Abstract
After birth, animals are colonized by a diverse community of microorganisms. The digestive tract is known to contain the largest number of microbiome in the body. With emergence of the gut-brain axis, the importance of gut microbiome and its metabolites in host health has been extensively studied in recent years. The establishment of organoid culture systems has contributed to studying intestinal pathophysiology by replacing current limited models. Owing to their architectural and functional complexity similar to a real organ, co-culture of intestinal organoids with gut microbiome can provide mechanistic insights into the detrimental role of pathobiont and the homeostatic function of commensal symbiont. Here organoid-based bacterial co-culture techniques for modeling host-microbe interactions are reviewed. This review also summarizes representative studies that explore impact of enteric microorganisms on intestinal organoids to provide a better understanding of host-microbe interaction in the context of homeostasis and disease.
Keywords: Intestinal organoid, Microbiome, Pathobiont, Probiotics, Symbiont
INTRODUCTION
Growing evidence supports that microorganisms and their byproducts can affect an individual’s phenotype and vice versa (1). With advances in high-throughput sequencing technology in the last decade, great efforts have been devoted to understanding host-microbiome interactions. Numerous works have demonstrated that the microbiome not only shapes the host immune system, but also correlates with tissue homeostasis and pathophysiology of diseases (2). Of note, the gut is the most heavily colonized organ. It contains over 70% of total symbionts. Significant dysbiosis has been found in gut luminal and fecal microbiota according to disease cohort studies, suggesting a causal relationship between the gut and its microbiota in host health (3-5). For a mechanistic study, a germ-free mouse model has been widely used to assess impact of the microbiome on disease progression. However, significant differences in microbial tropism, cellular composition, and microenvironment cues such as metabolic pathways between human and mouse often hinder interpretation of results (6). In this aspect, organoid technology has brought great advances in modeling of host-microbiome interaction in vitro. Organoids are self-organizing 3D structures with multiple differentiated cells derived from tissue-specific stem cells (7). With support of niche factors and extracellular matrix (ECM), LGR5-expressing crypt columnar cells can generate intestinal organoids (IOs), the first established epithelial organoids that could recapitulate the crypt-villus axis and lumen structures (8). Besides structural similarity, they can mimic several physiological properties of the gut such as selective absorption, barrier function, and mucus production. In addition, optimized culture conditions ensure robust generation and establishment of personalized- or genetically manipulated IOs (9, 10), overcoming limitations of conventional in vitro models. In this mini-review, we will briefly introduce current methodology for microbe-IO co-culture. We then summarize representative findings describing the impact of symbiont and pathobiont as well as probiotic candidates in host health using co-culture systems to provide insights into the importance of cross-talk between host and microorganisms.
ORGANOID-MICROBIAL CO-CULTURE TECHNOLOGY
To investigate interactions between IOs and microbes, it is essential to mimic the naïve gut environment harboring microbes. In the gastrointestinal tract, microbes exist within the lumen and directly interact with the intestinal epithelium through the apical side. In contrast, typical IOs have basal-out structures. Therefore, co-culture methods that allow physiologically relevant interaction are required for modeling microbial infections.
Microinjection
Microinjection of bacteria directly into the lumen of IOs can facilitate bacterial contact with the apical side of the epithelium (11, 12). Since the closed lumen has low oxygen tension, microinjection can improve the infection efficiency of anaerobic bacteria (13). Given that manual injection is a highly labor-intensive, time-consuming procedure (14, 15), a high-throughput organoid microinjection platform has been developed (16). However, since it is not a perfect anaerobic co-culture system, maintenance of a long-term culture of IOs with anaerobic bacteria is limited.
Suspension culture with IOs
Microbes can be simply treated to organoid growth media or embedded with ECM during IO culture. It is the most common method to study host-microbe interaction so far. Organoids can be cultured with live- and heat-killed (HK) bacteria or with conditioned media containing their byproducts, including bacterial toxins and metabolites (17-19). However, this method restricts the access of bacteria to the apical side of IOs. To overcome this limitation, IOs can be mechanically shredded to expose the luminal side and then re-seeded into ECM following co-culture with live bacteria (20, 21).
Organoid-derived monolayers (ODMs)
ODMs are established by seeding dissociated 3D-grown IOs on a Transwell plate to expose the apical surface upward with media (22, 23). ODMs can recapitulate cell compositions of gastrointestinal epithelium such as enterocytes, goblet cells, Paneth cells, and other cell populations (24). This monolayer culture provides practical advantages of easy microbial access to the luminal side and convenient sample collection compared to ECM-embedded classical IOs. However, 2D-grown epithelial stem cells usually undergo differentiation. They cannot be sub-cultured or maintained for a long time. Thus, many cells are needed each time. In addition, it is hard to obtain morphological information with ODM method (25). ODM culture technique can be further modified by exposing the upper side of the layer to air to generate an oxygen gradient. With the air-liquid interface (ALI) culture method, in which the basolateral side and the apical side contact with media and surrounding air, respectively, ODMs can differentiate into more mature epithelial cells such as mucus-secreting goblet cells than 3D organoids (26, 27). In addition, IHACS (intestinal hemi-anaerobic co-culture system), composed of a hypoxic apical chamber sealed with a rubber plug and a basal chamber in normal oxygen concentration can facilitate the survival of both epithelial cells and microbes (28, 29).
Organoids with reversed polarity
The apical-out IO model is an alternative to microinjection which has limitations of laborious processes and requirement of special equipment. Reversion of epithelial polarity is performed by removing ECM and maintaining suspended IOs in the low-attachment plate, where spontaneous polarity changes from basal-out to apical-out occur (30). Apical-out IOs allow direct interactions between the epithelium and microbes. Functional assays for nutrient uptake and epithelial barrier integrity can also be performed since epithelial cells within apical-out IOs can differentiate into a more mature state than conventional IOs (30).
CULTURING IOs WITH MICROORGANISMS TO STUDY HOST-MICROBIOME INTERACTIONS
Co-culture with pathobionts
Pathobiont has a significant impact on host health. Enteric infections by bacterial pathogens are responsible for various diarrheal diseases, particularly in developing countries. Dysbiosis in the gut microbiome can lead to several enteric and systemic disorders (31). Moreover, the rapid increase of antibiotic-resistant pathogens has become a critical threat in recent years (32). Thus, numerous efforts have been made to evaluate and understand the detrimental impact of pathobionts on the gut using IOs (Table 1).
Table 1.
Bacteria | Source of organoid | Culture system | Key findings | References |
---|---|---|---|---|
Shigella flexneri | Human small intestine, colon | ODM | ↑IL-8 secretion | (24) |
Human (unspecified) | ODM | ↑IL-8 secretion | (34) | |
↑Muc2 expression | ||||
Human small intestine | ODM | Basolateral infection, | (35) | |
↑Pro-inflammatory signals | ||||
Human small intestine, colon | ODM | Testing the therapeutic effect of bacteriophage | (36) | |
Salmonella enterica | Human small intestine | 3D-microinjection | ↓Organoid growth | (38) |
↑NF-kB signaling | ||||
↑Pro-inflammatory cytokine, | ||||
↓LGR5 expression | ||||
Human/mouse small intestine | 3D-microinjection | Recapitulation of early infection cycle, TTSS-1 is required for colonization | (39) | |
Human iPSC | 3D-microinjection | ↑Proinflammatory cytokines, InvA-dependent invasion | (40) | |
Human ESC | 3D-microinjection | T3SS-1-dependent invasion, | (41) | |
↑Inflammatory chemokine | ||||
Human small intestine, colon | Apical-out | Cytoskeletal rearrangement | (30) | |
Human small intestine | ODM | YrbE-dependent inflammatory response | (42) | |
EHEC | Human colon | ODM | ↓Colonic mucus | (45) |
Brush border damage | ||||
Human colon | ODM | Change in active ion transport | (47) | |
ETEC | Human small intestine | ODM | PDE5-mediated restriction of intracellular cGMP accumulation | (50) |
Pig small intestine | ODM | F4-mediated adhesion | (51) | |
pks+ E. coli | Human colon | ODM | Long-term exposure caused mutational signature | (54) |
Human/mouse colon | ODM/Shredded 3D | ↑Proliferation Wnt-independent growth | (21) | |
Clostridium difficile | Human iPSC, ESC | 3D-microinjection | ↓Epithelial barrier function | (11) |
Human iPSC | 3D-microinjection | ↓NHE3 expression | (56) | |
Human/mouse colon | 3D derived from infected mice/3D-toxin treatment | ↓Adherens junction, | (57) | |
↓Epithelial regeneration | ||||
Human small intestine | ODM | Adherence mechanism in human ODM model | (58) | |
Human iPSC | 3D-toxin treatment | ↓Transmembrane adhesion protein | (18) | |
Human iPSC | 3D-toxin treatment | Protective effect of HSA | (59) | |
Human (unspecified) | 3D-toxin treatment | Protective effect of antibiotic Bacitracin | (60) | |
Human/mouse iPSC | 3D-microinjection | Protective effect of Paneth cells on C. difficile-infected IO | (19) | |
Vibrio cholerae | Mouse small intestine | 3D-toxin treatment | ↑cAMP pathway | (62) |
Human small intestine | 3D-toxin treatment | Testing CT inhibitor with swelling assay | (63) | |
Human small intestine | 3D-toxin treatment | O-blood group exhibited different responses to CT | (64) | |
Listeria monocytogenes | Mouse small intestine | Shredded 3D | ↑Organoid growth | (20) |
↓Lgr5+ ISCs | ||||
↑Paneth cells | ||||
Mouse small intestine | Shredded 3D | ↑TNFa | (66) | |
↑Paneth cell, goblet cell | ||||
↓Notch signaling | ||||
Mouse small intestine | Shredded 3D | ↑TLR 2/4 signaling | (67) | |
Human small intestine, colon | Apical-out | Binding with basolateral receptor | (30) | |
Mouse small intestine | 3D-microinjection | InlA-Ecad-dependent translocation through goblet cells | (68) | |
Mouse (unspecified) | Shredded 3D | TMT-based quantitative proteomic analysis in different strains | (69) | |
Campylobacter jejuni | Mouse small intestine | 3D-bacterial lysate | DNA damage | (72) |
Fusobacterium nucleatum | Human (unspecified) | ODM-OMV treatment | ↑TNF, NF-κB, MAPK signaling | (74) |
Porphyromonas gingivalis | Mouse small intestine | 3D | Regulation of cell composition | (75) |
Shigella flexceri: Shigella flexneri infection is a leading cause of acute diarrhea, fever, and stomach pain in humans, particularly young children (33). ODM culture is the most frequently used in vitro model to study the role of S. flexneri in intestinal epithelial injury. After administration, S. flexneri exhibits high bacterial adherence rates in ODMs and induces maturation of M cells with upregulation of pro-inflammatory signals (24, 34, 35). Infection by S. flexneri can also trigger IL-8 secretion and mucin glycoprotein MUC2 expression (24, 35). S. flexneri can invade ODMs via the basolateral side, while disruption of the tight junctions in the epithelial barrier allows S. flexneri to invade ODMs via the apical surface (36). In addition, the killing potency of bacteriophages targeting S. flexneri has been tested in an ODM-based co-culture model to find alternatives to current antibiotics (34). Interestingly, bacterial adherence and invasion capacity in ODMs are significantly inhibited by the presence of bacteriophage.
Salmonella enterica: Salmonella enterica is a major pathogen for food-borne diarrheal diseases such as typhoid fever known to be caused by S. enterica serovar Typhimurium (37). Zhang et al. have investigated S. Typhimurium pathogenesis in the intestine using a microinjection method (38). In their study, disruption of epithelial tight junction and increment of inflammatory cytokine by NF-κB activation were observed in IOs upon administration of S. Typhimurium. Moreover, infection by Salmonella led to reduced Lgr5 expression, suggesting suppression of stem cell population. In an apical-out organoid model, S. Typhimurium invaded the apical epithelial surface more efficiently than invading the basolateral surface, which induced actin ruffles (30). Of note, key components for bacterial invasion have been revealed with a co-culture system. For instance, colonization of S. Typhimurium after microinjection into the lumen of IOs is dependent on an important virulence factor, type III secretion system 1 (TTSS-1), and its flagellar motility (39). The invasion capacity of S. Typhimurium is also mediated by invA (40) and Pathogenicity Island (SPI)-derived Type 3 secretion systems (T3SS) (41). Interestingly, a deficiency of phospholipid transporter YrbE in S. enterica serovar Typhi can lead to upregulation of flagellin, which enhances pro-inflammatory IL-8 expression in ODM (42).
Escherichia coli: Most Escherichia coli strains are commensal bacteria in the large intestine. However, several pathogenic strains are important causes of diarrheal illness and food poisoning (43). For example, enterohemorrhagic E. coli (EHEC) serotype O157:H7 is responsible for fatal foodborne diarrheal diseases (44, 45). After co-culturing with human ODMs, EHEC can readily colonize differentiated human ODMs. It especially targets MUC2 and microvillar resident protein PCDH24 at the early stage of infection (46). EHEC can also secrete extracellular serine protease EspP that exhibits enterotoxin activity by stimulating an electrogenic ion transporter, leading to reduced PCDH24 and brush border damage, while the enterotoxin-producing activity of EHEC in human IOs is independent of EspP activity (47).
Meanwhile, enterotoxigenic E. coli (ETEC) infection is associated with high mortalities in developing countries (48). ETEC can secrete heat-stable enterotoxin (ST) and heat-labile enterotoxin (LT) into the intestinal epithelium, which can induce cGMP and cAMP stimulation (49). Indeed, ST-induced cGMP synthesis followed by apical efflux of cGMP into the basolateral space has been observed in the human ODM model. However, disruption of phosphodiesterase PDE5 can reverse this phenomenon, revealing that cyclic nucleotide export and degradation can be initiated by enterotoxins (50). Given their importance in domestic animal farms, IOs have been established from porcine small intestine to generate ODM to investigate ETEC pathogenesis in pig gut (51). ETEC exhibits F4 fimbriae-mediated adhesion to porcine ODM as observed in vivo, indicating the utility of porcine IOs and co-culture systems to study enteric pathobionts in industrial animals.
Recent studies have shown that genotoxic colibactin-secreting E. coli strains are more abundantly detected in colorectal cancer (CRC) tissues than in healthy ones (52) and that pks genome is responsible for colibactin production (53). Indeed, long-term exposure (over five months) of human IOs to pks+ E. coli performed by luminal microinjection can lead to accumulation of genetic mutations in epithelial cells (54). Interestingly, organoids upon short-term exposure to pks+ E. coli also exhibit DNA damage, enhanced proliferation, and Wnt-independent abnormal growth (21). These studies demonstrate a hypothesis regarding the pks+ E. coli can mediate the tumorigenic process in CRC development.
Clostridium difficile: Clostridium difficile accounts for a significant proportion of antibiotic-induced diarrhea and colitis (55). Microinjection of C. difficile into IOs can lead to a reduction of Na+/H+ exchanger 3 (NHE3) with organoid swelling, which can recapitulate the in vivo situation of C. difficile-induced chronic diarrhea (56). Main virulence factors of C. difficile including C. difficile toxin A (TcdA), C. difficile toxin B (TcdB), and C. difficile transferase (CDT) can collapse the adherens junction through disruption E-Cadherin and actin-cytoskeleton (18, 57). Similarly, human ODMs express a high level of TcdA receptor (58). TcdA can disrupt the barrier function of human IOs upon C. difficile microinjection into the lumen for up to 12 h (11). TcdB can further inhibit epithelial regeneration by impairing stem cell functions in IOs established from C. difficile-infected mice (57). In this context, several studies have targeted C. difficile toxins to neutralize the harmful impact of C. difficile and found that administration of human serum albumin (HSA) and antibiotic bacitracin could prevent the toxic effect of TcdA and TcdB in IOs (59, 60). Co-culture of C. difficile and IOs has also provided insights into the physiological response of epithelial cells to resist microbial infection as reported by Liu et al. (19). In their work, the protective role of Paneth cells during C. difficile infection was investigated in murine and human IOs. It was shown that constitutive activation of signal transducer and activator of transcription 5 (STAT5) signaling could potentiate anti-bacterial and niche-supporting functions of Paneth cells in response to inflammatory cytokines and bacterial toxin, thus reducing C. difficile cytotoxicity.
Vibrio cholerae: Vibrio cholerae is an important cause of epidemic diarrhea, which is mediated by cholera toxin (CT) (61). Several studies have reported that treatment of IOs with CT can activate cAMP pathway, which induces acute swelling of IOs due to fluid accumulation in the lumen (62, 63). In this aspect, organoid swelling assay has been used for testing CT inhibitors (63). Interestingly, IOs derived from O-blood group show more elevated cAMP response upon CT administration than IOs derived from A-blood groups (64).
Listeria monocytogenes: Listeria monocytogenes is an opportunistic food-borne pathogen that causes listeriosis in immunocompromised individuals (65). In vitro infection of L. monocytogenes in IOs can stimulate organoid growth and induce differentiation into Paneth cells by regulating the expression of transcriptional factors Math1 and Sox9 (20). Another study has shown that L. monocytogenes can lead to Paneth cell induction in IOs by inhibiting Notch signaling and activating the toll-like receptor (TLR) 2/4 pathway with upregulation of opsonin protein CCN1 (66, 67). When the infection efficiency of L. monocytogenes was assessed in IOs exhibiting different polarity, basal-out IOs were more susceptible to L. monocytogenes invasion than apical-out ones because L. monocytogenes could bind to basolateral receptors after targeting cell extrusion regions (30). Interestingly, luminal microinjection of L. monocytogenes into IOs also demonstrated that crosslinking between bacterial internalin A (InlA) and E-cadherin on goblet cells could mediate the invasive process of Listeria, allowing its entry from lumen to the basal side despite the presence of epithelial barriers (68). Meanwhile, quantitative proteomic analysis has revealed that L. monocytogenes could differentially regulate the transcriptional activity and metabolism of IOs depending on their strains and serotypes (69).
Other pathogenic bacteria: A positive correlation between the abundance of Campylobacter species in CRC tissues and CRC development has been confirmed (70). Specifically, Campylobacter jejuni can induce DNA damage and intestinal inflammation by producing genotoxin (71). In line with previous studies, treatment with bacterial lysate from C. jejuni can lead to accumulation of DNA damage with increased γH2AX induction in human IOs, while ablation of cytolethal distending toxin (CDT) can abrogate pro-inflammatory- and genotoxic impact of C. jejuni (72).
Since the gut is part of the digestive tract, oral pathogenic bacteria can be detrimental to the intestine. For instance, Fusobacterium nucleatum in the gastrointestinal tract is positively associated with the development of gut inflammation and CRC (73). Interestingly, treatment with outer membrane vesicles (OMV) produced by F. nucleatum subsp. polymorphum can promote proinflammatory responses by stimulating tumor necrosis factor (TNF) secretion and NF-κB activation in human ODMs (74). Lipopolysaccharides (LPS) derived from Porphyromonas gingivalis, another major oral bacterium responsible for periodontitis, can regulate differentiated epithelial cell marker expression in murine IOs (75).
Co-culture with commensal bacteria and probiotics
Recent evidence has shown that probiotics can provide beneficial effects on the host by improving the balance of gut microbiota composition and promoting intestinal mucosal barrier function, indicating their therapeutic potential to treat a variety of intestinal disorders as shown below (Table 2) (76, 77).
Table 2.
Bacteria | Source of organoid | Culture system | Key findings | References |
---|---|---|---|---|
Lactobacillus reuteri D8 | Mouse small intestine | 3D | ↑Intestinal epithelial regeneration | (79) |
Mouse small intestine | 3D | ↑Proliferation of intestinal epithelial stem cells | (80) | |
Lactobacillus reuteri | Mouse small intestine | 3D | ↑Dendritic cell maturation and IL-10 production | (81) |
Lactobacillus rhamnosus GG | Mouse small intestine | 3D | ↑Expression of TLR3 | (82) |
Mouse small intestine, colon | 3D-microinjection | ↑Epithelial barrier function | (83) | |
Lactobacillus acidophilus | Mouse small intestine | 3D | ↑Protects the intestinal mucosa against pathogen | (84) |
Mouse small intestine | 3D -HK bacteria | ↑Intestinal epithelial function and differentiation | (85) | |
Bifidobacterium adolescentis | Human colon | ODM | ↑Differentiation of goblet cell and stem cell | (29) |
Escherichia coli Nissle | Human small intestine | 3D-microinjection | ↑Epithelial barrier function | (87) |
Nonpathogenic E. coli | Human small intestine | 3D-microinjection | ↑Epithelial proliferation & secretion of anti-microbial peptide | (88) |
Lactobacillus: Most Lactobacillus species including L. reuteri, L. rhamnosus, and L. acidophilus are regarded as important probiotics in the intestine. They have been reported to be able to improve proliferation, regeneration, and maturation of IOs (78).
L. reuteri can enhance the recovery of Lgr5+ cells and epithelial barrier after TNF-induced intestinal damage by activating the Wnt/β-catenin pathway in IOs (79). When L. reuteri D8 was co-cultured with murine IOs in the presence of lamina propria lymphocytes (LPLs), L. reuteri D8 could stimulate the proliferation of stem cells and Paneth cell induction by up-regulating the secretion of IL-22 mediated by STAT3 signaling activation (80). The role of L. reuteri in modulating host immunomodulation has also been investigated in an IOs-dendritic cell (DC) co-culture system (81). It was verified that both L. reuteri and its bacterial surface components could promote IL-10 production and DC maturation (81).
Administration of L. rhamnosus GG (LGG) to an IO culture system can provide some protection against rotavirus infection by up-regulating antiviral secretory factors such as interferon-α (IFN-α) and CXC motif chemokine ligand 1 (CXCL1) via activation of TLR3 pathway in epithelial cells (82). Han et al. have also investigated the therapeutic potential of LGG in irritable bowel syndrome (IBS) using IO-based co-culture systems with a focus on barrier function (83). Interestingly, they found that LGG significantly upregulated junctional marker expression and prevented the increase in organoid permeability in response to treatment with IFN-γ or fecal supernatants obtained from IBS patients, indicating a beneficial role of LGG in the maintenance of gut barrier integrity.
The protective effect of Lactobacillus species on intestinal epithelial damage has been also exhibited by L. acidophilus. To evaluate whether L. acidophilus could suppress the detrimental impact of pathobionts on IOs, L. acidophilus and S. typhimurium were treated simultaneously to dissociated IOs. Cellular properties of IOs were then assessed (84). In that work, S. Typhimurium significantly reduced the organoid-forming efficiency, which could be reversed by co-treatment with L. acidophilus. L. acidophilus could also enhance the differentiation into secretory lineage cells and, which in turn increased the production of mucus and antibacterial peptides to strengthen the mucosal barrier by regulating S. typhimurium-mediated hyperactivation of TLR2- and Wnt/β-catenin signaling pathways. Of note, Sittipo et al. have revealed dynamic changes in the prevalence of Lactobacillus species in murine stool samples after irradiation exposure using 16S rRNA-based oligotyping analysis (85) and reported the contribution of L. acidophilus to functional recovery of radiation-induced epithelial injury both in vitro and in vivo. Treatment of irradiated IOs with HK L. acidophilus can significantly enhance the organoid formation capacity with goblet cell enrichment, suggesting that administration of L. acidophilus and its derivatives might be beneficial to restoring intestinal homeostasis and barrier function impeded by pathogenic bacterial infection or irradiation.
Bifidobacterium: Bifidobacterium is another commensal probiotic bacterium that participates in the homeostasis of the gut microorganism community (86). Bifidobacterium, the dominant species in the large intestine, requires an anaerobic environment (78). In this context, IHACS, which mimics physiological gut anaerobic conditions in vitro, is useful for culturing IOs with Bifidobacterium (29). With this oxygen-controlled co-culture system, B. adolescentis could be successfully propagated with ODM. Importantly, only live B. adolescentis, not heat-killed bacteria or bacterial culture supernatant, could increase the expression of stem cell and goblet cell markers of ODM upon co-culture, demonstrating the importance of viability of bacteria in studying host-microbe interactions.
Escherichia coli: Benefits of commensal bacteria E. coli Nissle against dysbiosis have been confirmed in IOs (87). E. coli Nissle can protect human IOs from pathogenic E. coli-mediated disruption of the epithelial barrier, increased oxidative stress, and apoptosis. In addition, microinjection of a non-pathogenic E. coli strain ECOR2 into human IOs can lead to transient changes in the oxygen concentration without causing any harmful impact on epithelial cells (88). After colonization, E. coli strain ECOR2 can increase the production of antimicrobial peptides and improve tissue maturation of IOs.
FUTURE PERSPECTIVES
Organoid technology holds great potential to overcome limitations of conventional models such as 2D cell lines and experimental animals for modeling human anatomy and physiology. However, several challenging issues have to be solved to achieve advanced modeling of host-microorganism interactions with IOs. First, the absence of other cellular components except for epithelial cells is the main limitation of present IOs. Compared to pluripotent stem cell-derived organoids, which exhibit diverse cellular complexity, most adult stem cell-derived organoids like IOs consist of restricted lineage-derived cells that impede modeling of naïve microenvironment (89). Considering that microorganisms usually elicit an immune response and a repair process, which are predominantly mediated by regional immune cells and stromal cells respectively, the addition of these cells to an IO-microbe co-culture system would be necessary to recapitulate in vivo circumstances. Optimization of culture conditions is another important challenging issue. Although IOs are generally maintained under neutral, normoxic conditions, a dynamic range of oxygen concentration and pH found in the gut can significantly influence bacterial colonization patterns (90). Since oxygen availability strictly regulates bacterial behavior including growth, metabolism, and stress resistance, providing adequate oxygen gradient to both mammalian cells and microorganisms using advanced techniques such as microfluidic systems would be required to recapitulate in vivo situations. These improvements can also help us evaluate IO responses to a polymicrobial infection, which will provide clues about the role of dysbiosis in enteric disorder progression and greatly contribute to the understanding of disease mechanisms and establishing an effective therapeutic strategy.
ACKNOWLEDGEMENTS
This study was supported by the National Research Foundation of Korea (NRF), funded by the Ministry of Science and ICT (2018-R1A5A2023879, 2019-R1A2C2085876) and the Ministry of Education (2021-R1I1A1A01055654). Korean Fund for Regenerative Medicine (KFRM) grant funded by the Ministry of Science and ICT and the Ministry of Health & Welfare (22A0205L1) also supported this project.
Footnotes
CONFLICTS OF INTEREST
The authors have no conflicting interests.
REFERENCES
- 1.Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006;124:837–848. doi: 10.1016/j.cell.2006.02.017. [DOI] [PubMed] [Google Scholar]
- 2.Lynch JB, Hsiao EY. Microbiomes as sources of emergent host phenotypes. Science. 2019;365:1405–1408. doi: 10.1126/science.aay0240. [DOI] [PubMed] [Google Scholar]
- 3.Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol. 2018;11:1–10. doi: 10.1007/s12328-017-0813-5. [DOI] [PubMed] [Google Scholar]
- 4.Baumler AJ, Sperandio V. Interactions between the microbiota and pathogenic bacteria in the gut. Nature. 2016;535:85–93. doi: 10.1038/nature18849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Park EM, Chelvanambi M, Bhutiani N, Kroemer G, Zitvogel L, Wargo JA. Targeting the gut and tumor microbiota in cancer. Nat Med. 2022;28:690–703. doi: 10.1038/s41591-022-01779-2. [DOI] [PubMed] [Google Scholar]
- 6.Nguyen TLA, Vieira-Silva S, Liston A, Raes J. How informative is the mouse for human gut microbiota research? Dis Model Mech. 2015;8:1–16. doi: 10.1242/dmm.017400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kim J, Koo BK, Knoblich JA. Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol. 2020;21:571–584. doi: 10.1038/s41580-020-0259-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sato T, Vries RG, Snippert HJ, et al. 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]
- 9.Menche C, Farin HF. Strategies for genetic manipulation of adult stem cell-derived organoids. Exp Mol Med. 2021;53:1483–1494. doi: 10.1038/s12276-021-00609-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Min S, Kim S, Cho SW. Gastrointestinal tract modeling using organoids engineered with cellular and microbiota niches. Exp Mol Med. 2020;52:227–237. doi: 10.1038/s12276-020-0386-0.2bb77a6b61684d79a3525dbeddc5abb9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Leslie JL, Huang S, Opp JS, et al. Persistence and toxin production by Clostridium difficile within human intestinal organoids result in disruption of epithelial paracellular barrier function. Infect Immun. 2015;83:138–145. doi: 10.1128/IAI.02561-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zhang Y, Yu LC. Microinjection as a tool of mechanical delivery. Curr Opin Biotechnol. 2008;19:506–510. doi: 10.1016/j.copbio.2008.07.005. [DOI] [PubMed] [Google Scholar]
- 13.Poletti M, Arnauts K, Ferrante M, Korcsmaros T. Organoid-based models to study the role of host-microbiota Interactions in IBD. J Crohns Colitis. 2021;15:1222–1235. doi: 10.1093/ecco-jcc/jjaa257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ginga NJ, Slyman R, Kim GA, et al. Perfusion system for modification of luminal contents of human intestinal organoids and realtime imaging analysis of microbial populations. Micromachines 13. 2022;(Basel):131. doi: 10.3390/mi13010131.8e5d4698ca5b49bba31b2498c1b4d7d1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Puschhof J, Pleguezuelos-Manzano C, Martinez-Silgado A, et al. Intestinal organoid cocultures with microbes. Nat Protoc. 2021;16:4633–4649. doi: 10.1038/s41596-021-00589-z. [DOI] [PubMed] [Google Scholar]
- 16.Williamson IA, Arnold JW, Samsa LA, et al. 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]
- 17.Chen Y, Cao K, Liu H, et al. Heat killed Salmonella typhimurium protects intestine against radiation injury through wnt signaling pathway. J Oncol. 2021;2021:5550956. doi: 10.1155/2021/5550956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Fischer S, Uckert AK, Landenberger M, et al. Human peptide alpha-defensin-1 interferes with Clostridioides difficile toxins TcdA, TcdB, and CDT. FASEB J. 2020;34:6244–6261. doi: 10.1096/fj.201902816R. [DOI] [PubMed] [Google Scholar]
- 19.Liu R, Moriggl R, Zhang D, et al. Constitutive STAT5 activation regulates Paneth and Paneth-like cells to control Clostridium difficile colitis. Life Sci Alliance. 2019;2:e201900296. doi: 10.26508/lsa.201900296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Huang J, Zhou C, Zhou G, Li H, Ye K. Effect of Listeria monocytogenes on intestinal stem cells in the co-culture model of small intestinal organoids. Microb Pathog. 2021;153:104776. doi: 10.1016/j.micpath.2021.104776. [DOI] [PubMed] [Google Scholar]
- 21.Iftekhar A, Berger H, Bouznad N, et al. Genomic aberrations after short-term exposure to colibactin-producing E. coli transform primary colon epithelial cells. Nat Commun. 2021;12:1003. doi: 10.1038/s41467-021-21162-y.6216b08bac074ebf879284a8ec39ba76 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sato T, Stange DE, Ferrante M, et al. 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]
- 23.Thorne CA, Chen IW, Sanman LE, Cobb MH, Wu LF, Altschuler SJ. Enteroid monolayers reveal an autonomous WNT and BMP circuit controlling intestinal epithelial growth and organization. Dev Cell. 2018;44:624–633. doi: 10.1016/j.devcel.2018.01.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Nickerson KP, Llanos-Chea A, Ingano L, et al. A versatile human intestinal organoid-derived epithelial monolayer model for the study of enteric pathogens. Microbiol Spectr. 2021;9:e0000321. doi: 10.1128/Spectrum.00003-21.72c9a39e47f844738b9c99362443b4b3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zhang J, Hernandez-Gordillo V, Trapecar M, et al. Coculture of primary human colon monolayer with human gut bacteria. Nat Protoc. 2021;16:3874–3900. doi: 10.1038/s41596-021-00562-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Nossol C, Diesing AK, Walk N, et al. Air-liquid interface cultures enhance the oxygen supply and trigger the structural and functional differentiation of intestinal porcine epithelial cells (IPEC) Histochem Cell Biol. 2011;136:103–115. doi: 10.1007/s00418-011-0826-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Sachs N, Papaspyropoulos A, Zomer-van Ommen DD, et al. Long-term expanding human airway organoids for disease modeling. EMBO J. 2019;38:e100300. doi: 10.15252/embj.2018100300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kim R, Attayek PJ, Wang Y, et al. An in vitro intestinal platform with a self-sustaining oxygen gradient to study the human gut/microbiome interface. Biofabrication. 2019;12:015006. doi: 10.1088/1758-5090/ab446e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Sasaki N, Miyamoto K, Maslowski KM, Ohno H, Kanai T, Sato T. Development of a scalable coculture system for gut anaerobes and human colon epithelium. Gastroenterology. 2020;159:388–390. doi: 10.1053/j.gastro.2020.03.021. [DOI] [PubMed] [Google Scholar]
- 30.Co JY, Margalef-Catala M, Li X, et al. Controlling epithelial polarity: a human enteroid model for host-pathogen interactions. Cell Rep. 2019;26:2509–2520. doi: 10.1016/j.celrep.2019.01.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Troeger C, Blacker BF, Khalil IA, et al. Estimates of the global, regional, and national morbidity, mortality, and aetiologies of diarrhoea in 195 countries: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Infect Dis. 2018;18:1211–1228. doi: 10.1016/S1473-3099(18)30362-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hazen TH, Michalski J, Nagaraj S, Okeke IN, Rasko DA. Characterization of a large antibiotic resistance plasmid found in enteropathogenic escherichia coli strain B171 and its relatedness to plasmids of diverse E. coli and shigella strains. Antimicrob Agents Chemother. 2017;61:e00995-17. doi: 10.1128/AAC.00995-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Livio S, Strockbine NA, Panchalingam S, et al. Shigella isolates from the global enteric multicenter study inform vaccine development. Clin Infect Dis. 2014;59:933–941. doi: 10.1093/cid/ciu468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Llanos-Chea A, Citorik RJ, Nickerson KP, et al. Bacteriophage therapy testing against shigella flexneri in a novel human intestinal organoid-derived infection model. J Pediatr Gastroenterol Nutr. 2019;68:509–516. doi: 10.1097/MPG.0000000000002203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ranganathan S, Doucet M, Grassel CL, Delaine-Elias B, Zachos NC, Barry EM. Evaluating shigella flexneri pathogenesis in the human enteroid model. Infect Immun. 2019;87:e00740-18. doi: 10.1128/IAI.00740-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Koestler BJ, Ward CM, Fisher CR, Rajan A, Maresso AW, Payne SM. Human intestinal enteroids as a model system of shigella pathogenesis. Infect Immun. 2019;87:e00733-18. doi: 10.1128/IAI.00733-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Jajere SM. A review of Salmonella enterica with particular focus on the pathogenicity and virulence factors, host specificity and antimicrobial resistance including multidrug resistance. Vet World. 2019;12:504–521. doi: 10.14202/vetworld.2019.504-521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhang YG, 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]
- 39.Geiser P, Di Martino ML, Samperio Ventayol P, et al. Salmonella enterica serovar typhimurium exploits cycling through epithelial cells to colonize human and murine enteroids. mBio. 2021;12:e02684-20. doi: 10.1128/mBio.02684-20.0404c004b7a948ba8fd42e2c92a17f39 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Forbester JL, Goulding D, Vallier L, et al. Interaction of Salmonella enterica serovar typhimurium with intestinal organoids derived from human induced pluripotent stem cells. Infect Immun. 2015;83:2926–2934. doi: 10.1128/IAI.00161-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lawrence AE, Abuaita BH, Berger RP, et al. Salmonella enterica serovar typhimurium SPI-1 and SPI-2 shape the global transcriptional landscape in a human intestinal organoid model system. mBio. 2021;12:e00399-21. doi: 10.1128/mBio.00399-21.03f9d926e18e4f8f85620941b17d954e [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Verma S, Prescott RA, Ingano L, et al. The YrbE phospholipid transporter of Salmonella enterica serovar Typhi regulates the expression of flagellin and influences motility, adhesion and induction of epithelial inflammatory responses. Gut Microbes. 2020;11:526–538. doi: 10.1080/19490976.2019.1697593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Blount ZD. The unexhausted potential of E. coli. Elife. 2015;4:e05826. doi: 10.7554/eLife.05826.0dec92df648d4db59dd563d07a49a524 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kaper JB, Nataro JP, Mobley HL. Pathogenic Escherichia coli. Nat Rev Microbiol. 2004;2:123–140. doi: 10.1038/nrmicro818. [DOI] [PubMed] [Google Scholar]
- 45.Takeda Y. Enterohaemorrhagic Escherichia coli. World Health Stat Q. 1997;50:74–80. doi: 10.18535/ijetst/v3i11.05. [DOI] [PubMed] [Google Scholar]
- 46.In J, Foulke-Abel J, Zachos NC, et al. Enterohemorrhagic Escherichia coli reduce mucus and intermicrovillar bridges in human stem cell-derived colonoids. Cell Mol Gastroenterol Hepatol. 2016;2:48–62. doi: 10.1016/j.jcmgh.2015.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Tse CM, In JG, Yin J, et al. Enterohemorrhagic E. coli (EHEC)-secreted serine protease EspP stimulates electrogenic ion transport in human colonoid monolayers. Toxins (Basel) 2018;10:351. doi: 10.3390/toxins10090351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kotloff KL, Nataro JP, Blackwelder WC, et al. Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicenter Study, GEMS): a prospective, case-control study. Lancet. 2013;382:209–222. doi: 10.1016/S0140-6736(13)60844-2. [DOI] [PubMed] [Google Scholar]
- 49.Evans DJ, Jr, Evans DG. Three characteristics associated with enterotoxigenic Escherichia coli isolated from man. Infect Immun. 1973;8:322–328. doi: 10.1128/iai.8.3.322-328.1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Foulke-Abel J, Yu H, Sunuwar L, et al. Phosphodiesterase 5 (PDE5) restricts intracellular cGMP accumulation during enterotoxigenic Escherichia coli infection. Gut Microbes. 2020;12:1752125. doi: 10.1080/19490976.2020.1752125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Vermeire B, Gonzalez LM, Jansens RJJ, Cox E, Devriendt B. Porcine small intestinal organoids as a model to explore ETEC-host interactions in the gut. Vet Res. 2021;52:94. doi: 10.1186/s13567-021-00961-7.8d83712726cf4c89819507d8bd9e82b1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Dejea CM, Fathi P, Craig JM, et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science. 2018;359:592–597. doi: 10.1126/science.aah3648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Escobar-Paramo P, Le Menac'h A, Le Gall T, et al. Identification of forces shaping the commensal Escherichia coli genetic structure by comparing animal and human isolates. Environ Microbiol. 2006;8:1975–1984. doi: 10.1111/j.1462-2920.2006.01077.x. [DOI] [PubMed] [Google Scholar]
- 54.Pleguezuelos-Manzano C, Puschhof J, Rosendahl Huber A, et al. 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]
- 55.Chandrasekaran R, Lacy DB. The role of toxins in Clostridium difficile infection. FEMS Microbiol Rev. 2017;41:723–750. doi: 10.1093/femsre/fux048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Engevik MA, Engevik KA, Yacyshyn MB, et al. Human Clostridium difficile infection: inhibition of NHE3 and microbiota profile. Am J Physiol Gastrointest Liver Physiol. 2015;308:G497–G509. doi: 10.1152/ajpgi.00090.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mileto SJ, Jarde T, Childress KO, et al. Clostridioides difficile infection damages colonic stem cells via TcdB, impairing epithelial repair and recovery from disease. Proc Natl Acad Sci U S A. 2020;117:8064–8073. doi: 10.1073/pnas.1915255117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Engevik MA, Danhof HA, Chang-Graham AL, et al. Human intestinal enteroids as a model of Clostridioides difficile-induced enteritis. Am J Physiol Gastrointest Liver Physiol. 2020;318:G870–G888. doi: 10.1152/ajpgi.00045.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.di Masi A, Leboffe L, Polticelli F, et al. Human serum albumin is an essential component of the host defense mechanism against clostridium difficile intoxication. J Infect Dis. 2018;218:1424–1435. doi: 10.1093/infdis/jiy338. [DOI] [PubMed] [Google Scholar]
- 60.Zhu Z, Schnell L, Muller B, Muller M, Papatheodorou P, Barth H. The antibiotic bacitracin protects human intestinal epithelial cells and stem cell-derived intestinal organoids from clostridium difficile toxin TcdB. Stem Cells Int. 2019;2019:4149762. doi: 10.1155/2019/4149762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Sigman M, Luchette FA. Cholera: something old, something new. Surg Infect (Larchmt) 2012;13:216–222. doi: 10.1089/sur.2012.127. [DOI] [PubMed] [Google Scholar]
- 62.Foulke-Abel J, In J, Yin J, et al. Human enteroids as a model of upper small intestinal ion transport physiology and pathophysiology. Gastroenterology. 2016;150:638–649. doi: 10.1053/j.gastro.2015.11.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zomer-van Ommen DD, Pukin AV, Fu O, et al. Functional characterization of cholera toxin inhibitors using human intestinal organoids. J Med Chem. 2016;59:6968–6972. doi: 10.1021/acs.jmedchem.6b00770. [DOI] [PubMed] [Google Scholar]
- 64.Kuhlmann FM, Santhanam S, Kumar P, Luo Q, Ciorba MA, Fleckenstein JM. Blood group O-dependent cellular responses to cholera toxin: parallel clinical and epidemiological links to severe cholera. Am J Trop Med Hyg. 2016;95:440–443. doi: 10.4269/ajtmh.16-0161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Radoshevich L, Cossart P. Listeria monocytogenes: towards a complete picture of its physiology and pathogenesis. Nat Rev Microbiol. 2018;16:32–46. doi: 10.1038/nrmicro.2017.126. [DOI] [PubMed] [Google Scholar]
- 66.Zhou C, Zhang Y, Bassey A, Huang J, Zou Y, Ye K. Expansion of intestinal secretory cell population induced by listeria monocytogenes infection: accompanied with the inhibition of NOTCH pathway. Front Cell Infect Microbiol. 2022;12:793335. doi: 10.3389/fcimb.2022.793335.1bac6f2d3e614fa4a79b9f45ac9c565c [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Zhou C, Zou Y, Zhang Y, Teng S, Ye K. Involvement of CCN1 protein and TLR2/4 signaling pathways in intestinal epithelial cells response to listeria monocytogenes. Int J Mol Sci. 2022;23:2739. doi: 10.3390/ijms23052739.b81f760ed20d472e88d266449328da97 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kim M, Fevre C, Lavina M, Disson O, Lecuit M. Live imaging reveals listeria hijacking of E-cadherin recycling as it crosses the intestinal barrier. Curr Biol. 2021;31:1037–1047. doi: 10.1016/j.cub.2020.11.041. [DOI] [PubMed] [Google Scholar]
- 69.Zhou C, Zou Y, Huang J, et al. TMT-based quantitative proteomic analysis of intestinal organoids infected by listeria monocytogenes strains with different virulence. Int J Mol Sci. 2022;23:6231. doi: 10.3390/ijms23116231.f0225e3eda1a40dbade6940b11ffc2e6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Wu N, Yang X, Zhang R, et al. Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb Ecol. 2013;66:462–470. doi: 10.1007/s00248-013-0245-9. [DOI] [PubMed] [Google Scholar]
- 71.Phongsisay V. The immunobiology of Campylobacter jejuni: innate immunity and autoimmune diseases. Immunobiology. 2016;221:535–543. doi: 10.1016/j.imbio.2015.12.005. [DOI] [PubMed] [Google Scholar]
- 72.He Z, Gharaibeh RZ, Newsome RC, et al. Campylobacter jejuni promotes colorectal tumorigenesis through the action of cytolethal distending toxin. Gut. 2019;68:289–300. doi: 10.1136/gutjnl-2018-317200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Shang FM, Liu HL. Fusobacterium nucleatum and colorectal cancer: a review. World J Gastrointest Oncol. 2018;10:71–81. doi: 10.4251/wjgo.v10.i3.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Engevik MA, Danhof HA, Ruan W, et al. Fusobacterium nucleatum secretes outer membrane vesicles and promotes intestinal inflammation. mBio. 2021;12:e02706–20. doi: 10.1128/mBio.02706-20.ebd421ea1ed24b248d975fb71e9bd169 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Seo Y, Oh SJ, Ahn JS, Shin YY, Yang JW, Kim HS. Implication of Porphyromonas gingivalis in colitis and homeostasis of intestinal epithelium. Lab Anim Res. 2019;35:26. doi: 10.1186/s42826-019-0029-6.845dcab180984dada3443db77cd866d5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Khan R, Petersen FC, Shekhar S. Commensal bacteria: an emerging player in defense against respiratory pathogens. Front Immunol. 2019;10:1203. doi: 10.3389/fimmu.2019.01203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Lordan C, Thapa D, Ross RP, Cotter PD. Potential for enriching next-generation health-promoting gut bacteria through prebiotics and other dietary components. Gut Microbes. 2020;11:1–20. doi: 10.1080/19490976.2019.1613124.86c9abd4fe1e41118035d35a7711d4cc [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Park NY, Koh A. From the dish to the real world: modeling interactions between the gut and microorganisms in gut organoids by tailoring the gut milieu. Int J Stem Cells. 2022;15:70–84. doi: 10.15283/ijsc21243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wu H, Xie S, Miao J, et al. Lactobacillus reuteri maintains intestinal epithelial regeneration and repairs damaged intestinal mucosa. Gut Microbes. 2020;11:997–1014. doi: 10.1080/19490976.2020.1734423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hou QH, Ye LL, Liu HF, et al. Lactobacillus accelerates ISCs regeneration to protect the integrity of intestinal mucosa through activation of STAT3 signaling pathway induced by LPLs secretion of IL-22. Cell Death Differ. 2018;25:1657–1670. doi: 10.1038/s41418-018-0070-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Engevik MA, Ruan W, Esparza M, et al. Immunomodulation of dendritic cells by Lactobacillus reuteri surface components and metabolites. Physiol Rep. 2021;9:e14719. doi: 10.14814/phy2.14719.8c4a9a94d3f64f9196bbf623bcc6e880 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Aoki-Yoshida A, Saito S, Fukiya S, et al. Lactobacillus rhamnosus GG increases Toll-like receptor 3 gene expression in murine small intestine ex vivo and in vivo. Benef Microbes. 2016;7:421–429. doi: 10.3920/BM2015.0169. [DOI] [PubMed] [Google Scholar]
- 83.Han X, Lee A, Huang S, Gao J, Spence JR, Owyang C. Lactobacillus rhamnosus GG prevents epithelial barrier dysfunction induced by interferon-gamma and fecal supernatants from irritable bowel syndrome patients in human intestinal enteroids and colonoids. Gut Microbes. 2019;10:59–76. doi: 10.1080/19490976.2018.1479625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Lu X, Xie S, Ye L, Zhu L, Yu Q. Lactobacillus protects against S. typhimurium-induced intestinal inflammation by determining the fate of epithelial proliferation and differentiation. Mol Nutr Food Res. 2020;64:e1900655. doi: 10.1002/mnfr.201900655. [DOI] [PubMed] [Google Scholar]
- 85.Sittipo P, Pham HQ, Park CE, et al. Irradiation-induced intestinal damage is recovered by the indigenous gut bacteria lactobacillus acidophilus. Front Cell Infect Microbiol. 2020;10:415. doi: 10.3389/fcimb.2020.00415.befcf30e3bcb4ad0aaa19d6db5c70874 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Pino A, Benkaddour B, Inturri R, et al. Characterization of Bifidobacterium asteroides Isolates. Microorganisms. 2022;10:655. doi: 10.3390/microorganisms10030655.1fdd2e91eb2f42988c86c1d1df574a02 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Pradhan S, Weiss AA. Probiotic properties of escherichia coli nissle in human intestinal organoids. mBio. 2020;11:e01470–20. doi: 10.1128/mBio.01470-20.1955a920ace14e4284eef470d4c668ab [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hill DR, Huang S, Nagy MS, et al. Bacterial colonization stimulates a complex physiological response in the immature human intestinal epithelium. Elife. 2017;6:e29132. doi: 10.7554/eLife.29132.4f3e225ce6c5433682f7583fb2e61dd3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Kim J, Koo BK, Knoblich JA. Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol. 2020;21:571–584. doi: 10.1038/s41580-020-0259-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Lee JY, Tsolis RM, Baumler AJ. The microbiome and gut homeostasis. Science. 2022;377:eabp9960. doi: 10.1126/science.abp9960. [DOI] [PubMed] [Google Scholar]