetti M.F., Zachos N.C. A primary human macrophage-enteroid co-culture model to investigate mucosal gut physiology and host-pathogen interactions. Sci Rep. 2017;7:1–14. doi: 10.1038/srep45270. [DOI] [PMC free article] [PubMed] [Google Scholar]


