Review Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Aug 7, 2013; 19(29): 4671-4678
Published online Aug 7, 2013. doi: 10.3748/wjg.v19.i29.4671
Update on small intestinal stem cells
Valentina Tesori, Department of Internal Medicine and Gastroenterology, Università Cattolica del Sacro Cuore, Rome, Largo A. Gemelli 8, 00197 Rome, Italy
Valentina Tesori, Maria Ausiliatrice Puglisi, Antonio Gasbarrini, Department of Internal Medicine, Gemelli Hospital, 00168 Rome, Italy
Wanda Lattanzi, Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
Giovanni Battista Gasbarrini, Medical Research Foundation ONLUS, Galleria Falcone Borsellino 2, 40123 Bologna, Italy
Author contributions: Tesori V and Puglisi MA contributed equally to this work; Tesori V, Puglisi MA and Gasbarrini A contributed to the article design, writing and revision; Lattanzi W and Gasbarrini GB contributed to the literature research and revision; all authors approved the version to be published.
Correspondence to: Valentina Tesori, PhD, Department of Internal Medicine, Università Cattolica del Sacro Cuore, Rome, Largo A. Gemelli 8, 00197 Rome, Italy. valentina.tesori@libero.it
Telephone: +39-6-30156018 Fax: +39-6-30157249
Received: January 30, 2013
Revised: April 18, 2013
Accepted: May 7, 2013
Published online: August 7, 2013

Abstract

Among somatic stem cells, those residing in the intestine represent a fascinating and poorly explored research field. Particularly, somatic stem cells reside in the small intestine at the level of the crypt base, in a constant balance between self-renewal and differentiation. Aim of the present review is to delve into the mechanisms that regulate the delicate equilibrium through which intestinal stem cells orchestrate intestinal architecture. To this aim, special focus will be addressed to identify the integrating signals from the surrounding niche, supporting a model whereby distinct cell populations facilitate homeostatic vs injury-induced regeneration.

Key Words: Intestinal stem cells, Organoids, Intestinal regeneration, Lgr5, Niche

Core tip: Among somatic stem cells, those residing in the intestine represent a fascinating and poorly explored research field. Aim of the present review is to delve into the mechanisms that regulate the delicate equilibrium through which intestinal stem cells orchestrate intestinal architecture, integrating signals from the surrounding niche and supports a model whereby distinct cell populations facilitate homeostatic vs injury-induced regeneration.



INTRODUCTION

In an adult organism, stem cells are characterized by their ability to generate multiple differentiated cell types while maintaining their capacity for long-term self-renewal[1,2]. These are generally known as “adult” or “somatic” stem cells, including all stem cells residing in adult organs, regardless of the age of the individual. These include mesenchymal stem cells[3-7], residing in the connective stroma of most organs, and haematopoietic stem cells[8,9] among the best known and characterized, that are already being tested in clinical trials[10-14].

The amazing renewal capacity of the intestinal epithelium[1] has made this organ an attractive site to study stem-cell regulation. The intestinal tract is anatomically subdivided into the small intestine and large intestine. The inner mucosal surface, composed by an absorptive and secretory epithelium, is folded into repeated units comprising finger-like invaginations (called crypts of Lieberkühn) associated with numerous protrusions (villi), which increase the surface area, allowing efficient absorption of nutrients from the bowel lumen[2].

In normal homeostasis, the specialized differentiated cell types that orchestrate the uptake of nutrients into the body, are routinely and rapidly turned over. In fact, the intestinal epithelium is the most rapidly self-renewing tissue in the human body, with a 3-5 d turnover rate[2]. It is widely accepted that this complex process is regulated, via a highly regulated process of self-renewal, by a population of multipotent stem cells, residing within the bottom of the crypt namely the intestinal stem cells (ISCS)[15-19].

The number and location of these cells are still debated. Clonal analysis has demonstrated the existence of multiple stem cells in each crypt[20], with an estimated number in the 4-6 cells per crypt range[21]. ISCs have the properties of self-renewing and generating rapidly dividing transit-amplifying (TA) daughter cells, via asymmetric cell division[22]. TA cells undergo rapid cell division and migrate upwards into the villus. During their migration, TA cells start differentiating and finally localize at the surface of the villus epithelium as either mature absorptive enterocytes, which represent the main cell type, or mucous secreting goblet cells, or hormone-producing enteroendocrine cells[22]. Upon completing their life cycle, TA die and are discarded into the lumen[23,24].

A distinct cell type, the Paneth cell, evades this upward migration program, completing the differentiation at the base of the crypt, where it start producing lysozyme, maintaining the sterile environment of the crypt, and regulating the stem cell compartment[25-27].

Converging evidence suggests the existence of two distinct populations of intestinal stem cells: one that remains quiescent for a long time and one that actively proliferate[28]. The actively dividing ISCs provide to the baseline regeneration, whereas quiescent stem cells represent a reserve subpopulation that copes to injuries. These two subpopulations are located in adjacent sites within the crypt and are probably maintained by specific signals from the surrounding niche. Nonetheless, the precise identity of the ISCS is still a matter of debate. Two alternative models are currently proposed in the literature: the label-retaining cells (LRC) + 4 model, which identifies the quiescent stem cells, and the crypt base columnar (CBC) cells model, which identifies the actively cycling stem cells.

According to the LRC+4 model, the ISCS should be located specifically at the +4 position from the bottom of the intestinal crypt region, precisely at the origin of the migratory epithelial cell column[29]. This prediction was supported by Potten et al[30], who showed that cells most commonly found in this position, are characteristically label-retaining and extremely sensitive to X- and γ-radiation, two features ascribed to stem cells. Furthermore, the expression of Bmi1, a gene thought to be involved in stem cell maintenance, was shown to be elevated in the +4 cells[31]. Alternatively, the CBC cell model is based on a series of electron microscopy studies on the crypts of the small intestine, showing slender, immature, cycling cells interspersed between Paneth cells at positions 1-4, hence termed crypt base columnar cells. To support the hypothesis of CBC as the ISCs, mutagenesis studies demonstrated that 90% of the crypts, that contained a mixed population of mutant cells of different epithelial lineages, also contained mutant CBC cells, indicating the CBC cells as the common source of these different lineages[32]. Further studies, based on targeted lineage tracing strategies, have definitively identified the CBC as the intestinal stem cells, and have revealed the strategy by which the balance between proliferation and differentiation is maintained[33]. Taken together, these studies suggested that ISCs operate within a complex and dynamic environment, in which stochastic cell loss is compensated by the proliferation of neighboring stem cells.

INTESTINAL STEM CELLS MARKERS

The crypt stem cells responsible for the renewal capacity of the intestinal epithelium, represent a minority of the whole intestinal population, therefore, their identification is extremely troublesome[14]. Indeed, until relatively recently, ISCs could be identified only by indirect measurements. The recent discovery of specific ISC markers has allowed their isolation and paved the way towards a clearer understanding of their biology and role in tissue homeostasis, repair, and cancer[14].

Among the various ISCS markers, the best characterized one is the leucine rich-repeat containing G-protein coupled receptor (Lgr5), a Wnt-target gene that expressed by the cycling crypt base columnar cells, interspersed between Paneth cells[22]. Lgr5 encodes an orphan G-protein-coupled receptor, characterized by a large leucine-rich extracellular domain[34].

Barker and co-workers demonstrated that CBC cells are capable of long-term maintenance and support the epithelium self-renewal, using the lineage tracing technique (i.e., introducing permanent genetic marker into candidate stem cell genes in situ, thus allowing the visualization of the modified stem cells and their progeny over time)[34]. One of the major advantages of in vivo lineage tracing, compared to transplantation-based methods, is the lack of a physical manipulation of the candidate stem cell, so that ISCS are studied in their physiological niche[34]. In particular, the visualization and isolation of putative ISCs was obtained by targeting the Lgr5/Gpr49 gene locus by knock-in of a dual expression cassette encoding enhanced green fluorescent protein (EGFP) and CreERT2. This resulted in the tamoxifen-induced expression of EGFP in the Lgr5+ fraction, which allowed the in vivo lineage tracing when combined with inducible reporter strains such as R26RLacZ[34]. This study hence showed that Lgr5 expression was confined to CBCs, and that these cells give rise to the variety of epithelial cells present in crypts, proving that CBCs function as ISCs as well[34].

In addition, it has been demonstrated that Lgr5+ cells form self-renewing epithelial organoids in ex vivo culture assays, resembling the in vivo structure and composition of crypt/villus epithelial units. It is also worth noticing that cells expressing low, if any, Lgr5 were unable to produce organoids[35]. Gene expression analysis of purified Lgr5+ stem cells, indicated that they express additional specific markers, such as Olfm4 and the Achaete scutelike 2 (Ascl2)[36]. Ascl2 is a basic helix-loop-helix transcription factor[19]; its expression in the intestinal epithelium is regulated by the Wnt pathway and is restricted to Lgr5+ stem cells. Ascl2 deletion results in the complete loss of Lgr5+ ISCs, whereas transgenic Ascl2 expression induces crypt hyperplasia[36].

Moreover, several lines of evidence have demonstrated the existence of another putative intestinal stem cells marker, the Polycomb family member Bmi1[31]. The Polycomb family plays a key role in the development and in the regulation of progenitor self-renewal in several tissues, including the nervous system[37], the retina[38] and hematopoietic organs[39]. In vivo lineage tracing technique, showed that Bmi1+ cells are mainly located at the +4 position in the crypts of the small intestine, contributing to the long-term maintenance of all its epithelial cell types[31]. In addition, the selective ablation of the Bmi1+ population led to a disorganization of the intestinal mucosa, resulting in the absence of the crypts[31]. Unexpectedly, Bmi1 transgenic expression was restricted only to a minority of the crypts in the proximal small intestine, while being completely absent in the distal tract[31]. This could be possibly due to the existence of Bmi1-negative ISC populations in other regions of the intestine, or, perhaps, to an inaccurate reporting of endogenous Bmi1 expression, as a result of the variegated transgene activity, frequently observed in the intestine. Interestingly, microarray analysis showed that sorted Lgr5+ cells express Bmi1, raising the question whether the two markers really characterize independent stem cells populations. Yan et al[40] clarified this issue by demonstrating that Bmi1 and Lgr5 mark two functionally distinct ISCs in vivo. Lgr5 identifies actively cycling ISCs that are sensitive to Wnt modulation, involved in homeostatic regeneration and markedly ablated by irradiation, i.e., the CBC cells. Conversely, Bmi1 is expressed by quiescent ISCS insensitive to Wnt modulations, that contribute partly to homeostatic regeneration, and are resistant to radiation injury, namely the LRC stem cells[40].

Another interesting molecule in the scenario of ISCS putative markers, is the CD133/Prominin 1(Prom1), originally discovered as novel glycoprotein expressed on neural[41] and hematopoietic stem cells[42,43]. More recently, CD133 has been described as marker of epithelial stem/progenitor cells in human kidney tubules[44] and in the prostate[45]. In the study by Zhu et al[46], a knock-in allele was used that integrated a CreERT2-IRES-nLacZ cassette at the first ATG codon of Prom1 (Prom1C-L). This allowed demonstrating a wide expression pattern for Prom1 in the colon; on the other hand, the expression in the small intestine, appeared to be restricted to the crypt base, overlapping with the Lgr5+ CBC cells[46] The Prom1+ cells were self-renewing, multipotent adult stem cells. By contrast with these findings, Snippert et al[47] reported that Prom 1 expression occurred in Lgr5+ stem cells as well as in their TA progenitors. A possible explanation for this discrepancy may reside in the different sensitivity of detection methods used in the two studies.

The RNA-binding protein Musashi 1 (Msi1), a regulator of asymmetric cell division[48], is also involved in stem cell maintenance[49,50]. Particularly, in neural stem cells Msi1 is able to maintain stemness properties through Notch pathway activation[51]. Independent immunoistochemical and in situ hybridization analyses, demonstrated that, Msi1 is expressed in the CBC cells immediately above the Paneth cells[52-54].

Moreover, Msi1 overexpression in the intestine increases both Wnt and Notch pathways, and induces the upregulation of Lgr5 and Bmi1[55]. Interestingly, although Msi1 is expressed in putative ISCS, in knockout mice lacking this marker, no defects in the development of the intestine are detected[56]. Taken together, these observations demonstrated that Msi1 is not a specific ISCS marker, but is expressed in both ISCS and in their early progeny[57].

To sum up, different markers point to distinct stem cells within the crypt: the marker Lgr5 points to the crypt base columnar cells located in between the Paneth cells at the crypt bottom[22], whereas the markers BMI1 identify the +4 position in the crypt, just above the Paneth cells[31]. The existence and interdependency of these different types of ISCs remain a matter of debate.

STEM CELL NICHE: HOMEOSTASIS AND MORPHOGENESIS SIGNALS

A key role in the dynamics of ISCS is ascribed to the niche, a complex and dynamic setting, that adapts in response to environmental stimuli and provides the cells essential signals, including the morphogenetic pathways, such as Wnt, Notch, bone morphogenetic proteins (BMPs) and Hedgehog[21,58-63]. The microenvironment of the niche surrounding ISCs features extracellular matrix, neural cells, lymphocytes, macrophages, endothelial cells, fibroblasts, smooth muscle cells, and myofibroblasts, that generate signals able to regulate stem cells properties and behavior[64-67].

A wide range of evidence indicates that the Wnt pathway has a crucial role in intestinal proliferation and ISC maintenance[68-74].

The Wnt pathway molecules are evolutionary conserved intracellular signaling molecules which regulate cellular fate in the crypt-villus axis in normal gut epithelium, and are implicated in stem cells self-renewal[75]. Indeed, loss of Wnt signaling in vivo effectively blocks cell proliferation in the intestinal crypts, destroying the epithelium[76]. Moreover, when the Wnt secretion inhibitor (IWP1) was added to organoids, the LacZ signal derived from Lgr5+ cells, was completely lost, and the proliferation was inhibited; this inhibition could be overcome administrating exogenous Wnt3A[76]. Recent evidences have also demonstrated that Paneth cells residing next to ISCs are crucial for their maintenance and serve as the stem cell niche[77]. Paneth cells are known to secrete a variety of bactericidal products, such as cryptidins/defensins and lysozyme, epidermal growth factor (EGF), transforming growth factor β (TGF-β), and represent the main sources of Wnt3a. Indeed, the ablation of Paneth cells, decreases the number of ISCs in the crypt[78] confirming that an active Wnt signal is crucial for ISC maintenance[79].

A Wnt signaling gradient exists along the crypt-villus axis. When cells migrate away from the Wnt source at the base of the crypt, they progressively lose their proliferative capacity and differentiate. The activity of the Wnt pathway, in conjunction with other pathways such as Notch and bone morphogenetic protein (BMP), is vital for the proper organization of the colic epithelium.

In the small intestine, Notch activity determines lineage differentiation between enterocytes and secretory cell differentiation; indeed, Notch inhibition results in an increase of goblet cells, while its activation results in goblet cells depletion[78]. Recent data support the idea that Notch promotes proliferation when Wnt activity is high, while induces enterocyte differentiation when Wnt activity decreases at the top of the crypt[29]. Given that Notch receptors are membrane-bound, it would appear that only the neighboring Paneth cells can maintain active Notch signaling in Lgr5 stem cells.

BMP belongs to a family of ligands which comprises BMP and TGF-β family members, acting through the SMAD intracellular signaling cascade[80-82].

In the intestine, BMP2 and BMP3, are expressed by mesenchymal cells and are able to arrest proliferation at the crypt-villus edge, rather than promoting differentiation[29].

In fact, both mice lacking the BMP receptor (Bmpr1a), and mice overexpressing the BMP inhibitor noggin, present hyperproliferation and crypt fission[80].

Under physiological conditions, the amount of stem cells within the niche remains constant, thus these processes need to be highly regulated, probably through negative feedback mechanisms[40]. In fact, stem cells may divide: (1) asymmetrically, giving rise to another stem cell, which remains in the niche, and to a daughter cell which form a progenitor cell, that migrates upwards in the crypt and differentiate into a mature element; and (2) symmetrically, giving rise to two daughter stem cells, or two daughter non-stem progenitor cells, the latter phenomenon leading to stem cells exhaustion[75].

Overall, the current scenario indicates a niche organized into a complex network of morphogenetic signals, each crucial for ISCS and crypt maintenance[28].

STEM CELLS DERIVED ORGANOIDS: THERAPEUTIC APPLICATIONS FOR GASTROINTESTINAL DISEASES

Rapid progress in the field of intestinal stem cell biology now lends promise to the application of adult stem cell therapy in gastroenterology. The apparently unlimited scale at which these stem cells can be expanded in vitro offers particularly exciting therapeutic possibilities[83-85].

In particular, organoids, derived from in vitro expansion of a single adult colonic stem cell, can be used to repair damaged colon tissue.

Indeed, as discussed above, intestinal organoid cultures, with a gut like structure, and containing all epithelial cell types, can be derived from single Lgr5+ sorted stem cells[86]; the resulting organoids can be expanded efficiently and over long periods of time, without losing tissue identity. To date, protocols for the efficient generation of organoids from stomach, human small bowel and colon have been developed[87-90].

Interestingly, the growth factors used to supplement the culture medium are the natural growth factors to which the stem cells are exposed in vivo, suggesting a high clinical-grade biocompatibility of this approach[87]. Moreover, no genetic manipulation required, making the entire procedure extremely safe.

As a first step toward the development of stem cell transplantation, it has been shown that, using the colonic organoids culture system, significant amounts of tissue can be grown in vitro from a single adult colon stem cell[91,92].

Colon organoids were reintroduced into superficially damaged recipient colons of immunocompromised (rag2-/-) mice, pretreated with dextran sulphate sodium (DSS), which induces superficial mucosal lesions. The engrafted organoids RFP+, were able to readily integrate into the existing epithelium (RFP-), and generated histologically and functionally normal crypts containing all differentiated cell types, covering the area that lacked epithelium in recipient mice. At 4 wk after transplantation, the donor-derived cells constituted a single-layered epithelium, which formed self-renewing crypts that were functionally and histologically normal. In long term studies, carried out at 25 wk after transplantation, the grafts still contributed to the epithelium without any sign of adenomatous or dysplastic transformation[91,93].

Moreover, transplanted mice displayed a significant lower weight loss than control mice[91]. These data showed the feasibility of colon stem-cell therapy based on the in vitro expansion of a single adult colonic stem cell; graft rejection can be managed by standard approaches, i.e., by leukocyte antigens matching of donor and acceptor and by immunosuppressive therapy, as currently used for organ transplantation.

Protocols have also been developed to expand human small intestine and colon organoids from small biopsies[94]. As a first application, Cleavers and his collaborators have transplanted the organoid-derived small intestinal epithelium into the bowel of patients affected by the microvillus inclusion disease[48]. This is a rare hereditary defect of the enterocyte brush border resulting in insufficient nutrients’ assimilation[95-97], requiring colon transplantation as the unique therapeutic strategy. Intestinal organoids technology may allow a novel venue into gene therapy approaches, that involves the introduction of DNA sequences into the genomes of cells of the pertinent patient. The achievement of a safe gene transfer represents the major hurdle, which has largely hampered the introduction of gene therapy into the clinic despite three decades of intensive efforts. On this regard, retrovirus- and lentivirus-mediated gene transfer has already been proven to be feasible in organoids systems[98,99].

These viral vectors are though associated to documented risk for insertional mutagenesis. As organoids can be grown from single sorted stem cells, one could envisage an approach in which individual stem cells are analyzed after integration of the recombinant DNA sequences. Only stem cells with safe integrations could then be expanded clonally for subsequent transplantation.

Overall, adult stem-cell therapy holds promise for the treatment of gastrointestinal diseases, using tissues “harvested” from a single living donor, overcoming the difficulties of the organ transplantation, that is still limited by the availability of donor.

Clinical application of this protocol still waits the translation of the technical procedures to the good clinical practice standards, to generate the adequate amounts of tissue to treat human subjects, and the development of efficient transplantation approaches.

CONCLUSION

ISCs could be reasonably considered the key players that orchestrate the high-rate regenerative capacity of the intestinal epithelium. The understanding of the interplay between the ISCs and their niche, led by a complex molecular network, will pave the way for the future development ISCS based therapy especially to the application in gastroenterology.

Footnotes

P- Reviewer Ip YT S- Editor Song XX L- Editor A E- Editor Zhang DN

References
1.  Lin SA, Barker N. Gastrointestinal stem cells in self-renewal and cancer. J Gastroenterol. 2011;46:1039-1055.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
2.  Heath JP. Epithelial cell migration in the intestine. Cell Biol Int. 1996;20:139-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 139]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
3.  Puglisi MA, Tesori V, Lattanzi W, Piscaglia AC, Gasbarrini GB, D’Ugo DM, Gasbarrini A. Therapeutic implications of mesenchymal stem cells in liver injury. J Biomed Biotechnol. 2011;2011:860578.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
4.  Lattanzi W, Geloso MC, Saulnier N, Giannetti S, Puglisi MA, Corvino V, Gasbarrini A, Michetti F. Neurotrophic features of human adipose tissue-derived stromal cells: in vitro and in vivo studies. J Biomed Biotechnol. 2011;2011:468705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
5.  Bernardini C, Saulnier N, Parrilla C, Pola E, Gambotto A, Michetti F, Robbins PD, Lattanzi W. Early transcriptional events during osteogenic differentiation of human bone marrow stromal cells induced by Lim mineralization protein 3. Gene Expr. 2010;15:27-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
6.  Saulnier N, Puglisi MA, Lattanzi W, Castellini L, Pani G, Leone G, Alfieri S, Michetti F, Piscaglia AC, Gasbarrini A. Gene profiling of bone marrow- and adipose tissue-derived stromal cells: a key role of Kruppel-like factor 4 in cell fate regulation. Cytotherapy. 2011;13:329-340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
7.  Saulnier N, Lattanzi W, Puglisi MA, Pani G, Barba M, Piscaglia AC, Giachelia M, Alfieri S, Neri G, Gasbarrini G. Mesenchymal stromal cells multipotency and plasticity: induction toward the hepatic lineage. Eur Rev Med Pharmacol Sci. 2009;13 Suppl 1:71-78.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Alenzi FQ, Alenazi BQ, Ahmad SY, Salem ML, Al-Jabri AA, Wyse RK. The haemopoietic stem cell: between apoptosis and self renewal. Yale J Biol Med. 2009;82:7-18.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Mountford JC. Human embryonic stem cells: origins, characteristics and potential for regenerative therapy. Transfus Med. 2008;18:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 74]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
10.  Hutton JF, D’Andrea RJ, Lewis ID. Potential for clinical ex vivo expansion of cord blood haemopoietic stem cells using non-haemopoietic factor supplements. Curr Stem Cell Res Ther. 2007;2:229-237.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Kapoor S, Wilson AG, Sharrack B, Lobo A, Akil M, Sun L, Dalley CD, Snowden JA. Haemopoietic stem cell transplantation--an evolving treatment for severe autoimmune and inflammatory diseases in rheumatology, neurology and gastroenterology. Hematology. 2007;12:179-191.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Hough R, Cooper N, Veys P. Allogeneic haemopoietic stem cell transplantation in children: what alternative donor should we choose when no matched sibling is available? Br J Haematol. 2009;147:593-613.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
13.  Gimble JM, Bunnell BA, Casteilla L, Jung JS, Yoshimura K. Phases I-III Clinical Trials Using Adult Stem Cells. Stem Cells Int. 2011;2010:604713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
14.  Sanz-Ruiz R, Gutiérrez Ibañes E, Arranz AV, Fernández Santos ME, Fernández PL, Fernández-Avilés F. Phases I-III Clinical Trials Using Adult Stem Cells. Stem Cells Int. 2010;2010:579142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 38]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
15.  Bjerknes M, Cheng H. Intestinal epithelial stem cells and progenitors. Methods Enzymol. 2006;419:337-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 327]  [Cited by in F6Publishing: 301]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
16.  Marshman E, Booth C, Potten CS. The intestinal epithelial stem cell. Bioessays. 2002;24:91-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 426]  [Cited by in F6Publishing: 403]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
17.  Molofsky AV, Pardal R, Morrison SJ. Diverse mechanisms regulate stem cell self-renewal. Curr Opin Cell Biol. 2004;16:700-707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 237]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
18.  Casali A, Batlle E. Intestinal stem cells in mammals and Drosophila. Cell Stem Cell. 2009;4:124-127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 133]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
19.  Leedham SJ, Brittan M, McDonald SA, Wright NA. Intestinal stem cells. J Cell Mol Med. 2005;9:11-24.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Wong MH, Saam JR, Stappenbeck TS, Rexer CH, Gordon JI. Genetic mosaic analysis based on Cre recombinase and navigated laser capture microdissection. Proc Natl Acad Sci USA. 2000;97:12601-12606.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Shaker A, Rubin DC. Intestinal stem cells and epithelial-mesenchymal interactions in the crypt and stem cell niche. Transl Res. 2010;156:180-187.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Barker N, Clevers H. Leucine-rich repeat-containing G-protein-coupled receptors as markers of adult stem cells. Gastroenterology. 2010;138:1681-1696.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  van der Flier LG, Clevers H. Stem cells, self-renewal, and differentiation in the intestinal epithelium. Annu Rev Physiol. 2009;71:241-260.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Barker N, van de Wetering M, Clevers H. The intestinal stem cell. Genes Dev. 2008;22:1856-1864.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 433]  [Cited by in F6Publishing: 451]  [Article Influence: 28.2]  [Reference Citation Analysis (0)]
25.  Simons BD, Clevers H. Stem cell self-renewal in intestinal crypt. Exp Cell Res. 2011;317:2719-2724.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Clevers HC, Bevins CL. Paneth cells: maestros of the small intestinal crypts. Annu Rev Physiol. 2013;75:289-311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 427]  [Cited by in F6Publishing: 448]  [Article Influence: 40.7]  [Reference Citation Analysis (0)]
27.  Garabedian EM, Roberts LJ, McNevin MS, Gordon JI. Examining the role of Paneth cells in the small intestine by lineage ablation in transgenic mice. J Biol Chem. 1997;272:23729-23740.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Li L, Clevers H. Coexistence of quiescent and active adult stem cells in mammals. Science. 2010;327:542-545.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Medema JP, Vermeulen L. Microenvironmental regulation of stem cells in intestinal homeostasis and cancer. Nature. 2011;474:318-326.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Potten CS, Kovacs L, Hamilton E. Continuous labelling studies on mouse skin and intestine. Cell Tissue Kinet. 1974;7:271-283.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Sangiorgi E, Capecchi MR. Bmi1 is expressed in vivo in intestinal stem cells. Nat Genet. 2008;40:915-920.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Bjerknes M, Cheng H. Clonal analysis of mouse intestinal epithelial progenitors. Gastroenterology. 1999;116:7-14.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Scoville DH, Sato T, He XC, Li L. Current view: intestinal stem cells and signaling. Gastroenterology. 2008;134:849-864.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449:1003-1007.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, van Es JH, Abo A, Kujala P, Peters PJ. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262-265.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  van der Flier LG, van Gijn ME, Hatzis P, Kujala P, Haegebarth A, Stange DE, Begthel H, van den Born M, Guryev V, Oving I. Transcription factor achaete scute-like 2 controls intestinal stem cell fate. Cell. 2009;136:903-912.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF, Morrison SJ. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature. 2003;425:962-967.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Chatoo W, Abdouh M, Duparc RH, Bernier G. Bmi1 distinguishes immature retinal progenitor/stem cells from the main progenitor cell population and is required for normal retinal development. Stem Cells. 2010;28:1412-1423.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Konuma T, Oguro H, Iwama A. Role of the polycomb group proteins in hematopoietic stem cells. Dev Growth Differ. 2010;52:505-516.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Yan KS, Chia LA, Li X, Ootani A, Su J, Lee JY, Su N, Luo Y, Heilshorn SC, Amieva MR. The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proc Natl Acad Sci USA. 2012;109:466-471.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Weigmann A, Corbeil D, Hellwig A, Huttner WB. Prominin, a novel microvilli-specific polytopic membrane protein of the apical surface of epithelial cells, is targeted to plasmalemmal protrusions of non-epithelial cells. Proc Natl Acad Sci USA. 1997;94:12425-12430.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Miraglia S, Godfrey W, Yin AH, Atkins K, Warnke R, Holden JT, Bray RA, Waller EK, Buck DW. A novel five-transmembrane hematopoietic stem cell antigen: isolation, characterization, and molecular cloning. Blood. 1997;90:5013-5021.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Yin AH, Miraglia S, Zanjani ED, Almeida-Porada G, Ogawa M, Leary AG, Olweus J, Kearney J, Buck DW. AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood. 1997;90:5002-5012.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Lindgren D, Boström AK, Nilsson K, Hansson J, Sjölund J, Möller C, Jirström K, Nilsson E, Landberg G, Axelson H. Isolation and characterization of progenitor-like cells from human renal proximal tubules. Am J Pathol. 2011;178:828-837.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Richardson GD, Robson CN, Lang SH, Neal DE, Maitland NJ, Collins AT. CD133, a novel marker for human prostatic epithelial stem cells. J Cell Sci. 2004;117:3539-3545.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Zhu L, Gibson P, Currle DS, Tong Y, Richardson RJ, Bayazitov IT, Poppleton H, Zakharenko S, Ellison DW, Gilbertson RJ. Prominin 1 marks intestinal stem cells that are susceptible to neoplastic transformation. Nature. 2009;457:603-607.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Snippert HJ, van Es JH, van den Born M, Begthel H, Stange DE, Barker N, Clevers H. Prominin-1/CD133 marks stem cells and early progenitors in mouse small intestine. Gastroenterology. 2009;136:2187-2194.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 176]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
48.  Okano H, Imai T, Okabe M. Musashi: a translational regulator of cell fate. J Cell Sci. 2002;115:1355-1359.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Kawahara H, Imai T, Imataka H, Tsujimoto M, Matsumoto K, Okano H. Neural RNA-binding protein Musashi1 inhibits translation initiation by competing with eIF4G for PABP. J Cell Biol. 2008;181:639-653.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Okano H, Kawahara H, Toriya M, Nakao K, Shibata S, Imai T. Function of RNA-binding protein Musashi-1 in stem cells. Exp Cell Res. 2005;306:349-356.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Sakakibara S, Imai T, Hamaguchi K, Okabe M, Aruga J, Nakajima K, Yasutomi D, Nagata T, Kurihara Y, Uesugi S. Mouse-Musashi-1, a neural RNA-binding protein highly enriched in the mammalian CNS stem cell. Dev Biol. 1996;176:230-242.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Siegl G, Gautschi M. Multiplication of parvovirus LuIII in a synchronized culture system. III. Replication of viral DNA. J Virol. 1976;17:841-853.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Kayahara T, Sawada M, Takaishi S, Fukui H, Seno H, Fukuzawa H, Suzuki K, Hiai H, Kageyama R, Okano H. Candidate markers for stem and early progenitor cells, Musashi-1 and Hes1, are expressed in crypt base columnar cells of mouse small intestine. FEBS Lett. 2003;535:131-135.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Siddall NA, McLaughlin EA, Marriner NL, Hime GR. The RNA-binding protein Musashi is required intrinsically to maintain stem cell identity. Proc Natl Acad Sci USA. 2006;103:8402-8407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 89]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
55.  Rezza A, Skah S, Roche C, Nadjar J, Samarut J, Plateroti M. The overexpression of the putative gut stem cell marker Musashi-1 induces tumorigenesis through Wnt and Notch activation. J Cell Sci. 2010;123:3256-3265.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Sakakibara S, Nakamura Y, Yoshida T, Shibata S, Koike M, Takano H, Ueda S, Uchiyama Y, Noda T, Okano H. RNA-binding protein Musashi family: roles for CNS stem cells and a subpopulation of ependymal cells revealed by targeted disruption and antisense ablation. Proc Natl Acad Sci USA. 2002;99:15194-15199.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Montgomery RK, Breault DT. Small intestinal stem cell markers. J Anat. 2008;213:52-58.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Sémont A, François S, Mouiseddine M, François A, Saché A, Frick J, Thierry D, Chapel A. Mesenchymal stem cells increase self-renewal of small intestinal epithelium and accelerate structural recovery after radiation injury. Adv Exp Med Biol. 2006;585:19-30.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Sémont A, Mouiseddine M, François A, Demarquay C, Mathieu N, Chapel A, Saché A, Thierry D, Laloi P, Gourmelon P. Mesenchymal stem cells improve small intestinal integrity through regulation of endogenous epithelial cell homeostasis. Cell Death Differ. 2010;17:952-961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 110]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
60.  Yen TH, Wright NA. The gastrointestinal tract stem cell niche. Stem Cell Rev. 2006;2:203-212.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Ishizuya-Oka A, Hasebe T. Sonic hedgehog and bone morphogenetic protein-4 signaling pathway involved in epithelial cell renewal along the radial axis of the intestine. Digestion. 2008;77 Suppl 1:42-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
62.  Ishizuya-Oka A, Hasebe T, Shimizu K, Suzuki K, Ueda S. Shh/BMP-4 signaling pathway is essential for intestinal epithelial development during Xenopus larval-to-adult remodeling. Dev Dyn. 2006;235:3240-3249.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Ishizuya-Oka A, Ueda S, Amano T, Shimizu K, Suzuki K, Ueno N, Yoshizato K. Thyroid-hormone-dependent and fibroblast-specific expression of BMP-4 correlates with adult epithelial development during amphibian intestinal remodeling. Cell Tissue Res. 2001;303:187-195.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Li L, Xie T. Stem cell niche: structure and function. Annu Rev Cell Dev Biol. 2005;21:605-631.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Ohlstein B, Kai T, Decotto E, Spradling A. The stem cell niche: theme and variations. Curr Opin Cell Biol. 2004;16:693-699.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Nusse R, Fuerer C, Ching W, Harnish K, Logan C, Zeng A, ten Berge D, Kalani Y. Wnt signaling and stem cell control. Cold Spring Harb Symp Quant Biol. 2008;73:59-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 186]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
67.  Lin H. The stem-cell niche theory: lessons from flies. Nat Rev Genet. 2002;3:931-940.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Clevers H. Wnt/beta-catenin signaling in development and disease. Cell. 2006;127:469-480.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Fodde R, Brabletz T. Wnt/beta-catenin signaling in cancer stemness and malignant behavior. Curr Opin Cell Biol. 2007;19:150-158.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Pinto D, Clevers H. Wnt, stem cells and cancer in the intestine. Biol Cell. 2005;97:185-196.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Ogaki S, Shiraki N, Kume K, Kume S. Wnt and Notch signals guide embryonic stem cell differentiation into the intestinal lineages. Stem Cells. 2013;31:1086-1096.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 72]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
72.  Kühl SJ, Kühl M. On the role of Wnt/β-catenin signaling in stem cells. Biochim Biophys Acta. 2013;1830:2297-2306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 75]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
73.  Farin HF, Van Es JH, Clevers H. Redundant sources of Wnt regulate intestinal stem cells and promote formation of Paneth cells. Gastroenterology. 2012;143:1518-1529.e7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 464]  [Cited by in F6Publishing: 475]  [Article Influence: 39.6]  [Reference Citation Analysis (0)]
74.  Kim BM, Mao J, Taketo MM, Shivdasani RA. Phases of canonical Wnt signaling during the development of mouse intestinal epithelium. Gastroenterology. 2007;133:529-538.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Yeung TM, Chia LA, Kosinski CM, Kuo CJ. Regulation of self-renewal and differentiation by the intestinal stem cell niche. Cell Mol Life Sci. 2011;68:2513-2523.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  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;19:379-383.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  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. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature. 2011;469:415-418.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Hardwick JC, Kodach LL, Offerhaus GJ, van den Brink GR. Bone morphogenetic protein signalling in colorectal cancer. Nat Rev Cancer. 2008;8:806-812.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Pellegrinet L, Rodilla V, Liu Z, Chen S, Koch U, Espinosa L, Kaestner KH, Kopan R, Lewis J, Radtke F. Dll1- and dll4-mediated notch signaling are required for homeostasis of intestinal stem cells. Gastroenterology. 2011;140:1230-1240.e1-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 299]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
80.  Hardwick JC, Kodach LL, Offerhaus GJ, van den Brink GR. Bone morphogenetic protein signalling in colorectal cancer. Nat Rev Cancer. 2008;8:806-812.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 152]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
81.  Kodach LL, Wiercinska E, de Miranda NF, Bleuming SA, Musler AR, Peppelenbosch MP, Dekker E, van den Brink GR, van Noesel CJ, Morreau H. The bone morphogenetic protein pathway is inactivated in the majority of sporadic colorectal cancers. Gastroenterology. 2008;134:1332-1341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 125]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
82.  Blanco Calvo M, Bolós Fernández V, Medina Villaamil V, Aparicio Gallego G, Díaz Prado S, Grande Pulido E. Biology of BMP signalling and cancer. Clin Transl Oncol. 2009;11:126-137.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Hu J, Chen X, Zhou Z. [Intestinal stem cells and tissue engineering technique used in treating intestinal diseases]. Zhongguo Xiu Fu Chong Jian Wai Ke Zazhi. 2007;21:175-179.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Day RM. Epithelial stem cells and tissue engineered intestine. Curr Stem Cell Res Ther. 2006;1:113-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
85.  Polak JM, Bishop AE. Stem cells and tissue engineering: past, present, and future. Ann N Y Acad Sci. 2006;1068:352-366.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Barker N, Huch M, Kujala P, van de Wetering M, Snippert HJ, van Es JH, Sato T, Stange DE, Begthel H, van den Born M. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 2010;6:25-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1056]  [Cited by in F6Publishing: 1103]  [Article Influence: 78.8]  [Reference Citation Analysis (0)]
87.  Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, Van Houdt WJ, Pronk A, Van Gorp J, Siersema PD. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141:1762-1772.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Ootani A, Li X, Sangiorgi E, Ho QT, Ueno H, Toda S, Sugihara H, Fujimoto K, Weissman IL, Capecchi MR. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche. Nat Med. 2009;15:701-706.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 616]  [Cited by in F6Publishing: 638]  [Article Influence: 42.5]  [Reference Citation Analysis (0)]
89.  Kim TH, Escudero S, Shivdasani RA. Intact function of Lgr5 receptor-expressing intestinal stem cells in the absence of Paneth cells. Proc Natl Acad Sci USA. 2012;109:3932-3937.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 186]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
90.  Huch M, Dorrell C, Boj SF, van Es JH, Li VS, van de Wetering M, Sato T, Hamer K, Sasaki N, Finegold MJ. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature. 2013;494:247-250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 966]  [Cited by in F6Publishing: 1023]  [Article Influence: 93.0]  [Reference Citation Analysis (0)]
91.  Yui S, Nakamura T, Sato T, Nemoto Y, Mizutani T, Zheng X, Ichinose S, Nagaishi T, Okamoto R, Tsuchiya K. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5⁺ stem cell. Nat Med. 2012;18:618-623.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Sato T, Clevers H. Primary mouse small intestinal epithelial cell cultures. Methods Mol Biol. 2013;945:319-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 183]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
93.  Barker N, van Es JH, Jaks V, Kasper M, Snippert H, Toftgård R, Clevers H. Very long-term self-renewal of small intestine, colon, and hair follicles from cycling Lgr5+ve stem cells. Cold Spring Harb Symp Quant Biol. 2008;73:351-356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 86]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
94.  Li VS, Clevers H. In vitro expansion and transplantation of intestinal crypt stem cells. Gastroenterology. 2012;143:30-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
95.  Müller T, Hess MW, Schiefermeier N, Pfaller K, Ebner HL, Heinz-Erian P, Ponstingl H, Partsch J, Röllinghoff B, Köhler H. MYO5B mutations cause microvillus inclusion disease and disrupt epithelial cell polarity. Nat Genet. 2008;40:1163-1165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 281]  [Cited by in F6Publishing: 293]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
96.  Ruemmele FM, Müller T, Schiefermeier N, Ebner HL, Lechner S, Pfaller K, Thöni CE, Goulet O, Lacaille F, Schmitz J. Loss-of-function of MYO5B is the main cause of microvillus inclusion disease: 15 novel mutations and a CaCo-2 RNAi cell model. Hum Mutat. 2010;31:544-551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 108]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
97.  Ruemmele FM, Schmitz J, Goulet O. Microvillous inclusion disease (microvillous atrophy). Orphanet J Rare Dis. 2006;1:22.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Koo BK, Stange DE, Sato T, Karthaus W, Farin HF, Huch M, van Es JH, Clevers H. Controlled gene expression in primary Lgr5 organoid cultures. Nat Methods. 2012;9:81-83.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Turner VM, Gardam S, Brink R. Lineage-specific transgene expression in hematopoietic cells using a Cre-regulated retroviral vector. J Immunol Methods. 2010;360:162-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]