Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Jun 26, 2024; 16(6): 619-622
Published online Jun 26, 2024. doi: 10.4252/wjsc.v16.i6.619
Gut microbiota modulating intestinal stem cell differentiation
Lin He, Xiang-Feng Zhou, Kuan Li, Department of Alcohol and Drug Dependence Treatment, The Mental Hospital of Yunnan Province, Kunming 650224, Yunnan Province, China
Chen Zhu, Department of Physical Education, Kunming Medical University, Kunming 650500, Yunnan Province, China
Shu-E Zeng, Department of Geriatric Psychiatry, The Mental Hospital of Yunnan Province, Kunming 650224, Yunnan Province, China
Le Zhang, Sleep Medicine Center, The Mental Hospital of Yunnan Province, Kunming 650224, Yunnan Province, China
ORCID number: Kuan Li (0000-0001-7662-3093).
Author contributions: He L, Zhu C, Zhou XF, Zeng SE, Zhang L, and Li K contributed to this paper; He L, Zhu C, and Li K designed the overall concept and outline of the manuscript; Zhou XF, Zeng SE, and Zhang L contributed to the discussion and design of the manuscript; Zhu C and Li K contributed to the writing, and editing the manuscript, illustrations, and review of literature.
Supported by Scientific Research Fund Project of Education Department of Yunnan Province, No. 2023J0346; the Kunming Health Commission Kunming Health Science and Technology Personnel Training Project, No. 2021-SW-75; and the Medical and Health Science and Technology Project of Kunming Health Committee, No. 2022-03-09-008.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Kuan Li, PhD, Doctor, Department of Alcohol and Drug Dependence Treatment, The Mental Hospital of Yunnan Province, No. 733 Chuanjin Road, Panlong District, Kunming 650224, Yunnan Province, China. n_kli_sxwang@163.com
Received: February 22, 2024
Revised: May 6, 2024
Accepted: May 20, 2024
Published online: June 26, 2024
Processing time: 124 Days and 2.5 Hours

Abstract

Proliferation and differentiation of intestinal stem cell (ISC) to replace damaged gut mucosal epithelial cells in inflammatory states is a critical step in ameliorating gut inflammation. However, when this disordered proliferation continues, it induces the ISC to enter a cancerous state. The gut microbiota on the free surface of the gut mucosal barrier is able to interact with ISC on a sustained basis. Microbiota metabolites are able to regulate the proliferation of gut stem and progenitor cells through transcription factors, while in steady state, differentiated colonocytes are able to break down such metabolites, thereby protecting stem cells at the gut crypt. In the future, the gut flora and its metabolites mediating the regulation of ISC differentiation will be a potential treatment for enteropathies.

Key Words: Intestinal stem cells, Gut microbiota, Gut stem niche, Microenvironment, Probiotics

Core Tip: The dysbiosis may cause intestinal cancer. when the proliferation of the stem cells attempting to repair the loss of integrity of the gut barrier. The correction of the gut stem niche dysbiosis by the assumption of some beneficial microbiota could be a specific therapy of this disease.



INTRODUCTION

Inflammatory bowel disease and irritable bowel syndrome have become global diseases. In the inflammatory state that occurs, proliferation and differentiation of intestinal stem cells (ISCs) to replace damaged gut mucosal epithelial cells is a critical step in ameliorating gut inflammation[1]. ISCs at the gut crypts have the ability to continuously proliferate and differentiate into different types of gut mucosal epithelium during their migration towards the apical part of the gut villi. The microenvironment in which the ISCs reside is known as the stem cell niche, which directs the function of ISCs in homeostasis and repair processes and influences cancer development. Current studies have shown that intrinsic and extrinsic factors and diet can regulate stem cell niche; ISCs can gain competitive advantages when mutated and can regulate the local microenvironment, which drives tumorigenesis and clonal expansion; tumor stem cells control tumor growth and progression in colorectal cancer, and the function of these cells also depends on the microenvironment in which they reside; mutant stem cells are difficult to eliminate, and an understanding of the interactions between mutant stem cells and their microenvironment could help to develop new technologies for colorectal cancer[2].

STEM CELL NICHE REGULATE ISCS

Competitive inhibition: Mutations in the oncogene Apc are present in about 80% of colon cancers, and ISCs carrying the Apc mutation have a strong competitive advantage in the face of normal ISCs. This advantage is due to the fact that Apc mutant cells can secrete WNT antagonist factors, which inhibit the activity of the normal ISCs and promote their differentiation; the inhibitory effect of WNT antagonist factors on the normal ISCs is effectively counteracted[3]. Balance of renewal and differentiation: In addition, ISCs promote gut homeostasis through the balance between self-renewal and differentiation, and the secretory matrix protein CCN1 at the base of the crypts interacts with integrins αvβ3/αvβ5 to regulate ISCs homeostasis through two different pathways downstream of it, regulating Notch and Wnt signaling, respectively[4]. T cell-expressed integrin αEβ7 binds to E-cadherin, an adhesion signal expressed by ISCs and transiently proliferating (TA) cells, triggering endocytosis of E-cadherin and modulating Wnt and Notch signaling changes. Blocking Eβ7-E-cadherinα adhesion inhibits Wnt signaling and promotes Notch signaling in ISC and TA cells, leading to defective ISC differentiation. αEβ7+ T cells regulate ISC differentiation at the single-cell level through cell-cell contact-mediated αEβ7-E-cadherin adhesion signaling, emphasizing the important role of T-cell-stem cell/TA cell contact in maintaining homeostasis in the intestine[5]. Cellular supportive role: ISCs located at the base of the gut crypt are dependent on various factors in their surrounding stem cell niche to function properly, however the cellular source of these factors and how they play a supportive role for ISCs is not fully understood. Identifies two types of cells in the ISCs microenvironment - lymphatic endothelial cells and RSPO3+GREM1+ fibroblasts, which are the main cellular source of RSPO3, a key factor in the ISC microenvironment, and which play an important supportive role for the ISCs under gut homeostasis and during regeneration of the gut epithelium[6].

GUT MICROBIOTA REGULATE STEM CELL NICHE

The gut harbors a large number of microorganisms that interact with epithelial cells to maintain a healthy physiological state in the host. These gut microbiota are involved in the fermentation of non-digestible nutrients and produce beneficial metabolites that regulate the host’s homeostasis, metabolism and immune response. Lactobacillus acidophilus not only inhibits pathogen invasion, but also determines the fate of the gut epithelium, thereby protecting the gut mucosa from overactivation of the Wnt signaling pathway, aberrant proliferation of crypts, and overconsumption of secretory cells in Salmonella typhimurium infection[7]. Lactobacillus reuteri stimulates gut epithelial cell proliferation and thereby activating the Wnt/β-catenin pathway, effectively maintaining gut epithelial regeneration and homeostasis in vivo, as well as repairing gut damage after pathologic injury[8]. On the other hand, butyric acid, at certain physiological concentrations, is able to inhibit the proliferation of gut stem and progenitor cells in dependence on the transcription factor FoxO3; whereas, at homeostasis, differentiated colonocytes are able to metabolize butyric acid and reduce the concentration of butyric acid in the gut tract, in order to protect stem cells at the gut saphenous fossa[9].

Ha et al[10] proposed that restoration of microbial composition, enhancement of gut barrier integrity, induction of apoptosis in cancer cells, inactivation of carcinogens, and modulation of host immune response through probiotics. Reducing the incidence of colorectal cancer, attenuating treatment-related side effects, and enhancing the efficacy of anticancer therapies are key to successful translation into clinical practice. However, before their use in the clinic, it is important to assess the potential risks, optimize the method of administration, and consider the changes in the baseline gut microbiology in the patient’s body[10].

Ha et al[10] proposed that restoration of microbial composition, enhancement of gut barrier integrity, induction of apoptosis in cancer cells, inactivation of carcinogens, and modulation of host immune response through probiotics. Reducing the incidence of colorectal cancer, attenuating treatment-related side effects, and enhancing the efficacy of anticancer therapies are key to successful translation into clinical practice. However, before their use in the clinic, it is important to assess the potential risks, optimize the method of administration, and consider the changes in the baseline gut microbiology in the patient’s body[10].

CONCLUSION

In the future single-cell sequencing, cell lineage tracing, and metabolomics approaches effectively reveal the impact of ISCs composition, cellular characteristics, and function, as well as signaling pathway changes in associated cancers, providing new insights into gut microbes modulating ISCs differentiation for the treatment of gastrointestinal tumors[11]. Gut epithelial cells form organoids in the medium of three-dimensional scaffolds containing stem and differentiated cells, and the use of organoids will be effective in revealing the relationship between the complex three-dimensional structure of the intestine and the microphysiological system in gut tumors[12].

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cell and tissue engineering

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade A

Novelty: Grade A

Creativity or Innovation: Grade A

Scientific Significance: Grade A

P-Reviewer: Salvadori M, Italy S-Editor: Wang JJ L-Editor: A P-Editor: Zheng XM

References
1.  Holmberg FE, Seidelin JB, Yin X, Mead BE, Tong Z, Li Y, Karp JM, Nielsen OH. Culturing human intestinal stem cells for regenerative applications in the treatment of inflammatory bowel disease. EMBO Mol Med. 2017;9:558-570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 61]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
2.  Ramadan R, van Driel MS, Vermeulen L, van Neerven SM. Intestinal stem cell dynamics in homeostasis and cancer. Trends Cancer. 2022;8:416-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
3.  van Neerven SM, de Groot NE, Nijman LE, Scicluna BP, van Driel MS, Lecca MC, Warmerdam DO, Kakkar V, Moreno LF, Vieira Braga FA, Sanches DR, Ramesh P, Ten Hoorn S, Aelvoet AS, van Boxel MF, Koens L, Krawczyk PM, Koster J, Dekker E, Medema JP, Winton DJ, Bijlsma MF, Morrissey E, Léveillé N, Vermeulen L. Apc-mutant cells act as supercompetitors in intestinal tumour initiation. Nature. 2021;594:436-441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 81]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
4.  Won JH, Choi JS, Jun JI. CCN1 interacts with integrins to regulate intestinal stem cell proliferation and differentiation. Nat Commun. 2022;13:3117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
5.  Chen S, Zheng Y, Ran X, Du H, Feng H, Yang L, Wen Y, Lin C, Wang S, Huang M, Yan Z, Wu D, Wang H, Ge G, Zeng A, Zeng YA, Chen J. Integrin αEβ7(+) T cells direct intestinal stem cell fate decisions via adhesion signaling. Cell Res. 2021;31:1291-1307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
6.  Goto N, Goto S, Imada S, Hosseini S, Deshpande V, Yilmaz ÖH. Lymphatics and fibroblasts support intestinal stem cells in homeostasis and injury. Cell Stem Cell. 2022;29:1246-1261.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 33]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
7.  Lu X, Xie S, Ye L, Zhu L, Yu Q. Lactobacillus Protects Against S. Typhimurium-Induced Intestinal Inflammation by Determining the Fate of Epithelial Proliferation and Differentiation. Mol Nutr Food Res. 2020;64:e1900655.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
8.  Wu H, Xie S, Miao J, Li Y, Wang Z, Wang M, Yu Q. Lactobacillus reuteri maintains intestinal epithelial regeneration and repairs damaged intestinal mucosa. Gut Microbes. 2020;11:997-1014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 157]  [Article Influence: 39.3]  [Reference Citation Analysis (0)]
9.  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;165:1708-1720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 452]  [Cited by in F6Publishing: 415]  [Article Influence: 51.9]  [Reference Citation Analysis (0)]
10.  Ha S, Zhang X, Yu J. Probiotics intervention in colorectal cancer: From traditional approaches to novel strategies. Chin Med J (Engl). 2024;137:8-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
11.  Aliluev A, Tritschler S, Sterr M, Oppenländer L, Hinterdobler J, Greisle T, Irmler M, Beckers J, Sun N, Walch A, Stemmer K, Kindt A, Krumsiek J, Tschöp MH, Luecken MD, Theis FJ, Lickert H, Böttcher A. Diet-induced alteration of intestinal stem cell function underlies obesity and prediabetes in mice. Nat Metab. 2021;3:1202-1216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 50]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
12.  Antfolk M, Jensen KB. A bioengineering perspective on modelling the intestinal epithelial physiology in vitro. Nat Commun. 2020;11:6244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]