925
Views
0
CrossRef citations to date
0
Altmetric
Review

Host-microbiota interaction in intestinal stem cell homeostasis

, , , , &
Article: 2353399 | Received 02 Feb 2024, Accepted 06 May 2024, Published online: 17 May 2024

ABSTRACT

Intestinal stem cells (ISCs) play a pivotal role in gut physiology by governing intestinal epithelium renewal through the precise regulation of proliferation and differentiation. The gut microbiota interacts closely with the epithelium through myriad of actions, including immune and metabolic interactions, which translate into tight connections between microbial activity and ISC function. Given the diverse functions of the gut microbiota in affecting the metabolism of macronutrients and micronutrients, dietary nutrients exert pronounced effects on host-microbiota interactions and, consequently, the ISC fate. Therefore, understanding the intricate host-microbiota interaction in regulating ISC homeostasis is imperative for improving gut health. Here, we review recent advances in understanding host-microbiota immune and metabolic interactions that shape ISC function, such as the role of pattern-recognition receptors and microbial metabolites, including lactate and indole metabolites. Additionally, the diverse regulatory effects of the microbiota on dietary nutrients, including proteins, carbohydrates, vitamins, and minerals (e.g. iron and zinc), are thoroughly explored in relation to their impact on ISCs. Thus, we highlight the multifaceted mechanisms governing host–microbiota interactions in ISC homeostasis. Insights gained from this review provide strategies for the development of dietary or microbiota-based interventions to foster gut health.

1. Introduction

In the intricate ecosystem of the intestines, the dense and diverse community of microbiota plays a pivotal role in regulating various key physiological functions of the host, including intestinal epithelial maturation, modulation of the immune system, and maintenance of metabolic balance.Citation1 The continuous renewal and differentiation of intestinal epithelial cells driven by ISCs in crypts are essential for maintaining the structural integrity and functionality of the intestines. Therefore, elucidating specific mechanisms regulating ISC homeostasis and investigating the interplay between the microbiota and the host in regulating ISC homeostasis are essential for advancing the understanding of gut health and developing targeted interventions for maintaining intestinal homeostasis.

The crypt–villus structure of the intestine is essential for efficient digestion, absorption, and reliable pathogen resistance.Citation2 Continual intestinal epithelial cell self-renewal, supported by ISCs inside the crypt, is imperative for coping with persistent luminal challenges.Citation3 ISCs feed daughter cells into the transit-amplifying compartment, and then TA cells (or progenitor cells) rapidly proliferate and move out of the crypt to differentiate into mature intestinal epithelial cells including absorptive cells (enterocytes and M cells) and secretory cells (Paneth, goblet, enteroendocrine, and tuft cells), each of these cell types carries out unique and specialized functions.Citation4 The ISC niche regulates the proliferation and differentiation of mammalian ISCs.Citation5

Stomach, small intestine, and large intestine harbor distinct microbial communities that participate in tissue homeostasis.Citation6,Citation7 Intestinal microbiota can regulate ISCs through pattern-recognition receptors (PRRs) or by modulating the redox state and oxygen concentration in the intestine.Citation8–11 On the other hand, there is growing evidence that bioactive metabolites derived from the intestinal microbiota, such as lactate, short-chain fatty acids (SCFAs), and secondary bile acids (SBAs), can influence various physiological functions in the host, including ISC activity.Citation12,Citation13 Dietary nutrients can be used as an intervention to control the makeup of the intestinal microbiota, serving not only as a source of energy for ISC metabolism and a means of regulating ISC fate through nutrient-sensing pathways,Citation14–16 but also influence ISC function through interactions between nutrients and the intestinal microbiome.Citation17 Therefore, gaining a deeper understanding of connections between dietary nutrients, intestinal microbiome, and ISCs, and their regulatory mechanisms, may provide new perspectives on strategies for manipulating the intestinal microbiota to promote intestinal health.

ISC-derived organoids containing all main types of epithelial cells mimic physiological functions of intact intestinal epithelium, including nutrient absorption, ion transport, secretion, and mucus production.Citation18–20 This advance has addressed limitations of animal studies and cell lines in understanding diet-host and microbiome-host interactions.Citation21,Citation22 Over the past decade, an increasing number of technologies have been applied to enhance the availability of intestinal organoids, such as 2D culture of organoids,Citation23 enhanced epithelial polarization,Citation24 and co-culture of organoids with intestinal immune cells, intestinal mesenchymal cells, and bacteria.Citation25 In addition, micro-engineered and high-throughput automated organoid culture has enhanced our understanding of the effects and mechanisms of nutrients and microbiomes on ISCs.Citation26 In this study, we summarize the latest progress in understanding the crosstalk between nutrients, hosts, and intestinal microbiota in regulating ISC homeostasis with the aim of elucidating how the intestinal microbiome regulates host intestinal health.

2. Principal signaling mechanisms in ISC fate regulation

2.1. ISCs

The mammalian intestine harbors two populations of ISCs. One is the crypt base columnar cells (CBCs), also called ‘activated ISCs,’ which are intercalated with the granular Paneth cells at the crypt base.Citation27 LGR5 (leucine-rich repeat-containing G-protein-coupled receptor 5) is one of the most prominent target genes of the Wnt signaling pathway and is exclusively expressed in CBCs at the bottom of the crypt.Citation28,Citation29 Lineage-tracing experiments have demonstrated that LGR5+ CBCs meet two criteria for stemness: long-term self-renewal and differentiation into all epithelial lineages.Citation27 Another population of ISC, known as ‘+4 ISCs’ or ‘quiescent ISCs’ is situated at the fourth position above the crypt base, expressing markers such as Hopx, Bmi1, mTert, and Lrig1.Citation30–33 Subsequent studies have shown that + 4 ISCs can undergo rapid proliferation and give rise to active ISCs that promote intestinal epithelial repair when active ISCs are subject to injury conditions.Citation34,Citation35 Thus, some researchers have defined + 4 ISCs as reserve intestinal stem cells (rISCs) because they can replenish the pool of cycling CBC cells as needed; however, this concept remains controversial. Recent studies have shown that the dedifferentiation of absorptive and secretory progenitor cells is the principle means for ISC restoration.Citation36,Citation37()

Figure 1. ISCs and differentiated progeny in the small intestine. (a) Active ISCs feed daughter cells into the transit-amplifying compartment, and TA cells differentiate into mature intestinal epithelial cells, including absorptive and secretory cells. Quiescent ISCs can be converted to active ISCs to promote intestinal epithelial repair. (b) Villus-crypt axis structure of the small intestine. Intensity gradient of the four crucial signaling pathways for ISC maintenance along the villus-crypt axis. This figure was drawn using online Figdraw software (https://www.figdraw.com/#/).

Figure 1. ISCs and differentiated progeny in the small intestine. (a) Active ISCs feed daughter cells into the transit-amplifying compartment, and TA cells differentiate into mature intestinal epithelial cells, including absorptive and secretory cells. Quiescent ISCs can be converted to active ISCs to promote intestinal epithelial repair. (b) Villus-crypt axis structure of the small intestine. Intensity gradient of the four crucial signaling pathways for ISC maintenance along the villus-crypt axis. This figure was drawn using online Figdraw software (https://www.figdraw.com/#/).

2.2. Key signaling pathways for ISC fate determination

Unique characteristics and functions of ISCs depend on a supportive microenvironment that includes Paneth cells, intestinal subepithelial myofibroblasts, and intestinal stromal cells.Citation38 In addition, this microenvironment is regulated by multiple factors, including the endocrine system, intestinal microbes, and enteral dietary nutrients.Citation38 Several signal pathways, such as Wnt, Notch, and BMP from the ISC microenvironment coordinate to control ISC fate and functionCitation18 ().

Figure 2. Essential signaling pathway regulating ISC fate. The principal Wnt, Notch, BMP, and EGF signaling cascades collectively regulate ISC behavior and homeostasis. Further details are provided in the main text. This figure was drawn using online Figdraw software (https://www.figdraw.com/#/).

Figure 2. Essential signaling pathway regulating ISC fate. The principal Wnt, Notch, BMP, and EGF signaling cascades collectively regulate ISC behavior and homeostasis. Further details are provided in the main text. This figure was drawn using online Figdraw software (https://www.figdraw.com/#/).

2.2.1. Wnt signaling

The Wnt signaling pathway plays an essential role in maintaining ISC proliferation and controlling ISC fate. Wnt ligands are produced by both Paneth cells and the intestinal mesenchymal cells.Citation39 The binding of Wnt ligands to the Frizzled-LRP5/LRP6 receptor complex prevents the continuous degradation of β-catenin by a multiprotein ‘destruction complex’ comprising Axin, adenomatous polyposis coli (APC), casein kinase I (CKI), and glycogen synthase kinase 3β (GSK3β).Citation40 Unphosphorylated β-catenin, which is not degraded, accumulates in the cytoplasm and translocates into the nucleus, where it binds to T-cell factor (TCFs, also called lymphoid enhancer factor, LEF) family of transcription factors to regulate expression of target genes that activate ISCs.Citation40 Zinc and ring finger 3 (ZNRF3) and Ring finger protein 43 (RNF43), as target genes of Wnt signaling, translocate to the plasma membrane, where they recognize and induce the ubiquitination and degradation of Frizzled through Dishevelled (Dvl), shutting off Wnt signaling. R-spondin, which is an essential cytokine for intestinal organoid culture in vitro, binds to LGR4/5 and ZNRF3/RNF43 and further induces ubiquitination and degradation of Wnt receptors to reinitiate Wnt signaling.Citation41

Global knockout of Tcf4 in neonatal mice or conditional deletion of Tcf4 in adult mice in the intestinal epithelium contributes to ISC loss, similar to the results observed with overexpression of Dickkopf-related protein 1 (Dkk1), an inhibitor of the Wnt signaling pathway, by adenoviral transfection of the intestinal epithelium or via genetic modification.Citation42,Citation43 These results suggest that Wnt signaling is essential for the development and maintenance of ISCs.Citation44,Citation45

2.2.2. Notch signaling

Notch signaling is important for maintaining the ISC pool and controlling the balance between secretory and absorptive lineages. Direct membrane contact between two cells is essential for the activation of Notch signaling, whereby one cell expresses Notch ligands (such as DLL1 or DLL4) and the other expresses Notch receptors (NOTCH1–4).Citation46 Once membrane-bound Notch receptors and their ligands bind, Notch signaling is activated. Notch receptors undergo γ-secretase mediated cleavage, ultimately releasing the Notch intracellular domain (NICD) into the cytoplasm.Citation47 NICD is then transported into the nucleus and binds to CSL (CBF-1/RBP-Jκ, Su(H), Lag-1) to form a transcriptional activator complex resulting in the expression of Notch target genes.

The hairy and enhancer of split (HES) family genes are main target genes of Notch signaling. HES1, HES5, and HES7 proteins are major HES proteins expressed in the intestinal epithelium.Citation47 Once expressed HES family proteins repress transcription of another basic helix-loop-helix transcription factor, ATOH1. HES1 null embryos develop secretory cell hyperplasia at the expense of absorptive enterocytes, whereas ATOH1 loss leads to the inability to generate secretory-type cells.Citation48–50 Hence, ATOH1 plays a role opposite to that of Notch/HES1 in ISC differentiation. Blocking Notch signaling with a Notch antibody induces secretory lineage hyperplasia by repression of the Wnt signaling, while attenuation of Wnt signaling rescues the phenotype associated with Notch blockade.Citation51 These results indicate that Notch and Wnt signaling jointly regulate ISC activity and differentiation through a negative feedback regulatory mechanism.

2.2.3. EGF signaling

Epidermal growth factor (EGF) is a critical component that drives ISC proliferation. EGF and TGFα (transforming growth factor-alpha) produced by Paneth cells act as ligands for the EGF receptor (EGFR) expressed by CBC stem cells.Citation39 Overactivation of EGF signaling causes an increase in cell division rate of ISCs and, ultimately, could cause cancer.Citation52 Therefore, the activity of this pathway must be tightly regulated. Knockout of Lgr1, which serves as a negative feedback regulator of the EGFR in CBC cells, contributes to duodenal adenomas with significant intestinal crypt expansion, emphasizing the importance of EGF signaling in regulating the rate of intestinal epithelial turnover.Citation31,Citation53 However, EGF signaling appears to be unnecessary for maintaining ISC identity. Blocking EGF signaling leads proliferative ISCs to enter a quiescent state and stops the growth of organoids, whereas restoring EGF signaling enables their reentry into the proliferative state.Citation54

2.2.4. BMP signaling

Bone morphogenetic proteins (BMPs) restrict ISC expansion to maintain intestinal homeostasis and prevent ISC hyperproliferation following damage. BMP ligands bind to type II receptors (BMPRII), leading to the phosphorylation and activation of type I receptors (BMPRI).Citation55 Phosphorylated BMPRI further phosphorylates and activates R-Smads (Smad1, 5, and 8) and forms a complex with a co-Smad (Smad4) to translocate into the nucleus and regulate target gene expression.Citation55,Citation56 BMP2 and BMP4 are the main ligands for BMP receptors in the small intestine.Citation57,Citation58 The BMP signal antagonist Noggin, generated by myofibroblasts and smooth muscle cells in the submucosa, is an essential factor for the culture of intestinal organoids in vitro, thus emphasizing the importance of BMP signaling in maintaining ISC homeostasis.Citation18,Citation59

BMP signals exhibit an increasing concentration gradient along the crypt-villus axis, in contrast to Wnt signaling.Citation60 Overexpression of the BMP inhibitor Noggin and conditional deletion of BMP’s main receptor Bmpr1α lead to crypt expansion.Citation61,Citation62 These findings resemble the phenotype observed in juvenile patients with polyposis and mutations in the BMP pathway.Citation63 An earlier study indicated that BMP signaling inhibits the nuclear accumulation of β-catenin to suppress Wnt signaling via the PTEN-PI3K-AKT pathway.Citation62 Nevertheless, a 2017 study challenged this conclusion by demonstrating that BMP tightly governs ISC expansion via the regulation of stem cell signature genes, including Lgr5, Sox9, and Cdk6, through SMAD-mediated recruitment of HDAC1.Citation64

3. Interplay between host and microbiota in ISC homeostasis

The intestinal lumen and mucosa harbor a variety of microorganisms, including bacteria, fungi, archaea, bacteriophages, and protists, that collectively form the gastrointestinal microbiome. In the last two decades, progress in microbial culture and high-throughput sequencing technologies has significantly enhanced our comprehension of the composition and functionality of the intestinal microbiota. Analysis of bacterial communities in the gastrointestinal tracts of different mammals has revealed a coevolutionary relationship between the bacterial community structure and mammalian lineages, resulting in a mutualistic symbiotic ecological structure. In this section, we provide an overview of the latest advancements in understanding the relationship between the intestinal microbiota and the maintenance of ISC homeostasis.

3.1. Immune function

Intestinal epithelial cells and antigen-presenting cells (such as dendritic cells) express numerous types of PRRs, including Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), which recognize microorganism-associated molecular patterns (MAMPs) in bacteria, including pathogenic bacteria and beneficial symbiotic bacteria.Citation65,Citation66 Several studies have revealed the crosstalk between the intestinal microbiota and ISCs through PRRs.Citation8 In addition, the intestinal microbiota regulates ISCs by modulating the redox state and oxygen concentration in the intestine.Citation9–11

3.1.1. PRRs

3.1.1.1. Microbiota regulation of ISCs through TLRs

Intestinal epithelial TLR signaling plays an essential role in crypt dynamics by altering ISC proliferation and differentiation. Peptidoglycan (PG) and lipoteichoic acid (LTA), from bacteria including Lactobacillus spp., Bifidobacterium spp., and Bacillus subtilis, facilitate Toll-like receptor 2 (TLR2) signaling.Citation67 Lactobacillus rhamnosus GG (LGG) releases LTA to activate TLR2 on macrophages, thereby protecting ISCs from radiation by stimulating macrophages to secrete chemokines and induce migration of prostaglandin E2 (PGE2)-secreting MSCs.Citation68 Hou et al. reported that Bacillus subtilis induces ISCs differentiation through inhibiting the Notch pathway in an LTA-TLR2-dependent manner.Citation69 The expression of Toll-like receptor 4 (TLR4) was first observed in LGR5+ ISCs, and its activation reduced proliferation and increased apoptosis of ISCs via a p53-up-regulated modulator of apoptosis, both in vivo and in ISC-based organoid culture.Citation70 TLR4 also controls ISC fate by modulating Notch and Wnt signaling. TLR4 has been reported to inhibit Wnt signaling by suppressing the activation of the Wnt receptor LRP6 and blocking the protective effect of Wnt3a ligands.Citation71 Additionally, deletion of TLR4 in the intestinal epithelium in mice and in intestinal organoids both lead to increased goblet cells.Citation72 Lipopolysaccharide (LPS) derived from crypt-specific core microbiota (e.g., Acinetobacter, Delftia, and Stenotrophomonas) in mice inhibits ISC proliferation and promotes differentiation of goblet cell lineages in a TLR4-dependent manner, further highlighting the role of microbiota-derived LPS-mediated TLR4 activation of ISCs.Citation73 In conclusion, the evidence presented above strongly suggests interactions among intestinal microbiota, TLR, Wnt, and Notch signaling pathways, impacting the fate of ISCs .

3.1.1.2. Microbiota regulation of ISCs through NLRs

NOD2, a member of the NLR subfamily, plays a critical role in recognizing conserved bacterial peptidoglycan motifs and triggering host immune responses.Citation74 MDP (muramyl dipeptide) is a commonly found peptidoglycan motif in all bacteria and activates NOD2.Citation75 LGR5+ ISCs constitutively express NOD2 at substantially higher levels than Paneth cells within the intestinal crypt,Citation76 and MDP strongly protects ISCs from oxidative stress-mediated cell death and promotes epithelial regeneration via a NOD2-dependent pathway.Citation76 A subsequent study further showed NOD2 facilitates a cytoprotective process by the removal of the lethal excess of ROS molecules through mitochondrial mitophagy.Citation77 This mechanism is activated by synergistic activation of NOD2 and ATG16L1 via a nuclear factor κB (NF-κB)-independent pathway. Another study has demonstrated that NOD2 supports crypt survival and intestinal epithelial regeneration after irradiation-induced ISC damage.Citation78 These results illustrate intestinal microbe-derived molecules trigger ISC survival to promote intestinal epithelial recovery in a NOD2-dependent manner.

3.1.2. Reactive oxygen species and hypoxia

Multiple studies have suggested that indigenous bacteria in the gut interacts synergistically with reactive oxygen species (ROS) in epithelial cells, while exogenous bacteria produces ROS and upregulates the innate gut immune response, leading to increased ROS generation to shape ISC development.Citation79 ROS modulate ISC fate by controlling a cascade of signal responses, including Wnt, BMP, and Notch pathways.Citation80 In addition, ROS autonomously govern various epigenetic changes that impact ISCs. These changes include CpG island methylation, histone acetylation on lysine tails, and deacetylation through SIRTs, all of which contribute to ISC fate.Citation81,Citation82 It has been conceived that at low ROS concentration, ISC remain inactive and undifferentiated, preserving their stem-like properties. Higher ROS levels promote ISC proliferation and differentiation, while an excessive increase in ROS ultimately triggers apoptosis.Citation83–86 Recent studies have also demonstrated that some intestinal symbiotic bacteria, such as Lactobacillus plantarum, stimulates ROS production to enhance ISC proliferation through activation of the Nrf2/Keap1 pathway both in mice and Drosophila .Citation10,Citation86 However, excessive ROS generation shortens Drosophila lifespan.Citation87

The intestinal epithelium can experience prolonged hypoxia, with significant pO2 fluctuations overtime. Such hypoxia can lead to ROS-mediated ISC proliferation. Hypoxia induces ROS production, which stimulates Extracellular regulated kinase 1/2 (ERK1/2) phosphorylation and activates IκB kinase, resulting in the release of NF-κB from IκB and leading to increased hypoxia-inducible factor-1α (HIF-1α) levels.Citation88,Citation89 Similarly, HIF-1α promotes maintenance of gut barrier and ISC growth.Citation90–92 It is well established that HIF-1α and ROS interact to maintain ISC homeostasis.Citation93 Overexpression of HIF-1α functions as a suppressor of ROS production during periods of excessive ISC progeny generation, while HIF-1α knockdown results in higher ROS levels in the Caco-2 intestinal epithelial cells when treated with the hypoxia-inducing agent CoCl2.Citation94

3.2. Metabolic interaction

Recent studies have demonstrated that complex metabolic interactions between the intestinal microbiota, their metabolites, and the host are important for maintaining intestinal homeostasis. Here, we provide a concise overview of key discoveries concerning the impact of intestinal microbiota-derived metabolites on regulating ISC function, aiming to enhance our comprehension of the dynamic equilibrium within the intestines ( and ).

Figure 3. The effects of key microbiota-derived metabolites on ISCs and the pathways that control gut homeostasis. Microbiota-derived metabolites, such as SCFAs, lactate, succinate, indoles and their derivatives, and bile acids, play a crucial role in regulating ISC homeostasis and associated signaling pathways. Further details are provided in the main text. This figure was drawn using online Figdraw software (https://www.figdraw.com/#/).

Figure 3. The effects of key microbiota-derived metabolites on ISCs and the pathways that control gut homeostasis. Microbiota-derived metabolites, such as SCFAs, lactate, succinate, indoles and their derivatives, and bile acids, play a crucial role in regulating ISC homeostasis and associated signaling pathways. Further details are provided in the main text. This figure was drawn using online Figdraw software (https://www.figdraw.com/#/).

Table 1. Effects of intestinal microbiota-derived metabolites on ISCs.

3.2.1. SCFAs, lactate, and succinate

SCFAs, comprising approximately 60% acetate, 25% propionate, and 15% butyrate, are the primary end products resulting from the fermentation of complex carbohydrate fibers by anaerobic symbiotic bacteria in the intestines.Citation114 SCFAs, especially butyrate, serve as an energy source for colonocytes and have various direct or indirect physiological effects on the host, such as the epithelial barrier, immune responses, and energy metabolism. These effects are mediated through their role as ligands for metabolite-sensing G protein-coupled receptors (GPCRs) and histone deacetylases (HDACs) inhibitor.Citation115–117

Acetate, the main final metabolite of carbohydrates, has no impact on the growth, proliferation, or passaging capacity of intestinal organoids under physiological conditions. However, acetate supports the formation, growth, and budding of organoids by inhibition of β-oxidation when acetyl-CoA concentration is low.Citation95,Citation118

Propionate also regulates ISC function. Propionate supplementation reserves chemical injury-induced loss of aISC markers LGR5 and OLFM4 expression.Citation96 Another research revealed that the supplementation of fucose increases the production of propionic acid by Akkermansia muciniphila, which further promotes the stemness of ISCs through a Gpr41/Gpr43-dependent mechanism.Citation97

Butyrate, the least abundant of the three main SCFAs, serves as a significant energy source for colonocytes, stimulating ISC proliferation.Citation115 However, the effects of butyrate on ISC proliferation remain controversial. Yin et al. reported butyrate promotes ISC amplification in intestinal organoids by inhibiting HDAC.Citation99 However, another study has found that butyrate suppresses colonic stem cell proliferation at physiological concentrations in a FOXO3-dependent manner.Citation100 One possible reason for these controversial results is that butyrate exerts varying effects on the intestinal epithelial cells, depending on the specific segment of the intestine being studied.

Intermediate metabolites such as lactate and succinate, which are produced as end products by some intestinal microbes under certain conditions, also regulate ISC function.Citation119 Previous studies have reported that lactate enhances intestinal proliferation in the small intestine and cecum.Citation101,Citation102 Another study further demonstrated that Lactobacillus-derived lactate promotes ISC-mediated intestinal proliferation by activating the Wnt/β-catenin pathway in a GPR81-dependent manner.Citation103 The effect of lactate on ISC-mediated intestinal proliferation may partly explain the positive effects that Lactobacillus has on intestinal homeostasis. The exact effect of succinate on intestinal proliferation and homeostasis remains controversial. Some studies indicated that succinate may inhibit intestinal epithelial proliferation and induce mucosal damage in the colon.Citation104,Citation105 However, Li et al. demonstrated that succinate improves inflammation responses and intestinal barrier function in mice and pigs.Citation106,Citation107 Further experiments treating animals or intestinal organoids with physiological concentrations of succinate in the intestinal lumen would be more conducive to exploring the effects of succinate on ISC function.

3.2.2. Indoles and derivatives

Tryptophan (Trp), an essential aromatic amino acid, is metabolized either by the gut microbiota through the indole pathway or by the host cells through the kynurenine and serotonin pathways.Citation120 Indole and its derivatives modulate intestinal epithelial cell physiology, immune homeostasis, ISC function, and neurotransmission by interacting with the aryl hydrocarbon receptor (AhR) on host cells. Intestinal microbiota such as Escherichia coli, Lactobacillus spp., and Serratia marcescens express tryptophanase, which metabolizes Trp into indoles and their derivatives, including indole‐3‐acetaldehyde, indole‐3‐aldehyde, indole‐3‐acetic-acid, indole‐3‐propionic acid and indoleacrylic acid.Citation121–123 These molecules can activate AhR signaling to directly or indirectly regulate ISC fate.

AhRs are highly expressed in mouse LGR5+ ISCs and play important roles in controlling ISC proliferation.Citation124 Indole-3-carbinol, as an AhR ligand, promotes ISC differentiation into secretory lineages by activating Wnt/β-catenin signaling and suppressing Notch signaling.Citation109 In addition, the regulatory effect of indoles and their derivatives on ISCs partly depends on AhR-driven mechanisms in immune cells.Citation125 Hou et al. also confirmed that Lactobacillus-derived indole-3-aldehyde activates innate lymphoid cells type 3 (ILC3) cells through AhR ligands to produce IL-22, which further increases ISC proliferation in a STAT3-dependent manner.Citation25

3.2.3. Bile acids

Liver cells synthesize primary bile acids (BAs) from cholesterol, which are then released into the bile ducts and small intestine with glycin or taurine-conjugation to facilitate nutrient digestion and absorption.Citation126 The majority of BAs are reabsorbed in the terminal ileum and transported back to the liver, while a smaller portion reaches the colon. In the colon, the resident microbiota produces bile salt hydrolase (BSH) to convert BAs into different SBAs, including deoxycholic acid (DCA) and lithocholic acid (LCA).Citation127 Thus, the composition and structure of colonic microbiota define the BAs signature.Citation128 BAs regulate host metabolism and intestinal barrier function through several host cell receptors, including farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5).Citation126,Citation129

BAs exhibit a paradoxical effect on ISCs, potentially connected to their ecological niche and dosage. Physiological levels of BAs promote ISC-mediated intestinal epithelium regeneration after injury by activating TGR5 signaling, resulting in the activation of Src and Yes-associated protein (YAP) and their target genes.Citation112 In addition, BAs reduce intestinal inflammation, which is dependent on the TGR5.Citation130 These results indicate that BAs have the potential to enhance intestinal barrier function. However, SBAs, especially DCA and LCA, increase colon cancer stemness and invasiveness of colonic epithelial cells by influencing muscarinic 3 receptor (M3R) and Wnt/β-catenin signaling pathways.Citation110,Citation111 In line with this, some studies have reported that a high fat diet (HFD) increases BA concentrations and further activates the BA-FXR axis to induce hyperproliferation of colon crypts.Citation113,Citation131 These results indicate that a basal level of BAs maintains ISC proliferation, whereas HFD-induced higher concentrations of BAs may increase the risk of colorectal cancer, further highlighting the importance of a balanced diet.

3.3. Microbiota-nutrient interaction in regulation of ISC homeostasis

Dietary nutrients affect microbial distribution and alter microbial metabolism, thereby regulating the stability of the intestinal microenvironment. Additionally, the intestinal microbiota influences absorption, metabolism, and utilization of nutrients. Nutritional elements ingested by the body are digested, absorbed, and utilized by the host and intestinal microbiota to regulate intestinal health.

3.3.1. Macronutrients

3.3.1.1. Protein

Protein forms and levels affect the structure and metabolism patterns of the intestinal microbiota, which may further modulate intestinal homeostasis. It was discovered that the microbiota in the porcine small intestine exhibited a preference for utilizing peptides over free amino acids for bacterial protein synthesis in vitro .Citation132 Additional research has indicated that the presence of peptide-bound amino acids contributes to the prevalence of L. amylovorus and metabolic patterns characterized by lactate production, underscoring the impact of amino acid utilization on intestinal microbial distribution.Citation133 The intestinal microbiota actively participates in the digestion, absorption, and metabolism of amino acids in the body, as demonstrated by Dai et al., who used subculture and isotope tracing techniques to illustrate that the intestinal microbiota can utilize dietary amino acids to synthesize bacterial proteins.Citation134,Citation135

Amino acids are essential nutrients that directly regulate ISC homeostasis. The evidence to date suggests that the gut microbiota affects the metabolic fate of amino acids, including glutamate, glutamine, and arginine,Citation134, Citation136, Citation137 which further regulates ISCs function. Notably, L-glutamate, one of the most abundant amino acids, plays a vital role in balancing amino acids in the body and regulating intestinal function.Citation138 L-glutamate has been found to regulate ISC fate through complex mechanisms, involving the EGFR-ERK-mTORC1 pathwayCitation139 and the amplification of β-catenin through the switching of the membrane receptor Frizzled7.Citation140 In Drosophila, L-glutamate stimulates ISC fate through regulating calcineurin and CREB-regulated transcriptional co-activator via Ca2+ signaling.Citation141 Glutamine, another crucial amino acid, serves as the preferred energy substrate for intestinal epithelial cells, promoting ISC activity to accelerate intestinal epithelial regeneration through enhanced Wnt signaling.Citation142 Similarly, L-arginine and L-methionine have been shown to influence ISC function in response to injury, regulating the ISC niche, proliferation, and differentiation balance.Citation143–147 In Drosophila, methionine and its derivative S-adenosylmethionine reduce midgut mitosis by controlling protein synthesis autonomously in ISCs and induction of the JAK/STAT ligand Unpaired 3 non-autonomously in enterocytes (ECs).Citation148 These findings underscore critical roles of amino acids in sustaining intestinal epithelial homeostasis.

3.3.1.2. Carbohydrate

Food rich in dietary fiber alters composition of the intestinal microbiota.Citation149 It can promote colonization of fiber-degrading microbiota and the production of SCFAs, which in turn support intestinal mucosal homeostasis and host health.Citation149 Conversely, dietary fiber deficiency decreases the abundance of fiber-degrading microbiota in the intestine, potentially leading to the generation of mucus-degrading enzymes by the intestinal microbiota, thereby impairing the mucus barrier.Citation150 In addition, hindgut nutrient substrate availability, especially the ratio of carbohydrate/nitrogenous compounds, alters microbe-related SBAs metabolism and modulates intestinal barrier function.Citation151 These studies suggest that carbohydrates, as essential nutritional substrates, modulate the composition and metabolic patterns of the gut microbiota.

Additionally, human milk oligosaccharides (HMO) play an essential role in intestinal development and maturation of neonates.Citation152 Clinical studies demonstrate that fucosylated HMO such as α2’-fucosyllactose supplementation modifies intestinal microbiota profile in infants and adults.Citation153,Citation154 Furthermore, feeding sialylated oligosaccharides to newborn piglets increases intestinal crypt depth and proliferation, and reduces diarrhea rate.Citation155 Interestingly, when specific microbial-colonized mice were fed a Malawian diet along with sialylated oligosaccharide supplementation, early growth and development were improved.Citation156 Notably, this effect cannot be observed in germ-free mice, highlighting the involvement of the intestinal microbiota in the regulation of intestinal growth and development by sialylated oligosaccharides.

3.3.1.3. Lipid

The interaction between intestinal microbiota and saturated fatty acids plays a vital role in host health under various physiological conditions. HFD increases the relative abundances of Clostridium, Turicibacter, and Peptostreptococcaceae, while notably decreasing the relative abundances of Bifidobacterium, Allobaculum, and Bacteroides.Citation157 Supplementation with long-chain saturated fatty acids led to increased relative abundances of total Lactobacillus species and Lactobacillus rhamnosus, which reduced alcohol-induced liver injury in mice.Citation158 Nevertheless, administering a high saturated fatty acid diet to normal mice increases the abundance of hydrogen sulfide-producing Desulfovibrio in fecal samples and colonic permeability, ultimately resulting in mesenteric inflammation.Citation159

The intestinal microbiota affects the availability of lipids in the gut. Germ-free mice fed an HFD have higher levels of lipids in the feces than normal mice.Citation160 Additionally, antibiotic-treated rats exhibited a reduction in lipid content in the lymphoid tissue after being subjected to HFD.Citation161 In Drosophila, HFD induces transient activation of ISCs through modulating the composition of indigenous microbiota.Citation162 The availability of environmental lipids and the fundamental processes of fatty acid metabolism significantly influence ISC function. Various dietary components, such as arachidonic acid, beta-hydroxybutyrate, have been shown to influence various signaling pathways and metabolic programs in ISCs, affecting their self-renewal, differentiation, and susceptibility to tumor formation.Citation163–166 For example, cholesterol has been shown to control ISC differentiation toward the endocrine lineage by modulating Notch signaling in an Hr96-dependent manner.Citation167 In addition, cholesterol has been shown to be necessary for ISC mitosis by acting as a precursor of steroid hormones and its abnormal intracellular trafficking leads to gut dysbiosis in Drosophila.Citation168 Notably, excess delivery of external lipids, fatty acids, or cholesterol through the diet renders mice ISCs more susceptible toward intestinal tumor formation.Citation169

3.3.2. Micronutrients

3.3.2.1. Vitamins

Vitamins are essential cofactors for myriad of enzymes involved in fat and carbohydrate metabolism. Studies have indicated that certain vitamins, when administered in high doses or targeted to the large intestine, can positively impact on the gut microbiome. This includes increasing the abundance of presumed commensals, enhancing microbial diversity and richness, and promoting SCFA production.Citation170

Humans lack the biosynthetic capacity for most vitamins, and these must thus be provided exogenously through diet and synthesis by the intestinal microbiome.Citation171 Previous studies have shown that intestinal microbiota can synthesize vitamins C and K and the B group vitamins.Citation172,Citation173 In monogastric animals, vitamins produced by the intestinal microbiota are primarily absorbed in the colon, whereas dietary vitamins are absorbed in the proximal small intestine.Citation174,Citation175 Vitamin A and its metabolite, retinoic acid, enhance ISC stemness and promote ISC differentiation, respectively,Citation176,Citation177 whereas vitamin B9 rescues the reduction in cell metabolic activity in small intestinal organoids caused by the chemotherapeutic agent methotrexate.Citation178 The effects of 1,25-dihydroxyvitamin D3 (vitamin D3) on ISC function are controversial, with studies reporting contrasting impacts.Citation179,Citation180 Vitamin D receptor in enterocytes in the intestine of Drosophila is essential for ISC proliferation and enteroendocrine cell differentiation.Citation181 Vitamin B7 is also critical for ISC maintenance and tumorigenesis in Drosophila. In particular, biotin provided through the diet or the microbiota is necessary for mitosis and homeostasis maintenance in ISCs and its absence leads to gut dysbiosis in Drosophila .Citation182 These results underscore the vital role of dietary vitamins and microbial metabolism in regulating ISC homeostasis.

3.3.2.2. Minerals

Dietary mineral deficiencies influence the composition and behavior of the intestinal microbiota. For instance, magnesium deficiency alters the composition of the intestinal microbiota and induces anxiety-like behavior,Citation183 while zinc deficiency causes taxonomic alterations and decreases overall species richness and diversity in the cecum of broiler chickens.Citation184 Supplementation of zinc-amino acid conjugates in mice with zinc deficiency during pregnancy rescued the abnormal microbiota composition and gut physiology status.Citation185 Parabacteroides and Lactobacillus show a negative correlation with increased iron stores, while members of the Clostridia class exhibit a positive correlation with iron stores.Citation186 These results indicate that dietary mineral availability directly influences the intestinal microbial composition and metabolism.

Minerals in food can exist in free or bound forms, with free minerals being directly absorbed and bound minerals being released slowly and absorbed by digestive enzymes and the intestinal microbiota. The probiotic Lactobacillus plantarum 299 v increases non-heme dietary iron absorption.Citation187 Dietary supplementation with iron and Vitamin A increases villus height and intestinal surface area in suckling piglets.Citation188 Selenium-enriched Bifidobacterium longum can biotransform inorganic selenium (Na2SeO3) into more bioactive organic selenium forms (e.g., selenomethionine [Se-Met]) for efficient utilization by the host.Citation189 Se-Met notably elevates the population of LGR5+ and PCNA+ cells and concomitantly increases the number of goblet cells, Paneth cells, and absorptive cells compared with deoxynivalenol (DON) treatment alone.Citation190 Additionally, zinc L-aspartate, in particular Zn, enhances ISC activity to safeguard the integrity of the intestinal epithelium against DON by activating the Wnt/β-catenin signaling pathway in in vivo (mouse) and ex vivo (mouse enteroid) models.Citation191 These findings indicate that the intestinal microbiota is essential for the absorption and metabolism of minerals in the intestine, as well as for maintaining ISC homeostasis.

4. Intestinal organoids: a model for investigating the effect of host – microbiota interaction on ISC homeostasis

In 2009, Clevers et al. first established a murine intestinal organoid model using single LGR5+ ISCs from mouse intestinal crypts in vitro .Citation18 Subsequently, the human intestinal organoid model was successfully established by the same group.Citation192 After 15 years, the technology for intestinal organoid cultures derived from ISCs has become increasingly advanced and has been applied to various experimental animals including rats,Citation193 rabbits,Citation194 pigs,Citation195 chickens,Citation196 cows,Citation197 and sheepCitation198 ().

Figure 4. The establishment and engineering improvement of the intestinal organoid model. (a) Flowchart of the establishment of the mammalian intestinal organoid model. Intestinal crypts were isolated from intestinal tissue, and further embedded in Matrigel® with culture medium to form intestinal organoids. (b) Engineering improvement of the intestinal organoid model. (a) 2D organoids; (b) intestinal organoid polarization; (c) co-culture of intestinal organoids with intestinal mesenchymal and immune cells. (d) High-throughput automated organoid culture. Phenotypic analysis, RT-PCR, imaging, single-cell RNA sequencing, and other indicators can be used to evaluate organoid function. This figure was drawn using online Figdraw software (https://www.figdraw.com/#/).

Figure 4. The establishment and engineering improvement of the intestinal organoid model. (a) Flowchart of the establishment of the mammalian intestinal organoid model. Intestinal crypts were isolated from intestinal tissue, and further embedded in Matrigel® with culture medium to form intestinal organoids. (b) Engineering improvement of the intestinal organoid model. (a) 2D organoids; (b) intestinal organoid polarization; (c) co-culture of intestinal organoids with intestinal mesenchymal and immune cells. (d) High-throughput automated organoid culture. Phenotypic analysis, RT-PCR, imaging, single-cell RNA sequencing, and other indicators can be used to evaluate organoid function. This figure was drawn using online Figdraw software (https://www.figdraw.com/#/).

4.1. Advantages of intestinal organoids

Intestinal organoids contain major types of epithelial cells, which can mimic physiological functions such as nutrient absorption, transport and secretion.Citation18–20 Additionally, intestinal organoids can maintain more stable phenotypic and genetic characteristics during continuous passage than intestinal cell lines.Citation199,Citation200 Primary cells and ex vivo xenografts have a low expansion potential and are not amenable to cryopreservation and thawing, preventing their widespread use in mechanistic research.Citation201 Compared to animal models, intestinal organoids are easier to manipulate with shorter culture cycles, and reduced ethical concerns.Citation19 These advantages make them an excellent model for nutritional and microbial research. Intestinal organoids have been widely utilized to study the impact of diet patterns and nutrients on intestinal health, nutrient transport and absorption functions, interactions between the microbiota and host, and location-specific functions of the intestine.

4.2. Limitations of intestinal organoids

Although intestinal organoids simulate the physiological structure of the intestine, some limitations and challenges remain. The 3D geometric architecture and apical membrane face the inside of the organoid structure to prevent direct contact between nutrients, intestinal microbiota, and bioactive and toxic compounds in the apical epithelium.Citation202,Citation203 As organoids grow, the efficiency of nutrient supply and waste removal decreases, and the organoids must be re-fragmented and reseeded. In addition, limited one-week lifespan of organoids is inadequate for robust differentiation into the full spectrum of differentiated cell types found in vivo .Citation204 In addition, conventional organoid models lack mesenchymal cells and immune cells derived from various non-epithelial lineages.Citation205 However, epithelial development, homeostasis, and disease rely on intricate interactions between different cell types to establish and sustain normal intestinal physiological functions, making intestinal organoid model insufficient in mimicking all aspects of intestinal biology. In recent years, various strategies have been developed to overcome these limitations ().

4.3. Engineering improvement of intestinal organoids

4.3.1. Human intestinal/colonic organoids (HIOs/HCOs)

The methods for generating Human pluripotent stem cell (hPSC)-derived small intestinal organoids (HIOs) were first established in 2011,Citation206 enabling research on human development,Citation207 modeling genetic intestinal diseases,Citation208 understanding enteric pathogenesis,Citation209 and elucidating mechanisms of intestinal physiology.Citation210 Helmarth et al. further developed an in vivo HIO engraftment model to generate mature and functional human intestinal tissues,Citation211 while also containing a functional enteric nervous system (ENS)Citation212and immune cells.Citation213 A notable limitation in the widespread adoption of hPSC-derived gastrointestinal organoid technologies is the requirement for initial differentiation of hPSCs and reliance on spontaneous morphogenesis to form detached spheroids.Citation206 To address this challenge, Mayhew et al. introduced a straightforward, reproducible, and scalable approach for generating HIOs through aggregating cryopreservable hPSC-derived mid-hindgut endoderm (MHE) monolayers, significantly enhancing HIO production by approximately tenfold.Citation214 Given the high incidence of diseases that impact the large intestine such as colitis and colon cancer, Wells et al. detailed the differentiation of human colonic organoids (HCOs) from hPSCs through transient activation of BMP signaling. This innovative approach further expands the utility of HCO technology in studying colonic pathologies.Citation215

4.3.2. 2D culture of organoids

Microinjection and mechanical disruption of intestinal organoids into fragments enable direct contact between the apical surface and luminal nutrients and microbes.Citation77,Citation216,Citation217 However, ensuring synchronous exposure and uniform injection volumes poses a significant challenge.Citation218 Therefore, some researchers have established 2D monolayer from intestinal organoids by mechanically disrupting or partially enzymatically dissociating 3D organoids and subsequently seeding organoid fragments into tissue culture plates or Transwells, enabling the study of intestinal epithelial permeability and responses to nutrients and microbiome.Citation219–221 2D monolayers have been employed in a range of studies of intestinal barrier function, nutrient absorption, and pathogenic infections.Citation23,Citation139,Citation222 However, previously reported methods have resulted in slower growth and higher variability between different wells.Citation202 Moreover, production and maturation of 2D monolayers require more single cells and several days of culture.Citation24 To rapidly obtain a confluent and stable monolayer of cells, several parameters, such as seeding density and culture time, need to be standardized.

4.3.3. Epithelial polarization

Another technique to allow direct contact between the apical side of the intestinal organoids and the experimental treatment is to reverse the polarity of the enteroids. Co et al. successfully generated apical-out enteroids for the first time by removing the extracellular matrix proteins and suspension culture.Citation24 Apical-out enteroids still maintain epithelial barrier integrity and functional characteristics of enteroids.Citation24 Additionally, the apical side of the epithelium is readily accessible for interactions with nutrients and pathogens.Citation24 Subsequently, Li et al. applied this method to preserve the epithelial polarity of porcine jejunal enteroids and investigated the interactions between a transmissible gastroenteritis virus and the intestine.Citation223 However, approximately 20% of the organoids failed to preserve epithelial polarity, suggesting phenotypic variability under specific culture conditions.Citation223

4.3.4. Co-culture of organoids with intestinal mesenchymal and immune cells

Recent studies demonstrated that intestinal stem cell niches including intestinal mesenchymal cells,Citation224,Citation225 Paneth cells,Citation39 and immune cellsCitation226,Citation227 play a crucial role in regulating ISC fate. Hou et al. discovered that L-arginine treatment did not directly target ISCs but rather increased ISC function by stimulating the secretion of Wnt2b by CD90+ stromal cells.Citation144 Lepr+ mesenchymal cells surrounding intestinal crypts sense dietary changes and maintain ISC function via the leptin-Igf1 axis.Citation225 L. reuteri D8 accelerated ISC regeneration to maintain the intestinal barrier by inducing lamina propria lymphocyte secretion of IL-22.Citation25 Utilizing an in vitro co-culture model of organoids with immune or mesenchymal cells offers an in-depth and systematic approach to understanding the mechanisms by which luminal active substances, such as nutrients and microbiota, affect ISC activity.

4.3.5. Micro-engineered and high-throughput automated organoid culture

Micro-engineered and high-throughput automated organoid culture technologies have been used to address complex biological problems. An image-based screening platform for organoids cultured from single cells has been developed to characterize the phenotypic landscape of organoid development.Citation26 Organoid microarrays dynamically simulate the functional units of human tissues and organs in vitro by combining microfluidic microarray technology with 3D organoid culture technology.Citation228 Different cells or microorganisms can be added to study cell-cell and cell-microbe interactions.Citation228 Researchers have also developed a microfluidic platform called IFlowPlate that can be used to culture 128 colon organoids in vitro, providing new possibilities for modeling relevant diseases and screening potential therapeutic targets.Citation229

5. Conclusions and perspectives

Intestinal homeostasis is maintained by a dynamic interplay between ISC self-renewal and differentiation, which is directly or indirectly regulated by the ISC niche, enteral microbiota, and nutrients. In this review, we provide a detailed discussion of various mechanisms through which host-microbiota interactions regulate ISC function, including immune function, metabolic interactions, and the interplay between microbiota and dietary nutrients, including macronutrients and micronutrients. Furthermore, we provide a summary of the most recent advances in intestinal organoid modeling techniques and their potential applications in the study of nutrients, microbiota, and intestinal health.

Further research should explore the specific mechanisms by which nutrients modulate ISC fate and how gut microbes mediate these effects. It is worth noting that the gut microbiota may indirectly influence the stem cell niche by modulating other cells, such as the immune and nervous systems.Citation230 Thus, further investigations are needed to explore and improve organoid models, including more accurately mimicking the intestinal microenvironment and enhancing model complexity and diversity to simulate the complexity of interactions between the gut microbiota and the host.

List of abbreviations

ACC1=

Acetyl-CoA-carboxylase

AHR=

Aryl hydrocarbon receptor

APC=

Adenomatous polyposis coli

BAs=

Bile acids

BMPs=

Bone morphogenetic proteins

BSH=

Bile salt hydrolase

CBCs=

Crypt-base columnar cells

CSL=

CBF-1/RBP-Jκ, Su(H), Lag-1

DCA=

Deoxycholic acid

Dkk1=

Dickkopf-related protein 1

DON=

Deoxynivalenol

ECs=

Enterocytes

EEs=

Enteroendocrine cells

EGF=

Epidermal growth factor

EGFR=

Epidermal growth factor receptor

ERK1/2=

Extracellular regulated kinase 1/2

FXR=

Farnesoid X receptor

GCPRs=

G protein-coupled receptors

HADCs=

Histone deacetylases

HCOs=

Human colonic organoids

HES=

Hairy and Enhancer of split

HFD=

High-fat diet

HIF-1α=

Hypoxia-inducible factor-1α

HIOs=

HPSC-derived small intestinal organoids

HMO=

Human milk oligosaccharides

HPSC=

Human pluripotent stem cell

ILC3=

innate lymphoid cells type 3

ISCs=

Intestinal stem cells

LCA=

Lithocholic acid

LGR5=

Leucine-rich repeat-containing G-protein-coupled receptor 5

LPS=

lipopolysaccharide

LTA=

Lipoteichoic acid

M3R=

muscarinic 3 receptor

MAMPs=

Microorganism-associated molecular patterns

MDP=

Muramyl dipeptide

MHE=

Mid-hindgut endoderm

NF-κB=

Nuclear factor κB

NICD=

Notch intracellular domain

NLRs=

Nucleotide-binding oligomerization domain (NOD)-like receptors

PG=

peptidoglycan

PGE2=

Prostaglandin E2

PPARα=

Peroxisome proliferator-activated receptor alpha

PPARδ=

Peroxisome proliferator-activated receptor delta

PRRs=

Pattern recognition receptors

rISCs=

Reserve intestinal stem cells

RNF43=

Ring finger protein 43

ROS=

Reactive oxygen species

SBAs=

Secondary bile acids

Se-Met=

Selenomethionine

SFCAs=

Short-chain fatty acids

TCFs=

T-cell factors

TGFα=

Transforming growth factor-a

TGR5=

Takeda G protein-coupled receptor 5

TLR2=

Toll-like receptor 2

TLR4=

Toll-like receptor 4

TLRs=

Toll-like receptors

Trp=

Tryptophan

YAP=

Yes-associated protein

ZNRF3=

ZNRF3

Author contributions

WZ conceived and designed this review, edited the manuscript, and secured funding for the study. HW wrote the manuscript draft. CM participated in the concept development, edited and revised the manuscript. LX helped collect literature and draw the diagram. KY provided advice on the review structure. LS edited and proofread the manuscript. All authors contributed to the article and approved the final version of this manuscript.

Acknowledgments

We extend our sincere admiration to the researchers in this field and within our laboratories for their unwavering dedication and hard work. We regret that we could not include citations of all the valuable works of scientists in this field owing to space constraints.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data Availability statement

Data sharing is not applicable to this article, as no datasets were generated or analyzed in the current study. were created using Figdraw software (www.figdraw.com).

Additional information

Funding

This work was supported by the National Natural Science Foundation of China [under Grant number 32030104] and the National Key R&D Program of China [under Grant number 2022YFD1300402].

References

  • Mu C, Zhu W. Understanding the relationship between the microbiome and the structure and function of the pig gastrointestinal tract. Understanding gut microbiomes as targets for improving pig gut health. 2022. doi:10.19103/AS.2021.0089.06.
  • Gehart H, Clevers H. Tales from the crypt: new insights into intestinal stem cells. Nat Rev Gastroenterol Hepatol. 2019;16(1):19–26. doi:10.1038/s41575-018-0081-y.
  • Barker N. Adult intestinal stem cells: critical drivers of epithelial homeostasis and regeneration. Nat Rev Mol Cell Biol. 2014;15(1):19–33. doi:10.1038/nrm3721.
  • van der Flier LG, Clevers H. Stem cells, self-renewal, and differentiation in the intestinal epithelium. Annu Rev Physiol. 2009;71(1):241–260. doi:10.1146/annurev.physiol.010908.163145.
  • Santos AJM, Lo Y, Mah AT, Kuo CJ. The intestinal stem cell niche: homeostasis and adaptations. Trends Cell Biol. 2018;28(12):1062–1078. doi:10.1016/j.tcb.2018.08.001.
  • Ohland CL, Jobin C. Microbial activities and intestinal homeostasis: a delicate balance between health and disease. Cell Mol Gastroenterol Hepatol. 2015;1(1):28–40. doi:10.1016/j.jcmgh.2014.11.004.
  • Wu H, Wu E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes. 2012;3(1):4–14. doi:10.4161/gmic.19320.
  • Bansal K, Trinath J, Chakravortty D, Patil SA, Balaji KN. Pathogen-specific TLR2 protein activation programs macrophages to induce wnt-beta-catenin signaling. J Biol Chem. 2011;286(42):37032–37044. doi:10.1074/jbc.M111.260414.
  • Wang K, Zhang T, Dong Q, Nice EC, Huang C, Wei Y. Redox homeostasis: the linchpin in stem cell self-renewal and differentiation. Cell Death Disease. 2013;4(3):e537. doi:10.1038/cddis.2013.50.
  • Hochmuth CE, Biteau B, Bohmann D, Jasper H. Redox regulation by Keap1 and Nrf2 controls intestinal stem cell proliferation in Drosophila. Cell Stem Cell. 2011;8(2):188–199. doi:10.1016/j.stem.2010.12.006.
  • Pral LP, Fachi JL, Corrêa RO, Colonna M, Vinolo MAR. Hypoxia and HIF-1 as key regulators of gut microbiota and host interactions. Trends Immunol. 2021;42(7):604–621. doi:10.1016/j.it.2021.05.004.
  • Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020;30(6):492–506. doi:10.1038/s41422-020-0332-7.
  • Cox TO, Lundgren P, Nath K, Thaiss CA. Metabolic control by the microbiome. Genome Med. 2022;14(1):80–93. doi:10.1186/s13073-022-01092-0.
  • Alonso S, Yilmaz ÖH. Nutritional regulation of intestinal stem cells. Annu Rev Nutr. 2018;38(1):273–301. doi:10.1146/annurev-nutr-082117-051644.
  • Mattila J, Kokki K, Hietakangas V, Boutros M. Stem cell intrinsic hexosamine metabolism regulates intestinal adaptation to nutrient content. Dev Cell. 2018;47(1):112–121. doi:10.1016/j.devcel.2018.08.011.
  • Aliluev A, Tritschler S, Sterr M, Oppenländer L, Hinterdobler J, Greisle T, Irmler M, Beckers J, Sun N, Walch A. et al. Diet-induced alteration of intestinal stem cell function underlies obesity and prediabetes in mice. Nat Metab. 2021;3(9):1202–1216. doi:10.1038/s42255-021-00458-9.
  • Yao C, Gou X, Tian C, Zhou L, Hao R, Wan L, Wang Z, Li M, Tong X. Key regulators of intestinal stem cells: diet, microbiota, and microbial metabolites. J Genet Genomics. 2023;50(10):735–774. doi:10.1016/j.jgg.2022.12.002.
  • Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, van Es JH, Abo A, Kujala P, Peters PJ. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459(7244):262–265. doi:10.1038/nature07935.
  • Zachos NC, Kovbasnjuk O, Foulke-Abel J, In J, Blutt SE, de Jonge HR, Estes MK, Donowitz M. Human Enteroids/Colonoids and intestinal organoids functionally recapitulate normal intestinal physiology and pathophysiology. J Biol Chem. 2016;291(8):3759–3766. doi:10.1074/jbc.R114.635995.
  • Fair KL, Colquhoun J, Hannan NRF. Intestinal organoids for modelling intestinal development and disease. Phil Trans R Soc B. 2018;373(1750):20170217. doi:10.1098/rstb.2017.0217.
  • Yin Y, Guo S, Wan D, Wu X, Yin Y. Enteroids: promising in vitro models for studies of intestinal physiology and nutrition in farm animals. J Agric Food Chem. 2019;67(9):2421–2428. doi:10.1021/acs.jafc.8b06908.
  • Rubert J, Schweiger PJ, Mattivi F, Tuohy K, Jensen KB, Lunardi A. Intestinal organoids: a tool for modelling diet-microbiome-host interactions. Trends Endocrinol Metab. 2020;31(11):848–858. doi:10.1016/j.tem.2020.02.004.
  • Haynes J, Palaniappan B, Tsopmegha E, Sundaram U. Regulation of nutrient and electrolyte absorption in human organoid-derived intestinal epithelial cell monolayers. Transl Res. 2022;248:22–35. doi:10.1016/j.trsl.2022.04.008.
  • Co JY, Margalef-Català M, Li X, Mah AT, Kuo CJ, Monack DM, Amieva MR. Controlling epithelial polarity: a human enteroid model for host-pathogen interactions. Cell Rep. 2019;26(9):2509–2520.e4. doi:10.1016/j.celrep.2019.01.108.
  • Hou Q, Ye L, Liu H, Huang L, Yang Q, Turner JR, Yu Q. Lactobacillus accelerates ISCs regeneration to protect the integrity of intestinal mucosa through activation of STAT3 signaling pathway induced by LPLs secretion of IL-22. Cell Death Differ. 2018;25(9):1657–1670. doi:10.1038/s41418-018-0070-2.
  • Lukonin I, Serra D, Challet Meylan L, Volkmann K, Baaten J, Zhao R, Meeusen S, Colman K, Maurer F, Stadler MB. et al. Phenotypic landscape of intestinal organoid regeneration. Nature. 2020;586(7828):275–280. doi:10.1038/s41586-020-2776-9.
  • Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449(7165):1003–1007. doi:10.1038/nature06196.
  • van de Wetering M, Sancho E, Verweij C, de Lau W, Oving I, Hurlstone A, van der Horn K, Batlle E, Coudreuse D, Haramis A-P. et al. The β-Catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells. Cell. 2002;111(2):241–250. doi:10.1016/s0092-8674(02)01014-0.
  • Van der Flier LG, Sabates-Bellver J, Oving I, Haegebarth A, De Palo M, Anti M, Van Gijn ME, Suijkerbuijk S, Van de Wetering M, Marra G. et al. The intestinal Wnt/TCF signature. Gastroenterology. 2007;132(2):628–632. doi:10.1053/j.gastro.2006.08.039.
  • Montgomery RK, Carlone DL, Richmond CA, Farilla L, Kranendonk MEG, Henderson DE, Baffour-Awuah NY, Ambruzs DM, Fogli LK, Algra S. et al. Mouse telomerase reverse transcriptase (mTert) expression marks slowly cycling intestinal stem cells. Proc Natl Acad Sci USA. 2011;108(1):179–184. doi:10.1073/pnas.1013004108.
  • Powell AE, Wang Y, Li Y, Poulin EJ, Means AL, Washington MK, Higginbotham J, Juchheim A, Prasad N, Levy S. et al. The pan-ErbB negative regulator Lrig1 is an intestinal stem cell marker that functions as a tumor suppressor. Cell. 2012;149(1):146–158. doi:10.1016/j.cell.2012.02.042.
  • Sangiorgi E, Capecchi MR. Bmi1 is expressed in vivo in intestinal stem cells. Nat Genet. 2008;40(7):915–920. doi:10.1038/ng.165.
  • Breault DT, Min IM, Carlone DL, Farilla LG, Ambruzs DM, Henderson DE, Algra S, Montgomery RK, Wagers AJ, Hole N. et al. Generation of mTert-GFP mice as a model to identify and study tissue progenitor cells. Proc Natl Acad Sci U S A. 2008;105(30):10420–10425. doi:10.1073/pnas.0804800105.
  • Yan KS, Chia LA, Li X, Ootani A, Su J, Lee JY, Su N, Luo Y, Heilshorn SC, Amieva MR. et al. The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proc Natl Acad Sci U S A. 2012;109(2):466–471. doi:10.1073/pnas.1118857109.
  • Tian H, Biehs B, Warming S, Leong KG, Rangell L, Klein OD, de Sauvage FJ. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature. 2011;478(7368):255–259. doi:10.1038/nature10408.
  • Murata K, Jadhav U, Madha S, van Es J, Dean J, Cavazza A, Wucherpfennig K, Michor F, Clevers H, Shivdasani RA. et al. Ascl2-dependent cell dedifferentiation drives regeneration of ablated intestinal stem cells. Cell Stem Cell. 2020;26(3):377–390. doi:10.1016/j.stem.2019.12.011.
  • Shivdasani RA, Clevers H, de Sauvage FJ. Tissue regeneration: reserve or reverse? Science. 2021;371(6531):784–786. doi:10.1126/science.abb6848.
  • Hageman JH, Heinz MC, Kretzschmar K, van der Vaart J, Clevers H, Snippert HJG. Intestinal regeneration: regulation by the microenvironment. Dev Cell. 2020;54(4):435–446. doi:10.1016/j.devcel.2020.07.009.
  • Sato T, van Es JH, Snippert HJ, Stange DE, Vries RG, van den Born M, Barker N, Shroyer NF, van de Wetering M, Clevers H. et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature. 2011;469(7330):415–418. doi:10.1038/nature09637.
  • Nusse R, Clevers H. Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell. 2017;169(6):985–999. doi:10.1016/j.cell.2017.05.016.
  • Hao H, Jiang X, Cong F. Control of wnt receptor turnover by R-spondin-ZNRF3/RNF43 signaling module and its dysregulation in cancer. Cancers Basel. 2016;8(6):54–66. doi:10.3390/cancers8060054.
  • Kuhnert F, Davis CR, Wang HT, Chu P, Lee M, Yuan J. et al. Essential requirement for wnt signaling in proliferation of adult small intestine and colon revealed by adenoviral expression of dickkopf-1. Proc Natl Acad Sci U S A. 2004;101(1):266–271. doi:10.1073/pnas.2536800100.
  • Pinto D, Gregorieff A, Begthel H, Clevers H. Canonical wnt signals are essential for homeostasis of the intestinal epithelium. Genes Dev. 2003;17(14):1709–1713. doi:10.1101/gad.267103.
  • Korinek V, Barker N, Moerer P, van Donselaar E, Huls G, Peters PJ, Clevers H. Depletion of epithelial stem-cell compartments in the small intestine of mice lacking tcf-4. Nat Genet. 1998;19(4):379–383. doi:10.1038/1270.
  • van Es JH, Haegebarth A, Kujala P, Itzkovitz S, Koo B-K, Boj SF, Korving J, van den Born M, van Oudenaarden A, Robine S. et al. A critical role for the wnt effector Tcf4 in adult intestinal homeostatic self-renewal. Mol Cellr Biol. 2012;32(10):1918–1927. doi:10.1128/MCB.06288-11.
  • Sancho R, Cremona CA, Behrens A. Stem cell and progenitor fate in the mammalian intestine: notch and lateral inhibition in homeostasis and disease. EMBO Rep. 2015;16(5):571–581. doi:10.15252/embr.201540188.
  • Noah TK, Shroyer NF. Notch in the intestine: regulation of homeostasis and pathogenesis. Annu Rev Physiol. 2013;75(1):263–288. doi:10.1146/annurev-physiol-030212-183741.
  • Yang Q, Bermingham NA, Finegold MJ, Zoghbi HY. Requirement of Math1 for secretory cell lineage commitment in the mouse intestine. Science. 2001;294(5549):2155–2158. doi:10.1126/science.1065718.
  • Jensen J, Pedersen EE, Galante P, Hald J, Heller RS, Ishibashi M, Kageyama R, Guillemot F, Serup P, Madsen OD. et al. Control of endodermal endocrine development by hes-1. Nat Genet. 2000;24(1):36–44. doi:10.1038/71657.
  • Suzuki K, Fukui H, Kayahara T, Sawada M, Seno H, Hiai H, Kageyama R, Okano H, Chiba T. Hes1-deficient mice show precocious differentiation of Paneth cells in the small intestine. Biochem Biophys Res Commun. 2005;328(1):348–352. doi:10.1016/j.bbrc.2004.12.174.
  • Tian H, Biehs B, Chiu C, Siebel CW, Wu Y, Costa M, de Sauvage F, Klein O. Opposing activities of notch and wnt signaling regulate intestinal stem cells and gut homeostasis. Cell Rep. 2015;11(1):33–42. doi:10.1016/j.celrep.2015.03.007.
  • Snippert HJ, Schepers AG, van Es JH, Simons BD, Clevers H. Biased competition between Lgr5 intestinal stem cells driven by oncogenic mutation induces clonal expansion. EMBO Rep. 2014;15(1):62–69. doi:10.1002/embr.201337799.
  • Wong VWY, Stange DE, Page ME, Buczacki S, Wabik A, Itami S, van de Wetering M, Poulsom R, Wright NA, Trotter MWB. et al. Lrig1 controls intestinal stem-cell homeostasis by negative regulation of ErbB signalling. Nat Cell Biol. 2012;14(4):401–408. doi:10.1038/ncb2464.
  • Basak O, Beumer J, Wiebrands K, Seno H, van Oudenaarden A, Clevers H. Induced quiescence of Lgr5+ stem cells in intestinal organoids enables differentiation of hormone-producing enteroendocrine cells. Cell Stem Cell. 2017;20(2):177–190. doi:10.1016/j.stem.2016.11.001.
  • Shi Y, Massagué J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell. 2003;113(6):685–700. doi:10.1016/s0092-8674(03)00432-x.
  • Feng X, Derynck R. Specificity and versatility in tgf-beta signaling through Smads. Annu Rev Cell Dev Biol. 2005;21(1):659–693. doi:10.1146/annurev.cellbio.21.022404.142018.
  • Hardwick JCH, Van Den Brink GR, Bleuming SA, Ballester I, Van Den Brande JMH, Keller JJ, Offerhaus GJA, Van Deventer SJH, Peppelenbosch MP. Bone morphogenetic protein 2 is expressed by, and acts upon, mature epithelial cells in the colon. Gastroenterology. 2004;126(1):111–121. doi:10.1053/j.gastro.2003.10.067.
  • Haramis A, Begthel H, van den Born M, van Es J, Jonkheer S, Offerhaus GJA, Clevers H. De Novo crypt formation and juvenile polyposis on BMP inhibition in mouse intestine. Science. 2004;303(5664):1684–1686. doi:10.1126/science.1093587.
  • Kosinski C, Li VSW, Chan ASY, Zhang J, Ho C, Tsui WY, Chan TL, Mifflin RC, Powell DW, Yuen ST. et al. Gene expression patterns of human colon tops and basal crypts and BMP antagonists as intestinal stem cell niche factors. Proc Natl Acad Sci U S A. 2007;104(39):15418–15423. doi:10.1073/pnas.0707210104.
  • Beumer J, Puschhof J, Yengej FY, Zhao L, Martinez-Silgado A, Blotenburg M, Begthel H, Boot C, van Oudenaarden A, Chen Y-G. et al. BMP gradient along the intestinal villus axis controls zonated enterocyte and goblet cell states. Cell Rep. 2022;38(9):110438–110458. doi:10.1016/j.celrep.2022.110438.
  • Davis H, Irshad S, Bansal M, Rafferty H, Boitsova T, Bardella C, Jaeger E, Lewis A, Freeman-Mills L, Giner FC. et al. Aberrant epithelial GREM1 expression initiates colonic tumorigenesis from cells outside the stem cell niche. Nat Med. 2015;21(1):62–70. doi:10.1038/nm.3750.
  • He X, Zhang J, Tong W, Tawfik O, Ross J, Scoville DH, Tian Q, Zeng X, He X, Wiedemann LM. et al. BMP signaling inhibits intestinal stem cell self-renewal through suppression of wnt–β-catenin signaling. Nat Genet. 2004;36(10):1117–1121. doi:10.1038/ng1430.
  • Ma H, Brosens LAA, Offerhaus GJA, Giardiello FM, de Leng WWJ, Montgomery EA. Pathology and genetics of hereditary colorectal cancer. Pathology. 2018;50(1):49–59. doi:10.1016/j.pathol.2017.09.004.
  • Qi Z, Li Y, Zhao B, Xu C, Liu Y, Li H, Zhang B, Wang X, Yang X, Xie W. et al. BMP restricts stemness of intestinal Lgr5+ stem cells by directly suppressing their signature genes. Nat Commun. 2017;8(1):13824–13828. doi:10.1038/ncomms13824.
  • Hou Q, Ye L, Huang L, Yu Q. The research progress on intestinal stem cells and its relationship with intestinal microbiota. Front Immunol. 2017;8:599–608. doi:10.3389/fimmu.2017.00599.
  • Mu C, Yang Y, Zhu W. Crosstalk between the immune receptors and gut microbiota. Curr Protein Pept Sci. 2015;16(7):622–631. doi:10.2174/1389203716666150630134356.
  • Lebeer S, Vanderleyden J, De Keersmaecker SCJ. Host interactions of probiotic bacterial surface molecules: comparison with commensals and pathogens. Nat Rev Microbiol. 2010;8(3):171–184. doi:10.1038/nrmicro2297.
  • Riehl TE, Alvarado D, Ee X, Zuckerman A, Foster L, Kapoor V, Thotala D, Ciorba MA, Stenson WF. Lactobacillus rhamnosus GG protects the intestinal epithelium from radiation injury through release of lipoteichoic acid, macrophage activation and the migration of mesenchymal stem cells. Gut. 2019;68(6):1003–1013. doi:10.1136/gutjnl-2018-316226.
  • Hou Q, Jia J, Lin J, Zhu L, Xie S, Yu Q, Li Y. Bacillus subtilis programs the differentiation of intestinal secretory lineages to inhibit salmonella infection. Cell Rep. 2022;40(13):111416. doi:10.1016/j.celrep.2022.111416.
  • Neal MD, Sodhi CP, Jia H, Dyer M, Egan CE, Yazji I, Good M, Afrazi A, Marino R, Slagle D. et al. Toll-like receptor 4 is expressed on intestinal stem cells and regulates their proliferation and apoptosis via the p53 up-regulated modulator of apoptosis. J Biol Chem. 2012;287(44):37296–37308. doi:10.1074/jbc.M112.375881.
  • Yi H, Patel AK, Sodhi CP, Hackam DJ, Hackam AS, Rodrigues MM. Novel role for the innate immune receptor Toll-like receptor 4 (TLR4) in the regulation of the wnt signaling pathway and photoreceptor apoptosis. PLos One. 2012;7(5):e36560. doi:10.1371/journal.pone.0036560.
  • Sodhi CP, Neal MD, Siggers R, Sho S, Ma C, Branca MF, Prindle T, Russo AM, Afrazi A, Good M. et al. Intestinal epithelial Toll-like receptor 4 regulates goblet cell development and is required for necrotizing enterocolitis in mice. Gastroenterology. 2012;143(3):708–718. doi:10.1053/j.gastro.2012.05.053.
  • Naito T, Mulet C, De Castro C, Molinaro A, Saffarian A, Nigro G, Bérard M, Clerc M, Pedersen AB, Sansonetti PJ. et al. Lipopolysaccharide from crypt-specific core microbiota modulates the colonic epithelial proliferation-to-differentiation balance. mBio. 2017;8(5):e01680–17. doi:10.1128/mBio.01680-17.
  • Petnicki-Ocwieja T, Hrncir T, Liu Y, Biswas A, Hudcovic T, Tlaskalova-Hogenova H, Kobayashi KS. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc Natl Acad Sci U S A. 2009;106(37):15813–15818. doi:10.1073/pnas.0907722106.
  • Girardin SE, Boneca IG, Viala J, Chamaillard M, Labigne A, Thomas G, Philpott DJ, Sansonetti PJ. Nod2 is a general sensor of peptidoglycan through muramyl dipeptide (MDP) detection. J Biol Chem. 2003;278(11):8869–8872. doi:10.1074/jbc.C200651200.
  • Nigro G, Rossi R, Commere P, Jay P, Sansonetti PJ. The cytosolic bacterial peptidoglycan sensor Nod2 affords stem cell protection and links microbes to gut epithelial regeneration. Cell Host Microbe. 2014;15(6):792–798. doi:10.1016/j.chom.2014.05.003.
  • Levy A, Stedman A, Deutsch E, Donnadieu F, Virgin HW, Sansonetti PJ, Nigro G. Innate immune receptor NOD2 mediates LGR5+ intestinal stem cell protection against ROS cytotoxicity via mitophagy stimulation. Proc Natl Acad Sci U S A. 2020;117(4):1994–2003. doi:10.1073/pnas.1902788117.
  • Lee C, Choi C, Kang HS, Shin S, Kim S, Park HC, Hong SN. NOD2 supports crypt survival and epithelial regeneration after radiation-induced injury. Int J Mol Sci. 2019;20(17):4297–4310. doi:10.3390/ijms20174297.
  • Lee K, Kim S, Kim E, Ha E, You H, Kim B, Kim M-J, Kwon Y, Ryu J-H, Lee W-J. et al. Bacterial-derived uracil as a modulator of mucosal immunity and gut-microbe homeostasis in Drosophila. Cell. 2013;153(4):797–811. doi:10.1016/j.cell.2013.04.009.
  • Radisky DC, Levy DD, Littlepage LE, Liu H, Nelson CM, Fata JE, Leake D, Godden EL, Albertson DG, Angela Nieto M. et al. Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability. Nature. 2005;436(7047):123–127. doi:10.1038/nature03688.
  • Bigarella CL, Liang R, Ghaffari S. Stem cells and the impact of ROS signaling. Development. 2014;141(22):4206–4218. doi:10.1242/dev.107086.
  • Nath A, Chakrabarti P, Sen S, Barui A. Reactive oxygen species in modulating intestinal stem cell dynamics and function. Stem Cell Rev Rep. 2022;18(7):2328–2350. doi:10.1007/s12015-022-10377-1.
  • Burtenshaw D, Hakimjavadi R, Redmond EM, Cahill CP. Nox, reactive oxygen species and regulation of vascular cell fate. Antioxidants (Basel). 2017;6(4):90–112. doi:10.3390/antiox6040090.
  • Bhattacharyya A, Chattopadhyay R, Mitra S, Crowe SE. Oxidative stress: an essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol Rev. 2014;94(2):329–354. doi:10.1152/physrev.00040.2012.
  • Morris O, Jasper H. Reactive oxygen species in intestinal stem cell metabolism, fate and function. Free Radic Biol Med. 2021;166:140–146. doi:10.1016/j.freeradbiomed.2021.02.015.
  • Jones RM, Luo L, Ardita CS, Richardson AN, Kwon YM, Mercante JW, Alam A, Gates CL, Wu H, Swanson PA. et al. Symbiotic lactobacilli stimulate gut epithelial proliferation via Nox-mediated generation of reactive oxygen species. EMBO J. 2013;32(23):3017–3028. doi:10.1038/emboj.2013.224.
  • Iatsenko I, Boquete J, Lemaitre B. Microbiota-derived lactate activates production of reactive oxygen species by the intestinal NADPH oxidase Nox and shortens Drosophila Lifespan. Immunity. 2018;49(5):929–942. doi:10.1016/j.immuni.2018.09.017.
  • Lamberti MJ, Pansa MF, Vera RE, Fernández-Zapico ME, Rumie Vittar NB, Rivarola VA, Karhausen J. Transcriptional activation of HIF-1 by a ROS-ERK axis underlies the resistance to photodynamic therapy. PLoS One. 2017;12(5):e0177801. doi:10.1371/journal.pone.0177801.
  • Minatel IO, Francisqueti FV, Corrêa CR, Lima GPP. Antioxidant activity of γ-oryzanol: a complex network of Interactions. Int J Mol Sci. 2016;17(8):1107–1122. doi:10.3390/ijms17081107.
  • Walaas GA, Gopalakrishnan S, Bakke I, Skovdahl HK, Flatberg A, Østvik AE, Sandvik AK, Bruland T. Physiological hypoxia improves growth and functional differentiation of human intestinal epithelial organoids. Front Immunol. 2023;14:1095812. doi:10.3389/fimmu.2023.1095812.
  • Wang RX, Henen MA, Lee JS, Vögeli B, Colgan SP. Microbiota-derived butyrate is an endogenous HIF prolyl hydroxylase inhibitor. Gut Microbes. 2021;13(1):1938380. doi:10.1080/19490976.2021.1938380.
  • Kim YI, Yi EJ, Kim YD, Lee AR, Chung J, Ha HC, Cho JM, Kim S-R, Ko H-J, Cheon J-H. et al. Local stabilization of hypoxia-inducible factor-1α controls intestinal inflammation via enhanced gut barrier function and immune regulation. Front Immunol. 2020;11:609689. doi:10.3389/fimmu.2020.609689.
  • Shao Y, Wang K, Xiong X, Liu H, Zhou J, Zou L, Qi M, Liu G, Huang R, Tan Z. et al. The landscape of interactions between hypoxia-inducible factors and reactive oxygen species in the gastrointestinal tract. Oxid Med Cell Longev. 2021;2021(1):1–9. doi:10.1155/2021/8893663.
  • Liu Y, Wang C, Wang Y, Ma Z, Xiao J, McClain C, Li X, Feng W. Cobalt chloride decreases fibroblast growth factor-21 expression dependent on oxidative stress but not hypoxia-inducible factor in caco-2 cells. Toxicol Appl Pharmacol. 2012;264(2):212–221. doi:10.1016/j.taap.2012.08.00.
  • Stine RR, Sakers AP, TeSlaa T, Kissig M, Stine ZE, Kwon CW, Cheng L, Lim H-W, Kaestner KH, Rabinowitz JD. et al. PRDM16 maintains homeostasis of the intestinal epithelium by controlling Region-Specific Metabolism. Cell Stem Cell. 2019;25(6):830–845. doi:10.1016/j.stem.2019.08.017.
  • Bajic D, Niemann A, Hillmer A-K, Mejias-Luque R, Bluemel S, Docampo M, Funk MC, Tonin E, Boutros M, Schnabl B. et al. Gut microbiota-derived propionate regulates the expression of Reg3 mucosal lectins and ameliorates experimental colitis in mice. J Crohns Colitis. 2020;14(10):1462–1472. doi:10.1093/ecco-jcc/jjaa065.
  • Duan C, Wu J, Wang Z, Tan C, Hou L, Qian W, Han C, Hou X. Fucose promotes intestinal stem cell-mediated intestinal epithelial development through promoting Akkermansia-related propanoate metabolism. Gut Microbes. 2023;15(1):2233149. doi:10.1080/19490976.2023.2233149.
  • Bartram HP, Scheppach W, Schmid H, Hofmann A, Dusel G, Richter F, Richter A, Kasper H. Proliferation of human colonic mucosa as an intermediate biomarker of carcinogenesis: effects of butyrate, deoxycholate, calcium, ammonia, and pH. Cancer Res. 1993;53(14):3283–3288. doi:10.1016/j.aap.2005.11.012.
  • Yin X, Farin HF, van Es JH, Clevers H, Langer R, Karp JM. Niche-independent high-purity cultures of Lgr5+ intestinal stem cells and their progeny. Nat Methods. 2014;11(1):106–112. doi:10.1038/nmeth.2737.
  • Kaiko GE, Ryu SH, Koues OI, Collins PL, Solnica-Krezel L, Pearce EJ, Pearce EL, Oltz EM, Stappenbeck TS. The colonic crypt protects stem cells from microbiota-derived metabolites. Cell. 2016;165(7):1708–1720. doi:10.1016/j.cell.2016.05.018.
  • Ichikawa H, Sakata T. Effect of L-lactic acid, short-chain fatty acids, and pH in cecal infusate on morphometric and cell kinetic parameters of rat cecum. Dig Dis Sci. 1997;42(8):1598–1610. doi:10.1023/a:1018884625737.
  • Okada T, Fukuda S, Hase K, Nishiumi S, Izumi Y, Yoshida M, Hagiwara T, Kawashima R, Yamazaki M, Oshio T. et al. Microbiota-derived lactate accelerates colon epithelial cell turnover in starvation-refed mice. Nat Commun. 2013;4(1):1654. doi:10.1038/ncomms2668.
  • Lee Y, Kim T, Kim Y, Lee S, Kim S, Kang SW, Yang J-Y, Baek I-J, Sung YH, Park Y-Y. et al. Microbiota-derived lactate accelerates intestinal stem-cell-mediated epithelial development. Cell Host Microbe. 2018;24(6):833–846.e6. doi:10.1016/j.chom.2018.11.002.
  • Fukui S, Shimoyama T, Tamura K, Yamamura M, Satomi M. Mucosal blood flow and generation of superoxide in rat experimental colitis induced by succinic acid. J Gastroenterol. 1997;32(4):464–471. doi:10.1007/BF02934084.
  • Inagaki A, Ichikawa H, Sakata T. Inhibitory effect of succinic acid on epithelial cell proliferation of colonic mucosa in rats. J Nutr Sci Vitaminol (Tokyo). 2007;53(4):377–379. doi:10.3177/jnsv.53.377.
  • Li X, Huang G, Zhang Y, Ren Y, Zhang R, Zhu W, Yu K. Succinate signaling attenuates high-fat diet-induced metabolic disturbance and intestinal barrier dysfunction. Pharmacol Res. 2023;194:106865. doi:10.1016/j.phrs.2023.106865.
  • Li X, Mao M, Zhang Y, Yu K, Zhu W. Succinate modulates intestinal barrier function and inflammation response in pigs. Biomolecules. 2019;9(9):486–500. doi:10.3390/biom9090486.
  • Venkatesh M, Mukherjee S, Wang H, Li H, Sun K, Benechet AP, Qiu Z, Maher L, Redinbo M, Phillips R. et al. Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity. 2014;41(2):296–310. doi:10.1016/j.immuni.2014.06.014.
  • Park J, Lee J, Lee E, Hwang W, Kim D. Indole-3-Carbinol promotes goblet-cell differentiation regulating Wnt and Notch signaling pathways AhR-dependently. Mol Cells. 2018;41(6):290–300. doi:10.14348/molcells.2018.2167.
  • Pai R, Tarnawski AS, Tran T. Deoxycholic acid activates beta-catenin signaling pathway and increases colon cell cancer growth and invasiveness. Mol Biol Cell. 2004;15(5):2156–2163. doi:10.1091/mbc.e03-12-0894.
  • Farhana L, Nangia-Makker P, Arbit E, Shango K, Sarkar S, Mahmud H, Hadden T, Yu Y, Majumdar APN. Bile acid: a potential inducer of colon cancer stem cells. Stem Cell Res Ther. 2016;7(1):181–191. doi:10.1186/s13287-016-0439-4.
  • Sorrentino G, Perino A, Yildiz E, El Alam G, Bou Sleiman M, Gioiello A, Pellicciari R, Schoonjans K. Bile acids signal via TGR5 to activate intestinal stem cells and epithelial regeneration. Gastroenterology. 2020;159(3):956–968.e8. doi:10.1053/j.gastro.2020.05.067.
  • Fu T, Coulter S, Yoshihara E, Oh TG, Fang S, Cayabyab F, Zhu Q, Zhang T, Leblanc M, Liu S. et al. FXR regulates intestinal cancer stem cell proliferation. Cell. 2019;176(5):1098–1112. doi:10.1016/j.cell.2019.01.036.
  • Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. 2016;165(6):1332–1345. doi:10.1016/j.cell.2016.05.041.
  • Donohoe DR, Garge N, Zhang X, Sun W, O’Connell TM, Bunger MK, Bultman S. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011;13(5):517–526. doi:10.1016/j.cmet.2011.02.018.
  • Liu H, Wang J, He T, Becker S, Zhang G, Li D, Ma X. Butyrate: a double-edged sword for health? Adv Nutr. 2018;9(1):21–29. doi:10.1093/advances/nmx009.
  • Yang W, Yu T, Huang X, Bilotta AJ, Xu L, Lu Y, Sun J, Pan F, Zhou J, Zhang W. et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat Commun. 2020;11(1):4457. doi:10.1038/s41467-020-18262-6.
  • Chen L, Vasoya RP, Toke NH, Parthasarathy A, Luo S, Chiles E, Flores J, Gao N, Bonder EM, Su X. et al. HNF4 regulates fatty acid oxidation and is required for renewal of intestinal stem cells in mice. Gastroenterology. 2020;158(4):985–999. doi:10.1053/j.gastro.2019.11.031.
  • Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol. 2014;12(10):661–672. doi:10.1038/nrmicro3344.
  • Gao K, Mu C, Farzi A, Zhu W. Tryptophan metabolism: a link between the gut microbiota and brain. Adv Nutr. 2020;11(3):709–723. doi:10.1093/advances/nmz127.
  • Agus A, Planchais J, Sokol H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe. 2018;23(6):716–724. doi:10.1016/j.chom.2018.05.003.
  • Wlodarska M, Luo C, Kolde R, d’Hennezel E, Annand JW, Heim CE, Krastel P, Schmitt EK, Omar AS, Creasey EA. et al. Indoleacrylic acid produced by commensal peptostreptococcus species suppresses inflammation. Cell Host Microbe. 2017;22(1):25–37. doi:10.1016/j.chom.2017.06.007.
  • Lamas B, Richard ML, Leducq V, Pham H-P, Michel M-L, Da Costa G, Bridonneau C, Jegou S, Hoffmann TW, Natividad JM. et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med. 2016;22(6):598–605. doi:10.1038/nm.4102.
  • Park J, Choi A, Kim SJ, Cheong SW, Jeong S. AhR activation by 6-formylindolo[3,2-b]carbazole and 2,3,7,8-tetrachlorodibenzo-p-dioxin inhibit the development of mouse intestinal epithelial cells. Environ Toxicol Pharmacol. 2016;43:44–53. doi:10.1016/j.etap.2016.02.007.
  • Cervantes-Barragan L, Chai JN, Tianero MD, Di Luccia B, Ahern PP, Merriman J, Cortez VS, Caparon MG, Donia MS, Gilfillan S. et al. Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science. 2017;357(6353):806–810. doi:10.1126/science.aah5825.
  • Hegyi P, Maléth J, Walters JR, Hofmann AF, Keely SJ. Guts and Gall: bile acids in regulation of intestinal epithelial function in health and disease. Physiol Rev. 2018;98(4):1983–2023. doi:10.1152/physrev.00054.2017.
  • Ferrebee CB, Dawson PA. Metabolic effects of intestinal absorption and enterohepatic cycling of bile acids. Acta Pharm Sin B. 2015;5(2):129–134. doi:10.1016/j.apsb.2015.01.001.
  • Ridlon JM, Kang DJ, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. J Lipid Res. 2006;47(2):241–259. doi:10.1194/jlr.R500013-JLR200.
  • Xiang J, Zhang Z, Xie H, Zhang C, Bai Y, Cao H, Che Q, Guo J, Su Z. Effect of different bile acids on the intestine through enterohepatic circulation based on FXR. Gut Microbes. 2021;13(1):1949095. doi:10.1080/19490976.2021.1949095.
  • Sinha SR, Haileselassie Y, Nguyen LP, Tropini C, Wang M, Becker LS, Sim D, Jarr K, Spear ET, Singh G. et al. Dysbiosis-induced secondary bile acid deficiency promotes intestinal inflammation. Cell Host Microbe. 2020;27(4):659–670.e5. doi:10.1016/j.chom.2020.01.021.
  • Kim T, Kim S, Kim Y, Lee Y, Lee S, Lee S, Kweon M-N. A high-fat diet activates the BAs-FXR Axis and triggers cancer-associated fibroblast properties in the colon. Cell Mol Gastroenterol Hepatol. 2022;13(4):1141–1159. doi:10.1016/j.jcmgh.2021.12.015.
  • Liu J, Mu C, Yu K, Zhu W. Effect of two different casein hydrolysates on small intestinal bacteria of growing pigs. Acta Microbiol Sin. 2018;58:63–72. doi:10.13343/j.cnki.wsxb.20170024.
  • Jing Y, Mu C, Wang H, Shen J, Zoetendal EG, Zhu W. Amino acid utilization allows intestinal dominance of Lactobacillus amylovorus. ISME J. 2022;16(11):2491–2502. doi:10.1038/s41396-022-01287-8.
  • Dai Z, Zhang J, Wu G, Zhu W. Utilization of amino acids by bacteria from the pig small intestine. Amino Acids. 2010;39(5):1201–1215. doi:10.1007/s00726-010-0556-9.
  • Dai Z, Li X, Xi P, Zhang J, Wu G, Zhu W. Metabolism of select amino acids in bacteria from the pig small intestine. Amino Acids. 2012;42(5):1597–1608. doi:10.1007/s00726-011-0846-x.
  • Burrin DG, Stoll B. Metabolic fate and function of dietary glutamate in the gut. Am J Clin Nutr. 2009;90:850S–856S. doi:10.3945/ajcn.2009.27462Y.
  • Nüse B, Holland T, Rauh M, Gerlach RG, Mattner J. L-arginine metabolism as pivotal interface of mutual host-microbe interactions in the gut. Gut Microbes. 2023;15(1):2222961. doi:10.1080/19490976.2023.2222961.
  • Rezaei R, Knabe DA, Tekwe CD, Dahanayaka S, Ficken MD, Fielder SE, Eide SJ, Lovering SL, Wu G. Dietary supplementation with monosodium glutamate is safe and improves growth performance in postweaning pigs. Amino Acids. 2013;44(3):911–923. doi:10.1007/s00726-012-1420-x.
  • Zhu M, Qin Y, Gao C, Yan H, Wang X. L-glutamate drives porcine intestinal epithelial renewal by increasing stem cell activity via upregulation of the EGFR-ERK-mTORC1 pathway. Food Funct. 2020;11(3):2714–2724. doi:10.1039/c9fo03065d.
  • Qin Y, Zhou J, Zhu M, Zan G, Gao C, Yan H, Li X-G, Wang X-Q. L-glutamate requires β-catenin signalling through Frizzled7 to stimulate porcine intestinal stem cell expansion. Cell Mol Life Sci. 2022;79(10):523. doi:10.1007/s00018-022-04545-2.
  • Deng H, Gerencser AA, Jasper H. Signal integration by Ca(2+) regulates intestinal stem-cell activity. Nature. 2015;528(7581):212–217. doi:10.1038/nature16170.
  • Tian J, Li Y, Bao X, Yang F, Tang X, Jiang Q, Yang C, Yin Y, Yao K. Glutamine boosts intestinal stem cell-mediated small intestinal epithelial development during early weaning: involvement of WNT signaling. Stem Cell Rep. 2023;18(7):1451–1467. doi:10.1016/j.stemcr.2023.05.012.
  • Hou Q, Dong Y, Yu Q, Wang B, Le S, Guo Y, Zhang B. Regulation of the Paneth cell niche by exogenous L-arginine couples the intestinal stem cell function. FASEB J. 2020;34(8):10299–10315. doi:10.1096/fj.201902573RR.
  • Hou Q, Dong Y, Huang J, Liao C, Lei J, Wang Y, Lai Y, Bian Y, He Y, Sun J. et al. Exogenous L-arginine increases intestinal stem cell function through CD90+ stromal cells producing mTORC1-induced Wnt2b. Commun Biol. 2020;3(1):611–624. doi:10.1038/s42003-020-01347-9.
  • Wang Y, Hou Q, Wu Y, Xu Y, Liu Y, Chen J, Xu L, Guo Y, Gao S, Yuan J. et al. Methionine deficiency and its hydroxy analogue influence chicken intestinal 3-dimensional organoid development. Anim Nutr. 2022;8(1):38–51. doi:10.1016/j.aninu.2021.06.001.
  • Saito Y, Iwatsuki K, Hanyu H, Maruyama N, Aihara E, Tadaishi M, Shimizu M, Kobayashi-Hattori K. Effect of essential amino acids on enteroids: methionine deprivation suppresses proliferation and affects differentiation in enteroid stem cells. Biochem Biophys Res Commun. 2017;488(1):171–176. doi:10.1016/j.bbrc.2017.05.029.
  • Zhou J, Wang Z, Zhang S, Lin H, Gao C, Zhao J, Yang C, Wang X-Q. Methionine and its hydroxyl analogues improve stem cell activity to eliminate deoxynivalenol-induced intestinal injury by reactivating Wnt/β-catenin signaling. J Agric Food Chem. 2019;67(41):11464–11473. doi:10.1021/acs.jafc.9b04442.
  • Obata F, Tsuda-Sakurai K, Yamazaki T, Nishio R, Nishimura K, Kimura M, Funakoshi M, Miura M. Nutritional Control of Stem Cell Division through S-Adenosylmethionine in Drosophila Intestine. Dev Cell. 2018;44(6):741–751.e3. doi:10.1016/j.devcel.2018.02.017.
  • Ye S, Shah BR, Li J, Liang H, Zhan F, Geng F. et al. A critical review on interplay between dietary fibers and gut microbiota. Trends Food Sci Technol. 2022;124:237–249. doi:10.1016/j.tifs.2022.04.010.
  • Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, Pudlo NA, Kitamoto S, Terrapon N, Muller A. et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell. 2016;167(5):1339–1353.e21. doi:10.1016/j.cell.2016.10.043.
  • Pi Y, Mu C, Gao K, Liu Z, Peng Y, Zhu W, Langille MGI. Increasing the hindgut carbohydrate/protein ratio by cecal infusion of corn starch or casein hydrolysate drives gut microbiota-related bile acid metabolism to stimulate colonic barrier function. mSystems. 2020;5(3):e00176–20. doi:10.1128/mSystems.00176-20.
  • Walsh C, Lane JA, van Sinderen D, Hickey RM. Human milk oligosaccharides: shaping the infant gut microbiota and supporting health. J Funct Foods. 2020;72:104074. doi:10.1016/j.jff.2020.104074.
  • Yu Z, Chen C, Kling DE, Liu B, McCoy JM, Merighi M, Heidtman M, Newburg DS. The principal fucosylated oligosaccharides of human milk exhibit prebiotic properties on cultured infant microbiota. Glycobiology. 2013;23(2):169–177. doi:10.1093/glycob/cws138.
  • Elison E, Vigsnaes LK, Rindom Krogsgaard L, Rasmussen J, Sørensen N, McConnell B, Hennet T, Sommer MOA, Bytzer P. Oral supplementation of healthy adults with 2′- O -fucosyllactose and lacto- N -neotetraose is well tolerated and shifts the intestinal microbiota. Br J Nutr. 2016;116(8):1356–1368. doi:10.1017/S0007114516003354.
  • Yang C, Zhang P, Fang W, Chen Y, Zhang N, Qiao Z, Troy FA, Wang B. Molecular mechanisms underlying how sialyllactose intervention promotes intestinal maturity by upregulating GDNF through a CREB-Dependent pathway in neonatal piglets. Mol Neurobiol. 2019;56(12):7994–8007. doi:10.1007/s12035-019-1628-9.
  • Charbonneau MR, O’Donnell D, Blanton LV, Totten SM, Davis JCC, Barratt MJ, Cheng J, Guruge J, Talcott M, Bain J. et al. Sialylated milk oligosaccharides promote microbiota-dependent growth in models of infant undernutrition. Cell. 2016;164(5):859–871. doi:10.1016/j.cell.2016.01.024.
  • Martinez-Guryn K, Hubert N, Frazier K, Urlass S, Musch MW, Ojeda P, Pierre JF, Miyoshi J, Sontag TJ, Cham CM. et al. Small intestine microbiota regulate Host digestive and absorptive adaptive responses to dietary lipids. Cell Host Microbe. 2018;23(4):458–469.e5. doi:10.1016/j.chom.2018.03.011.
  • Chen P, Torralba M, Tan J, Embree M, Zengler K, Stärkel P, van Pijkeren J-P, DePew J, Loomba R, Ho SB. et al. Supplementation of saturated long-chain fatty acids maintains intestinal eubiosis and reduces ethanol-induced liver injury in mice. Gastroenterology. 2015;148(1):203–214.e16. doi:10.1053/j.gastro.2014.09.014.
  • Lam YY, Ha CWY, Hoffmann JMA, Oscarsson J, Dinudom A, Mather TJ, Cook DI, Hunt NH, Caterson ID, Holmes AJ. et al. Effects of dietary fat profile on gut permeability and microbiota and their relationships with metabolic changes in mice. Obesity (Silver Spring). 2015;23(7):1429–1439. doi:10.1002/oby.21122.
  • Rabot S, Membrez M, Bruneau A, Gérard P, Harach T, Moser M, Raymond F, Mansourian R, Chou CJ. Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB J. 2010;24(12):4948–4959. doi:10.1096/fj.10.164921.
  • Sato H, Zhang LS, Martinez K, Chang EB, Yang Q, Wang F, Howles PN, Hokari R, Miura S, Tso P. et al. Antibiotics suppress activation of intestinal mucosal mast cells and reduce dietary lipid absorption in Sprague-Dawley rats. Gastroenterology. 2016;151(5):923–932. doi:10.1053/j.gastro.2016.07.009.
  • von Frieling J, Faisal MN, Sporn F, Pfefferkorn R, Nolte SS, Sommer F, Rosenstiel P, Roeder T. A high-fat diet induces a microbiota-dependent increase in stem cell activity in the Drosophila intestine. PloS Genet. 2020;16(5):e1008789. doi:10.1371/journal.pgen.1008789.
  • Beyaz S, Mana MD, Roper J, Kedrin D, Saadatpour A, Hong S-J, Bauer-Rowe KE, Xifaras ME, Akkad A, Arias E. et al. High-fat diet enhances stemness and tumorigenicity of intestinal progenitors. Nature. 2016;531(7592):53–58. doi:10.1038/nature17173.
  • Wang Q, Lin Y, Sheng X, Xu J, Hou X, Li Y, Zhang H, Guo H, Yu Z, Ren F. et al. Arachidonic acid promotes intestinal regeneration by activating WNT signaling. Stem Cell Rep. 2020;15(2):374–388. doi:10.1016/j.stemcr.2020.06.009.
  • Cheng C, Biton M, Haber AL, Gunduz N, Eng G, Gaynor LT, Tripathi S, Calibasi-Kocal G, Rickelt S, Butty VL. et al. Ketone body signaling mediates intestinal stem cell homeostasis and adaptation to diet. Cell. 2019;178(5):1115–1131.e15. doi:10.1016/j.cell.2019.07.048.
  • Li S, Lu C, Diem EC, Li W, Guderian M, Lindenberg M, Kruse F, Buettner M, Floess S, Winny MR. et al. Acetyl-CoA-carboxylase 1-mediated de novo fatty acid synthesis sustains Lgr5+ intestinal stem cell function. Nat Commun. 2022;13(1):3998–4013. doi:10.1038/s41467-022-31725-2.
  • Obniski R, Sieber M, Spradling AC. Dietary lipids modulate notch signaling and influence adult intestinal development and metabolism in Drosophila. Dev Cell. 2018;47(1):98–111.e5. doi:10.1016/j.devcel.2018.08.013.
  • Neophytou C, Soteriou E, Pitsouli C. The sterol transporter Npc2c controls intestinal stem cell mitosis and host-microbiome interactions in Drosophila. Metabolites. 2023;13(10):1084. doi:10.3390/metabo13101084.
  • Wang B, Rong X, Palladino END, Wang J, Fogelman AM, Martín MG, Alrefai WA, Ford DA, Tontonoz P. Phospholipid remodeling and cholesterol availability regulate intestinal stemness and tumorigenesis. Cell Stem Cell. 2018;22(2):206–220.e4. doi:10.1016/j.stem.2017.12.017.
  • Pham VT, Dold S, Rehman A, Bird JK, Steinert RE. Vitamins, the gut microbiome and gastrointestinal health in humans. Nutr Res. 2021;95:35–53. doi:10.1016/j.nutres.2021.09.001.
  • LeBlanc JG, Milani C, de Giori GS, Sesma F, van Sinderen D, Ventura M. Bacteria as vitamin suppliers to their host: a gut microbiota perspective. Curr Opin Biotechnol. 2013;24(2):160–168. doi:10.1016/j.copbio.2012.08.005.
  • Magnúsdóttir S, Ravcheev D, de Crécy-Lagard V, Thiele I. Systematic genome assessment of B-vitamin biosynthesis suggests co-operation among gut microbes. Front Genet. 2015;6:148–166. doi:10.3389/fgene.2015.00148.
  • Chang Y, Rossetti M, Vlamakis H, Casero D, Sunga G, Harre N, Miller S, Humphries R, Stappenbeck T, Simpson KW. et al. A screen of Crohn’s disease-associated microbial metabolites identifies ascorbate as a novel metabolic inhibitor of activated human T cells. Mucosal Immunol. 2019;12(2):457–467. doi:10.3389/fgene.2015.00148.
  • Said HM, Mohammed ZM. Intestinal absorption of water-soluble vitamins: an update. Curr Opin Gastroenterol. 2006;22(2):140–146. doi:10.1097/01.mog.0000203870.22706.52.
  • Ichihashi T, Takagishi Y, Uchida K, Yamada H. Colonic absorption of menaquinone-4 and menaquinone-9 in rats. J Nutr. 1992;122(3):506–512. doi:10.1093/jn/122.3.506.
  • Wang Z, Li J, Wang Y, Wang L, Yin Y, Yin L, Yang H, Yin Y. Dietary vitamin a affects growth performance, intestinal development, and functions in weaned piglets by affecting intestinal stem cells. J Anim Sci. 2020;98(2):skaa020. doi:10.1093/jas/skaa020.
  • Qu N, Jeffcoat B, Maity P, Christensen RK, Múnera JO. Retinoic acid promotes the in vitro growth, patterning and improves the cellular composition of human pluripotent stem-cell-derived intestinal organoids. Int J Mol Sci. 2022;23(15):8624–8637. doi:10.3390/ijms23158624.
  • da Silva Ferreiram AR, van der Aa SAJ, Wehkamp T, Wardill HR, Ten Klooster JP, Garssen J, Harthoorn LF, Hartog A, Harmsen HJM, Tissing WJE. et al. Development of a self-limiting model of methotrexate-induced mucositis reinforces butyrate as a potential therapy. Sci Rep. 2021;11(1):22911–22922. doi:10.1038/s41598-021-02308-w.
  • Khan AA, Dragt BS, Porte RJ, Groothuis GMM. Regulation of VDR expression in rat and human intestine and liver–consequences for CYP3A expression. Toxicol In Vitro. 2010;24(3):822–829. doi:10.1016/j.tiv.2009.12.011.
  • Li W, Peregrina K, Houston M, Augenlicht LH. Vitamin D and the nutritional environment in functions of intestinal stem cells: implications for tumorigenesis and prevention. J Steroid Biochem Mol Biol. 2020;198:105556–105575. doi:10.1016/j.jsbmb.2019.105556.
  • Park J, Na H, Kim Y. The anti-aging effect of vitamin D and vitamin D receptor in drosophila midgut. Aging (Albany NY). 2024;16(3):2005–2025. doi:10.18632/aging.205518.
  • Neophytou C, Pitsouli C. Biotin controls intestinal stem cell mitosis and host-microbiome interactions. Cell Rep. 2022;38(10):110505–110531. doi:10.1016/j.celrep.2022.110505.
  • Pyndt Jørgensen B, Winther G, Kihl P, Nielsen DS, Wegener G, Hansen AK, Sørensen DB. Dietary magnesium deficiency affects gut microbiota and anxiety-like behaviour in C57BL/6N mice. Acta Neuropsychiatr. 2015;27(5):307–311. doi:10.1017/neu.2015.10.
  • Reed S, Neuman H, Moscovich S, Glahn RP, Koren O, Tako E. Chronic zinc deficiency alters chick gut microbiota composition and function. Nutrients. 2015;7(12):9768–9784. doi:10.3390/nu7125497.
  • Sauer AK, Grabrucker AM. Zinc deficiency during pregnancy leads to altered microbiome and elevated inflammatory markers in mice. Front Neurosci. 2019;13:1295–1311. doi:10.3389/fnins.2019.01295.
  • La Carpia F, Wojczyk BS, Annavajhala MK, Rebbaa A, Culp-Hill R, D’Alessandro A, Freedberg DE, Uhlemann A-C, Hod EA. Transfusional iron overload and intravenous iron infusions modify the mouse gut microbiota similarly to dietary iron. NPJ Biofilms Microbiomes. 2019;5(1):26–37. doi:10.1038/s41522-019-0097-2.
  • Vonderheid SC, Tussing-Humphreys L, Park C, Pauls H, OjiNjideka Hemphill N, LaBomascus B, McLeod A, Koenig MD. A systematic review and meta-analysis on the effects of probiotic species on iron absorption and iron status. Nutrients. 2019;11(12):2938–2954. doi:10.3390/nu11122938.
  • Zhou J, Qin Y, Xiong X, Wang Z, Wang M, Wang Y, Wang QY, Yang HS, Yin Y. Effects of iron, vitamin A, and the interaction between the two nutrients on intestinal development and cell differentiation in piglets. J Anim Sci. 2021;99(10):1–9. doi:10.1093/jas/skab258.
  • Zhu H, Zhou Y, Qi Y, Ji R, Zhang J, Qian Z, Wu C, Tan J, Shao L, Chen D. et al. Preparation and characterization of selenium enriched-Bifidobacterium longum DD98, and its repairing effects on antibiotic-induced intestinal dysbacteriosis in mice. Food Funct. 2019;10(8):4975–4984. doi:10.1039/c9fo00960d.
  • Zhu C, Liang S, Zan G, Wang X, Gao C, Yan H, Wang X, Zhou J. Selenomethionine alleviates DON-Induced oxidative stress via modulating Keap1/Nrf2 signaling in the small intestinal epithelium. J Agric Food Chem. 2023;71(1):895–904. doi:10.1021/acs.jafc.2c07885.
  • Zhou J, Lin H, Wang Z, Zhang S, Huang D, Gao C, Yan H-C, Wang X-Q. Zinc L-Aspartate enhances intestinal stem cell activity to protect the integrity of the intestinal mucosa against deoxynivalenol through activation of the Wnt/β-catenin signaling pathway. Environ Pollut. 2020;262:114290. doi:10.1016/j.envpol.2020.114290.
  • Sato T, Stange DE, Ferrante M, Vries RGJ, Van Es JH, Van den Brink S, van Houdt WJ, Pronk A, van Gorp J, Siersema PD. et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141(5):1762–1772. doi:10.1053/j.gastro.2011.07.050.
  • Hedrich WD, Panzica-Kelly JM, Chen S-J, Strassle B, Hasson C, Lecureux L, Wang L, Chen W, Sherry T, Gan J. et al. Development and characterization of rat duodenal organoids for ADME and toxicology applications. Toxicology. 2020;446:152614–152627. doi:10.1016/j.tox.2020.152614. Epub 2020 Oct 24.
  • Kardia E, Frese M, Smertina E, Strive T, Zeng X, Estes M, Hall RN. Culture and differentiation of rabbit intestinal organoids and organoid-derived cell monolayers. Sci Rep. 2021;11(1):5401–5413. doi:10.1038/s41598-021-84774-w.
  • Gonzalez LM, Williamson I, Piedrahita JA, Blikslager AT, Magness ST, Singh SR. Cell lineage identification and stem cell culture in a porcine model for the study of intestinal epithelial regeneration. PLoS One. 2013;8(6):e66465. doi:10.1371/journal.pone.0066465.
  • Zhao D, Farnell MB, Kogut MH, Genovese KJ, Chapkin RS, Davidson LA, Berghman LR, Farnell YZ. From crypts to enteroids: establishment and characterization of avian intestinal organoids. Poultry Sci. 2022;101(3):101642–101653. doi:10.1016/j.psj.2021.101642.
  • Hamilton CA, Young R, Jayaraman S, Sehgal A, Paxton E, Thomson S, Katzer F, Hope J, Innes E, Morrison LJ. et al. Development of in vitro enteroids derived from bovine small intestinal crypts. Vet Res. 2018;49(1):54–69. doi:10.1186/s13567-018-0547-5.
  • Smith D, Price DRG, Burrells A, Faber MN, Hildersley KA, Chintoan-Uta C, Chapuis AF, Stevens M, Stevenson K, Burgess STG. et al. The development of ovine gastric and intestinal organoids for studying ruminant host-pathogen interactions. Front Cell Infect Microbiol. 2021;11:733811–733829. doi:10.3389/fcimb.2021.733811.
  • Zhang M, Lv L, Cai H, Li Y, Gao F, Yu L, Jiang Y, Tong W, Li L, Li G. et al. Long-term expansion of porcine intestinal organoids serves as an in vitro model for swine enteric coronavirus infection. Front Microbiol. 2022;13:865336–865349. doi:10.3389/fmicb.2022.865336.
  • Sato T, Clevers H. Growing self-organizing mini-guts from a single intestinal stem cell: mechanism and applications. Science. 2013;340(6137):1190–1194. doi:10.1126/science.1234852.
  • Yin Y, de Jonge HR, Wu X, Yin Y. Enteroids for nutritional studies. Mol Nutr Food Res. 2019;63(16):e1801143. doi:10.1002/mnfr.201801143.
  • van der Hee B, Loonen LMP, Taverne N, Taverne-Thiele JJ, Smidt H, Wells JM. Optimized procedures for generating an enhanced, near physiological 2D culture system from porcine intestinal organoids. Stem Cell Res. 2018;28:165–171. doi:10.1016/j.scr.2018.02.013. Epub 2018 Feb 20.
  • Kozuka K, He Y, Koo-McCoy S, Kumaraswamy P, Nie B, Shaw K, Chan P, Leadbetter M, He L, Lewis JG. et al. Development and characterization of a human and mouse intestinal epithelial cell monolayer platform. Stem Cell Rep. 2017;9(6):1976–1990. doi:10.1016/j.stemcr.2017.10.013.
  • Hofer M, Lutolf MP. Engineering organoids. Nat Rev Mater. 2021;6(5):402–420. doi:10.1038/s41578-021-00279-y.
  • Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, Liu IH, Chiou S-H, Salahudeen AA, Smith AR. et al. Organoid modeling of the tumor immune microenvironment. Cell. 2018;175(7):1972–1988. doi:10.1016/j.cell.2018.11.021.
  • Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, Tolle K, Hoskins EE, Kalinichenko VV, Wells SI, Zorn AM. et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro, Nature. 2011;470(7332):105–109. doi:10.1038/nature09691. Epub 2010 Dec 12.
  • Du A, McCracken KW, Walp ER, Terry NA, Klein TJ, Han A, Wells JM, May CL. Arx is required for normal enteroendocrine cell development in mice and humans. Dev Biol. 2012;365(1):175–188. doi:10.1016/j.ydbio.2012.02.024.
  • Sommer CA, Capilla A, Molina-Estevez FJ, Gianotti-Sommer A, Skvir N, Caballero I, Chowdhury S, Mostoslavsky G. Modeling APC mutagenesis and familial adenomatous polyposis using human iPS cells. PLos One. 2018;13(7):e0200657. doi:10.1371/journal.pone.0200657.
  • Son YS, Ki SJ, Thanavel R, Kim J, Lee M, Kim J, Jung C-R, Han T-S, Cho H-S, Ryu C-M. et al. Maturation of human intestinal organoids in vitro facilitates colonization by commensal lactobacilli by reinforcing the mucus layer. FASEB J. 2020;34(8):9899–9910. doi:10.1096/fj.202000063R.
  • McCauley HA, Matthis AL, Enriquez JR, Nichol JT, Sanchez JG, Stone WJ, Sundaram N, Helmrath MA, Montrose MH, Aihara E. et al. Enteroendocrine cells couple nutrient sensing to nutrient absorption by regulating ion transport. Nat Commun. 2020;11(1):4791–4794. doi:10.1038/s41467-020-18536-z.
  • Watson CL, Mahe MM, Múnera J, Howell JC, Sundaram N, Poling HM, Schweitzer JI, Vallance JE, Mayhew CN, Sun Y. et al. An in vivo model of human small intestine using pluripotent stem cells. Nat Med. 2014;20(11):1310–1314. doi:10.1038/nm.3737.
  • Workman MJ, Mahe MM, Trisno S, Poling HM, Watson CL, Sundaram N, Chang C-F, Schiesser J, Aubert P, Stanley EG. et al. Engineered human pluripotent-stem-cell-derived intestinal tissues with a functional enteric nervous system. Nat Med. 2017;23(1):49–59. doi:10.1038/nm.4233.
  • Bouffi C, Wikenheiser-Brokamp KA, Chaturvedi P, Sundaram N, Goddard GR, Wunderlich M, Brown NE, Staab JF, Latanich R, Zachos NC. et al. In vivo development of immune tissue in human intestinal organoids transplanted into humanized mice. Nat Biotechnol. 2023;41(6):824–831. doi:10.1038/s41587-022-01558-x.
  • Pitstick AL, Poling HM, Sundaram N, Lewis PL, Kechele DO, Sanchez JG, Scott MA, Broda TR, Helmrath MA, Wells JM. et al. Aggregation of cryopreserved mid-hindgut endoderm for more reliable and reproducible hPSC-derived small intestinal organoid generation. Stem Cell Rep. 2022;17(8):1889–1902. doi:10.1016/j.stemcr.2022.06.011.
  • Múnera JO, Sundaram N, Rankin SA, Hill D, Watson C, Mahe M, Vallance JE, Shroyer NF, Sinagoga KL, Zarzoso-Lacoste A. et al. Differentiation of human pluripotent stem cells into colonic organoids via transient activation of BMP signaling. Cell Stem Cell. 2017;21(1):51–54. doi:10.1016/j.stem.2017.05.020.
  • Dutta D, Clevers H. Organoid culture systems to study host-pathogen interactions. Curr Opin Immunol. 2017;48:15–22. doi:10.1016/j.coi.2017.07.012.
  • Puschhof J, Pleguezuelos-Manzano C, Martinez-Silgado A, Akkerman N, Saftien A, Boot C, de Waal A, Beumer J, Dutta D, Heo I. et al. Intestinal organoid cocultures with microbes. Nat Protoc. 2021;16(10):4633–4649. doi:10.1038/s41596-021-00589-z.
  • Aguilar C, Alves da Silva M, Saraiva M, Neyazi M, Olsson IAS, Bartfeld S. Organoids as host models for infection biology – a review of methods. Exp Mol Med. 2021;53(10):1471–1482. doi:10.1038/s12276-021-00629-4.
  • VanDussen KL, Marinshaw JM, Shaikh N, Miyoshi H, Moon C, Tarr PI, Ciorba MA, Stappenbeck TS. Development of an enhanced human gastrointestinal epithelial culture system to facilitate patient-based assays. Gut. 2015;64(6):911–920. doi:10.1136/gutjnl-2013-306651.
  • Moon C, VanDussen KL, Miyoshi H, Stappenbeck TS. Development of a primary mouse intestinal epithelial cell monolayer culture system to evaluate factors that modulate IgA transcytosis. Mucosal Immunol. 2014;7(4):818–828. doi:10.1038/mi.2013.98.
  • Wang Y, DiSalvo M, Gunasekara DB, Dutton J, Proctor A, Lebhar MS, Williamson IA, Speer J, Howard RL, Smiddy NM. et al. Self-renewing monolayer of primary colonic or rectal epithelial cells. Cell Mol Gastroenterol Hepatol. 2017;4(1):165–182. doi:10.1016/j.jcmgh.2017.02.011.
  • Roodsant T, Navis M, Aknouch I, Renes IB, van Elburg RM, Pajkrt D, Wolthers KC, Schultsz C, van der Ark KCH, Sridhar A. et al. A human 2D primary organoid-derived epithelial monolayer model to study host-pathogen interaction in the small intestine. Front Cell Infect Microbiol. 2020;10:272–286. doi:10.3389/fcimb.2020.00272.
  • Li Y, Yang N, Chen J, Huang X, Zhang N, Yang S, Liu G, Liu G. Next-generation porcine intestinal organoids: an apical-out organoid model for swine enteric virus infection and immune response investigations. J Virol. 2020;94(21):e01006–20. doi:10.1128/JVI.01006-20.
  • Jardé T, Chan WH, Rossello FJ, Kaur Kahlon T, Theocharous M, Kurian Arackal T, Flores T, Giraud M, Richards E, Chan E. et al. Mesenchymal niche-derived neuregulin-1 drives intestinal stem cell proliferation and regeneration of damaged epithelium. Cell Stem Cell. 2020;27(4):646–662.e7. doi:10.1016/j.stem.2020.06.021.
  • Deng M, Guerrero-Juarez CF, Sheng X, Xu J, Wu X, Yao K, Li M, Yang X, Li G, Xiao J. et al. Lepr+ mesenchymal cells sense diet to modulate intestinal stem/progenitor cells via leptin–Igf1 axis. Cell Res. 2022;32(7):670–686. doi:10.1038/s41422-022-00643-9.
  • Biton M, Haber AL, Rogel N, Burgin G, Beyaz S, Schnell A, Ashenberg O, Su C-W, Smillie C, Shekhar K. et al. T helper cell cytokines modulate intestinal stem cell renewal and differentiation. Cell. 2018;175(5):1307–1320.e22. doi:10.1016/j.cell.2018.10.008.
  • Lindemans CA, Calafiore M, Mertelsmann AM, O’Connor MH, Dudakov JA, Jenq RR, Velardi E, Young LF, Smith OM, Lawrence G. et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature. 2015;528(7283):560–564. doi:10.1038/nature16460.
  • Low LA, Mummery C, Berridge BR, Austin CP, Tagle DA. Organs-on-chips: into the next decade. Nat Rev Drug Discov. 2021;20(5):345–361. doi:10.1038/s41573-020-0079-3.
  • Rajasekar S, Lin DSY, Abdul L, Liu A, Sotra A, Zhang F, Zhang B. IFlowPlate—A customized 384-well plate for the culture of perfusable vascularized colon organoids. Adv Mater. 2020;32(46):e2002974. doi:10.1002/adma.202002974.
  • Zhu P, Lu T, Wu J, Fan D, Liu B, Zhu X, Guo H, Du Y, Liu F, Tian Y. et al. Gut microbiota drives macrophage-dependent self-renewal of intestinal stem cells via niche enteric serotonergic neurons. Cell Res. 2022;32(6):555–569. doi:10.1038/s41422-022-00645-7.