The small intestinal microbiome: vibing with intestinal stem cells

in Microbiota and Host
Authors:
Victoria Poplaski Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA

Search for other papers by Victoria Poplaski in
Current site
Google Scholar
PubMed
Close
,
Faith Sawyer Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA

Search for other papers by Faith Sawyer in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-5125-6941
, and
Sarah E Blutt Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA

Search for other papers by Sarah E Blutt in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-4062-709X

Correspondence should be addressed to S E Blutt; Email: sb691007@bcm.edu
Open access
Sign up for journal news

The epithelial lining of the small intestine mediates its absorptive and secretory function and thus is a critical component of human health. Regeneration and renewal of the epithelium is the result of proliferation of intestinal stem cells (ISCs). Many cell types and molecular factors are known to regulate the ability of ISCs to proliferate, including adjacent neighboring epithelial cells and the underlying, supportive stromal cells. The microbiome resides in the lumen of the small intestine and is in close contact with the epithelium. Due to its proximity to ISCs, it has been hypothesized that species within the microbiome have the capacity to regulate ISC proliferation and differentiation. This review highlights research that probes interactions between ISCs and the microbiome in the small intestine to detail the current understanding of microbial regulation of ISCs. Results from these studies provide important knowledge that can be exploited to identify therapeutic targets or develop novel preventative treatments to treat intestinal diseases.

Abstract

The epithelial lining of the small intestine mediates its absorptive and secretory function and thus is a critical component of human health. Regeneration and renewal of the epithelium is the result of proliferation of intestinal stem cells (ISCs). Many cell types and molecular factors are known to regulate the ability of ISCs to proliferate, including adjacent neighboring epithelial cells and the underlying, supportive stromal cells. The microbiome resides in the lumen of the small intestine and is in close contact with the epithelium. Due to its proximity to ISCs, it has been hypothesized that species within the microbiome have the capacity to regulate ISC proliferation and differentiation. This review highlights research that probes interactions between ISCs and the microbiome in the small intestine to detail the current understanding of microbial regulation of ISCs. Results from these studies provide important knowledge that can be exploited to identify therapeutic targets or develop novel preventative treatments to treat intestinal diseases.

Introduction

The small intestinal epithelium is a single layer of diverse types of secretory and absorptive cells that mediate intestinal function. The epithelium is divided into two microdomains or compartments: the villus and the crypt. The crypt region is characterized by invaginating pocket-like structures that are surrounded by the underlying stroma, while the villus region protrudes into the intestinal lumen to increase surface area for nutrient absorption. The crypt is home to intestinal stem cells (ISCs), that undergo two types of cell division. Symmetric division generates new ISCs for self-renewal, and asymmetric division gives rise to daughter cells that migrate from the crypt region into the villus region, where they undergo differentiation to functional secretory and absorptive cells. ISCs continuously proliferate and differentiate, replacing the mature cells that line the villus region every 3–5 days. The microenvironment of the small intestinal crypt where ISCs reside is termed the ‘ISC niche.’ Components of the ISC niche include the surrounding differentiated epithelial cells that remain in the crypt instead of migrating to the villus, and the underlying stromal cells, a heterogeneous group of cells that reside in the connective tissue including mesenchymal, immune, neuronal, and vascular cells. The epithelial and stromal components of the ISC niche serve as a well-known, major source of signals that modulate ISC proliferation and differentiation (Santos et al. 2018, Greicius & Virshup 2019). Recently, the human microbiome has emerged as another source of factors that regulate the function of ISCs. The advent of the Human Microbiome Project resulted in a wealth of information regarding the putative microbial composition in the small intestine (Human Microbiome Project 2012), and a recent focus on the functionality of these organisms has resulted in the discovery that the microbiome and ISCs have an intimate relationship (Peck et al. 2017). This review will discuss the regulation of ISCs, the current understanding of the landscape of the small intestinal microbiome and its products, and the evidence that the microbiome is a source of signals for ISCs and thereby a major component of the ISC niche. It is important to comprehend how ISCs interact with the small intestinal microbiota and are influenced by bacterial products, as this knowledge enhances our understanding of the factors that assist in maintaining epithelial homeostasis and contribute to renewal and repair in the context of small intestinal damage.

Regulation of the intestinal stem cell

ISCs in the small intestine undergo continuous rounds of cell division approximately every 24 h (Potten 1998). As they divide in the base of the crypt, undifferentiated daughter cells migrate upward through the transit-amplifying (TA) zone near the crypt–villus axis, where they continue to divide as they progress upward toward the villus. As the cells move up the villus, they differentiate into their respective functional cell types, including enteroendocrine cells, which secrete hormones, enterocytes, which absorb nutrients, tuft cells, which perform chemosensory functions, and goblet cells, which secrete mucus. The exception to this upward movement of differentiating cells are Paneth cells, which reside in the base of the crypts, where they secrete antimicrobial factors and shape the intestinal microbial composition (Fig. 1) (van der Flier & Clevers 2009, Bevins & Salzman 2011, Tian et al. 2011, Barker et al. 2012, Clevers 2013, van Clevers & Bevins 2013, Metcalfe et al. 2014). Apart from Paneth cells and a few enteroendocrine and tuft cells that remain in the crypt region, differentiated daughter cells migrate to the tip of the villus, where they are eventually shed into the small intestinal lumen by a process called anoikis. As ISCs are primarily responsible for this process of generating new daughter cells that differentiate into the mature intestinal cell types, regulation of ISCs is tightly controlled to ensure normal epithelial homeostasis and renewal, and conversely, is dysregulated in many small intestinal diseases.

Figure 1
Figure 1

Landscape of the intestinal epithelium. The intestinal epithelium is made up of diverse cell types organized into microdomains termed the crypt and villus. The microbiome lies adjacent to the intestinal epithelium in close contact with a mucous layer produced by the epithelium. The villus contains differentiated cells including enterocytes, goblet cells, enteroendocrine cells, and tuft cells. The crypt contains the undifferentiated intestinal stem cells (ISCs) and transient amplifying (TA) cells. The Paneth cells and few enteroendocrine and tuft cells reside alongside the ISC in the crypt. The stromal cells reside directly beneath the epithelium in the crypt region and provide essential regulatory factors to the ISC. Created in BioRender.

Citation: Microbiota and Host 1, 1; 10.1530/MAH-23-0012

Many markers have been used to identify ISCs, facilitating the study of ISC regulation. Currently in the small intestine, the most well-studied crypt ISC marker is a cell surface receptor named LGR5 (leucine-rich-repeat-containing G-protein-coupled receptor 5, also known as Gpr49). Other markers that have been associated with ISCs include OLFM4, KI67, PCNA, SOX9, ASCL2, and AXIN2 (Barker et al. 2012, Burclaff et al. 2022). Postulated quiescent or reserved stem cells – also called +4 ISCs – reside directly above the terminal Paneth cells in the beginning of the TA zone, and are marked by BMI1, HOPX, TERT, and LRIG1 (Yan et al. 2012, Duckworth 2021, Burclaff et al. 2022). The existence of quiescent ISCs is controversial, and unlike the actively dividing LGR5+ ISCs, quiescent ISCs are not thought to be the primary source of epithelial renewal in homeostatic conditions, but are rather believed to play a major role in regenerative repair in the context of LGR5+ ISC damage, especially following radiation exposure (Metcalfe et al. 2014, Andersson-Rolf et al. 2017, Dheer & Young 2021). Control of LGR5+ ISC proliferation and differentiation in the small intestine has been linked to conserved signaling molecules and pathways, including wingless/integrated (WNT), R-spondin, epidermal growth factor (EGF), Hedgehog, Notch, Gremlin, and bone morphogenetic proteins (Vanuytsel et al. 2013). Key factors in these pathways are provided to ISCs by various cells that comprise the ISC niche (Santos et al. 2018, Greicius & Virshup 2019, Duckworth 2021), namely Paneth cells, which reside in between the actively dividing LGR5+ ISCs (Fig. 1) and provide WNT and Notch ligands to ISCs (Clevers & Bevins 2013), and stromal cells, such as telocytes and myofibroblasts, which reside underneath the epithelium and serve as a source of WNTs, R-spondin proteins, and EGF (Gregorieff et al. 2005, Kabiri et al. 2014, Greicius et al. 2018, Greicius & Virshup 2019). The canonical WNT pathway is the most well-established regulator of ISCs (Barker et al. 2007, 2008, 2012, Logan & Nusse 2004, Katoh 2007, Katoh & Katoh 2007), and WNT proteins secreted from stromal or Paneth cells locally activate intracellular signaling cascades within neighboring ISCs in a paracrine fashion (Theodosiou & Tabin 2003, Katoh 2007, Katoh & Katoh 2007, Sato et al. 2011, Kabiri et al. 2014, Greicius et al. 2018, Greicius & Virshup 2019). WNT target genes play an indispensable role in maintaining stemness and promoting proliferation in the small intestine, and WNT signaling thereby serves as the master regulator of proliferation in the ISC niche (Logan & Nusse 2004, Katoh & Katoh 2007, Biechele & Moon 2008). In addition to their role in small intestinal homeostasis, many of these proliferation-promoting factors, such as WNT, have been shown to play a vital role in ISC recovery after damage (Santos et al. 2018, Gehart & Clevers 2019, Greicius & Virshup 2019, Duckworth 2021).

The microbiome

The microbiome consists of more than 300 trillion bacteria, fungi, and viruses (Dave et al. 2012, Human Microbiome Project 2012) that colonize the gastrointestinal tract. The small intestinal organisms are dynamic due to the changing conditions in the small intestine caused by intake of food, the intermittent secretion of enzymes and factors, and short transit time (Kastl et al. 2020). The composition of organisms varies by region with increasing amounts and diversity from the proximal to distal portions (Kastl et al. 2020). Interactions between these organisms and ISCs could occur through two possible points of contact: direct communication between the organism and ISCs or communication with a niche cell that then indirectly signals ISCs (Nelson & Mata 1970, Savage & Blumershine 1974, Peck et al. 2017). Although tantalizing data suggests small intestinal microbes play a role in regulating ISCs, the underlying mechanisms that form the basis for microbiome–ISC interactions have not been elucidated. Filling this knowledge gap has been challenging, partly due to the lack of in vitro models in which microbe–epithelium interactions can be interrogated, the diversity of the gut microbiome and factors it produces, and the difficulty in pinpointing exactly what organisms are present in the small intestine. Most of the studies investigating small intestinal microbe–ISC interactions have utilized murine models. Although much has been learned from these studies, the composition of the murine microbiome is distinct from humans due to many factors, including host genetics, differences in the composition of outer intestinal mucosa layer that affects mucus-associated bacteria survival, lower intestinal pH values, and differences in diet (Arrieta et al. 2016, Hugenholtz & de Vos 2018). Humans and mice share only 15% of intestinal bacteria lineages, and while murine models can provide valuable insights into ISC–microbiome interactions, the many differences between the human and mouse intestinal microbiomes highlight the need to study these interactions in a complimentary human model system (Arrieta et al. 2016). Human intestinal organoids provide a new model system in which human microbe–ISC interactions can be interrogated ex vivo, as they enable the culturing of human ISCs, model differentiation of the human epithelium, and facilitate the exploration of microbial–ISC interactions in a reductionist environment (Foulke-Abel et al. 2014, Zachos et al. 2016, Blutt et al. 2018). New advances in human organoid cultures will be addressed below.

The landscape of the intestinal microbiome and its metabolites

The composition of the small intestinal microbiome is highly variable based on the genetics of the individual and environmental factors such as age, diet, and antibiotic use (Hasan & Yang 2019, Kurilshikov et al. 2021). The microbiome produces many factors that appear to modulate its functional role in the intestine, including short chain fatty acids (Boffa et al. 1992, Ichikawa & Sakata 1997, Lee et al. 2017, 2018), tryptophan metabolites (Roager & Licht 2018), polyamines (Wang et al. 1991), secondary bile acids (Kozoni et al. 2000, Pai et al. 2004), vitamins (Lai et al. 2021), reactive oxygen species (Morris & Jasper 2021), and hydrogen sulfide (Xing et al. 2020). Most of the research characterizing the overall intestinal microbiome has utilized 16S gene content analysis in stool, which is easily collected via noninvasive methods (Dave et al. 2012, Human Microbiome Project 2012, Tang et al. 2020) but reveals very little about what might be present in the small intestine. Recent studies have revealed differences between the composition of the stool microbiome compared to the microbiome found at the intestinal epithelial surface (Vasapolli et al. 2019, Vuik et al. 2019). Differences in bacterial composition also exist between the luminal microbiota and the epithelial-associated microbiota, potentially because epithelial-associated organisms have unique properties allowing them to utilize nutrients and adhere to glycans within the mucous layer that coats the epithelial surface (Robbe et al. 2004, Juge 2012). The composition of the microbiome also varies by region in the intestine and increases in diversity from the small intestine to the large intestine, with a smaller bacterial load and less-diverse microbiota associated within the upper duodenal regions of the small intestine and the largest numbers and greatest diversity of bacteria found in the terminal large intestine (Zhang et al. 2014, Martinez-Guryn et al. 2019). The diversity of the bacterial communities along the different regions of the intestine also contributes to the spectrum of products produced by microbes in each intestinal region, thereby affecting the local interactions of microbes and their metabolites with the epithelium (Wang et al. 2005, Stearns et al. 2011).

While researchers are making strides in gaining a deeper understanding of which bacteria inhabit the upper and lower regions of the human gastrointestinal tract, the composition of the human microbiota that colonizes the middle portions of the small intestine – the jejunum and upper ileum – has remained elusive (Dave et al. 2012, Kastl et al. 2020, Tang et al. 2020). Difficulties in assessing the microbiota in this region is mainly a result of technical challenges in sampling these regions through traditional endoscopy and colonoscopy methods, as the middle regions of the small intestine are not easily reached by scope equipment. Additionally, utilizing animal models to make these discoveries is limited by the substantial variations in microbial composition between animals and humans. Advancements in identifying which bacteria reside in the small intestine may be achieved by examining small intestinal tissues obtained through organ donation. By scraping the intestinal tissue, organ donor samples can be used to isolate microbes found in these regions from healthy individuals (Sartor 2015).

Role of the microbiome on intestinal stem cell regulation

The idea that small intestinal microbiome plays a role in modulating ISC activity results from its proximity to the epithelium and ability to secrete factors that can modulate ISC biology. Indirect evidence for microbial regulation of ISCs originated from observations by Gordon and Bruckner-Kardoss in 1961 that germ-free mice showed reduced intestinal surface area compared to that of conventionally raised mice (Gordon & Bruckner-Kardoss 1961). Many other studies have observed the phenomena of reduced villus height, crypt depth, and mitotic indices in the small intestine of germ-free or antibiotic-treated mice when compared to mice with a unaltered microbiome (Lesher et al. 1964, Khoury et al. 1969, Greig et al. 2018), suggesting that the microbiome can control epithelial regeneration and renewal. Further support for a link between the microbiome and ISCs comes from reports that colonization with microbes restores normal intestinal epithelial histology in germ-free mice, rats, and Drosophila (Gordon & Bruckner-Kardoss 1961, Banasaz et al. 2002, Buchon et al. 2009a ). Evidence for direct interactions between microbes and ISCs has also emerged from studies by Lee et al. where microbially produced lactate was shown to enhance proliferation in the murine small intestine via the stimulation of the LGR5 receptor on ISCs (Lee et al. 2018). It remains to be seen whether this activation of LGR5 ISCs occurs because microbially derived lactate interacts directly with ISCs or indirectly via the stimulation of other cell types of the ISC niche (Table 1). Although an in-depth mechanism of how the microbiome regulates ISCs remains to be fully elucidated, evidence from the literature suggests that a complex network of microbes and their metabolites are clearly involved in regulating ISC activity through various niche pathways, in the context of both homeostasis and damage.

Table 1

Summary of the known effects of microbial species and strains on ISC regulation.

Microbe/Strain Model Effects on ISCs Mechanisms Reference
Akkermansia muciniphila
 ATCC BAA-835 and AK32 Normal mouse, radiation and methotrexate mouse damage model Stimulates proliferation of Lgr5+ ISCs, promotes differentiation to Paneth cells and goblet cells, promotes gut repair following damage Activates Wnt/β-catenin pathway by promoting Wnt3 secretion Kim et al. (2021), Duan et al. (2023)
Bacillus subtilis
 JNFE0126 DSS-induced mouse colitis model Stimulates proliferation, increases Lgr5+ ISC numbers Protects ISCs from inflammatory injury; rebalances intestinal microbiota Zhang et al. (2020)
Bacteroides fragilis
 ZY-312 DSS-induced mouse colitis model Promotes colonic proliferation Promotes colonic mucosal regeneration in colitis via IL-22-induced STAT3 phosphorylation Zhang et al. (2023)
 086-5443-2-2 Human colonic epithelial HT29/C1 cells Promotes proliferation and c-myc expression Activates β-catenin/TCF signaling pathway via Wu et al. (2003)
Erwinia carotovora carotovora 15
 Ecc15 Ecc15-infected Drosophila model Stimulates ISC proliferation and differentiation to enteroendocrine cells JAK-STAT pathway induction of ISC proliferation via Upd3 release from enterocytes Buchon et al. (2009b), Liu et al. (2022)
Lactobacillus acidophilus
 ATCC4356 Salmonella typhimurium-infected mouse intestinal organoids Inhibits proliferation, inhibits excessive differentiation of Paneth cells and goblet cells Inhibits overactivation of Wnt/β-catenin pathway Lu et al. (2020)
Lactobacillus casei
 ATCC334 Radiation-induced rat injury model Stimulates proliferation, increases Lgr5+ ISC numbers Promotes proliferation via alpha-linolenic acid production and IL-22 Hua et al. (2023)
 ATCC334 Normal mouse and porcine epithelial cells Stimulates epithelial cell proliferation Induces epithelial expression of Reg3a to inhibit pathogens and stimulate damage repair Bai et al. (2021)
Lactobacillus plantarum
Normal Drosophila Stimulates ISC proliferation Nox1-dependant reactive oxygen species generation Jones et al. (2013)
Lactobacillus reuteri
 D8 C. rodentium-infected mouse, DSS-induced mouse colitis model, TNF-damaged mouse organoids Stimulates proliferation, induces differentiation of Paneth cells, increases Lgr5+ ISC numbers Induces R-spondin expression and activates Wnt/β-catenin pathway; Wu et al. (2020)
 D8 Stimulates lamina propria lymphocyte secretion of IL-22 and STAT3 signaling pathway Hou et al. (2018)
 17938 Normal neonatal mice Stimulates proliferation Increases microbial phylogenetic diversity Preidis et al. (2012)
Lactobacillus rhamnosus
 GG Normal mouse colonic organoids, normal mouse Stimulates colonic epithelial proliferation Microbe-induced leptin expression triggers JAK-STAT signaling in a nox1 and leptin receptor-dependent manner Jones et al. (2013), Darby et al. (2020)
 GG Normal mouse Stimulates ISC proliferation Nox1-dependant reactive oxygen species generation Lyu et al. (2022)
 GG Intrauterine growth retardation rat pups Stimulates proliferation and goblet cell differentiation Stimulates WNT/β-catenin signaling Banasaz et al. (2002)

Many species and strains of intestinal bacteria have been implicated in affecting ISC proliferation and differentiation (Table 1). Due to the amount of genetic variability between strains of a bacterial species – especially in those that were isolated from different hosts (Frese et al. 2011) – it is probable that many of the observed effects of a given microbe are strain-specific. For example, in mice, Lactobacillus reuteri strain 17938 increases murine small intestinal crypt cell proliferation, but strain 6475 does not (Preidis et al. 2012). Comparative genomics reveals that although these strains are members of the same species, they share only 70% of their genes (Saulnier et al. 2011). As these strains differ in their ability to induce crypt cell proliferation, these genetic differences may code for factors that modulate ISCs, potentially providing key targets for further exploring the relationship between ISCs and the microbiome. Strain-specific differences are particularly relevant when drawing conclusions from studies linking specific microbial metabolites or factors from a microbial species to ISC regulation, as not all strains of a species may produce the same factor.

Many different mechanisms have been proposed to explain how microbes regulate ISCs (Table 1 and 2), suggesting that microbial regulation of ISCs is most likely complex and multifactorial, involving multiple pathways and mechanisms. Of the major niche pathways, the mechanism often proposed for microbial stimulation of ISC proliferation is the activation of WNT/β-catenin signaling (Wu et al. 2020, Kim et al. 2021, Lyu et al. 2022), which is unsurprising, as this pathway is the major proliferation-stimulating pathway in LGR5+ ISCs, and without proper WNT/β-catenin signaling, the intestinal epithelium is unable to self-renew (Korinek et al. 1998, Pinto et al. 2003, Kuhnert et al. 2004). Whether microbes induce these pathways directly, with microbial cells themselves interacting with the epithelial surface, or indirectly, via microbial metabolites, secreted proteins, or outer membrane vesicles remains poorly understood. Due to the presence of the mucus barrier that overlays the intestinal epithelium in the absence of injury or disease, many studies have concluded that microbes are unable to physically interact with the epithelium, hypothesizing that the ISC niche is a sterile environment, free from microbes themselves (Johansson et al. 2008, 2014, Hansson & Johansson 2010). However, the crypt location would be ideal for microbial modulation of ISC pathways through direct microbial communication with the various ISC niche components – potentially through interaction of microbial cell wall components with the epithelium or through the release of outer membrane vesicles or metabolites that can locally interact with the niche – yet whether these direct interactions occur remains unknown.

Table 2

Summary of the known effects of microbial metabolites and factors on ISC regulation.

Microbial factor Description Effects on ISCs Mechanisms Reference
Butyrate SCFA Stimulates proliferation Stimulates MEK-ERK signaling Park et al. (2016)
Stimulates proliferation Unspecified Boffa et al. (1992)
Inhibits proliferation Unspecified Kaiko et al. (2016)
Deoxycholic acid Secondary bile acid Inhibits proliferation Possible FXR inactivation of EGFR signaling Dossa et al. (2016)
Flagellin Flagella component Stimulates proliferation Upregulation of NOX1 and stimulation of EGFR signaling van der Post et al. (2021)
Indole-3-aldehyde Tryptophan metabolite Promotes proliferation and Paneth cell differentiation Stimulation of IL-22 secretion and STAT3-dependent proliferation stimulation Hou et al. (2018)
Promotes proliferation and goblet cell differentiation Promotes IL-10 signaling Powell et al. (2020)
Indoleacetic acid Tryptophan metabolite Inhibits proliferation Aryl hydrocarbon receptor (AhR) E3 ubiquitin ligase degradation of β-catenin Kawajiri et al. (2009)
Lactate Organic acid Stimulates proliferation, enhances survival of Lgr5+ ISCs Gpr81-dependent Wnt3 expression and WNT/β-catenin pathway stimulation Lee et al. (2018)
Lipopolysaccharide Gram-negative bacterial cell membrane Inhibits proliferation, promotes cell death, promotes goblet cell differentiation TLR4-induced apoptosis Naito et al. (2017)
Muramyl-dipeptide Peptidoglycan (cell wall) motif Stimulates proliferation, enhances survival of Lgr5+ ISCs Nod2-mediated cytoprotection Nigro et al. (2014), Levy et al. (2020)
Propionate SCFA Stimulates proliferation, increases ISC stemness, increased LGR5+ cells Stimulates MEK-ERK signaling and WNT signaling Park et al. (2016), Duan et al. (2023)
Succinic acid Organic acid Inhibits proliferation Unspecified Inagaki et al. (2007)
Valproic acid SCFA Stimulates proliferation, suppresses secretory lineage differentiation Stimulates Notch pathway Yin et al. (2014)
Stimulates proliferation Stimulates MEK-ERK signaling Park et al. (2016)

In addition to WNT/β-catenin signaling, other pathways of microbial regulation of ISCs have also been suggested. ISCs robustly express the pattern recognition receptor NOD2, which recognizes the peptidoglycan motif muramyl dipeptide (MDP) that is a cell wall component in all bacteria (Ogura et al. 2003, Nigro et al. 2014). This finding serves as a clue that microbial cells may be able to physically interact directly with ISCs. MDP has been linked to beneficial effects in epithelial repair and has recently emerged as a microbial-responsive mechanism that aids in ISC survival in murine and organoid studies. Nod2 has been shown to trigger a pathway of ISC cytoprotection when stimulated by MDP; a mechanism thought to enhance the ability of ISCs to regenerate crypts upon exposure to cytotoxic stressors such as reactive oxygen species, radiation damage, or the chemotherapeutic agent doxorubicin (Nigro et al. 2014, Lee et al. 2019, Levy et al. 2020).

Many microbially derived metabolites and secreted products are thought to provide important components to the ISC niche (Table 2). Recognition that small intestinal microbial products such as MDP and lactate can maintain the proliferative capacity of the ISC niche in the context of damage highlights the importance of maintaining a diverse, healthy microbiome. Several strains promote proliferation and facilitate epithelial renewal following damage, as seen in DSS-induced mouse colitis models, radiation-induced rat injury models, and during pathogenic infection (Lu et al. 2020, Wu et al. 2020, Zhang et al. 2020, Hua et al. 2023, Zhang et al. 2023) (Table 1). These findings demonstrate that microbes play an important role in repairing the small intestinal epithelium following damage. Commensal microbes, such as Lactobacillus reuteri D8, can stimulate epithelial proliferation and repair to reduce intestinal pro-inflammatory cytokine secretion and serum lipopolysaccharide (LPS) concentrations (Wu et al. 2020). Additionally, in the context of damage due to inflammation or radiation injury, commensal microbes – especially lactobacilli – have been documented to play a very important role in facilitating small intestinal epithelial renewal by stimulating ISC proliferation via IL-22 secretion from immune cells (Qiu et al. 2017, Hou et al. 2018, Ge et al. 2022, Hamade et al. 2022, Hua et al. 2023, Zhang et al. 2023). In contrast, other evidence suggests that certain microbes might enhance damage. The microbial endotoxin LPS, which is often present in higher amounts during pathogenic infection, has been shown to inhibit cellular proliferation and increase apoptosis in bot the small intestine upon binding to the toll-like receptor 4 (TLR4) (Neal et al. 2012, Naito et al. 2017). Further work is necessary to understand both the positive and detrimental effects of the small intestinal microbial composition on ISC regulation.

Ways to study microbial regulation of intestinal stem cells

Most of the research studying the pro-proliferative effects of various microbes on the ISC has been conducted in murine models. However, it has not yet been ascertained how applicable these discoveries in murine systems are to human biology. The expanded use of human intestinal organoid model systems provides the opportunity to overcome the hurdles of host and species-specific differences. In 2011, Sato et al. pioneered the development of ex vivo tissue-derived human small intestinal organoids (HIOs), which allow the direct cultivation of human ISCs in vitro (Sato et al. 2011). Unlike transformed cell lines, HIOs are genetically stable and closely model the cellular makeup of the in vivo intestinal epithelium (Sato et al. 2009, 2011, Foulke-Abel et al. 2014, Blutt et al. 2018), which presents many advantages. HIOs can be grown in multiple formats, facilitating the study of many scientific questions involving microbe–epithelial interactions (Fig. 2). In a 3D format, bacteria can be microinjected into the lumen of the HIO to assess how live microbes affect the rate of organoid growth and ISC proliferation (Poletti et al. 2021). Williamson et al. recently developed a high-throughput method of 3D organoid microinjection to evaluate how various microbes influenced gastrointestinal physiology (Williamson et al. 2018). Additional studies in 3D HIOs have analyzed the impacts of microbes or their secreted products on various aspects of epithelial biology (Co et al. 2019, Dheer & Young 2021). HIOs can also be grown in a 2D monolayer or transwell format, which allows easy access to the epithelial apical surface for the application of microbes or their products and more closely mimics the physiologic contact of the epithelium and the intestinal microbiome (VanDussen et al. 2015, Wilke et al. 2020). Transwells also facilitate access to the basolateral side of the epithelium to address microbial–cell interactions that may occur in this region and model systemic infections (Wang et al. 2018). Work in organoid models has allowed researchers to determine the colonization patterns of microbes and how microbial factors affect the apical surface of the epithelial cells (Rajan et al. 2018, Zhang et al. 2020). As HIOs have the advantage of preserving the host’s genetic makeup in vitro, microbial–HIO studies can encompass multiple individuals with a variety of genetic backgrounds, permitting the assessment of demographic factors such as sex, ethnicity, and age to be studied in the context of the microbiome. The advent of HIOs presents a powerful tool that can be used to gain a comprehensive understanding of the intricate interplay between the human small intestinal microbiome and ISCs. To circumvent disparities in the microbiota between mice and humans, future research endeavors should supplement rodent models with the integration of microbiome components into HIOs.

Figure 2
Figure 2

Formats of human intestinal organoids. Human intestinal organoids (HIOs) can be used in a variety of formats depending on the scientific questions to be explored. Created in BioRender.

Citation: Microbiota and Host 1, 1; 10.1530/MAH-23-0012

Additional methods to study complex communities of bacteria are being developed that, when combined with organoid technology, can be used to assess host–microbe and ISC–microbe interactions. Robert Britton’s laboratory pioneered the development of mini-bioreactor arrays (MBRAs) that can be utilized to cultivate complex human small intestinal microbial communities and metabolites (Auchtung et al. 2015). Using the MBRA system, stable microbial communities can be reliably and reproducibly cultured within 8 days, with approximately 94% of the microbes present from the original sample represented in the MBRA community (Auchtung et al. 2015). Supernatants from these communities can be collected and their biological effects on ISCs assessed using several different assays. Other laboratories are actively expanding the repertoire of culturable bacteria; an advancement that, when paired with HIO studies, will substantially contribute to the scientific community’s ability to uncover the mechanistic relationship between complex communities of microbes and ISCs (Rettedal et al. 2014, Kim et al. 2016, 2022, Wang et al. 2018, Afrizal et al. 2022).

Microbial regulation of the large intestine stem cell

Unlike the small intestinal epithelial landscape, which has a crypt and villus architecture, the colonic epithelial landscape is much different, and only consists of deep glandular crypt regions. The colon is home to the greatest and most diverse population of microbes in the body; therefore, it is also important to understand how the resident bacteria in the colon may be impacting the regeneration processes of the colonic stem cell. Due to the ease of sampling and characterizing the microbiome in the large intestine, there is a plethora of research examining whether large intestinal microbial communities modulate ISCs. Driving this work is the association of colorectal cancer and the microbial composition in the large intestine. Large intestinal microbial dysbiosis strongly correlates with the development of cancer in the large intestine (Kim & Lee 2021), and there is much focus on understanding the effect of the large intestinal microbiome on epithelial proliferation.

Several groups have found that various large intestinal microbes and microbial products can influence ISC proliferation in the colon including Bacteroides fragilis, and Lactobacillus rhamnosus GG (Wu et al. 2003, Darby et al. 2020, Zhang et al. 2023). Colonic organisms have been postulated to interact with the colonic ISC via several different mechanisms, including a toxin-mediated destruction of epithelial E-cadherin which subsequently triggers an epithelial repair response characterized by β-catenin translocation to the nucleus, a hallmark of ISC activation (Wu et al. 2003). Other mechanisms include activation of STAT2 signaling via the induction of IL-22 (Zhang et al. 2023), leptin mediated induction of epithelial proliferation, and proliferation via pathways that involve Nox (Darby et al. 2020). Most of the work linking specific large intestinal microbial metabolites to ISC regulation involve short-chain fatty acids (SCFAs), such as butyrate, acetate, propionate, and valproic acid, which are produced by microbial fermentation of dietary polysaccharides and are found primarily in the colon. The most well-studied SCFA is butyrate, which serves as a direct energy source for colonocytes and has been shown to modulate inflammation (Donohoe et al. 2011, Chang et al. 2014, Lee et al. 2017, Dou et al. 2020). Butyrate can stimulate proliferation in healthy colonic epithelium but inhibits proliferation in tumor cell lines (Whitehead et al. 1986, Sakata 1987, Frankel et al. 1994, Kien et al. 2007), a phenomenon known as the ‘butyrate paradox’ (Mariadason et al. 2001, Comalada et al. 2006). Although detailed mechanisms explaining how colonic microbial metabolites regulate ISCs have not been fully elucidated, evidence from the literature suggests that many microbial metabolites can regulate epithelial proliferation. With the high prevalence of colonic cancers worldwide (Boustany et al. 2023), it is important to understand the relationship between resident colonic microbiota and their influences on epithelial proliferation. Recent work has identified a crypt-specific microbiota that resides deep within murine colonic crypts (Pedron et al. 2012, Saffarian et al. 2019) which implies local interactions between the two entities. Further research is needed to fully understand and appreciate whether organisms that live in the large intestinal crypt region provide local signals that regulate ISCs.

Discussion and Conclusion

The intestinal lumen is a highly dynamic and changing environment that is home to trillions of bacteria that constitute the intestinal microbiome, a complex ecosystem of organisms that live in symbiosis with the intestinal epithelium. Epithelial maintenance is critical for intestinal health and originates from ISCs. The relationship between the intestinal microbiota and ISCs is highly dynamic and understanding its complexity is still in its infancy. Rodent, zebrafish, and Drosophila model systems provide solid evidence that the microbiome plays an important regulatory role in ISC functioning. In addition to functioning under homeostasis, the microbiome has also been linked to enhanced intestinal repair mechanisms that occur following injury. A deep focus on the role of the microbiome in regeneration and renewal following intestinal damage will begin to shed light on the utilization and manipulation of the microbiome to treat intestinal disease. Collectively, this area of research implies that the intestinal microbiome holds tremendous potential as a therapeutic target. In addition, the microbiome can be regulated by many external and internal factors including dietary factors (Perler et al. 2023) such as fiber (Myhrstad et al. 2020) and sugars (Di Rienzi & Britton 2020), prebiotic (Bedu-Ferrari et al. 2022), probiotic (Hemarajata & Versalovic 2013) and antibiotic use (Fishbein et al. 2023), stress (Segerberg-Konttinen 1988), physical activity (Holzhausen et al. 2022), sleep (Klimashina et al. 1989), aging (Badal et al. 2020), genetics (Hall et al. 2017), and disease (Durack & Lynch 2019). Understanding how these variables indirectly affect ISC dynamics and whether they have the potential to modulate intestinal regeneration and repair via their effects on the microbiome is an emerging area of research. It is tantalizing to speculate as to whether simple interventions such a modulation of diet might modulate ISC renewal in humans, as this has been demonstrated in mice (Hou et al. 2021). Deeper knowledge of microbiome–ISC interactions will provide an understanding of the potential application of the microbiome to human health. We predict that the field will continue to advance with the use of new model systems to fully understand how the microbiome ‘vibes’ with ISCs in humans and further define the human ISC niche.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

Funding

This work was supported by the National Institute of Health grants U01-DK103168, and U19-AI116497, and the Translational Research Institute for Space Health grant NNX16AO69A.

Author contribution statement

FS, VP, and SEB wrote the manuscript.

Acknowledgements

We are grateful for the support and guidance from Dr Mary K. Estes in the completion of the manuscript by providing feedback and editing the manuscript. Images were created using BioRender.

References

  • Afrizal A, Hitch TCA, Viehof A, et al.2022 Anaerobic single-cell dispensing facilitates the cultivation of human gut bacteria. Environmental Microbiology 24 38613881. (https://doi.org/10.1111/1462-2920.15935)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Andersson-Rolf A, Zilbauer M, Koo BK, et al.2017 Stem cells in repair of gastrointestinal epithelia. Physiology 32 278289. (https://doi.org/10.1152/physiol.00005.2017)

  • Arrieta MC, Walter J & & Finlay BB 2016 Human microbiota-associated mice: a model with challenges. Cell Host and Microbe 19 575578. (https://doi.org/10.1016/j.chom.2016.04.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Auchtung JM, Robinson CD & & Britton RA 2015 Cultivation of stable, reproducible microbial communities from different fecal donors using minibioreactor arrays (MBRAs). Microbiome 3 42. (https://doi.org/10.1186/s40168-015-0106-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Badal VD, Vaccariello ED, Murray ER, et al.2020 The gut microbiome, aging, and longevity: a systematic review. Nutrients 12. (https://doi.org/10.3390/nu12123759)

  • Bai Y, Huang Y, Li Y, et al.2021 The murine Reg3a stimulated by Lactobacillus casei promotes intestinal cell proliferation and inhibits the multiplication of porcine diarrhea causative agent in vitro. Frontiers in Microbiology 12 675263. (https://doi.org/10.3389/fmicb.2021.675263)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Banasaz M, Norin E, Holma R, et al.2002 Increased enterocyte production in gnotobiotic rats mono-associated with Lactobacillus rhamnosus GG. Applied and Environmental Microbiology 68 30313034. (https://doi.org/10.1128/AEM.68.6.3031-3034.2002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barker N, van Es JH, Kuipers J, et al.2007 Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449 10031007. (https://doi.org/10.1038/nature06196)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barker N, Van de Wetering M & & Clevers H 2008 The intestinal stem cell. Genes and Development 22 18561864. (https://doi.org/10.1101/gad.1674008)

  • Barker N, Van Oudenaarden A & & Clevers H 2012 Identifying the stem cell of the intestinal crypt: strategies and pitfalls. Cell Stem Cell 11 452460. (https://doi.org/10.1016/j.stem.2012.09.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bedu-Ferrari C, Biscarrat P, Langella P, et al.2022 Prebiotics and the human gut microbiota: from breakdown mechanisms to the impact on metabolic health. Nutrients 14. (https://doi.org/10.3390/nu14102096)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bevins CL & & Salzman NH 2011 Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis. Nature Reviews. Microbiology 9 356368. (https://doi.org/10.1038/nrmicro2546)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Biechele TL & & Moon RT 2008 Assaying beta-catenin/TCF transcription with beta-catenin/TCF transcription-based reporter constructs. Methods in Molecular Biology 468 99110. (https://doi.org/10.1007/978-1-59745-249-6_8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Blutt SE, Crawford SE, Ramani S, et al.2018 Engineered human gastrointestinal cultures to study the microbiome and infectious diseases. Cellular and Molecular Gastroenterology and Hepatology 5 241251. (https://doi.org/10.1016/j.jcmgh.2017.12.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Boffa LC, Lupton JR, Mariani MR, et al.1992 Modulation of colonic epithelial cell proliferation, histone acetylation, and luminal short chain fatty acids by variation of dietary fiber (wheat bran) in rats. Cancer Research 52 59065912.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Boustany A, Onwuzo S, Almomani A, et al.2023 Epidemiology and risk of colorectal cancer in patients with a history of Helicobacter pylori infection: a population-based study. Annals of Gastroenterology 36 203207. (https://doi.org/10.20524/aog.2023.0783)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buchon N, Broderick NA, Chakrabarti S, et al.2009a Invasive and indigenous microbiota impact intestinal stem cell activity through multiple pathways in Drosophila. Genes and Development 23 23332344. (https://doi.org/10.1101/gad.1827009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buchon N, Broderick NA, Poidevin M, et al.2009b Drosophila intestinal response to bacterial infection: activation of host defense and stem cell proliferation. Cell Host and Microbe 5 200211. (https://doi.org/10.1016/j.chom.2009.01.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Burclaff J, Bliton RJ, Breau KA, et al.2022 A proximal-to-distal survey of healthy adult human small intestine and colon epithelium by single-cell transcriptomics. Cellular and Molecular Gastroenterology and Hepatology 13 15541589. (https://doi.org/10.1016/j.jcmgh.2022.02.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chang PV, Hao L, Offermanns S, et al.2014 The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. PNAS 111 22472252. (https://doi.org/10.1073/pnas.1322269111)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clevers H 2013 The intestinal crypt, a prototype stem cell compartment. Cell 154 274284. (https://doi.org/10.1016/j.cell.2013.07.004)

  • Clevers HC & & Bevins CL 2013 Paneth cells: maestros of the small intestinal crypts. Annual Review of Physiology 75 289311. (https://doi.org/10.1146/annurev-physiol-030212-183744)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Co JY, Margalef-Catala M, Li X, et al.2019 Controlling epithelial polarity: a human enteroid model for host-pathogen interactions. Cell Reports 26 25092520.e4. (https://doi.org/10.1016/j.celrep.2019.01.108)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Comalada M, Bailon E, de Haro O, et al.2006 The effects of short-chain fatty acids on colon epithelial proliferation and survival depend on the cellular phenotype. Journal of Cancer Research and Clinical Oncology 132 487497. (https://doi.org/10.1007/s00432-006-0092-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Darby TM, Naudin CR, Luo L, et al.2020 Lactobacillus rhamnosus GG-induced expression of leptin in the intestine orchestrates epithelial cell proliferation. Cellular and Molecular Gastroenterology and Hepatology 9 627639. (https://doi.org/10.1016/j.jcmgh.2019.12.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dave M, Higgins PD, Middha S, et al.2012 The human gut microbiome: current knowledge, challenges, and future directions. Translational Research 160 246257. (https://doi.org/10.1016/j.trsl.2012.05.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dheer R & & Young VB 2021 Stem-cell-derived models: tools for studying role of microbiota in intestinal homeostasis and disease. Current Opinion in Gastroenterology 37 1522. (https://doi.org/10.1097/MOG.0000000000000691)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Di Rienzi SC & & Britton RA 2020 Adaptation of the gut microbiota to modern dietary sugars and sweeteners. Advances in Nutrition 11 616629. (https://doi.org/10.1093/advances/nmz118)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Donohoe DR, Garge N, Zhang X, et al.2011 The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metabolism 13 517526. (https://doi.org/10.1016/j.cmet.2011.02.018)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dossa AY, Escobar O, Golden J, et al.2016 Bile acids regulate intestinal cell proliferation by modulating EGFR and FXR signaling. American Journal of Physiology. Gastrointestinal and Liver Physiology 310 G81G92. (https://doi.org/10.1152/ajpgi.00065.2015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dou X, Gao N, Yan D, et al.2020 Sodium butyrate alleviates mouse colitis by regulating gut microbiota dysbiosis. Animals 10. (https://doi.org/10.3390/ani10071154)

  • Duan C, Wu J, Wang Z, et al.2023 Fucose promotes intestinal stem cell-mediated intestinal epithelial development through promoting Akkermansia-related propanoate metabolism. Gut Microbes 15 2233149. (https://doi.org/10.1080/19490976.2023.2233149)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Duckworth CA 2021 Identifying key regulators of the intestinal stem cell niche. Biochemical Society Transactions 49 21632176. (https://doi.org/10.1042/BST20210223)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Durack J & & Lynch SV 2019 The gut microbiome: relationships with disease and opportunities for therapy. Journal of Experimental Medicine 216 2040. (https://doi.org/10.1084/jem.20180448)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fishbein SRS, Mahmud B & & Dantas G 2023 Antibiotic perturbations to the gut microbiome. Nature Reviews. Microbiology. (https://doi.org/10.1038/s41579-023-00933-y)

  • Foulke-Abel J, In J, Kovbasnjuk O, et al.2014 Human enteroids as an ex-vivo model of host-pathogen interactions in the gastrointestinal tract. Experimental Biology and Medicine 239 11241134. (https://doi.org/10.1177/1535370214529398)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Frankel WL, Zhang W, Singh A, et al.1994 Mediation of the trophic effects of short-chain fatty acids on the rat jejunum and colon. Gastroenterology 106 375380. (https://doi.org/10.1016/0016-5085(9490595-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Frese SA, Benson AK, Tannock GW, et al.2011 The evolution of host specialization in the vertebrate gut symbiont Lactobacillus reuteri. PLOS Genetics 7 e1001314. (https://doi.org/10.1371/journal.pgen.1001314)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ge Y, Sun H, Xu L, et al.2022 The amelioration of alcohol-induced liver and intestinal barrier injury by Lactobacillus rhamnosus Gorbach-Goldin (LGG) is dependent on interleukin 22 (IL-22) expression. Bioengineered 13 1265012660. (https://doi.org/10.1080/21655979.2022.2070998)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gehart H & & Clevers H 2019 Tales from the crypt: new insights into intestinal stem cells. Nature Reviews. Gastroenterology and Hepatology 16 1934. (https://doi.org/10.1038/s41575-018-0081-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gordon HA & & Bruckner-Kardoss E 1961 Effect of the normal microbial flora on various tissue elements of the small intestine. Acta Anatomica 44 210225. (https://doi.org/10.1159/000141723)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gregorieff A, Pinto D, Begthel H, et al.2005 Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology 129 626638. (https://doi.org/10.1016/j.gastro.2005.06.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greicius G & & Virshup DM 2019 Stromal control of intestinal development and the stem cell niche. Differentiation; Research in Biological Diversity 108 816. (https://doi.org/10.1016/j.diff.2019.01.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greicius G, Kabiri Z, Sigmundsson K, et al.2018 PDGFRalpha(+) pericryptal stromal cells are the critical source of Wnts and RSPO3 for murine intestinal stem cells in vivo. PNAS 115 E3173E3181. (https://doi.org/10.1073/pnas.1713510115)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greig CJ, Alper A, Goodman AL, et al.2018 Mucosal homeostasis is altered in the ileum of gnotobiotic mice. Journal of Surgical Research 231 331337. (https://doi.org/10.1016/j.jss.2018.05.055)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hall AB, Tolonen AC & & Xavier RJ 2017 Human genetic variation and the gut microbiome in disease. Nature Reviews. Genetics 18 690699. (https://doi.org/10.1038/nrg.2017.63)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hamade DF, Espinal A, Yu J, et al.2022 Lactobacillus reuteri releasing IL-22 (LR-IL-22) facilitates intestinal radioprotection for whole-abdomen irradiation (WAI) of ovarian cancer. Radiation Research 198 89105. (https://doi.org/10.1667/RADE-21-00224.1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hansson GC & & Johansson ME 2010 The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Gut Microbes 1 5154. (https://doi.org/10.4161/gmic.1.1.10470)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hasan N & & Yang H 2019 Factors affecting the composition of the gut microbiota, and its modulation. PeerJ 7 e7502. (https://doi.org/10.7717/peerj.7502)

  • Hemarajata P & & Versalovic J 2013 Effects of probiotics on gut microbiota: mechanisms of intestinal immunomodulation and neuromodulation. Therapeutic Advances in Gastroenterology 6 3951. (https://doi.org/10.1177/1756283X12459294)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Holzhausen EA, Malecki KC, Sethi AK, et al.2022 Assessing the relationship between physical activity and the gut microbiome in a large, population-based sample of Wisconsin adults. PLoS One 17 e0276684. (https://doi.org/10.1371/journal.pone.0276684)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hou Q, Ye L, Liu H, et al.2018 Lactobacillus accelerates ISCs regeneration to protect the integrity of intestinal mucosa through activation of STAT3 signaling pathway induced by LPLs secretion of IL-22. Cell Death and Differentiation 25 16571670. (https://doi.org/10.1038/s41418-018-0070-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hou Y, Wei W, Guan X, et al.2021 A diet-microbial metabolism feedforward loop modulates intestinal stem cell renewal in the stressed gut. Nature Communications 12 271. (https://doi.org/10.1038/s41467-020-20673-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hua Q, Zhang H, Xu R, et al.2023 Lacticaseibacillus casei ATCC334 Ameliorates Radiation-Induced intestinal Injury in Rats by Targeting Microbes and Metabolites. Molecular Nutrition and Food Research 67 e2200337. (https://doi.org/10.1002/mnfr.202200337)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hugenholtz F & & de Vos WM 2018 Mouse models for human intestinal microbiota research: a critical evaluation. Cellular and Molecular Life Sciences 75 149160. (https://doi.org/10.1007/s00018-017-2693-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Human Microbiome Project 2012 Structure, function and diversity of the healthy human microbiome. Nature 486 207214. (https://doi.org/10.1038/nature11234)

  • Ichikawa H & & Sakata T 1997 Effect of L-lactic acid, short-chain fatty acids, and pH in cecal infusate on morphometric and cell kinetic parameters of rat cecum. Digestive Diseases and Sciences 42 15981610. (https://doi.org/10.1023/a:1018884625737)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Inagaki A, Ichikawa H & & Sakata T 2007 Inhibitory effect of succinic acid on epithelial cell proliferation of colonic mucosa in rats. Journal of Nutritional Science and Vitaminology 53 377379. (https://doi.org/10.3177/jnsv.53.377)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Johansson ME, Phillipson M, Petersson J, Velcich A, Holm L & & Hansson GC 2008 The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. PNAS 105 1506415069. (https://doi.org/10.1073/pnas.0803124105)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Johansson ME, Gustafsson JK, Holmen-Larsson J, et al.2014 Bacteria penetrate the normally impenetrable inner colon mucus layer in both murine colitis models and patients with ulcerative colitis. Gut 63 281291. (https://doi.org/10.1136/gutjnl-2012-303207)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jones RM, Luo L, Ardita CS, et al.2013 Symbiotic lactobacilli stimulate gut epithelial proliferation via Nox-mediated generation of reactive oxygen species. EMBO Journal 32 30173028. (https://doi.org/10.1038/emboj.2013.224)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Juge N 2012 Microbial adhesins to gastrointestinal mucus. Trends in Microbiology 20 3039. (https://doi.org/10.1016/j.tim.2011.10.001)

  • Kabiri Z, Greicius G, Madan B, et al.2014 Stroma provides an intestinal stem cell niche in the absence of epithelial Wnts. Development 141 22062215. (https://doi.org/10.1242/dev.104976)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kaiko GE, Ryu SH, Koues OI, et al.2016 The colonic crypt protects stem cells from microbiota-derived metabolites. Cell 165 17081720. (https://doi.org/10.1016/j.cell.2016.05.018)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kastl AJ, Terry NA, Wu GD, et al.2020 The structure and function of the human small intestinal microbiota: current understanding and future directions. Cellular and Molecular Gastroenterology and Hepatology 9 3345. (https://doi.org/10.1016/j.jcmgh.2019.07.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Katoh M 2007 Networking of WNT, FGF, Notch, BMP, and Hedgehog signaling pathways during carcinogenesis. Stem Cell Reviews 3 3038. (https://doi.org/10.1007/s12015-007-0006-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Katoh M & & Katoh M 2007 Wnt signaling pathway and stem cell signaling network. Clinical Cancer Research 13 40424045. (https://doi.org/10.1158/1078-0432.CCR-06-2316)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kawajiri K, Kobayashi Y, Ohtake F, et al.2009 Aryl hydrocarbon receptor suppresses intestinal carcinogenesis in ApcMin/+ mice with natural ligands. PNAS 106 1348113486. (https://doi.org/10.1073/pnas.0902132106)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Khoury KA, Floch MH & & Hersh T 1969 Small intestinal mucosal cell proliferation and bacterial flora in the conventionalization of the germfree mouse. Journal of Experimental Medicine 130 659670. (https://doi.org/10.1084/jem.130.3.659)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kien CL, Blauwiekel R, Bunn JY, et al.2007 Cecal infusion of butyrate increases intestinal cell proliferation in piglets. Journal of Nutrition 137 916922. (https://doi.org/10.1093/jn/137.4.916)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim J & & Lee HK 2021 Potential role of the gut microbiome in colorectal cancer progression. Frontiers in Immunology 12 807648. (https://doi.org/10.3389/fimmu.2021.807648)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim HJ, Lee J, Choi JH, et al.2016 Co-culture of living microbiome with microengineered human intestinal villi in a gut-on-a-chip microfluidic device. Journal of Visualized Experiments 114. (https://doi.org/10.3791/54344)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim S, Shin YC, Kim TY, et al.2021 Mucin degrader Akkermansia muciniphila accelerates intestinal stem cell-mediated epithelial development. Gut Microbes 13 120. (https://doi.org/10.1080/19490976.2021.1892441)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim MB, Hwangbo S, Jang S, et al.2022 Bioengineered Co-culture of organoids to recapitulate host-microbe interactions. Materials Today. Bio 16 100345. (https://doi.org/10.1016/j.mtbio.2022.100345)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Klimashina MM, Viktorovskii IV, Mikhailova NP, et al.1989 Substrate specificity of the biotransformation enzymes in a Nocardia erythropolis culture. Izvestiia Akademii Nauk SSSR. Seriia Biologicheskaia 3 429434.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Korinek V, Barker N, Moerer P, et al.1998 Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nature Genetics 19 379383. (https://doi.org/10.1038/1270)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kozoni V, Tsioulias G, Shiff S, et al.2000 The effect of lithocholic acid on proliferation and apoptosis during the early stages of colon carcinogenesis: differential effect on apoptosis in the presence of a colon carcinogen. Carcinogenesis 21 9991005. (https://doi.org/10.1093/carcin/21.5.999)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kuhnert F, Davis CR, Wang HT, et al.2004 Essential requirement for Wnt signaling in proliferation of adult small intestine and colon revealed by adenoviral expression of Dickkopf-1. Proceedings of the National Academy of Sciences of the United States of America 101 266271. (https://doi.org/10.1073/pnas.2536800100)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kurilshikov A, Medina-Gomez C, Bacigalupe R, et al.2021 Large-scale association analyses identify host factors influencing human gut microbiome composition. Nature Genetics 53 156165. (https://doi.org/10.1038/s41588-020-00763-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lai Y, Masatoshi H, Ma Y, et al.2021 Role of vitamin K in intestinal health. Frontiers in Immunology 12 791565. (https://doi.org/10.3389/fimmu.2021.791565)

  • Lee C, Kim BG, Kim JH, et al.2017 Sodium butyrate inhibits the NF-kappa B signaling pathway and histone deacetylation, and attenuates experimental colitis in an IL-10 independent manner. International Immunopharmacology 51 4756. (https://doi.org/10.1016/j.intimp.2017.07.023)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee YS, Kim TY, Kim Y, et al.2018 Microbiota-derived lactate accelerates intestinal stem-cell-mediated epithelial development. Cell Host and Microbe 24 833846.e6. (https://doi.org/10.1016/j.chom.2018.11.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee C, Choi C, Kang HS, et al.2019 NOD2 supports crypt survival and epithelial regeneration after radiation-induced injury. International Journal of Molecular Sciences 20. (https://doi.org/10.3390/ijms20174297)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lesher S, Walburg HE & & Sacher GA 1964 Generation cycle in the duodenal crypt cells of Germ-free and conventional mice. Nature 202 884886. (https://doi.org/10.1038/202884a0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Levy A, Stedman A, Deutsch E, et al.2020 Innate immune receptor NOD2 mediates LGR5(+) intestinal stem cell protection against ROS cytotoxicity via mitophagy stimulation. PNAS 117 19942003. (https://doi.org/10.1073/pnas.1902788117)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X, Nagy P, Bonfini A, et al.2022 Microbes affect gut epithelial cell composition through immune-dependent regulation of intestinal stem cell differentiation. Cell Reports 38 110572. (https://doi.org/10.1016/j.celrep.2022.110572)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Logan CY & & Nusse R 2004 The Wnt signaling pathway in development and disease. Annual Review of Cell and Developmental Biology 20 781810. (https://doi.org/10.1146/annurev.cellbio.20.010403.113126)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lu X, Xie S, Ye L, et al.2020 Lactobacillus protects against S. Typhimurium-induced intestinal inflammation by determining the fate of epithelial proliferation and differentiation. Molecular Nutrition and Food Research 64 e1900655. (https://doi.org/10.1002/mnfr.201900655)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lyu L, Zhou X, Zhang M, et al.2022 Lactobacillus derived from breast milk facilitates intestinal development in IUGR rats. Journal of Applied Microbiology 133 503514. (https://doi.org/10.1111/jam.15576)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mariadason JM, Velcich A, Wilson AJ, et al.2001 Resistance to butyrate-induced cell differentiation and apoptosis during spontaneous Caco-2 cell differentiation. Gastroenterology 120 889899. (https://doi.org/10.1053/gast.2001.22472)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martinez-Guryn K, Leone V & & Chang EB 2019 Regional diversity of the gastrointestinal microbiome. Cell Host and Microbe 26 314324. (https://doi.org/10.1016/j.chom.2019.08.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Metcalfe C, Kljavin NM, Ybarra R, et al.2014 Lgr5+ stem cells are indispensable for radiation-induced intestinal regeneration. Cell Stem Cell 14 149159. (https://doi.org/10.1016/j.stem.2013.11.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Morris O & & Jasper H 2021 Reactive oxygen species in intestinal stem cell metabolism, fate and function. Free Radical Biology and Medicine 166 140146. (https://doi.org/10.1016/j.freeradbiomed.2021.02.015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Myhrstad MCW, Tunsjo H, Charnock C, et al.2020 Dietary fiber, gut microbiota, and metabolic regulation-current status in human randomized trials. Nutrients 12. (https://doi.org/10.3390/nu12030859)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Naito T, Mulet C, de Castro C, et al.2017 Lipopolysaccharide from crypt-specific core microbiota modulates the colonic epithelial proliferation-to-differentiation balance. mBio 8. (https://doi.org/10.1128/mBio.01680-17)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Neal MD, Sodhi CP, Jia H, et al.2012 Toll-like receptor 4 is expressed on intestinal stem cells and regulates their proliferation and apoptosis via the p53 up-regulated modulator of apoptosis. Journal of Biological Chemistry 287 3729637308. (https://doi.org/10.1074/jbc.M112.375881)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nelson DP & & Mata LJ 1970 Bacterial flora associated with the human gastrointestinal mucosa. Gastroenterology 58 5661. (https://doi.org/10.1016/S0016-5085(7080093-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nigro G, Rossi R, Commere PH, et al.2014 The cytosolic bacterial peptidoglycan sensor Nod2 affords stem cell protection and links microbes to gut epithelial regeneration. Cell Host and Microbe 15 792798. (https://doi.org/10.1016/j.chom.2014.05.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ogura Y, Lala S, Xin W, et al.2003 Expression of NOD2 in Paneth cells: a possible link to Crohn's ileitis. Gut 52 15911597. (https://doi.org/10.1136/gut.52.11.1591)

  • Pai R, Tarnawski AS & & Tran T 2004 Deoxycholic acid activates beta-catenin signaling pathway and increases colon cell cancer growth and invasiveness. Molecular Biology of the Cell 15 21562163. (https://doi.org/10.1091/mbc.e03-12-0894)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Park JH, Kotani T, Konno T, et al.2016 Promotion of intestinal epithelial cell turnover by commensal bacteria: role of short-chain fatty acids. PLoS One 11 e0156334. (https://doi.org/10.1371/journal.pone.0156334)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Peck BCE, Shanahan MT, Singh AP, et al.2017 Gut microbial influences on the mammalian intestinal stem cell niche. Stem Cells International 2017 5604727. (https://doi.org/10.1155/2017/5604727)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pedron T, Mulet C, Dauga C, et al.2012 A crypt-specific core microbiota resides in the mouse colon. mBio 3. (https://doi.org/10.1128/mBio.00116-12)

  • Perler BK, Friedman ES & & Wu GD 2023 The role of the gut microbiota in the relationship between diet and human health. Annual Review of Physiology 85 449468. (https://doi.org/10.1146/annurev-physiol-031522-092054)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pinto D, Gregorieff A, Begthel H, et al.2003 Canonical Wnt signals are essential for homeostasis of the intestinal epithelium. Genes and Development 17 17091713. (https://doi.org/10.1101/gad.267103)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Poletti M, Arnauts K, Ferrante M, et al.2021 Organoid-based models to study the role of host-microbiota interactions in IBD. Journal of Crohn’s and Colitis 15 12221235. (https://doi.org/10.1093/ecco-jcc/jjaa257)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Potten CS 1998 Stem cells in gastrointestinal epithelium: numbers, characteristics and death. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences 353 821830. (https://doi.org/10.1098/rstb.1998.0246)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Powell DN, Swimm A, Sonowal R, et al.2020 Indoles from the commensal microbiota act via the AHR and IL-10 to tune the cellular composition of the colonic epithelium during aging. PNAS 117 2151921526. (https://doi.org/10.1073/pnas.2003004117)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Preidis GA, Saulnier DM, Blutt SE, et al.2012 Probiotics stimulate enterocyte migration and microbial diversity in the neonatal mouse intestine. FASEB Journal 26 19601969. (https://doi.org/10.1096/fj.10-177980)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qiu Y, Jiang Z, Hu S, et al.2017 Lactobacillus plantarum Enhanced IL-22 Production in Natural Killer (NK) cells that protect the integrity of intestinal epithelial cell barrier damaged by enterotoxigenic Escherichia coli. International Journal of Molecular Sciences 18. (https://doi.org/10.3390/ijms18112409)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rajan A, Vela L, Zeng XL, et al.2018 Novel segment- and host-specific patterns of enteroaggregative Escherichia coli adherence to human intestinal enteroids. mBio 9. (https://doi.org/10.1128/mBio.02419-17)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rettedal EA, Gumpert H & & Sommer MO 2014 Cultivation-based multiplex phenotyping of human gut microbiota allows targeted recovery of previously uncultured bacteria. Nature Communications 5 4714. (https://doi.org/10.1038/ncomms5714)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Roager HM & & Licht TR 2018 Microbial tryptophan catabolites in health and disease. Nature Communications 9 3294. (https://doi.org/10.1038/s41467-018-05470-4)

  • Robbe C, Capon C, Coddeville B, et al.2004 Structural diversity and specific distribution of O-glycans in normal human mucins along the intestinal tract. Biochemical Journal 384 307316. (https://doi.org/10.1042/BJ20040605)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Saffarian A, Mulet C, Regnault B, et al.2019 Crypt- and mucosa-associated core microbiotas in humans and their alteration in colon cancer patients. mBio 10. (https://doi.org/10.1128/mBio.01315-19)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sakata T 1987 Stimulatory effect of short-chain fatty acids on epithelial cell proliferation in the rat intestine: a possible explanation for trophic effects of fermentable fibre, gut microbes and luminal trophic factors. British Journal of Nutrition 58 95103. (https://doi.org/10.1079/bjn19870073)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Santos AJM, Lo YH, Mah AT, et al.2018 The intestinal stem cell niche: homeostasis and adaptations. Trends in Cell Biology 28 10621078. (https://doi.org/10.1016/j.tcb.2018.08.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sartor RB 2015 Gut microbiota: optimal sampling of the intestinal microbiota for research. Nature Reviews. Gastroenterology and Hepatology 12 253254. (https://doi.org/10.1038/nrgastro.2015.46)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sato T, Vries RG, Snippert HJ, et al.2009 Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459 262265. (https://doi.org/10.1038/nature07935)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sato T, van Es JH, Snippert HJ, et al.2011 Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 469 415418. (https://doi.org/10.1038/nature09637)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Saulnier DM, Santos F, Roos S, et al.2011 Exploring metabolic pathway reconstruction and genome-wide expression profiling in Lactobacillus reuteri to define functional probiotic features. PLoS One 6 e18783. (https://doi.org/10.1371/journal.pone.0018783)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Savage DC & & Blumershine RV 1974 Surface-surface associations in microbial communities populating epithelial habitats in the murine gastrointestinal ecosystem: scanning electron microscopy. Infection and Immunity 10 240250. (https://doi.org/10.1128/iai.10.1.240-250.1974)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Segerberg-Konttinen M 1988 Malignant tumors found during medicolegal postmortem examinations. American Journal of Forensic Medicine and Pathology 9 357. (https://doi.org/10.1097/00000433-198812000-00019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stearns JC, Lynch MD, Senadheera DB, et al.2011 Bacterial biogeography of the human digestive tract. Scientific Reports 1 170. (https://doi.org/10.1038/srep00170)

  • Tang Q, Jin G, Wang G, et al.2020 Current sampling methods for gut microbiota: a call for more precise devices. Frontiers in Cellular and Infection Microbiology 10 151. (https://doi.org/10.3389/fcimb.2020.00151)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Theodosiou NA & & Tabin CJ 2003 Wnt signaling during development of the gastrointestinal tract. Developmental Biology 259 258271. (https://doi.org/10.1016/s0012-1606(0300185-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tian H, Biehs B, Warming S, et al.2011 A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature 478 255259. (https://doi.org/10.1038/nature10408)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • van der Flier LG & & Clevers H 2009 Stem cells, self-renewal, and differentiation in the intestinal epithelium. Annual Review of Physiology 71 241260. (https://doi.org/10.1146/annurev.physiol.010908.163145)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • van der Post S, Birchenough GMH & & Held JM 2021 NOX1-dependent redox signaling potentiates colonic stem cell proliferation to adapt to the intestinal microbiota by linking EGFR and TLR activation. Cell Reports 35 108949. (https://doi.org/10.1016/j.celrep.2021.108949)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • VanDussen KL, Marinshaw JM, Shaikh N, et al.2015 Development of an enhanced human gastrointestinal epithelial culture system to facilitate patient-based assays. Gut 64 911920. (https://doi.org/10.1136/gutjnl-2013-306651)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vanuytsel T, Senger S, Fasano A, et al.2013 Major signaling pathways in intestinal stem cells. Biochimica et Biophysica Acta 1830 24102426. (https://doi.org/10.1016/j.bbagen.2012.08.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vasapolli R, Schutte K, Schulz C, et al.2019 Analysis of transcriptionally active bacteria throughout the gastrointestinal tract of healthy individuals. Gastroenterology 157 10811092.e3. (https://doi.org/10.1053/j.gastro.2019.05.068)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vuik F, Dicksved J, Lam SY, et al.2019 Composition of the mucosa-associated microbiota along the entire gastrointestinal tract of human individuals. United European Gastroenterology Journal 7 897907. (https://doi.org/10.1177/2050640619852255)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang JY, Mccormack SA, Viar MJ, et al.1991 Stimulation of proximal small intestinal mucosal growth by luminal polyamines. American Journal of Physiology 261 G504G511. (https://doi.org/10.1152/ajpgi.1991.261.3.G504)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang M, Ahrne S, Jeppsson B, et al.2005 Comparison of bacterial diversity along the human intestinal tract by direct cloning and sequencing of 16S rRNA genes. FEMS Microbiology Ecology 54 219231. (https://doi.org/10.1016/j.femsec.2005.03.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang Y, Kim R, Gunasekara DB, et al.2018 Formation of human colonic crypt array by application of chemical gradients across a shaped epithelial monolayer. Cellular and Molecular Gastroenterology and Hepatology 5 113130. (https://doi.org/10.1016/j.jcmgh.2017.10.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Whitehead RH, Young GP & & Bhathal PS 1986 Effects of short chain fatty acids on a new human colon carcinoma cell line (LIM1215). Gut 27 14571463. (https://doi.org/10.1136/gut.27.12.1457)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wilke G, Wang Y, Ravindran S, et al.2020 In vitro culture of Cryptosporidium parvum using stem cell-derived intestinal epithelial monolayers. Methods in Molecular Biology 2052 351372. (https://doi.org/10.1007/978-1-4939-9748-0_20)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Williamson IA, Arnold JW, Samsa LA, et al.2018 A high-throughput organoid microinjection platform to study gastrointestinal microbiota and luminal physiology. Cellular and Molecular Gastroenterology and Hepatology 6 301319. (https://doi.org/10.1016/j.jcmgh.2018.05.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu S, Morin PJ, Maouyo D, et al.2003 Bacteroides fragilis enterotoxin induces c-Myc expression and cellular proliferation. Gastroenterology 124 392400. (https://doi.org/10.1053/gast.2003.50047)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu H, Xie S, Miao J, et al.2020 Lactobacillus reuteri maintains intestinal epithelial regeneration and repairs damaged intestinal mucosa. Gut Microbes 11 9971014. (https://doi.org/10.1080/19490976.2020.1734423)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xing PY, Pettersson S & & Kundu P 2020 Microbial metabolites and intestinal stem cells tune intestinal homeostasis. Proteomics 20 e1800419. (https://doi.org/10.1002/pmic.201800419)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yan KS, Chia LA, Li X, et al.2012 The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proceedings of the National Academy of Sciences of the United States of America 109 466471. (https://doi.org/10.1073/pnas.1118857109)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yin X, Farin HF, van Es JH, et al.2014 Niche-independent high-purity cultures of Lgr5+ intestinal stem cells and their progeny. Nature Methods 11 106112. (https://doi.org/10.1038/nmeth.2737)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zachos NC, Kovbasnjuk O, Foulke-Abel J, et al.2016 Human Enteroids/Colonoids and intestinal organoids functionally recapitulate normal intestinal physiology and pathophysiology. Journal of Biological Chemistry 291 37593766. (https://doi.org/10.1074/jbc.R114.635995)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang Z, Geng J, Tang X, et al.2014 Spatial heterogeneity and co-occurrence patterns of human mucosal-associated intestinal microbiota. ISME Journal 8 881893. (https://doi.org/10.1038/ismej.2013.185)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang X, Tong Y, Lyu X, et al.2020 Prevention and alleviation of dextran sulfate sodium salt-induced inflammatory bowel disease in mice with Bacillus subtilis-fermented milk via inhibition of the inflammatory responses and regulation of the intestinal flora. Frontiers in Microbiology 11 622354. (https://doi.org/10.3389/fmicb.2020.622354)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang W, Zhou Q, Liu H, et al.2023 Bacteroides fragilis strain ZY-312 facilitates colonic mucosa regeneration in colitis via motivating STAT3 signaling pathway induced by IL-22 from ILC3 secretion. Frontiers in Immunology 14 1156762. (https://doi.org/10.3389/fimmu.2023.1156762)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
Get Permissions
  • Figure 1

    Landscape of the intestinal epithelium. The intestinal epithelium is made up of diverse cell types organized into microdomains termed the crypt and villus. The microbiome lies adjacent to the intestinal epithelium in close contact with a mucous layer produced by the epithelium. The villus contains differentiated cells including enterocytes, goblet cells, enteroendocrine cells, and tuft cells. The crypt contains the undifferentiated intestinal stem cells (ISCs) and transient amplifying (TA) cells. The Paneth cells and few enteroendocrine and tuft cells reside alongside the ISC in the crypt. The stromal cells reside directly beneath the epithelium in the crypt region and provide essential regulatory factors to the ISC. Created in BioRender.

  • Figure 2

    Formats of human intestinal organoids. Human intestinal organoids (HIOs) can be used in a variety of formats depending on the scientific questions to be explored. Created in BioRender.

  • Afrizal A, Hitch TCA, Viehof A, et al.2022 Anaerobic single-cell dispensing facilitates the cultivation of human gut bacteria. Environmental Microbiology 24 38613881. (https://doi.org/10.1111/1462-2920.15935)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Andersson-Rolf A, Zilbauer M, Koo BK, et al.2017 Stem cells in repair of gastrointestinal epithelia. Physiology 32 278289. (https://doi.org/10.1152/physiol.00005.2017)

  • Arrieta MC, Walter J & & Finlay BB 2016 Human microbiota-associated mice: a model with challenges. Cell Host and Microbe 19 575578. (https://doi.org/10.1016/j.chom.2016.04.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Auchtung JM, Robinson CD & & Britton RA 2015 Cultivation of stable, reproducible microbial communities from different fecal donors using minibioreactor arrays (MBRAs). Microbiome 3 42. (https://doi.org/10.1186/s40168-015-0106-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Badal VD, Vaccariello ED, Murray ER, et al.2020 The gut microbiome, aging, and longevity: a systematic review. Nutrients 12. (https://doi.org/10.3390/nu12123759)

  • Bai Y, Huang Y, Li Y, et al.2021 The murine Reg3a stimulated by Lactobacillus casei promotes intestinal cell proliferation and inhibits the multiplication of porcine diarrhea causative agent in vitro. Frontiers in Microbiology 12 675263. (https://doi.org/10.3389/fmicb.2021.675263)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Banasaz M, Norin E, Holma R, et al.2002 Increased enterocyte production in gnotobiotic rats mono-associated with Lactobacillus rhamnosus GG. Applied and Environmental Microbiology 68 30313034. (https://doi.org/10.1128/AEM.68.6.3031-3034.2002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barker N, van Es JH, Kuipers J, et al.2007 Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449 10031007. (https://doi.org/10.1038/nature06196)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barker N, Van de Wetering M & & Clevers H 2008 The intestinal stem cell. Genes and Development 22 18561864. (https://doi.org/10.1101/gad.1674008)

  • Barker N, Van Oudenaarden A & & Clevers H 2012 Identifying the stem cell of the intestinal crypt: strategies and pitfalls. Cell Stem Cell 11 452460. (https://doi.org/10.1016/j.stem.2012.09.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bedu-Ferrari C, Biscarrat P, Langella P, et al.2022 Prebiotics and the human gut microbiota: from breakdown mechanisms to the impact on metabolic health. Nutrients 14. (https://doi.org/10.3390/nu14102096)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bevins CL & & Salzman NH 2011 Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis. Nature Reviews. Microbiology 9 356368. (https://doi.org/10.1038/nrmicro2546)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Biechele TL & & Moon RT 2008 Assaying beta-catenin/TCF transcription with beta-catenin/TCF transcription-based reporter constructs. Methods in Molecular Biology 468 99110. (https://doi.org/10.1007/978-1-59745-249-6_8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Blutt SE, Crawford SE, Ramani S, et al.2018 Engineered human gastrointestinal cultures to study the microbiome and infectious diseases. Cellular and Molecular Gastroenterology and Hepatology 5 241251. (https://doi.org/10.1016/j.jcmgh.2017.12.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Boffa LC, Lupton JR, Mariani MR, et al.1992 Modulation of colonic epithelial cell proliferation, histone acetylation, and luminal short chain fatty acids by variation of dietary fiber (wheat bran) in rats. Cancer Research 52 59065912.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Boustany A, Onwuzo S, Almomani A, et al.2023 Epidemiology and risk of colorectal cancer in patients with a history of Helicobacter pylori infection: a population-based study. Annals of Gastroenterology 36 203207. (https://doi.org/10.20524/aog.2023.0783)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buchon N, Broderick NA, Chakrabarti S, et al.2009a Invasive and indigenous microbiota impact intestinal stem cell activity through multiple pathways in Drosophila. Genes and Development 23 23332344. (https://doi.org/10.1101/gad.1827009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buchon N, Broderick NA, Poidevin M, et al.2009b Drosophila intestinal response to bacterial infection: activation of host defense and stem cell proliferation. Cell Host and Microbe 5 200211. (https://doi.org/10.1016/j.chom.2009.01.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Burclaff J, Bliton RJ, Breau KA, et al.2022 A proximal-to-distal survey of healthy adult human small intestine and colon epithelium by single-cell transcriptomics. Cellular and Molecular Gastroenterology and Hepatology 13 15541589. (https://doi.org/10.1016/j.jcmgh.2022.02.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chang PV, Hao L, Offermanns S, et al.2014 The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. PNAS 111 22472252. (https://doi.org/10.1073/pnas.1322269111)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clevers H 2013 The intestinal crypt, a prototype stem cell compartment. Cell 154 274284. (https://doi.org/10.1016/j.cell.2013.07.004)

  • Clevers HC & & Bevins CL 2013 Paneth cells: maestros of the small intestinal crypts. Annual Review of Physiology 75 289311. (https://doi.org/10.1146/annurev-physiol-030212-183744)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Co JY, Margalef-Catala M, Li X, et al.2019 Controlling epithelial polarity: a human enteroid model for host-pathogen interactions. Cell Reports 26 25092520.e4. (https://doi.org/10.1016/j.celrep.2019.01.108)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Comalada M, Bailon E, de Haro O, et al.2006 The effects of short-chain fatty acids on colon epithelial proliferation and survival depend on the cellular phenotype. Journal of Cancer Research and Clinical Oncology 132 487497. (https://doi.org/10.1007/s00432-006-0092-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Darby TM, Naudin CR, Luo L, et al.2020 Lactobacillus rhamnosus GG-induced expression of leptin in the intestine orchestrates epithelial cell proliferation. Cellular and Molecular Gastroenterology and Hepatology 9 627639. (https://doi.org/10.1016/j.jcmgh.2019.12.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dave M, Higgins PD, Middha S, et al.2012 The human gut microbiome: current knowledge, challenges, and future directions. Translational Research 160 246257. (https://doi.org/10.1016/j.trsl.2012.05.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dheer R & & Young VB 2021 Stem-cell-derived models: tools for studying role of microbiota in intestinal homeostasis and disease. Current Opinion in Gastroenterology 37 1522. (https://doi.org/10.1097/MOG.0000000000000691)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Di Rienzi SC & & Britton RA 2020 Adaptation of the gut microbiota to modern dietary sugars and sweeteners. Advances in Nutrition 11 616629. (https://doi.org/10.1093/advances/nmz118)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Donohoe DR, Garge N, Zhang X, et al.2011 The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metabolism 13 517526. (https://doi.org/10.1016/j.cmet.2011.02.018)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dossa AY, Escobar O, Golden J, et al.2016 Bile acids regulate intestinal cell proliferation by modulating EGFR and FXR signaling. American Journal of Physiology. Gastrointestinal and Liver Physiology 310 G81G92. (https://doi.org/10.1152/ajpgi.00065.2015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dou X, Gao N, Yan D, et al.2020 Sodium butyrate alleviates mouse colitis by regulating gut microbiota dysbiosis. Animals 10. (https://doi.org/10.3390/ani10071154)

  • Duan C, Wu J, Wang Z, et al.2023 Fucose promotes intestinal stem cell-mediated intestinal epithelial development through promoting Akkermansia-related propanoate metabolism. Gut Microbes 15 2233149. (https://doi.org/10.1080/19490976.2023.2233149)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Duckworth CA 2021 Identifying key regulators of the intestinal stem cell niche. Biochemical Society Transactions 49 21632176. (https://doi.org/10.1042/BST20210223)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Durack J & & Lynch SV 2019 The gut microbiome: relationships with disease and opportunities for therapy. Journal of Experimental Medicine 216 2040. (https://doi.org/10.1084/jem.20180448)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fishbein SRS, Mahmud B & & Dantas G 2023 Antibiotic perturbations to the gut microbiome. Nature Reviews. Microbiology. (https://doi.org/10.1038/s41579-023-00933-y)

  • Foulke-Abel J, In J, Kovbasnjuk O, et al.2014 Human enteroids as an ex-vivo model of host-pathogen interactions in the gastrointestinal tract. Experimental Biology and Medicine 239 11241134. (https://doi.org/10.1177/1535370214529398)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Frankel WL, Zhang W, Singh A, et al.1994 Mediation of the trophic effects of short-chain fatty acids on the rat jejunum and colon. Gastroenterology 106 375380. (https://doi.org/10.1016/0016-5085(9490595-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Frese SA, Benson AK, Tannock GW, et al.2011 The evolution of host specialization in the vertebrate gut symbiont Lactobacillus reuteri. PLOS Genetics 7 e1001314. (https://doi.org/10.1371/journal.pgen.1001314)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ge Y, Sun H, Xu L, et al.2022 The amelioration of alcohol-induced liver and intestinal barrier injury by Lactobacillus rhamnosus Gorbach-Goldin (LGG) is dependent on interleukin 22 (IL-22) expression. Bioengineered 13 1265012660. (https://doi.org/10.1080/21655979.2022.2070998)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gehart H & & Clevers H 2019 Tales from the crypt: new insights into intestinal stem cells. Nature Reviews. Gastroenterology and Hepatology 16 1934. (https://doi.org/10.1038/s41575-018-0081-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gordon HA & & Bruckner-Kardoss E 1961 Effect of the normal microbial flora on various tissue elements of the small intestine. Acta Anatomica 44 210225. (https://doi.org/10.1159/000141723)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gregorieff A, Pinto D, Begthel H, et al.2005 Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology 129 626638. (https://doi.org/10.1016/j.gastro.2005.06.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greicius G & & Virshup DM 2019 Stromal control of intestinal development and the stem cell niche. Differentiation; Research in Biological Diversity 108 816. (https://doi.org/10.1016/j.diff.2019.01.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greicius G, Kabiri Z, Sigmundsson K, et al.2018 PDGFRalpha(+) pericryptal stromal cells are the critical source of Wnts and RSPO3 for murine intestinal stem cells in vivo. PNAS 115 E3173E3181. (https://doi.org/10.1073/pnas.1713510115)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greig CJ, Alper A, Goodman AL, et al.2018 Mucosal homeostasis is altered in the ileum of gnotobiotic mice. Journal of Surgical Research 231 331337. (https://doi.org/10.1016/j.jss.2018.05.055)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hall AB, Tolonen AC & & Xavier RJ 2017 Human genetic variation and the gut microbiome in disease. Nature Reviews. Genetics 18 690699. (https://doi.org/10.1038/nrg.2017.63)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hamade DF, Espinal A, Yu J, et al.2022 Lactobacillus reuteri releasing IL-22 (LR-IL-22) facilitates intestinal radioprotection for whole-abdomen irradiation (WAI) of ovarian cancer. Radiation Research 198 89105. (https://doi.org/10.1667/RADE-21-00224.1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hansson GC & & Johansson ME 2010 The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Gut Microbes 1 5154. (https://doi.org/10.4161/gmic.1.1.10470)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hasan N & & Yang H 2019 Factors affecting the composition of the gut microbiota, and its modulation. PeerJ 7 e7502. (https://doi.org/10.7717/peerj.7502)

  • Hemarajata P & & Versalovic J 2013 Effects of probiotics on gut microbiota: mechanisms of intestinal immunomodulation and neuromodulation. Therapeutic Advances in Gastroenterology 6 3951. (https://doi.org/10.1177/1756283X12459294)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Holzhausen EA, Malecki KC, Sethi AK, et al.2022 Assessing the relationship between physical activity and the gut microbiome in a large, population-based sample of Wisconsin adults. PLoS One 17 e0276684. (https://doi.org/10.1371/journal.pone.0276684)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hou Q, Ye L, Liu H, et al.2018 Lactobacillus accelerates ISCs regeneration to protect the integrity of intestinal mucosa through activation of STAT3 signaling pathway induced by LPLs secretion of IL-22. Cell Death and Differentiation 25 16571670. (https://doi.org/10.1038/s41418-018-0070-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hou Y, Wei W, Guan X, et al.2021 A diet-microbial metabolism feedforward loop modulates intestinal stem cell renewal in the stressed gut. Nature Communications 12 271. (https://doi.org/10.1038/s41467-020-20673-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hua Q, Zhang H, Xu R, et al.2023 Lacticaseibacillus casei ATCC334 Ameliorates Radiation-Induced intestinal Injury in Rats by Targeting Microbes and Metabolites. Molecular Nutrition and Food Research 67 e2200337. (https://doi.org/10.1002/mnfr.202200337)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hugenholtz F & & de Vos WM 2018 Mouse models for human intestinal microbiota research: a critical evaluation. Cellular and Molecular Life Sciences 75 149160. (https://doi.org/10.1007/s00018-017-2693-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Human Microbiome Project 2012 Structure, function and diversity of the healthy human microbiome. Nature 486 207214. (https://doi.org/10.1038/nature11234)

    • PubMed