5,195
Views
5
CrossRef citations to date
0
Altmetric
Review

In vitro and ex vivo modeling of enteric bacterial infections

ORCID Icon, ORCID Icon & ORCID Icon
Article: 2158034 | Received 25 Oct 2022, Accepted 07 Dec 2022, Published online: 28 Dec 2022

ABSTRACT

Enteric bacterial infections contribute substantially to global disease burden and mortality, particularly in the developing world. In vitro 2D monolayer cultures have provided critical insights into the fundamental virulence mechanisms of a multitude of pathogens, including Salmonella enterica serovars Typhimurium and Typhi, Vibrio cholerae, Shigella spp., Escherichia coli and Campylobacter jejuni, which have led to the identification of novel targets for antimicrobial therapy and vaccines. In recent years, the arsenal of experimental systems to study intestinal infections has been expanded by a multitude of more complex models, which have allowed to evaluate the effects of additional physiological and biological parameters on infectivity. Organoids recapitulate the cellular complexity of the human intestinal epithelium while 3D bioengineered scaffolds and microphysiological devices allow to emulate oxygen gradients, flow and peristalsis, as well as the formation and maintenance of stable and physiologically relevant microbial diversity. Additionally, advancements in ex vivo cultures and intravital imaging have opened new possibilities to study the effects of enteric pathogens on fluid secretion, barrier integrity and immune cell surveillance in the intact intestine. This review aims to present a balanced and updated overview of current intestinal in vitro and ex vivo methods for modeling of enteric bacterial infections. We conclude that the different paradigms are complements rather than replacements and their combined use promises to further our understanding of host-microbe interactions and their impacts on intestinal health.

This article is part of the following collections:
Enteric Bacterial Infections

1. Introduction

The intestine of humans and animals is a fascinating organ characterized by an intricate mutual interplay between the host and the commensal microbiome that covers approximately 200 m2, making it the largest organ surface area of the human body. The vast majority of intestinal microbes are autochthonous and beneficial for human health whereas others can cause intestinal injury or disease via an arsenal of specific virulence factors that include the production of toxins, toxic metabolites or the invasion of intestinal epithelial cells.Citation1,Citation2 The gastrointestinal microbiome plays a predominant role in colonization resistance throughout the animal kingdom.Citation3,Citation4 Tightly controlled interactions between the host and the microbiome are thus essential to prevent dysbiosis and maintain intestinal health.Citation5 However, a wide variety of Gram-negative and Gram-positive pathogens from diverse branches of the phylogenetic tree can cause infectious disease along the gastrointestinal tract. Besides acute infections, long-term subclinical dysbiosis in combination with environmental, nutritional and genetic factors can initiate a wide variety of inflammatory and autoimmune diseases, metabolic disorders, but also neuropsychiatric diseases and cancers.Citation6–9 Furthermore, reduction of microbiome diversity through excessive use of antibiotics or impairment of its regenerative capacity, e.g. by appendectomy, can lead to imbalances in inter-microbe and microbiome-host crosstalk.

The intestinal barrier separates the gut lumen, the outside environment, from underlying tissues and controls the uptake of nutrients and xenobiotics, while restricting invasion of pathogens. The innermost layer of the intestinal mucosa is the epithelium, a lining consisting of a single layer of cells that are tightly connected by different cell junction complexes to provide an efficient barrier.Citation10 This epithelial layer is primarily composed of enterocytes, but also includes mucin-secreting goblet cells, enteroendocrine cells, as well as various specialized cell types involved in antimicrobial defense, such as Paneth cells, which secrete antimicrobial factors, M cells, which sample and transport antigens to the underlying lymphoid tissue, as well as chemosensory tuft cells that protect against parasitic infections.Citation11 The epithelial cell lining is apically covered by mucin layers, which are differentially functionalized with antimicrobial tools and protect epithelial integrity.Citation12,Citation13 The basolateral side in the lamina propria is speckled by resting immune cells. Microbiome-immune cell crosstalk promotes immune homeostasis, for instance by facilitating the extrathymic development of CD4+ regulatory T cells with immunosuppressive activity.Citation14

Pathogens can traverse these barriers via paracellular or transcellular pathways. Paracellular crossing of the intestinal lining constitutes a process in which bacteria breach the tight connections between enterocytes. Paracellular permeability is increased upon inflammation, and, once integrity is compromised, leads to the infiltration of pathogens into the underlying submucosal layers or into the circulation.Citation15,Citation16 Alternatively, pathogens can infiltrate transcellularly via apical invasion of enterocytes followed by basolateral exocytosis. Both autochthonous and invading microbes can affect mucin production and alter the composition of the mucus layers with important consequences for epithelial maturation, barrier function and immune cell activation.Citation17,Citation18

Emulating the human intestine in vitro is of importance for mechanistic investigations into enteric infections, as well as for drug discovery programs focused on gastrointestinal diseases. For more than five decades, 2D monolayer cultures of human immortalized cell lines have served as the main in vitro paradigm to study infectious disease biology. While reductionistic, those systems have provided fundamental insights into the virulence and pathogenesis of various enteric bacterial infections and remain valuable tools for high throughput investigations that unravel molecular mechanisms of infection processes with unprecedented detail. In recent years, these models were complemented with a variety of bioengineered intestinal in vitro and ex vivo systems, including organoid cultures, scaffold-based 3D models and microfluidic systems. These models allow to capture intestinal phenotypes at the cellular and molecular level more accurately. In addition, they enable the investigation of effects related to mucus production and composition, symbiont-pathogen interactions, immune crosstalk and intestinal peristalsis – features that are not possible to study in conventional 2D in vitro models. However, these advantages come at the expense of increased complexity and reduced throughput. In this review, we provide an updated overview of in vitro and ex vivo models of enteric bacterial infections and discuss key examples where these different systems have advanced our understanding of the pathogenesis of infections with Salmonella enterica serovars Typhimurium and Typhi, Vibrio cholerae, Shigella spp., Escherichia coli and Campylobacter jejuni. For other intestinal pathogens we refer the interested reader to informative recent reviews.Citation19–22 We furthermore highlight current frontiers where emerging complex intestinal model systems can provide added value.

2. Intestinal monolayer culture models

Historically, 2D cultures of human immortalized cell lines have served as the first in vitro systems to study infectious disease.Citation23 In this approach, cells are cultured as monolayers on flat stiff surfaces, e.g. in petri dishes, tissue culture flasks or multi-well plates. For facultative intracellular pathogens, application of the aminoglycoside antibiotic gentamicin, which does not penetrate host cells and thus exclusively inhibits extracellular bacteria, allows to distinguish between intra- and extracellular replication. This gentamicin protection assay constitutes a classical method for quantifying pathogen invasiveness and intracellular replication that has led to the identification of multiple bacterial virulence factors by comparing wild type and mutant bacterial cells.Citation24 While monolayer culture formats are highly scalable, inexpensive and amenable to high-throughput analyses, the phenotype of these cells is not fully differentiated. Furthermore, these models do not allow easy distinction between events occurring at the apical or basolateral side, rendering them unsuitable for studies involving cell polarity. To ameliorate these limitations, transwell cultures have been developed in which monolayers are cultured on nano porous membranes that separate two compartments.Citation25 Transwell culture promotes the in vitro development of a polarized intestinal epithelium and these systems are extensively used for investigations of intestinal permeability and polarity.Citation26 However, both flat culture dishes and transwells are mostly made from polystyrene or other thermoplastics, which are not oxygen permeable, have unfavorable mechanical properties and are prone to drug absorption of hydrophobic molecules.Citation27,Citation28

The most popular cell models for the study of enteric infectious disease biology are undifferentiated cell lines, such as the cervical epithelial cell line HeLa, Henle-407 and the kidney epithelial cell line HEK293, as well as the enterocyte-derived colorectal adenocarcinoma cell lines Caco-2 and HT29. Upon reaching confluency, Caco-2 cells exhibit features such as tight junction-mediated barrier function, apical brush border formation and elevated expression of intestinal enzymes, receptors and transporters.Citation29 Caco-2 cells can furthermore be induced to transdifferentiate into M cells by co-culture with Raji B cells, a human B lymphoblastoid cell line Citation30 while HT29 cells and particularly the HT29-MTX subclone, can differentiate into mucus-producing goblet cells. Immortalized macrophage-like cell lines such as murine RAW264.7 and human J774.1 and THP-1 are frequently applied to assess interactions between enteric pathogens and basolaterally recruited immune cells that also cross the epithelial barrier to target pathogens in the lumen.

The advantages of immortalized cell lines are that they provide continuous, inexpensive and readily available supply of isogenic cellular material. Overall, however, cell lines are highly dedifferentiated, as evidenced by reduced or absent expression of multiple enterocyte-specific genes, including SLC5A1, MTTP, VLDLR, PXR (NR1I2) and MUC4.Citation31,Citation32 Immortalized cell lines also do not mimic the expression and diversity of in vivo toll-like receptor expression, which can impact the observed responses to microbial stimuli.Citation33 Furthermore, they exhibit significantly lower levels of inflammasome activation and pyroptotic cell death, resulting in impaired extrusion of infected cells from polarized epithelia and, eventually, higher pathogen loads.Citation34,Citation35

As an alternative to epithelial cell lines, primary intestinal cells or dissociated intestinal organoids can be used, while bone marrow-derived macrophages (BMDM) provide an alternative to immortalized immune cells. The molecular signatures of these cells more closely resemble their in vivo counterparts and allow to investigate effects of inter-individual differences.Citation36,Citation37 However, they are more difficult to acquire, limited in material and the cells lose their phenotypic resemblance after a few passages.

A main advantage of monolayers is the standardized experimental setup, reproducibility, and applicability for drug studies. They have been successfully utilized to pioneer the delineation of different mechanisms of infection employed by a variety of bacterial enteropathogens and their toxins (). However, most monolayer cultures comprise only enterocytes with only few studies attempting to integrate also other intestinal cell types, such as goblet cells and immune cells.Citation38–40 Additionally, the vast majority of monolayer cultures are conducted under static conditions, which results in rapid bacterial overgrowth, thus strongly limiting culture times and impairing the investigation of stable host-microbiome interactions.

Figure 1. Overview of key infectious mechanisms of enteric bacteria revealed using 2D monolayer cultures. Illustration showing the mechanism of invasion and bacteria-host interactions for Vibrio cholerae, Campylobacter jejuni, Shigella flexneri and Salmonella Typhimurium. AC = adenylate cyclase; CdtB = virulence factor with DNase I activity; CjeCas9 = virulence factor causing unspecific DNA damage; CtxA = cholera toxin A; GPCR = G-protein coupled receptor; Mφ = macrophage; PMN = polymorphonuclear leukocyte; T3SS = type III secretion system.

Figure 1. Overview of key infectious mechanisms of enteric bacteria revealed using 2D monolayer cultures. Illustration showing the mechanism of invasion and bacteria-host interactions for Vibrio cholerae, Campylobacter jejuni, Shigella flexneri and Salmonella Typhimurium. AC = adenylate cyclase; CdtB = virulence factor with DNase I activity; CjeCas9 = virulence factor causing unspecific DNA damage; CtxA = cholera toxin A; GPCR = G-protein coupled receptor; Mφ = macrophage; PMN = polymorphonuclear leukocyte; T3SS = type III secretion system.

3. Modeling of enteric bacterial infections in monolayer cultures

Reductionistic monolayer cultures can aid in the characterization and dissection of the molecular events occurring upon infection. However, despite their relatively low complexity, multiple experimental parameters remain important to consider. Infectivity of pathogens can be strongly affected by different growth conditions prior to infection. One example of this phenomenon is displayed by S. Typhimurium, which is less prone to invade epithelial cell lines when grown on an agar plate where it forms multicellular aggregates and does not express its major virulence factor, the type III secretion system-1 (T3SS-1), whereas it is substantially more invasive when grown in liquid culture.Citation41

Media composition, CO2 levels and nutrient supply also impact a multitude of molecular events, including signal transduction by bacterial two-component signaling systems based on the phosphotransfer from a histidine protein kinase to a response regulator, second messenger cyclic di-GMP and cyclic AMP signaling networks and chemotactic signaling. For instance, culture conditions that differ by ionic strength and the presence of digestive enzymes thereby mimicking the ileal or colonic environment, were sufficient to rewire metabolic pathways and production of virulence factors including Shiga toxin of enterohemorrhagic E. coli.Citation42 In addition to differences in virulence factor expression, environmental conditions and nutrient supply can determine the extent of immunostimulation by microbial associated molecular patterns,Citation43 such as regulation of flagellin secretion by cyclic di-GMP,Citation44 as well as the decision to adhere to epithelial cells and form biofilms, all of which affect disease outcome.Citation45,Citation46

Infectivity is majorly modulated by the commensal microbiome and, thus, interaction with non-pathogenic microbes is also important to consider in vitro. Colonization resistance of the residing microbiota is reliant on various mechanisms such as production of colicins, inhibition of pathogens by metabolic products and the type VI secretion system (T6SS).Citation47–49 A major protective mechanism of the microbial flora against pathogens is nutritional competition. In vitro studies found that commensal E. coli metabolize mono- and disaccharides available in the mucus, which was subsequently demonstrated to determine colonization success. Provision of mucus sugars to a commensal and an E. coli O157:H7 strain, which competes for the mucus niche, showed that the pathogenic strain used a distinct saccharide pool, which might provide a competitive colonization advantage.Citation50 The T6SS, a needle-like nano-machine injecting effector proteins into target microbes and phagocytic cells facilitates virulence by altering cellular motility,Citation51 culling bacterial competition Citation52 and killing host macrophages.Citation53 Mucus activates the T6SS of Vibrio cholerae, which, in turn, is counteracted by the resident microbiota via modification of bile acids, which exhibit inhibitory functions.Citation54 Notably, the T6SS can be used by both pathogenic and commensal bacterial to compete for enteric niches.Citation55

Inter-experimental variability can moreover be founded in genetic differences of the utilized bacterial strains. For instance, even a single nucleotide polymorphism can alter fitness and interaction with host cells. One example is a missense mutation found in the cache 1 signaling domain of the di-guanylate cyclase STM1987 of the invasive Salmonella enterica serovar Typhimurium ST131 clone that recently emerged in sub-Saharan Africa.Citation56 This variant was shown to drastically reduce cellulose production and displayed increased fitness both in vitro and in vivo virulence models.

Combined, these examples illustrate that, in addition to the selection of the cell model and culture system, genetic differences between bacterial strains, nutrient and media compositions as well as interactions with commensal bacteria constitute important factors that need to be considered for investigations of enteric infections. In the following, we highlight key insights into the pathobiology of enteric bacterial infections resulting from in vitro studies in monolayer cultures.

3.1 Shigella spp

The Escherichia coli subspecies Shigella spp., evolved multiple times through significant genome alterations and the acquisition of a large virulence plasmid. Shigella spp. causes shigellosis, a bacillary dysentery characterized by invasion of the epithelial lining with subsequent transcellular spreading. Shigella spp. infection is hallmarked by the uptake of bacteria into M cells, transcytosis, and subsequent macrophage uptake, followed by rapid escape via pyroptosis and basolateral invasion of enterocytes. In addition, using high-resolution microscopy of Caco-2/Raji B transwell cultures infected with S. flexneri, it was recently demonstrated that the bacteria can also directly spread from M cells to neighboring enterocytes, thereby evading adaptive immunity.Citation57 Once in enterocytes, Shigella hijacks the host actin polymerization machinery leading to high intracellular motility and inflammation-mediated disruption of the intestinal epithelium. Initially, Shiga toxin produced by Shigella dysenteriae 1 was assumed to be the reason for the disease symptoms. Then, in a series of seminal experiments, the first published in 1964, cellular invasion of the HeLa cell line by the virulent wild type S. flexneri 2a, but not by its avirulent colony variant has been identified as a key virulence property.Citation58 Subsequent early in vitro cell line experiments conducted most likely with spontaneous mutant strains of S. dysenteriae 1 moreover allowed to dissect the contributions of different virulence traits, such as host cell invasion and Shiga toxin production, on bacterial pathogenicity.Citation59,Citation60 Only almost two decades later, the HeLa invasion assay led to the initial identification of the T3SS located on the virulence plasmid of the model strain S. flexneri 2a required for invasion.Citation61,Citation62 The T3SS, a complex bacterial structure that enables the injection of effector proteins directly into the target cells' cytoplasm via specific translocator proteins, has later been identified as a major virulence factor in multiple human, animal and plant pathogens.

Subsequent experiments assigned additional loci on the Shigella virulence plasmid to specific virulence events, such as the intracellular escape from the phagocytic vacuole, multiplication in and early killing of host cells.Citation63–66 Such highly reductionistic approaches have not only aided the discovery of novel microbial virulence traits and vice versa novel functions of the host cells, but also allowed the detailed dissection of molecular mechanisms of infectious disease processes until today.Citation67,Citation68 For example, an RNA interference screen led to the identification of the chemokine receptor CCR5 as a stimulator of pore formation by the T3SS of Yersinia pseudotuberculosis.Citation69

Upon uptake into macrophages, Shigella causes rapid pyroptotic cell death after escape from the phagocytic vacuole by activation of inflammasomes and T3SS effector ubiquitin ligases, which cause degradation of human antimicrobial guanylate-binding proteins.Citation70,Citation71 The battery of virulence factors common to all phylogenetically distinct Shigella lines has been recently summarized.Citation72 Seminal in vitro work revealed that intra- and intercellular movement of Shigella is determined by actin polymerization via N-WASP on IcsA, a multifunctional type V secretion system autotransporter also involved in cell adhesion, escape from autophagy and extracellular biofilm formation.Citation73,Citation74 The IcsA autotransporter delicately interacts with the T3SS to promote virulence and immune escape.Citation75 Upon reactivation of the T3SS, the translocon pore protein IcaC interacts with the adherens junction protein β-catenin to reduce membrane tension and promote the formation of cellular protrusions, which are engulfed by adjacent cells resulting in intercellular spread.Citation76 While rupturing of the double membrane by Listeria monocytogenes, which similarly spreads using the cellular actin polymerization network, is achieved by the pore-forming toxin LLO and phosphatidylinositide phospholipase C, the role of the T3SS effectors IcsB and VirA in the vacuolar escape of Shigella needs to be further clarified.Citation77

3.2 Salmonella enterica serovar Typhimurium

Upon contact with animals or ingestion of contaminated produce and animal products, non-typhoidal Salmonella serovars, such as S. Typhimurium and Salmonella enterica serovar Enteritidis, cause a self-limiting gastroenteritis/colitis in immunocompetent individuals characterized by massive influx of neutrophils. In young children and immunocompromised individuals however, invasive disease can manifest, characterized by systemic infection with bacteremia, meningitis and osteomyelitis.Citation78,Citation79 In sub-Saharan Africa, such cases have recently seen a dramatic increase in frequency driven by pseudogene formation and chromosomal alterations followed by clonal replacements, resulting in the emergence of the S. Typhimurium ST313 clone.Citation80,Citation81

S. Typhimurium infections are characterized by a massive proliferation of the bacteria in the intestinal lumen. To fuel this overgrowth S. Typhimurium has developed chemotactic mechanisms that aid in identifying intestinal niches rich in microbiota- and host-derived electron acceptors, such as the microbial fermentation product 1,2-propanediol.Citation82,Citation83 Furthermore, nutrients and metabolites released from dying host cells induce a specific transcriptional response in Salmonella that involves expression of the pyruvate-formate lyase, resulting in the promotion of bacterial growth.Citation84 While the majority of bacterial cells reside in the gut lumen, few S. Typhimurium cells penetrate the mucus and breach the epithelial lining mainly via T3SS-1-mediated invasion of enterocytes. Monolayer culture systems have proven particularly useful in this context due to their compatibility with high-resolution time lapse microscopy, which allowed to directly image pathogen-host interactions at the epithelial cell lining.Citation85 A second type III secretion system (T3SS-2) is required for apical-to-basolateral migration and cell egress.Citation86 Upon escape from the Salmonella containing vacuole, bacteria are marked for autophagy by ubiquitinoylation of their lipopolysaccharide coat by the ring finger protein 213 E3 ubiquitin ligase.Citation87

Major virulence determinants of S. Typhimurium, adhesion to and invasion of enterocytes and flagellin-provoked induction of a proinflammatory cytokine response in enterocytes can be emulated in vitro using cell lines. Seminal studies have resulted in the identification of the T3SS-1,Citation88 as well as of fimbriae that mediate adhesion to specific cell types.Citation89 Infection of polarized epithelial cells furthermore led to the discovery that flagellin, the subunit of flagella, was rapidly transcytosed, which subsequently stimulated the basolateral secretion of proinflammatory cytokines and the recruitment of polymorphonuclear lymphocytes into the gut lumen.Citation90 More recently, dual RNA sequencing of bacteria and human cells (HeLa) upon infection facilitated the profiling of the transcriptomic alterations during the infection process, which was followed up by live cell fluorescent imaging combined with pharmacological interference in both microbes and the host.Citation67 The results reveal a novel bacterial small RNA that controls the expression of virulence genes as well as of coding and noncoding host transcripts, thus demonstrating that even simple cellular paradigms remain powerful tools for the dissection of fundamental infectious disease mechanisms.

After infiltration of the intestinal epithelium by few bacterial cells in the context of invasive disease, S. Typhimurium can survive in macrophages, which transport microbes to inner organs and are a reservoir for bacteria during chronic subclinical infections.Citation91 BMDMs showed a heterogeneity with respect to the response to infection depending on the bacterial status.Citation92 Specifically, macrophages display an M1-like pro-inflammatory phenotype with non-growing Salmonella, whereas macrophages harboring growing bacteria have an anti-inflammatory phenotype. Within a single BMDM cell, heterogeneity of bacterial replication and cyclic di-GMP second messenger concentration has been observed with a green fluorescent protein dilution and a FRET-based receptor binding assay, respectively, using confocal microscopy. A slow growing population is most affected by (over)production of the exopolysaccharide cellulose, which aids persistence of the organism by virulence downregulation.Citation93,Citation94 Activated by the regulator CsgD, the ancient rdar (red, dry and rough colony morphotype upon agar plate growth) biofilm and cyclic di-GMP signaling thus trade off acute virulence versus environmental and host persistence of S. Typhimurium.Citation44,Citation95 Although not traced back to a specific biofilm phenotype, in vitro findings in macrophages suggest that the switch into a transient non-proliferative state allows Salmonella to persist during environmental stress such as treatment with antibiotics and reemerge once antibiotic pressure is relieved with important implications for the understanding of recrudescent infection and the prevention of disease relapse.Citation92,Citation96

3.3 Salmonella enterica serovar Typhi

As S. Typhimurium and S. Enteritidis, Salmonella Typhi is a distinct pathovar of the species Salmonella enterica but, in contrast to the former, is restricted to humans.Citation97 S. Typhi elicits a severe systemic disease phenotype involving the lymphatic tissue of the gastrointestinal tract and internal organs. Unique virulence factors of S. Typhi determinative for the disease phenotype include the virulence-promoting Vi antigen capsule, a linear polymer consisting of α-1,4-linked N-acetylgalactosaminuronate, as well as the typhoid toxin (). In vitro studies showed that the Vi antigen capsule inhibits phagocytosis and limits the induction of proinflammatory cytokines by epithelial cells and resting macrophages.Citation98–100 Typhoid toxin is a composite A2B5 toxin that is only produced by S. Typhi when residing in the Salmonella containing vacuoles in host cells. The toxin is a modular assembly of two catalytic A units with distinct catalytic activity, CtdB and PltA, and a pentamer of two alternative B units (PltB or PltC). Although both toxin variants are synthesized, the heteromeric toxin supported by the PltB scaffold is selectively sorted and exported from the vacuole into the extracellular space via vesicles.Citation68 Using monolayer cultures of enterocytes, vascular endothelial cells and immune cells, it was shown that the typhoid toxin engages different receptors on different target cell types, including the CD34 family member podocalyxin-like protein 1 (PODXL) on the apical side of epithelial cells, the glycan receptor N-acetylneuraminic acid (Neu5Ac) and the receptor-type phosphatase CD45 expressed on macrophages.Citation101 The CdtB catalytic subunit, which exerts DNase I activity subsequently translocates into the nucleus to induce single-strand DNA breaks, triggering DNA damage response and cell cycle arrest.Citation102

Figure 2. Schematic overview of typhoid toxin secretion and export, and intracellular trafficking of the typhoid toxin. A) Typhoid toxin expression commences upon invasion of S. Typhi into host cells. Specifically, typhoid toxin consists of two enzymatic subunits, PltA and CdtB, which bind to pentamers of PltB or PltC. Upon assembly, the toxin subunits are secreted into the bacterial periplasm (inset). The bacterial transpeptidase YcbB and muramidase TtsA are required for typhoid toxin secretion into the lumen of the Salmonella containing vacuole. Subsequently, typhoid toxin is packaged into vesicle carrier intermediates, which transport the toxin to the plasma membrane where it gets released into the extracellular space. B) The fully assembled PltB toxin binds to the N-acetylneuraminic acid cell surface receptor (Neu5Ac) which results in the endocytosis and retrograde trafficking to the Golgi complex and endoplasmic reticulum (ER) where the CdtB component is released from its pentameric structure and proceeds into the nucleus to induce DNA damage via its DNase I activity. T3SS = type III secretion system.

Figure 2. Schematic overview of typhoid toxin secretion and export, and intracellular trafficking of the typhoid toxin. A) Typhoid toxin expression commences upon invasion of S. Typhi into host cells. Specifically, typhoid toxin consists of two enzymatic subunits, PltA and CdtB, which bind to pentamers of PltB or PltC. Upon assembly, the toxin subunits are secreted into the bacterial periplasm (inset). The bacterial transpeptidase YcbB and muramidase TtsA are required for typhoid toxin secretion into the lumen of the Salmonella containing vacuole. Subsequently, typhoid toxin is packaged into vesicle carrier intermediates, which transport the toxin to the plasma membrane where it gets released into the extracellular space. B) The fully assembled PltB toxin binds to the N-acetylneuraminic acid cell surface receptor (Neu5Ac) which results in the endocytosis and retrograde trafficking to the Golgi complex and endoplasmic reticulum (ER) where the CdtB component is released from its pentameric structure and proceeds into the nucleus to induce DNA damage via its DNase I activity. T3SS = type III secretion system.

3.4 Vibrio cholerae

Cholera, watery diarrhea caused by distinct strains of the marine and freshwater environmental species Vibrio cholerae, is a non-invasive disease of the small intestine. Major virulence factors of the O1 serogroup strains that caused seven pandemics since 1817 are CTX phage derived AB5 cholera toxin and the toxin-coregulated pilus TCP located on the vibrio pathogenicity island-1. While the six previous pandemics have been caused by the classical biotype, the seventh ongoing pandemic is caused by the El Tor biotype that differs in virulence factor expression.Citation103 After chemotaxis towards the intestinal mucus, V. cholerae penetrates the mucus layer via flagellar motility and adheres to enterocytes where it forms biofilms aided by the TCP pilus and other adhesins.Citation104–107 The mucin layer is, however, not only a barrier, but directly contributes to the population expansion of V. cholerae under aerobic conditions.Citation108 Further emphasizing the delicate molecular crosstalk between microbial physiology and the host, microbial proteins such as the recombinant TcpA subunit of the TCP pilus upregulate transcription and production of mucin from 2D cultured HT29 enterocytes and, in turn, mucin exerts a feedback on the expression of the bacterial chitin-binding protein GbpA.Citation109,Citation110 Recombinant TcpA further significantly upregulates transcription of Toll-like pattern-recognition receptors (TLRs), proinflammatory cytokines in particular TNF-α and the CEACAM1 adhesin in co-cultures of Caco-2 cells with -peripheral blood mononuclear cells.Citation111

The immunogenic neuraminidase (sialidase) recruits additional monoganglioside GM1 glycosphingolipid receptors to the host cell plasma membrane of 2D cultured primary human colon cells, which further facilitates uptake of cholera toxin into enterocytes.Citation112,Citation113 Alternatively, cholera toxin can also be delivered to intestinal epithelial cells in outer membrane vesicles that protect it from intestinal proteases.Citation114 Upon entry, the catalytic subunit of cholera toxin ADP-ribosylates the α subunit of GTP-binding proteins (Gs) to irreversibly inhibit its GTPase activity, resulting in constitutive activation of adenylate cyclase. Excessive cyclic AMP concentrations then activate protein kinase A (PKA), which phosphorylates its target, the chloride channel CFTR, leading to Cl, HCO3 and water efflux.Citation115 Na+ counterions are recruited via the paracellular route which cumulatively leads to massive dehydration, thus directly linking the molecular mechanisms dissected in vitro to the observed disease symptoms. Besides this mechanism cholera toxin exerts additional functions such as targeting Rab11, the major regulatory hub of intracellular membrane trafficking, via cyclic AMP which leads to inhibition of exocyst trafficking and disruption of barrier integrity.Citation116

3.5 Pathogenic Escherichia coli strains

The role of the species Escherichia coli in interaction with humans is multifactorial.Citation117 E. coli can even be considered a symbiont, as it is among the first species colonizing the gastrointestinal tract, which stimulates the innate immune response and significantly protects against the development of autoimmunity.Citation118 However, with a tenuous transition to a pathogenic lifestyle via acquisition of accessory genomic islands bearing distinct colonization and virulence factors, E. coli has diversified into numerous intra- and extra-intestinal pathovars.Citation119 Conventional Caco-2 monolayer cultures have had notable success in the development of anti-infectives that act via blocking the bacterial machinery for enterocyte entry. Specifically, the pharmacological inhibition of the bacterial Tir protein that upon injection into enterocytes remodels the cytoskeleton to provide susceptibility to enteropathogenic E. coli (EPEC) infection, blocked infection in vitro and protected mice from EPEC-induced diarrhea.Citation120

E. coli is also associated with the development of chronic inflammatory diseases, such as Crohn’s disease, and with the development of gastrointestinal cancer. Inflammatory disease manifestations are associated with the adherent-invasive pathovar that displays distinct biological features, including the formation of intracellular colonies in immune cells.Citation121 In contrast, development of colorectal cancer is associated with specific E. coli strains that encode genotoxins, such as colibactin, which can alkylate DNA in cell culture and in vivo directly leading to genomic instability.Citation122,Citation123

3.6 Campylobacter jejuni

The helically shaped polarly flagellated Gram-negative bacterium Campylobacter jejuni is the most prevalent food-born pathogen with one in ten individuals globally affected each year. Colonizing warm-blooded animals including birds, the organism is transmitted by consumption of undercooked poultry, but also milk and water. C. jejuni strains have been found to cause disease with a broad range of symptoms from watery to bloody mucoid dysentery. Additionally, infections can be followed by a number of autoimmune diseases, such as Guillain-Barré syndrome. Although the disease symptoms had been recognized already by Theodor Escherich in the 19th century, Campylobacter was isolated only in the 1970ʹs.Citation124 Molecularly, the periplasmic protease HtrA is the virulence factor that enables rapid paracellular translocation of C. jejuni by cleavage of the adherens and tight junction proteins, such as E-cadherin and occludin, without however altering overall membrane integrity, as evidenced by unchanged transepithelial resistance in polarized epithelial transwell cultures upon infection.Citation125

Adhesion of C. jejuni to epithelial cells is in part mediated by interactions between host heparin and the bacterial flagellar tip protein FliD, as well as by direct, high-affinity interactions of the O-side chain of Campylobacter lipooligosaccharide with host glycans.Citation126,Citation127 Following binding, the fibronectin-binding protein CadF activates the integrin-FAK signaling axis to trigger actin rearrangement and bacterial invasion.Citation128 Using an infection model based on co-cultured human enterocytes and HT29-MTX mucin-producing goblet-like cells in transwells, Campylobacter adhesion and invasion was shown to require N-linked glycosylation of C. jejuni proteins and a unique O-methyl phosphoramidate modification of its capsular polysaccharide.Citation129,Citation130

Interestingly, C. jejuni lacks a T3SS that is common to most enteropathogenic bacterial species. In its absence, cytotoxicity to different human cell culture models, including HeLa and Caco-2, is exerted by a variety of factors, including functional flagella, secretion of cytolethal distending toxin (CDT), as well as membranous short-chain lysophosphatidylethanolamine that permeabilize host cells.Citation131,Citation132 Another unconventional virulence factor is the CRISPR/Cas related endonuclease CjeCas9 that causes unspecific DNA cleavage and programmed cell death upon its translocation into the nucleus.Citation133 Furthermore, the Campylobacter metabolite ADP-heptose, a core oligosaccharide precursor of lipopolysaccharide, strongly activated NFκB signaling in enterocytes, without the need for a type III or type IV injection machinery.Citation134 Similar effects have been observed in other pathogens, including Helicobacter pylori and Akkermansia muciniphila, suggesting that ADP-heptoses constitute a pathogen-associated molecular pattern.Citation135–137

4. Advanced intestinal culture systems

As illustrated above, 2D monolayer cultures of human intestinal cells – either as enterocyte monocultures or co-cultured with goblet cells or immune cells – have contributed, and continue to contribute, tremendously to our mechanistic understanding of the pathogenic processes underlying enteric bacterial infections. Specifically, these culture systems have proven very useful for 1) the systematic identification of host factors required for or protective against infection, 2) the determination of virulence factors and their regulatory pathways, as well as 3) the uncovering of metabolites and post-translational modifications that link infection to cellular pathogenesis. To these ends, RNA interference, library screens based on CRISPR/Cas or transposons and dual RNA sequencing complement previously used fluorescence microscopy, pharmacological interference and biochemical strategies such as systematic mapping of protein-protein interactions for the detailed dissection of host regulatory pathways.

Notably however, despite these undisputable technological advancements, monolayer cultures are not well suited for discovery studies that require long-term maintenance, accurate cellular and molecular phenotypes (e.g. to support complex pathogen lifecycles or to mimic changes in infectivity along the gastrointestinal tract), crosstalk between different enteric cell types, peristaltic motion or interactions between the pathogen and a complex commensal microbiome. Furthermore, studies using cell lines are not well positioned to investigate effects of inter-individual differences. To overcome these limitations, a wide variety of diverse organotypic and microphysiological in vitro and ex vivo models have been developed in recent years. Importantly, we would like to emphasize that we see these more complex systems not as a replacement for conventional monolayer studies, but rather as a useful complement to address selected aspects that cannot be interrogated in the more simplified culture systems.

4.1 Intestinal organoid culture

Advances in stem cell research have facilitated the development of 3D cell culture systems that aim to narrow the gap between 2D monotypic cultures and ex vivo tissue models. Specifically, intestinal organoids have provided an important breakthrough for the emulation of intestinal phenotypes and functions.Citation138,Citation139 Intestinal organoids can be derived from tissue-resident adult stem cells, embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) using Matrigel as a scaffold. In culture medium containing specific mitogens and differentiation factors, these cells proliferate and, following a series of divisions, self-organize into microtissues containing enterocytes, as well as goblet cells, Paneth cells and enteroendocrine cells.Citation140 Intestinal stem cells can furthermore be induced to differentiate into M cells by exposure to the TNF family cytokine RANKL.Citation141 Organoids exhibit the morphology of the folded intestinal epithelium including villi and crypt-like domains the latter of which host a functional stem cell niche at their base.

In recent years, organoids have been adopted for modeling infection with pathogens. These systems hold promise for addressing novel questions in host-microbe interactions, infectious diseases, and the resulting inflammatory conditions. As organoids capture the cellular complexity of the human intestinal epithelium, they are well suited to investigate pathogens with narrow cell-type tropism and the corresponding mechanisms of their cell-specific interactions. For instance, M cells are the preferred cell type for S. flexneri invasion and the presence of these cells in the intestinal organoid enabled the holistic analysis of this infection.Citation142 Importantly, however, when conventional organoid protocols are used, the apical side faces inwards, which requires that microbiome and pathogen need to be microinjected. Multiple pathogens have already been injected into organoids to understand intestinal disease mechanisms, including enterohemorrhagic E. coli,Citation143 Cryptosporidium parvum Citation144 and S. enteritica.Citation145,Citation146 Furthermore, organoids can support the luminal culture of microbial communities from human fecal isolates including both aerobic and obligately anaerobic taxa for at least four days.Citation147 However, microinjection is labor intensive, and the multiplicity of infection (MOI) is difficult to control given the differing numbers of cells per organoid. While there are elegant technical solutions to inject organoids at scale, these methods require considerable technical expertise as well as investments into robotic infrastructure.Citation147 To ameliorate these issues, protocols for the generation of inverted, apical-out organoids have been established.Citation148 These methods are based on the removal of extracellular Matrigel and subsequent culture in low attachment plates, resulting in reversed polarity while functionality and barrier integrity stayed intact. These inverted organoids recapitulated the preferential infection with S. Typhimurium and L. monocytogenes on the apical and basolateral sides, respectively.Citation149 However, these methods instead render the basolateral side inaccessible and do not solve issues pertaining to controlling MOI.

To address these limitations, an alternative approach is the monolayer culture of organoid-derived cells. For this approach, organoids are enzymatically dissociated into single cells and then seeded on transwells, thereby ensuring appropriate polarity and barrier formation while retaining the cellular complexity of the intestinal epithelium and granting ready access to both apical and basolateral sides. By generating organoids from patient-derived samples with region-specific development into mature epithelial lineages, this approach has been successfully used for infections with S. flexneri, different pathogenic E. coli strains as well as with S. Typhi and S. Typhimurium.Citation37,Citation150,Citation151 Specifically, the cultures closely recapitulated in vivo tissue tropism, inter-individual differences in infectivity as well as pathogen-specific infection patterns. For Shigella, organoid-derived monolayer cultures revealed basolateral invasion preference in the absence of M cells,Citation152 whereas in the presence of M cells also direct lateral M cell-to-enterocyte spread can be observed.Citation57 Transwell culture of dissociated organoids has moreover identified the infection dynamics and coordinated contractile responses of reconstituted intestinal epithelium upon sensing of the T3SS and flagellar ligands of S. Typhimurium.Citation153,Citation154 Organoid-derived monolayers also allow to study microbial uptake by M cells, as demonstrated for the enteric pathogen Yersinia pseudotuberculosis, which exploits M cells to gain access to the underlying lymphatic system.Citation155

The physiologically relevant phenotypes are showcased by the possibility to culture previously non-cultivatable pathogens in monolayer cultures of dissociated organoids, such as norovirus Citation156,Citation157 and rotavirus,Citation158,Citation159 which are among the most common causes of gastroenteritis. Furthermore, culture of intact or dissociated organoids on air-liquid interface transwells allowed for the first time to support the entire life cycle of Cryptosporidium spp, apicomplexan parasites, which infect enterocytes and propagate via rounds of release and reinvasion with both sexual and asexual replication stages.Citation144,Citation160 As such, this culture paradigm is highly promising to enable studies into the molecular biology of meiotic divisions and for progressing genetic analyses of complex apicomplexan phenotypes.

Intestinal organoids are easy to maintain, can be readily established from individual patients for personalized testing and provide physiologically relevant and versatile tools that closely mimic the human intestinal epithelium. However, notable hurdles and limitations intrinsic to the method remain. It is important to consider that Matrigel, which constitutes the most commonly used scaffold for intestinal organoids, commonly contains gentamicin, which interferes with bacterial cultures.Citation161 Furthermore, while organoids accurately recapitulate the composition of the epithelial lining, they lack immune cells, neuronal connections and vascularization, as well as flow and peristaltic motion. Thus, combination of organoid culture and tissue engineering approaches, as demonstrated for the reconstruction of tubular epithelia with an accessible lumen connected to external micropumps (), could be promising to combine cellular complexity with an appropriate biophysical microenvironment, thereby further approximating intestinal architecture and function.

Figure 3. Examples of organotypic and microphysiological culture methods to mimic enteric infections. A) Schematic of a fluidic device based on dissociated intestinal organoids. Inset shows the hydrogel-based microchannel. Immunofluorescence of C. parvum undergoing its major epicellular stages in the mini-guts (right-top) and scanning electron microscopy image of distinct stages of the C. parvum life cycle at 72 h post infection (right-bottom). Figure obtained with permission from.Citation162 B) Illustration showing a 3D villi model infected with S. Typhimurium. Scanning electron microscopy image of the fabricated villi (top right), and fluorescence micrographs showing S. Typhimurium in 2D (bottom left), the crypt section of 3D villi (bottom-middle) and the villus tips of 3D villi (bottom right) 20 days after infection. Image obtained with permission from.Citation163 C) Schematic of a cross-section of an intestinal microphysiological system (left top). Micrographs showing cross-sections of uninfected (control) and chips infected with GFP expressing Shigella (bottom left). Phase contrast images, and fluorescence confocal micrographs (vertical cross-sectional views) of villi showing intestinal villus damage upon infection with enteropathogenic E. coli (right). Images in this panel were obtained with permission from.Citation164,Citation165 D) Examples of intravital microscopy (IVM). Top: IVM analyses of TCRγδGFP reporter mice after infection with S. Typhimurium. Arrows show tracked flossing movements on one villus (left) and 4D tracking of TCRγδGFP cells (right). The intraepithelial compartment is outlined. Reproduced from Citation166 with permission. Bottom: IVM analyses of Salmonella uptake by CD103+ dendritic cells showing that the immune cells extend dendrites through the epithelium while crawling above the basement membrane (arrow on left); the dendrites engulf Salmonella (circled on right), and retract them toward the cell’s soma. Images adopted with permission from.Citation167

Figure 3. Examples of organotypic and microphysiological culture methods to mimic enteric infections. A) Schematic of a fluidic device based on dissociated intestinal organoids. Inset shows the hydrogel-based microchannel. Immunofluorescence of C. parvum undergoing its major epicellular stages in the mini-guts (right-top) and scanning electron microscopy image of distinct stages of the C. parvum life cycle at 72 h post infection (right-bottom). Figure obtained with permission from.Citation162 B) Illustration showing a 3D villi model infected with S. Typhimurium. Scanning electron microscopy image of the fabricated villi (top right), and fluorescence micrographs showing S. Typhimurium in 2D (bottom left), the crypt section of 3D villi (bottom-middle) and the villus tips of 3D villi (bottom right) 20 days after infection. Image obtained with permission from.Citation163 C) Schematic of a cross-section of an intestinal microphysiological system (left top). Micrographs showing cross-sections of uninfected (control) and chips infected with GFP expressing Shigella (bottom left). Phase contrast images, and fluorescence confocal micrographs (vertical cross-sectional views) of villi showing intestinal villus damage upon infection with enteropathogenic E. coli (right). Images in this panel were obtained with permission from.Citation164,Citation165 D) Examples of intravital microscopy (IVM). Top: IVM analyses of TCRγδGFP reporter mice after infection with S. Typhimurium. Arrows show tracked flossing movements on one villus (left) and 4D tracking of TCRγδGFP cells (right). The intraepithelial compartment is outlined. Reproduced from Citation166 with permission. Bottom: IVM analyses of Salmonella uptake by CD103+ dendritic cells showing that the immune cells extend dendrites through the epithelium while crawling above the basement membrane (arrow on left); the dendrites engulf Salmonella (circled on right), and retract them toward the cell’s soma. Images adopted with permission from.Citation167

4.2 Models on bioengineered scaffolds

With the emergence of tissue engineering in the 1990s, models have been developed in which cells are cultured on specific 3D scaffolds that resemble the structure of the tissue in situ. These models are specifically a breakthrough for intestinal studies given the complex native crypt-villus geometry, which is required for efficient intestinal stem cells self-renewal and for the generation of biochemical gradients that mediate cell communication, differentiation and proliferation.Citation168,Citation169 A variety of biomaterials have been used to create the scaffolds, ranging from biocompatible hydrogels based on natural or synthetic biopolymers with tunable physiochemical properties Citation170,Citation171 to decellularized extracellular matrix.Citation172 Besides the biochemical scaffold composition, also its stiffness impacts the morphology and differentiation of intestinal epithelial cells. Specifically, stiff substrates result in flattening of the cells whereas physiological matrix elasticity results in columnar organization and the maintenance of cellular polarity.Citation173 Decellularized scaffolds offer the natural composition and physical properties of the tissue for cells and the intestinal submucosa scaffold is widely used to model intestinal epithelium in vitro. Seeded with Caco-2 cells, these scaffolds were used to investigate the infection with C. jejuni. It was observed that non-capsulated bacteria can adhere and internalize into the cells in 3D, whereas this effect was not seen in monolayers cultured on flat topologies.Citation174 This model was later advanced by the incorporation of endothelial cells and monocytes. When the co-culture system was infected by S. enterica, the pathogens were confirmed to target epithelial cells only, while the endothelial cells remained intact.Citation175

In addition to these native scaffolds, bioengineered 3D structures are widely used for modeling bacterial infections in order to increase standardization and avoid the dependence on fresh biological specimens. These scaffolds are produced using synthetic polymers such as poly(lactic-co-glycolic acid) (PLGA) Citation176,Citation177 or poly(ethylene)glycol diacrylate (PEGDA) Citation178 which are then coated with hydrogels or ECM derived proteins.Citation163,Citation171 Using such an approach, collagen-based 3D villi scaffolds were fabricated and seeded with Caco-2 cells to investigate interactions between the human intestinal epithelium and enteric pathogens (). This reconstitution of the intestinal topology resulted in increased expression of mucins scaffold-cultured cells compared to 2D culture.Citation163,Citation179 The mucin layer protected the epithelial cells from infection with the prototypic adherent-invasive E. coli strain LF82 or invasive S. Typhimurium, which was reversible by knock-down of the mucin MUC17. 3D scaffolds have also been used for the culture of cells derived from dissociated intestinal organoids, resulting in the development of a physiologically relevant mimic of the human small intestinal epithelium with crypt-villus architecture, appropriate cell-lineage compartmentalization and dedicated stem cell niches.Citation171 3D bioengineered scaffolds can furthermore be integrated with the culture of intact organoids. Specifically, a 3D collagen scaffold with physiological topography was presented in which primary intestinal fibroblasts were integrated to mimic the intestinal stroma.Citation180 Culture or intestinal organoids on this scaffold resulted in the epithelialization of the scaffold and the correct formation of functionally distinct villi and crypt domains without the need for organoid dissociation, thus providing a versatile means for future infectious disease studies.

An interesting recent development is the bioprinting on 3D scaffolds. Using bioinks containing a decellularized small intestine submucosa, Caco-2 cells and human umbilical vein endothelial cells (HUVECs) were printed to reconstruct an intestinal epithelium with high aspect ratio 3D villi and a connected underlying capillary network.Citation181,Citation182 The model closely mimicked the geometry of human intestinal villi, exhibited enhanced barrier function and expressed higher levels of intestinal enzymes (ALP and ANPEP) and mucins (MUC17), markers of mature enterocytes, compared to either conventional monolayers or 3D models without endothelial bed. While such bioprinted systems are promising to further increase the level of detail at which human intestinal physiology can be modeled, they have not yet been utilized for the study of enteric infections. Furthermore, lack of flow, shear and peristalsis, as well as the impedance of diffusion caused by the polymeric matrix remain important to consider for studies based on scaffold-based 3D models.

4.3 Microfluidic devices (“Gut-on-a-chip”)

Advances in microengineering technologies have resulted in the development of microphysiological systems in which cells are cultured in a perfused environment with hydrostatic forces and mechanical deformations. Arguably the first perfused intestinal model was based on a system comprising two microchannels which were divided by a rigid semipermeable membrane on which Caco-2 cells were seeded.Citation183 The model presented an important step forward in the development of biomimetic in vitro models, but it still lacked relevant physiological features such as villi structures, cellular complexity and peristaltic motions. To address these limitations, the rigid polyester membrane was replaced with a porous flexible polycarbonate membrane and two vacuum chambers were integrated on the sides of the device to introduce cyclic strain and emulate peristaltic motion.Citation184 Application of microfluidic flow and mechanical strain resulted in improved enterocytic phenotypes, mucus production, metabolic activity and the formation of crypt-villi microstructures with subsets of cells differentiating into Goblet and enteroendocrine cells.Citation185,Citation186 Upon infection with S. flexneri, bacteria rapidly colonized the crypt-like invaginations resulting in high rates of infectivity with minimal bacterial loads in the microphysiological system, which were further amplified by peristaltic forces (). In contrast, infectivity in monolayer culture was 10,000-fold lower.Citation164

Model complexity was further increased by co-culture with endothelial cells at the opposite side of the membrane. Strikingly, exposure of such co-cultures to bacterial lipopolysaccharides on the endothelial (basolateral) side resulted in increased production of pro-inflammatory cytokines and loss of barrier integrity, whereas exposure on the apical side did not result in inflammation, but even further increased epithelial tightness.Citation187 Such selective immunotolerance is critical for meaningful studies of host-microbe interactions. The relevance and potential of these systems was furthermore demonstrated by co-cultures with commensal microbes. Specifically, colonization of the microphysiological model with a probiotic formulation of eight strains of beneficial bacteria (VSL#3 formulation) resulted in more in vivo-like transcriptomic profiles, as well as inhibition of pathogenic E. coli growth and resulting epithelial injury.Citation165 Importantly, microfluidic models are moreover compatible with the stable co-culture of communities of aerobic and anaerobic human gut microbiota and application of a physiological oxygen gradient in an engineered anaerobic chamber allowed to sustain relevant levels of microbial diversity, consisting of over 200 unique operational taxonomic units from 11 genera.Citation188

These results demonstrate that the continuous or frequent-pulse flow in microfluidic systems that replenishes nutrients and removes metabolic waste products as well as non-adherent bacteria, prevents bacterial overgrowth and extends model stability from a few hours to multiple days, thereby providing powerful tools for the emulation of microbial homeostasis and imbalance. Combined, these engineered models allow to combine 3D intestinal architecture, biochemical gradients, mechanical cues and microbial complexity to stimulate the organization and differentiation of primary cells. These models thus provide the most biomimetic in vitro models for bacterial infection available to date.

4.4 Enteric ex vivo models

In vitro assays constitute reductionistic approaches that allow to probe and functionally challenge intestinal biology and pathobiology in accessible systems. It is important to note though that these technologies are reductionistic and even the most advanced culture systems do not include the diverse populations of innate and adaptive immune cells present in the human mucosa and cannot mimic innervation, vascularization or muscular coating. For this reason, ex vivo cultures, ranging from the short-term culture of intestinal slices or explants to intravital analyses of intestinal loops in anesthetized animals, constitute important complements to the arsenal of intestinal in vitro systems. Specifically, ileal loops have served as an important model for the study of enteric bacterial infections for many decades. In this paradigm, animals, mostly rabbits or mice, are laparotomized, followed by the exposure of a few centimeters of intestinal loops, which are carefully ligated and injected with a defined bacterial suspension. Subsequent histological and biochemical evaluation of the infected tissues and non-infected control loops have revealed important disease mechanisms and host responses and have enabled the development of protective strategies against various enteric infections, including S. flexneri, Clostridium perfringens, enteropathogenic E. coli and C. jejuni.Citation189–193

The arguably most common ex vivo culture model is based on the Ussing chamber system, an experimental apparatus in which an epithelium, e.g. obtained from an intestinal biopsy, is mounted vertically to separate two chambers. By measuring electric potential differences between the electrodes in the adjacent compartments, the setup can quantify epithelial barrier properties, permeability as well as ion transport. Ussing chambers have been used to study the effects of various enteric pathogens on intestinal integrity. For S. flexneri infections, Ussing chamber experiments have facilitated the identification of enterotoxins and demonstrated their rapid and dose-dependent effects of intestinal absorption.Citation194,Citation195 Moreover, enterohemorrhagic but not non-pathogenic E. coli was identified to increase expression of the receptor for the neuropeptide galanin 1 and subsequent exposure to galanin increased short-circuit currents in the Ussing chamber by 20-fold, linking the molecular pathogenesis to the excessive fluid secretion in bacterial dysentery.Citation196 Similar secretory responses with disruption of intestinal integrity were found for C. jejuni,Citation197 Clostridioides difficile Citation198 and Enterococcus faecalis.Citation199 Furthermore, ex vivo infection of mounted human ileal biopsies with S. Typhi followed by transcriptomic profiling revealed that the host mucosal immune response was downregulated already during the first two hours of invasion.Citation200

Cultures of precision-cut slices from mice, rat, chicken and human intestines have emerged as model systems for the evaluation of intestinal metabolism and toxicity.Citation201 These systems accurately reflect the cellular and structural complexity of the tissue in situ with myenteric and submucosal neuronal plexuses and functional interstitial cells of Cajal.Citation202 However, their functional stability is restricted to a few hours and, as such, these systems are currently not in widespread use for studies of enteric infections.

Intravital microscopy (IVM) of intestinal loops plays a significant role in elucidating the host immune response of macrophages, dendritic cells, T cells, B cells, and various innate-like lymphocytes to enteric infections.Citation203 These methods draw on the extensive repertoire of fluorescent imaging methods to study intestinal barrier transport of pharmacologically active peptides.Citation204 Upon infection with S. Typhimurium, multiphoton imaging of these loops showed that intraepithelial lymphocytes augmented scanning behavior by increasing their motility and squeezing in between the intestinal cells.Citation166 Furthermore, dendritic cells concentrated in the epithelium and efficiently phagocytose bacteria by pulling them from the lumen using intraepithelial dendrites.Citation167 Similarly, CX3CR1+ cells were observed to rapidly migrate into the intestinal lumen in the vicinity of the Salmonella clusters and control the initial infection 205. Notably, peristalsis and the resulting movement of the tissue challenges the acquisition of stable images and, thus, advanced software tools are required to correct and improve the visualization of dynamic events, such as cell-cell interactions.Citation205

Open questions

As outlined above, intestinal in vitro and ex vivo models have contributed extensively to our understanding of the pathobiology of enteric diseases. However, multiple important frontiers remain. Although pathogen-host interactions have been studied in great detail, much less is known about the roles of the microbiota in colonization resistance, but also in promotion of infections upon dysbiosis. Recently developed models which allow for more long-term concomitant exposures to microbiome components and pathogens are starting to shed light on these intra-kingdom interactions Citation206–208 and we anticipate that these and other investigations will provide important experimental insights into the interplay between the microbial flora and intestinal health. Along similar lines, unravelling of the molecular mechanisms underlying the beneficial roles of probiotics on the human host beyond the simple prevention of pathogen exclusion can contribute to new insights into human well-being.

Another important aspect that we find to be currently understudied is the impact of genetic and epigenetic factors in both microbes and host on infectivity and pathogenesis. Polymorphisms in multiple human immune-related genes, including various interleukins and genes encoding the major histocompatibility complex, have been implicated in the susceptibility to enteric infections.Citation209 However, these genetic association studies are often underpowered, lack replication in independent cohorts and are not supported by functional assays, which is of particular importance for synonymous variations or variants in non-coding regions of the genome. We believe that organoid culture that is amenable to genetic editing provides an important opportunity to directly probe the effects of genetic variations on infectious disease phenotypes. Similarly, in vitro systems allow for the systematic investigations of genetic variability at the level of the pathogen.

Concluding remarks

Enteric infections remain a main global health concern accounting for >1.6 million deaths per year, more than 25% of which in children under the age of five.Citation210 While minimizing malnutrition, contaminated drinking water and unsafe sanitation constitute the main means for the prevention of severe disease, obtaining a deeper mechanistic understanding of gut homeostasis and pathogenesis is of paramount importance for the development of new therapies. Experimental model systems that faithfully reflect infectious disease biology constitute essential tools in this regard. Over the past 60 years, conventional 2D monolayer cultures have played critical roles in the identification of factors in both microbes and host cells that are critical for infectivity. More recently, this culture method has been complemented with a multitude of 3D in vitro and ex vivo systems that differ in complexity, stability, versatility and function ().

Figure 4. Representative schematic of biological models applicable to study enteric bacterial infections and host-pathogen interactions in the intestine. The specific features, as well as advantages and limitations of intestinal model systems are highlighted and compared with respect to complexity, physiological relevance and scalability. Note that transwells, engineered scaffolds and microphysiological systems are compatible with the culture of cell lines, primary intestinal cells as well as dissociated organoidsCitation211.

Figure 4. Representative schematic of biological models applicable to study enteric bacterial infections and host-pathogen interactions in the intestine. The specific features, as well as advantages and limitations of intestinal model systems are highlighted and compared with respect to complexity, physiological relevance and scalability. Note that transwells, engineered scaffolds and microphysiological systems are compatible with the culture of cell lines, primary intestinal cells as well as dissociated organoidsCitation211.

Monolayer cultures remain the method of choice for high-throughput applications, such as large-scale screening based on transposon mutagenesis, RNA interference, genetic editing or pharmacological modulation. Intestinal organoids provide unprecedented opportunities to reconstruct the cellular complexity of the human intestine in vitro and they provide critical building blocks for bioengineered systems. Major advantages of 3D scaffolds and microphysiological systems are the ability to reconstitute intestinal tissue architecture and integrate biochemical gradients, flow and mechanical cues, which extends the functional lifespan of in vitro cultures, allows the cultivation of previously non-cultivatable pathogens and facilitates the incorporation of a physiologically relevant and stable microbiome. Lastly, ex vivo cultures of intact tissue explants and intravital imaging of ligatured intestinal loops have facilitated discoveries into the effects of enteric infections on fluid secretion, as well as into the behavior of immune cells upon infection in their native microenvironment. Thus, instead of being replacements for conventional 2D cultures, we see these emerging, complex in vitro and ex vivo systems as complements that each allow to address unique sets of questions.

Disclosure statement

VML is CEO and shareholder of HepaPredict AB, as well as co-founder and shareholder of PersoMedix AB. The other authors declare no conflicts of interest.

Additional information

Funding

The work received support by the Swedish Research Council [grant agreement numbers: 2019-01837], by the EU/EFPIA/OICR/McGill/KTH/Diamond Innovative Medicines Initiative 2 Joint Undertaking (EUbOPEN grant number 875510), by the Robert Bosch Foundation, Stuttgart, Germany and by the Lillian Sagens and Curt Ericssons Research Foundation.

References

  • Pickard JM, Zeng MY, Caruso R, Núñez G. Gut microbiota: role in pathogen colonization, immune responses, and inflammatory disease. Immunol Rev. 2017;279(1):70–26. doi:10.1111/imr.12567.
  • Kamada N, Chen GY, Inohara N, Núñez G. Control of pathogens and pathobionts by the gut microbiota. Nat Immunol. 2013;14(7):685–690. doi:10.1038/ni.2608.
  • Rogers AWL, Tsolis RM, Bäumler AJ. Salmonella versus the microbiome. Microbiol Mol Biol R. 2020;85:e00027–19.
  • Steele MI, Motta EVS, Gattu T, Martinez D, Moran NA. The gut microbiota protects bees from invasion by a bacterial pathogen. Microbiol Spectr. 2021;9(2):e00394–21. doi:10.1128/Spectrum.00394-21.
  • Yurist-Doutsch S, Arrieta M-C, Vogt SL, Finlay BB. Gastrointestinal microbiota–mediated control of enteric pathogens. Annu Rev Genet. 2014;48(1):1–22. doi:10.1146/annurev-genet-120213-092421.
  • Sepich-Poore GD, Zitvogel L, Straussman R, Hasty J, Wargo JA, Knight R. The microbiome and human cancer. Science. 2021;371(6536). doi:10.1126/science.abc4552.
  • Luca FD, Shoenfeld Y. The microbiome in autoimmune diseases. Clin Exp Immunol. 2019;195(1):74–85. doi:10.1111/cei.13158.
  • Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–71. doi:10.1038/s41579-020-0433-9.
  • Nikolova VL, Smith MRB, Hall LJ, Cleare AJ, Stone JM, Young AH. Perturbations in gut microbiota composition in psychiatric disorders. JAMA Psychiatry. 2021;78(12):1343–1354. doi:10.1001/jamapsychiatry.2021.2573.
  • Ulluwishewa D, Anderson RC, McNabb WC, Moughan PJ, Wells JM, Roy NC. Regulation of tight junction permeability by intestinal bacteria and dietary components. J Nutrition. 2011;141(5):769–776. doi:10.3945/jn.110.135657.
  • Allaire JM, Crowley SM, Law HT, Chang S-Y, Ko H-J, Vallance BA. The intestinal epithelium: central coordinator of mucosal immunity. Trends Immunol. 2018;39(9):677–696. doi:10.1016/j.it.2018.04.002.
  • Yoo JY, Groer M, Dutra SVO, Sarkar A, McSkimming DI. Gut microbiota and immune system interactions. Microorganisms. 2020;8(10):1587. doi:10.3390/microorganisms8101587.
  • Hansson GC. Mucins and the microbiome. Annu Rev Biochem. 2020;89(1):1–25. doi:10.1146/annurev-biochem-011520-105053.
  • Campbell C, McKenney PT, Konstantinovsky D, Isaeva OI, Schizas M, Verter J, Mai C, Jin W-B, Guo C-J, Violante S, et al. Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells. Nature. 2020;581(7809):475–479. doi:10.1038/s41586-020-2193-0.
  • Guttman JA, Finlay BB. Tight junctions as targets of infectious agents. Biochim Biophys Acta. 2009;1788(4):832–841. doi:10.1016/j.bbamem.2008.10.028.
  • Paradis T, Bègue H, Basmaciyan L, Dalle F, Bon F. Tight junctions as a key for pathogens invasion in intestinal epithelial cells. Int J Mol Sci. 2021;22(5):2506. doi:10.3390/ijms22052506.
  • Engevik MA, Yacyshyn MB, Engevik KA, Wang J, Darien B, Hassett DJ, Yacyshyn BR, Worrell RT. Human Clostridium difficile infection: altered mucus production and composition. Am J Physiol Gastrointest Liver Physiol. 2015;308(6):G510–G524. doi:10.1152/ajpgi.00091.2014.
  • Hill DR, Huang S, Nagy MS, Yadagiri VK, Fields C, Mukherjee D, Bons B, Dedhia PH, Chin AM, Tsai Y-H, et al. Bacterial colonization stimulates a complex physiological response in the immature human intestinal epithelium. eLife. 2017;6:237ra65. doi:10.7554/eLife.29132.
  • Radoshevich L, Cossart P. Listeria monocytogenes: towards a complete picture of its physiology and pathogenesis. Nat Rev Microbiol. 2018;16(1):32–46. doi:10.1038/nrmicro.2017.126.
  • Czepiel J, Dróżdż M, Pituch H, Kuijper EJ, Perucki W, Mielimonka A, Goldman S, Wultańska D, Garlicki A, Biesiada G, et al. Clostridium difficile infection: review. Eur J Clin Microbiol Infect Dis. 2019;38(7):1211–1221. doi:10.1007/s10096-019-03539-6.
  • Shoaib M, Shehzad A, Raza H, Niazi S, Khan IM, Akhtar W, Safdar W, Wang Z. A comprehensive review on the prevalence, pathogenesis and detection of Yersinia enterocolitica. RSC Adv. 2019;9(70):41010–41021. doi:10.1039/C9RA06988G.
  • Fernández-Bravo A, Figueras MJ. An update on the genus aeromonas: taxonomy, epidemiology, and pathogenicity. Microorganisms. 2020;8(1):129. doi:10.3390/microorganisms8010129.
  • Scherer WF, Syverton JT. Studies on the propagation in vitro of poliomyelitis viruses. III. The propagation of poliomyelitis viruses in tissue cultures devoid of nerve cells. J Exp Med. 1952;96(4):389–400. doi:10.1084/jem.96.4.389.
  • Kaneko M, Emoto Y, Emoto MA. Simple reproducible inexpensive, yet old-fashioned method for determining phagocytic and bactericidal activities of macrophages. Yonsei Med J. 2016;57(2):283–290. doi:10.3349/ymj.2016.57.2.283.
  • Hidalgo IJ, Raub TJ, Borchardt RT. Characterization of the human colon carcinoma cell line (Caco-2) as a model system for intestinal epithelial permeability. Gastroenterology. 1989;96(2):736–749. doi:10.1016/S0016-5085(89)80072-1.
  • Youhanna S, Lauschke VM. The past, present and future of intestinal in vitro cell systems for drug absorption studies. J Pharm Sci. 2021;110(1):50–65. doi:10.1016/j.xphs.2020.07.001.
  • Shen JX, Youhanna S, Shafagh RZ, Kele J, Lauschke VM. Organotypic and microphysiological models of liver, gut, and kidney for studies of drug metabolism, pharmacokinetics, and toxicity. Chem Res Toxicol. 2020;33(1):38–60. doi:10.1021/acs.chemrestox.9b00245.
  • Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, et al. Organotypic and microphysiological human tissue models for drug discovery and development—current state-of-the-art and future perspectives. Pharmacol Rev. 2022;74(1):141–206. doi:10.1124/pharmrev.120.000238.
  • Sambuy Y, De Angelis I, Ranaldi G, Scarino ML, Stammati A, Zucco F. The Caco-2 cell line as a model of the intestinal barrier: influence of cell and culture-related factors on Caco-2 cell functional characteristics. Cell Biol Toxicol. 2005;21(1):1–26. doi:10.1007/s10565-005-0085-6.
  • Gullberg E, Leonard M, Karlsson J, Hopkins AM, Brayden D, Baird AW, Artursson P. Expression of specific markers and particle transport in a new human intestinal M-cell model. Biochem Biophys Res Commun. 2000;279(3):808–813. doi:10.1006/bbrc.2000.4038.
  • Kasendra M, Tovaglieri A, Sontheimer-Phelps A, Jalili-Firoozinezhad S, Bein A, Chalkiadaki A, Scholl W, Zhang C, Rickner H, Richmond CA, Li H. Development of a primary human small intestine-on-a-chip using biopsy-derived organoids. Sci Rep. 2018;8(1):2871–14. doi:10.1038/s41598-018-21201-7.
  • Takahashi Y, Noguchi M, Inoue Y, Sato S, Shimizu M, Kojima H, Okabe T, Kiyono H, Yamauchi Y, Sato R. Organoid-derived intestinal epithelial cells are a suitable model for preclinical toxicology and pharmacokinetic studies. IScience. 2022;25(7):104542. doi:10.1016/j.isci.2022.104542.
  • Price AE, Shamardani K, Lugo KA, Deguine J, Roberts AW, Lee BL, Barton GM. A map of toll-like receptor expression in the intestinal epithelium reveals distinct spatial, cell type-specific, and temporal patterns. Immunity. 2018;49(3):560–575.e6. doi:10.1016/j.immuni.2018.07.016.
  • Kobayashi T, Ogawa M, Sanada T, Mimuro H, Kim M, Ashida H, Akakura R, Yoshida M, Kawalec M, Reichhart J-M, et al. The shigella OspC3 effector inhibits caspase-4, antagonizes inflammatory cell death, and promotes epithelial infection. Cell Host Microbe. 2013;13(5):570–583. doi:10.1016/j.chom.2013.04.012.
  • Knodler LA, Crowley S, Sham H, Yang H, Wrande M, Ma C, Ernst R, Steele-Mortimer O, Celli J, Vallance B, et al. Noncanonical inflammasome activation of Caspase-4/Caspase-11 mediates epithelial defenses against enteric bacterial pathogens. Cell Host Microbe. 2014;16(2):249–256. doi:10.1016/j.chom.2014.07.002.
  • Kozuka K, He Y, Koo-McCoy S, Kumaraswamy P, Nie B, Shaw K, Chan P, Leadbetter M, He L, Lewis JG, et al. Development and characterization of a human and mouse intestinal epithelial cell monolayer platform. Stem Cell Reports. 2017;9(6):1976–1990. doi:10.1016/j.stemcr.2017.10.013.
  • VanDussen KL, Marinshaw JM, Shaikh N, Miyoshi H, Moon C, Tarr PI, Ciorba MA, Stappenbeck TS. Development of an enhanced human gastrointestinal epithelial culture system to facilitate patient-based assays. Gut. 2015;64(6):911–920. doi:10.1136/gutjnl-2013-306651.
  • Pontier C, Pachot J, Botham R, Lenfant B, Arnaud P. HT29-MTX and Caco-2/TC7 monolayers as predictive models for human intestinal absorption: role of the mucus layer. J Pharm Sci. 2001;90(10):1608–1619. doi:10.1002/jps.1111.
  • Bazes A, Nollevaux G, Coco R, Joly A, Sergent T, Schneider Y-J. Development of a triculture based system for improved benefit/risk assessment in pharmacology and human food. BMC Proc. 2011;5(8):67. doi:10.1186/1753-6561-5-S8-P67.
  • Antunes F, Andrade F, Araújo F, Ferreira D, Sarmento B. Establishment of a triple co-culture in vitro cell models to study intestinal absorption of peptide drugs. Eur J Pharm Biopharm. 2013;83(3):427–435. doi:10.1016/j.ejpb.2012.10.003.
  • Lamprokostopoulou A, Römling U. Yin and yang of biofilm formation and Cyclic di-GMP signaling of the gastrointestinal pathogen Salmonella enterica serovar Typhimurium. J Innate Immun. 2021;14(4):275–292. doi:10.1159/000519573.
  • Polzin S, Huber C, Eylert E, Elsenhans I, Eisenreich W, Schmidt H. Growth media simulating ileal and colonic environments affect the intracellular proteome and carbon fluxes of enterohemorrhagic Escherichia coli O157:H7 Strain EDL933. Appl Environ Microb. 2013;79(12):3703–3715. doi:10.1128/AEM.00062-13.
  • Karaolis DKR, Means TK, Yang D, Takahashi M, Yoshimura T, Muraille E, Philpott D, Schroeder JT, Hyodo M, Hayakawa Y, et al. Bacterial c-di-GMP is an immunostimulatory molecule. J Immunol. 2007;178(4):2171–2181. doi:10.4049/jimmunol.178.4.2171.
  • Lamprokostopoulou A, Monteiro C, Rhen M, Römling U. Cyclic di-GMP signalling controls virulence properties of Salmonella enterica serovar Typhimurium at the mucosal lining. Environ Microbiol. 2010;12(1):40–53. doi:10.1111/j.1462-2920.2009.02032.x.
  • Römling U, Galperin MY, Gomelsky M. Cyclic di-GMP: the first 25 years of a universal bacterial second messenger. Microbiol Mol Biol Rev. 2013;77:1–52.
  • Dey AK, Bhagat A, Chowdhury R. Host cell contact induces expression of virulence factors and VieA, a Cyclic di-GMP phosphodiesterase, in vibrio cholerae. J Bacteriol. 2013;195(9):2004–2010. doi:10.1128/JB.02127-12.
  • Sassone-Corsi M, Raffatellu M. No vacancy: how beneficial microbes cooperate with immunity to provide colonization resistance to pathogens. J Immunol. 2015;194(9):4081–4087. doi:10.4049/jimmunol.1403169.
  • Ducarmon QR, Zwittink RD, Hornung BV, Van Schaik W, Young VB, Kuijper EJ. Gut microbiota and colonization resistance against bacterial enteric infection. Microbiol Mol Biol R. 2019;83(3):e00007–e000019.
  • Samuels AN, Roggiani M, Smith KA, Zhu J, Goulian M, Kohli RM. Deciphering the role of colicins during colonization of the mammalian gut by Commensal E. coli. Microorganisms. 2020;8(5):664. doi:10.3390/microorganisms8050664.
  • Fabich AJ, Jones SA, Chowdhury FZ, Cernosek A, Anderson A, Smalley D, McHargue JW, Hightower GA, Smith JT, Autieri SM, Leatham MP. Comparison of carbon nutrition for pathogenic and commensal Escherichia coli strains in the mouse intestine. Infect Immun. 2008;76(3):1143–1152. doi:10.1128/IAI.01386-07.
  • Frederick A, Huang Y, Pu M, Rowe-Magnus DA. Vibrio cholerae Type VI activity alters motility behavior in mucin. J Bacteriol. 2020;202(24). doi:10.1128/JB.00261-20.
  • Joshi A, Kostiuk B, Rogers A, Teschler J, Pukatzki S, Yildiz FH. Rules of engagement: the Type VI secretion system in vibrio cholerae. Trends Microbiol. 2017;25(4):267–279. doi:10.1016/j.tim.2016.12.003.
  • Pukatzki S, Ma AT, Sturtevant D, Krastins B, Sarracino D, Nelson WC, Heidelberg JF, Mekalanos JJ. Identification of a conserved bacterial protein secretion system in Vibrio cholerae using the Dictyostelium host model system. Proc Natl Acad Sci USA. 2006;103(5):1528–1533. doi:10.1073/pnas.0510322103.
  • Bachmann V, Kostiuk B, Unterweger D, Diaz-Satizabal L, Ogg S, Pukatzki S. Bile salts modulate the mucin-activated type VI secretion system of pandemic vibrio cholerae. PLoS Neglect Trop Dis. 2015;9(8):e0004031. doi:10.1371/journal.pntd.0004031.
  • Serapio-Palacios A, Woodward SE, Vogt SL, Deng W, Creus-Cuadros A, Huus KE, Cirstea M, Gerrie M, Barcik W, Yu H, Finlay BB. Type VI secretion systems of pathogenic and commensal bacteria mediate niche occupancy in the gut. Cell Rep. 2022;39(4):110731. doi:10.1016/j.celrep.2022.110731.
  • Sokaribo AS, Balezantis LR, MacKenzie KD, Wang Y, Palmer MB, Chung B, Herman NJ, McCarthy MC, Chen JM, White AP, et al. A SNP in the cache 1 signaling domain of diguanylate cyclase STM1987 leads to increased in vivo fitness of invasive salmonella strains. Infect Immun. 2021;89(4):e00810–20. doi:10.1128/IAI.00810-20.
  • Rey C, Chang -Y-Y, Latour-Lambert P, Varet H, Proux C, Legendre R, Coppée J-Y, Enninga J. Transcytosis subversion by M cell-to-enterocyte spread promotes Shigella flexneri and Listeria monocytogenes intracellular bacterial dissemination. PLoS Pathog. 2020;16(4):e1008446. doi:10.1371/journal.ppat.1008446.
  • Labrec EH, Schneider H, Magnani TJ, Formal SB. Epithelial cell penetration as as essential step in the pathogenesis of bacillary dysentery. J Bacteriol. 1964;88(5):1503–1518. doi:10.1128/jb.88.5.1503-1518.1964.
  • Gemski P, Takeuchi A, Washington O, Formal SB. Shigellosis due to shigella dysenteriae 1: relative importance of mucosal invasion versus toxin production in pathogenesis. J Infect Dis. 1972;126(5):523–530. doi:10.1093/infdis/126.5.523.
  • Lampel KA, Formal† SB, Maurelli AT, Kaper J. A brief history of shigella. EcoSal Plus. 2018;8(1). doi:10.1128/ecosalplus.ESP-0006-2017.
  • Sansonetti PJ, Kopecko DJ, Formal SB. Involvement of a plasmid in the invasive ability of Shigella flexneri. Infect Immun. 1982;35(3):852–860. doi:10.1128/iai.35.3.852-860.1982.
  • Maurelli AT, Baudry B, d’Hauteville H, Hale TL, Sansonetti PJ. Cloning of plasmid DNA sequences involved in invasion of HeLa cells by Shigella flexneri. Infect Immun. 1985;49(1):164–171. doi:10.1128/iai.49.1.164-171.1985.
  • Clerc P, Ryter A, Mounier J, Sansonetti PJ. Plasmid-mediated intracellular multiplication of Shigella flexneri. Ann Inst Pasteur Microbiol. 1986;137:315–320.
  • Sansonetti PJ, Ryter A, Clerc P, Maurelli AT, Mounier J. Multiplication of Shigella flexneri within HeLa cells: lysis of the phagocytic vacuole and plasmid-mediated contact hemolysis. Infect Immun. 1986;51(2):461–469. doi:10.1128/iai.51.2.461-469.1986.
  • Clerc PL, Ryter A, Mounier J, Sansonetti PJ. Plasmid-mediated early killing of eucaryotic cells by Shigella flexneri as studied by infection of J774 macrophages. Infect Immun. 1987;55(3):521–527. doi:10.1128/iai.55.3.521-527.1987.
  • Bernardini ML, Mounier J, d’Hauteville H, Coquis-Rondon M, Sansonetti PJ. Identification of icsA, a plasmid locus of Shigella flexneri that governs bacterial intra- and intercellular spread through interaction with F-actin. Proc Natl Acad Sci USA. 1989;86(10):3867–3871. doi:10.1073/pnas.86.10.3867.
  • Westermann AJ, Förstner KU, Amman F, Barquist L, Chao Y, Schulte LN, Müller L, Reinhardt R, Stadler PF, Vogel J, et al. Dual RNA-seq unveils noncoding RNA functions in host–pathogen interactions. Nature. 2016;529(7587):496–501. doi:10.1038/nature16547.
  • Chang S-J, Hsu Y-T, Chen Y, Lin -Y-Y, Lara-Tejero M, Galan JE. Typhoid toxin sorting and exocytic transport from Salmonella Typhi-infected cells. Elife. 2022;11:e78561. doi:10.7554/eLife.78561.
  • Sheahan K-L, Isberg RR, Miller JF. Identification of mammalian proteins that collaborate with type iii secretion system function: involvement of a chemokine receptor in supporting translocon activity. mBio. 2015;6(1):e02023–14. doi:10.1128/mBio.02023-14.
  • Suzuki S, Mimuro H, Kim M, Ogawa M, Ashida H, Toyotome T, Franchi L, Suzuki M, Sanada T, Suzuki T, Tsutsui H. Shigella IpaH7.8 E3 ubiquitin ligase targets glomulin and activates inflammasomes to demolish macrophages. Proc Natl Acad Sci USA. 2014;111(40):E4254–E4263. doi:10.1073/pnas.1324021111.
  • Li P, Jiang W, Yu Q, Liu W, Zhou P, Li J, Xu J, Xu B, Wang F, Shao F, et al. Ubiquitination and degradation of GBPs by a Shigella effector to suppress host defence. Nature. 2017;551(7680):378–383. doi:10.1038/nature24467.
  • Mattock E, Blocker AJ. How do the virulence factors of shigella work together to cause disease? Front Cell Infect Microbiol. 2017;7:64. doi:10.3389/fcimb.2017.00064.
  • Agaisse H. Molecular and cellular mechanisms of shigella flexneri dissemination. Front Cell Infect Microbiol. 2016;6:29. doi:10.3389/fcimb.2016.00029.
  • Köseoğlu VK, Agaisse H, Freitag NE. Evolutionary perspectives on the moonlighting functions of bacterial factors that support actin-based motility. mBio. 2019;10(4):e01520–19. doi:10.1128/mBio.01520-19.
  • Qin J, Hong Y, Morona R, Totsika M. Cysteine-dependent conformational heterogeneity of shigella flexneri autotransporter IcsA and implications of its function. Microbiol Spectr. 2022:e03410–22. doi:10.1128/spectrum.03410-22.
  • Duncan-Lowey JK, Wiscovitch AL, Wood TE, Goldberg MB, Russo BC. Shigella flexneri disruption of cellular tension promotes intercellular spread. Cell Rep. 2020;33(8):108409. doi:10.1016/j.celrep.2020.108409.
  • Campbell-Valois F-X, Sachse M, Sansonetti PJ, Parsot C. Escape of actively secreting shigella flexneri from ATG8/LC3-positive vacuoles formed during cell-to-cell spread is facilitated by IcsB and VirA. mBio. 2015;6:e02567–14.
  • Wen SC, Best E, Nourse C. Non-typhoidal Salmonella infections in children: review of literature and recommendations for management. J Paediatr Child Health. 2017;53(10):936–941. doi:10.1111/jpc.13585.
  • Gordon MA. Salmonella infections in immunocompromised adults. J Infection. 2008;56(6):413–422. doi:10.1016/j.jinf.2008.03.012.
  • Kingsley RA, Msefula CL, Thomson NR, Kariuki S, Holt KE, Gordon MA, Harris D, Clarke L, Whitehead S, Sangal V, et al. Epidemic multiple drug resistant Salmonella Typhimurium causing invasive disease in sub-Saharan Africa have a distinct genotype. Genome Res. 2009;19(12):2279–2287. doi:10.1101/gr.091017.109.
  • Jong HKD, Parry CM, Poll TVD, Wiersinga WJ, Chitnis CE. Host–pathogen interaction in invasive salmonellosis. PLoS Pathog. 2012;8(10):e1002933. doi:10.1371/journal.ppat.1002933.
  • Rivera-Chávez F, Winter SE, Lopez CA, Xavier MN, Winter MG, Nuccio S-P, Russell JM, Laughlin RC, Lawhon SD, Sterzenbach T, et al. Salmonella uses energy taxis to benefit from intestinal inflammation. PLoS Pathog. 2013;9(4):e1003267. doi:10.1371/journal.ppat.1003267.
  • Faber F, Thiennimitr P, Spiga L, Byndloss MX, Litvak Y, Lawhon S, Andrews-Polymenis HL, Winter SE, Bäumler AJ. Respiration of microbiota-derived 1,2-propanediol drives salmonella expansion during colitis. PLoS Pathog. 2017;13(1):e1006129. doi:10.1371/journal.ppat.1006129.
  • Anderson CJ, Medina CB, Barron BJ, Karvelyte L, Aaes TL, Lambertz I, Perry JSA, Mehrotra P, Gonçalves A, Lemeire K, et al. Microbes exploit death-induced nutrient release by gut epithelial cells. Nature. 2021;596(7871):262–267. doi:10.1038/s41586-021-03785-9.
  • Schlumberger MC, Müller AJ, Ehrbar K, Winnen B, Duss I, Stecher B, Hardt W-D. Real-time imaging of type III secretion: salmonella SipA injection into host cells. Proc Natl Acad Sci USA. 2005;102(35):12548–12553. doi:10.1073/pnas.0503407102.
  • Fulde M, van Vorst K, Zhang K, Westermann AJ, Busche T, Huei YC, Welitschanski K, Froh I, Pägelow D, Plendl J, et al. SPI2 T3SS effectors facilitate enterocyte apical to basolateral transmigration of Salmonella -containing vacuoles in vivo. Gut Microbes. 2021;13(1):1973836. doi:10.1080/19490976.2021.1973836.
  • Otten EG, Werner E, Crespillo-Casado A, Boyle KB, Dharamdasani V, Pathe C, Santhanam B, Randow F. Ubiquitylation of lipopolysaccharide by RNF213 during bacterial infection. Nature. 2021;594(7861):111–116. doi:10.1038/s41586-021-03566-4.
  • Galán JE, Curtiss R. Cloning and molecular characterization of genes whose products allow Salmonella typhimurium to penetrate tissue culture cells. Proc Natl Acad Sci USA. 1989;86(16):6383–6387. doi:10.1073/pnas.86.16.6383.
  • Bäumler AJ, Tsolis RM, Heffron F. Contribution of fimbrial operons to attachment to and invasion of epithelial cell lines by Salmonella typhimurium. Infect Immun. 1996;64(5):1862–1865. doi:10.1128/iai.64.5.1862-1865.1996.
  • Gewirtz AT, Simon PO, Schmitt CK, Taylor LJ, Hagedorn CH, O’Brien AD, Neish AS, Madara JL. Salmonella typhimurium translocates flagellin across intestinal epithelia, inducing a proinflammatory response. J Clin Invest. 2001;107(1):99–109. doi:10.1172/JCI10501.
  • Monack DM, Bouley DM, Falkow S. Salmonella typhimurium persists within macrophages in the mesenteric lymph nodes of chronically infected Nramp1+/+ mice and can be reactivated by IFNγ neutralization. J Exp Med. 2004;199(2):231–241. doi:10.1084/jem.20031319.
  • Saliba A-E, Westermann AJ, Appenzeller S, Stapels DA, Schulte LN, Helaine S, Vogel J. Single-cell RNA-seq ties macrophage polarization to growth rate of intracellular Salmonella. Nat Microbiol. 2016;2(2]):16206. doi:10.1038/nmicrobiol.2016.206.
  • Pontes MH, Lee E-J, Choi J, Groisman EA. Salmonella promotes virulence by repressing cellulose production. Proc Natl Acad Sci USA. 2015;112(16):5183–5188. doi:10.1073/pnas.1500989112.
  • Petersen E, Mills E, Miller SI. Cyclic-di-GMP regulation promotes survival of a slow-replicating subpopulation of intracellular Salmonella Typhimurium. Proc Natl Acad Sci USA. 2019;116:6335–6340.
  • Ahmad I, Lamprokostopoulou A, Le Guyon S, Streck E, Barthel M, Peters V, Hardt WD, Römling U. Complex c-di-GMP signaling networks mediate transition between virulence properties and biofilm formation in salmonella enterica serovar typhimurium. PLoS One. 2011;6(12):e28351. doi:10.1371/journal.pone.0028351.
  • Stapels DAC, Hill PWS, Westermann AJ, Fisher RA, Thurston TL, Saliba A-E, Blommestein I, Vogel J, Helaine S. Salmonella persisters undermine host immune defenses during antibiotic treatment. Science. 2018;362(6419):1156–1160. doi:10.1126/science.aat7148.
  • Eng S-K, Pusparajah P, Ab Mutalib N-S, Ser H-L, Chan K-G, Lee L-H. Salmonella : a review on pathogenesis, epidemiology and antibiotic resistance. Front Life Sci. 2015;8(3):284–293. doi:10.1080/21553769.2015.1051243.
  • Hirose K, Ezaki T, Miyake M, Li T, Khan AQ, Kawamura Y, Yokoyama H, Takami T. Survival of Vi‐capsulated and Vi‐deleted Salmonella typhi strains in cultured macrophage expressing different levels of CD14 antigen. FEMS Microbiol Lett. 1997;147(2):259–265. doi:10.1111/j.1574-6968.1997.tb10251.x.
  • Sharma A, Qadri A. Vi polysaccharide of Salmonella typhi targets the prohibitin family of molecules in intestinal epithelial cells and suppresses early inflammatory responses. Proc Natl Acad Sci USA. 2004;101(50):17492–17497. doi:10.1073/pnas.0407536101.
  • Parween F, Yadav J, Qadri A. The virulence polysaccharide of salmonella typhi suppresses activation of Rho family GTPases to limit inflammatory responses from epithelial cells. Front Cell Infect Microbiol. 2019;9:141. doi:10.3389/fcimb.2019.00141.
  • Song J, Gao X, Galán JE. Structure and function of the Salmonella Typhi chimaeric A2B5 typhoid toxin. Nature. 2013;499(7458):350–354. doi:10.1038/nature12377.
  • Grasso F, Frisan T. Bacterial genotoxins: merging the DNA damage response into infection biology. Biomolecules. 2015;5(3):1762–1782. doi:10.3390/biom5031762.
  • Kanungo S, Azman AS, Ramamurthy T, Deen J, Dutta S. Cholera. Lancet. 2022;399(10333):1429–1440. doi:10.1016/S0140-6736(22)00330-0.
  • Sengupta C, Mukherjee O, Chowdhury R. Adherence to intestinal cells promotes biofilm formation in vibrio cholerae. J Infect Dis. 2016;214(10):1571–1578. doi:10.1093/infdis/jiw435.
  • Schluter J, Nadell CD, Bassler BL, Foster KR. Adhesion as a weapon in microbial competition. ISME J. 2015;9(1):139–149. doi:10.1038/ismej.2014.174.
  • Valiente E, Davies C, Mills DC, Getino M, Ritchie JM, Wren BW. Vibrio cholerae accessory colonisation factor AcfC: a chemotactic protein with a role in hyperinfectivity. Sci Rep. 2018;8(1):8390. doi:10.1038/s41598-018-26570-7.
  • Grognot M, Mittal A, Mah’moud M, Taute KM, Alexandre G. Vibrio cholerae motility in aquatic and mucus-mimicking environments. Appl Environ Microbiol. 2021;87(20):e01293–21. doi:10.1128/AEM.01293-21.
  • Alst AJV, DiRita VJ. Aerobic metabolism in vibrio cholerae is required for population expansion during infection. mBio. 2020;11(5):e01989–20. doi:10.1128/mBio.01989-20.
  • Bhowmick R, Ghosal A, Das B, Koley H, Saha DR, Ganguly S, Nandy RK, Bhadra RK, Chatterjee NS. Intestinal adherence of vibrio cholerae involves a coordinated interaction between colonization factor GbpA and mucin. Infect Immun. 2008;76(11):4968–4977. doi:10.1128/IAI.01615-07.
  • Ghasemi M, Bakhshi B, Khashei R, Soudi S. Modulatory effect of Vibrio cholerae toxin co-regulated pilus on mucins, toll-like receptors and NOD genes expression in co-culture model of Caco-2 and peripheral blood mononuclear cells (PBMC). Microb Pathog. 2020;149:104566. doi:10.1016/j.micpath.2020.104566.
  • Ghasemi M, Bakhshi B, Khashei R, Soudi S, Boustanshenas M. Vibrio cholerae toxin coregulated pilus provokes inflammatory responses in Coculture model of Caco-2 and peripheral blood mononuclear cells (PBMC) leading to increased colonization. Microbiol Immunol. 2021;65(6):238–244. doi:10.1111/1348-0421.12889.
  • Kaisar MH, Bhuiyan MS, Akter A, Saleem D, Iyer AS, Dash P, Hakim A, Chowdhury F, Khan AI, Calderwood SB, et al. Vibrio cholerae Sialidase-specific immune responses are associated with protection against cholera. mSphere. 2021;6(2):e01232–20. doi:10.1128/mSphere.01232-20.
  • Detzner J, Püttmann C, Pohlentz G, Müthing J. Ingenious action of vibrio cholerae neuraminidase recruiting additional GM1 cholera toxin receptors for primary human colon epithelial cells. Microorganisms. 2022;10(6):1255. doi:10.3390/microorganisms10061255.
  • Zingl FG, Thapa HB, Scharf M, Kohl P, Müller AM, Schild S. Outer membrane vesicles of vibrio cholerae protect and deliver active cholera toxin to host cells via porin-dependent uptake. mBio. 2021;12(3):e00534–21. doi:10.1128/mBio.00534-21.
  • Sánchez J, Holmgren J. Cholera toxin structure, gene regulation and pathophysiological and immunological aspects. Cell Mol Life Sci. 2008;65(9):1347–1360. doi:10.1007/s00018-008-7496-5.
  • Guichard A, Cruz-Moreno B, Aguilar B, Van sorge N, Kuang J, Kurkciyan A, Wang Z, Hang S, Pineton de chambrun G, McCole D, et al. Cholera toxin disrupts barrier function by inhibiting exocyst-mediated trafficking of host proteins to intestinal cell junctions. Cell Host Microbe. 2013;14(3):294–305. doi:10.1016/j.chom.2013.08.001.
  • Blount ZD. The unexhausted potential of E. coli. Elife. 2015;4:e05826. doi:10.7554/eLife.05826.
  • Vatanen T, Kostic A, d’Hennezel E, Siljander H, Franzosa E, Yassour M, Kolde R, Vlamakis H, Arthur T, Hämäläinen A-M, et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell. 2016;165(4):842–853. doi:10.1016/j.cell.2016.04.007.
  • Krall LJ, Klein S, Boutin S, Wu CC, Sähr A, Stanifer ML, Boulant S, Heeg K, Nurjadi D, Hildebrand D, et al. Invasiveness of Escherichia coli is associated with an IncFII plasmid. Pathogens. 2021;10(12):1645. doi:10.3390/pathogens10121645.
  • Qiu J, Nie Y, Zhao Y, Zhang Y, Li L, Wang R, Wang M, Chen S, Wang J, Li Y-Q, et al. Safeguarding intestine cells against enteropathogenic Escherichia coli by intracellular protein reaction, a preventive antibacterial mechanism. Proc Natl Acad Sci USA. 2020;117(10):5260–5268. doi:10.1073/pnas.1914567117.
  • Prudent V, Demarre G, Vazeille E, Wery M, Quenech’Du N, Ravet A, Dauverd - Girault J, van Dijk E, Bringer M-A, Descrimes M, et al. The Crohn’s disease-related bacterial strain LF82 assembles biofilm-like communities to protect itself from phagolysosomal attack. Commun Biol. 2021;4(1):627. doi:10.1038/s42003-021-02161-7.
  • Dejea CM, Fathi P, Craig JM, Boleij A, Taddese R, Geis AL, Wu X, DeStefano Shields CE, Hechenbleikner EM, Huso DL, et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science. 2018;359(6375):592–597. doi:10.1126/science.aah3648.
  • Wilson MR, Jiang Y, Villalta PW, Stornetta A, Boudreau PD, Carrá A, Brennan CA, Chun E, Ngo L, Samson LD, et al. The human gut bacterial genotoxin colibactin alkylates DNA. Science. 2019;363(6428). doi:10.1126/science.aar7785.
  • Sheppard SK, Maiden MCJ. The evolution of Campylobacter jejuni and Campylobacter coli. Cold Spring Harb Perspect Biol. 2015;7(8):a018119. doi:10.1101/cshperspect.a018119.
  • Boehm M, Hoy B, Rohde M, Tegtmeyer N, Bæk KT, Oyarzabal OA, Brøndsted L, Wessler S, Backert S. Rapid paracellular transmigration of Campylobacter jejuni across polarized epithelial cells without affecting TER: role of proteolytic-active HtrA cleaving E-cadherin but not fibronectin. Gut Pathog. 2012;4(3). doi:10.1186/1757-4749-4-3.
  • Day CJ, Tran EN, Semchenko EA, Tram G, Hartley-Tassell LE, Ng PSK, King RM, Ulanovsky R, McAtamney S, Apicella MA, et al. Glycan:glycan interactions: high affinity biomolecular interactions that can mediate binding of pathogenic bacteria to host cells. Proc Natl Acad Sci USA. 2015;112(52):E7266–E7275. doi:10.1073/pnas.1421082112.
  • Freitag CM, Strijbis K, Putten JPMV. Host cell binding of the flagellar tip protein of Campylobacter jejuni. Cell Microbiol. 2017;19(6):e12714. doi:10.1111/cmi.12714.
  • Fukushima S, Shimohata T, Inoue Y, Kido J, Uebanso T, Mawatari K, Takahashi A. Recruitment of LC3 by campylobacter jejuni to bacterial invasion site on host cells via the Rac1-mediated signaling pathway. Front Cell Infect Microbiol. 2022;12:829682. doi:10.3389/fcimb.2022.829682.
  • Taylor ZW, Raushel FM. Cytidine diphosphoramidate kinase: an enzyme required for the biosynthesis of the O -methyl phosphoramidate modification in the capsular polysaccharides of Campylobacter jejuni. Biochemistry. 2018;57(15):2238–2244. doi:10.1021/acs.biochem.8b00279.
  • Zamora CY, Ward EM, Kester JC, Chen WLK, Velazquez JG, Griffith LG, Imperiali B. Application of a gut-immune co-culture system for the study of N -glycan-dependent host–pathogen interactions of Campylobacter jejuni. Glycobiology. 2020;30(6):374–381. doi:10.1093/glycob/cwz105.
  • Zheng J, Meng J, Zhao S, Singh R, Song W. Campylobacter -induced interleukin-8 secretion in polarized human intestinal epithelial cells requires campylobacter -secreted cytolethal distending toxin- and toll-like receptor-mediated activation of NF-κB. Infect Immun. 2008;76(10):4498–4508. doi:10.1128/IAI.01317-07.
  • Cao X, Lest CHAVD, Huang LZX, Putten JP, van M, Wösten MMSM. Campylobacter jejuni permeabilizes the host cell membrane by short chain lysophosphatidylethanolamines. Gut Microbes. 2022;14(1):2091371. doi:10.1080/19490976.2022.2091371.
  • Saha C, Mohanraju P, Stubbs A, Dugar G, Hoogstrate Y, Kremers G-J, van Cappellen WA, Horst-Kreft D, Laffeber C, Lebbink JHG, et al. Guide-free Cas9 from pathogenic Campylobacter jejuni bacteria causes severe damage to DNA. Sci Adv. 2020;6(25):eaaz4849. doi:10.1126/sciadv.aaz4849.
  • Cui J, Duizer C, Bouwman LI, van Rooijen KS, Voogdt CGP, van Putten JPM, de Zoete MR. The ALPK1 pathway drives the inflammatory response to Campylobacter jejuni in human intestinal epithelial cells. PLoS Pathog. 2021;17(8):e1009787. doi:10.1371/journal.ppat.1009787.
  • Zhou P, She Y, Dong N, Li P, He H, Borio A, Wu Q, Lu S, Ding X, Cao Y, et al. Alpha-kinase 1 is a cytosolic innate immune receptor for bacterial ADP-heptose. Nature. 2018;561(7721):122–126. doi:10.1038/s41586-018-0433-3.
  • Pfannkuch L, Hurwitz R, Trauisen J, Sigulla J, Poeschke M, Matzner L, Kosma P, Schmid M, Meyer TF. ADP heptose, a novel pathogen-associated molecular pattern identified in Helicobacter pylori. FASEB J. 2019;33(8):9087–9099. doi:10.1096/fj.201802555R.
  • Martin-Gallausiaux C, Garcia-Weber D, Lashermes A, Larraufie P, Marinelli L, Teixeira V, Rolland A, Béguet-Crespel F, Brochard V, Quatremare T, et al. Akkermansia muciniphil upregulates genes involved in maintaining the intestinal barrier function via ADP-heptose-dependent activation of the ALPK1/TIFA pathway. Gut Microbes. 2022;14(1):2110639. doi:10.1080/19490976.2022.2110639.
  • Günther C, Winner B, Neurath MF, Stappenbeck TS. Organoids in gastrointestinal diseases: from experimental models to clinical translation. Gut. 2022;71(9):1892–1908. doi:10.1136/gutjnl-2021-326560.
  • Taelman J, Diaz M, Guiu J. Human intestinal organoids: promise and challenge. Front Cell Dev Biol. 2022;10:854740. doi:10.3389/fcell.2022.854740.
  • Haber AL, Biton M, Rogel N, Herbst RH, Shekhar K, Smillie C, Burgin G, Delorey TM, Howitt MR, Katz Y, et al. A single-cell survey of the small intestinal epithelium. Nature. 2017;551(7680):333–339. doi:10.1038/nature24489.
  • Rouch JD, Scott A, Lei NY, Solorzano-Vargas RS, Wang J, Hanson EM, Kobayashi M, Lewis M, Stelzner MG, Dunn JCY, et al. Development of functional Microfold (M) cells from intestinal stem cells in primary human enteroids. PLoS One. 2016;11(1):e0148216. doi:10.1371/journal.pone.0148216.
  • 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(4):e00740–18. doi:10.1128/IAI.00740-18.
  • Pradhan S, Weiss AA, Barbour AG. Probiotic properties of Escherichia coli Nissle in human intestinal organoids. mBio. 2020;11(4):e01470–20. doi:10.1128/mBio.01470-20.
  • Heo I, Dutta D, Schaefer DA, Iakobachvili N, Artegiani B, Sachs N, Boonekamp KE, Bowden G, Hendrickx APA, Willems RJL, et al. Modelling Cryptosporidium infection in human small intestinal and lung organoids. Nat Microbiol. 2018;3(7):814–823. doi:10.1038/s41564-018-0177-8.
  • Forbester JL, Lees EA, Goulding D, Forrest S, Yeung A, Speak A, Clare S, Coomber EL, Mukhopadhyay S, Kraiczy J, et al. Interleukin-22 promotes phagolysosomal fusion to induce protection against Salmonella enterica Typhimurium in human epithelial cells. Proc Natl Acad Sci USA. 2018;115(40):10118–10123. doi:10.1073/pnas.1811866115.
  • Geiser P, Di Martino ML, Samperio Ventayol P, Eriksson J, Sima E, Al-Saffar AK, Ahl D, Phillipson M, Webb D-L, Sundbom M, et al. Salmonella enterica serovar Typhimurium exploits cycling through epithelial cells to colonize human and murine enteroids. mBio. 2021;12(1):e02684–20. doi:10.1128/mBio.02684-20.
  • Williamson IA, Arnold JW, Samsa LA, Gaynor L, DiSalvo M, Cocchiaro JL, Carroll I, Azcarate-Peril MA, Rawls JF, Allbritton NL, et al. A high-throughput organoid microinjection platform to study gastrointestinal microbiota and luminal physiology. Cell Mol Gastroenterol Hepatol. 2018;6(3):301–319. doi:10.1016/j.jcmgh.2018.05.004.
  • Co JY, Margalef-Català M, Monack DM, Amieva MR. Controlling the polarity of human gastrointestinal organoids to investigate epithelial biology and infectious diseases. Nat Protoc. 2021;16(11):5171–5192. doi:10.1038/s41596-021-00607-0.
  • Co JY, Margalef-Català M, Li X, Mah AT, Kuo CJ, Monack DM, Amieva MR. Controlling epithelial polarity: a human enteroid model for host-pathogen interactions. Cell Rep. 2019;26(9):2509–2520.e4. doi:10.1016/j.celrep.2019.01.108.
  • Rajan A, Vela L, Zeng X-L, Yu X, Shroyer N, Blutt SE, Poole NM, Carlin LG, Nataro JP, Estes MK, et al. Novel segment- and host-specific patterns of enteroaggregative Escherichia coli adherence to human intestinal enteroids. mBio. 2018;9(1). doi:10.1128/mBio.02419-17.
  • Nickerson KP, Llanos-Chea A, Ingano L, Serena G, Miranda-Ribera A, Perlman M, Lima R, Sztein MB, Fasano A, Senger S, et al. A versatile human intestinal organoid-derived epithelial monolayer model for the study of enteric pathogens. Microbiol Spectr. 2021;9(1):e00003–21. doi:10.1128/Spectrum.00003-21.
  • 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(4). doi:10.1128/IAI.00733-18.
  • Ventayol PS, Geiser P, Di Martino ML, Florbrant A, Fattinger SA, Walder N, Sima E, Shao F, Gekara NO, Sundbom M, et al. Bacterial detection by NAIP/NLRC4 elicits prompt contractions of intestinal epithelial cell layers. Proc Natl Acad Sci USA. 2021;118(16):e2013963118. doi:10.1073/pnas.2013963118.
  • Rijn J, van M, Grüttner J, Sundbom M, Webb D-L, Hellström PM, Svärd SG, Sellin ME. High-definition DIC imaging uncovers transient stages of pathogen infection cycles on the surface of human adult stem cell-derived intestinal epithelium. mBio. 2022;13(1):e00022–22. doi:10.1128/mbio.00022-22.
  • Fasciano AC, Dasanayake GS, Estes MK, Zachos NC, Breault DT, Isberg RR, Tan S, Mecsas J. Yersinia pseudotuberculosis YopE prevents uptake by M cells and instigates M cell extrusion in human ileal enteroid-derived monolayers. Gut Microbes. 2021;13(1):1988390. doi:10.1080/19490976.2021.1988390.
  • Ettayebi K, Crawford SE, Murakami K, Broughman JR, Karandikar U, Tenge VR, Neill FH, Blutt SE, Zeng X-L, Qu L, et al. Replication of human noroviruses in stem cell-derived human enteroids. Science. 2016;353(6306):1387–1393. doi:10.1126/science.aaf5211.
  • Hosmillo M, Chaudhry Y, Nayak K, Sorgeloos F, Koo B-K, Merenda A, Lillestol R, Drumright L, Zilbauer M, Goodfellow I, et al. Norovirus replication in human intestinal epithelial cells is restricted by the interferon-induced JAK/STAT signaling pathway and RNA polymerase II-mediated transcriptional responses. mBio. 2020;11(2):e00215–20. doi:10.1128/mBio.00215-20.
  • Saxena K, Blutt SE, Ettayebi K, Zeng X-L, Broughman JR, Crawford SE, Karandikar UC, Sastri NP, Conner ME, Opekun AR, et al. Human intestinal enteroids: a new model to study human rotavirus infection, host restriction, and pathophysiology. J Virol. 2016;90(1):43–56. doi:10.1128/JVI.01930-15.
  • RePass MAD, Chen Y, Lin Y, Zhou W, Kaplan DL, Ward HD. Novel bioengineered three-dimensional human intestinal model for long-term infection of Cryptosporidium parvum. Infect Immun. 2017;65(3): e00731–16.
  • Wilke G, Funkhouser-Jones LJ, Wang Y, Ravindran S, Wang Q, Beatty WL, Baldridge MT, VanDussen KL, Shen B, Kuhlenschmidt MS, et al. A stem-cell-derived platform enables complete cryptosporidium development in vitro and genetic tractability. Cell Host Microbe. 2019;26(1):123–134.e8. doi:10.1016/j.chom.2019.05.007.
  • Bartfeld S. Modeling infectious diseases and host-microbe interactions in gastrointestinal organoids. Dev Biol. 2016;420(2):262–270. doi:10.1016/j.ydbio.2016.09.014.
  • Nikolaev M, Mitrofanova O, Broguiere N, Geraldo S, Dutta D, Tabata Y, Elci B, Brandenberg N, Kolotuev I, Gjorevski N, et al. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature. 2020;585(7826):574–578. doi:10.1038/s41586-020-2724-8.
  • Kim SH, Chi M, Yi B, Kim SH, Oh S, Kim Y, Park S, Sung JH. Three-dimensional intestinal villi epithelium enhances protection of human intestinal cells from bacterial infection by inducing mucin expression. Integr Biol. 2014;6(12):1122–1131. doi:10.1039/c4ib00157e.
  • Grassart A, Malardé V, Gobaa S, Sartori-Rupp A, Kerns J, Karalis K, Marteyn B, Sansonetti P, Sauvonnet N. Bioengineered human organ-on-chip reveals intestinal microenvironment and mechanical forces impacting shigella infection. Cell Host Microbe. 2019;26(3):435–444.e4. doi:10.1016/j.chom.2019.08.007.
  • Kim HJ, Li H, Collins JJ, Ingber DE. Contributions of microbiome and mechanical deformation to intestinal bacterial overgrowth and inflammation in a human gut-on-a-chip. Proc Natl Acad Sci USA. 2016;113(1):E7–15. doi:10.1073/pnas.1522193112.
  • Konijnenburg D, van PH, Pedicord VA, Farache J, Victora GD, Mucida D. Intestinal epithelial and intraepithelial T cell crosstalk mediates a dynamic response to infection. Cell. 2017;171(4):783–794.e13. doi:10.1016/j.cell.2017.08.046.
  • Farache J, Koren I, Milo I, Gurevich I, Kim K-W, Zigmond E, Furtado G, Lira S, Shakhar G. Luminal bacteria recruit CD103+ dendritic cells into the intestinal epithelium to sample bacterial antigens for presentation. Immunity. 2013;38(3):581–595. doi:10.1016/j.immuni.2013.01.009.
  • Garreta E, Kamm RD, Chuva de Sousa Lopes SM, Lancaster MA, Weiss R, Trepat X, Hyun I, Montserrat N. Rethinking organoid technology through bioengineering. Nat Mater. 2021;20(2):145–155. doi:10.1038/s41563-020-00804-4.
  • Pentinmikko N, Lozano R, Scharaw S, Andersson S, Englund JI, Castillo-Azofeifa D, Gallagher A, Broberg M, Song K-Y, Sola Carvajal A, et al. Cellular shape reinforces niche to stem cell signaling in the small intestine. Sci Adv. 2022;8(41):eabm1847. doi:10.1126/sciadv.abm1847.
  • Taebnia N, Zhang R, Kromann EB, Dolatshahi-Pirouz A, Andresen TL, Larsen NB. Dual-material 3D-printed intestinal model devices with integrated villi-like scaffolds. ACS Appl Mater Inter. 2021;13(49):58434–58446. doi:10.1021/acsami.1c22185.
  • Wang Y, Gunasekara DB, Reed MI, DiSalvo M, Bultman SJ, Sims CE, Magness ST, Allbritton NL. A microengineered collagen scaffold for generating a polarized crypt-villus architecture of human small intestinal epithelium. Biomaterials. 2017;128:44–55. doi:10.1016/j.biomaterials.2017.03.005.
  • McCrary MW, Bousalis D, Mobini S, Song YH, Schmidt CE. Decellularized tissues as platforms for in vitro modeling of healthy and diseased tissues. Acta Biomater. 2020;111:1–19. doi:10.1016/j.actbio.2020.05.031.
  • Xi W, Saleh J, Yamada A, Tomba C, Mercier B, Janel S, Dang T, Soleilhac M, Djemat A, Wu H, et al. Modulation of designer biomimetic matrices for optimized differentiated intestinal epithelial cultures. Biomaterials. 2022;282:121380. doi:10.1016/j.biomaterials.2022.121380.
  • Alzheimer M, Svensson SL, König F, Schweinlin M, Metzger M, Walles H, Sharma CM. A three-dimensional intestinal tissue model reveals factors and small regulatory RNAs important for colonization with Campylobacter jejuni. PLoS Pathog. 2020;16(2):e1008304. doi:10.1371/journal.ppat.1008304.
  • Schulte LN, Schweinlin M, Westermann AJ, Janga H, Santos SC, Appenzeller S, Walles H, Vogel J, Metzger M. An advanced human intestinal coculture model reveals compartmentalized host and pathogen strategies during salmonella infection. mBio. 2020;11(1):e03348–19. doi:10.1128/mBio.03348-19.
  • Costello CM, Hongpeng J, Shaffiey S, Yu J, Jain NK, Hackam D, March JC. Synthetic small intestinal scaffolds for improved studies of intestinal differentiation. Biotechnol Bioeng. 2014;111(6):1222–1232. doi:10.1002/bit.25180.
  • Costello CM, Sorna RM, Goh Y-L, Cengic I, Jain NK, March JC. 3-D intestinal scaffolds for evaluating the therapeutic potential of probiotics. Mol Pharm. 2014;11(7):2030–2039. doi:10.1021/mp5001422.
  • Creff J, Courson R, Mangeat T, Foncy J, Souleille S, Thibault C, Besson A, Malaquin L. Fabrication of 3D scaffolds reproducing intestinal epithelium topography by high-resolution 3D stereolithography. Biomaterials. 2019;221:119404. doi:10.1016/j.biomaterials.2019.119404.
  • García-Díaz M, Cendra MDM, Alonso-Roman R, Urdániz M, Torrents E, Martínez E. Mimicking the intestinal host–pathogen interactions in a 3D in vitro model: the role of the mucus layer. Pharmaceutics. 2022;14(8):1552. doi:10.3390/pharmaceutics14081552.
  • Verhulsel M, Simon A, Bernheim-Dennery M, Gannavarapu VR, Gérémie L, Ferraro D, Krndija D, Talini L, Viovy J-L, Vignjevic DM, et al. Developing an advanced gut on chip model enabling the study of epithelial cell/fibroblast interactions. Lab Chip. 2020;21(2):365–377. doi:10.1039/D0LC00672F.
  • Kim W, Kim G. Intestinal villi model with blood capillaries fabricated using collagen-based bioink and dual-cell-printing process. ACS Appl Mater Inter. 2018;10(48):41185–41196. doi:10.1021/acsami.8b17410.
  • Kim W, Kim GH. An intestinal model with a finger-like villus structure fabricated using a bioprinting process and collagen/SIS-based cell-laden bioink. Theranostics. 2020;10(6):2495–2508. doi:10.7150/thno.41225.
  • Kimura H, Yamamoto T, Sakai H, Sakai Y, Fujii T. An integrated microfluidic system for long-term perfusion culture and on-line monitoring of intestinal tissue models. Lab Chip. 2008;8(5):741–746. doi:10.1039/b717091b.
  • Kim HJ, Huh D, Hamilton G, Ingber DE. Human gut-on-a-chip inhabited by microbial flora that experiences intestinal peristalsis-like motions and flow. Lab Chip. 2012;12(12):2165–10. doi:10.1039/c2lc40074j.
  • Kim HJ, Ingber DE. Gut-on-a-Chip microenvironment induces human intestinal cells to undergo villus differentiation. Integr Biol. 2013;5(9):1130. doi:10.1039/c3ib40126j.
  • Delon LC, Guo Z, Oszmiana A, Chien -C-C, Gibson R, Prestidge C, Thierry B. A systematic investigation of the effect of the fluid shear stress on Caco-2 cells towards the optimization of epithelial organ-on-chip models. Biomaterials. 2019;225:119521. doi:10.1016/j.biomaterials.2019.119521.
  • Maurer M, Gresnigt MS, Last A, Wollny T, Berlinghof F, Pospich R, Cseresnyes Z, Medyukhina A, Graf K, Gröger M, et al. A three-dimensional immunocompetent intestine-on-chip model as in vitro platform for functional and microbial interaction studies. Biomaterials. 2019;220:119396. doi:10.1016/j.biomaterials.2019.119396.
  • Jalili-Firoozinezhad S, Gazzaniga FS, Calamari EL, Camacho DM, Fadel CW, Bein A, Swenor B, Nestor B, Cronce MJ, Tovaglieri A, et al. A complex human gut microbiome cultured in an anaerobic intestine-on-a-chip. Nat Biomed Eng. 2019;3(7):520–531. doi:10.1038/s41551-019-0397-0.
  • Duncan CL, Strong DH. Ileal loop fluid accumulation and production of diarrhea in rabbits by cell-free products of clostridium perfringens. J Bacteriol. 1969;100(1):86–94. doi:10.1128/jb.100.1.86-94.1969.
  • Punyashthiti K, Finkelstein RA. Enteropathogencity of Escherichia coli I. Evaluation of mouse intestinal loops. Infect Immun. 1971;4(4):473–478. doi:10.1128/iai.4.4.473-478.1971.
  • Everest PH, Goossens H, Sibbons P, Lloyd DR, Knutton S, Leece R, Ketley JM, Williams PH. Pathological changes in the rabbit ileal loop model caused by Campylobacter jejuni from human colitis. J Med Microbiol. 1993;38(5):316–321. doi:10.1099/00222615-38-5-316.
  • Flamant M, Aubert P, Rolli-Derkinderen M, Bourreille A, Neunlist MR, Mahe MM, Meurette G, Marteyn B, Savidge T, Galmiche JP, et al. Enteric glia protect against Shigella flexneri invasion in intestinal epithelial cells: a role for S-nitrosoglutathione. Gut. 2011;60(4):473. doi:10.1136/gut.2010.229237.
  • Yum LK, Byndloss MX, Feldman SH, Agaisse H. Critical role of bacterial dissemination in an infant rabbit model of bacillary dysentery. Nat Commun. 2019;10(1):1826. doi:10.1038/s41467-019-09808-4.
  • Fasano A, Noriega FR, Liao FM, Wang W, Levine MM. Effect of shigella enterotoxin 1 (ShET1) on rabbit intestine in vitro and in vivo. Gut. 1997;40(4):505. doi:10.1136/gut.40.4.505.
  • Faherty CS, Harper JM, Shea-Donohue T, Barry EM, Kaper JB, Fasano A, Nataro JP. Chromosomal and plasmid-encoded factors of shigella flexneri induce secretogenic activity ex vivo. PLoS One. 2012;7(11):e49980. doi:10.1371/journal.pone.0049980.
  • Hecht G, Marrero JA, Danilkovich A, Matkowskyj KA, Savkovic SD, Koutsouris A, Benya RV. Pathogenic Escherichia coli increase Cl– secretion from intestinal epithelia by upregulating galanin-1 receptor expression. J Clin Invest. 1999;104(3):253–262. doi:10.1172/JCI6373.
  • Beltinger J, Del Buono J, Skelly MM, Thornley J, Spiller RC, Stack WA, Hawkey CJ. Disruption of colonic barrier function and induction of mediator release by strains of Campylobacter jejuni that invade epithelial cells. World J Gastroenterol. 2008;14(48):7345–7352. doi:10.3748/wjg.14.7345.
  • Jafari NV, Kuehne SA, Minton NP, Allan E, Bajaj-Elliott M. Clostridium difficile-mediated effects on human intestinal epithelia: modelling host-pathogen interactions in a vertical diffusion chamber. Anaerobe. 2016;37:96–102. doi:10.1016/j.anaerobe.2015.12.007.
  • Isenmann R, Schwarz M, Rozdzinski E, Marre R, Beger HG. Aggregation substance promotes colonic mucosal invasion of enterococcus faecalis in an ex vivo model. J Surg Res. 2000;89(2):132–138. doi:10.1006/jsre.1999.5813.
  • Nickerson KP, Senger S, Zhang Y, Lima R, Patel S, Ingano L, Flavahan WA, Kumar DKV, Fraser CM, Faherty CS, et al. 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.
  • Li M, Graaf IAMD, Groothuis GMM. Precision-cut intestinal slices: alternative model for drug transport, metabolism, and toxicology research. Expert Opin Drug Metab Toxicol. 2016;12(2):175–190. doi:10.1517/17425255.2016.1125882.
  • Schwerdtfeger LA, Ryan EP, Tobet SA. An organotypic slice model for ex vivo study of neural, immune, and microbial interactions of mouse intestine. Am J Gastrointest Liver Physiol. 2016;310(4):G240–8. doi:10.1152/ajpgi.00299.2015.
  • Kolesnikov M, Farache J, Shakhar G. Intravital two-photon imaging of the gastrointestinal tract. J Immunol Methods. 2015;421:73–80. doi:10.1016/j.jim.2015.03.008.
  • Larsen JB, Taebnia N, Dolatshahi-Pirouz A, Eriksen AZ, Hjørringgaard C, Kristensen K, Larsen NW, Larsen NB, Marie R, Mündler A-K, et al. Imaging therapeutic peptide transport across intestinal barriers. RSC Chem Biol. 2021;2(4):1115–1143. doi:10.1039/D1CB00024A.
  • Soulet D, Paré A, Coste J, Lacroix S, Sensi SL. Automated filtering of intrinsic movement artifacts during two-photon intravital microscopy. PLoS One. 2013;8(1):e53942. doi:10.1371/journal.pone.0053942.
  • McGrath CJ, Laveckis E, Bell A, Crost E, Juge N, Schüller S. Development of a novel human intestinal model to elucidate the effect of anaerobic commensals on Escherichia coli infection. Dis Model Mech. 2022;15(4). doi:10.1242/dmm.049365.
  • Deng L, Tan KSW. Interactions between Blastocystis subtype ST4 and gut microbiota in vitro. Parasit Vector. 2022;15(1):80. doi:10.1186/s13071-022-05194-x.
  • Luchan J, Choi C, Carrier RL. Reactive oxygen species limit intestinal mucosa-bacteria homeostasis in vitro. Sci Rep. 2021;11(1):23727. doi:10.1038/s41598-021-02080-x.
  • Flores J, Okhuysen PC. Genetics of susceptibility to infection with enteric pathogens. Curr Opin Infect Dis. 2009;22(5):471–476. doi:10.1097/QCO.0b013e3283304eb6.
  • Troeger C, Blacker BF, Khalil IA, Rao PC, Cao S, Zimsen SR, Albertson SB, Stanaway JD, Deshpande A, Abebe Z, 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(11):1211–1228. doi:10.1016/S1473-3099(18)30362-1.
  • Man, A. L., Gicheva, N., Regoli, M., Rowley, G., De Cunto, G., Wellner, N., Bassity, E., Gulisano, M., Bertelli, E., Nicoletti, C., et al. CX 3 CR1 + Cell–Mediated Salmonella Exclusion Protects the Intestinal Mucosa during the Initial Stage of Infection. J Immunol. 2017;198(1):335–343. doi:10.4049/jimmunol.1502559