Skip to main content

Harnessing 3D models to uncover the mechanisms driving infectious and inflammatory disease in the intestine

Abstract

Representative models of intestinal diseases are transforming our knowledge of the molecular mechanisms of disease, facilitating effective drug screening and avenues for personalised medicine. Despite the emergence of 3D in vitro intestinal organoid culture systems that replicate the genetic and functional characteristics of the epithelial tissue of origin, there are still challenges in reproducing the human physiological tissue environment in a format that enables functional readouts. Here, we describe the latest platforms engineered to investigate environmental tissue impacts, host-microbe interactions and enable drug discovery. This highlights the potential to revolutionise knowledge on the impact of intestinal infection and inflammation and enable personalised disease modelling and clinical translation.

Background

Authentic models are required to decipher the complexity of intestinal disease

The intestinal tract undergoes significant morphological changes from development through to adulthood [1,2,3] generating regions with distinct cellular composition and function in digestion and nutrient absorption within the small intestine and colon [4]. The intestine is lined with a rapidly regenerating epithelial layer where resident stem cells located within crypts contribute to tissue homeostasis [5] and repair following injury [6, 7] (Fig. 1). The apical surface of the epithelium is continuously exposed to microbes and by-products in the lumen that can cause tissue damage contributing to diseases such as inflammatory bowel diseases (IBD). IBD and other diseases can vary between individuals [8, 9], suggesting disease modelling should be conducted using pre-clinical systems that can test responses from multiple patients. Immortalised cell lines [10] and animal models have been extensively utilised and have contributed to our understanding of cellular mechanisms and signalling pathways but they fail to recapitulate numerous physiological and phenotypic aspects of human disease [11].

Fig. 1
figure 1

Modelling epithelial and niche signals in intestinal organoids to mimic cellular diversity. The intestinal epithelium consists of stem and differentiated cells and is surrounded by mesenchymal stromal and immune cells that secrete key signals to support epithelial self-renewal, proliferation and differentiation. This complexity can be modelled in organoids by adding specific niche factors or cell types. Briefly, organoids are typically cultured in standard growth medium. To drive cell differentiation and diversity, organoids are cultured in a differentiation medium supplemented with reduced WNT3a (WNT3aLo) and RSPO1 (RSPO1Lo), and with niche factors like NRG1, or with stromal cells only. To model inflammation, organoids can be exposed to an inflammatory medium where standard growth medium is supplemented with different pro-inflammatory cytokines and bacterial components, or by introducing immune cells while maintaining standard organoid culture conditions

The ability to grow primary human intestinal cells as three-dimensional (3D) organoids [12, 13] has opened new avenues to more accurately model numerous aspects of organ physiology including cellular fate transitions, cell-to-cell communication and host-microbe interactions. Adult stem cell (ASC)-derived organoids were initially established from the mouse small intestine [13] and consist solely of epithelial cells. These organoids have the remarkable capacity to self-organise to closely resemble crypts in vivo where stems cells are intercalated between Paneth cells in buds and regions of proliferation and differentiation can be clearly identified [13, 14] (Fig. 1). Epithelial organoids also retain the regional-specific characteristics from where they were isolated enabling the mechanisms underlying regional identity to be examined [15]. Although intestinal organoids better resemble endogenous tissue compared to two-dimensional (2D) models, human intestinal organoid models still do not fully recapitulate the cellular diversity and differentiation states found in vivo [12, 16, 17]. It is also clear that mechanical signals and spatial interactions with other cell types including stromal, immune, endothelial and lymphatic cells modulate epithelial cell fate [7, 18,19,20,21,22,23,24,25,26,27,28,29,30]. In this review, we discuss the latest approaches in utilising organoids to characterise the impact of cellular signals and microbes on epithelial tissue, engineering approaches to advance organ-on-a-chip models, innovations in high-throughput screening, organoid phenotyping and artificial intelligence (AI). We further discuss these current advances in the field, evaluating challenges and applications to clinical translation.

Main text

Recapitulating the cellular diversity of the intestinal epithelium in vitro

The adult small intestinal epithelium is composed primarily of absorptive enterocytes with secretory cells including goblet, Paneth, enteroendocrine and tuft cells [31, 32] (Fig. 1). Crypts that invaginate into the underlying mesenchyme and house stem and progenitor cells are present throughout the GI tract, while villi that protrude into the lumen contain differentiated cells are only present in the small intestine [31]. In contrast, the adult colonic epithelium is predominantly composed of enterocytes and goblet cells, with a notably higher proportion of goblet cells compared to the small intestine [33, 34], reflecting its role in mucin secretion and water absorption. Additionally, the colon lacks Paneth cells but retains other secretory cell types such as enteroendocrine cells, which contribute to local hormonal signalling [33, 34]. The absence of villi in the colon results in a different structural organisation of the epithelium, which consists of deeper crypts and a thicker mucosal layer [35]. Overall, while both the small intestine and colon serve crucial roles in digestion and absorption, their cellular compositions and structures reflect their distinct physiological functions.

Recent single-cell and spatial transcriptomic analyses have revealed cellular composition and transcriptional profiles vary considerably along the proximal–distal axis in humans. This includes the characterisation of cell types such as Bestrophin 4 (BEST4) cells, a subset of the absorptive lineage that is present in humans, not mice [4, 36,37,38,39] (Fig. 1). Stem cells fuel the renewal of the intestinal lining every 4–5 days [40], with new cells proliferating within the transit-amplifying (TA) zone before terminally differentiating and migrating upwards along the crypt-villus axis [5] (Fig. 1).

A key breakthrough in 2009 demonstrated that intestinal organoids could be established from crypts or isolated Lgr5+ stem cells from mice, and when embedded in gel containing extracellular matrix, self-organised into stem and differentiation zones [13]. This study recapitulated supportive niche signals by the addition of R-spondin1, Noggin and WNT3a that are normally secreted from surrounding cell types [18, 23, 25, 30] (Fig. 1). These organoids supported the growth of cells that maintained apical-basal polarity, modelled the cellular heterogeneity of in vivo tissue and retained the transcriptional profile typical of their regional origin [15]. The methodology was then adapted to culture human tissue [12]. Although tissue can be grown, there are many more difficulties and variabilities in growing human intestinal organoid cultures, with cellular composition, level of maturity and organisation not fully representative of the repertoire of cell types and spatial organisation in the adult human intestine [12, 16, 41, 42]. Improvements in culture conditions are developing [17], but require further optimisation to generate a highly reproducible culture system that faithfully mirrors that of in vivo tissue. For instance, a recent study demonstrated that incorporating insulin-like growth factor 1 (IGF1) and fibroblast growth factor 2 (FGF2) into the culture medium promoted simultaneous multi-lineage differentiation and self-renewal of human intestinal organoids [43] (Fig. 1). These and other environmental factors discussed below can influence organoid phenotypes (Fig. 1). These changes can be quantitatively assessed through different screening protocols with machine learning aiding both tracking of organoids and image analysis [44,45,46,47].

Modelling interactions in the tissue microenvironment

In recent years, efforts to understand the regulation of intestinal stem cell plasticity, critical for epithelial barrier repair, mesenchymal remodelling and regeneration following damage, have driven continuous advancements in intestinal organoid co-culture with various resident cell populations and supportive scaffolds. Traditionally, elucidating aspects of cell–cell interactions relied on animal models; however, they fail to identify short-distance interactions. In vitro co-culture systems bridge this gap, providing a clearer understanding of key contributors by simplifying the complex in vivo environment. Several studies have demonstrated the feasibility of integrating induced pluripotent stem cell (iPSC)-derived organoids with neurons [48, 49], vascular endothelial cells [48, 50] and adipocytes [51]. However, limited studies are available on the co-culture of patient-derived organoids (PDOs) with niche cells, primarily due to the technical complexities involved in such cultures (reviewed in [52]). Some of these challenges include determining the type of interaction between the desired cell types (also defined as direct or indirect co-cultures), selecting the most suitable co-culture medium to support the growth of all relevant cell types and choosing the correct plasticware. For instance, transwells are mainly used for studying cell–cell interactions [53], while ultra-low attachment plates are better suited to grow apical-out organoids, useful for investigating microbe-epithelial cell interactions [54]. In contrast, tissue culture-treated plates are used to grow and expand organoids within an extracellular matrix, closely mimicking the tissue environment [13]. Building on this, selecting the correct matrix is crucial. Utilising synthetic matrices in place of the commonly used, but poorly defined basement membrane extracts like Matrigel, offers the potential to more accurately replicate the native tissue environment [55, 56]. This can be achieved by fine-tuning the biophysical properties of the synthetic matrix, such as stiffness, which is frequently heterogeneous in diseased tissues (reviewed in [57, 58]).

Despite these limitations, several significant studies have identified key regulatory signals that impact the activity of epithelial cells by co-culture with immune cells, fibroblasts and microbial components. A detailed overview of co-culturing epithelial cells with various components of the microenvironment is presented in the sections below.

Intestinal organoid co-cultures with stromal cells

Intestinal stem and progenitor cells are tightly regulated by niche factors produced by stromal cells that reside underneath the epithelium (reviewed in [18]). Fibroblasts are essential drivers of epithelial regeneration following damage [6, 7, 23,24,25]. Therefore, defining the activity of signalling molecules that are crucial for mucosal repair may have therapeutic potential in treating degenerative intestinal diseases, such as IBD, where the epithelial barrier is impaired.

Stromal cells have been identified as potential sources of key signals by molecular profiling that has identified the presence of WNT and BMP agonists and antagonists, in stromal cells along the crypt-villus axis [23,24,25, 30, 59, 60] (Fig. 1). Other signals such as members of the epidermal growth factor (EGF) family of ligands, secreted from stromal cells, have also been directly applied to organoid cultures in order to model in vivo biological activity [7, 17, 42, 61] (Fig. 1). Sources of these essential signals in the mesenchyme were proposed to be supplied by stromal cells marked by expression of platelet-derived growth factor receptor alpha (PDGFRɑ), subepithelial telocytes, that localise in close proximity to cells within crypts and villi, and trophocytes located near the base of crypts marked by expression of CD81 (Fig. 1). The function of these cells has been verified by co-culture experiments, enabling the assessment of stromal-epithelial interactions. Most methods involve embedding 3D organoids in Matrigel on top of a layer of fibroblasts, allowing direct contact for morphological assessment [62, 63]. While this method facilitates direct interaction, it poses challenges in separating the compartments for downstream analysis, such as secretome profiling. To address this, transwell systems can be used, where organoids are seeded in Matrigel in the apical compartment, and stromal cells as monolayers in the basal compartment [64]. Building on this, co-culturing intestinal organoids with specific stromal cell types has proven successful in enabling the long-term culture of organoids in the absence of growth factors that are typically essential for their survival (Fig. 1). For example, telocytes [23, 25, 60, 65] and PDGFRɑ-low trophocytes, source of crucial stem cell growth factors GREM1, WNT2b and R-spondin3 [30], fully support organoid growth in culture ex vivo. These studies have identified the function of specific cell types and insight into the contributions of specific growth factors. However, this approach fails to recreate the complexity of the stromal niche that establishes a spatial morphogen gradient in vivo [30] (Fig. 1). This has been partially addressed with intestinal assembloid cultures that combine diverse stromal cell types with epithelial organoids to produce self-organising structures with distinct stem and differentiated cellular compartments [66]. This highlights the importance of bi-directional signals between epithelial and stromal cells.

Intestinal organoid co-cultures with immune cells

In addition to stromal cells, immune cells play a crucial role in maintaining homeostasis by protecting against pathogens, regulating inflammation and supporting epithelial barrier function. As key components of the intestinal mesenchyme, immune cells are activated in response to injury and infection, playing a crucial role in protecting and repairing the epithelium. Understanding the interaction between immune and epithelial cells is critical for identifying novel therapeutic targets, particularly in the context of anti-inflammatory therapies. Disruption in this communication can have significant pathophysiological consequences, emphasising the importance of studying these interactions in disease.

A common approach to modelling inflammation as seen in diseases like IBD involves supplementing the organoid medium with one or a cocktail of pro-inflammatory molecules. Several studies on mouse and human intestinal organoids identified that adding tumour necrosis factor alpha (TNFα), interleukin-1 beta (IL-1β), lipopolysaccharide (LPS), Flagellin and interferon-gamma (INF-γ), either individually or in combination, induces an inflammatory response that mimics certain aspects of IBD [67,68,69,70] (Fig. 1). While this approach allows for direct study of specific cytokines’ effects on organoids, it lacks the complexity provided by co-culturing methods.

Establishing immune cell-human organoid co-cultures is challenging; however, several methods have been developed using various immune cells such as T-lymphocytes, innate lymphoid cells, macrophages, neutrophils, dendritic cells and intraepithelial lymphocytes [26, 71,72,73,74]. Depending on the aim of the experiment, co-cultures can be allogeneic or autologous, where donors of immune and epithelial cells are different or the same, respectively. For translational purposes, researchers rely on autologous immune cells, obtained from different sources. Peripheral blood mononuclear cells (PBMCs) are commonly employed due to their accessibility. Tumour immunology can also be modelled with co-culture of immune cells and organoids derived from cancer patients [22, 75]. While immune cells, such as T-lymphocytes, can be isolated from biopsy samples [76], this method presents challenges, necessitating collection of multiple samples from the same donor to ensure enough material is generated and demanding more specialised skills. Co-culture protocols involve the pairing of immune cells with either 3D organoids [21], which preserve the key features of the original tissue, or alternatively, with organoid-derived monolayers (Fig. 1) [74]. The choice of co-culturing method depends on the experimental goals. For instance, co-culturing immune cells with 3D organoids is ideal when the focus is on understanding how immune cells influence the spatial organisation and behaviour of epithelial cells in a more in vivo-like environment. For example, the integration of T-cells into the epithelial cell layer was observed in 3D organoids alongside changes in T-cell morphology and epithelial gene expression [77]. Alternatively, co-culturing immune and epithelial cells as monolayers using a transwell system separates luminal and basal compartments, while establishing the natural polarity of epithelium. This configuration is particularly useful for studying concurrently host–pathogen interactions, immune responses to enteric pathogens and immune-epithelial communication, as it enables the examination of both apical and basolateral signalling. For example, human macrophages seeded on the basal side of organoid-derived epithelial monolayers have been observed to generate projections that stretch through the epithelial layer when bacteria were seeded on the apical surface [78]. The ability to segregate these compartments enhances the ability to measure secreted factors and observe interactions on the basal and apical sides of the epithelium that represent in vivo tissue dynamics.

When considering immune-epithelial cell co-culture, all cell types require specific culture medium. This makes it often difficult to find a balanced combination of factors that allows survival of all populations within the same experiment. For instance, co-culturing T-cells with epithelial cells is feasible but challenging. Activation of T-cells requires supplementing the co-culture medium with interlukin-2 (IL-2) [21], which, while necessary for T-cell function, it promotes epithelial cell maturation and differentiation [79]. This shift in cell composition in organoids can complicate studies targeting stem cell function. Another significant challenge is the use of foetal bovine serum (FBS). To minimise the impact of unknown FBS components on epithelial cells, organoids should ideally be cultured in media with low FBS (< 1%) or in FBS-free conditions [80]. However, immune cells typically require FBS to prevent stress responses like autophagy, which can impair their cytokine production [81]. Despite these challenges, ongoing advancements in co-culture techniques, such as the development of more refined media formulations, and the potential for machine learning to guide selection of growth conditions [82] offer promising solutions. These innovations may allow for precise control over the cellular microenvironment, reducing unwanted influences on both epithelial and immune cell behaviour.

Utilising organoids to assess epithelial-microbe interactions

In addition to the internal intestinal microenvironment, defined by the intricate interactions amongst immune, stromal and epithelial cells, understanding how the external microenvironment influences epithelial cells is essential for promoting tissue regeneration and maintaining homeostasis. Growing evidence suggests that elements of the human microbiome play a significant role in either supporting a healthy epithelial barrier or exacerbating intestinal diseases including IBD [83]. Most of this evidence has been correlative, but with recent breakthroughs in the capability to culture a diversity of microbes from the human intestinal tract [84], functional studies are now possible. To gain mechanistic insight into how enteric microbiota, both commensal and pathogenic, interact with epithelial cells, protocols have been developed to culture human intestinal organoids with microbes [85]. For instance, Lamers et al. highlighted the potential of using human intestinal organoids as an infection model for studying the SARS-CoV-2 virus [86]. To accurately replicate the interactions and infection pathways between epithelial cells and microbes, selecting the appropriate co-culture model is essential. Organoids typically self-organise such that the apical surface is located on the inside representing the lumen. This requires microinjection of microbes into the lumen or strategies to “flip” cell polarity [87, 88], so the apical surface is on the outside to accurately mimic epithelial-microbe interactions. There are also co-culture protocols that utilise organoids plated in 2D in transwells to facilitate analysis of host-microbe interactions. These approaches are summarised in Fig. 2 and are discussed in more detail below.

Fig. 2
figure 2

Current models of microbe-epithelial interactions. Bacteria and toxins can be introduced to organoids using different approaches depending on the type of interaction being investigated. This includes the use of 3D models, microinjection, apical-out organoids, organoids grown as a 2D monolayer and microfluidic platforms

  1. (i)

    Studying metabolites and toxins.

Microbes that reside within the GI tract can modulate epithelial tissue directly or through the secretion of metabolites and toxins. The complexity of these interactions can be difficult to decipher but the specific impact of individual factors can be assessed by direct addition to organoids. Metabolites such as short chain fatty acids and bile acids can modulate the proliferation of intestinal stem cells [89], and the activity and receptor specificity of bacterial toxins can be experimentally tested in organoids [90].

  1. (i)

    Microinjection.

Microbes may be injected directly into the lumen if maintenance of the structural integrity of the intestinal organoid is required. This is typically performed under a microscope via a specialised needle that can puncture individual organoids and dispense drugs or bacteria [85]. To facilitate the ease of injection and visualisation of the dispense of bacteria, the organoids are usually first induced into a cystic shape by promoting stem cell proliferation. Cystic organoids have a relatively larger lumen, a thinner epithelial layer and less luminal debris, all of which are crucial to enable injection efficiency. Similar to other co-culture models, dedicated media conditions must be established beforehand to fulfil the needs of both microbial and epithelial cell growth. Optimising the multiplicity of infection is also essential to prevent bacterial overgrowth within the lumen [85]. Furthermore, antibiotics may be added to the medium to restrict bacterial growth external to the organoids [91]. This methodology has been utilised by several groups to study infection of Listeria monocytogenes in mouse small intestinal organoids [92]; Helicobacter pylori in mouse and human gastric organoids [93, 94]; Salmonella enterica serovar Typhimurium (S. Typhimurium) in mouse small intestinal organoids [95] and human iPSC-derived intestinal organoids [96]; and Cryptosporidium parvum oocytes in ASC-derived intestinal organoids [97], amongst others. One group successfully transplanted two different bacteria (Escherichia coli and Yersinia pseudotuberculosis, each fluorescently labelled red and green, respectively) into a single organoid and visualised both within the same lumen [91]. This was subsequently repeated with whole faecal-derived microbiota, highlighting the scalability of this method depending on the desired output [91]. In addition to infection, microinjection has been used to measure barrier permeability by quantifying the amount of fluorescently labelled dextran that is transferred from the lumen of the organoid in a given amount of time [98]. In contrast to other culturing methods such as 2D organoid culture in which bacteria are added to the medium, microinjecting directly into the lumen permits researchers to control the amount of bacteria within each organoid. Another advantage of microinjection as a methodology to study epithelial-microbe interactions is the hypoxic environment within an organoid lumen which supports the growth of anaerobic bacteria and is thus more biologically relevant [91]. Despite these benefits, organoid microinjection is technically challenging, requires advanced equipment and is not amendable to high-throughput experimentation as only one organoid can be microinjected at one time [54].

  1. (i)

    Apical-out organoid.

In addition to microinjection, several groups have recently reversed the epithelial polarity within organoid models so that the apical surface is facing outwards and is in contact with the medium [54, 99, 100]. As epithelial polarity is largely regulated via extracellular membrane (ECM) proteins within the basement membrane in vivo or Matrigel in culture, intestinal organoid polarity is rapidly reversed to an apical-out state if transferred to a suspension medium lacking any ECM components. Supplementation of basement membrane extract to these apical-out organoids in suspension restored the basal-out phenotype in a dose-dependent manner [54, 99]. Importantly, Co et al. determined that these apical-out organoids retained key features of basal-out organoids including intact barrier integrity, cellular differentiation and selective diffusion [54, 99]. Furthermore, when co-cultured with the pathogenic bacteria S. Typhimurium, the bacterium preferentially bound to the apical surface (now accessible) and compromised barrier function of the organoids [54, 99]. These results highlight the potential of apical-out organoids in studying the relationship between the intestinal epithelium and pathogenic bacteria that act through the apical surface. Although these apical-out organoids maintain a 3D structure and do not require the microinjection of microbes into the lumen, they are hindered by their short life span in the reversed state, the propensity to aggregate and the inability to change the medium while in suspension [101].

(iv) 2D monolayer organoids.

Although a key benefit of organoid models is the ability to exist in three dimensions and thus recapitulate a physiologically relevant environment, for certain research questions, a 2D model may be more appropriate. This can be achieved by seeding intestinal crypts on a thin layer of dilute Matrigel [87, 88], or more commonly, directly on transwell cell culture inserts [102,103,104,105,106]. The resulting monolayer maintains key features of 3D organoid models including epithelial polarisation [88, 102, 103], proliferation and self-renewal [88, 105], cellular heterogeneity [88, 102,103,104,105,106], barrier integrity [88, 102, 103, 105, 106] and a pathophysiological response to various stimuli [102, 103, 106]. For instance, Ettayebi et al. reported the first in vitro culture of Norovirus in human epithelial cells using 2D monolayer organoids [107], which could not be achieved in transformed cell lines [108]. Roodsant et al. infected human monolayer organoids with a pathogenic virus and bacterium, Enterovirus A71 (EV-A71) and Listeria monocytogenes, respectively, to demonstrate their effectiveness in studying epithelial-microbe interactions [102]. Both virus and bacterium demonstrated the ability to translocate from the apical to the basal side of the epithelial monolayer, with EV-A71 disrupting barrier function and L. monocytogenes inducing inflammation in the process [102]. This was similarly observed by Holthaus et al. who co-infected mouse monolayer organoids with two parasitic protozoans, Toxoplasma gondii and Giardia duodenalis, with a marked loss of barrier integrity observed only in the presence of G. duodenalis [103]. This highlights the capacity of 2D organoid cultures to easily facilitate infection with a variety of microbes and organisms, and thus reinforces this method as a possible alternative to 3D organoid models to study disease with a strong microbial component such as IBD. However, despite their advantages, 2D organoids still present challenges for long-term culture. Prolonged culture results in low oxygen tension, which leads to cellular stress and altered secretory cell differentiation [101, 109]. To address this, an air–liquid interface (ALI) method using a transwell system can alleviate oxygen stress by exposing only the basolateral side of the cells to the culture medium while keeping the apical side exposed to air [109, 110]. This approach promotes a higher degree of differentiation and produces long-lived models ideal for studying chronic infections [111,112,113]. For example, Boccellato et al. successfully used an ALI co-culture system to study H. pylori infection for up to 4 weeks [113]. Another challenge in co-culturing systems is the selection of a suitable growth medium, as different cell types and microbes often require specialised conditions for growth. However, transwell systems, which separate the apical and basal compartments, allow for the optimisation of culture conditions for both bacteria and epithelial cells, creating a microenvironment that better mimics in vivo conditions [114]. With recent advances in microfluidic technology, more physiologically relevant conditions, such as oxygen gradient, pH level and specific growth factors, can be better recapitulated.

Advances in bioengineering organoids

Despite these advances in culture conditions to improve the physiological relevance of organoid models, they still lack some fundamental features of the in vivo environment due to their growth as spherical structures. This includes the tissue geometry and physical forces, fluid flow across the luminal surface and variations in oxygen concentration. These forces are important for the patterning of tissue and measuring bacterial interactions [20, 115]. To address this, intestine-on-a-chip model systems have been generated in which intestinal epithelial cells (IECs) or whole organoids are seeded on a microfluidic platform to which endothelial cells, immune cells and the patient’s microbiome may be added [116, 117] (Fig. 2). Typically, the microfluidic platform is composed of two channels between which the IECs are seeded in a polarised manner so that one channel is representative of the lumen and the other of the basolateral surface [118]. Therefore, unlike in organoid models, the apical side is easily accessible in intestine-on-a-chip model systems for administration of bacteria, cytokines and drugs in a physiologically relevant manner [119]. Peristaltic-like movement can be artificially induced in intestine-on-a-chip models via the addition of a vacuum, while the flow of medium over the cells more accurately reflects the in vivo intestinal environment by providing similar mechanical cues and allows for removal of waste products produced by the IECs [116, 117]. Furthermore, by controlling the oxygen content of the supplied medium, an artificial oxygen gradient may also be established to more accurately reflect the in vivo intestine in which varying oxygen gradients are essential for nutrient absorption and maintenance of a complex microbiota [117, 118].

Due to their complexity, intestine-on-a-chip models are limited by their physical and financial demands and are thus only suitable for more nuanced questions that require the presence of the complete microenvironment, mechanical stress from peristalsis or a controllable oxygen gradient while still being accessible in vitro. These advantages of intestine-on-a-chip models make them highly valuable for researching complex, multifactorial diseases like IBD, but perhaps are limiting when more direct interactions or high-throughput screening approaches are being investigated. Intestine-on-a-chip models are already being utilised to answer a broad range of questions relating to intestinal biology and disease. For example, Liu et al. established a controllable oxygen gradient across a microfluidic platform to successfully co-culture the highly oxygen sensitive anaerobic bacterium Bifidobacterium bifidum with aerobic human intestinal epithelial cells (Caco-2) [120]. Beaurivage and colleagues were also successful in generating a microfluidic intestine-on-a-chip culture from Caco-2 cells which were likewise stimulated with inflammation-inducing cytokines to model IBD [121]. Within 4 days of establishment, the Caco-2 cells had already formed an effective barrier that was subsequently lost after treatment with inflammatory cytokines [121]. In the following year, Beaurivage and colleagues further optimised this methodology by using PDOs to more accurately model the heterogenous intestinal epithelium and introduced monocyte-derived macrophages which were induced to produce cytokines via addition of LPS and IFN-γ [119]. Induction of an inflamed state was subsequently assessed via RNA-sequencing which confirmed upregulation of genes relating to the innate immune response in microfluidic cultures treated with LPS and IFN-γ [119]. Kim et al. cultured pathogenic and non-pathogenic E. coli on an intestine-on-a-chip system [122]. From this, Kim et al. observed that non-pathogenic E. coli had no detectable effect on the intestinal epithelium, while pathogenic E. coli induced loss of barrier integrity and tissue architecture in the intestine-on-a-chip models, a result not detected with LPS endotoxin alone [122]. This paper highlights the importance of external factors (i.e. the gut microbiota and the immune system) when modelling infection or features of IBD, and thus intestine-on-a-chip systems provide an alternative approach to studying intestinal disease and facilitating and drug discovery when compared to strictly epithelial systems such as organoid models.

Furthermore, these intestine-on-a-chip systems may be optimised by combining the technology with 3D bioprinting [123]. 3D bioprinting involves the deposition of a 3D scaffold (using biomaterial inks) onto which heterogenous cell cultures are seeded [124,125,126] or alternatively, cells may be directly 3D printed (using bioinks) to generate complex microenvironments in vitro [125]. Recent work on 3D printed intestinal tissue arranged in either a polarised monolayer with protrusions [127,128,129] or in a cylindrical tube [130, 131] has highlighted the effectiveness of establishing and maintaining a selectively permeable barrier with this methodology [101, 127,128,129, 132], the potential for co-culture with non-epithelial cells or bacteria/viruses [128, 129, 131] and the possibility to replicate a diseased state [132, 133]. Analysis of data in these systems also benefits from utilising AI.

Ultimately, the goal of bioprinting is to generate whole organs in vitro for research or transplantation purposes; however, this is currently limited by its resolution capacity to generate small structures such as capillaries [126]. Until this is overcome, bioprinting can be used instead to help generate intestine-on-a-chip systems and enhance reproducibility. Thus, these next generation engineered models incorporate spatial and mechanical cues to establish human systems that mimic human mucosa even more closely, and make the complex architecture of the in vivo intestine more accessible in vitro for the study of multi-cellular diseases such as IBD [134]. This includes reproducing crypt structure, morphogen gradients, luminal flow and cell turnover. These bioengineered systems can model tumour initiation and disease states [135].

Utilising high-throughput organoid screening to facilitate personalised medicine

A valuable feature of PDOs is the ability to perform chemical screening to identify potential therapeutics across a broad range of patients. To this extent, human organoid models have an advantage over cell lines and animal models as they more readily recapitulate human genetic diversity. One of the first drug screens performed on an organoid biobank was performed by van de Wetering and colleagues using patient-derived colorectal tumour (n = 22) and normal organoids (n = 19) [136] where they successfully tested 83 compounds with varying degrees of sensitivity utilising the CellTitre-Glo (CTG) cell viability assay. More recently, Luo et al. identified four potential hits out of a total of 139 compounds which reduced the viability of high-risk colorectal adenoma organoids [137]. Phenotyping approaches to analyse impacts of drugs is summarised in Fig. 3. Alternatively, with more specific questions, a single compound may be tested on a larger cohort of organoids to determine the effect of a single drug of interest across a range of genetic backgrounds. For example, Nishimura and colleagues have utilised human normal colon organoids (supplemented with cytokines and bacteria to replicate IBD) to investigate the effect of the novel drug, KAG-308, in relation to IBD [138]. Using this model system, the authors found that KAG-308 supressed many of the inflammation-associated genes and instead supported the intestinal stem cell population as observed by the increase in proliferation and organoid establishment after treatment with KAG-308.

Fig. 3
figure 3

Current screening methods for quantifying organoid growth and morphology. Phenotypic changes in organoids over time or in response to specific treatments can be quantified by assessing growth, morphology, size and viability. These changes can be visualised using brightfield imaging of live/fixed organoids or by labelling live/fixed organoids with different cell markers or stains, depending on the experimental objective. High-content imaging systems (e.g. ImageXpress Pico Automated Cell Imaging System) enable data acquisition, with analysis performed using software such as ImageJ. A Growth, measured by perimeter, can be plotted to show changes over time. In this example, growth of mouse small intestinal (SI) organoids was measured at day 2 compared to day 5, and human SI organoids at day 5 compared to day 9 (paired t-test). B Morphology can be assessed through circularity which determines the degree of differentiation. In this example, morphological heterogeneity of mouse SI organoids is observed within the same well. Organoids exhibiting a cystic morphology are less differentiated (value closer to 1), whereas non-cystic organoids are more differentiated (value closer to 0) (paired t-test). C Organoid size can be measured using brightfield imaging or by labelling organoids with a fluorescent marker for better segmentation analysis to enable the evaluation of size changes in response to specific treatments. In this example, individual human colorectal cancer organoids have been masked based on their Calcein AM signal and the resulting area quantified (expressed as fold change from the untreated control, one-way ANOVA). D Viability analysis is performed to assess the proportion of live vs dead cells in organoids under different treatment conditions. Cell death can be measured by using different cell death stains such as ethidium homodimer III (EthD-III). In this example, the number of dead cells in colorectal cancer organoids upon addition of chemotherapy drug has been plotted to evaluate treatment impact on survival (unpaired t-test)

Metabolism-based assays such as CTG are common outputs of organoid drug screens; however, drug response in organoid models can additionally be measured through image-based phenotypic screens which quantify the effect of a drug based on a visual change in the organoids (Fig. 3) [139, 140]. Typically, image-based phenotypic screens use fluorescent markers to measure organoid size as a representation of cell viability which has been confirmed to be comparable to a CTG assay [140]. However, alternative fluorescent markers may be chosen depending on the desired read-out of the assay, e.g. live/dead cells, proliferative cells, cell type-specific markers or a combination of several markers for multiplex imaging, and thus image-based phenotypic screens can provide a more complex picture of the effect of candidate drugs on organoid models [139] (Fig. 3). Furthermore, if combined with the emerging advances in machine learning that can both integrate multi-omic data and predict drug response, this process could become even more informative and streamlined in the future [141, 142] (Fig. 4).

Fig. 4
figure 4

Overview of advancing methodologies to model IBD. Different approaches to model IBD include traditional or co-culture organoid systems, 2D organoid culture and intestine-on-a-chip. Currently, these models serve as a tool for more accurate personalised medicine, next generation imaging and high-throughput drug screening, while advancements in AI and machine learning could further enhance the quantity and quality of data generated from these models

Conclusions

Future perspectives

Since the initial discovery of conditions that support the growth and self-organisation of native intestinal tissue, there have been significant advances in both the range and complexity of systems to analyse the function of the human gut. As highlighted in Fig. 4, these innovative models have enormous potential for translational applications including the discovery of new drug and bacterial treatments for IBD and screening for effective treatments for pathological infections such as Clostridium difficile. The ability to establish organoids efficiently from individual patients, advances in high-throughput screening, AI and machine learning paves the way for personalised medicine approaches to improve effective patient treatment.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

2D:

Two dimensional

3D:

Three dimensional

AI:

Artificial intelligence

ALI:

Air-liquid interface

ASC:

Adult stem cell

BEST4:

Bestrophin 4

CTG:

CellTitre-Glo

ECM:

Extracellular matrix

EGF:

Epidermal growth factor

EthD-III:

Ethidium homodimer III

EV-A71:

Enterovirus A71

FBS:

Foetal bovine serum

FGF2:

Fibroblast growth factor 2

IBD:

Inflammatory bowel disease

IEC:

Intestinal epithelial cell

IGF1:

Insulin-like growth factor 1

IL-1β:

Interleukin-1 beta

IL-2:

Interleukin-2

INF-γ:

Interferon-gamma

iPSC:

Induced pluripotent stem cell

LPS:

Lipopolysaccharide

PBMC:

Peripheral blood mononuclear cell

PDGFRα:

Platelet-derived growth factor receptor alpha

PDO:

Patient-derived organoid

S. Typhimurium:

Salmonella enterica Serovar Typhimurium

SI:

Small intestine

TA:

Transit-amplifying

TNFα:

Tumour necrosis factor alpha

References

  1. Huycke TR, Hakkinen TJ, Miyazaki H, Srivastava V, Barruet E, McGinnis CS, et al. Patterning and folding of intestinal villi by active mesenchymal dewetting. Cell. 2024;187(12):3072–89.e20.

    Article  PubMed  Google Scholar 

  2. Hansen SL, Larsen HL, Pikkupeura LM, Maciag G, Guiu J, Muller I, et al. An organoid-based CRISPR-Cas9 screen for regulators of intestinal epithelial maturation and cell fate. Sci Adv. 2023;9(28):eadg4055.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Guiu J, Jensen KB. From definitive endoderm to gut-a process of growth and maturation. Stem Cells Dev. 2015;24(17):1972–83.

    Article  PubMed  Google Scholar 

  4. Zwick RK, Kasparek P, Palikuqi B, Viragova S, Weichselbaum L, McGinnis CS, et al. Epithelial zonation along the mouse and human small intestine defines five discrete metabolic domains. Nat Cell Biol. 2024;26(2):250–62.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449(7165):1003–7.

    Article  PubMed  Google Scholar 

  6. Hageman JH, Heinz MC, Kretzschmar K, van der Vaart J, Clevers H, Snippert HJG. Intestinal regeneration: regulation by the microenvironment. Dev Cell. 2020;54(4):435–46.

    Article  PubMed  Google Scholar 

  7. Jarde T, Chan WH, Rossello FJ, Kaur Kahlon T, Theocharous M, Kurian Arackal T, et al. Mesenchymal niche-derived neuregulin-1 drives intestinal stem cell proliferation and regeneration of damaged epithelium. Cell Stem Cell. 2020;27(4):646-62 e7.

    Article  PubMed  Google Scholar 

  8. Yu YR, Rodriguez JR. Clinical presentation of Crohn’s, ulcerative colitis, and indeterminate colitis: symptoms, extraintestinal manifestations, and disease phenotypes. Semin Pediatr Surg. 2017;26(6):349–55.

    Article  PubMed  Google Scholar 

  9. Dhaliwal J, Walters TD, Mack DR, Huynh HQ, Jacobson K, Otley AR, et al. Phenotypic variation in paediatric inflammatory bowel disease by age: a multicentre prospective inception cohort study of the Canadian children IBD network. J Crohns Colitis. 2020;14(4):445–54.

    Article  PubMed  Google Scholar 

  10. Langerholc T, Maragkoudakis PA, Wollgast J, Gradisnik L, Cencic A. Novel and established intestinal cell line models - an indispensable tool in food science and nutrition. Trends Food Sci Technol. 2011;22:S11–20.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Zhou A, Yuan Y, Yang M, Huang Y, Li X, Li S, et al. Crosstalk between the gut microbiota and epithelial cells under physiological and infectious conditions. Front Cell Infect Microbiol. 2022;12: 832672.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141(5):1762–72.

    Article  PubMed  Google Scholar 

  13. Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459(7244):262–5.

    Article  PubMed  Google Scholar 

  14. Serra D, Mayr U, Boni A, Lukonin I, Rempfler M, Challet Meylan L, et al. Self-organization and symmetry breaking in intestinal organoid development. Nature. 2019;569(7754):66–72.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Kraiczy J, Nayak KM, Howell KJ, Ross A, Forbester J, Salvestrini C, et al. DNA methylation defines regional identity of human intestinal epithelial organoids and undergoes dynamic changes during development. Gut. 2019;68(1):49–61.

    Article  PubMed  Google Scholar 

  16. Fujii M, Matano M, Toshimitsu K, Takano A, Mikami Y, Nishikori S, et al. Human intestinal organoids maintain self-renewal capacity and cellular diversity in niche-inspired culture condition. Cell Stem Cell. 2018;23(6):787-93 e6.

    Article  PubMed  Google Scholar 

  17. Oost KC, Kahnwald M, Barbiero S, de Medeiros G, Suppinger S, Kalck V, et al. Dynamics and plasticity of stem cells in the regenerating human colonic epithelium. bioRxiv. 2023.12.18.572103.

  18. Abud HE, Amarasinghe SL, Micati D, Jarde T. Stromal niche signals that orchestrate intestinal regeneration. Cell Mol Gastroenterol Hepatol. 2024;17(5):679–85.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Nikolaev M, Mitrofanova O, Broguiere N, Geraldo S, Dutta D, Tabata Y, et al. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature. 2020;585(7826):574–8.

    Article  PubMed  Google Scholar 

  20. Gjorevski N, Nikolaev M, Brown TE, Mitrofanova O, Brandenberg N, DelRio FW, et al. Tissue geometry drives deterministic organoid patterning. Science. 2022;375(6576):eaaw9021.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Takashima S, Martin ML, Jansen SA, Fu Y, Bos J, Chandra D, et al. T cell-derived interferon-gamma programs stem cell death in immune-mediated intestinal damage. Sci Immunol. 2019;4(42):eaay8556.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Dekkers JF, Alieva M, Cleven A, Keramati F, Wezenaar AKL, van Vliet EJ, et al. Uncovering the mode of action of engineered T cells in patient cancer organoids. Nat Biotechnol. 2023;41(1):60–9.

    Article  PubMed  Google Scholar 

  23. Farin HF, Van Es JH, Clevers H. Redundant sources of Wnt regulate intestinal stem cells and promote formation of Paneth cells. Gastroenterology. 2012;143(6):1518-29 e7.

    Article  PubMed  Google Scholar 

  24. Greicius G, Kabiri Z, Sigmundsson K, Liang C, Bunte R, Singh MK, et al. PDGFRalpha(+) pericryptal stromal cells are the critical source of Wnts and RSPO3 for murine intestinal stem cells in vivo. Proc Natl Acad Sci U S A. 2018;115(14):E3173–81.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Kabiri Z, Greicius G, Madan B, Biechele S, Zhong Z, Zaribafzadeh H, et al. Stroma provides an intestinal stem cell niche in the absence of epithelial Wnts. Development. 2014;141(11):2206–15.

    Article  PubMed  Google Scholar 

  26. Staab JF, Lemme-Dumit JM, Latanich R, Pasetti MF, Zachos NC. Co-culture system of human enteroids/colonoids with innate immune cells. Curr Protoc Immunol. 2020;131(1): e113.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, et al. Organoid modeling of the tumor immune microenvironment. Cell. 2018;175(7):1972-88 e16.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Niec RE, Chu T, Schernthanner M, Gur-Cohen S, Hidalgo L, Pasolli HA, et al. Lymphatics act as a signaling hub to regulate intestinal stem cell activity. Cell Stem Cell. 2022;29(7):1067-82 e18.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Goto N, Goto S, Imada S, Hosseini S, Deshpande V, Yilmaz OH. Lymphatics and fibroblasts support intestinal stem cells in homeostasis and injury. Cell Stem Cell. 2022;29(8):1246-61 e6.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Kraiczy J, McCarthy N, Malagola E, Tie G, Madha S, Boffelli D, et al. Graded BMP signaling within intestinal crypt architecture directs self-organization of the Wnt-secreting stem cell niche. Cell Stem Cell. 2023;30(4):433-49 e8.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Clevers H. The intestinal crypt, a prototype stem cell compartment. Cell. 2013;154(2):274–84.

    Article  PubMed  Google Scholar 

  32. Barker N, van Oudenaarden A, Clevers H. Identifying the stem cell of the intestinal crypt: strategies and pitfalls. Cell Stem Cell. 2012;11(4):452–60.

    Article  PubMed  Google Scholar 

  33. Bowcutt R, Forman R, Glymenaki M, Carding SR, Else KJ, Cruickshank SM. Heterogeneity across the murine small and large intestine. World J Gastroenterol. 2014;20(41):15216–32.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Podolsky DK, Fournier DA, Lynch KE. Human colonic goblet cells. Demonstration of distinct subpopulations defined by mucin-specific monoclonal antibodies. J Clin Invest. 1986;77(4):1263–71.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Atuma C, Strugala V, Allen A, Holm L. The adherent gastrointestinal mucus gel layer: thickness and physical state in vivo. Am J Physiol Gastrointest Liver Physiol. 2001;280(5):G922–9.

    Article  PubMed  Google Scholar 

  36. Ito G, Okamoto R, Murano T, Shimizu H, Fujii S, Nakata T, et al. Lineage-specific expression of bestrophin-2 and bestrophin-4 in human intestinal epithelial cells. PLoS One. 2013;8(11): e79693.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Parikh K, Antanaviciute A, Fawkner-Corbett D, Jagielowicz M, Aulicino A, Lagerholm C, et al. Colonic epithelial cell diversity in health and inflammatory bowel disease. Nature. 2019;567(7746):49–55.

    Article  PubMed  Google Scholar 

  38. Elmentaite R, Ross ADB, Roberts K, James KR, Ortmann D, Gomes T, et al. Single-cell sequencing of developing human gut reveals transcriptional links to childhood Crohn’s disease. Dev Cell. 2020;55(6):771-83 e5.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Moor AE, Harnik Y, Ben-Moshe S, Massasa EE, Rozenberg M, Eilam R, et al. Spatial reconstruction of single enterocytes uncovers broad zonation along the intestinal villus axis. Cell. 2018;175(4):1156-67 e15.

    Article  PubMed  Google Scholar 

  40. Stevens CE, Leblond CP. Rate of renewal of the cells of the intestinal epithelium in the rat. Anat Rec. 1947;97(3):373.

    PubMed  Google Scholar 

  41. Holloway EM, Czerwinski M, Tsai YH, Wu JH, Wu A, Childs CJ, et al. Mapping development of the human intestinal niche at single-cell resolution. Cell Stem Cell. 2021;28(3):568-80 e4.

    Article  PubMed  Google Scholar 

  42. Childs CJ, Holloway EM, Sweet CW, Tsai YH, Wu A, Vallie A, et al. EPIREGULIN creates a developmental niche for spatially organized human intestinal enteroids. JCI Insight. 2023;8(6):e165566.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Fujii M, Matano M, Toshimitsu K, Takano A, Mikami Y, Nishikori S, et al. Human intestinal organoids maintain self-renewal capacity and cellular diversity in niche-inspired culture condition. Cell Stem Cell. 2018;23(6):787-93.e6.

    Article  PubMed  Google Scholar 

  44. Bian X, Li G, Wang C, Liu W, Lin X, Chen Z, et al. A deep learning model for detection and tracking in high-throughput images of organoid. Comput Biol Med. 2021;134: 104490.

    Article  PubMed  Google Scholar 

  45. Matthews JM, Schuster B, Kashaf SS, Liu P, Ben-Yishay R, Ishay-Ronen D, et al. OrganoID: a versatile deep learning platform for tracking and analysis of single-organoid dynamics. PLoS Comput Biol. 2022;18(11): e1010584.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Abdul L, Xu J, Sotra A, Chaudary A, Gao J, Rajasekar S, et al. D-CryptO: deep learning-based analysis of colon organoid morphology from brightfield images. Lab Chip. 2022;22(21):4118–28.

    Article  PubMed  Google Scholar 

  47. Gritti N, Lim JL, Anlas K, Pandya M, Aalderink G, Martinez-Ara G, et al. MOrgAna: accessible quantitative analysis of organoids with machine learning. Development. 2021;148(18):dev199611.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Park CS, Nguyen LP, Yong D. Development of colonic organoids containing enteric nerves or blood vessels from human embryonic stem cells. Cells. 2020;9(10):2209.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Workman MJ, Mahe MM, Trisno S, Poling HM, Watson CL, Sundaram N, et al. Engineered human pluripotent-stem-cell-derived intestinal tissues with a functional enteric nervous system. Nat Med. 2017;23(1):49–59.

    Article  PubMed  Google Scholar 

  50. Kasendra M, Tovaglieri A, Sontheimer-Phelps A, Jalili-Firoozinezhad S, Bein A, Chalkiadaki A, et al. Development of a primary human small intestine-on-a-chip using biopsy-derived organoids. Sci Rep. 2018;8(1):2871.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Takahashi Y, Sato S, Kurashima Y, Lai CY, Otsu M, Hayashi M, et al. Reciprocal inflammatory signaling between intestinal epithelial cells and adipocytes in the absence of immune cells. EBioMedicine. 2017;23:34–45.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Mason J, Ohlund D. Key aspects for conception and construction of co-culture models of tumor-stroma interactions. Front Bioeng Biotechnol. 2023;11: 1150764.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Rasouli M, Safari F. Principles of indirect co-culture method using transwell. Methods Mol Biol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/7651_2024_537. Epub ahead of print.

  54. Co JY, Margalef-Català M, Li X, Mah AT, Kuo CJ, Monack DM, et al. Controlling epithelial polarity: a human enteroid model for host-pathogen interactions. Cell Rep. 2019;26(9):2509-20.e4.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Kim S, Min S, Choi YS, Jo SH, Jung JH, Han K, et al. Tissue extracellular matrix hydrogels as alternatives to Matrigel for culturing gastrointestinal organoids. Nat Commun. 2022;13(1):1692.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Takahashi Y, Inoue Y, Sato S, Okabe T, Kojima H, Kiyono H, et al. Drug cytotoxicity screening using human intestinal organoids propagated with extensive cost-reduction strategies. Sci Rep. 2023;13(1):5407.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Kozlowski MT, Crook CJ, Ku HT. Towards organoid culture without Matrigel. Commun Biol. 2021;4(1):1387.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Luo X, Fong ELS, Zhu C, Lin QXX, Xiong M, Li A, et al. Hydrogel-based colorectal cancer organoid co-culture models. Acta Biomater. 2021;132:461–72.

    Article  PubMed  Google Scholar 

  59. McCarthy N, Kraiczy J, Shivdasani RA. Cellular and molecular architecture of the intestinal stem cell niche. Nat Cell Biol. 2020;22(9):1033–41.

    Article  PubMed  Google Scholar 

  60. Shoshkes-Carmel M, Wang YJ, Wangensteen KJ, Toth B, Kondo A, Massasa EE, et al. Subepithelial telocytes are an important source of Wnts that supports intestinal crypts. Nature. 2018;557(7704):242–6.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Lemmetyinen TT, Viitala EW, Wartiovaara L, Kaprio T, Hagstrom J, Haglund C, et al. Fibroblast-derived EGF ligand neuregulin 1 induces fetal-like reprogramming of the intestinal epithelium without supporting tumorigenic growth. Dis Model Mech. 2023;16(4):dmm049692.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Karpus ON, Westendorp BF, Vermeulen JLM, Meisner S, Koster J, Muncan V, et al. Colonic CD90+ crypt fibroblasts secrete semaphorins to support epithelial growth. Cell Rep. 2019;26(13):3698-708 e5.

    Article  PubMed  Google Scholar 

  63. Wallisch S, Neef SK, Denzinger L, Monch D, Koch J, Marzi J, et al. Protocol for establishing a coculture with fibroblasts and colorectal cancer organoids. STAR Protoc. 2023;4(3): 102481.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Hautefort I, Poletti M, Papp D, Korcsmaros T. Everything you always wanted to know about organoid-based models (and never dared to ask). Cell Mol Gastroenterol Hepatol. 2022;14(2):311–31.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Lahar N, Lei NY, Wang J, Jabaji Z, Tung SC, Joshi V, et al. Intestinal subepithelial myofibroblasts support in vitro and in vivo growth of human small intestinal epithelium. PLoS One. 2011;6(11): e26898.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Lin M, Hartl K, Heuberger J, Beccaceci G, Berger H, Li H, et al. Establishment of gastrointestinal assembloids to study the interplay between epithelial crypts and their mesenchymal niche. Nat Commun. 2023;14(1):3025.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Arnauts K, Verstockt B, Ramalho AS, Vermeire S, Verfaillie C, Ferrante M. Ex vivo mimicking of inflammation in organoids derived from patients with ulcerative colitis. Gastroenterology. 2020;159(4):1564–7.

    Article  PubMed  Google Scholar 

  68. Hibiya S, Tsuchiya K, Hayashi R, Fukushima K, Horita N, Watanabe S, et al. Long-term inflammation transforms intestinal epithelial cells of colonic organoids. J Crohns Colitis. 2017;11(5):621–30.

    PubMed  Google Scholar 

  69. Lee C, Hong SN, Kim ER, Chang DK, Kim YH. Epithelial regeneration ability of Crohn’s disease assessed using patient-derived intestinal organoids. Int J Mol Sci. 2021;22(11):6013.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Niklinska-Schirtz BJ, Venkateswaran S, Anbazhagan M, Kolachala VL, Prince J, Dodd A, et al. Ileal derived organoids from Crohn’s disease patients show unique transcriptomic and secretomic signatures. Cell Mol Gastroenterol Hepatol. 2021;12(4):1267–80.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Rogoz A, Reis BS, Karssemeijer RA, Mucida D. A 3-D enteroid-based model to study T-cell and epithelial cell interaction. J Immunol Methods. 2015;421:89–95.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Lemme-Dumit JM, Doucet M, Zachos NC, Pasetti MF. Epithelial and neutrophil interactions and coordinated response to shigella in a human intestinal enteroid-neutrophil coculture model. mBio. 2022;13(3):e0094422.

    Article  PubMed  Google Scholar 

  73. Tsuruta S, Kawasaki T, Machida M, Iwatsuki K, Inaba A, Shibata S, et al. Development of human gut organoids with resident tissue macrophages as a model of intestinal immune responses. Cell Mol. Gastroenterol Hepatol. 2022;14(3):726-9 e5.

    Google Scholar 

  74. Jowett GM, Read E, Roberts LB, Coman D, Vila Gonzalez M, Zabinski T, et al. Organoids capture tissue-specific innate lymphoid cell development in mice and humans. Cell Rep. 2022;40(9): 111281.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Dijkstra KK, Cattaneo CM, Weeber F, Chalabi M, van de Haar J, Fanchi LF, et al. Generation of tumor-reactive T cells by co-culture of peripheral blood lymphocytes and tumor organoids. Cell. 2018;174(6):1586-98 e12.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Giles EM, Sanders TJ, McCarthy NE, Lung J, Pathak M, MacDonald TT, et al. Regulation of human intestinal T-cell responses by type 1 interferon-STAT1 signaling is disrupted in inflammatory bowel disease. Mucosal Immunol. 2017;10(1):184–93.

    Article  PubMed  Google Scholar 

  77. Recaldin T, Steinacher L, Gjeta B, Harter MF, Adam L, Kromer K, et al. Human organoids with an autologous tissue-resident immune compartment. Nature. 2024;633(8028):165–73.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Noel G, Baetz NW, Staab JF, Donowitz M, Kovbasnjuk O, Pasetti MF, et al. A primary human macrophage-enteroid co-culture model to investigate mucosal gut physiology and host-pathogen interactions. Sci Rep. 2017;7: 45270.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Jung KB, Lee H, Son YS, Lee MO, Kim YD, Oh SJ, et al. Interleukin-2 induces the in vitro maturation of human pluripotent stem cell-derived intestinal organoids. Nat Commun. 2018;9(1):3039.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Zheng X, Baker H, Hancock WS, Fawaz F, McCaman M, Pungor E Jr. Proteomic analysis for the assessment of different lots of fetal bovine serum as a raw material for cell culture. Part IV. Application of proteomics to the manufacture of biological drugs. Biotechnol Prog. 2006;22(5):1294–300.

    Article  PubMed  Google Scholar 

  81. Rahmani M, Mohammadnia-Afrouzi M, Nouri HR, Fattahi S, Akhavan-Niaki H, Mostafazadeh A. Human PBMCs fight or flight response to starvation stress: increased T-reg, FOXP3, and TGF-beta1 with decreased miR-21 and constant miR-181c levels. Biomed Pharmacother. 2018;108:1404–11.

    Article  PubMed  Google Scholar 

  82. Gjorevski N, Sachs N, Manfrin A, Giger S, Bragina ME, Ordonez-Moran P, et al. Designer matrices for intestinal stem cell and organoid culture. Nature. 2016;539(7630):560–4.

    Article  PubMed  Google Scholar 

  83. Tsolis RM, Baumler AJ. Gastrointestinal host-pathogen interaction in the age of microbiome research. Curr Opin Microbiol. 2020;53:78–89.

    Article  PubMed  Google Scholar 

  84. Browne HP, Forster SC, Anonye BO, Kumar N, Neville BA, Stares MD, et al. Culturing of ‘unculturable’ human microbiota reveals novel taxa and extensive sporulation. Nature. 2016;533(7604):543–6.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Puschhof J, Pleguezuelos-Manzano C, Martinez-Silgado A, Akkerman N, Saftien A, Boot C, et al. Intestinal organoid cocultures with microbes. Nat Protoc. 2021;16(10):4633–49.

    Article  PubMed  Google Scholar 

  86. Lamers MM, Beumer J, van der Vaart J, Knoops K, Puschhof J, Breugem TI, et al. SARS-CoV-2 productively infects human gut enterocytes. Science. 2020;369(6499):50–4.

    Article  PubMed  Google Scholar 

  87. Bvd Hee, Loonen LMP, Taverne N, Taverne-Thiele JJ, Smidt H, Wells JM. Optimized procedures for generating an enhanced, near physiological 2D culture system from porcine intestinal organoids. Stem Cell Res. 2018;28:165–71.

    Article  Google Scholar 

  88. Thorne CA, Chen IW, Sanman LE, Cobb MH, Wu LF, Altschuler SJ. Enteroid monolayers reveal an autonomous WNT and BMP circuit controlling intestinal epithelial growth and organization. Dev Cell. 2018;44(5):624-33.e4.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Yao C, Gou X, Tian C, Zhou L, Hao R, Wan L, et al. Key regulators of intestinal stem cells: diet, microbiota, and microbial metabolites. J Genet Genomics. 2023;50(10):735–46.

    Article  PubMed  Google Scholar 

  90. Mileto SJ, Jarde T, Childress KO, Jensen JL, Rogers AP, Kerr G, et al. Clostridioides difficile infection damages colonic stem cells via TcdB, impairing epithelial repair and recovery from disease. Proc Natl Acad Sci U S A. 2020;117(14):8064–73.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Williamson IA, Arnold JW, Samsa LA, Gaynor L, DiSalvo M, Cocchiaro JL, et al. A high-throughput organoid microinjection platform to study gastrointestinal microbiota and luminal physiology. Cell Mol Gastroenterol Hepatol. 2018;6(3):301–19.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Kim M, Fevre C, Lavina M, Disson O, Lecuit M. Live imaging reveals listeria hijacking of E-cadherin recycling as it crosses the intestinal barrier. Curr Biol. 2021;31(5):1037-47 e4.

    Article  PubMed  Google Scholar 

  93. Bartfeld S, Clevers H. Organoids as model for infectious diseases: culture of human and murine stomach organoids and microinjection of Helicobacter pylori. J Vis Exp. 2015;105:2015.

    Google Scholar 

  94. Palframan SL, Mahmud MT, Tan KS, Grinter R, Xin V, Dunstan RA, et al. Helicobacter pylori vacuolating cytotoxin A exploits human endosomes for intracellular activation. bioRxiv. 2022.08.22.504206

  95. Wilson SS, Tocchi A, Holly MK, Parks WC, Smith JG. A small intestinal organoid model of non-invasive enteric pathogen–epithelial cell interactions. Mucosal Immunol. 2015;8(2):352–61.

    Article  PubMed  Google Scholar 

  96. Forbester JL, Goulding D, Vallier L, Hannan N, Hale C, Pickard D, et al. Interaction of Salmonella enterica serovar typhimurium with intestinal organoids derived from human induced pluripotent stem cells. Infect Immun. 2015;83(7):2926–34.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Dutta D, Heo I, O’Connor R. Studying cryptosporidium infection in 3D tissue-derived human organoid culture systems by microinjection. J Vis Exp. 2019;151:e59610.

    Google Scholar 

  98. Hill DR, Huang S, Tsai Y-H, Spence JR, Young VB. Real-time measurement of epithelial barrier permeability in human intestinal organoids. J Vis Exp. 2017;130:1–10.

    Google Scholar 

  99. 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):5171–92.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Kakni P, López-Iglesias C, Truckenmüller R, Habibović P, Giselbrecht S. Reversing epithelial polarity in pluripotent stem cell-derived intestinal organoids. Front Bioeng Biotechnol. 2022;10:879024.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Han X, Mslati MA, Davies E, Chen Y, Allaire JM, Vallance BA. Creating a more perfect union: modeling intestinal bacteria-epithelial interactions using organoids. Cell Mol Gastroenterol Hepatol. 2021;12(2):769–82.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Roodsant T, Navis M, Aknouch I, Renes IB, Elburg RMV, Pajkrt D, et al. A human 2D primary organoid-derived epithelial monolayer model to study host-pathogen interaction in the small intestine. Front Cell Infect Microbiol. 2020;10:272.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Holthaus D, Delgado-Betancourt E, Aebischer T, Seeber F, Klotz C. Harmonization of protocols for multi-species organoid platforms to study the intestinal biology of Toxoplasma gondii and other protozoan infections. Front Cell Infect Microbiol. 2021;10: 610368.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Hanyu H, Sugimoto S, Sato T. Visualization of differentiated cells in 3D and 2D intestinal organoid cultures. Methods Mol Biol. 2023;2650:141–53.

    Article  PubMed  Google Scholar 

  105. van der Hee B, Loonen LMP, Taverne N, Taverne-Thiele JJ, Smidt H, Wells JM. Optimized procedures for generating an enhanced, near physiological 2D culture system from porcine intestinal organoids. Stem Cell Res. 2018;28:165–71.

    Article  PubMed  Google Scholar 

  106. Hoffmann P, Schnepel N, Langeheine M, Künnemann K, Grassl GA, Brehm R, et al. Intestinal organoid-based 2D monolayers mimic physiological and pathophysiological properties of the pig intestine. PLoS One. 2021;16(8): e0256143.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Ettayebi K, Crawford SE, Murakami K, Broughman JR, Karandikar U, Tenge VR, et al. Replication of human noroviruses in stem cell-derived human enteroids. Science. 2016;353(6306):1387–93.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Moore MD, Goulter RM, Jaykus LA. Human norovirus as a foodborne pathogen: challenges and developments. Annu Rev Food Sci Technol. 2015;6:411–33.

    Article  PubMed  Google Scholar 

  109. Wang Y, Chiang IL, Ohara TE, Fujii S, Cheng J, Muegge BD, et al. Long-term culture captures injury-repair cycles of colonic stem cells. Cell. 2019;179(5):1144-59 e15.

    Article  PubMed  PubMed Central  Google Scholar 

  110. DiMarco RL, Su J, Yan KS, Dewi R, Kuo CJ, Heilshorn SC. Engineering of three-dimensional microenvironments to promote contractile behavior in primary intestinal organoids. Integr Biol (Camb). 2014;6(2):127–42.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Aguilar C, Silva MAd, Saraiva M, Neyazi M, Olsson IAS, Bartfeld S. Organoids as host models for infection biology – a review of methods. Exp Mol Med. 2021;53(10):1471–82.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Li X, Nadauld L, Ootani A, Corney DC, Pai RK, Gevaert O, et al. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat Med. 2014;20(7):769–77.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Boccellato F, Woelffling S, Imai-Matsushima A, Sanchez G, Goosmann C, Schmid M, et al. Polarised epithelial monolayers of the gastric mucosa reveal insights into mucosal homeostasis and defence against infection. Gut. 2019;68(3):400–13.

    Article  PubMed  Google Scholar 

  114. Kozuka K, He Y, Koo-McCoy S, Kumaraswamy P, Nie B, Shaw K, et al. Development and characterization of a human and mouse intestinal epithelial cell monolayer platform. Stem Cell Rep. 2017;9(6):1976–90.

    Article  Google Scholar 

  115. 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):2165–74.

    Article  PubMed  Google Scholar 

  116. Donkers JM, Eslami Amirabadi H, van de Steeg E. Intestine-on-a-chip: next level in vitro research model of the human intestine. Curr Opin Toxicol. 2021;25:6–14.

    Article  Google Scholar 

  117. Zhang D, Qiao L. Intestine-on-a-chip for intestinal disease study and pharmacological research. View. 2023;4(1):20220037.

    Article  Google Scholar 

  118. Thomas DP, Zhang J, Nguyen NT, Ta HT. Microfluidic gut-on-a-chip: fundamentals and challenges. Biosensors (Basel). 2023;13(1):136.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Beaurivage C, Kanapeckaite A, Loomans C, Erdmann KS, Stallen J, Janssen RAJ. Development of a human primary gut-on-a-chip to model inflammatory processes. Sci Rep. 2020;10(1):21475.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Liu J, Lu R, Zheng X, Hou W, Wu X, Zhao H, et al. Establishment of a gut-on-a-chip device with controllable oxygen gradients to study the contribution of Bifidobacterium bifidum to inflammatory bowel disease. Biomater Sci. 2023;11(7):2504–17.

    Article  PubMed  Google Scholar 

  121. Beaurivage C, Naumovska E, Chang YX, Elstak ED, Nicolas A, Wouters H, et al. Development of a gut-on-a-chip model for high throughput disease modeling and drug discovery. Int J Mol Sci. 2019;20(22):5661.

    Article  PubMed  PubMed Central  Google Scholar 

  122. 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 U S A. 2016;113(1):E7-15.

    Article  PubMed  Google Scholar 

  123. Mazrouei R, Velasco V, Esfandyarpour R. 3D-bioprinted all-inclusive bioanalytical platforms for cell studies. Sci Rep. 2020;10(1):14669.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Fetah K, Tebon P, Goudie MJ, Eichenbaum J, Ren L, Barros N, et al. The emergence of 3D bioprinting in organ-on-chip systems. Prog Biomed Eng. 2019;1(1): 012001.

    Article  Google Scholar 

  125. Yu F, Choudhury D. Microfluidic bioprinting for organ-on-a-chip models. Drug Discov Today. 2019;24(6):1248–57.

    Article  PubMed  Google Scholar 

  126. Barreiro Carpio M, Dabaghi M, Ungureanu J, Kolb MR, Hirota JA, Moran-Mirabal JM. 3D bioprinting strategies, challenges, and opportunities to model the lung tissue microenvironment and its function. Front Bioeng Biotechnol. 2021;9: 773511.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Madden LR, Nguyen TV, Garcia-Mojica S, Shah V, Le AV, Peier A, et al. Bioprinted 3D primary human intestinal tissues model aspects of native physiology and ADME/tox functions. iScience. 2018;2:156–67.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Torras N, Zabalo J, Abril E, Carré A, García-Díaz M, Martínez E. A bioprinted 3D gut model with crypt-villus structures to mimic the intestinal epithelial-stromal microenvironment. Biomater Adv. 2023;153: 213534.

    Article  PubMed  Google Scholar 

  129. Macedo MH, Torras N, García-Díaz M, Barrias C, Sarmento B, Martínez E. The shape of our gut: dissecting its impact on drug absorption in a 3D bioprinted intestinal model. Biomater Adv. 2023;153:213564.

    Article  PubMed  Google Scholar 

  130. Han H, Park Y, Choi YM, Yong U, Kang B, Shin W, et al. A bioprinted tubular intestine model using a colon-specific extracellular matrix bioink. Adv Healthc Mater. 2022;11(2): e2101768.

    Article  PubMed  Google Scholar 

  131. Cheng L, Liu T, Liu Q, Lian L, Tang G, Mille LS, et al. A 3D bioprinted gut anaerobic model for studying bacteria-host interactions. Research (Wash D C). 2023;6:0058.

    PubMed  Google Scholar 

  132. Almutary AG, Alnuqaydan AM, Almatroodi SA, Bakshi HA, Chellappan DK, Tambuwala MM. Development of 3D-bioprinted colitis-mimicking model to assess epithelial barrier function using albumin nano-encapsulated anti-inflammatory drugs. Biomimetics (Basel). 2023;8(1):41.

    Article  PubMed  Google Scholar 

  133. Sbirkov Y, Molander D, Milet C, Bodurov I, Atanasov B, Penkov R, et al. A colorectal cancer 3D bioprinting workflow as a platform for disease modeling and chemotherapeutic screening. Front Bioeng Biotechnol. 2021;9: 755563.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Mitrofanova O, Broguiere N, Nikolaev M, Lutolf MP. Bioengineered human colon organoids with in vivo-like complexity and function. Cell Stem Cell. 2024;31(8):1175–86.e7.

    Article  PubMed  Google Scholar 

  135. Lorenzo-Martin LF, Hubscher T, Bowler AD, Broguiere N, Langer J, Tillard L, et al. Spatiotemporally resolved colorectal oncogenesis in mini-colons ex vivo. Nature. 2024;629(8011):450–7.

    Article  PubMed  PubMed Central  Google Scholar 

  136. van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161(4):933–45.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Luo Z, Wang B, Luo F, Guo Y, Jiang N, Wei J, et al. Establishment of a large-scale patient-derived high-risk colorectal adenoma organoid biobank for high-throughput and high-content drug screening. BMC Med. 2023;21(1):336.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Nishimura R, Shirasaki T, Tsuchiya K, Miyake Y, Watanabe Y, Hibiya S, et al. Establishment of a system to evaluate the therapeutic effect and the dynamics of an investigational drug on ulcerative colitis using human colonic organoids. J Gastroenterol. 2019;54(7):608–20.

    Article  PubMed  Google Scholar 

  139. Lukonin I, Zinner M, Liberali P. Organoids in image-based phenotypic chemical screens. Exp Mol Med. 2021;53(10):1495–502.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Betge J, Rindtorff N, Sauer J, Rauscher B, Dingert C, Gaitantzi H, et al. The drug-induced phenotypic landscape of colorectal cancer organoids. Nat Commun. 2022;13(1):3135.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Chandrasekaran SN, Ceulemans H, Boyd JD, Carpenter AE. Image-based profiling for drug discovery: due for a machine-learning upgrade? Nat Rev Drug Discov. 2021;20(2):145–59.

    Article  PubMed  Google Scholar 

  142. Kong J, Lee H, Kim D, Han SK, Ha D, Shin K, et al. Network-based machine learning in colorectal and bladder organoid models predicts anti-cancer drug efficacy in patients. Nat Commun. 2020;11(1):5485.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Health and Medical Research Council of Australia, projects 1188689, 2021181 and the Australian Research Council, Discovery Project 200103589. Figures were created in BioRender, https://BioRender.com/l74w601 (Figure 1), https://BioRender.com/w50u622 (Figure 2), https://BioRender.com/g75z931 (Figure 3), https://BioRender.com/x07h185 (Figure 4).

Funding

This work was supported by grants from the National Health and Medical Research Council of Australia, projects 1188689 and 2021181, and the Australian Research Council, Discovery Project 200103589.

Author information

Authors and Affiliations

Authors

Contributions

H.A. generated the idea for the article. H.A., D.M., S.H., and W.H.C. developed content, literature search and drafted the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Helen E. Abud.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Micati, D., Hlavca, S., Chan, W.H. et al. Harnessing 3D models to uncover the mechanisms driving infectious and inflammatory disease in the intestine. BMC Biol 22, 300 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12915-024-02092-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12915-024-02092-9

Keywords